Enroll-HD
Enroll-HD
Sponsor: CHDI
Enroll-HD is an observational study for HD families actively recruiting in North America, Latin America, Europe, Australia, New Zealand and some countries in Asia. The Enroll-HD dataset and biosamples are available for research. More.
Enroll-HD platform studies
The Enroll-HD study can serve as a platform for other research studies which makes it faster and more efficient for researchers to set up new projects.
HD Clarity
Sponsor: UCL
This is a multi-site cerebrospinal fluid collection initiative to facilitate identification of biomarkers in Huntington’s disease. More.
The HD Young Adult Study (HD-YAS)
Sponsor: UCL
Aim: To find the earliest time point at which any HD-related changes can be found in young-adult gene carriers. More.
Clinical studies
The EHDN supports a wide variety of clinical studies in HD that have either been endorsed by the EHDN or are collaborations with CHDI and other entities.
Current EHDN Endorsed Trials and Studies
Registration ID (CT.gov) | Sponsor | Trial Name | Phase | Investi- gational Product | Mode of Action | Delivery | Treatment Goal | Target Enrolment | Location(s) | Status |
---|---|---|---|---|---|---|---|---|---|---|
NCT06585449 | Alnylam Pharma-ceuticals | ALN-HTT02-001 | 1 | ALN-HTT02 | Htt lowering; siRNA | Intrathecal | Disease modification | 54 | Canada, Germany, US, UK | Not yet recruiting |
NCT05541627 | BrainVectis, a subsidiary of AskBio | ASK-HD-01-CS-101 | 1/2 | AB-1001 | Restoration of cholesterol metabolism dysfunction (AAV delivered gene therapy) | Surgical, intrastriatal | Disease modification | 18 | France | Active, not recruiting |
NCT05686551 | Hoffmann-La Roche | GENERATION HD2 | 2 | Tominersen | Htt lowering; ASO | Intrathecal | Disease modification | 300 | Argentina, Australia, Austria, Canada, Denmark, France, Germany, Italy, New Zealand, Poland, Portugal, Spain, Switzerland, UK, USA | Recruiting |
NCT05358717 | PTC Therapeutics | PIVOT-HD | 2 | PTC518 | Htt lowering; mRNA splicing modifier | Oral | Disease modification | 162 | Australia, Austria, Canada, France, Germany, Italy, Netherlands, New Zealand, Spain, UK, USA | Active, not recruiting |
NCT05655520 | Sage Therapeutics | PURVIEW | 3/OLE* | SAGE-718 | NMDA receptor modulator | Oral | Symptomatic | 300 | Australia, Canada, UK, USA | Recruiting |
NCT05243017 | UniQure | HD GeneTRX2 | 1b/2 | rAAV5-miHTT | Htt lowering; miRNA AAV delivered gene therapy | Surgical, intrastriatal | Disease modification | 15 | Germany, Poland, UK | Recruiting |
Completed EHDN Endorsed Trials and Studies
Registration ID (CT.gov) | Sponsor | Trial Name | Phase | Investi- gational Product | Mode of Action | Delivery | Treatment Goal | Target Enrolment | Location(s) | Status |
---|---|---|---|---|---|---|---|---|---|---|
NCT00146211 | Amarin Neuroscience Ltd | TREND-HD | 3 | Ethyl-EPA | Unknown | Oral | Symptomatic | 300 | North America, Europe | Completed |
ISRCTN12103732 | Cardiff University | DOMINO-HD | N/A | N/A | N/A | N/A | Feasibility and symptomatic | 300 | Poland, Spain, Switzerland, UK | Completed |
NCT03344601 | Cardiff University | PACE-HD | N/A | Physical Therapy | N/A | N/A | Feasibility and acceptance | 116 | UK, US, Spain, Germany | Completed |
ISRCTN94284668 | Cardiff University | TRAIN-HD | 2 | Physical therapy | Unknown | Physical activity | Symptomatic | 30 | UK | Completed |
ISRCTN65378754 | Cardiff University | ENGAGE-HD | N/A | Physical therapy | Unknown | Physical activity | Feasibility | 46 | UK | Completed |
ISRCTN11392629 | Cardiff University | ExeRT-HD | 2 | Physical therapy | Unknown | Physical activity | Feasibility | 32 | UK | Completed |
NCT01914965 | Charite University | ACTION-HD | 2 | Bupropion | Dual inhibition of norepinephrine and dopamine reuptake | Oral | Symptomatic | 40 | Germany | Completed |
NCT01357681 | Charite University | ETON-STUDY | 2 | Epigallocatechin Gallate | Unknown | Oral | Disease modification | 40 | Germany | Completed |
NCT02061722 | CHDI Foundation, Inc. | PEARL-HD | 1 | [18F]MNI-659 | PDE10A enzyme availability | Intravenous | Biomarker | 90 | Denmark, Netherlands, Norway, Sweden | Completed |
NCT02956148 | CHDI Foundation, Inc. | LONG PDE10A | 1 | [18F]MNI-659 | PDE10A enzyme availability | Intravenous | Biomarker | 45 | Denmark, Netherlands, Norway, Sweden | Completed |
NCT01590589 | EHDN | REGISTRY - an Observational Study of the European Huntington's Disease Network (EHDN) | N/A | N/A | N/A | N/A | Natural History | 14860 | Europe | Completed |
NCT02535884 | Heinrich-Heine University, Duesseldorf | HD-DBS | 2 | Deep brain stimulation | High-frequency stimulation of the Globus Pallidus | Surgical, implant | Symptomatic | 50 | Austria, Germany, Switzerland | Completed |
NCT03761849 | Hoffmann-La Roche | GENERATION-HD1 | 3 | RO7234292 (RG6042) | htt lowering ASO | Intrathecal | Disease modification | 899 | North America, Europe, Oceania, Japan, Latin America | Completed |
NCT03842969 | Hoffmann-La Roche | GEN-EXTEND | OLE* | RO7234292 (RG6042) | htt lowering ASO | Intrathecal | Disease modification | 236 | North America, Europe | Completed |
NCT02519036 | Ionis Pharmaceuticals, Inc. | IONIS-HTTRx | 1 | ISIS 443139 (tominersen) | htt lowering ASO | Intrathecal | Disease modification | 46 | Canada, Germany, UK | Completed |
NCT00920946 | Medivation, Inc | Horizon | 3 | Dimebon/ latrepirdine | Developed as antihistamine, /cholinesterase inhibitor | Oral | Disease modification | 403 | US, UK, Denmark, Canada, Australia, Germany, Sweden | Completed |
NCT05111249 | Novartis | VIBRANT-HD | 2b, OLE* | Branaplam | mRNA splicing modifier | Oral | Disease modification | 75 | Belgium, Canada, Germany, Hungary, Italy, Spain, UK, USA | Completed |
NCT02197130 | Pfizer | Amaryllis | 2 | PF-02545920 | Phosphodiesterase 10A Inhibitor | Oral | Disease modification | 272 | North America, Europe | Completed |
NCT04556656 | Prilenia Therapeutics | PROOF-HD | 3/OLE* | Pridopidine | Sigma-1 receptor agonist | Oral | Disease modification | 480 | Austria, Canada, Czech Republic, France, Germany, Italy, Netherlands, Poland, Spain, Switzerland, UK, USA | Completed |
NCT05107128 | Sage Therapeutics | DIMENSION | 2 | SAGE-718 | NMDA receptor modulator | Oral | Symptomatic | 178 | Australia, Canada, UK, USA | Completed |
NCT05358821 | Sage Therapeutics | SURVEYOR | 2 | SAGE-718 | NMDA receptor modulator | Oral | Symptomatic | 80 | Canada, USA | Completed |
NCT01521585 | Siena BioTech | Selisistat | 2 | Selisistat, SEN0014196 | SirT1 inhibitor | Oral | Disease modification | 144 | Germany | Completed |
NCT05475483 | SOM Biotech | SOMCT03 | 2b | SOM3355 (bevantolol hydrochloride) | VMAT2 inhibitor | Oral | Symptomatic | 129 | France, Germany, Italy, Poland, Spain, Switzerland, UK | Completed |
NCT02215616 | Teva | LEGATO-HD | 2 | Laquinimod | Immunomodulatory and neuroprotective effects | Oral | Disease modification | 352 | North America, Europe | Completed |
NCT02006472 | Teva | PRIDE-HD | 2 | Pridopidine | Dopaminergic stabiliser | Oral | Symptomatic | 408 | North America, Europe, Australia | Completed |
NCT02494778 | Teva | OPEN PRIDE-HD | OLE* | Pridopidine | Dopaminergic stabiliser | Oral | Symptomatic | 248 | North America, Europe, Australia | Completed |
NCT04406636 | Triplet Therapeutics/ CHDI Foundation, Inc | SHIELD-HD | N/A | N/A | N/A | N/A | Natural History | 70 | US, Germany, UK, Canada, France | Completed |
NCT05032196 | Wave Life Sciences | SELECT-HD | 1b/2a | WVE-003 | Allele-selective htt lowering ASO | Intrathecal | Disease modification | 54 | Australia, Canada, France, Germany, Italy, Netherlands, Poland, Spain, Switzerland, UK | Completed |
NCT03225833 | Wave Life Sciences | PRECISION-HD1 | 1b/2a | WVE-120101 | Allele-selective htt lowering ASO | Intrathecal | Disease modification | 61 | Australia, Canada, Denmark, France, Germany, Poland, UK | Completed |
NCT03225846 | Wave Life Sciences | PRECISION-HD2 | 1b/2a | WVE-120102 | Allele-selective htt lowering ASO | Intrathecal | Disease modification | 88 | Australia, Canada, Denmark, France, Germany, Poland, UK | Completed |
NCT04617847 | Wave Life Sciences | PRECISION-HD1 OLE* | 1b/2a, OLE* | WVE-120101 | Allele-selective htt lowering ASO | Intrathecal | Disease modification | 27 | Australia, Canada, Denmark, France, Germany, Poland, UK | Completed |
NCT04617860 | Wave Life Sciences | PRECISION-HD2 OLE* | 1b/2a, OLE* | WVE-120102 | Allele-selective htt lowering ASO | Intrathecal | Disease modification | 36 | Australia, Canada, Denmark, France, Germany, Poland, UK | Completed |
Projects supported by EHDN
The EHDN supports basic and clinical research in HD by offering seed funds, making available the clinical data and biosamples of several thousands participants from the two observational studies Enroll-HD and Registry, and creating a collaborative platform through the EHDN working groups.
Basic Research Projects:
Modifiers
Enteric Nervous System in Huntington’s Disease (HD): neglected phenotypes as possible new disease modifiers
While brain alterations have been deeply characterized in HD, a number of less described, but strongly disabling symptoms, such as weight loss and gastrointestinal (GI) dysfunction, significantly impact patients’ life quality. Here, we propose to characterize the alterations in enteric nervous system (ENS) – controlling GI motility, secretions, and functionality – taking advantage of the HD mice, which faithfully mirror the genetics of the HD mutation. This study will be crucial not only for disentangling a new piece of HD biology, but furthermore to identify new targets to alleviate HD peripheral symptoms, thus ultimately improving HD patients’ life quality.
Author: Marta Biagioli
Approved: January 2023
Prenatal effects of mutant Huntingtin
Even though most Huntington’s (HD) symptoms appear in later life, the genetic cause is present from conception and may impact fetal and placental development. Early developmental changes may sow the seed for clinical symptoms experienced in adulthood, but very little is known about the role of huntingtin during this time.
This project will use a Huntington’s mouse model to look for differences in the embryonic brain and its placenta for the first time.
Evidence that mutant Huntingtin could alter how the placenta influences brain development could provide a new direction for potential preventative treatment options.
Author: David Harrison
Approved: January 2022
Investigation into the interplay between prion-like spreading of mutant huntingtin and somatic CAG expansion
HD results from inheritance of a mutant mHTT gene with CAG repeats of >=36. Additionally, through a patient’s life, two processes increase the mHTT pathogenic burden. Firstly, continued repeat expansion occurs such that tissues become mosaic with increasing numbers of cells expressing mHTT with much longer CAG repeats. Secondly, pathogenic mHTT protein transfers from one cell to another to spread disease across brain regions. This project uses stem cell models to address a novel hypothesis that mHTT protein transfer interacts with DNA repair modifier pathways and acts as a driver to increase CAG repeat expansion rates in receiving cells.
Author: Nicholas Allen
Approved: May 2021
Examining Fyn/Src in Huntington’s disease cell models
Huntington’s disease (HD) is a genetic neurodegenerative disorder caused by expression of mutant huntingtin (mHTT) that largely affects the striatum and later the cortex. Striatal glutamatergic synapse deregulation and mitochondrial dysfunction linked to redox changes are relevant HD pathological events. Interestingly, these processes are also influenced by Fyn/Src kinases. However, the role of Fyn/Src in HD-related neuronal dysfunction is scarce. Thus, in this study, we aim to define the impact of Fyn/Src levels in HD models by focusing on glutamatergic dendrites and as mitochondrial modulator(s). This project is expected to expand the current understanding of early neuropathological mechanism(s) in HD.
Author: Ana Cristina Carvalho REGO
Approved: January 2021
Astrocytes at the hub of neuronal dysfunction in Huntington´s disease: Dissecting the role of ARMS/kidins 220 on astrocyte secretome
Most aspects of nervous system function rely on neuron-glia crosstalk, particularly astrocytes. This neuronal-astrocyte communication implies the release of different factors that may influence both cell types and modulate neuronal function. This proposal is aimed to understand the contribution of astrocyte-secretome alterations in HD striatal pathology. We are interested to decipher whether HD pathology relies on neuronal-autonomous processes or whether astrocyte-neuron crosstalk is playing an essential role. We postulate that deficient BDNF release by astrocytes as well as abnormal astrocytic inflammatory signature due to aberrant ARMS/kidins220 expression would play a major role in loss of striatal neuroprotection and therefore striatal dysfunction.
Author: Silvia Gines Padrós
Approved: May 2020
circHTT, a circular RNA from the HTT locus: functional characterization and possible therapeutic applications
Innovative strategies aimed at lowering mutant huntingtin are under careful investigation as therapeutic intervention for Huntington’s Disease (HD). Our work frames into this focus area and proposes to evaluate circHTT, highly stable circularized RNA molecule originated from the HTT transcript, as possible modulator of mutant huntingtin levels. Thus, our work represents a compelling hypothesis to define new HD biology, but also, to potentially identify a new tool of intervention to modulate HD pathogenesis.
Author: Marta Biagioli
Approved: January 2019
Whole genome sequencing to identify novel rare variants that modify Huntington’s disease onset
The overall goal of this study is to identify genetic variants with effects on HD disease onset. We will perform whole-genome sequencing to identify novel variants, which while rare in the general population, may be important modifiers of age of onset in HD. The power to detect association will be maximized by using the HD cohort collected at the University of Copenhagen, an exclusive family-based HD cohort, and by implementing a careful study design and advanced bioinformatic approaches to analyze the data. This work will identify novel HD modifier genes and provide new basic mechanistic insights into the disease etiology of HD and will thus open new venues for targeted drug design.
Authors: Kristina Becanovic, Anne Nørremølle
Approved: January 2019
Role of primary cilia in striatal neurons for their vulnerability to degeneration in Huntington’s disease
Primary cilia are non-motile microtubule-based organelles resembling a cellular antenna that can act as a reservoir for mutant huntingtin (mHTT). Increased mHTT results in increased ciliogenesis. Here we will test the hypothesis that primary cilia pathology plays a role in the differential vulnerability of striatal medium spiny neurons (MSNs) in HD. We will monitor neuron- and stage-specific changes of primary cilia structure in HD mouse models. Concomitantly, we will analyze the neuropathological effects of primary cilia deficits in HD mice lacking primary cilia in MSNs. The results could help to identify new factors causing MSN degeneration and modifying HD progression.
Author: Rosanna Parlato
Approved: April 2017
The role of microRNA regulation in a Drosophila model of Huntington Disease
Recent studies in mice, monkeys and humans show that small non-coding RNAs (microRNAs) are differentially expressed in Huntington Disease (HD) brains. Yet, the biological roles exerted by microRNAs in HD remain largely unclear. Here we investigate this problem using a Drosophila model of HD where we will test the roles played by microRNAs on the disease. Drosophila is an excellent system to study this problem due to its unmatched genetics, imaging tools and the detailed understanding of neural development in this organism. Our work will contribute to establish whether microRNA-based approaches might be of potential use in future HD therapies.
Author: Claudio Alonso
Approved: May 2016
The role of SMAD-dependent TGFβ signalling in transcriptional dysregulation in early stage models of HD
SMAD proteins are major regulators of gene expression that act within the transforming growth factor-beta (TGFβ) signalling pathway. Preliminary data has revealed potential differences in this pathway in a cell model of HD. This project aims to characterise the role of SMAD proteins in mouse and human cell models of HD by comparing their expression, localisation and activity, as well as by manipulating the TGFβ pathway in order to determine whether it may contribute to the altered gene expression that is observed in HD.
Author: Kathryn Bowles
Approved: January 2014
Unbiased prioritarization of neuroprotective targets for HD
A project developed by the Biological Modifiers Working Group at EHDN
The aim of this project is to develop data integration and network analysis to prioritize neuroprotective targets in HD. Huntington’s disease is extensively studied thanks to models that were developed in several species and that recapitulate complementary components of HD pathogenesis. Genome-wide analyses in these models have generated a large amount of data with high potential for target selection. The comprehensive and unbiased integration of ‘omics data’ on HD will allow better decisions in candidate target selection to be reached. Selecting for gene networks, modules and strings that are consistently highlighted by different model systems as ‘targets’ and/or ‘modifiers’ of mutant huntingtin activity may provide a strong basis to develop neuroprotective strategies that may be efficient against several stages and/or key features of HD. This concerted action involves the laboratories of Juan Botas, Bob Hughes, Lesley Jones, Ruth Luthi-Carter, Christian Neri and Shen Zhang.
Author: Christian Neri
Approved: January 2009
Biomarkers
Exploring the Role of Meningeal Immune Alteration in Huntington’s Disease
At the Crossroads of Brain and Body: Exploring the Meninges in HD
This research explores the role of meningeal immunity in Huntington’s disease (HD). While the immune system and inflammation are known to influence HD, the specific involvement of meningeal immunity has not yet been studied. Using a mouse model and flow cytometry analysis, we examine changes in meningeal immune cells. The meninges, previously seen as just a protective barrier, are now known to play a key role in brain health, immune surveillance, and inflammation. By studying this in the context of HD, our work aims to uncover new insights into HD’s complex pathology.
Author: Maria Björkqvist
Approved: January 2024
A pilot study of a novel molecular assay to quantify DNA repair synthesis in the HTT exon 1 as readout of somatic instability in Huntington´s Disease
Huntington’s disease (HD) worsens as CAG repeats in the huntingtin gene (HTT) expand. DNA repair might be involved, making it a target for treatments. But measuring these changes in peripheral tissues is tough. We propose a new test based on the idea that if DNA repair in HTT repeats or nearby happens more often then the repeat itself gets longer. In this project we will explore this innovative approach that measures the incorporation of a molecule called EdU during repair activity of HTT exon1. This new assay might offer a potentially new and sensitive way to track HD progression in peripheral tissues such as blood and skin fibroblasts, even before onset.
Author: Veronica Britto
Approved: April 2023
Identification of regional neuronal and glial cell-specific proteome disturbances in HD mice
The brain cells including neurons, astrocytes, microglia, and oligodendrocytes play specific roles in Huntington’s disease, but the extent of their contributions to disease progression is not completely clear. We aim to identify regional and cell-specific protein changes to pinpoint causal events leading to selective cellular and regional impairment. Using mass-spectrometry, proteins from striatal and cortical brain cells isolated from adult HD mice will be investigated to advance our understanding of the complex interplay between these cells, define new targets for therapeutic intervention, and discover novel biomarkers for monitoring disease progression.
Author: Niels Henning Skotte
Approved: January 2023
Bayesian deep-learning-based de-novo prediction and affinity-based mass spectrometry identification of protein interactors of huntingtin and HAP40
Until recently, predicting the shape of a protein from its amino acid sequence was considered impossible. However, scientists have recently achieved this with using artificial intelligence (AI). With the aim, to identify new proteins interacting with Huntingtin (HTT) and its partner protein huntingtin-associated protein 40 (HAP40), both proteins forming a tight complex in cells, we will generate a novel computational model for predicting such interactions and we will experimentally verify the data in the laboratory. In parallel to this work on artificial intelligence, we will also use biochemical methods as the starting point to identify novel protein interactors of HTT and HAP40. The confirmation of known and the identification of so far unknown interactors may help to understand their function in health and disease, connected to HTT and HAP40, to discover new HD biomarkers and to identify new therapeutic targets.
Author: Stefan Kochanek
Approved: May 2022
Development and evaluation of a method for live-cell imaging of Ca2+ fluxes in myoblasts
There is no process in the cell that does not involve intracellular calcium (Ca2+). Ca2+ acts as a signaling molecule, and in mature muscles, Ca2+ rises control cell contractility. The fusion of satellite cells forms the skeletal muscle, and the satellite cells are responsible for both the development and renewal of muscles. Thus the loss of Ca2+ homeostasis may be causative of muscle pathology observed in Huntington’s disease (HD). We will establish a quantitative analytical platform to investigate intracellular Ca2+ fluxes in live satellite cells from HD mouse models and HD patients to analyze the changes during disease progression.
Author: Rana Soylu Kucharz
Approved: May 2020
Exploring striatal postsynaptic SAPAP3 in Huntington’s disease
SAPAP3 is highly expressed in striatum, a brain area selectively affected in Huntington’s disease (HD). Modified SAPAP3 was associated with obsessive-compulsive disorder, an early/pre-motor symptom exhibited by HD patients. However, the role of SAPAP3 in HD was not explored yet. Here we will monitor changes in SAPAP3 in striatal versus cortical postsynaptic density and determine how SAPAP3 affects glutamatergic dendrites and related mitochondrial function and dynamics in animal and cell models of HD, and postmortem HD brain samples. The proposal is expected to unravel SAPAP3 as a novel player in striatal glutamatergic postsynapse and mitochondrial dysfunction in early HD stages.
Author: Ana Cristina Carvalho REGO
Approved: January 2018
The association of nucleolar activity with mutant huntingtin protein species in HD
Transcriptional dysregulation is known in HD. Here we will test the hypothesis that changes in ribosomal DNA (rDNA) transcription in the nucleolus are early signs of transcriptional dysregulation by mutant huntingtin (mHTT). By a systematic analysis of nucleolar transcription in HD models and tissue biopsies at different stages, we aim at identifying a sensitive metabolic marker linked to mHTT. The results could help to test the efficacy of ongoing therapeutic strategies aiming at lowering mHTT levels in specific cells. Concomitantly, the tissue-specific impact of mHTT on rRNA transcriptional dysregulation might account for the still unexplained variability in HD onset.
Author: Rosanna Parlato
Approved: September 2015
The role of microRNA-34 in cancer defense- and HD pathogenic mechanisms
The frequency of cancer in HD patients is only half of what is expected from population studies, and this applies to all forms of cancer. This is also true for other polyQ disease patients, which indicates that CAG repeat expansions have protective effects against the initial steps in tumor formation. The microRNA, miR-34, is well-known as a tumor suppressor that protects against uncontrolled cell growth, and it may be involved, in some situations, in cell death. We investigate the role of miR-34 in cancer defense and neurodegeneration by studying HD patients, transgenic HD mice and a cell model.
Author: Lis Hasholt
Approved: January 2014
Muscle pathology in HD as a source for biomarkers?
Peripheral manifestations of HD may provide an easily accessible and valuable source of HD biomarkers. Muscle wasting is a well-recognised phenomenon in patients with HD and worsens with disease progression. Interestingly, inflammatory processes can be linked to the development of muscle atrophy and immune activation in peripheral blood monocytes can be seen in HD. In this project, we will investigate the relationship between immune activation and muscle wasting in HD. We will study muscle pathology in relation to inflammatory cytokines in HD mouse models and in patients with juvenile onset of HD.
Author: Maria Bjorkqvist
Approved: July 2010
Treatments
Visualising the peptidase IDE at work in Huntington’s Disease
HD is hallmarked by the aggregation of mutant huntingtin protein fragments. Improving the degradation of these toxic fragments would be a therapeutic strategy to delay or prevent disease onset. Recently, we discovered that Insulin Degrading Enzyme (IDE) is well capable to degrade the mutant sequence, yet we lack specific activity based probes (ABPs) to visualize and quantify IDE activity in cells, tissues and biochemical assays. Monique Mulder (LUMC), Sabine Schipper-Krom and Eric Reits (AmsterdamUMC) will develop and synthesize selective IDE ABPs to study the role of IDE in degrading mutant huntingtin in HD neuronal cell and tissue models.
Author: Eric Reits
Approved: May 2022
Nanoparticle based CRSIPR/Cas gene editing system to treat Huntington’s disease
The CRISPR/Cas system represents a pioneering gene editing technology for the treatment of monogenic disorders including HD. One major obstacle is the efficient delivery of the components into the target cells in the brain. In this project, we aim at targeting the HD gene by use of nanoparticles in vitro in murine and porcine cell lines and human ipSCs, as well as in vivo in the HD mouse model. Since we will use the data from this project in subsequent trials in the pig model of HD, the project contributes to the development of safe gene therapy approaches for HD.
Author: Knut Stieger
Approved: January 2018
Developing a new class of antisense oligonucleotides (ASOs) with tricyclo-DNA technology to specifically suppress mHTT expression as therapeutic approach
Currently, the most promising strategy for an effective treatment for Huntington disease is to specifically target the cause of the disease: the mutant gene. Research in HD animal models has demonstrated that this can be achieved with gene silencing methods such ASO. One of the major challenges is getting the silencing molecules into the brain. Here we will test a new class of ASOs, tricyclo-DNA-ASOs, which are reported to have superior properties such as an unprecedented uptake in many tissues. We will evaluate three novel tc-DNA-ASOs in a rat model for HD and compare their efficiencies in lowering mutant HTT.
Author: Hoa Nguyen
Approved: September 2017
Sphingosine-1-Phosphate metabolism/axis: toward new therapeutic perspectives for Huntington disease
Huntington disease (HD) is the most common dominantly inherited neurodegenerative disorder with a complex pathogenetic profile and no cure available, yet.
Over the past few years, metabolic changes of sphingolipids, major components of brain cell membranes, have been frequently associated with HD. Evidence suggests that aberrant sphingolipid metabolism may play a prominent role in the pathogenesis of the disease.
The main objective of this research proposal is to collect evidence that modulation of sphingolipid metabolism may be beneficial in HD and eventually represent a concrete drug target for the development of novel and effective therapeutic interventions for the disease.
Author: Alba Di Pardo
Approved: April 2017
Sterols /oxysterols lipidome of neurons and astrocytes after restoration of CYP46A1, the cholesterol degradation enzyme, in the striatum of R6/2mice
No efficacy to slow the evolution was demonstrated in clinical trials in HD. Studies from human and mice indicate that altered sterol synthesis is implicated in neurodegenerative processes and could offer new therapeutic strategies. We recently showed that restoration of cholesterol 24-hydroxylase (CYP46A1) is neuroprotective in R6/2 mice. In the brain, cholesterol synthesis relies mostly on astrocytes, whereas neurons eliminate cholesterol oxidized into 24S-hydroxycholesterol (24SOHC) by CYP46A1. We propose to investigate
how neuronal restoration of CYP46A1 in HD regulates the cholesterol and lipid metabolism in these two cell populations using FACS followed by mass spectrometry analysis.
Author: Sandrine Betuing
Approved: January 2017
Defining the assembly, pathogenicity and modes of regulation of small fibrillar aggregation intermediates in Huntington’s disease pathology by optical nanoscopy
A pathological hallmark of Huntington’s disease (HD) is the accumulation of misfolded, polyglutamine-expanded Huntingtin (Htt) proteins into large aggregates. Yet, rather than aggregates themselves, intermediate Htt species formed ‘en route’ or ‘in parallel’ to aggregate formation are proposed to provoke neurotoxicity. In this project, the presence, pathogenic nature, and cellular fate of intermediate Htt species at different stages of HD pathology will be studied in detail using super-resolution nanoscopy. Additionally, their potential modulation by quality control activities of molecular chaperones and protein clearance machineries will be investigated. The obtained knowledge may advance targeted therapeutic design to ultimately delay HD onset and progression.
Authors: Willianne I.M. Vonk, Steffen J. Sahl
Approved: May 2016
Ghrelin, potential treatment of Huntington’s disease pathology
In addition to classical neurological symptoms, Huntington’s disease (HD) is complicated by peripheral pathology, including weight loss, altered metabolism and muscle atrophy. In this project we will evaluate a treatment strategy using ghrelin administration in a HD mouse model. Ghrelin, is a peptide hormone that has multi-tissue effects. Ghrelin has for example been shown to improve muscle wasting and prevent weight loss. In addition, ghrelin analogues (ie ghrelin similar compounds), have been shown to have beneficial effects on cognition. We believe that by targeting multiple tissues, we will both get increased understanding of the complex HD pathology as well as possibly find new treatment strategies.
Author: Maria Björkqvist
Approved: September 2015
Functional Studies on Glutamate Signaling, in a Drosophila model for HD, using analogs of Glutamate or Glutamine to Ameliorate the Pathology of the Disease
Glutamate that is responsible for neuronal communication is also the culprit for their death. Neuronal death in Huntington’s disease results in involuntary movement, dementia, until death. We are using the fruit fly or Drosophila melanogaster as a model of Huntington’s disease. We are trying to identify small molecules with similar structure as the glutamate that allow the neurons to communicate but avoid their death. The new drugs will be administrated in the diet and their effect analyzed on animal climbing. Because of the short generation time of flies we hope to quickly address the efficiency of these new molecules on neuronal death and to extend our studies to other neuronal degeneration diseases.
Author: Paola Bellosta
Approved: May 2015
Repurposing an enzyme inhibitor for dual therapeutic benefits in Huntington’s disease
Huntington’s disease (HD) stems from mutation in the HD gene. These genetic abnormalities cause the disease, including motor defects and cognitive and memory decline. Surprisingly, one enzyme in the HD brain seems to be involved both at the DNA level and in the disease progression. This double activity offers the chance to inhibit the enzyme for dual therapeutic benefits. This project will repurpose an existing inhibitor of the enzyme and evaluate its effects in preclinical models for protection of the HD gene and in rescuing long-term memory defects and cognitive decline.
Author: Robert Lahue
Approved: July 2014
Analysis and recovery of neuronal microcircuit activity in Huntington’s disease using two-photon microscopy and optogenetics
Intracellular Ca2+ homeostasis is altered in HD and is involved in the degeneration of striatal neurons. Prior to degeneration, however, the functionality of the neuronal network could already be impaired. We will study such network alterations in a HD mouse model as this could give insights into disease mechanisms and may provide a therapeutic target for potential corrective measures aimed at restoring normal network functionality. We will first analyze spontaneous cortical activity and then we will selectively stimulate/inhibit regions of interest from local neuronal populations to single cells or even subcellular structures to correct the altered network activity.
Authors: Axel Methner, Albrecht Stroh
Approved: January 2014
A comparative MRI study to determine the affect of extensive cognitive training on disease progression in HD mice
There is evidence from HD mouse studies that environmental enrichment can slow disease progression. In the present study we were interested to see whether long term cognitive training, so called “brain training” could slow the onset of, or reduce the severity of the disease in the YAC128 HD mouse line. Mice will be tested on a number of tests of motor and cognitive function and will then be scanned using magnetic resonance imaging (MRI) to determine whether “brain training” improves performance and whether the physiological changes underlying any performance benefit, can be identified with the MRI. This study could provide evidence as to the important brain structures responsible for the beneficial effects of environmental enrichment.
Author: Simon Brooks
Approved: May 2012
A translational study on the beneficial effects of exercise on Huntington’s Disease symptomatology: The development of a mouse correlate of an ongoing patient trial.
There is a considerable body of evidence from studies on Parkinson’s and Alzheimer’s disease, that exercise can reduce symptom severity in patients. As a consequence, a study was designed to determine whether this was true for Huntington’s disease. Based on this human study, we have designed a comparable study in mouse models of the disease to see whether a model system of exercise intervention could be developed. A model system for mouse models of the disease would permit us to optimise human exercise programmes and maximise the potential of this intervention strategy.
Author: Simon Brooks
Approved: November 2010
Clinical Research Projects:
Modifiers
Generating unified HD progression measures in Registry participants
Although Huntington Disease is caused by a single gene, other genes can influence the disease’s speed, severity, and perhaps the mixture of features. Most studies to identify these genes have relied on variation in the age of initial clinical illness. In contrast, my collaborators and I previously used the Track-HD data and a subset of Enroll to discover an important secondary gene by testing relationships with a combination of HD measures that change with time. Our sample size was relatively small. We will now use a larger portion of Registry data to refine these measures and search for additional modifier genes.
Author: Douglas Langbehn
Approved: December 2018
Anti-inflammatory drugs, and synergistic interactions with anti-diabetic drugs, and the progression of Huntington’s disease
Inflammation is associated with many neurodegenerative disorders, including Huntington’s disease. In this project, we aim, through a longitudinal observational study of the Registry and Enroll-HD databases, to explore if the intake of nonsteroidal anti-inflammatory substances would modify the progression of the disease in HD patients. Many HD patients, aside of suffering this neurodegenerative condition have other chronical diseases such as arthrosis, cardiac disease or diabetes. For some of these conditions they are prescribed drugs chronically, and we do believe that these drugs may affect the progression of HD. This information may have interest for future clinical trials.
Author: Rafael Vazquez Manrique
Approved: April 2017
Does the precise CAG repeat sequence of intermediate and reduced penetrance alleles of HTT influence likelihood of expansion into the pathogenic range?
Although many HD patients have a family history of the disease, there is a low rate of de ‚novo’ mutation, which are thought to arise from intergenerational expansion of the CAG repeat in HTT from the intermediate allele range (CAG 27-35 repeats) into the disease allele range (CAG ≥36 repeats). A recent study has shown that approximately 6% of the general population carries an intermediate allele and so it would appear that only a small proportion of all intermediate alleles expand on transmission into the disease range. We will study individuals with ‚no family history’ identifed through REGISTRY and families with intermediate/reduced penetrance alleles using SNP haplotype analysis and next generation sequencing technologies to study the sequence and architecture of the CAG gene to determine their influence on CAG repeat stability. Understanding these mechanisms better will improve the accuracy of genetic counselling in patients with intermediate/reduced penetrance alleles, as well as advancing our knowledge of HD pathogenesis.
Authors: Darren Monckton, Nayana Lahiri
Approved: January 2017
Perinatal asphyxia as a modulator of Huntington’s disease onset and progression
There is a large variation in the age-of-onset of Huntington’s disease (HD), even among persons with the same CAG-repeat length. There are other genetic and environmental factors which play a role in the disease. Oxygen-shortage during birth, called perinatal asphyxia, affects 8.5 babies per 1000 live births world-wide. These babies often have learning and communication difficulties like autism and attention-deficit-hyperactivity disorder and movement problems like cerebral palsy. Our project will investigate whether birth complications, like perinatal asphyxia, decreases the age-of-onset of HD or affects the type of symptoms at disease-onset, thus whether birth complications are an environmental modifier of HD.
Author: Melinda Barkhuizen
Approved: October 2016
Analysis of expression levels of Htt-interacting proteins in human lymphoblasts from HD patients
The project will address the question, whether amounts of certain proteins binding to Htt in human cells are influenced by the presence of a polyQ expansion in Htt. Amounts of known Htt-interacting proteins will be determined in lymphoblasts from HD patients and from normal controls, as will be their turnover and the half-life of the corresponding mRNAs. We hypothesize that levels of some of the Htt-interacting proteins might be influenced by the presence or absence of the HD mutation.
Author: Stefan Kochanek
Approved: September 2016
Identification of coding variants in DNA repair genes affecting age at onset of Huntington’s Disease
The gene mutation causing HD influences the age at which disease symptoms start, and the speed at which they progress. Recent genetic studies have uncovered a possible role for DNA repair pathways in modifying disease onset. This project will stratify the Registry database by actual age at disease onset against predicted age at onset, and then look specifically (by exome sequencing) for DNA changes that alter the protein sequence in DNA repair enzymes. Identification of these changes will help us understand the factors that affect when HD starts.
Author: Tom Massey
Approved: May 2016
Analysis of select Lymphocytic cell lines with rare or alternate alleles that are associated with GWA signal for age at motor onset modification
GWA study of 3500 HD individuals revealed three significant chromosome loci that are modifiers of mAOO. By computing the halotype and SNP structure of individuals bearing these SNPs at each of the three loci, we identified a group of individuals who represent a strategically overlapping genotype structure around each of these loci. We will use the LCL’s from these individuals to capture and sequence the length of the chromosome loci as bounded by linkage disequilibrium, to compare and identify sequence level variants that likely confer the modifier effect in these individuals. This is one of critical steps to drill down from associated chromosome loci to causative modifier gene/genes- and ultimately advise on therapeutic strategies.
Author: Seung Kwak
Approved: March 2016
Do hormonal birth control pills and/or does pregnancy influence the onset and/or severity in HD?
To be able to answer frequently asked questions about the influence of hormonal anticonceptives and/or pregnancy on the course of HD, we want to check data collected within „Registry” and enroll-HD to look for differences among women with equal CAG repeats, but different patterns of either anticonception (hormonal-mechanical etc.) and women who were pregnant and gave birth.
The aim of our data-mining project is to determine if there are any significant differences between these groups, which will mean that hormonal changes might influence the course of HD.
Author: Janina Kieni
Approved: December 2015
Comparative analysis of mitochondrial genomes in HD
Mitochondria are critical for cellular energy production and many other vital biological functions. Mitochondrion is the only organelle with its own genome in animals. Perturbations in mitochondrial function have been routinely observed in HD patients. We propose to investigate whether mtDNA variations and mutations can contribute to onset and progression of HD. By sequencing at high depth the mitochondrial genomes from HD patients and healthy controls we will be able to discern mutations, variation, and the proportion of pathogenic mutations in HD patients. We will be able to determine whether mtDNA variations/mutations play a role in HD progression.
Author: Zhenglong Gu
Approved: July 2015
Evaluating possible effects of HD on epigenetic acceleration of DNA aging in peripheral blood cells
Our coapplicant S. Horvath (UCLA) recently developed a highly accurate biological clock based on DNA methylation levels. The compound signature using multiple methyl-DNA marks in post mortem HD brains have provided insight on how cells bearing HD mutation manifest DNA marks reminiscent of premature DNA aging (See project “Correlation of DNA sequence and epigenetic status…”). Here we sought to use a set of whole blood DNA, employing to ascertain epigenetic signature in blood of HD for evidence of methy-DNA age accelaration
Author: Seung Kwak
Approved: November 2014
Correlation of DNA sequence and epigenetic status between blood and lymphocytic cell line
Horvath et al recently developed a highly accurate biological clock based on DNA methylation levels. This epigenetic clock turns out to be substantially more accurate than existing biological clocks including telomere length and p16INK4A levels. They applied epigenetic clock to a number of human brain samples from HD subjects and suitable controls (unpublished observations). Strikingly, they see a highly significant age acceleration (respective to control brains of similar age) associated with HD in several brain regions including cerebellum, cingulate gyrus, and possibly motor cortex and parietal cortex. Here we plan to determine 1) whether similar epigenetic accelerated aging effects can also be observed in HD blood DNA, and 2) if aspects of epigenetic aging acceleration can be modeled in patient derived cell lines for in vitro experimentation
Author: Seung Kwak
Approved: November 2014
SNP typing of 96 highest value loci of Registry samples identified from the GWA Study effort
The variance in the neurological age of onset in HD cannot be explained by CAG repeat size alone. We have previously performed a GWA study and successfully identified significant SNPs that modify neurological age of on-set. Many other sites were nominally associated but not significant. We intend to survey 5000 additional samples at 96 top loci as means to cross validate, and new modifier genes arising from this effort should give rise to high value drug targets.
Author: Seung Kwak
Approved: July 2014
Identification of epigenetic signatures in HD striatum
Transcriptional dysregulation is an early process that is central to HD pathogenesis. Compounds aimed to improve gene expression through chromatin structure modulation are considered as promising. HDAC inhibitors are such compounds. However they lack selectivity and are relatively toxic. The goal of the project is to better characterize HD epigenome using genome-wide and high-resolution technique (e.g. ChIP-seq) and identify epigenetic signatures that may be useful to define new targets for epigenetic therapies. Our data obtained in HD R6/1 mouse striatum reveal a specific epigenetic signature associating with disease. We will determine whether this signature is conserved in HD brain tissues.
Author: Karine Merienne
Approved: July 2014
Analysis of genes participating in the dopamine and serotonin neurotransmitter system as potential modifier genes for Huntington disease pathogenesis
The pathogenesis of Huntington disease is provoked by the CAG repeat expansion in the HTT gene, but seems to be determined by a wide spectrum of pathways. To date, different studies showed an involvement of proteins in the dopamine and serotonin system in the pathological process of HD. Alterations in the dopamine and serotonin metabolism have been detected in HD animal models as well as in HD patients. In the present study, we intend to test various components of the dopamine and serotonin system (MAO-A, SLC6A3, SLC6A4, DRD2, DRD3, DRD4, ANKK1, HTR2A) as putative HD genetic modifiers. The identification of phenotype-associated polymorphisms may improve prognosis prediction and evaluation of medical intervention during patient assessment.
Author: Silke Metzger
Approved: July 2014
Can genetic imprinting be a factor in the pathogenesis of Huntington’s disease?
This study aims at verifying if genetic imprinting may modify the age at onset (AO) in Huntington’s disease (HD). The proportion of variation in the AO explained by the sex of parent affected with HD in a large population of patients, with the dominant impact of (CAG)n repeats included in the analyses, will be determined. Simple linear regression analyses and nested multiple models will be performed. The AO will be treated as the outcome measure while the number of CAG repeats, parental sex and parental age at conception will be included in the analyses as the predictor variables.
Author: Anna Stanisławska-Sachadyn
Approved: July 2014
Investigating the significance of novel huntingtin splice variants in Huntington’s disease patients
Screening the HTT gene in HD and control brain revealed many novel splice variants. A pool of HTT RNA species therefore exists with the potential to encode novel HTT protein isoforms. This study aims to determine whether these different protein isoforms exist and whether they associate with HD pathogenesis. The normal and pathogenic roles of HTT are not fully understood. Characterisation of any novel HTT protein isoform could contribute to a better understanding of the function of HTT in the cell. Furthermore, a comprehensive knowledge of alternative HTT RNA species could have implications for the design of RNA silencing therapies in HD.
Author: Alis Hughes
Approved: May 2014
CAG repeat length polymorphisms as modifiers of clinical phenotype: Huntington’s disease and the general population
Recently it was shown that subjects with intermediate HTT alleles experience significantly more depressive symptoms than controls. As in the general population the frequency of intermediate alleles can be as high as 6%, these findings could have important implications for both Huntington’s disease patients and the community at large. Therefore, we aim to systematically evaluate 1) the role of HTT CAG repeat polymorphisms in the normal range as modifiers of mental health, cognition, metabolism and aging in the general population, and 2) the effects of CAG repeat polymorphisms in other polyglutamine disease-associated genes on the clinical features of Huntington’s disease patients.
Author: Ahmad Aziz
Approved: January 2014
The role of SMAD-dependent TGFβ signalling in transcriptional dysregulation in early stage models of HD
SMAD proteins are major regulators of gene expression that act within the transforming growth factor-beta (TGFβ) signalling pathway. Preliminary data has revealed potential differences in this pathway in a cell model of HD. This project aims to characterise the role of SMAD proteins in mouse and human cell models of HD by comparing their expression, localisation and activity, as well as by manipulating the TGFβ pathway in order to determine whether it may contribute to the altered gene expression that is observed in HD.
Author: Kathryn Bowles
Approved: January 2014
Analysis of the Registry Clinical Characteristics Questionnaire data
The Clinical Characteristics Questionnaire (CCQ) records age of onset in a range of HD symptoms. To inform our use of CCQ information in our ongoing genome-wide association study we wish to obtain data from all subjects in the database who have a filled out CCQ. The two main aims of asking for data on all subjects that are available are to:
• Model the relationship of each symptom to CAG repeat length;
• Explore whether symptoms recorded are an inevitable part of the disease progression in all or most HD subjects, or whether some symptoms can be regarded as dichotomous.
Author: Lesley Jones
Approved: May 2013
Investigation of polymorphisms affecting the dopaminergic system on age of onset and disease progression in human Huntington’s disease
The abnormal gene in HD contains a large repeat that codes for an abnormal form of the huntingtin protein and the size of this repeat governs to some extent when people get the disease. However, there are other influences on this and we are investigating whether changes in a specific transmitter system in the brain could be involved. In order to do this we are testing small variations in the genes coding for dopamine receptors and analysing whether it affects when people get HD. If this does prove to be the case then it may open up new treatment strategies.
Author: Roger Barker
Approved: May 2013
An investigation into gender influence on anxiety and depression in Huntington’s Disease (HD) across disease-stage
The proposed study aims to examine gender differences in depression and anxiety in HD. A consistent finding among the general population is that women are more likely to experience anxiety and depression than men. While there is some evidence of a higher frequency of depression in females with HD when compared to men with HD, the results are inconsistent. Moreover, anxiety remains a relatively neglected area in HD research and little is known about any impact of gender on anxiety symptoms. This study proposes to use REGISTRY data from a UK sample, with the Hospital Anxiety and Depression scale (HADs) as a measure of symp-tomology. Due to the variable course of anxiety and depression in HD, an analysis of gender by disease-stage will be conducted.
Author: Maria Dale
Approved: April 2013
The use of propensity scores to draw inferences about how lifestyle factors might be associated with HD progression and clinical severity
The overarching aim of this study is to develop statistical methods for the analysis of large scale longitudinal data sets for the ENROLL database. The study will use currently available data from the European Huntington Disease Network Registry („Registry”) database to evaluate whether use of causal modeling techniques (namely propensity score weights) might prove helpful for increasing understanding of the potential environmental modifiers of HD. Specific moderating variables of interest include: education, socioeconomic status, smoking, alcohol, drug use, statin use, aspirin use, nutritional supplements, and antidepressant drug use.
Author: Beth Ann Griffin
Approved: March 2013
Replication study of non-genetic factors that can influence the age at onset (AO)
It is estimated that 60% of the age at onset of Huntington’s disease variability is due to environmental factors; however, few studies have analysed the influence of that factors. Previously, we have detected some association between two different environmental factors and age at onset, but in a small HD patient’s sample. Therefore, the aim of this work is to carry out a replication study in a larger sample in order to confirm or reject the previously obtained results. The relationship between premorbid factors (such as Diabetes mellitus, arterial hypertension, allergies, etc.), life style habits (such as tobacco smoking, alcohol abuse, drug consume…) and age at onset will be studied.
Author: Leire Valcarcel Ocete
Approved: January 2013
Does the presence of the HD mutation affect the development of the human striatum?
The aim of this project is to set-up a network across six University Hospital sites in the UK for the collection of fetal tissue carrying the HD mutation. Consent will be obtained by a specially trained research nurse and tissue will be collected following both surgical and medical termination of pregnancy. Tissue will be returned to the School of Biosciences at Cardiff University where it will be used to collect preliminary data on the HD mutation, including gene expression profiles, CNS anatomy, and cellular characteristics.
Author: Anne Rosser
Approved: September 2012
Heritability of age at onset and progression in Huntington’s disease: heritability of symptom onset and progression and identification of the most informative subjects for revealing genetic modifiers of Huntington’s disease
In HD both onset and progression are variable: the variability in HD onset can be attributed to HTT CAG length but also other genetic differences between subjects. The influence of CAG length on disease progression unclear and its heritability poorly understood. To inform the search for modifiers of disease onset and progression we wish to establish the correlation of onset ages of multiple symptoms from the Clinical Characteristics Questionnaire, establish the heritability of progression and onset amongst relat-ed subjects and examine Registry for discordant subject relatives in which to search for genetic modifiers of onset and progression of HD.
Author: Lesley Jones
Approved: May 2012
The role of phosphorylation pathways in the oligomerization and toxicity of mutant huntingtin
Aggregation of the huntingtin protein is a central event in Huntington’s disease. While mutations in huntingtin affect its aggregation, it is becoming evident that post-translational modifications, such as phosphorylation, may also influence the aggregation propensity and toxicity of the protein. In order to understand how phosphorylation modulates huntingtin aggregation and toxicity we will search for genetic and pharmacological modifiers of the initial steps of aggregation and toxicity using an assay we recently developed, known as bimolecular fluorescence complementation. Ultimately, our findings may enable the development of novel strategies for therapeutic intervention by modulating huntingtin phosphorylation.
Author: Tiago Fleming Outeiro
Approved: December 2011
Analysis of potential modifier genes involved in intracellular trafficking and mitochondrial function
The expanded CAG repeat in the HTT gene causes Huntington disease (HD), but other genetic factors additionally influence the course of the disease. We identified HAP1 as one of these genes modifying the age-at-onset. So, we aim at verifying this effect of HAP1 in an additionally patient cohort and examining the association between HAP1 and the body mass index of these patients. Additionally, we intend to search for further modifier genes participating in intracellular transport and mitochondrial function. As both aspects seem to be closely related to the pathogenesis of HD, modifying effects of these genes may influence the disease.
Author: Silke Metzger
Approved: January 2011
Are there specific lifestyle factors that could be associated with the HD clinical characteristics and progression thereof: a retrospective data mining study?
This data mining study aims to use currently available data from Registry to evaluate whether there are any specific environmental factors that could be associated with progression of symptoms in the first instance and ultimately with onset of symptoms. This will not prove causal links but may identify a starting point to aid further understanding of environmental modifiers. We will investigate any associations between a) education and occupation, b) concomitant disorders c) smoking, d) alcohol e) drug use and HD clinical characteristics using multiple regression analysis. Classification and regression tree (CART) analysis will also be utilised.
Author: Monica Busse
Approved: December 2010
Identification of genetic modifiers of age of onset in HD. Candidate gene study of CR1, CLU, PICAM, PRNP and APOE
We aim to identify potential genetic modifiers of HD age at onset. There are many parallels between different neurodegenerative diseases; the processes which occur in Alzheimers disease and HD may, for example, involve similar pathways. For that reason we will search for polymorphisms in several genes/proteins of HD patients that are known to play a role in the pathogenesis of Alzheimers Disease. We will investigate 1 SNP (punctual variabilities in the DNA) in each of the following genes: CLU, CR1, PICALM and PRNP and 2 SNPs in APOE.
Author: Sarah Tabrizi
Approved: December 2009
Disease Progression Modeling in HD
This is a collaborative effort between CHDI, FDA and the HD research community to develop a disease progression model, using REGISTRY data and data from other observational studies (COHORT, PREDICT, TRACK and ENROLL) and selected clinical trials. The model will be used in the planning of future clinical trials. We model the age at first appearance and rate of progression of various signs and symptoms of HD, search for genetic, environmental, and life-style related variables that explain variability in disease progression and attempt to characterize the diverse patterns of disease progression that appear in the study population.
Author: John Warner
Approved: November 2009
An assessment of CAG repeat length and age at onset on rate of disease progression in Huntington´s disease
While CAG repeat length influences onset age, studies have been insufficiently powered to evaluate the effect on rate of disease progression. We propose to study individuals followed longitudinally for at least three years, and with at least three neurological examinations, to evaluate whether the CAG repeat length and age at onset are independent predictors of rate of progression or if one or the other singly accounts for association to rate of disease progression. These studies may be relevant to clinical trials, to control for the effects of factors that influence rate of disease progression.
Author: Richard Myers
Approved: April 2009
Tau haplotype and its relationship to age of onset and disease progression in Huntington’s disease (HD)
Factors influencing the age of onset of HD are largely unknown outside of CAG repeat length. What affects speed of disease progression is also unclear. This project will investigate whether tau haplotype has an influence on either of these aspects of HD, as we have shown that the H1 tau haplotype is associated with the early onset of dementia in Parkinsons disease. We will compare tau haplotype against CAG repeat length, age of disease onset and evidence of disease progression over a two year period using Total Functional Capacity measures.
Author: Roger Barker
Approved: April 2009
Copy number variation of potential genetic modifiers of Huntington’s Disease
Genetic modifiers of Huntington’s disease (HD) influence when and how HD manifests and how HD progresses. We have identified several potential modifier genes that show copy number variation (CNV). Various gene mutations can lead to CNV from the standard gene dose of two copies (one from each parent). CNV of candidate genes influences resistance or susceptibility e.g. for psoriasis and HIV-1 infection. This project will investigate the association of CNV of potential genetic modifiers of HD with age-of-onset and progression. If CNV in a given candidate modifier gene were associated with e.g. a delay of age-of-onset or progression, this may help elucidate further the pathogenesis of HD, as well as offer new therapeutic targets for HD.
Author: Flaviano Giorgini
Approved: February 2009
Unbiased prioritarization of neuroprotective targets for HD
A project developed by the Biological Modifiers Working Group at EHDN
The aim of this project is to develop data integration and network analysis to prioritize neuroprotective targets in HD. Huntington’s disease is extensively studied thanks to models that were developed in several species and that recapitulate complementary components of HD pathogenesis. Genome-wide analyses in these models have generated a large amount of data with high potential for target selection. The comprehensive and unbiased integration of ‘omics data’ on HD will allow better decisions in candidate target selection to be reached. Selecting for gene networks, modules and strings that are consistently highlighted by different model systems as ‘targets’ and/or ‘modifiers’ of mutant huntingtin activity may provide a strong basis to develop neuroprotective strategies that may be efficient against several stages and/or key features of HD. This concerted action involves the laboratories of Juan Botas, Bob Hughes, Lesley Jones, Ruth Luthi-Carter, Christian Neri and Shen Zhang.
Author: Christian Neri
Approved: January 2009
Search for candidate genes implicated in Huntington’s disease (HD) age of onset (AOO)
The aim of this work is the identification of Huntington´s disease AOO genetic modifiers, different from the CAG repeat length in the HD gene. For this purpose we will carry out an association analysis with 115 SNPs from 20 candidate genes in 250 samples collected by the REGISTRY project. The chosen candidate genes are located in several chromosomes and have been selected for being genes which encode proteins that interact directly with huntingtin or because they take part in the pathogenesis of the disease. The selected SNPs are located in coding, intron and 5´ and 3´ regions of the selected genes.
Author: ANA AGUIRRE ESCOBAL
Approved: December 2008
Genome-Wide Genotyping of EHDN Samples
The variance of the age at onset (AAO) of first signs of HD cannot be explained by CAG repeat size alone. A substantial proportion of the residual variability is highly heritable indicating that other genes also influence AAO. In order to identify such genetic variants we will undertake genome-wide genotyping in REGISTRY participants where the appropriate clinical information is available. This will employ powerful analysis tools with the aim of genotyping millions of gene loci in thousands of REGISTRY participants. The identification of genetic variants that modify AAO of HD may open new avenues for developing novel therapeutics.
Author: Seung Kwak
Approved: October 2008
Influence of gender on progression of HD
Several factors other than the CAG repeat length very likely influence when Huntington’s disease manifests and how it progresses. One such factor may be gender. We will assess in a cross-sectional CAG repeat matched sample whether there is any difference between men and women in the rater estimates of age at onset. In the second part of the study we will look at longitudinal data (at least 3 REGISTRY visits) to test for any gender related differences in the progression of HD signs (motor, behaviour, TFC, cognition). Any difference between the sexes may point to gen-der specific modifying factors.
Author: Daniel Zielonka
Approved: October 2008
PGC-1a as a potential modifier gene of age of onset of HD
The Huntington’s disease (HD) gene determines to some extent when HD signs appear. However, other genes, so called modifier genes, also influence the age of onset. Recent studies implicate PGC-1a, an important regulator of the cell’s main energy generator, the mitochondrion, as a potential modifier gene. We have already found an association of PGC-1a gene variants with age of onset of motor symptoms in an Italian HD population. These findings we intend to replicate in a larger sample of data collected in REGISTRY. If confirmed, it may be worthwhile to explore the potential of PGC-1a as a therapeutic target.
Author: Patrick Weydt
Approved: August 2008
Search for genetic modifiers in HD: exploring the Foxo network
In Huntington’s disease (HD) the CAG repeat length explains much, but not all, of the variance in the age at onset (AO). Other as yet unknown genes also contribute. In mouse and worm models we identified potential modifiers of mutant huntingtin cytotoxicity in the Foxo network. FOXO transcription factors are important regulators of cell survival in response to a variety of stress stimuli such as oxidative stress, DNA damage, and nutrient deprivation. To look at the relevance of our findings in HD model systems for HD patients using REGISTRY data we will investigate the relationship of gene alterations within several FOXO members with the age at onset of HD signs, and progression of HD.
Author: Christian Neri
Approved: November 2007
Hunt for Huntington modifiers with systems biology approaches
Genes other than the Huntington’s disease (HD) gene influence when HD signs appear. These genes are called modifier genes. We previously identified potential modifier genes, and mutations within these genes. In this project, we intend to associate the mutations with the variation in age at onset. To this end we will study genotypes of 1000 HD patients and correlate genetic findings with clinical characteristics. Once the potential modifier genes and its mutations have been determined, they could serve as important targets for therapy development to delay the onset of HD.
Author: Erich Wanker
Approved: November 2007
Sex-specific differences in AO
The CAG repeat length accounts for about half of the variance in the age of onset (AO) in Huntington’s disease (HD). Genes other than the HD gene (Modifier genes) may account for an additional 40% of the variation. N-Methyl-d-aspartate (NMDA) receptor-mediated excitotoxicity has been proposed to play a role in the pathogenesis of HD. We previously found substantial differences of the C2664T SNP in GRIN2B (glutamate receptor; ionotropic NMDA) between men and women, in particular in premenopausal patients. If confirmed in a larger sample of data collected in REGISTRY hormonal factors might prove a possible therapeutic target.
Author: Carsten Saft
Approved: November 2007
Genome Wide Association Scan to Identify Modifiers of HD Onset
The length of the HD CAG trinucleotide repeat plays the major role in determining age at neurologic onset but does not explain all of the variation observed. In an extensive collaborative effort, we are searching the entire human genome for genetic variants that modify age at neurologic onset in HD to identify genes capable of altering HD pathogenesis. A first phase of 1200 HD samples has been genotyped using a high density microarray (Affymetrix GeneChip v6.0) and a second phase of genotyping of 1600-2,000 independent HD samples, is gearing up, to confirm or refute results from phase 1 and to add additional power to detect genetic modifiers that could provide therapeutic targets.
Author: James Gusella
Approved: November 2007
Impact of education on age at onset and progression of HD
Genetic and environmental factors other than the Huntington’s disease (HD) gene influence when HD signs appear and how HD progresses. Education protects from Alzheimer´s disease, and development in an enriched environment slows the disease in mouse models of HD. We aim to investigate the impact of education on 1) the age at onset and 2) the progression of HD. For the first aim we use a cross-sectional sample and for the second longitudinal data from patients with 3 or more REGISTRY visits. If education modified HD onset or progression paying attention to education could improve the prognosis.
Author: Justo García de Yébenes
Approved: October 2007
Biomarkers
Understanding how conformation-specific antibodies target misfolded huntingtin states
The huntingtin protein that is mutated in Huntington disease (HD) forms multiple types of misfolded protein deposits in symptomatic and pre-symptomatic patients, in model animals, and in vitro. In clinical trials focused on lowering the amount of mutant protein, the mutant protein deposits represent a crucial biomarker given its association with disease onset and progression. In our project we aim to develop a molecular understanding of how particular HTT-specific antibodies bind and recognize distinct types of protein deposits, to support their use as ongoing and future use as diagnostic tools in preclinical research and beyond.
Author: Patrick van der Wel
Approved: May 2021
Untargeted proteomics analysis and proteomic profiling in Cypriot Huntington’s disease patients
Protein-protein interactions can be studied in order to understand how mutant huntingtin (mHTT) and/or wild-type HTT (wtHTT) proteins interact with other proteins and how abnormal protein expression changes in different HD disease stages. The study will utilize discovery proteomics using a Liquid chromatography-mass spectrometry (LC-MS) based approach on serum samples obtained from asymptomatic, symptomatic and advanced HD patients versus gender/age matched controls. The proteomics data will be analysed using bioinformatics tools and databases, to describe and identify the biological pathways and underlying mechanisms involved in HD phenoconversion and progression.
Author: Christiana Christodoulou
Approved: March 2021
Huntington’s disease brain pathology signatures in exosomes: potential markers of disease progression
Exosomes, small vesicles secreted by most cell types, including brain cells, contain biological material specific to the state of the cells of origin. Thus, they have emerged as promising carriers of biomarkers in HD. However, investigation is needed to identify disease-specific signatures in brain exosomes. We will isolate exosomes from postmortem HD and control brain samples and measure changes in protein composition in order to identify HD-specific signatures that correlate with brain pathology. Given that exosomes can cross the blood-brain barrier and reach the bloodstream, they could be accessed and serve as a source of biomarkers for HD brain-related processes.
Author: Rocio Perez Gonzalez
Approved: January 2020
Pioneering the next generation of biofluid biomarkers for Huntington’s disease: the NEVADA-HD Study
We have made considerable progress in biofluid biomarkers for HD, but we still need better biomarkers to tell us what is happening inside the brain. Exosomes are tiny vesicles released by most cell types under normal and pathological conditions. They harbour proteins/RNA/lipids that reflect the functionality of the host cell. Exosomes can be isolated from biological fluids and their content can be measured as a fingerprint of the tissue of origin. This study aims to optimize the isolation of exosomes of neuronal origin, both from cerebrospinal fluid and blood of subjects with pre-manifest and manifest HD, comparing different techniques; and to evaluate them as a new source for biomarker discovery in HD.
Author: Edward Wild, Rosanna Tortelli
Approved: May 2019
The role of the ribosome in the pathogenesis of HD
Huntington´s disease (HD) is a proteinopathy, like most neurodegenerative diseases. In proteinopathy, the balance of protein synthesis, maintenance and degradation (protein homeostasis) is severely disturbed. In this project we would like to investigate if huntingtin (htt) is a transcription factor involved in ribosomal biogenesis by RNA polymerase I. We will analyse synthesis, processing, assembly and performance of the ribosomes of HD patients. Following a recent description by our group in an unrelated childhood progeria with neurodegeneration, we hypothesize that disturbances in protein synthesis at the ribosome might contribute to the loss of protein homeostasis in HD.
Author: Sebastian Iben
Approved: May 2019
CLEAR-HD: Cortical Layer Examination At high Resolution in Huntington’s Disease
With a number of therapies emerging that can directly target the Huntington’s disease (HD) gene mutation, there is now potential to slow or even cure the disease. Many of these treatments are either injected into the cerebrospinal fluid, where they achieve greatest uptake in the cortex, the outermost layer of the brain, or are delivered by injection into the basal ganglia, a deep brain structure. In the CLEAR-HD study we are using cutting edge brain imaging techniques to look at these structures at a level of detail that has never been possible before using a combination of ultra-high field 7T MRI and magnetocephalography.
Author: Peter McColgan
Approved: January 2019
Exploring potential cerebrospinal fluid biomarkers for reverse transsulfuration pathway impairment and redox imbalance in Huntington’s disease – relevance for cysteamine treatment?
Oxidative stress may play a role in neurodegeneration. The sulfur-containing amino acid cysteine is made from methionine via transsulfuration from cystathionine. Cysteine is important for the synthesis of the antioxidant glutathione. Due to their sulfhydryl group, cysteine and glutathione, so-called aminothiols, can neutralize pro-oxidants. In the HD brain, cysteine synthesis seems to be impaired. Cysteamine, an EMA-approved drug, can boost cysteine levels. A clinical trial indicated that cysteamine might be beneficial in HD.
In this project, we aim to explore whether aminothiols are changed in the HD cerebrospinal fluid indicating cysteine deficiency and/or oxidative stress. Demonstrating alterations in aminothiols in HD would substantiate the rationale for cysteamine treatment.
Author: Jan Lewerenz
Approved: July 2018
Association Between Iron Dysregulation, Neuroinflammation and Clinical Measures in Huntington’s Disease
In Huntington’s disease (HD), as in other neurodegenerative diseases such as Alzheimer’s disease (AD) and Parkinson’s disease (PD), damage to neurons appears to go hand in hand with inflammation in the affected brain region. Inflammation in the brain (neuroinflammation) activates specific immune cells called microglia. Another aspect common to neurodegenerative disease is accumulation of iron in brain areas affected by the disease. There is evidence that in HD, as well as in PD and AD, much of this excess iron is found in activated microglia. We wish to establish the role of iron accumulation in brain regions involved in HD and its link with neurodegeneration and neuroinflammation, as well as to examine the potential of using iron accumulation as a sensitive imaging marker for disease progression. We plan to achieve these goals by quantitatively measuring the distribution of iron in the brain using MRI at ultrahigh field (7 Tesla). This sensitive measure for iron in the brain will then be correlated with cerebrospinal fluid (CSF) markers for neuroinflammation and neurodegeneration on the one hand, and with clinical measures for disease progression and severity on the other.
Author: Itamar Ronen
Approved: May 2018
Leucocyte telomere length as a biomarker in Huntington’s disease
Peripheral blood biomarkers, predicting age of onset and disease progression, are crucial points both in genetic counselling and in trials for new drug delaying disease onset and progression. Telomeres are repetitive sequences at the ends of chromosomes with a role in genomic stability maintenance. Reduced leucocyte telomere length (LTL) is associated with neuroinflammation and neurodegeneration in aging and Alzheimer’s Disease. Our project aims at verifying whether LTL could be used as a biomarker for HD progression. The study of LTL shortening in a large sample of HD subjects, might provide useful data to be correlated with disease onset and progression.
Author: Liana Veneziano
Approved: January 2018
Mutant huntingtin associated changes in exosome content composition
Tissues in the body communicate with each other by releasing parcels into the blood stream. These parcels, called exosomes, contain various biological materials including proteins and mRNA. Exosomes can cross the blood-brain-barrier and thus examining them might help us learn what is going on within the brain including in Huntington’s disease. This could be important in the context of clinical interventions where we will need a good measure of their efficacy, in particular within the brain. We propose that exosomes can be used to this end in particular as measures of the molecular effects of mtHTT.
Author: Andreas Neueder
Approved: January 2017
The association of nucleolar activity with mutant huntingtin protein species in HD
Transcriptional dysregulation is known in HD. Here we will test the hypothesis that changes in ribosomal DNA (rDNA) transcription in the nucleolus are early signs of transcriptional dysregulation by mutant huntingtin (mHTT). By a systematic analysis of nucleolar transcription in HD models and tissue biopsies at different stages, we aim at identifying a sensitive metabolic marker linked to mHTT. The results could help to test the efficacy of ongoing therapeutic strategies aiming at lowering mHTT levels in specific cells. Concomitantly, the tissue-specific impact of mHTT on rRNA transcriptional dysregulation might account for the still unexplained variability in HD onset.
Author: Rosanna Parlato
Approved: September 2015
The role of microRNA-34 in cancer defense- and HD pathogenic mechanisms
The frequency of cancer in HD patients is only half of what is expected from population studies, and this applies to all forms of cancer. This is also true for other polyQ disease patients, which indicates that CAG repeat expansions have protective effects against the initial steps in tumor formation. The microRNA, miR-34, is well-known as a tumor suppressor that protects against uncontrolled cell growth, and it may be involved, in some situations, in cell death. We investigate the role of miR-34 in cancer defense and neurodegeneration by studying HD patients, transgenic HD mice and a cell model.
Author: Lis Hasholt
Approved: January 2014
The Alterations in Transcranial Sonography of Huntington Disease Patients with Psychiatric Symptoms
Transcranial sonography (TCS) is a noninvasive method that provides an insight into the pathology of movement and affective disorders. TCS findings could play a role in differential diagnostics and serve as markers of disease progression. The aim of the project is to prospectively screen a population of HD patients in search of abnormal TCD findings (altered echogenicity of nucleus raphe, substantia nigra, caudate nucleus) and to correlate them with symptoms like depression and apathy. The main goal is to check, if TCS findings may serve as a biomarker and a predictor of depression in HD patients, also those treated with dopamine depleting agents.
Author: Katarzyna Jachińska
Approved: September 2013
Apathy, cognition and oculomotor impairment in HD
Individuals with Huntington’s disease (HD) often suffer from apathy, a condition involving reduced motivation and initiation. Apathy has negative consequences for both patient and family, and has been associated with cognitive (thinking) changes in HD. In healthy adults and individuals with other conditions, apathy has been associated with abnormal eye movements while processing information. HD patients and gene carriers are known to show abnormal eye movements, but little is currently known about how changes in eye movement relate to changes in cognition and behaviour. This project will explore relationships between data on eye movements, apathy and cognitive performance in HD.
Author: Judith Bek
Approved: January 2013
Mitochondrial dysfunction in Huntington’s disease
Mitochondria are the subcellular particles that are critical for the use of oxygen and glucose. A better understanding of their role in Huntington’s disease could be important for the discovery of new therapeutic approaches. Hence we will investigate mitochondria in blood cells and autopsied brains from patients, as well as in brains from mice that bear the gene mutation that causes HD in humans. Our plan is to measure activity, protein and message levels of the enzymes of the tricarboxylic acid (TCA) cycle, which is critical for several biosynthetic pathways. The results could reveal peripheral and central energetic alterations that may contribute to HD pathology.
Author: Gary Gibson
Approved: October 2012
Muscle pathology in HD as a source for biomarkers?
Peripheral manifestations of HD may provide an easily accessible and valuable source of HD biomarkers. Muscle wasting is a well-recognised phenomenon in patients with HD and worsens with disease progression. Interestingly, inflammatory processes can be linked to the development of muscle atrophy and immune activation in peripheral blood monocytes can be seen in HD. In this project, we will investigate the relationship between immune activation and muscle wasting in HD. We will study muscle pathology in relation to inflammatory cytokines in HD mouse models and in pa-tients with juvenile onset of HD.
Author: Maria Bjorkqvist
Approved: July 2010
The studies on the biochemical markers characterizing Huntington´s disease –determination of the profile of common amino acids in the serum of patients in different stages of Huntington´s disease
Decreased levels of branched amino acids (BCAA) correlate with the number of CAG repeats, UHDRS® scores and weight loss in HD patients. This study will determine the profile of common amino acids in the serum of HD patients and normal controls. The results might clarify the role of certain amino acids further, lead to metabolic markers of early disease stages and could be crucial for the development of special nutrition supplements.
Author: Beata Gruber
Approved: November 2009
Molecular mechanisms of BCAA and energy metabolism study in peripheral blood mononuclear cells of Huntington’s disease patients
Energy production impairment and mitochondrial dysfunction play an important role in the pathogenesis of Huntington’s disease (HD), within and outside the outside central nervous system. Bioenergetics defects on the cellular level and low levels of branched chain amino acids (BCAA) in serum plasma of HD patients were previously described. Difficulties of the clinical onset description and HD progress detection are also related to a lack of specific and sensitive HD biomarkers. The objective of this study is to investigate bioenergetics defects in peripheral mononuclear blood cells of HD patients on a molecular level including BCAA and energy metabolism by evaluation of the gene expression level in order to establish molecular biomarkers.
Author: Jolanta Krzyszton-Rusjan
Approved: November 2009
Parameter optimisation for diffusion tensor imaging in Huntington’s Disease
Striatum and cortex are affected in HD. Disruption of fibres connecting these structures may play a role in symptom development. In preparation for a large Europewide diffusion weighted imaging study, this pilot study aims to identify suitable scanning parameters as well as post-processing methods for the analysis of cerebral white matter. In addition, we attempt to identify possible gains in sensitivity of the technique at higher MRI field strengths. To this end, HD gene mutation carriers and matched controls will be studied using different MRI hardware at the Universities of Ulm and Freiburg, Germany.
Author: Jan Kassubek
Approved: November 2008
Treatments
A qualitative exploration of gaps in mental health support for people affected by Huntington’s disease in England and Wales
People who carry the HD gene expansion can experience many mental health difficulties, affecting quality of life. Many people affected by HD consider mental wellbeing a key issue. However, difficulties accessing mental healthcare are common, and evidence is lacking regarding intervention effectiveness. We aim to understand the mental wellbeing needs of people affected by HD in the UK, and the barriers to good mental healthcare. Accordingly, we will interview and analyse data from people carrying the HD gene, other HD family members, and healthcare workers supporting people affected by HD. We aim to generate evidence to improve mental health support.
Author: Sarah Gunn
Approved: April 2023
Huntington Partner in Balance: online self-management for partners/caregivers of persons with Huntington Disease (HD)
Overburdening the caregiver can lead to anxiety and depression and ultimately to inability to maintain their informal role in the treatment of HD. It is therefore of great importance to preventively increase the resilience of caregivers and in this way prevent overloading at a later stage of the care process. This project aims to develop and evaluate an online self-management program for partners, relatives, caregivers of HD patients. This intervention will be based on the blended care self-management program Partner in Balance (PiB) for partners of patients with dementia, which has proven to be effective (https://www.partnerinbalans.nl/home/en/).
Author: Annelien Duits
Approved: May 2020
Establishing and validating a SOP for state-of-the-art post-mortem analyses of fetal grafts in Huntington Disease patient’s brains
Replacing lost cells in Huntington’s Disease (HD) by transplantation seems like an intuitive treatment but bigger studies failed to prove efficacy. Analyzes of brains donated by HD patients who received transplants, demonstrated viable transplanted cells so the reason for this failure remains elusive. Only expanding our knowledge on graft integration can help us understand if this therapy can be made more effective. We therefore plan to analyze further donated post-mortem brains of HD patients by MRI imaging and state of the art histological analyzes to find out if and how strong grafted neurons integrated into the host’s brain.
Author: Philipp Capetian
Approved: January 2020
Exploring the efficacy of narrative interventions post predictive testing for HD: A pilot study
This study involves a pilot of a narrative group session to help support individuals post predictive testing for HD. The aims of the 2 hour session are to foster resilience and strengthen existing coping using narrative therapy principles. Three genetic counselling narrative groups will be held with 8-10 participants in each group. Individuals who consent to take part in the research will complete two short questionnaires before and after the session and will be invited to take part in a telephone interview two weeks later. The semi structured interview will explore individual experience of taking part in the group.
Author: Rhona MacLeod
Approved: April 2017
Investigating the therapeutic potential of manipulating DNA repair in Huntington’s disease
New evidence from genetics highlights DNA repair as a mechanism underlying HD and in this project we are exploring the changes in the DNA repair machinery. Intact DNA is vital in cells so all cells have machinery to fix DNA damage. To repair DNA the machinery needs to cut out the incorrect segment and replace it with the correct one. If it cuts in the CAG repeat, when the gap in the DNA is filled the machinery may loose count and inserts too many CAGs, leading to further expansion of the repeat. We know this expansion happens in HD and we are developing an assay to investigate this in many samples at once, in order to develop potential drugs to prevent CAG expansion.
Authors: Lesley Jones, Thomas Massey, Anne Rosser and Stephen Jackson
Approved: September 2015
Investigation of the effect of dopamine depletion on hippocampal-dependent cognitive function in Huntington’s disease patients
Huntington’s disease patients are commonly prescribed olanzapine, a serotonin and dopamine receptors antagonist due to its effect on chorea, sedation and weight. Previous work from our lab suggests that anti-dopaminergic drugs may also have an effect on cognition in HD.
The purpose of this study is to evaluate the effect of olanzapine and other anti-dopaminergic agents on cognitive function in early HD using the data collected in the REGISTRY study.
The study will enable us to get a better understanding of the impact of existing prescribing practices and dopaminergic modulation on the cognitive functions in Huntington’s disease.
Author: Roger Barker
Approved: August 2015
A pilot evaluation of mindfulness-based cognitive therapy for people with Huntington’s disease
It is acknowledged that many people at various stages of Huntington’s disease (pre-symptomatic and symptomatic) experience low mood, anxiety and other psychological difficulties. However, very little research has been conducted to investigate whether psychological therapies could help people with Huntington’s disease with these difficulties. While medication might be effective for some people, it is not suitable for all and psychological approaches should be developed. This study will provide the first indication of whether mindfulness-based cognitive therapy, a therapeutic approach with an established evidence base, would be acceptable and useful for people with Huntington’s disease with low mood.
Author: Jane Simpson
Approved: January 2014
Targeting the role of the Rab family GTPases in Huntington’s disease
The misfolding and aggregation of mutant huntingtin protein is a critical initiating factor in Huntington’s disease. Several cellular processes are disturbed due to this aggregation, including vesicle trafficking, which is required for movement of cargo within and between cells, and has been implicated in disease pathogenesis. Rab family GTPases play a crucial role in these cellular processes, and two members of this family are known to be neuroprotective in HD models. However, a systematic survey of these GTPases in models of Huntington’s models has not been performed, and therapeutic strategies targeting these genes are lacking. In this project will will implement genetic approaches to systematically interrogate the thera-peutic potential of Rab family GTPases in a human cell model of Huntington’s, with the aims of identifying novel therapeutic targets and clarifying the mechanisms underlying this process.
Author: Flaviano Giorgini
Approved: January 2013
Safety and tolerability of tetrabenazine in Huntington’s disease in clinical use – data from the REGISTRY cohort
Neuroleptics and Tetrabenazine are both used in order to treat chorea in HD patients. So far Tetrabenazine is not known to cause tardive dyskinesia – a severe possible complication of long use of neuroleptics. However, other side effects as depression and increased suicidality might be possi-ble. Little is known about interactions of Tetrabenazine with comedication, e.g. parallel use of tetrabenazine and neuroleptics. The aim of this retro-spective study will be to explore safety, side-effect profiles, duration of therapy, reasons for discontinuation, and data of exposure to co-medication in individuals with HD who used Tetrabenazine in comparison to Tiapride, which is frequently used to treat chorea off-label.
Author: Ralf Reilmann
Approved: September 2012
Improving function in Huntington’s disease through neurofeedback: using real-time fMRI to enhance cortical plasticity in early stages of the disease
The current project is a pilot study on the use of neurofeedback training using real-time functional MRI as a non-invasive intervention in Huntington’s Disease (HD). During neurofeedback training patients receive feed-back about the level of activity in a brain area, affected by the disease, and learn to selfmodulate their brain activation. The hypothesis is that by regulating and restoring function in affected brain regions, motor and cognitive function will also be restored. Following promising results in Parkinson’s Disease and depression, the current study will focus on early stage HD and collect preliminary data on the feasibility of the intervention.
Author: Sarah Tabrizi
Approved: September 2012
A survey of dietary intake in patients with Huntington’s disease
A deficient nutritional status may be an important determining factor in patients with Huntington’s disease, leading to increased comorbidity and social costs, and impaired quality of life. The aims of this study are to analyze the dietary intake of patients with presymtomatic and symptomatic Huntington’s disease, and to determine the association of the severity of motor, cognitive, psychiatric symptoms, and comorbidity with poor nutritional status in these patients. The determination of the nutritional status of patients with Huntington’s disease in its different stages, could help to introduce appropriate dietary measures to prevent complications, and to improve the quality of life of patients and caregiver burden.
Author: Esther Cubo
Approved: December 2011
Outreaching coordinated multidisciplinary care for Huntington’s disease patients
Most Huntington’s disease patients, together with their families, experience crisis-like events during the course of this progressive disease. Crises like fractures, resignation, divorce, or suicide attempts, seem related to the symptoms of the disorder. Experts in the field state that proactive multidisciplinary coordinated care for HD patients could reduce crises. This study aims to describe the crises prevalence and to investigate quality of life and caregiver burden. Multidisciplinary coordinated care will be provided to patients in their own homes. The study is set up to investigate the applicability and feasibility of a test protocol in preparation for a larger study on efficacy of this type of care.
Author: Ruth Veenhuizen
Approved: December 2011
Feasibility and benefit of inspiratory muscle training in people with Huntington’s disease.
A recent cross sectional study of respiratory function suggests that people with Huntington’s disease may have reduced strength of the breathing muscles, in the middle and late stages of the disease. Research in other neurodegenerative disease has shown that these muscles can be strengthened by using a small hand held device that provides resistance to the in-breath. This randomised, controlled feasibility study will investigate the benefit and perceptions of respiratory muscle training in 20 people with HD who have established respiratory muscle weakness. The primary outcome will be sniff nasal inspiratory pressure; adherence will be recorded by the device.
Author: Una Jones
Approved: July 2011
Development of predictive models to identify patients to recruit for clinical trials
Differences in the expression and progression of symptoms in individuals with Huntington Disease presents a significant challenge for clinical trials that are aimed at early intervention. It is important that individuals recruited into clinical trials include those that show measureable symptoms that progress within a reasonably short time period. We have used data from REGISTRY to identify a cohort of individuals who show a robust increase in motor symptoms over relatively short period of time. The aim of the present study is to develop predictive models to identify those that will progress in this manner.
Author: Anthony Vaccarino
Approved: December 2010
Memantine treatment in the REGISTRY cohort
The aim of this retrospective study will be to explore potential efficacy signals in individuals with HD who use memantine. We will review availability of datasets from the REGISTRY, number of visits available (follow-up duration), and doses applied. Our primary endpoint will be rate of decline in HDRS total motor score. Secondary endpoints will be rate of decline in UHRDS cognitive score, UHDRS® functional assessment and total functional capacity. We will review the feasibility to compare the clinical readouts available to a group of matched subjects from the REGISTRY cohort on no treatment with Memantine.
Author: Jan C. Frich
Approved: April 2010
Understanding prescribing habits of existing potential neuroprotective substances in HD
It is unknown how many patients receive antioxidants and potentially neuroprotective substances in what dosages and to what effect. Understanding existing prescription habits, and their differences, is important to develop effective clinical trials of both existing and emerging potential neuro-protective substances. We propose a two step analysis. First we would like to analyse prescribing habits in the various countries contributing to REG-ISTRY. Second, because the use of over-the-counter medications may be underreported, we would like to involve investigators in a subset of Cen-tres to take part in a more systematic survey of the use of over-the-counter medications.
Author: Herwig Lange
Approved: June 2009
Depression in Huntington’s disease, its treatment and associations
Depression is common and disabling in Huntington’s disease. This project aims to look at rates of depression in the different sites and countries of the EHDN. We will also examine how commonly depression is treated in HD and the types of treatments, dosages and duration of treatment. Our final aim is to examine what factors influence the severity of depression in HD. These factors may include the CAG repeat length, type of HD, previous psychiatric history or a variety of other demographic factors. The study may thus shed light on treatment practices across Europe and identify risk factors for depression in HD.
Author: Hugh Rickards
Approved: December 2008
Reliability and minimal detectable change of measures of participation, functional activities and impairments in individuals with Huntington’s disease
Huntington’s disease (HD) may be amenable to physiotherapy in terms of restoring or maintaining functional abilities. To date, there has been relatively little research to substantiate this suggestion. One of the first steps in developing intervention trials is to choose appropriate outcome measures. The purpose of this multi-centre project is to evaluate various potentially suitable measures. We will recruit a total of 80 patients with HD to undergo two assessments, with a one-week gap between, on a range of functional measures that have been chosen to reflect a range of impairments and activity limitations seen in people with HD.
Author: Monica Busse
Approved: October 2008
Survey of Pharmaceutical interventions in JHD
There are no guidelines for the management of JHD. As a prerequisite to further studies and future statements about the quality of care for JHD we want to know the current prescribing habits: two approaches have been used: interrogation of the REGISTRY data on medications which have prescribed and a survey of parent/carers in the UK. The results have been collected and a publication is planned.
Author: Oliver Quarrell
Approved: June 2008
Utilisation of rehabilitation services in Huntington’s Disease
Patients with HD are at risk of falls. However, they may not be referred enough to physiotherapy for management of their mobility problems. We therefore plan to investigate the referral of HD patients in Europe for re-habilitation interventions as well as types of help and home adaptations required. In order to investigate this, we will use Registry data including UHDRS® general history (age, age of onset, past medical history), UHDRS® motor & function and health economics data. We will specifically consider physiotherapy, occupational therapy and speech therapy requirements. This may have implications for planning and justifying physiotherapy interventions and understanding the rehabilitation needs of patients with HD.
Author: Monica Busse
Approved: November 2007
Prescription habits in Huntington’s disease
An evidence-based treatment in Huntington’s disease (HD) is flawed by the lack of proven efficacy regarding pharmacological interventions presently used for symptomatic control. Disease modifying interventions are unavailable at this stage. Physicians or patients attitudes, biological factors or other variables may individually and regionally affect the way physician treat HD-related symptoms. The investigators aim at studying prescription habits in HD across the REGISTRY cohort: a first step comprehends the description of therapeutic habits in the cohort followed by the analysis of demographic and clinical variables and their contribution for the observed prescription patterns.
Author: Tiago Mestre
Approved: November 2007
New assessment tools
Social cognition and quality of life in Huntington’s disease
People with Huntington’s and their families often report difficulties with social interaction. Research has also shown that HD can affect the ability to recognise emotions, and to reason about other people’s thoughts. This study will use cognitive tasks and questionnaires to show which social skills are most important for everyday functioning and the quality of life of both people with HD and their close others. This information will improve future care by supporting the design of a new assessment about the social side of HD. The assessment can then be used in medication trials and to develop new psychological treatments.
Author: Clare Eddy or Clare Eddy 2
Approved: April 2023
Monitoring prodromal hyperkinetic movements using body worn sensors in Huntington’s Disease
Motor symptoms may be of transient nature in prodromal and early HD. Thus, they may be missed upon clinic visits. Yet, there is a need for a more sensitive detection of early motor symptoms in HD, and for a sensitive progression marker in the transition from prodromal to motor HD. We use sensors to detect early motor signs of HD, aiming to facilitate an early motor diagnosis and monitor disease progression. Participants are equipped with 5 synchronized sensors to record involuntary movements while performing a set of different tasks. Coregistration of sensor data with video-recordings will allow us to develop algorithms that provide sensitive, objective and quantitative outcome parameters for detecting prodromal signs of HD.
Author: Franz Marxreiter
Approved: January 2021
Rasch Measurement Theory Analysis of longitudinal data of the UHDRS® Scales in the EHDN REGISTRY Legacy database
Data generated with rating scales can inform treatment decisions or be used as outcomes in clinical trials. It is therefore important to use scales that are fit for purpose; this requires a complete understanding of their performance. The Unified Huntington’s Disease Rating Scale (UHDRS®) was developed to measure the clinical features and course of Huntington’s disease (HD), a progressive neurodegenerative condition characterized by a mixed movement disorder, cognitive impairment and behavioural changes. HD. Based on an analysis of the UHDRS® using Rasch Measurement Theory we aim to provide recommendations for improving the performance and interpretability of the UHDRS®.
Author: Jean-Marc Burgunder
Approved: February 2019
Fitness-to-Drive in Huntington’s disease
Driving a car is important to people: it brings mobility and independence. This also holds true for people with HD. But, obviously, there comes a time when this is no longer possible. Very early on driving is not (yet) impaired; very late into the disease it is clearly impossible. But where is the divide?
We try to answer this by questioning partners and acquintances, by performing specialized psychological tests, and by assessing performance in a driving simulator and on-road. The psychological tests not only measure cognitive abilities (e.g. reaction speed, attention), but also so-called social-cognition tests that judge functions like self-reflection and risk-taking. Thus, we hope to come up with measures that provide a well-founded and personalized advice on fitness to drive for individuals with HD.
Author: Berry Kremer
Approved: January 2017
Skeletal muscle metabolic responses to physical exercise in clinical HD trials
Metabolic abnormalities and mitochondrial dysfunction in peripheral tissues such as skeletal muscle are components of the pathophysiology of HD. There is evidence to suggest that these differences in metabolic function lead to stimulation of anaerobic energy supply pathways associated with increased plasma lactate concentrations in response to exercise. The aims of this project are: a. to establish minimally invasive fine needle muscle biopsies as a tool to evaluate differences in metabolic gene expression profiles in response to acute exercise between HD patients and age matched controls and, b. to develop suitable endurance exercise programmes adapted to the capabilities of HD patients for future exercise training studies.
Author: Martina Velders
Approved: January 2014
The swallowing disorder in Huntington’s disease (HD): an observational, longitudinal study
Dysphagia is a common condition in Huntington´s Disease (HD). With the progression of the underlying disease the swallowing function worsens. In particular, the risk of aspiration increases, which is associated with bronchopulmonary infections and mortality. Little is known about onset, prevalence and characteristics of dysphagia in HD. Our aims are to describe frequency, onset and progression of dysphagia in HD and to determine feasibility and reliability of measurement methods that are potentially suitable for HD patients. Therefore we are currently investigating HD patients (2 centers, 60 patients) at a baseline visit and one follow-up visit using swallowing and nutrition questionnaires, a clinical swallowing examination, fiber endoscopic evaluation of swallowing and the UHDRS®.
Authors: Falk Schradt; Christina Lang
Approved: September 2013
Longitudinal evaluation of the Registry cognitive battery across the different stages of Huntington’s disease
The Registry study provides an opportunity to carry out a longitudinal, international study of the most commonly used cognitive tasks in Huntington’s disease (HD). In HD cognitive decline can start early, can be objectively measured and influences the functional independence of an individual. This indicates that cognitive measures have the potential to be used as outcome measures in future clinical trials. But first it is pertinent to evaluate the efficacy of the Registry assessment in terms of tracking disease progression through the stages. This information is extremely important for developing future clinical trials.
Author: Verena Rödig
Approved: July 2013
PHASE1-HD: a pilot phase-contrast MRI study of cerebrospinal fluid dynamics in Huntington’s disease
Cerebrospinal fluid, or CSF, is a clear liquid which surrounds the brain and spinal cord. Research in Huntington’s disease animal models suggests CSF flow might be altered in HD, which could be important for new drugs that may be injected into the CSF. A scanning technique called Phase Contrast MRI enables CSF flow to be studied non-invasively. PHASE1-HD will develop the Phase Contrast technique for HD and test it in 10 patients and 10 controls to help understand CSF flow and design larger trials of the technique.
Author: Ed Wild
Approved: January 2013
Smart phone App and optical motion sensing devices (Kinect/X-Box) as tools to assess motor function in HD – a Pilot Study
Our project aims to develop easy-to-use electronic tools to objectively assess the motor impairment in people with Huntington’s disease. Motor symptoms such as chorea are debilitating diseases symptoms and also important readouts for therapeutic and genetic studies. However, objectively recording and quantifying motor symptoms remains challenging. We have developed a prototype app that takes advantage of the sophisticated movement sensors built into standard smart phones to detect and quantify hand and limb movements. This tool is currently being optimized and validated for use in the clinical assessment of patients and controls.
Author: Patrick Weydt
Approved: May 2012
Theory of Mind Deficits in Huntington’s Disease
BACKGROUND: Studies suggest Huntington’s disease (HD) can be associated with changes in emotions and social behaviour. We are researching the changes that some patients show when they are asked to think about other people’s beliefs and emotions.
AIMS: This study will help us to understand the brain basis for patients’ difficulties on particular social cognitive tasks and ultimately, some of the behavioural changes seen in HD.
METHODS: People with HD will complete tasks which involve thinking about people’s emotions while undergoing brain scanning. Some of the patients taking part in the study have the HD gene but have not yet developed movement symptoms so that we can investigate relationships between social and emotional changes in HD and disease onset and progression.
Author: Clare M. Eddy
Approved: August 2011
Revisiting the Stroop test as a measure of cognitive deficits
The proposed project aims at a detailed analysis of UHDRS® cognitive assessments employed in the REGISTRY study. Particular focus shall be on the Stroop test. Here, it is our interest to compare the subscores from the colour, word and interference card, as well as other scoring methods derived from these raw scores.
On the one hand we would like to understand how the above scores develop as HD progresses. In addition, we hope to figure out the extent to which performance on these tests is influenced by other factors such as the UHDRS® Total Motor Score, age and education.
Thereby we hope to understand in more detail the dimensions of HD that are measured by the Stroop test and to provide potential optimising strategies for both, the testing procedure as well as its evaluation.
Author: Nathalia Weber
Approved: May 2011
Analysis of the behavioural UHDRS®
In our first publication (Rickards et al, 2010) the Behavioural Working Group of the EHDN defined distinct behavioural patterns within HD, comprising a depressive factor, a dysexecutive factor, an irritability factor and a psychosis factor. Starting from this point, we plan to analyze, as a next step, the dependence of those factors on important clinical variables like sex, stage of disease, motor and cognitive functions, age of onset, duration of the disease, psychiatric medications and psychiatric history. Furthermore, we are interested in regional differences of the behavioural patterns of the whole area of EHDN.
Author: Raphael Bonelli
Approved: March 2011
The relationship of chorea with function and cognition
HD is characterized by progressive motor impairment. These motor disturbances can be divided into two subgroups: the more rigid type (hypokinetic) and the more choreatic type (hyperkinetic). Patients with HD become increasingly more disabled in their activities of daily living as the disease progresses. Also, their cognitive capacities decline over time, eventually resulting in a dementia. The clinical impression of many neurologists is that those with severe chorea seem to function better compared to the more rigid patients. Up to date this has not been investigated thoroughly, therefore we make use of the extended REGISTRY-database of EHDN to study this clinical idea. In this project we aim to investigate the differences in general and cognitive functioning between predominantly choreatic and predominantly rigid HD patients.
Author: Raymund Roos
Approved: January 2011
Investigation of Time Course and Functional Impact of Voluntary Motor Function Impairment in Huntington’s Disease
The UHDRS® motor scale includes assessment for eye movement abnormalities, voluntary motor function and involuntary movements. The aim of this work is to examine the impact these three clinical domains on the functional decline HD as measured by UHDRS® functional assessments in 300 patients obtained from the REGISTRY data base. We will also measure the rate of decline in these measures over one year of progression of disease, as well as the impact of concomitant medications, demographic factors and CAG repeat length. It is hypothesized that voluntary motor function is more closely linked to functional impairment in HD than involuntary movements. The voluntary motor function score may thus be more useful for studying effects of experimental therapeutics and to monitor progression of HD over time.
Author: Joakim Tedroff
Approved: February 2009
The familiarity of psychiatric symptoms in Huntington´s disease
A wide range of behavioural problems are commonly seen in HD, including depression, anxiety, obsessive-compulsive symptoms, irritability, aggression, disinhibition and apathy. Improved understanding of the cause (or causes) of such psychiatric symptoms independent of the HD gene is critical to developing effective treatments and consequently improving the quality of life of patients. This study aims to determine whether certain psychiatric symptoms cluster in families affected with HD. This will be achieved by conducting a thorough, standardised interview of 80-100 families with sibling pairs with HD as well as any siblings without the HD gene.
Author: Jenny Keylock
Approved: February 2009
The accuracy of the estimation of age at onset of motor signs in HD
In Huntington’s disease (HD) the accurate determination of age at onset is important for counselling patients and critical if one tries to develop and evaluate therapies that aim to delay it. Here we compare the REGISTRY raters’ estimates of age at onset with estimates calculated using a number of algorithms such as the Langbehn or the Aylward fomula. A second aim is to calculate disease progression using data from at least three visits. Thirdly, we use longitudinal data for (backwards) extrapolation of actual age at onset and a comparison with rater’s estimates and formula estimates.
Author: Michael Orth
Approved: December 2008
Towards an improved Functional Rating Scale for Pre-Huntington’s Disease
Current functional ratings scales are insensitive and unsuitable for pre-manifest and early Huntington’s disease (HD) subjects. Because functional outcomes are very important in drug development a new functional rating scale is needed for this subgroup. We propose to use an item response theory (IRT) approach to characterize symptoms (including psychiatric, cognitive, motor and quality of life domains) in pre-HD and all stages of HD. With data from REGISTRY, we will analyze the relationship between the score assigned to a scale item and the overall severity of HD. This analysis will identify suitable items from pre-existing scales that are particularly relevant for pre-HD and early HD, and provide valuable information that will guide the development of a new and sensitive functional scale for this particular patient sub-group.
Author: Aileen Ho
Approved: October 2008
Reliability and minimal detectable change of measures of participation, functional activities and impairments in individuals with Huntington’s disease
Huntington’s disease (HD) may be amenable to physiotherapy in terms of restoring or maintaining functional abilities. To date, there has been relatively little research to substantiate this suggestion. One of the first steps in developing intervention trials is to choose appropriate outcome measures. The purpose of this multi-centre project is to evaluate various potentially suitable measures. We will recruit a total of 80 patients with HD to undergo two assessments, with a one-week gap between, on a range of functional measures that have been chosen to reflect a range of impairments and activity limitations seen in people with HD.
Author: Monica Busse
Approved: October 2008
Measuring outcomes in Huntington’s disease (HD)
Rating scales are important because they are the principal outcome measures used in drug trials. The REGISTRY data can be used to assess the performance of the rating scales. Two different analytic approaches were used: “traditional” psychometric methods and Rasch analysis. This enabled us to construct a detailed and sophisticated picture of the motor, total functional capacity and functional assessment subscales of the UHDRS®, identify their strengths and weaknesses, and provide recommends to maximise their ability to measure the impact of HD. The results are available as a report to EHDN and are the basis of a grant application to develop improved outcome measures for drug trials.
Author: Oliver Quarrell
Approved: June 2008
Late stage HD: phenotype and current management
The clinical characteristics of late-stage Huntington’s disease (LS-HD) have not been extensively documented. The prevalence of LS-HD may, however, increase since with better general health care and possibly better clinical management patients with HD will live longer. Knowledge of the magnitude and causes of patients` disability may allow to improve the care of LS-HD patients` and to focus therapeutic interventions. Using data from REGISTRY we therefore will first describe the phenotype of LS-HD and the current management practice. We will then analyse the contribution of each symptom complex (motor, behaviour, cognitive etc) to the phenomenology of LS-HD.
Author: Miguel Coelho
Approved: November 2007
The use of depression rating scales in patients with HD
Depression is a common, yet treatable complication of HD. Its recognition and assessment with the most valid rating scales is therefore very important. The Beck Depression Inventory (BDI) and the Hamilton Depression Rating Scale (HAM-D) are currently used in the REGISTRY study. However, they may not be ideal since they also measure e.g. physical symptoms of a degenerative disorder such as HD. Our goals are to evaluate the validity of the BDI and the HAM-D against the UHDRS® Behavioural Scale. In addition, Principal Components Analysis intends to find out how helpful individual items are in measuring depression.
Author: Jenny Keylock
Approved: November 2007
Epidemiology
Pain in Huntington’s Disease
Pain is often reported by patients with Huntington’s Disease (HD) and needs further study. The aim of this study is to assess the prevalence of pain, pain interference on daily activities, painful conditions, and the use of painkillers across the different stages of HD and to compare these prevalence to those without HD. Compared to the Enroll-HD dataset, the Registry dataset also includes a pain intensity scale, which provides a unique opportunity to more comprehensively assess the relationship between HD (severity) and pain, in order to get a better understanding of pain in HD and to improve pain management.
Author: Gregory Sprenger
Approved: January 2021
CAG and Age of Death: A Reanalysis
In HD, the length of the CAG trinucleotide expansion is clearly linked to the age of disease onset. However, there is controversy about the influence of CAG repeat length after the onset of disease. Recently, it was claimed that, after considering the age of onset, the CAG length plays no further role in predicting the lifespan of persons with HD. This claim was based partly on an analysis of the EHDN data. As a biostatistician with many years of HD research experience, I will reanalyze that data to see if I can draw the same conclusion.
Author: Douglas Langbehn
Approved: December 2018
Clinical characteristics of the Huntington disease mutation carriers in Russia
So far, no comprehensive clinical characterization of HD mutation carriers in Russia has been conducted. The proposed project is intended to provide a comprehensive clinical description of Russian HD mutation carriers based on the data collected within REGISTRY study — a multisite, prospective, observational study of HD mutation carriers and control subjects with annual follow-up visits. We seek to obtain genotype and phenotype characteristics of HD mutation carriers and to compare treatment approaches in Russia and other countries which participated in the REGISTRY study.
Author: Yury Seliverstov
Approved: December 2017
Steady State Simulations study in HD
Failure to renew the pool of active participants and the natural progression of ones recruited tend to drift the population in longitudinal observational studies towards older age and higher degrees of severity.
We seek to develop a set of numerical and statistical simulations that will help understand how manipulations of the entry and exit of participants from the active pool of Registry can impact in the long term stability of that pool in terms of distribution of types of participants and total number of participants. The goal is to understand the pattern of recruitment and dropout rate in Registry.
Author: Daisy de Abreu
Approved: August 2017
Prevalence and clinical correlates of homozygosity in Huntington´s disease
Because patients homozygous for Huntington´s disease (HD) receive the gain-of-function mutation in a double dose, one would expect a more toxic effect in homozygotes than in the heterozygotes. The goal of this study is to investigate the phenotypic differences between the two genetic groups (homozygote with both alleles > 36 CAG repeats, and heterozygote with one allele > 36 CAG repeats) in terms of onset of symptoms, phenotypic presentation and disease progression.
Author: Esther Cubo
Approved: December 2016
Evaluating the effect of substances abuse on disease progression in HD
Studies have shown that substances of abuse (tobacco, alcohol, drugs) lead to an earlier age of motor onset of HD. However, there is no data looking at how these substances affect patients with HD over time. Therefore, we are using the Registry dataset to see if these substances have an effect on motor scores, total functional capacity, and other markers of disease progression. If we are able to more accurately understand the effect that substances of abuse have on disease progression, we would like to look at other agents, such as medications that may impact progression of HD.
Author: Jordan Schultz
Approved: October 2016
Clinical and genetic characteristics of late onset Huntington’s disease
Age at onset of Huntington’s disease (HD) for most patients is in their forties. However for a substantial part of HD patients symptoms and signs start after 60 years of age. We wish to investigate this group of patients for phenotype, disease progression, CAG repeat length and family history. Several recent studies have been done, confirming that the main symptom in late onset HD are mild motor symptoms. However some confirm that negative family history is significantly higher in late onset others cannot support this. Diagnosing late onset HD might be challenging, especially when the family history is negative and chorea is the main feature. Our aim is to investigate if late-onset HD differs from usual onset in phenotype, disease progression, CAG repeat length and family history.
Author: Mayke Oosterloo
Approved: October 2016
Do autoimmune neuroinflammatory diseases influence the course of HD? Co-incidence of HD and MS, CIS or acute myelitis transversa
This project aim is to investigate the influence of clinical isolated syndrome (CIS), multiple sclerosis (ED), and acute myelitis transversa (MT) as typical neuroinflammatory diseases on i) the age of onset (AO) and ii) the course of HD. We expect only a small number of patients suffering from both HD and one of the following: CIS, ED or MT. As control group we ask for data of AO from other HD patients carrying the same CAG repeat length. It is still unclear if neuroinflammation in HD is a reactive process or if there is an active influence on disease progression. Further understanding of the influence of inflammation in HD may open various avenues for promising therapeutic approaches aiming at slowing disease progression or forestalling onset of disease (Ellrichmann et al., 2013).
Authors: Gisa Ellrichmann, Carsten Saft
Approved: August 2016
Pain in Juvenile Huntington’s disease
Some individuals manifest Huntington’s disease before 21 years-old and are classified as juvenile HD (JHD). Our clinical experience suggests that chronic intractable pain is an under-appreciated feature in JHD. The purpose of this study is to determine the frequency of pain in JHD using data from REGISTRY and the JHD substudy. Additionally, we will determine the influence of demographics, family history, genotype, phenotype, medication, co-morbidities on pain frequency. Ultimately we hope to build on these data to produce an evidence-based guideline for managing pain in JHD.
Author: Filipe Brogueira Rodrigues
Approved: May 2016
The effect of homozygosity of the huntingtin mutation
There is debate regarding whether, in the rare event that a person has a CAG expansion in both copies of the huntingtin gene (homozygosity), their age of onset and their progression in HD is affected. Among current HD databases, EHDN Registry possibly contains the largest number of such subjects. We will statistically model the effect of this second elongated HD allele in an attempt to clarify the clinical effect, if any.
Author: Douglas Langbehn
Approved: September 2015
Computational outlier analysis of GWA phenotypic dataset- a machine learning exercise
CHDI with collaboration of PsychoGenics (PGI) will explore the use of combinations of clinical features to identify phenotypically unusual individuals via machine learning approach, with an initial stage of analysis to involve patient classification by phenotypic feature values and differences between feature-to-feature relationships.
The objective is to have a model that explains the response variable in terms of how all other features would allow one to define explanatory features’ ranks, and will provide a qualitative insight of what’s important for defining HD severity by phenotypic characteristics. In addition, we will apply PGI exclusive outliers identification technique which defines an outlier as a data instance on which the classifier was trained on, and yet unable to recognize to that data instance.
Authors: Anna Begelfer-Ostrovski, Seung Kwak
Approved: September 2015
Modeling HD Clinical Assessments and Comprehensive HD Progression
Data will be used towards the following analyses, as part of CHDI/IBM collaboration:
Clinical Assessment – predictive statistical models combined with variable-selection will be applied to both imaging and non-imaging clinical datasets and used to detect most relevant groups of clinical variables to produce new biomarkers.
Machine Learning for Comprehensive HD Progression Modeling – advanced machine learning and data mining methodologies for data driven healthcare analytics developed by the IBM Healthcare Analytics Research Group will be applied to the multiple sets of longitudinal data covering multiple domains collected by the HD research community.
These models will support more effective clinical trial design, provide guidance to new experiments by identifying gaps, and provide researchers in different domains with a more integrated view of disease states and progression.
Authors: Anna Begelfer-Ostrovski, Cristina Sampaio
Approved: September 2015
Disease progression models for Huntington’s disease using population analysis
The development of disease progression models for Huntington’s disease (HD) will help to understand the disease mechanism, predict disease diagnosis and progression, and provide a framework to evaluate new therapeutic agents in HD that potentially slow down the disease progression, delay and eventually prevent the disease onset. By using population analysis approaches, we aim to quantitatively assess the longitudinal changes of various HD clinical rating scales and tests in domains of motor function, cognition, behavior and functional capacity. We will also determine and evaluate the possible demographic, genetic, and medical-related factors that contribute to the variability in disease progression.
Author: Xiaomeng Jiang
Approved: July 2015
The progression of different motor subtypes in Huntington’s disease and their relationship to general and cognitive functioning
The aim of the project is to investigate the progression of different motor subtypes in Huntington’s disease (HD) and their relationship to general and cognitive functioning. We want to investigate if the motor disturbances seen in HD change from predominantly choreatic to predominantly hypokinetic-rigid when the disease progresses. A previous cross-sectional study showed that predominantly choreatic patients perform better on general and cognitive assessments. The Registry database gives an opportunity to study this further on a large scale and longitudinally. Longitudinal study of the motor subtypes could provide a better understanding of the different motor phenotypes in HD and may have implications for future care and clinical trials.
Author: Milou Jacobs
Approved: May 2015
Huntington disease- prevalence and disease severity in Jämtland and Uppsala
The aim of our study is to estimate the prevalence of Huntington’s disease (HD) in the two countries Jämtland and Uppsala in Sweden. We want to compare the symptom severity between HD patients followed by a multi-disciplinary team in Uppsala and HD patients in Jämtland, who do not have access to similar care. Then we want to compare the disease severity in the Swedish patients and in patients from the rest of Europe, using the Registry data.
Author: Anna-Karin Roos
Approved: December 2014
Investigating irritability in Huntington’s Disease; is there a relationship between executive dysfunction and irritability (both internally and externally-directed)?
We are performing a study that looks at how people with Huntington’s Disease become irritable. We have divided irritability into that directed at other people, which can be recognised as shouting, snapping and sudden out-bursts; and that directed towards oneself. We know that some people with Huntington’s Disease have problems performing certain mental tasks. We predict that irritability directed at others may be related to problems with processing lots of information at once. To investigate this, we will use data from performance on cognitive tasks which probe the ability to deal with mental interference, and establish whether this relates to irritability.
Author: Sophie Green
Approved: July 2014
Estimating costs of health and social care resource use associated with Huntington’s disease in the United Kingdom
As Huntington’s disease (HD) progresses, intensive support from health and social care services is likely to be needed yet little is known about the economic cost of HD. This project will use REGISTRY data on health and social care use, informal care time and impact on employment to estimate an annual cost per person with HD in the UK, stratified by age and disease severity. Identifying the patterns and cost of service use associated with HD could potentially lead to improved resource allocation and ultimately care for people with HD.
Author: Monica Busse
Approved: July 2014
The Economics of Huntington’s Disease
Research underway will begin to examine under-explored economic aspects of HD by using individual participant data from the EHDN. The research has two streams: i) estimation of resource use/costs of care for people with HD and; ii) estimation of health outcomes data (health state values) suitable for use in health policy contexts. Such health state values are needed to calculate quality-adjusted life-years (QALYs), which are used for exploring the effectiveness of treatments and their value for money. This research aims to highlight the impact of HD and provide information for use in decision-making regarding the care of people with HD.
Author: Annie Hawton/Colin Green
Approved: April 2014
Psychological predictors of pain in people with Huntington’s Disease
The proposed project is a data mining study to identify psychological factors that predict pain severity in people with Huntington’s Disease (HD) at their most recent assessment and develop a model to account for the variance in pain severity. Factors to be investigated include low mood, anxiety, aggression, irritability, apathy, delusions and hallucinations with ordinal regression being used to analyse the data. The secondary aim of this research is to gain an understanding of the prevalence of pain in HD. This will be considered alongside the disease stage, to investigate whether pain severity differs according to disease stage.
Author: Mandy Underwood
Approved: January 2014
Prevalence and clinical correlates of the intermediate CAG repeats in Huntington’s disease
The underlying genetic cause of HD is a CAG trinucleotide repeat expansion in the HTT gene. CAG expansions above 39 account for the majority of HD presentations. CAG expansions in the 27-35 range are referred to as intermediate alleles. Based on last reports, it remains controversial whether intermediate alleles can cause HD. In this study, we will use data from the EHDN registry and include participants with intermediated CAG repeats (>27-35, cases) and controls (< 27 CAG repeats). The aim of this study is to determine the prevalence and clinical characteristics of intermediate CAG repeats carriers, which might have important implications for the pathogenesis of the disease and genetic counseling. Author: Esther Cubo
Approved: September 2013
Completed suicide in a European Huntington’s disease population
Although the prevalence of suicide in Huntington’s disease (HD) is four to eight times higher compared with the general population, no prospective studies on risk factors of completed suicide in HD have been carried out so far. In this study we aim to identify the incidence rate of completed suicide in the REGISTRY population. Furthermore, we aim to determine sociodemographic, clinical, and neuropsychiatric predictors of completed suicide in HD.
Author: A.A.M. Hubers
Approved: September 2013
Huntington’s disease causes of death: a prospective observational study
It is well known that Huntington’s disease (HD) patients experience premature death. Nevertheless, HD mortality has not been extensively studied and additional knowledge on causes of death is important to plan care and supportive services for HD patients and their families.
With this study we propose to evaluate HD patients causes and places of death, the time between symptoms onset/diagnosis and death and the overall survival of HD patients. Additionally, we intend to determine the influence of demographics, family history, genotype, phenotype, medication and co-morbidities on the above-mentioned variables.
Author: Filipe Brogueira Rodrigues
Approved: July 2013
Suicidal ideation and behaviour in a European Huntington’s disease population
In this study we aim to identify the lifetime prevalence, incidence rate, and severity of suicidal ideation and behaviour according to the Columbia Suicide Severity Rating Scale (CSSRS) in the Registry population. Furthermore, we also aim to determine sociodemographic, clinical, and neuropsy-chiatric associations and predictors of suicidal ideation and behaviour.
Author: Erik van Duijn
Approved: July 2013
Cancer prevalence in HD
Studies in Scandinavian cohorts have found that people with expanded repeats in their HD gene (HTT) and their SBMA gene (the androgen receptor, AR) have significantly reduced cancer incidence. We wish to explore these findings further and examine the relationship between the CAG repeat in HTT and cancer prevalence in the EHDN Registry population. We will access the medical history, comorbidity and medication data to examine cancer incidence and prevalence. Though it is unclear how increased CAG length could mediate cancer risk, determining the relationship between CAG length in HTT may reveal fundamental biology underlying both cancer risk and neurodegeneration.
Author: Lesley Jones
Approved: March 2013
Does age of onset affect the clinical phenotype and its progression in adult onset Huntington’s disease? An observational 5 year follow up study
It is established that the age at onset of Huntington’s disease is affected by the size of CAG repeats. It is also known that patients with juvenile onset (below the age of 20 years) have different physical characteristics such as more akintetic rigid (Parkinsonian) features. On the other hand, patients with late onset disease (over 50 years) could have more cognitive features. However, it is not clear whether age at onset affects the clinical manifestations of the disease in the most commonly affected age group (30-60 years). Understanding such differences would have implications in managing patients in terms of counselling on prognosis, and for measuring effects of drugs. This study aims at studying the effect of age at onset on the clinical features of HD (motor, psychiatric and cognitive) and the rate of symptom progression.
Author: Sundus Alusi
Approved: March 2013
Late-onset Huntington’s disease
Huntington’s disease (HD) is a neurodegenerative disease clinically characterized by the triad of an extrapyramidal movement disorder, cognitive decline, and behavioral changes. HD is caused by an abnormal expansion of CAG-triplets in the huntingtin gene. Carriers of this mutated HD gene typically first develop symptoms in their mid-thirties to mid-forties, but age of onset for HD ranges from early childhood to the seventies and eighties. Little is known on the natural history of late-onset HD. The aims of this project are to characterize the phenotype and to determine the progression of patients with late-onset HD.
Author: Fabienne Sprenger
Approved: January 2013
CAG repeat length and JHD
It is frequently stated that the patients with JHD have more than 60 repeats. This is partially correct. In fact the median CAG repeat length is around 60 and in some cases JHD patients may have a CAG repeat length in the 40’s. Descriptive data on the age of onset and CAG repeat length for JHD patients will be analysed. The negative correlation between age of onset and CAG repeat length is well established but in two populations Andressen et all (Ann Hum Genet 71:295-301, 2006) demonstrated a better fit with a two segment regression line. The inflection points were at 53 and 49 CAG repeats in the respective populations. The aim of this data mining project is to establish descriptive statistics for the spread of the age of onset of JHD patients and to obtain a third result for an inflection point in a two segment analysis of the correlation between age of onset and CAG repeat length.
Author: Oliver Quarrell
Approved: December 2012
COHORT vs REGISTRY – comparison of two similar observational cohort studies on two continents
As HD is relatively rare, we need advanced, multi-centre, multi national frameworks that allow us to study simultaneously multiple complementary aspects of HD. This includes the natural history of HD, its management and the collection of clinical information and biosamples for research. In the analysis of the Registry cross-sectional data it was shown that across different European regions CAG repeat genotypes and phenotypes were similar, suggesting that treatment results are generalizable across Europe. We do not know, whether this holds true for North-America. We therefore plan to investigate whether there are any differences in genotype, phenotype or treatment characteristics between Europe and North-American HD patients, using the Registry and Cohort databases.
Author: Christine Tritsch
Approved: September 2012
Prevalence rates and course of suicidality and its socio-demographic and clinical associations in persons with Huntington’s disease
Suicide is a relatively frequent cause of death among patients with Huntington’s disease. Various studies show that patients with Huntington’s disease commit suicide four to eight times more often than persons from the general population. The risk of suicide seems to be related to the development of the disease and the loss of independence, and may not always be related to depression. This REGISTRY data mining project aims to identify 1) prevalence rates and severity of suicidality using the UHDRS® behavior section, and 2) associated socio-demographic and clinical characteristics.
Author: Erik van Duijn
Approved: March 2011
The impact of biological and environmental factors on independence in Huntington’s Disease
Huntington’s disease invariably leads to debilitating motor signs, cognitive decline and sometimes behavioural signs. In the course of the illness patients loose the ability to live independently. The impact of HD on patients’ independence can have a great effect on the patients’ sense of well-being. Thus it is important to identify factors that influence independence in order to promote those factors that maintain independence and try and avoid those that negatively influence it. We wish to use the REGISTRY data to identify some of these factors. The results of the study may help clinicians in their management of HD patients.
Author: Michael Orth
Approved: January 2011
Using Registry data to inform choice of outcome measures and optimal design of future clinical trials in Huntington’s Disease
The aim of this project is to use statistical modelling of longitudinal data from the Registry cohort to inform the design of future randomised controlled trials in Huntington?s Disease. The project will explore the potential of a range of functional, cognitive, and motor score variables as possible clinical trial outcomes. Questions concerning the optimal length of trials, the ideal frequency of interim visits, the best inclusion criteria and the merits of alternative designs such as those in which all patients are ultimately given an active treatment, will all be addressed.
Author: Chris Frost
Approved: November 2010
A data mining project investigating psychosis in Huntington’s Disease
Psychotic symptoms have a higher prevalence rate in HD patients compared to the general population. This data mining project aims to identify the prevalence and incidences of delusions and hallucinations in the REGISTRY data. The data will then be compared to identify any differences between the data sets of those who report these symptoms and those who do not. Further analysis is also expected to be carried out between those who only report hallucinations and those who only report delusions. The data sets will comprise of demographics, history, CAG, medication, motor, cognition, behaviour, function and TFC subscales of the UHDRS®.
Author: Jennifer Crooks
Approved: July 2010
Data quality in REGISTRY: completeness, plausibility and the effect of monitoring
REGISTRY, EHDN’s observational study, collects phenotypic and biological data. REGISTRY aims for high quality data. To this end, REGISTRY adheres to the principles of GCP, similar to a clinical trial, and employs data monitoring. This project aims to appraise critically 1) data quality, and 2) the quality control measures employed in REGISTRY. First we will evaluate the completeness of data entered into the data base. Second, we wish to use the data to define what is ‘plausible’. Third, we wish to examine the effect of monitoring on the above. The outcome will help 1) define what is good quality data 2) improve the monitoring process.
Author: Michael Orth
Approved: April 2010
Defining the phenotype of Huntington’s disease in Europe: The European Huntington’s disease Network Registry Study
Registry aims to study the natural history and clinical care of individuals affected by and at risk of developing Huntington’s disease. The establishment of this cohort will facilitate trials of putative HD treatments and will enable meaningful analysis of both major topics requiring large absolute sample sizes, and topics requiring smaller numbers of patients from sub-groups of interest across centres. The organisation of Registry together with its emphasis on the collection of high quality of data has never been done in this way. Therefore, the focus of our analyses will be to describe the main characteristics of the population sample and the type and quality of data collection. In summary, this report will aim to provide an overview of the scale, scope and potential of Registry.
Author: European Huntington’s Disease Network
Approved: June 2008
Publications
Publications co-authored by EHDN members:
2024
- Afshari M, Hernandez AV, Joyce JM, Hauptschein AW, Trenkle KL, Stebbins GT, Goetz CG. A Novel Home-Based Telerehabilitation Program Targeting Fall Prevention in Parkinson Disease: A Preliminary Trial. Neurol Clin Pract. 2024 Feb;14(1):e200246. doi: 10.1212/CPJ.0000000000200246. Epub 2024 Jan 5. PMID: 38213401; PMCID: PMC10781563 (available on 2025-02-01)
- Ahmad S, Imtiaz MA, Mishra A, Wang R, Herrera-Rivero M, Bis JC, Fornage M, Roshchupkin G, Hofer E, Logue M, Longstreth WT Jr, Xia R, Bouteloup V, Mosley T, Launer LJ, Khalil M, Kuhle J, Rissman RA, Chene G, Dufouil C, Djoussé L, Lyons MJ, Mukamal KJ, Kremen WS, Franz CE, Schmidt R, Debette S, Breteler MMB, Berger K, Yang Q, Seshadri S, Aziz NA, Ghanbari M, Ikram MA. Genome-wide association study meta-analysis of neurofilament light (NfL) levels in blood reveals novel loci related to neurodegeneration. Commun Biol. 2024 Sep 9;7(1):1103. doi: 10.1038/s42003-024-06804-3. PMID: 39251807; PMCID: PMC11385583.
- Aiello EN, Solca F, Torre S, Lafronza A, Maranzano A, Bonetti R, Scheveger F, Maffi S, Ceccarelli C, Scocchia M, Casella M, Verde F, Migliore S, Silani V, Ticozzi N, Squitieri F, Ciammola A, Poletti B. Validity, diagnostics and feasibility of the Italian version of the Montreal Cognitive Assessment (MoCA) in Huntington’s disease. Neurol Sci. 2024 Mar;45(3):1079-1086. doi: 10.1007/s10072-023-07070-7. Epub 2023 Sep 28. PMID: 37770762.
- Aldous SG, Smith EJ, Landles C, Osborne GF, Cañibano-Pico M, Nita IM, Phillips J, Zhang Y, Jin B, Hirst MB, Benn CL, Bond BC, Edelmann W, Greene JR, Bates GP. A CAG repeat threshold for therapeutics targeting somatic instability in Huntington’s disease. Brain. 2024 February 22. doi: 10.1093/brain/awae063. PMID: 38387080.
- Anderson KE, Arbatti L, Hosamath A, Feigin A, Goldstein J, Kayson E, Kinsler BL, Falanga L, Denise L, Carlozzi NE, Frank S, Jackson K, Kostyk S, Purks JL, Serbin KP, Kinel S, Beck CA, Shoulson I. What Huntington’s Disease Patients Say About Their Illness: An Online Direct-to-Participant Pilot Study. J Huntingtons Dis. 2024 Apr 30. doi: 10.3233/JHD-231520. Epub ahead of print. PMID: 38701155.
- Andresen K, Cutting E, Apostolopoulos D, Evans AH, Oakley L, Dayimu A, Demiris N, Bongaerts K, Staples R, Gooding W, Rubinsztein D, Barker RA. Trial to assess the tolerability of using felodipine to upregulate autophagy as a treatment of Huntington’s disease (FELL-HD): a phase II, single-centre, open-label, dose-finding trial protocol. BMJ Open. 2024 Aug 21;14(8):e087983. doi: 10.1136/bmjopen-2024-087983. PMID: 39174070; PMCID: PMC11340714.
- Atkins KJ, Andrews SC, Stout JC, Chong TT. The effect of Huntington’s disease on cognitive and physical motivation. Brain. 2024 Jan 24:awae023. doi: 10.1093/brain/awae023. Epub ahead of print. PMID: 38266149; PMCID: PMC11224606.
- Bagherpoor Helabad M, Matlahov I, Kumar R, Daldrop JO, Jain G, Weingarth M, van der Wel PCA, Miettinen MS. Integrative determination of the atomic structure of mutant huntingtin exon 1 fibrils implicated in Huntington’s disease. bioRxiv [Preprint]. 2024 Sep 15:2023.07.21.549993. doi: 10.1101/2023.07.21.549993. PMID: 37502911; PMCID: PMC10370190.
- Bahat A, Itzhaki E, Weiss B, Tolmasov M, Tsoory M, Kuperman Y, Brandis A, Shurrush KA, Dikstein R. Lowering mutant huntingtin by small molecules relieves Huntington’s disease symptoms and progression. EMBO Mol Med. 2024 Mar;16(3):523-546. doi: 10.1038/s44321-023-00020-y. Epub 2024 Feb 19. PMID: 38374466; PCMID: PMC10940305.
- Bartl S, Xie Y, Potluri N, Kesineni R, Hencak K, Cengio LD, Balazs K, Oueslati A, Parth M, Salhat N, Siddu A, Smrzka O, Cicchetti F, Straffler G, Hayden MR, Southwell AL. Reducing huntingtin by immunotherapy delays disease progression in a mouse model of Huntington disease. Neurobiol Dis. 2024 Jan;190:106376. doi: 10.1016/j.nbd.2023.106376. Epub 2023 Dec 12. Erratum in: Neurobiol Dis. 2024 Apr;193:106444. PMID: 38092268; PMCID: PMC10940305.
- Bartl S, Xie Y, Potluri N, Kesineni R, Hencak K, Cengio LD, Balazs K, Oueslati A, Parth M, Salhat N, Siddu A, Smrzka O, Cicchetti F, Staffler G, Hayden MR, Southwell AL. Corrigendum to „Reducing huntingtin by immunotherapy delays disease progression in a mouse model of Huntington disease” [Neurobiology of Disease, 2024 Jan:190:106376]. Neurobiol Dis. 2024 Feb 23:106444. doi: 10.1016/j.nbd.2024.106444. Epub ahead of print. Erratum for: Neurobiol Dis. 2024 Jan;190:106376. PMID: 38402018.
- Bilal H, Harding IH, Stout JC. The relationship between disease-specific psychosocial stressors and depressive symptoms in Huntington’s disease. J Neurol. 2024 Jan;271(1):289-299. doi: 10.1007/s00415-023-11982-x. Epub 2023 Sep 11. PMID: 37695532; PMCID: PMC10769991.
- Bilal H, McDonald SJ, Stout JC, Harding IH. Associations of inflammatory cytokines and cortisol with nonmotor features of Huntington’s disease. Ann Clin Transl Neurol. 2024 Feb 14. doi: 10.1002/acn3.52016. Epub ahead of print. PMID: 38356101.
- Bocoum A, Ouologuem M, Cissé L, Essop F, Dit Papa Coulibaly S, Botha N, Cissé CAK, Dit Baneye Maiga A, Krause A, Landouré G; H3Africa consortium. The First Case of Huntington’s Disease like 2 in Mali, West Africa. Tremor Other Hyperkinet Mov (N Y). 2024 Apr 2;14:15. doi: 10.5334/tohm.859. PMID: 38617831; PMCID:PMC11011944.
- Bodai L, Borosta R, Ferencz Á, Kovács M, Zsindely N. The Role of miR-137 in Neurodegenerative Disorders. Int J Mol Sci. 2024 Jun 30;25(13):7229. doi: 10.3390/ijms25137229. PMID: 39000336; PMCID: PMC11241563.
- Bondulich MK, Phillips J, Cañibano-Pico M, Nita IM, Byrne LM, Wild EJ, Bates GP. Translatable plasma and CSF biomarkers for use in mouse models of Huntington’s disease. Brain Commun. 2024 February 7. 6 (1) :fcae030. doi: 10.1093/braincomms/fcae030; PMID: 38370446; PMCID: PMC10873584.
- Bonsor M, Ammar O, Schnoegl S, Wanker EE, Silva Ramos E. Polyglutamine disease proteins: Commonalities and differences in interaction profiles and pathological effects. Proteomics. 2024 Jun;24(12-13):e2300114. doi: 10.1002/pmic.202300114. Epub 2024 Apr 14. PMID: 38615323.
- Brady ST, Mesnard-Hoaglin NA, Mays S, Priego M, Dziechciowska J, Morris S, Kang M, Tsai MY, Purks JL, Klein A, Gaona A, Melloni A, Connors T, Hyman B, Song Y, Morfini GA. Toxic effects of mutant huntingtin in axons are mediated by its proline-rich domain. Brain. 2024 Jun 3;147(6):2098-2113. doi: 10.1093/brain/awad280. PMID: 37633260; PMCID: PMC11146425 (available on 2024-08-26).
- Burtscher J, Strasser B, Pepe G, Burtscher M, Kopp M, Di Pardo A, Maglione V, Khamoui AV. Brain-Periphery Interactions in Huntington’s Disease: Mediators and Lifestyle Interventions. Int J Mol Sci. 2024 Apr 25;25(9):4696. doi: 10.3390/ijms25094696. PMID: 38731912; PMCID: PMC11083237.
- Camerino I, Ferreira J, Vonk JM, Kessels RPC, de Leeuw FE, Roelofs A, Copland D, Piai V. Systematic Review and Meta-Analyses of Word Production Abilities in Dysfunction of the Basal Ganglia: Stroke, Small Vessel Disease, Parkinson’s Disease, and Huntington’s Disease. Neuropsychol Rev. 2024 Mar;34(1):1-26. doi: 10.1007/s11065-022-09570-3. Epub 2022 Dec 24. PMID: 36564612.
- Cano-Cano F, Martín-Loro F, Gallardo-Orihuela A, González-Montelongo MDC, Ortuño-Miquel S, Hervás-Corpión I, de la Villa P, Ramón-Marco L, Navarro-Calvo J, Gómez-Jaramillo L, Arroba AI, Valor LM. Retinal dysfunction in Huntington’s disease mouse models concurs with local gliosis and microglia activation. Sci Rep. 2024 Feb 20;14(1):4176. doi: 10.1038/s41598-024-54347-8. PMID: 38378796; PMCID: PMC10879138.
- Cao LX, Yin JH, Du G, Yang Q, Huang Y. Identifying and verifying Huntington’s disease subtypes: Clinical features, neuroimaging, and cytokine changes. Brain Behav. 2024 Mar;14(3):e3469. doi: 10.1002/brb3.3469. PMID: 38494708; PMCID: PMC10945031.
- Caron NS, Aly AE, Findlay Black H, Martin DDO, Schmidt ME, Ko S, Anderson C, Harvey EM, Casal LL, Anderson LM, Rahavi SMR, Reid GSD, Oda MN, Stanimirovic D, Abulrob A, McBride JL, Leavitt BR, Hayden MR. Systemic delivery of mutant huntingtin lowering antisense oligonucleotides to the brain using apolipoprotein A-I nanodisks for Huntington disease. J Control Release. 2024 Jan 24;367:27-44. doi: 10.1016/j.jconrel.2024.01.011. Epub ahead of print. PMID: 38215984.
- Cattaneo E, Barker RA. Brain cholesterol therapy for Huntington’s disease – Does it make sense? Clin Transl Med. 2024 Jul;14(7):e1746. doi: 10.1002/ctm2.1746. PMID: 38924677; PMCID: PMC11199055.
- Chang YC, Tsai YC, Chang EC, Hsu YC, Huang YR, Lee YH, Tsai YS, Chen YQ, Lee YC, Liao YC, Kuo JC, Su MT, Yang UC, Chern Y, Cheng TH. PIAS1 S510G variant acts as a genetic modifier of spinocerebellar ataxia type 3 by selectively impairing mutant ataxin-3 proteostasis. Int J Biochem Cell Biol. 2024 Sep 16;176:106662. doi: 10.1016/j.biocel.2024.106662. Epub ahead of print. PMID: 39293559.
- Chen S, Zhang H, Yu J, Cao X, Zhang S, Dong D. Sex-Specific Differences in the Progression of Huntington’s Disease Symptoms: A National Study in China. Neuroepidemiology. 2024 May 30. doi: 10.1159/000539131. Epub ahead of print. PMID: 38815560.
- Chenain L, Riad R, Fraisse N, Jubin C, Morgado G, Youssov K, Lunven M, Bachoud-Levi AC. Graph methods to infer spatial disturbances: Application to Huntington’s Disease’s speech. Cortex. 2024 Jul;176:144-160. doi: 10.1016/j.cortex.2024.04.014. Epub 2024 May 17. PMID: 38795650.
- Choi DE, Shin JW, Zeng S, Hong EP, Jang JH, Loupe JM, Wheeler VC, Stutzman HE, Kleinstiver B, Lee JM. Base editing strategies to convert CAG to CAA diminish the disease-causing mutation in Huntington’s disease. Elife. 2024 Jun 13;12:RP89782. doi: 10.7554/eLife.89782. PMID: 38869243; PMCID: PMC11175616.
- Coleman A, Langan MT, Verma G, Knights H, Sturrock A, Leavitt BR, Tabrizi SJ, Scahill RI, Hobbs NZ. Assessment of Perivascular Space Morphometry Across the White Matter in Huntington’s Disease Using MRI. J Huntingtons Dis. 2024;13(1):91-101. doi: 10.3233/JHD-231508. PMID: 38517798.
- Considine CM, Rossetti MA, Anderson K, Del Bene VA, Anderson SA, Celka AS, Edmondson MC, Sheese ALN, Piccolino A, Teixeira AL, Stout JC. Huntington study group’s neuropsychology working group position on best practice recommendations for the clinical neuropsychological evaluation of patients with Huntington disease. Clin Neuropsychol. 2024 May;38(4):984-1006. doi: 10.1080/13854046.2023.2267789. Epub 2023 Oct 18. PMID: 37849335.
- Cooper H, Simpson J, Dale M, Eccles FJR. Experiences of young people growing up in a family with Huntington’s disease: A meta-ethnography of qualitative research. J Genet Couns. 2024 Mar 12. doi: 10.1002/jgc4.1886. Epub ahead of print. PMID: 38469914.
- Cooper H, Simpson J, Dale M, Eccles FJR. Maintaining psychological well-being when living at risk of Huntington’s disease: An interpretative phenomenological analysis. J Genet Couns. 2024 Sep 9. doi: 10.1002/jgc4.1965. Epub ahead of print. PMID: 39252438.
- Czech MD, Badley D, Yang L, Shen J, Crouthamel M, Kangarloo T, Dorsey ER, Adams JL, Cosman JD. Improved measurement of disease progression in people living with early Parkinson’s disease using digital health technologies. Commun Med (Lond). 2024 Mar 15;4(1):49. doi: 10.1038/s43856-024-00481-3. PMID: 38491176; PMCID: PMC10942994.
- Dalene Skarping K, Arning L, Petersén Å, Nguyen HP, Gebre-Medhin S. Attenuated huntingtin gene CAG nucleotide repeat size in individuals with Lynch syndrome. Sci Rep. 2024 Feb 21;14(1):4300. doi: 10.1038/s41598-024-54277-5. PMID: 38383663; PMCID: PMC10881568.
- Dam T, Pagano G, Brumm MC, Gochanour C, Poston KL, Weintraub D, Chahine LM, Coffey C, Tanner CM, Kopil CM, Xiao Y, Chowdhury S, Concha-Marambio L, DiBiaso P, Foroud T, Frasier M, Jennings D, Kieburtz K, Merchant K, Mollenhauer B, Montine TJ, Nudelman K, Seibyl J, Sherer T, Singleton A, Stephenson D, Stern M, Soto C, Tolosa E, Siderowf A, Dunn B, Simuni T, Marek K; Parkinson’s Progression Markers Initiative. Neuronal alpha-Synuclein Disease Integrated Staging System performance in PPMI, PASADENA, and SPARK baseline cohorts. medRxiv [Preprint]. 2024 Sep 10:2024.02.14.24302818. doi: 10.1101/2024.02.14.24302818. Update in: NPJ Parkinsons Dis. 2024 Sep 27;10(1):178. doi: 10.1038/s41531-024-00789-w. PMID: 39314957; PMCID: PMC11419206.
- Dawson J, Kay C, Black HF, Bortnick S, Javier K, Xia Q, Sandhu A, Buchanan C, Hogg V, Chang FCF, Goto J, Arning L, Saft C, Bijlsma EK, Nguyen HP, Roxburgh R, Hayden MR. The frequency and clinical impact of synonymous HTT loss-of-interruption and duplication-of-interruption variants in a diverse HD cohort. Genet Med. 2024 Aug 10;26(11):101239. doi: 10.1016/j.gim.2024.101239. Epub ahead of print. PMID: 39140258.
- Delussi M, Valt C, Silvestri A, Ricci K, Ladisa E, Ammendola E, Rampino A, Pergola G, de Tommaso M. Auditory mismatch negativity in pre-manifest and manifest Huntington’s disease. Clin Neurophysiol. 2024 Jun;162:121-128. doi: 10.1016/j.clinph.2024.03.020. Epub 2024 Mar 26. PMID: 38603947.
- Diaz Escarcega R, Murambadoro K, Valencia R, Moruno-Manchon JF, Furr Stimming EE, Jung SY, Tsvetkov AS. Sphingosine kinase 2 regulates protein ubiquitination networks in neurons. Mol Cell Neurosci. 2024 Sep;130:103948. doi: 10.1016/j.mcn.2024.103948. Epub 2024 Jun 21. PMID: 38909878.
- Dickmann CGF, Milicevic Sephton S, Barker RA, Aigbirhio FI. PET Ligands for Imaging Mutant Huntingtin Aggregates: A Case Study in Non-For-Profit Scientific Management. Chembiochem. 2024 Jun 3;25(11):e202400152. doi: 10.1002/cbic.202400152. Epub 2024 May 2. PMID: 38695673.
- Dieter M, Kevin P, Tobias V, Andreas H, Lorenz N, Kathrin K, Nikolaus K, Juergen B, Jan R, Adrian D. Polysomnographic findings in the ultra-rare McLeod syndrome: further documentation of sleep apnea as a possible feature. J Clin Sleep Med. 2024 Mar 1;20(3):339-344. doi: 10.5664/jcsm.10854. PMID: 37811906.
- Doheny EP, Renerts K, Braun A, Werth E, Baumann C, Baumgartner P, Morgan-Jones P, Busse M, Lowery MM, Jung HH. Assessment of Fitbit Charge 4 for sleep stage and heart rate monitoring against polysomnography and during home monitoring in Huntington’s disease. J Clin Sleep Med. 2024 Mar 7. doi: 10.5664/jcsm.11098. Epub ahead of print. PMID: 38450553.
- Driscoll R, Hampton L, Abraham NA, Larigan JD, Joseph NF, Hernandez-Vega JC, Geisler S, Yang FC, Deninger M, Tran DT, Khatri N, Godinho BMDC, Kinberger GA, Montagna DR, Hirst WD, Guardado CL, Glajch KE, Arnold HM, Gallant-Behm CL, Weihofen A. Dose-dependent reduction of somatic expansions but not Htt aggregates by di-valent siRNA-mediated silencing of MSH3 in HdhQ111 mice. Sci Rep. 2024 Jan 24;14(1):2061. doi: 10.1038/s41598-024-52667-3. PMID: 38267530; PMCID: PMC10808119.
- Eaton JE, Claassen DO. Guidance on antipsychotic selection for agitation and aggressive behavior in persons with Huntington’s disease. Expert Rev Neurother. 2024 Oct;24(10):937-940. doi: 10.1080/14737175.2024.2376836. Epub 2024 Jul 9. PMID: 38982803.
- Ekwudo MN, Gubert C, Hannan AJ. The microbiota-gut-brain axis in Huntington’s disease: pathogenic mechanisms and therapeutic targets. FEBS J. 2024 Mar 1. doi: 10.1111/febs.17102. Epub ahead of print. PMID: 38426291.
- En-Hua Wang J, Simon NG, Brownstein MJ, Maibach HT, Maibach J, Anderson KE. The utility of the irritability scale in Huntington’s disease patients with evidence of irritability or aggression. Parkinsonism Relat Disord. 2024 Jun;123:106087. doi: 10.1016/j.parkreldis.2024.106087. Epub 2024 Mar 30. PMID: 38640832.
- Escudero-Cabarcas J, Pineda-Alhucema W, Martinez-Banfi M, Acosta-López JE, Cervantes-Henriquez ML, Mejía-Segura E, Jiménez-Figueroa G, Sánchez-Barros C, Puentes-Rozo PJ, Noguera-Machacón LM, Ahmad M, de la Hoz M, Vélez JI, Arcos-Burgos M, Pineda DA, Sánchez M. Theory of Mind in Huntington’s Disease: A Systematic Review of 20 Years of Research. J Huntingtons Dis. 2024;13(1):15-31. doi: 10.3233/JHD-230594. PMID: 38517797.
- Estevez-Fraga C, Tabrizi SJ, Wild EJ. Huntington’s Disease Clinical Trials Corner: March 2024. J Huntingtons Dis. 2024;13(1):1-14. doi: 10.3233/JHD-240017. PMID: 38489195; PMCID: PMC11091610.
- Evans TE, Vilor-Tejedor N, Operto G, Falcon C, Hofman A, Ibáñez A, Seshadari S, Tan LC, Weiner M, Alladi S, Anazodo U, Gispert Lopez JD; Alzheimer’s Disease Neuroimaging Initiative; Australian Imaging Biomarkers and Lifestyle flagship study of ageing; Adams HHH. Structural brain differences in the Alzheimer’s disease continuum: Insights into the heterogeneity from a large multi-site neuroimaging consortium. Biol Psychiatry Cogn Neurosci Neuroimaging. 2024 Jul 29:S2451-9022(24)00207-6. doi: 10.1016/j.bpsc.2024.07.019. Epub ahead of print. PMID: 39084525.
- Fahed VS, Doheny EP, Collazo C, Krzysztofik J, Mann E, Morgan-Jones P, Mills L, Drew C, Rosser AE, Cousins R, Witkowski G, Cubo E, Busse M, Lowery MM. Language-Independent Acoustic Biomarkers for Quantifying Speech Impairment in Huntington’s Disease. Am J Speech Lang Pathol. 2024 May;33(3):1390-1405. doi: 10.1044/2024_AJSLP-23-00175. Epub 2024 Mar 26. PMID: 38530396.
- Farag M, Coleman A, Knights H, Murphy MJ, Rajagopal S, Touzé A, Shoai M, Hearst C, Salanio DM, Wild EJ, Tabrizi SJ. Outcomes of Percutaneous Endoscopic Gastrostomy in Huntington’s Disease at a Tertiary Center. Mov Disord Clin Pract. 2024 Jun 9. doi: 10.1002/mdc3.14130. Epub ahead of print. PMID: 38853375.
- Feigin A, Evans EE, Fisher TL, Leonard JE, Smith ES, Reader A, Mishra V, Manber R, Walters KA, Kowarski L, Oakes D, Siemers E, Kieburtz KD, Zauderer M, Huntington Study Group SIGNAL investigators. Publisher Correction: Pepinemab antibody blockade of SEMA4D in early Huntington’s disease: a randomized, placebo-controlled, phase 2 trial. Nat Med. 2024 Feb;30(2):606. doi: 10.1038/s41591-022-02070-0. Erratum for: Nat Med. 2022 Oct;28(10):2183-2193. PMID: 36195687; PMCID: PMC10878960.
- Ferguson R, Goold R, Coupland L, Flower M, Tabrizi SJ. Therapeutic validation of MMR-associated genetic modifiers in a human ex vivo model of Huntington disease. Am J Hum Genet. 2024 Jun 6;111(6):1165-1183. doi: 10.1016/j.ajhg.2024.04.015. Epub 2024 May 14. PMID: 38749429; PMCID: PMC11179424.
- Festa BP, Siddiqi FH, Jimenez-Sanchez M, Rubinsztein DC. Microglial cytokines poison neuronal autophagy via CCR5, a druggable target. Autophagy. 2024 Apr;20(4):949-951. doi: 10.1080/15548627.2023.2221921. Epub 2023 Jun 26. PMID: 37358357; PMCID: PMC11062376.
- Field SE, Curle AJ, Barker RA. Inflammation and Huntington’s disease – a neglected therapeutic target? Expert Opin Investig Drugs. 2024 May;33(5):451-467. doi: 10.1080/13543784.2024.2348738. Epub 2024 May 29. PMID: 38758356.
- Fitzgerald ES, Manousakis JE, Glikmann-Johnston Y, Rankin M, Anderson C, Stout JC, Jackson ML. Sleep fragmentation despite intact rest-activity patterns in premanifest Huntington’s disease: An actigraphy study. Sleep Med. 2024 Aug 31;124:16-29. doi: 10.1016/j.sleep.2024.08.026. Epub ahead of print. PMID: 39250876.
- Fote GM, Eapen VV, Lim RG, Yu C, Salazar L, McClure NR, McKnight J, Nguyen TB, Heath MC, Lau AL, Villamil MA, Miramontes R, Kratter IH, Finkbeiner S, Reidling JC, Paulo JA, Kaiser P, Huang L, Housman DE, Thompson LM, Steffan JS. Huntingtin contains an ubiquitin-binding domain and regulates lysosomal targeting of mitochondrial and RNA-binding proteins. Proc Natl Acad Sci U S A. 2024 Aug 6;121(32):e2319091121. doi: 10.1073/pnas.2319091121. Epub 2024 Jul 29. PMID: 39074279; PMCID: PMC11317567.
- Frank S, Anderson KE, Fernandez HH, Hauser RA, Claassen DO, Stamler D, Factor SA, Jimenez-Shahed J, Barkay H, Wilhelm A, Alexander JK, Chaijale N, Barash S, Savola JM, Gordon MF, Chen M. Safety of Deutetrabenazine for the Treatment of Tardive Dyskinesia and Chorea Associated with Huntington Disease. Neurol Ther. 2024 Jun;13(3):655-675. doi: 10.1007/s40120-024-00600-1. Epub 2024 Apr 1. PMID: 38557959; PMCID: PMC11136929.
- Frank S, Anderson KE, Fernandez HH, Hauser RA, Claassen DO, Stamler D, Factor SA, Jimenez-Shahed J, Barkay H, Wilhelm A, Alexander JK, Chaijale N, Barash S, Savola JM, Gordon MF, Chen M. Correction: Safety of Deutetrabenazine for the Treatment of Tardive Dyskinesia and Chorea Associated with Huntington Disease. Neurol Ther. 2024 Sep 12. doi: 10.1007/s40120-024-00660-3. Epub ahead of print. Erratum for: Neurol Ther. 2024 Jun;13(3):655-675. doi: 10.1007/s40120-024-00600-1. PMID: 39266813; PMCID: PMC11541967.
- Franklin GL. My first dance. Postgrad Med J. 2024 Aug 16;100(1187):699-700. doi: 10.1093/postmj/qgad142. PMID: 38244551.
- Franklin GL, Teive HAG, Tensini FS, Camargo CHF, de Lima NSC, de Dos Santos DC, Meira AT, Tabrizi SJ. The Huntington’s Disease Gene Discovery. Mov Disord. 2024 Feb;39(2):227-234. doi: 10.1002/mds.29703. Epub 2024 Jan 5. PMID: 38179605.
- Fusco FR, Paldino E. Is GDNF to Parkinson’s disease what BDNF is to Huntington’s disease? Neural Regen Res. 2024 May;19(5):973-974. doi: 10.4103/1673-5374.385305. PMID: 37862194; PMCID: PMC10749623.
- Gaastra A, van Duijn E, Dols A. Is maintenance therapy warranted for recurrent mania in a woman with a positive family history of Huntington’s disease? Bipolar Disord. 2024 May;26(3):300-302. doi: 10.1111/bdi.13420. Epub 2024 Mar 14. PMID: 38485447.
- Galimberti M, Nucera MR, Bocchi VD, Conforti P, Vezzoli E, Cereda M, Maffezzini C, Iennaco R, Scolz A, Falqui A, Cordiglieri C, Cremona M, Espuny-Camacho I, Faedo A, Felsenfeld DP, Vogt TF, Ranzani V, Zuccato C, Besusso D, Cattaneo E. Huntington’s disease cellular phenotypes are rescued non-cell autonomously by healthy cells in mosaic telencephalic organoids. Nat Commun. 2024 Aug 2;15(1):6534. doi: 10.1038/s41467-024-50877-x. PMID: 39095390; PMCID: PMC11297310.
- Gao L, Bhattacharyya A, Beers B, Kaushik D, Bredlau AL, Kristensen A, Abd-Elaziz K, Grant R, Golden L, Kong R. Pharmacokinetics and pharmacodynamics of PTC518, an oral huntingtin lowering splicing modifier: A first-in-human study. Br J Clin Pharmacol. 2024 Aug 18. doi: 10.1111/bcp.16202. Epub ahead of print. PMID: 39155237.
- Geijtenbeek KW, Aranda AS, Sanz AS, Janzen J, Bury AE, Kors S, Al Amery N, Schmitz NCM, Reits EAJ, Schipper-Krom S. Insulin-Degrading Enzyme Efficiently Degrades polyQ Peptides but not Expanded polyQ Huntingtin Fragments. J Huntingtons Dis. 2024 Apr 16. doi: 10.3233/JHD-230583. Epub ahead of print. PMID: 38640164.
- Genetic Modifiers of Huntington’s Disease (GeM-HD) Consortium; Lee JM, McLean ZL, Correia K, Shin JW, Lee S, Jang JH, Lee Y, Kim KH, Choi DE, Long JD, Lucente D, Seong IS, Pinto RM, Giordano JV, Mysore JS, Siciliano J, Elezi E, Ruliera J, Gillis T, Wheeler VC, MacDonald ME, Gusella JF, Gatseva A, Ciosi M, Lomeikaite V, Loay H, Monckton DG, Wills C, Massey TH, Jones L, Holmans P, Kwak S, Sampaio C, Orth M, Bernhard Landwehrmeyer G, Paulsen JS, Ray Dorsey E, Myers RH. Genetic modifiers of somatic expansion and clinical phenotypes in Huntington’s disease reveal shared and tissue-specific effects. bioRxiv [Preprint]. 2024 Jun 18:2024.06.10.597797. doi: 10.1101/2024.06.10.597797. PMID: 38948755; PMCID: PMC11212857.
- Gentenaar M, Meulmeester FL, van der Burg XR, Hoekstra AT, Hunt H, Kroon J, van Roon-Mom WMC, Meijer OC. Glucocorticoid receptor antagonist CORT113176 attenuates motor and neuropathological symptoms of Huntington’s disease in R6/2 mice. Exp Neurol. 2024 Apr;374:114675. doi: 10.1016/j.expneurol.2024.114675. Epub 2024 Jan 10. PMID: 38216109.
- Gibbons CH, Levine T, Adler C, Bellaire B, Wang N, Stohl J, Agarwal P, Aldridge GM, Barboi A, Evidente VGH, Galasko D, Geschwind MD, Gonzalez-Duarte A, Gil R, Gudesblatt M, Isaacson SH, Kaufmann H, Khemani P, Kumar R, Lamotte G, Liu AJ, McFarland NR, Miglis M, Reynolds A, Sahagian GA, Saint-Hillaire MH, Schwartzbard JB, Singer W, Soileau MJ, Vernino S, Yerstein O, Freeman R. Skin Biopsy Detection of Phosphorylated α-Synuclein in Patients With Synucleinopathies. JAMA. 2024 Apr 16;331(15):1298-1306. doi: 10.1001/jama.2024.0792. PMID: 38506839; PMCID: PMC10955354 (available on 2024-09-20).
- Gijs M, Jorna N, Datson N, Beekman C, Dansokho C, Weiss A, Linden DEJ, Oosterloo M. High levels of mutant huntingtin protein in tear fluid from Huntington’s Disease Gene Expansion Carriers. J Mov Disord. 2024 Feb 21. doi: 10.14802/jmd.24014. Epub ahead of print. PMID: 38379425.
- Gil-Salcedo A, Massart R, de Langavant LC, Bachoud-Levi AC. Modifiable factors associated with Huntington’s disease progression in presymptomatic participants. Ann Clin Transl Neurol. 2024 Jun 10. doi: 10.1002/acn3.52120. Epub ahead of print. PMID: 38855890.
- Gonawala L, Wijekoon N, Attanayake D, Ratnayake P, Sirisena D, Gunasekara H, Dissanayake A, Keshavaraj A, Mohan C, Steinbusch HWM, Hoffman EP, Dalal A, de Silva KRD. Diagnostic outcome of pro bono neurogenetic diagnostic service in Sri Lanka: A wealth creation. Eur J Hum Genet. 2024 Jan 23. doi: 10.1038/s41431-023-01525-3. Epub ahead of print. PMID: 38253783.
- Grabska N, Wójcik-Pędziwiatr M, Sławek J, Sołtan W, Gawryluk J, Rudziński M, Szczudlik A, Rudzińska-Bar M. Reflexive and voluntary saccadic eye movements as biomarker of Huntington’s Disease. Neurol Neurochir Pol. 2024 May 31. doi: 10.5603/pjnns.95190. Epub ahead of print. PMID: 38818957.
- Greguletz P, Plötz M, Baade-Büttner C, Bien CG, Eisenhut K, Geis C, Handreka R, Klausewitz J, Körtvelyessy P, Kovac S, Kraft A, Lewerenz J, Malter M, Nagel M, von Podewils F, Prüß H, Rada A, Rau J, Rauer S, Rößling R, Seifert-Held T, Siebenbrodt K, Sühs KW, Tauber SC, Thaler F, Wagner J, Wickel J, Leypoldt F, Rittner HL, Sommer C, Villmann C, Doppler K; GENERATE study group. Different pain phenotypes are associated with anti-Caspr2 autoantibodies. J Neurol. 2024 May;271(5):2736-2744. doi: 10.1007/s00415-024-12224-4. Epub 2024 Feb 22. PMID: 38386048; PMCID: PMC11055745.
- Gu Y, Pope A, Smith C, Carmona C, Johnstone A, Shi L, Chen X, Santos S, Bacon-Brenes CC, Shoff T, Kleczko KM, Frydman J, Thompson LM, Mobley WC, Wu C. BDNF and TRiC-inspired reagent rescue cortical synaptic deficits in a mouse model of Huntington’s disease. Neurobiol Dis. 2024 Jun 1;195:106502. doi: 10.1016/j.nbd.2024.106502. Epub 2024 Apr 10. PMID: 38608784.
- Gubert C, Kong G, Costello C, Adams CD, Masson BA, Qin W, Choo J, Narayana VK, Rogers G, Renoir T, Furness JB, Hannan AJ. Dietary fibre confers therapeutic effects in a preclinical model of Huntington’s disease. Brain Behav Immun. 2024 Feb;116:404-418. doi: 10.1016/j.bbi.2023.12.023. Epub 2023 Dec 23. PMID: 38142919.
- Guilliot S, Wilson EN, Touchon J, Soto ME. Nanolithium, a New Treatment Approach to Alzheimer’s Disease: A Review of Existing Evidence and Clinical Perspectives. J Prev Alzheimers Dis. 2024;11(2):428-434. doi: 10.14283/jpad.2024.26. PMID: 38374749.
- Guzauskas GF, Tabrizi SJ, Long JD, Arnesen A, Hamilton JL, Claassen DO, Munetsi LR, Malik S, Rodríguez-Santana I, Ali TM, Zhang F. Long-Term Health Outcomes of Huntington Disease and the Impact of Future Disease-Modifying Treatments: A Decision-Modeling Analysis. Neurol Clin Pract. 2024 Oct;14(5):e200340. doi: 10.1212/CPJ.0000000000200340. Epub 2024 Aug 15. PMID: 39161748; PMCID: PMC11332983.
- Handley RR, Reid SJ, Burch Z, Jacobsen JC, Gillis T, Correia K, Rudiger SR, McLaughlin CJ, Bawden CS, MacDonald ME, Wheeler VC, Snell RG. Somatic CAG repeat stability in a transgenic sheep model of Huntington’s disease. J Huntingtons Dis. 2024 February 18. doi: 10.3233/JHD-231516. PMID: 38393920.
- Hannan AJ. Repeating themes of plastic genes and therapeutic schemes targeting the ‚tandem repeatome’. Brain Commun. 2024 Feb 19;6(2):fcae047. doi: 10.1093/braincomms/fcae047. PMID: 38449715; PMCID: PMC10917440.
- Haupeltshofer S, Mencl S, Szepanowski RD, Hansmann C, Casas AI, Abberger H, Hansen W, Blusch A, Deuschl C, Forsting M, Hermann DM, Langhauser F, Kleinschnitz C. Delayed plasma kallikrein inhibition fosters post-stroke recovery by reducing thrombo-inflammation. J Neuroinflammation. 2024 Jun 13;21(1):155. doi: 10.1186/s12974-024-03149-w. PMID: 38872149; PMCID: PMC11177352.
- Heim B, Mandler E, Buchmann A, Grossauer A, Peball M, Valent D, Carbone F, Schwarzová K, Djamshidian A, Mahlknecht P, Khalil M, Krismer F, Seppi K. Serum Neurofilament Light and Clinical Biomarkers for Disease Staging in Huntington’s Disease. Mov Disord Clin Pract. 2024 Mar 27. doi: 10.1002/mdc3.13993. Epub ahead of print. PMID: 38533634; PMCID: PMC11233851.
- Hendel RK, Hellem MNN, Larsen IU, Vinther-Jensen T, Hjermind LE, Nielsen JE, Vogel A. Impairments of social cognition significantly predict the progression of functional decline in Huntington’s disease: A 6-year follow-up study. Appl Neuropsychol Adult. 2024 Sep-Oct;31(5):777-786. doi: 10.1080/23279095.2022.2073824. Epub 2022 May 13. PMID: 35549503.
- Hermle D, Schubert R, Barallon P, Ilg W, Schüle R, Reilmann R, Synofzik M, Traschütz A. Multifeature quantitative motor assessment of upper limb ataxia including drawing and reaching. Ann Clin Transl Neurol. 2024 May;11(5):1097-1109. doi: 10.1002/acn3.52024. Epub 2024 Apr 8. PMID: 38590028; PMCID: PMC11093241.
- Hernández CA, Peikert K, Qiao M, Darras A, de Wilde JRA, Bos J, Leibowitz M, Galea I, Wagner C, Rab MAE, Walker RH, Hermann A, van Beers EJ, van Wijk R, Kaestner L. Osmotic gradient ektacytometry – a novel diagnostic approach for neuroacanthocytosis syndromes. Front Neurosci. 2024 Jul 18;18:1406969. doi: 10.3389/fnins.2024.1406969. PMID: 39091345; PMCID: PMC11292800.
- Hinchliffe C, Rehman RZU, Pinaud C, Branco D, Jackson D, Ahmaniemi T, Guerreiro T, Chatterjee M, Manyakov NV, Pandis I, Davies K, Macrae V, Aufenberg S, Paulides E, Hildesheim H, Kudelka J, Emmert K, Van Gassen G, Rochester L, van der Woude CJ, Reilmann R, Maetzler W, Ng WF, Del Din S; IDEA-FAST Consortium. Evaluation of walking activity and gait to identify physical and mental fatigue in neurodegenerative and immune disorders: preliminary insights from the IDEA-FAST feasibility study. J Neuroeng Rehabil. 2024 Jun 5;21(1):94. doi: 10.1186/s12984-024-01390-1. PMID: 38840208; PMCID: PMC11151484.
- Hobbs NZ, Papoutsi M, Delva A, Kinnunen KM, Nakajima M, Van Laere K, Vandenberghe W, Herath P, Scahill RI. Neuroimaging to Facilitate Clinical Trials in Huntington’s Disease: Current Opinion from the EHDN Imaging Working Group. J Huntingtons Dis. 2024 May 17. doi: 10.3233/JHD-240016. Epub ahead of print. PMID: 38788082.
- Hong EP, Ramos EM, Aziz NA, Massey TH, McAllister B, Lobanov S, Jones L, Holmans P, Kwak S, Orth M, Ciosi M, Lomeikaite V, Monckton DG, Long JD, Lucente D, Wheeler VC, Gillis T, MacDonald ME, Sequeiros J, Gusella JF, Lee JM. Modification of Huntington’s disease by short tandem repeats. Brain Commun. 2024 Jan 23;6(2):fcae016. doi: 10.1093/braincomms/fcae016. PMID: 38449714; PMCID: PMC10917446.
- Hopf S, Tüscher O, Schuster AK. Retinale OCT-Biomarker und neurodegenerative Erkrankungen des zentralen Nervensystems jenseits der Alzheimer-Krankheit [Retinal OCT biomarkers and neurodegenerative diseases of the central nervous system beyond Alzheimer’s disease]. Ophthalmologie. 2024 Feb;121(2):93-104. German. doi: 10.1007/s00347-023-01974-7. Epub 2024 Jan 23. PMID: 38263475.
- Hoschek F, Natan J, Wagner M, Sathasivam K, Abdelmoez A, von Einem B, Bates GP, Landwehrmeyer GB, Neueder A. Huntingtin HTT1a is generated in a CAG repeat-length-dependent manner in human tissues. Mol Med. 2024 Mar 8;30(1):36. doi: 10.1186/s10020-024-00801-2. PMID: 38459427; PMCID: PMC10924374.
- Huynh K, Georgiou-Karistianis N, Lampit A, Siddiqui MN, Stout JC, Jamadar SD. Computerized Cognitive Training Increases Gray Matter Volumes in Huntington’s Disease: A Pilot Study. Mov Disord. 2024 Aug 9. doi: 10.1002/mds.29972. Epub ahead of print. PMID: 39120126.
- Hwang YS, Jo S, Kim GH, Lee JY, Ryu HS, Oh E, Lee SH, Kim YS, Chung SJ. Clinical and Genetic Characteristics Associated With Survival Outcome in Late-Onset Huntington’s Disease in South Korea. J Clin Neurol. 2024 Apr 2. doi: 10.3988/jcn.2023.0329. Epub ahead of print. PMID: 38627228;PMCID: PMC11220345.
- Ibañez K, Jadhav B, Zanovello M, Gagliardi D, Clarkson C, Facchini S, Garg P, Martin-Trujillo A, Gies SJ, Deforie VG, Dalmia A, Hensman Moss DJ, Vandrovcova J, Rocca C, Moutsianas L, Marini-Bettolo C, Walker H, Turner C, Shoai M, Long JD; EUROSCA network; Fratta P, Langbehn DR, Tabrizi SJ, Caulfield MJ, Cortese A, Escott-Price V, Hardy J, Houlden H, Sharp AJ, Tucci A. Increased frequency of repeat expansion mutations across different populations. medRxiv [Preprint]. 2024 Jul 8:2023.07.03.23292162. doi: 10.1101/2023.07.03.23292162. PMID: 37461547; PMCID: PMC10350132.
- Ikefuama EC, Slaviero AN, Schalau R, Gott M, Tree MO, Dunbar GL, Rossignol J, Hochgeschwender U. Presymptomatic Targeted Circuit Manipulation for Ameliorating Huntington’s Disease Pathogenesis. bioRxiv [Preprint]. 2024 Jul 24:2024.07.24.604946. doi: 10.1101/2024.07.24.604946. PMID: 39091860; PMCID: PMC11291159.
- Ioakeimidis V, Busse M, Drew CJG, Pallmann P, Watson GB, Jones D, Palombo M, Schubert R, Rosser AE, Metzler-Baddeley C. Protocol for a randomised controlled unblinded feasibility trial of HD-DRUM: a rhythmic movement training application for cognitive and motor symptoms in people with Huntington’s disease. BMJ Open. 2024 Jul 31;14(7):e082161. doi: 10.1136/bmjopen-2023-082161. Erratum in: BMJ Open. 2024 Sep 20;14(9):e082161corr1. doi: 10.1136/bmjopen-2023-082161corr1. PMID: 39089721; PMCID: PMC11418498.
- Irakkam MPBD, Joseph JHM, Kandasamy M. Aberrant Hippocampal Neuroregenerative Plasticity in Schizophrenia: Reactive Neuroblastosis as a Possible Pathocellular Mechanism of Hallucination. Neurosignals. 2024 Jul 4;31(1):1-25. doi: 10.33594/000000712. PMID: 38967556.
- Isaacs DA, Hay KR, Hoadley J, McDonell KE, Brown AE, Wynn A, Claassen DO, Gibson J. Influence of anosognosia on patient-reported outcomes for psychiatric symptoms and quality of life in Huntington’s disease. Parkinsonism Relat Disord. 2024 Jun;123:106969. doi: 10.1016/j.parkreldis.2024.106969. Epub 2024 Apr 9. PMID: 38614044; PMCID: PMC11169743 (available on 2025-06-01).
- Jackson WS, Bauer S, Kaczmarczyk L, Magadi SS. Selective Vulnerability to Neurodegenerative Disease: Insights from Cell Type-Specific Translatome Studies. Biology (Basel). 2024 Jan 23;13(2):67. doi: 10.3390/biology13020067. PMID: 38392286; PMCID: PMC10886597.
- Jiang A, You L, Handley RR, Hawkins V, Reid SJ, Jacobsen JC, Patassini S, Rudiger SR, Mclaughlan CJ, Kelly JM, Verma PJ, Bawden CS, Gusella JF, MacDonald ME, Waldvogel HJ, Faull RLM, Lehnert K, Snell RG. Single nuclei RNA-seq reveals a medium spiny neuron glutamate excitotoxicity signature prior to the onset of neuronal death in an ovine Huntington’s disease model. Hum Mol Genet. 2024 May 22:ddae087. doi: 10.1093/hmg/ddae087. Epub ahead of print. PMID: 38776957; PMCID: PMC11336116.
- Jing Y, Dogan I, Reetz K, Romanzetti S. Neurochemical changes in the progression of Huntington’s disease: A meta-analysis of in vivo1H-MRS studies. Neurobiol Dis. 2024 Sep;199:106574. doi: 10.1016/j.nbd.2024.106574. Epub 2024 Jun 22. PMID: 38914172.
- Johnson S, Kantartjis M, Severson J, Dorsey R, Adams JL, Kangarloo T, Kostrzebski MA, Best A, Merickel M, Amato D, Severson B, Jezewski S, Polyak S, Keil A, Cosman J, Anderson D. Wearable Sensor-Based Assessments for Remotely Screening Early-Stage Parkinson’s Disease. Sensors (Basel). 2024 Aug 30;24(17):5637. doi: 10.3390/s24175637. PMID: 39275547; PMCID: PMC11397844.
- Joseph S, Robbins CB, Haystead A, Hemesath A, Allen A, Kundu A, Ma JP, Scott BL, Moore KPL, Agrawal R, Gunasan V, Stinnett SS, Grewal DS, Fekrat S. Characterizing differences in retinal and choroidal microvasculature and structure in individuals with Huntington’s Disease compared to healthy controls: A cross-sectional prospective study. PLoS One. 2024 Jan 30;19(1):e0296742. doi: 10.1371/journal.pone.0296742. PMID: 38289919; PMCID: PMC10826956.
- Kim KH, Hong EP, Lee Y, McLean ZL, Elezi E, Lee R, Kwak S, McAllister B, Massey TH, Lobanov S, Holmans P, Orth M, Ciosi M, Monckton DG, Long JD, Lucente D, Wheeler VC, MacDonald ME, Gusella JF, Lee JM. Posttranscriptional regulation of FAN1 by miR-124-3p at rs3512 underlies onset-delaying genetic modification in Huntington’s disease. Proc Natl Acad Sci U S A. 2024 Apr 16;121(16):e2322924121. doi: 10.1073/pnas.2322924121. Epub 2024 Apr 12. PMID: 38607933; PMCID: PMC11032436 (available on 2024-10-12).
- Kim R, Seong MW, Oh B, Shin HS, Lee JS, Park S, Jang M, Jeon B, Kim HJ, Lee JY. Analysis of HTT CAG repeat expansion among healthy individuals and patients with chorea in Korea. Parkinsonism Relat Disord. 2024 Jan;118:105930. doi: 10.1016/j.parkreldis.2023.105930. Epub 2023 Nov 21. PMID: 37992538.
- Knights H, Coleman A, Hobbs NZ, Tabrizi SJ, Scahill RI; HD-YAS investigators. Freesurfer Software Update Significantly Impacts Striatal Volumes in the Huntington’s Disease Young Adult Study and Will Influence HD-ISS Staging. J Huntingtons Dis. 2024;13(1):77-90. doi: 10.3233/JHD-231512. PMID: 38489194.
- Koneczny I, Macher S, Hutterer M, Seifert-Held T, Berger-Sieczkowski E, Blaabjerg M, Breu M, Dreyhaupt J, Dutra LA, Erdler M, Fae I, Fischer G, Frommlet F, Heidbreder A, Högl B, Klose V, Klotz S, Liendl H, Nissen MS, Rahimi J, Reinecke R, Ricken G, Stefani A, Süße M, Teive HAG, Weis S, Berger T, Sabater L, Gaig C, Lewerenz J, Höftberger R. HLA dependency and possible clinical relevance of intrathecally synthesized anti-IgLON5 IgG4 in anti-IgLON5 disease. Front Immunol. 2024 May 16;15:1376456. doi: 10.3389/fimmu.2024.1376456. PMID: 38827736; PMCID: PMC11141242.
- Konvalinkova R, Srp M, Doleckova K, Capek V, Gal O, Hoskovcova M, Kliment R, Muzik J, Ruzicka E, Klempir J. The impact of expiratory muscle strength training on voluntary cough effectiveness in Huntington’s disease. Eur J Neurol. 2024 Sep 30:e16500. doi: 10.1111/ene.16500. Epub ahead of print. PMID: 39344651.
- Korpela S, Sundblom J, Zetterberg H, Constantinescu R, Svenningsson P, Paucar M, Niemelä V. Cerebrospinal fluid glial fibrillary acidic protein, in contrast to amyloid beta protein, is associated with disease symptoms in Huntington’s disease. J Neurol Sci. 2024 Apr 15;459:122979. doi: 10.1016/j.jns.2024.122979. Epub 2024 Mar 30. PMID: 38569376.
- Krause A, Anderson DG, Ferreira-Correia A, Dawson J, Baine-Savanhu F, Li PP, Margolis RL. Huntington disease-like 2: insight into neurodegeneration from an African disease. Nat Rev Neurol. 2024 Jan;20(1):36-49. doi: 10.1038/s41582-023-00906-y. Epub 2023 Dec 19. PMID: 38114648.
- Kuijper EC, Overzier M, Suidgeest E, Dzyubachyk O, Maguin C, Pérot JB, Flament J, Ariyurek Y, Mei H, Buijsen RAM, van der Weerd L, van Roon-Mom W. Antisense oligonucleotide-mediated disruption of HTT caspase-6 cleavage site ameliorates the phenotype of YAC128 Huntington disease mice. Neurobiol Dis. 2024 Jan;190:106368. doi: 10.1016/j.nbd.2023.106368. Epub 2023 Nov 29. PMID: 38040383.
- Kruijthof C, de Boer ME, van Loon AM, Bredewold J, van Dusseldorp L. Experiences of Ambulatory Patients With Huntington’s Disease With Case Management: A Qualitative Study. Prof Case Manag. 2024 Jan-Feb 01;29(1):13-21. doi: 10.1097/NCM.0000000000000680. PMID: 37983776.
- Lee CY, Shin C, Hwang YS, Oh E, Kim M, Kim HS, Chung SJ, Sung YH, Yoon WT, Cho JW, Lee JH, Kim HJ, Chang HJ, Jeon B, Woo KA, Koh SB, Kwon KY, Moon J, Kim YE, Lee JY. Caregiver Burden of Patients With Huntington’s Disease in South Korea. J Mov Disord. 2024 Jan;17(1):30-37. doi: 10.14802/jmd.23134. Epub 2023 Sep 11. PMID: 37691330; PMCID: PMC10846961.
- Lefaucheur JP, Moro E, Shirota Y, Ugawa Y, Grippe T, Chen R, Benninger DH, Jabbari B, Attaripour S, Hallett M, Paulus W. Clinical neurophysiology in the treatment of movement disorders: IFCN handbook chapter. Clin Neurophysiol. 2024 May 23;164:57-99. doi: 10.1016/j.clinph.2024.05.007. Epub ahead of print. PMID: 38852434.
- Le Stanc L, Lunven M, Giavazzi M, Sliwinski A, Youssov K, Bachoud-Lévi AC, Jacquemot C. Cognitive reserve involves decision making and is associated with left parietal and hippocampal hypertrophy in neurodegeneration. Commun Biol. 2024 Jun 18;7(1):741. doi: 10.1038/s42003-024-06416-x. PMID: 38890487; PMCID: PMC11189446.
- Li H, Desai R, Quiles N, Quinn L, Friel C. Characterizing Heart Rate Variability Response to Maximal Exercise Testing in People with Huntington’s Disease. J Huntingtons Dis. 2024;13(1):67-76. doi: 10.3233/JHD-230593. PMID: 38489192.
- Lisowski P, Lickfett S, Rybak-Wolf A, Menacho C, Le S, Pentimalli TM, Notopoulou S, Dykstra W, Oehler D, López-Calcerrada S, Mlody B, Otto M, Wu H, Richter Y, Roth P, Anand R, Kulka LAM, Meierhofer D, Glazar P, Legnini I, Telugu NS, Hahn T, Neuendorf N, Miller DC, Böddrich A, Polzin A, Mayatepek E, Diecke S, Olzscha H, Kirstein J, Ugalde C, Petrakis S, Cambridge S, Rajewsky N, Kühn R, Wanker EE, Priller J, Metzger JJ, Prigione A. Mutant huntingtin impairs neurodevelopment in human brain organoids through CHCHD2-mediated neurometabolic failure. Nat Commun. 2024 Aug 22;15(1):7027. doi: 10.1038/s41467-024-51216-w. PMID: 39174523; PMCID: PMC11341898.
- Li X, Hernandez I, Koyuncu S, Kis B, Häggblad M, Lidemalm L, Abbas AA, Bendegúz S, Göblös A, Brautigam L, Lucas JJ, Carreras-Puigvert J, Hühn D, Pircs K, Vilchez D, Fernandez-Capetillo O. The anti-leprosy drug clofazimine reduces polyQ toxicity through activation of PPARγ. EBioMedicine. 2024 May;103:105124. doi: 10.1016/j.ebiom.2024.105124. Epub 2024 May 2. PMID: 38701619; PMCID: PMC11088276.
- Lin L, Cai M, Su F, Wu T, Yuan K, Li Y, Luo Y, Chen D, Pei Z. Real-world experience with Deutetrabenazine management in patients with Huntington’s disease using video-based telemedicine. Neurol Sci. 2024 May;45(5):2047-2055. doi: 10.1007/s10072-023-07179-9. Epub 2023 Nov 17. PMID: 37973627.
- Liu X, Wang F, Fan X, Chen M, Xu X, Xu Q, Zhu H, Xu A, Pouladi MA, Xu X. CHCHD2 up-regulation in Huntington disease mediates a compensatory protective response against oxidative stress. Cell Death Dis. 2024 Feb 10;15(2):126. doi: 10.1038/s41419-024-06523-x. PMID: 38341417; PMCID: PMC10858906.
- López-Molina L, Sancho-Balsells A, Al-Massadi O, Montalban E, Alberch J, Arranz B, Girault JA, Giralt A. Hippocampal Pyk2 regulates specific social skills: Implications for schizophrenia. Neurobiol Dis. 2024 May;194:106487. doi: 10.1016/j.nbd.2024.106487. Epub 2024 Mar 27. PMID: 38552722.
- Louçã M, El Akrouti D, Lemesle A, Louessard M, Dufour N, Baroin C, de la Fouchardière A, Cotter L, Jean-Jacques H, Redeker V, Perrier AL. Huntingtin lowering impairs the maturation and synchronized synaptic activity of human cortical neuronal networks derived from induced pluripotent stem cells. Neurobiol Dis. 2024 Oct 1;200:106630. doi: 10.1016/j.nbd.2024.106630. Epub 2024 Aug 5. PMID: 39106928.
- Louessard M, Cailleret M, Jarrige M, Bigarreau J, Lenoir S, Dufour N, Rey M, Saudou F, Deglon N, Perrier AL. Mono- and Biallelic Inactivation of Huntingtin Gene in Patient-Specific Induced Pluripotent Stem Cells Reveal HTT Roles in Striatal Development and Neuronal Functions. J Huntingtons Dis. 2024 Feb 24. doi: 10.3233/JHD-231509. Epub ahead of print. PMID: 38427495.
- Lozano-Garcia M, Doheny EP, Mann E, Morgan-Jones P, Drew C, Busse-Morris M, Lowery MM. Estimation of Gait Parameters in Huntington’s Disease using Wearable Sensors in the Clinic and Free-living Conditions. IEEE Trans Neural Syst Rehabil Eng. 2024 May 31;PP. doi: 10.1109/TNSRE.2024.3407887. Epub ahead of print. PMID: 38819972.
- Luykx JJ, van Duijn E, Geerdinck B, Burgers E, Kremer HPH, Veenhuizen R. Het diagnostisch proces bij chorea: het belang van heroverwegen en de familieanamnese [The value of re-evaluation and thorough family history taking for the diagnostic work-up of chorea]. Tijdschr Psychiatr. 2024;66(1):51-54. Dutch. PMID: 38380489.
- Machacek M, Garcia-Montoya E, McColgan P, Sanwald-Ducray P, Mazer NA. NfL concentration in CSF is a quantitative marker of the rate of neurodegeneration in aging and Huntington’s disease: a semi-mechanistic model-based analysis. Front Neurosci. 2024 Jul 3;18:1420198. doi: 10.3389/fnins.2024.1420198. PMID: 39022122; PMCID: PMC11253127.
- Maiuri T, Bazan CB, Harding RJ, Begeja N, Kam TI, Byrne LM, Rodrigues FB, Warner MM, Neuman K, Mansoor M, Badiee M, Dasovich M, Wang K, Thompson LM, Leung AKL, Andres SN, Wild EJ, Dawson TM, Dawson VL, Arrowsmith CH, Truant R. Poly ADP-ribose signaling is dysregulated in Huntington disease. Proc Natl Acad Sci U S A. 2024 Oct;121(40):e2318098121. doi: 10.1073/pnas.2318098121. Epub 2024 Sep 27. https://pubmed.ncbi.nlm.nih.gov/39331414/”>PMID: 39331414; PMCID: PMC11459172 (available on 2025-03-27).
- Mammen JR, Tyo M, Cadorette J, Adams JL, Xiao Y, Stephenson D, Bale C. Understanding what aspects of Parkinson’s disease matter most to patients and families. Sci Rep. 2024 Sep 11;14(1):21171. doi: 10.1038/s41598-024-71555-4. PMID: 39256441; PMCID: PMC11387791.
- Mangin A, Dion V, Menzies G. Developing small Cas9 hybrids using molecular modeling. Sci Rep. 2024 Jul 26;14(1):17233. doi: 10.1038/s41598-024-68107-1. PMID: 39060399; PMCID: PMC11282279.
- Martinez-Horta S, Perez-Perez J, Perez-Gonzalez R, Sampedro F, Horta-Barba A, Campolongo A, Rivas-Asensio E, Puig-Davi A, Pagonabarraga J, Kulisevsky J. Cognitive phenotype and neurodegeneration associated with Tau in Huntington’s disease. Ann Clin Transl Neurol. 2024 May;11(5):1160-1171. doi: 10.1002/acn3.52031. Epub 2024 Mar 27. PMID: 38544341; PMCID: PMC11093246.
- Martin-Solana E, Casado-Zueras L, Torres TE, Goya GF, Fernandez-Fernandez MR, Fernandez JJ. Disruption of the mitochondrial network in a mouse model of Huntington’s disease visualized by in-tissue multiscale 3D electron microscopy. Acta Neuropathol Commun. 2024 Jun 5;12(1):88. doi: 10.1186/s40478-024-01802-2. PMID: 38840253; PMCID: PMC11151585.
- Martin-Solana E, Diaz-Lopez I, Mohamedi Y, Ventoso I, Fernandez JJ, Fernandez-Fernandez MR. Progressive alterations in polysomal architecture and activation of ribosome stalling relief factors in a mouse model of Huntington’s disease. Neurobiol Dis. 2024 Jun 1;195:106488. doi: 10.1016/j.nbd.2024.106488. Epub 2024 Mar 31. PMID: 38565397.
- Mason SL, Barker RA, Andresen K, Gracey F, Ford C. The meaning of apathy in Huntington’s disease: A qualitative study of caregiver perspectives. Neuropsychol Rehabil. 2024 Aug 5:1-30. doi: 10.1080/09602011.2024.2384519. Epub ahead of print. PMID: 39102382.
- Massey TH, McLauchlan DJ. Huntington’s disease: A clinical primer for acute and general physicians. Clin Med (Lond). 2024 Mar;24(2):100200. doi: 10.1016/j.clinme.2024.100200. Epub 2024 Apr 6. PMID: 38588915; PMCID: PMC11061216.
- McColgan P, Tabrizi SJ, Doody RS; GENERATION HD1 Investigators. Concern about Tominersen in Patients with Huntington’s Disease. Reply. N Engl J Med. 2024 Mar 14;390(11):1059. doi: 10.1056/NEJMc2400161. PMID: 38478006.
- McDonnell EI, Xie S, Marder K, Cui F, Wang Y. Dynamic undirected graphical models for time-varying clinical symptom and neuroimaging networks. Stat Med. 2024 Sep 20;43(21):4131-4147. doi: 10.1002/sim.10143. Epub 2024 Jul 15. PMID: 39007408.
- McLean ZL, Gao D, Correia K, Roy JCL, Shibata S, Farnum IN, Valdepenas-Mellor Z, Kovalenko M, Rapuru M, Morini E, Ruliera J, Gillis T, Lucente D, Kleinstiver BP, Lee JM, MacDonald ME, Wheeler VC, Mouro Pinto R, Gusella JF. Splice modulators target PMS1 to reduce somatic expansion of the Huntington’s disease-associated CAG repeat. Nat Commun. 2024 Apr 12;15(1):3182. doi: 10.1038/s41467-024-47485-0. PMID: 38609352; PMCID: PMC11015039.
- Medina Escobar A, Pringsheim T, Gautreau S, Rivera-Duarte JD, Amorelli G, Cornejo-Olivas M, Rossi M. Epidemiology of Huntington’s Disease in Latin America: A Systematic Review and Meta-Analysis. Mov Disord. 2024 Jul 23. doi: 10.1002/mds.29929. Epub ahead of print. PMID: 39044616.
- Meoni S, Moro E. The impact of COVID-19 pandemic on patients with Huntington’s disease and care-givers: A French survey. eNeurologicalSci. 2024 Jul 20;36:100517. doi: 10.1016/j.ensci.2024.100517. PMID: 39161890; PMCID: PMC11332790.
- Mestre TA. Laquinimod, Huntington’s disease, and disease modification. Lancet Neurol. 2024 Mar;23(3):220-221. doi: 10.1016/S1474-4422(24)00001-2. Epub 2024 Jan 24. PMID: 38280390.
- Mestre TA, Stebbins GT, Stephenson D, Dexter D, Lee KK, Xiao Y, Dam T, Kopil CM, Simuni T. Patient-centered development of clinical outcome assessments in early Parkinson disease: key priorities and advances. NPJ Parkinsons Dis. 2024 May 14;10(1):101. doi: 10.1038/s41531-024-00716-z. PMID: 38744872; PMCID: PMC11094181.
- Migliore S, Bianco SD, Scocchia M, Maffi S, Busi LC, Ceccarelli C, Curcio G, Mazza T, Squitieri F. Prodromal Cognitive Changes as a Prognostic Indicator of Forthcoming Huntington’s Disease Severity: A Retrospective Longitudinal Study. Mov Disord Clin Pract. 2024 Jan 24. doi: 10.1002/mdc3.13975. Epub ahead of print. PMID: 38264920.
- Migliore S, Casella M, Tramontano C, Curcio G, Squitieri F. Virtual reality tolerability, sense of presence and usability in Huntington disease: a pilot study. Neurol Sci. 2024 Aug 6. doi: 10.1007/s10072-024-07726-y. Epub ahead of print. PMID: 39103734.
- Mills JA, Long JD, Vaidya JG, Gantman EC, Sathe S, Tabrizi SJ, Sampaio C. Time to Functional Loss as an Endpoint in Huntington’s Disease Trials: Enrichment and Sample Size. Mov Disord. 2024 Aug 5. doi: 10.1002/mds.29963. Epub ahead of print. PMID: 39101272.
- Morandell J, Monziani A, Lazioli M, Donzel D, Döring J, Oss Pegorar C, D’Anzi A, Pellegrini M, Mattiello A, Bortolotti D, Bergonzoni G, Tripathi T, Mattis VB, Kovalenko M, Rosati J, Dieterich C, Dassi E, Wheeler VC, Ellederová Z, Wilusz JE, Viero G, Biagioli M. CircHTT(2,3,4,5,6)- co-evolving with the HTT CAG-repeat tract – modulates Huntington’s disease phenotypes. Mol Ther Nucleic Acids. 2024 Jun 3;35(3):102234. doi: 10.1016/j.omtn.2024.102234. PMID: 38974999; PMCID: PMC11225910.
- Morris LA, Horne KL, Manohar S, Paermentier L, Buchanan C, MacAskill M, Myall D, Apps M, Roxburgh R, Anderson T, Husain M, Le Heron C. Decision cost hypersensitivity underlies Huntington’s disease apathy. Brain. 2024 Sep 12:awae296. doi: 10.1093/brain/awae296. Epub ahead of print. PMID: 39269457.
- Mousavi MA, Rezaee M, Pourhamzeh M, Salari M, Hossein-Khannazer N, Shpichka A, Nabavi SM, Timashev P, Vosough M. Translational Approach Using Advanced Therapy Medicinal Products for Huntington’s Disease. Curr Rev Clin Exp Pharmacol. 2024 May 23. doi: 10.2174/0127724328300166240510071548. Epub ahead of print. PMID: 38797903.
- Mühlbäck A, Hoffmann R, Pozzi NG, Marziniak M, Brieger P, Dose M, Priller J. Psychiatrische Symptome der Huntington-Krankheit [Psychiatric symptoms of Huntington’s disease]. Nervenarzt. 2024 Sep;95(9):871-884. German. doi: 10.1007/s00115-024-01728-z. Epub 2024 Aug 30. PMID: 39212681; PMCID: PMC11374876.
- Muthinja MJ, Guelngar CO, Fall M, Jama F, Shuja HA, Nambafu J, Massi DG, Ojo OO, Okubadejo NU, Taiwo FT, Diop AM, de Chacus CJDG, Cissé FA, Cissé A, Hooker J, Sokhi D, Houlden H, Rizig M. An exploration of the genetics of the mutant Huntingtin (mHtt) gene in a cohort of patients with chorea from different ethnic groups in sub-Saharan Africa. Ann Hum Genet. 2024 Apr 2. doi: 10.1111/ahg.12557. Epub ahead of print. PMID: 38563088.
- Narotam-Jeena H, Guttman M, van Hillegondsberg L, van Coller R, Krause A, Carr J. Atypical Presentations of Huntington Disease-like 2 in South African Individuals. Mov Disord Clin Pract. 2024 May 9. doi: 10.1002/mdc3.14052. Epub ahead of print. PMID: 38725192; PMCID: PMC11233840.
- Neema M, Schultz JL, Langbehn DR, Conrad AL, Epping EA, Magnotta VA, Nopoulos PC. Mutant Huntingtin Drives Development of an Advantageous Brain Early in Life: Evidence in Support of Antagonistic Pleiotropy. Ann Neurol. 2024 Aug 8. doi: 10.1002/ana.27046. Epub ahead of print. PMID: 39115048; PMCID: PMC11496017 (available on 2025-11-01).
- Neueder A, Kojer K, Gu Z, Wang Y, Hering T, Tabrizi S, Taanman JW, Orth M. Huntington disease affects mitochondrial network dynamics predisposing to pathogenic mtDNA mutations. Brain. 2024 Jan 9:awae007. doi: 10.1093/brain/awae007. Epub ahead of print. PMID: 38195181.
- Neueder A, Nitzschner P, Wagner R, Hummel J, Hoschek F, Wagner M, Abdelmoez A, von Einem B, Landwehrmeyer GB, Tabrizi SJ, Orth M. Huntington disease alters the actionable information in plasma extracellular vesicles. Clin Transl Med. 2024 Jan;14(1):e1525. doi: 10.1002/ctm2.1525. PMID: 38193625; PMCID: PMC10775183.
- Nou-Fontanet L, Nguyen QTR, Bachoud-Levi AC, Reinhard C; Chorea & Huntington Disease Group ERN-RND; Ortigoza-Escobar JD. Insights from European Reference Network for rare neurological disorders study surveys on diagnosis, treatment, and management of NKX2-1-related disorders. Eur J Paediatr Neurol. 2024 Jul;51:110-117. doi: 10.1016/j.ejpn.2024.06.007. Epub 2024 Jun 19. PMID: 38917695.
- Nunes AS, Pawlik M, Mishra RK, Waddell E, Coffey M, Tarolli CG, Schneider RB, Dorsey ER, Vaziri A, Adams JL. Digital assessment of speech in Huntington disease. Front Neurol. 2024 Jan 23;15:1310548. doi: 10.3389/fneur.2024.1310548. PMID: 38322583; PMCID: PMC10844459.
- Ogilvie AC, Carnahan RM, Mendizabal A, Gilbertson-White S, Seaman A, Chrischilles E, Schultz JL. Factors Influencing Discharges to Hospice for Patients With Late-Stage Huntington’s Disease. Am J Hosp Palliat Care. 2024 Aug 21:10499091241274725. doi: 10.1177/10499091241274725. Epub ahead of print. PMID: 39167632.
- Olechnowicz A, Blatkiewicz M, Jopek K, Isalan M, Mielcarek M, Rucinski M. Deregulated Transcriptome as a Platform for Adrenal Huntington’s Disease-Related Pathology. Int J Mol Sci. 2024 Feb 11;25(4):2176. doi: 10.3390/ijms25042176. PMID: 38396853; PMCID: PMC10888552.
- Oosterloo M, Touze A, Byrne LM, Achenbach J, Aksoy H, Coleman A, Lammert D, Nance M, Nopoulos P, Reilmann R, Saft C, Santini H, Squitieri F, Tabrizi S, Burgunder JM, Quarrell O; Pediatric Huntington Disease Working Group of the European Huntington Disease Network. Clinical Review of Juvenile Huntington’s Disease. J Huntingtons Dis. 2024 Apr 26. doi: 10.3233/JHD-231523. Epub ahead of print. PMID: 38669553.
- Paldino E, Migliorato G, Fusco FR. Neuroimmune pathways involvement in neurodegeneration of R6/2 mouse model of Huntington’s disease. Front Cell Neurosci. 2024 Feb 20;18:1360066. doi: 10.3389/fncel.2024.1360066. PMID: 38444595; PMCID: PMC10912295.
- Pardina-Torner H, De Paepe AE, Garcia-Gorro C, Rodriguez-Dechicha N, Vaquer I, Calopa M, Ruiz-Idiago J, Mareca C, de Diego-Balaguer R, Camara E. Disentangling the neurobiological bases of temporal impulsivity in Huntington’s disease. Brain Behav. 2024 Mar;14(3):e3335. doi: 10.1002/brb3.3335. PMID: 38450912; PMCID: PMC10918610.
- Parlato R, Volarić J, Lasorsa A, Bagherpoor Helabad M, Kobauri P, Jain G, Miettinen MS, Feringa BL, Szymanski W, van der Wel PCA. Photocontrol of the β-Hairpin Polypeptide Structure through an Optimized Azobenzene-Based Amino Acid Analogue. J Am Chem Soc. 2024 Jan 24;146(3):2062-2071. doi: 10.1021/jacs.3c11155. Epub 2024 Jan 16. PMID: 38226790; PMCID: PMC10811659.
- Park SJ, Son SM, Barbosa AD, Wrobel L, Stamatakou E, Squitieri F, Balmus G, Rubinsztein DC. Nuclear proteasomes buffer cytoplasmic proteins during autophagy compromise. Nat Cell Biol. 2024 Oct;26(10):1691-1699. doi: 10.1038/s41556-024-01488-7. Epub 2024 Aug 29. PMID: 39209961; PMCID: PMC11469956.
- Pengo M, Squitieri F. Beyond CAG Repeats: The Multifaceted Role of Genetics in Huntington Disease. Genes (Basel). 2024 Jun 19;15(6):807. doi: 10.3390/genes15060807. PMID: 38927742; PMCID: PMC11203031.
- Pérez-Oliveira S, Castilla-Silgado J, Painous C, Aldecoa I, Menéndez-González M, Blázquez-Estrada M, Corte D, Tomás-Zapico C, Compta Y, Muñoz E, Lladó A, Balasa M, Aragonès G, García-González P, Rosende-Roca M, Boada M, Ruíz A, Pastor P, De la Casa-Fages B, Rabano A, Sánchez-Valle R, Molina-Porcel L, Álvarez V. Huntingtin CAG repeats in neuropathologically confirmed tauopathies: Novel insights. Brain Pathol. 2024 Feb 28:e13250. doi: 10.1111/bpa.13250. Epub ahead of print. PMID: 38418081; PMCID: PMC11189778.
- Pérot JB, Brouillet E, Flament J. The contribution of preclinical magnetic resonance imaging and spectroscopy to Huntington’s disease. Front Aging Neurosci. 2024 Feb 13;16:1306312. doi: 10.3389/fnagi.2024.1306312. PMID: 38414634; PMCID: PMC10896846.
- Pierobon Mays G, Hett K, Eisma J, McKnight CD, Elenberger J, Song AK, Considine C, Richerson WT, Han C, Stark A, Claassen DO, Donahue MJ. Reduced cerebrospinal fluid motion in patients with Parkinson’s disease revealed by magnetic resonance imaging with low b-value diffusion weighted imaging. Fluids Barriers CNS. 2024 May 9;21(1):40. doi: 10.1186/s12987-024-00542-8. PMID: 38725029; PMCID: PMC11080257.
- Pierzynowska K, Podlacha M, Gaffke L, Rintz E, Wiśniewska K, Cyske Z, Węgrzyn G. Correction of symptoms of Huntington disease by genistein through FOXO3-mediated autophagy stimulation. Autophagy. 2024 May;20(5):1159-1182. doi: 10.1080/15548627.2023.2286116. Epub 2023 Nov 27. PMID: 37992314; PMCID: PMC11135876.
- Pizzorni N, Ciammola A, Pirola C, Nanetti L, Castaldo A, Poletti B, Mariotti C, Schindler A. Oropharyngeal Dysphagia Phenotypes Across Huntington’s Disease Stages: Endoscopic Findings and Tongue Pressure Analysis. J Huntingtons Dis. 2024 May 28. doi: 10.3233/JHD-231519. Epub ahead of print. PMID: 38820019.
- Pradhan S, Bush K, Zhang N, Pandita RK, Tsai CL, Smith C, Pandlebury DF, Gaikwad S, Leonard F, Nie L, Tao A, Russell W, Yuan S, Choudhary S, Ramos KS, Elferink C, Wairkar YP, Tainer JA, Thompson LM, Pandita TK, Sarkar PS. Chromatin remodeler BRG1 recruits huntingtin to repair DNA double-strand breaks in neurons. bioRxiv [Preprint]. 2024 Sep 20:2024.09.19.613927. doi: 10.1101/2024.09.19.613927. PMID: 39345557; PMCID: PMC11429940.
- Qian SX, Bao YF, Li XY, Dong Y, Zhang XL, Wu ZY. Multi-omics Analysis Reveals Key Gut Microbiota and Metabolites Closely Associated with Huntington’s Disease. Mol Neurobiol. 2024 Jun 8. doi: 10.1007/s12035-024-04271-9. Epub ahead of print. PMID: 38850348.
- Radulescu CI, Ferrari Bardile C, Garcia-Miralles M, Sidik H, Yusof NABM, Pouladi MA. Environmental Deprivation Effects on Myelin Ultrastructure in Huntington Disease and Wildtype Mice. Mol Neurobiol. 2024 Jul;61(7):4278-4288. doi: 10.1007/s12035-023-03799-6. Epub 2023 Dec 11. PMID: 38079108.
- Raschka T, Li Z, Gaßner H, Kohl Z, Jukic J, Marxreiter F, Fröhlich H. Unraveling progression subtypes in people with Huntington’s disease. EPMA J. 2024 May 28;15(2):275-287. doi: 10.1007/s13167-024-00368-2. PMID: 38841617; PMCID: PMC11148000.
- Ratié L, Humbert S. A developmental component to Huntington’s disease. Rev Neurol (Paris). 2024 May;180(5):357-362. doi: 10.1016/j.neurol.2024.04.001. Epub 2024 Apr 12. PMID: 38614929.
- Ratz-Wirsching V, Habermeyer J, Moceri S, Harrer J, Schmitz C, von Hörsten S. Gene-dosage- and sex-dependent differences in the prodromal-Like phase of the F344tgHD rat model for Huntington disease. Front Neurosci. 2024 Feb 7;18:1354977. doi: 10.3389/fnins.2024.1354977. PMID: 38384482; PMCID: PMC10879377.
- Reilmann R, Anderson KE, Feigin A, Tabrizi SJ, Leavitt BR, Stout JC, Piccini P, Schubert R, Loupe P, Wickenberg A, Borowsky B, Rynkowski G, Volkinshtein R, Li T, Savola JM, Hayden M, Gordon MF; LEGATO-HD Study Group. Safety and efficacy of laquinimod for Huntington’s disease (LEGATO-HD): a multicentre, randomised, double-blind, placebo-controlled, phase 2 study. Lancet Neurol. 2024 Mar;23(3):243-255. doi: 10.1016/S1474-4422(23)00454-4. Epub 2024 Jan 24. PMID: 38280392.
- Reilmann R. Concern about Tominersen in Patients with Huntington’s Disease. N Engl J Med. 2024 Mar 14;390(11):1058-1059. doi: 10.1056/NEJMc2400161. PMID: 38478005.
- Rivadeneyra-Posadas J, Simón-Vicente L, Castillo-Alvira D, Raya-González J, Soto-Celix M, Rodríguez-Fernández A, García-Bustillo A, Saiz-Rodríguez M, Vázquez-Sánchez F, Aguado-Garcia L, Gámez Leyva-Hernández G, Cubo E. How to estimate body composition in Huntington’s disease. A cross-sectional, observational study using multiple frequencies bioimpedance. Rev Neurol. 2024 Jan 1;78(1):17-25. Spanish, English. doi: 10.33588/rn.7801.2023224. PMID: 38112653.
- Roos AK, Stenvall E, Kockum ES, Grönlund KÅ, Alstermark H, Wuolikainen A, Andersen PM, Nordin A, Forsberg KME. Small striatal huntingtin inclusions in patients with motor neuron disease with reduced penetrance and intermediate HTT gene expansions. Hum Mol Genet. 2024 Sep 13:ddae137. doi: 10.1093/hmg/ddae137. Epub ahead of print. PMID: 39270726; PMCID: PMC11555821.
- Rossetti MA, Anderson KM, Hay KR, Del Bene VA, Celka AS, Piccolino A, Nelson Sheese AL, Huynh M, Zhu L, Claassen DO, Furr Stimming E, Considine CM. An Exploratory Pilot Study of Neuropsychological Performance in Two Huntington Disease Centers of Excellence Clinics. Arch Clin Neuropsychol. 2024 Jan 19;39(1):24-34. doi: 10.1093/arclin/acad054. PMID: 37530515.
- Ruiz de Sabando A, Ciosi M, Galbete A, Cumming SA; Spanish HD Collaborative Group; Monckton DG, Ramos-Arroyo MA. Somatic CAG repeat instability in intermediate alleles of the HTT gene and its potential association with a clinical phenotype. Eur J Hum Genet. 2024 Mar 4. doi: 10.1038/s41431-024-01546-6. Epub ahead of print. PMID: 38433266; PMCID: PMC11220145.
- Ruiz-Barrio I, Vázquez-Oliver A, Puig-Davi A, Rivas-Asensio E, Perez-Perez J, Fernandez-Vizuete C, Horta-Barba A, Olmedo-Saura G, Salvat-Rovira N, Sampedro F, Vacchi E, Melli G, Pagonabarraga J, Kulisevsky J, Martinez-Horta S. Skin Tau Quantification as a Novel Biomarker in Huntington’s Disease. Mov Disord. 2024 Aug 28. doi: 10.1002/mds.29989. Epub ahead of print. PMID: 39192729.
- Ryan L, Rubinsztein DC. The autophagy of stress granules. FEBS Lett. 2024 Jan;598(1):59-72. doi: 10.1002/1873-3468.14787. Epub 2023 Dec 21. PMID: 38101818.
- Saade J, Mestre TA. Huntington’s Disease: Latest Frontiers in Therapeutics. Curr Neurol Neurosci Rep. 2024 Jun 11. doi: 10.1007/s11910-024-01345-y. Epub ahead of print. PMID: 38861215.
- Salaun L, Bonduelle T, Ghorayeb I, Spampinato U, Debruxelles S, Guehl D, Goizet C. Intensification of Diurnal Abnormal Movements During Sleep in Huntington’s Disease. J Huntingtons Dis. 2024 May 18. doi: 10.3233/JHD-231518. Epub ahead of print. PMID: 38788081.
- Salem S, Kilgore MD, Anwer M, Maxan A, Child D, Bird TD, Keene CD, Cicchetti F, Latimer C. Evidence of mutant huntingtin and tau-related pathology within neuronal grafts in Huntington’s disease cases. Neurobiol Dis. 2024 Aug;198:106542. doi: 10.1016/j.nbd.2024.106542. Epub 2024 May 27. PMID: 38810948.
- Sampaio C. Huntington disease – Update on ongoing therapeutic developments and a look toward the future. Parkinsonism Relat Disord. 2024 Feb 15:106049. doi: 10.1016/j.parkreldis.2024.106049. Epub ahead of print. PMID: 38418319.
- Savva K, Zachariou M, Bourdakou MM, Dietis N, Spyrou GM. DReAmocracy: A Method to Capitalise on Prior Drug Discovery Efforts to Highlight Candidate Drugs for Repurposing. Int J Mol Sci. 2024 May 13;25(10):5319. doi: 10.3390/ijms25105319. PMID: 38791356; PMCID: PMC11121186.
- Sawant R, Paret K, Petrillo J, Koenig A, Wolowacz S, Ronquest N, Rickards H. Health state utility estimates for value assessments of novel treatments in Huntington’s disease: a systematic literature review. Health Qual Life Outcomes. 2024 Apr 16;22(1):33. doi: 10.1186/s12955-024-02242-1. PMID: 38627749; PMCID: PMC11020898.
- Scolz A, Vezzoli E, Villa M, Talpo F, Cazzola J, Raffin F, Cordiglieri C, Falqui A, Pepe G, Maglione V, Besusso D, Biella G, Zuccato C. Neuroprotection by ADAM10 inhibition requires TrkB signaling in the Huntington’s disease hippocampus. Cell Mol Life Sci. 2024 Aug 7;81(1):333. doi: 10.1007/s00018-024-05382-1. PMID: 39112663; PMCID: PMC11335257.
- Seefelder M, Kochanek S, Klein FAC. ProteinCoLoc streamlines Bayesian analysis of colocalization in microscopic images. Sci Rep. 2024 Jun 10;14(1):13277. doi: 10.1038/s41598-024-63884-1. PMID: 38858475; PMCID: PMC11164984.
- Shacham T, Offen D, Lederkremer GZ. Efficacy of therapy by MK-28 PERK activation in the Huntington’s disease R6/2 mouse model. Neurotherapeutics. 2024 Feb 16;21(2):e00335. doi: 10.1016/j.neurot.2024.e00335. Epub ahead of print. PMID: 38368172; PMCID: PMC10937961.
- Shin JH, Yang HJ, Ahn JH, Jo S, Chung SJ, Lee JY, Kim HS, Kim M; Korean Huntington’s Disease Society. Evidence-based review on symptomatic management of Huntington’s disease. J Mov Disord. 2024 Aug 9. doi: 10.14802/jmd.24140. Epub ahead of print. PMID: 39117301; PMCID: PMC11540544.
- Shin C, Kim R, Yoo D, Oh E, Moon J, Kim M, Lee JY, Kim JM, Koh SB, Kim M, Jeon B; Korean Huntington’s Disease Society. A practical guide for clinical approach to patients with Huntington’s disease in Korea. J Mov Disord. 2024 Mar 12. doi: 10.14802/jmd.24040. Epub ahead of print. PMID: 38467449.
- Sierra LA, Wynn A, Lanzaro E, Dzekon K, Russell A, Halko M, Claassen DO, Frank S, Considine CM, Laganiere S. Deciphering Cognitive Impairments in Huntington’s Disease: A Comparative Study of Stroop Test Variations. J Huntingtons Dis. 2024 May 11. doi: 10.3233/JHD-231528. Epub ahead of print. PMID: 38759020.
- Simón-Vicente L, Rodríguez-Fernández A, Rivadeneyra-Posadas J, Soto-Célix M, Raya-González J, Castillo-Alvira D, Calvo S, Mariscal N, García-Bustillo Á, Aguado L, Cubo E. Validation of ActiGraph and Fitbit in the assessment of energy expenditure in Huntington’s disease. Gait Posture. 2024 Mar;109:89-94. doi: 10.1016/j.gaitpost.2024.01.028. Epub 2024 Jan 28. PMID: 38286064.
- Simuni T, Chahine LM, Poston K, Brumm M, Buracchio T, Campbell M, Chowdhury S, Coffey C, Concha-Marambio L, Dam T, DiBiaso P, Foroud T, Frasier M, Gochanour C, Jennings D, Kieburtz K, Kopil CM, Merchant K, Mollenhauer B, Montine T, Nudelman K, Pagano G, Seibyl J, Sherer T, Singleton A, Stephenson D, Stern M, Soto C, Tanner CM, Tolosa E, Weintraub D, Xiao Y, Siderowf A, Dunn B, Marek K. A biological definition of neuronal α-synuclein disease: towards an integrated staging system for research. Lancet Neurol. 2024 Feb;23(2):178-190. doi: 10.1016/S1474-4422(23)00405-2. PMID: 38267190.
- Skwara J, Nowicki M, Sharif L, Milanowski Ł, Dulski J, Elert-Dobkowska E, Skrzypek K, Hoffman-Zacharska D, Koziorowski D, Sławek J. Differential diagnosis of Huntington’s disease- neurological aspects of NKX2-1-related disorders. J Neural Transm (Vienna). 2024 Sep;131(9):1013-1024. doi: 10.1007/s00702-024-02800-3. Epub 2024 Jun 25. PMID: 38916623; PMCID: PMC11365827.
- Snow ALB, Ciriegio AE, Watson KH, Pfalzer AC, Diehl S, Hale L, McDonell KE, Claassen DO, Compas BE. Stress in Huntington’s Disease: Characteristics and Correlates in Patients and At-Risk Individuals. J Huntingtons Dis. 2024 Apr 3. doi: 10.3233/JHD-231515. Epub ahead of print. PMID: 38578897.
- Sogorb-Gonzalez M, Landles C, Caron NS, Stam A, Osborne G, Hayden MR, Howland D, van Deventer S, Bates GP, Vallès A, Evers M. Exon 1-targeting miRNA reduces the pathogenic exon 1 HTT protein in Huntington disease models. Brain. 2024 Aug 18:awae266. doi: 10.1093/brain/awae266. Epub ahead of print. PMID: 39155061.
- Solana-Balaguer J, Garcia-Segura P, Campoy-Campos G, Chicote-González A, Fernández-Irigoyen J, Santamaría E, Pérez-Navarro E, Masana M, Alberch J, Malagelada C. Motor skill learning modulates striatal extracellular vesicles’ content in a mouse model of Huntington’s disease. Cell Commun Signal. 2024 Jun 11;22(1):321. doi: 10.1186/s12964-024-01693-9. PMID: 38863004; PMCID: PMC11167907.
- Sprenger GP, van Zwet EW, Bakels HS, Achterberg WP, Roos RA, de Bot ST. Prevalence and burden of pain across the entire spectrum of Huntington’s disease. J Neurol Neurosurg Psychiatry. 2024 Jan 30:jnnp-2023-332992. doi: 10.1136/jnnp-2023-332992. Epub ahead of print. PMID: 38290837.
- Steinhardt J, Zittel S, Tadic V, Tronnier V, Moll C, Bäumer T, Münchau A, Rasche D, Brüggemann N. GPi/GPe borderland- a potential sweet spot for deep brain stimulation for chorea in Huntington’s disease? Neurol Res Pract. 2024 May 23;6(1):28. doi: 10.1186/s42466-024-00316-5. PMID: 38778367; PMCID: PMC11112842.
- Suchy-Dicey AM, Longstreth WT Jr, Rhoads K, Umans J, Buchwald D, Grabowski T, Blennow K, Reiman E, Zetterberg H. Plasma biomarkers of Alzheimer’s disease and related dementias in American Indians: The Strong Heart Study. Alzheimers Dement. 2024 Jan 12. doi: 10.1002/alz.13664. Epub ahead of print. PMID: 38215191; PMCID: PMC10984473.
- Sun X, Liu L, Wu C, Li X, Guo J, Zhang J, Guan J, Wang N, Gu L, Yang XW, Li GM. Mutant huntingtin protein induces MLH1 degradation, DNA hyperexcision, and cGAS-STING-dependent apoptosis. Proc Natl Acad Sci USA. 2024 March 26. 121 (13). doi: 10.1073/pnas.2313652121. PMID: 38498709; PMCID: PMC10990133 (available on 2024-09-18).
- Stanisławska-Sachadyn A, Krzemiński M, Zielonka D, Krygier M, Ziętkiewicz E, Sławek J, Limon J; REGISTRY investigators of the European Huntington’s Disease Network (EHDN). Sex contribution to average age at onset of Huntington’s disease depends on the number of (CAG)n repeats. Sci Rep. 2024 Jul 8;14(1):15729. doi: 10.1038/s41598-024-64105-5. PMID: 38977715; PMCID: PMC11231309.
- Sun Z, Ware J, Dey S, Eyigoz E, Sathe S, Sampaio C, Hu J. Large-scale screening of clinical assessments to distinguish between states in the Integrated HD Progression Model (IHDPM). Front Aging Neurosci. 2024 Feb 13;16:1320755. doi: 10.3389/fnagi.2024.1320755. PMID: 38414632; PMCID: PMC10896990.
- Tan K, Alpaugh M, Ashton NJ, Chouinard S, Barker RA, Blennow K, Zetterberg H, Cicchetti F, Benedet AL. Plasma GFAP and its association with disease severity in Huntington’s disease. J Neurol. 2024 Apr;271(4):2108-2113. doi: 10.1007/s00415-023-12109-y. Epub 2023 Nov 18. PMID: 37979092.
- Tataridas-Pallas N, Aman Y, Williams R, Chapman H, Cheng KJH, Gomez-Paredes C, Bates GP, Labbadia J. Mitochondrial clearance and increased HSF-1 activity are coupled to promote longevity in fasted Caenorhabditis elegans. iScience. 2024 Apr 27;27(6):109834. doi: 10.1016/j.isci.2024.109834. PMID: 38784016; PMCID: PMC11112483.
- Tovar A, Perry S, Muñoz E, Painous C, Santacruz P, Ruiz-Idiago J, Mareca C, Hinzen W. Understanding of referential dependencies in Huntington’s disease. Neuropsychologia. 2024 Mar 4:108845. doi: 10.1016/j.neuropsychologia.2024.108845. Epub ahead of print. PMID: 38447638.
- Tripodi D, Vitarelli F, Spiti S, Leoni V. The Diagnostic Use of the Plasma Quantification of 24S-Hydroxycholesterol and Other Oxysterols in Neurodegenerative Disease. Adv Exp Med Biol. 2024;1440:337-351. doi: 10.1007/978-3-031-43883-7_17. PMID: 38036888.
- Tröger J, Dörr F, Schwed L, Linz N, König A, Thies T, Orozco-Arroyave JR, Rusz J. An automatic measure for speech intelligibility in dysarthrias-validation across multiple languages and neurological disorders. Front Digit Health. 2024 Jul 23;6:1440986. doi: 10.3389/fdgth.2024.1440986. PMID: 39108340; PMCID: PMC11300433.
- Turner M, Bartlett D, Poudel G, Zaenker P, Laws S, Lo J, Ziman M, Cruickshank T. Associations between Sleep Quality and Serum Levels of Neurofilament Light in Individuals with Premanifest Huntington Disease. Sleep Sci. 2024 Feb 15;17(2):e199-e202. doi: 10.1055/s-0043-1777783. PMID: 38846594; PMCID: PMC11152634.
- Twyning MJ, Tufi R, Gleeson TP, Kolodziej KM, Campesan S, Terriente-Felix A, Collins L, De Lazzari F, Giorgini F, Whitworth AJ. Partial loss of MCU mitigates pathology in vivo across a diverse range of neurodegenerative disease models. Cell Rep. 2024 Feb 27;43(2):113681. doi: 10.1016/j.celrep.2024.113681. Epub 2024 Jan 18. PMID: 38236772.
- van der Wel PCA. Solid-state nuclear magnetic resonance in the structural study of polyglutamine aggregation. Biochem Soc Trans. 2024 Apr 24;52(2):719-731. doi: 10.1042/BST20230731. PMID: 38563485; PMCID: PMC11088915.
- van Lonkhuizen PJC, Heemskerk AW, Meijer E, van Duijn E, de Bot ST, Klempir J, Landwehrmeyer GB, Mühlbäck A, Hoblyn J, Squitieri F, Chavannes NH, Vegt NJH; HEALTHE-RND consortium. Development of the Huntington Support App (HD-eHelp study): a human-centered and co-design approach. Front Neurol. 2024 Jul 1;15:1399126. doi: 10.3389/fneur.2024.1399126. PMID: 39011363; PMCID: PMC11246862.
- van Walsem MR, Howe EI, Andelic N, Frich JC. Primary health care professionals’ experiences with caring for patients with advanced Huntington’s disease: a qualitative study. BMC Prim Care. 2024 May 7;25(1):155. doi: 10.1186/s12875-024-02408-2. PMID: 38714964; PMCID: PMC11075220.
- Vasilkovska T, Salajeghe S, Vanreusel V, Van Audekerke J, Verschuuren M, Hirschler L, Warnking J, Pintelon I, Pustina D, Cachope R, Mrzljak L, Muñoz-Sanjuan I, Barbier EL, De Vos WH, Van der Linden A, Verhoye M. Longitudinal alterations in brain perfusion and vascular reactivity in the zQ175DN mouse model of Huntington’s disease. J Biomed Sci. 2024 April 16. 31 (1) :37. doi: 10.1186/s12929-024-01028-3. PMID: 38627751; PMCID: PMC11022401.
- Vercammen J, Terryn J, Van Daele S, Vermeer S, Vandenberghe W. A Case of Chorea with Slow Saccades Caused by NKX2-1 Mutation. Mov Disord Clin Pract. 2024 Mar 7. doi: 10.1002/mdc3.14013. Epub ahead of print. PMID: 38454250.
- Vidas-Guscic N, van Rijswijk J, Van Audekerke J, Jeurissen B, Nnah I, Tang H, Munoz-Sanjuan I, Pustina D, Cachope R, Van der Linden A, Bertoglio D, Verhoye M. Diffusion MRI marks progressive alterations in fiber integrity in the zQ175DN mouse model of Huntington’s disease. Neurobiol Dis. 2024 February 14. doi: 10.1016/j.nbd.2024.106438. PMID: 38365045.
- Vieira R, Mariani JN, Huynh NPT, Stephensen HJT, Solly R, Tate A, Schanz S, Cotrupi N, Mousaei M, Sporring J, Benraiss A, Goldman SA. Young glial progenitor cells competitively replace aged and diseased human glia in the adult chimeric mouse brain. Nat Biotechnol. 2024 May;42(5):719-730. doi: 10.1038/s41587-023-01798-5. Epub 2023 Jul 17. PMID: 37460676; PMCID: PMC11098747.
- Villegas LD, Chandrasekaran A, Andersen SAF, Nørremølle A, Schmid B, Pouladi MA, Freude K. Generation of three isogenic gene-edited Huntington’s disease human embryonic stem cell lines with DOX-inducible NGN2 expression cassette in the AAVS1 safe locus. Stem Cell Res. 2024 Jun;77:103408. doi: 10.1016/j.scr.2024.103408. Epub 2024 Mar 28. PMID: 38569398.
- Wagner M, Zhu G, Khalid F, Phan T, Maity P, Lupu L, Agyeman-Duah E, Wiese S, Lindenberg KS, Schön M, Landwehrmeyer GB, Penzo M, Kochanek S, Scharffetter-Kochanek K, Mulaw M, Iben S. General loss of proteostasis links Huntington disease to Cockayne syndrome. Neurobiol Dis. 2024 Sep 14;201:106668. doi: 10.1016/j.nbd.2024.106668. Epub ahead of print. PMID: 39284372.
- Wang N, Zhang S, Langfelder P, Ramanathan L, Plascencia M, Gao F, Vaca R, Gu X, Deng L, Dionisio LE, Prasad BC, Vogt T, Horvath S, Aaronson JS, Rosinski J, Yang XW. Msh3 and Pms1 Set Neuronal CAG-repeat Migration Rate to Drive Selective Striatal and Cortical Pathogenesis in HD Mice. bioRxiv [Preprint]. 2024 Jul 15:2024.07.09.602815. doi: 10.1101/2024.07.09.602815. PMID: 39026894; PMCID: PMC11257559.
- Wang X, Li Y, Li B, Shang H, Yang J. Gray matter alterations in Huntington’s disease: A meta-analysis of VBM neuroimaging studies. J Neurosci Res. 2024 Jul;102(7):e25366. doi: 10.1002/jnr.25366. PMID: 38953592.
- Wang X, Zhang Z, Ding Y, Chen T, Mucci L, Albanes D, Landi MT, Caporaso NE, Lam S, Tardon A, Chen C, Bojesen SE, Johansson M, Risch A, Bickeböller H, Wichmann HE, Rennert G, Arnold S, Brennan P, McKay JD, Field JK, Shete SS, Le Marchand L, Liu G, Andrew AS, Kiemeney LA, Zienolddiny-Narui S, Behndig A, Johansson M, Cox A, Lazarus P, Schabath MB, Aldrich MC, Hung RJ, Amos CI, Lin X, Christiani DC. Impact of individual level uncertainty of lung cancer polygenic risk score (PRS) on risk stratification. Genome Med. 2024 Feb 5;16(1):22. doi: 10.1186/s13073-024-01298-4. PMID: 38317189; PMCID: PMC10840262.
- Witzenberger M, Janowski R, Niessing D. Crystal structure of the RNA-recognition motif of Drosophila melanogaster tRNA (uracil-5-)-methyltransferase homolog A. Acta Crystallogr F Struct Biol Commun. 2024 Feb 1;80(Pt 2):36-42. doi: 10.1107/S2053230X24000645. Epub 2024 Jan 25. PMID: 38270511; PMCID: PMC10836426.
- Xia JQ, Cheng YF, Zhang SR, Ma YZ, Fu JJ, Yang TM, Zhang LY, Burgunder JM, Shang HF. The characteristic and prognostic role of blood inflammatory markers in patients with Huntington’s disease from China. Front Neurol. 2024 Mar 26;15:1374365. doi: 10.3389/fneur.2024.1374365. PMID: 38595854; PMCID: PMC11002148.
- Yadav M, Harding RJ, Li T, Xu X, Gall-Duncan T, Khan M, Bardile CF, Sequiera GL, Duan S, Chandrasekaran R, Pan A, Bu J, Yamazaki T, Hirose T, Prinos P, Tippett L, Turner C, Curtis MA, Faull RLM, Pouladi MA, Pearson CE, He HH, Arrowsmith CH. Huntingtin is an RNA binding protein and participates in NEAT1-mediated paraspeckles. Sci Adv. 2024 Jul 19;10(29):eado5264. doi: 10.1126/sciadv.ado5264. Epub 2024 Jul 19. PMID: 39028820; PMCID: PMC11259171.
- Yu-Taeger L, El-Ayoubi A, Qi P, Danielyan L, Nguyen HHP. Intravenous MSC-Treatment Improves Impaired Brain Functions in the R6/2 Mouse Model of Huntington’s Disease via Recovered Hepatic Pathological Changes. Cells. 2024 Mar 7;13(6):469. doi: 10.3390/cells13060469. PMID: 38534313; PMCID: PMC10969189.
- Yu Z, Teng Y, Yang J, Yang L. The role of exosomes in adult neurogenesis: implications for neurodegenerative diseases. Neural Regen Res. 2024 Feb;19(2):282-288. doi: 10.4103/1673-5374.379036. PMID: 37488879; PMCID: PMC10503605.
- Zadegan SA, Kupcha L, Patino J, Rocha NP, Teixeira AL, Furr Stimming E. Obsessive-compulsive and perseverative behaviors in Huntington’s disease. Behav Brain Res. 2024 Feb 26;458:114767. doi: 10.1016/j.bbr.2023.114767. Epub 2023 Nov 19. PMID: 37984520.
- Zadegan SA, Ramirez F, Reddy KS, Sahin O, Rocha NP, Teixeira AL, Furr Stimming E. Treatment of Depression in Huntington’s Disease: A Systematic Review. J Neuropsychiatry Clin Neurosci. 2024 Mar 26:appineuropsych20230120. doi: 10.1176/appi.neuropsych.20230120. Epub ahead of print. PMID: 38528808.
- Zajicek F, Verhaeghe J, De Lombaerde S, Van Eetveldt A, Miranda A, Munoz-Sanjuan I, Dominguez C, Khetarpal V, Bard J, Liu L, Staelens S, Bertoglio D. Preclinical evaluation of the novel [18F]CHDI-650 PET ligand for non-invasive quantification of mutant huntingtin aggregates in Huntington’s disease. Eur J Nucl Med Mol Imaging. 2024 Aug 27. doi: 10.1007/s00259-024-06880-x. Epub ahead of print. PMID: 39190197.
- Zarkali A, Hannaway N, McColgan P, Heslegrave AJ, Veleva E, Laban R, Zetterberg H, Lees AJ, Fox NC, Weil RS. Neuroimaging and plasma evidence of early white matter loss in Parkinson’s disease with poor outcomes. Brain Commun. 2024 Apr 16;6(3):fcae130. doi: 10.1093/braincomms/fcae130. PMID: 38715714; PMCID: PMC11073930.
- Zhang Z, Gehin C, Abriata LA, Dal Peraro M, Lashuel H. Differential Effects of Post-translational Modifications on the Membrane Interaction of Huntingtin Protein. ACS Chem Neurosci. 2024 May 16. doi: 10.1021/acschemneuro.4c00091. Epub ahead of print. PMID: 38752226; PMCID: PMC11191595.
2023
- Abjean L, Ben Haim L, Riquelme-Perez M, Gipchtein P, Derbois C, Palomares MA, Petit F, Hérard AS, Gaillard MC, Guillermier M, Gaudin-Guérif M, Aurégan G, Sagar N, Héry C, Dufour N, Robil N, Kabani M, Melki R, De la Grange P, Bemelmans AP, Bonvento G, Deleuze JF, Hantraye P, Flament J, Bonnet E, Brohard S, Olaso R, Brouillet E, Carrillo-de Sauvage MA, Escartin C. Reactive astrocytes promote proteostasis in Huntington’s disease through the JAK2-STAT3 pathway. Brain. 2023 Jan 5;146(1):149-166. doi: 10.1093/brain/awac068. PMID: 35298632
- Achenbach J, Stodt B, Saft C. Factors Influencing the Total Functional Capacity Score as a Critical Endpoint in Huntington’s Disease Research. Biomedicines. 2023 Dec 17;11(12):3336. doi: 10.3390/biomedicines11123336. PMID: 38137557; PMCID: PMC10741795.
- Almeida LM, Oliveira Â, Oliveira JMA, Pinho BR. Stress response mechanisms in protein misfolding diseases: Profiling a cellular model of Huntington’s disease. Arch Biochem Biophys. 2023 Sep 1;745:109711. doi: 10.1016/j.abb.2023.109711. Epub 2023 Aug 2. PMID: 37541563.
- Amini E, Rohani M, Habibi SAH, Azad Z, Yazdi N, Cubo E, Hummel T, Jalessi M. Underestimated Olfactory Domains in Huntington’s Disease: Odor Discrimination and Threshold. J Laryngol Otol. 2023 Jul 20:1-21. doi: 10.1017/S002221512300124X. Epub ahead of print. PMID: 37470108.
- Aracil-Bolaños I, Pérez-Pérez J, Martínez-Horta S, Horta-Barba A, Puig-Davi A, García-Cornet J, Olmedo-Saura G, Campolongo A, Pagonabarraga J, Kulisevsky J. Baseline Large-Scale Network Dynamics Associated with Disease Progression in Huntington’s Disease. Mov Disord. 2023 Dec 26. doi: 10.1002/mds.29655. Epub ahead of print. PMID: 38148511.
- Ayyildiz D, Bergonzoni G, Monziani A, Tripathi T, Döring J, Kerschbamer E, Di Leva F, Pennati E, Donini L, Kovalenko M, Zasso J, Conti L, Wheeler VC, Dieterich C, Piazza S, Dassi E, Biagioli M. CAG repeat expansion in the Huntington’s disease gene shapes linear and circular RNAs biogenesis. PLoS Genet. 2023 Oct 13;19(10):e1010988. doi: 10.1371/journal.pgen.1010988. PMID: 37831730; PMCID: PMC10617732.
- Bauer S, Chen CY, Jonson M, Kaczmarczyk L, Magadi SS, Jackson WS. Cerebellar granule neurons induce Cyclin D1 before the onset of motor symptoms in Huntington’s disease mice. Acta Neuropathol Commun. 2023 Jan 20;11(1):17. doi: 10.1186/s40478-022-01500-x. PMID: 36670467; PMCID: PMC9854201
- Bergh S, Gabery S, Tonetto S, Kirik D, Petersén Å, Cheong RY. Effects of mutant huntingtin in oxytocin neurons on non-motor features of Huntington’s disease. Neuropathol Appl Neurobiol. 2023 Apr;49(2):e12891. doi: 10.1111/nan.12891. PMID: 36776123
- Bergh S, Gabery S, Tonetto S, Kirik D, Petersén Å, Cheong RY. Effects of mutant huntingtin in oxytocin neurons on non-motor features of Huntington’s disease. Neuropathol Appl Neurobiol. 2023 Feb 12:e12891. doi: 10.1111/nan.12891. Epub ahead of print. PMID: 36776123
- Birolini G, Valenza M, Ottonelli I, Talpo F, Minoli L, Cappelleri A, Bombaci M, Caccia C, Canevari C, Trucco A, Leoni V, Passoni A, Favagrossa M, Nucera MR, Colombo L, Paltrinieri S, Bagnati R, Duskey JT, Caraffi R, Vandelli MA, Taroni F, Salmona M, Scanziani E, Biella G, Ruozi B, Tosi G, Cattaneo E. Chronic cholesterol administration to the brain supports complete and longlasting cognitive and motor amelioration in Huntington’s disease. Pharmacol Res. 2023 Jun 17:106823. doi: 10.1016/j.phrs.2023.106823. Epub ahead of print. PMID: 37336430
- Björkqvist M.Centrally and peripherally altered glucose transporters: is it time to revisit energy deficiency as a potential treatment strategy in Huntington’s disease? EBioMedicine. 2023 Dec;98:104882. doi: 10.1016/j.ebiom.2023.104882. Epub 2023 Nov 17. PMID: 37979315.
- Boersema-Wijma DJ, van Duijn E, Heemskerk AW, van der Steen JT, Achterberg WP. Palliative care in advanced Huntington’s disease: a scoping review. BMC Palliat Care. 2023 May 3;22(1):54. doi: 10.1186/s12904-023-01171-y. Erratum in: BMC Palliat Care. 2023 May 31;22(1):63. PMID: 37138329; PMCID: PMC10155365
- Bonassi G, Semprini M, Mandich P, Trevisan L, Marchese R, Lagravinese G, Barban F, Pelosin E, Chiappalone M, Mantini D, Avanzino L. Neural oscillations modulation during working memory in pre-manifest and early Huntington’s disease. Brain Res. 2023 Dec 1;1820:148540. doi: 10.1016/j.brainres.2023.148540. Epub 2023 Aug 19. PMID: 37598900.
- Bragina EY, Gomboeva DE, Saik OV, Ivanisenko VA, Freidin MB, Nazarenko MS, Puzyrev VP. Apoptosis Genes as a Key to Identification of Inverse Comorbidity of Huntington’s Disease and Cancer. Int J Mol Sci. 2023 May 27;24(11):9385. doi: 10.3390/ijms24119385. PMID: 37298337; PMCID: PMC10253782
- Buchanan DA, Brown AE, Osigwe EC, Pfalzer AC, Mann LG, Yan Y, Kang H, Claassen DO. Racial Differences in the Presentation and Progression of Huntington’s Disease. Mov Disord. 2023 Oct;38(10):1945-1949. doi: 10.1002/mds.29536. Epub 2023 Aug 10. PMID: 37559498.
- Burgunder JM. Mechanisms underlying phenotypic variation in neurogenetic disorders. Nat Rev Neurol. 2023 Jun;19(6):363-370. doi: 10.1038/s41582-023-00811-4. Epub 2023 May 18. PMID: 37202496
- Burtscher J, Pepe G, Maharjan N, Riguet N, Di Pardo A, Maglione V, Millet GP. Sphingolipids and impaired hypoxic stress responses in Huntington disease. Prog Lipid Res. 2023 Mar 8;90:101224. doi: 10.1016/j.plipres.2023.101224. Epub ahead of print. PMID: 36898481
- Chen S, Zhang H, Yu J, Cao X, Zhang S, Dong D. Economic Burden of Huntington’s Disease in China: Results from a National Wide Cross-Sectional Study. Neuroepidemiology. 2023 Dec 22. doi: 10.1159/000534564. Epub ahead of print. PMID: 38142687.
- Cheng YF, Liu KC, Yang TM, Xiao Y, Jiang QR, Huang JX, Zhang S, Wei QQ, Ou RW, Li CY, Gu XJ, Burgunder JM, Shang HF. Factors influencing cognitive function in patients with Huntington’s disease from China: A cross-sectional clinical study. Brain Behav. 2023 Nov;13(11):e3258. doi: 10.1002/brb3.3258. Epub 2023 Oct 17. PMID: 37849450; PMCID: PMC10636378.
- Christodoulou CC, Papanicolaou EZ. Integrated Bioinformatics Analysis of Shared Genes, miRNA, Biological Pathways and Their Potential Role as Therapeutic Targets in Huntington’s Disease Stages. Int J Mol Sci. 2023 Mar 2;24(5):4873. doi: 10.3390/ijms24054873. PMID: 36902304; PMCID: PMC10003639
- Christodoulou CC, Demetriou CA, Philippou E, Papanicolaou EZ. Investigating the Dietary Intake Using the CyFFQ Semi-Quantitative Food Frequency Questionnaire in Cypriot Huntington’s Disease Patients. Nutrients. 2023 Feb 23;15(5):1136. doi: 10.3390/nu15051136. PMID: 36904136; PMCID: PMC10005621
- Csehi R, Molnar V, Fedor M, Zsumbera V, Palasti A, Acsai K, Grosz Z, Nemeth G, Molnar MJ. The improvement of motor symptoms in Huntington’s disease during cariprazine treatment. Orphanet J Rare Dis. 2023 Dec 1;18(1):375. doi: 10.1186/s13023-023-02930-z. PMID: 38041194; PMCID: PMC10690981.
- Daemen MMJ, Boots LMM, Oosterloo M, de Vugt ME, Duits AA. Facilitators and barriers in caring for a person with Huntington’s disease: input for a remote support program. Aging Ment Health. 2023 Jul 6:1-10. doi: 10.1080/13607863.2023.2230949. Epub ahead of print. PMID: 37409463.
- Dale M, Eccles FJR, Melvin K, Khan Z, Jones L, Zarotti N, Kiani R, Johnson J, Wells R, Simpson J. Guided self-help for anxiety among Huntington’s disease gene expansion carriers (GUIDE-HD) compared to treatment as usual: a randomised controlled feasibility trial. Pilot Feasibility Stud. 2023 Sep 12;9(1):159. doi: 10.1186/s40814-023-01364-5. PMID: 37700320; PMCID: PMC10496323.
- Dash D, Mestre TA. Motor band sign in a Huntington disease phenocopy. Parkinsonism Relat Disord. 2023 Apr;109:105333. doi: 10.1016/j.parkreldis.2023.105333. Epub 2023 Feb 20. PMID: 36854213
- Dash D, Mestre TA. Motor band sign in a Huntington disease phenocopy. Parkinsonism Relat Disord. 2023 Feb 20;109:105333. doi: 10.1016/j.parkreldis.2023.105333. Epub ahead of print. PMID: 36854213
- Dickson E, Fryklund C, Soylu-Kucharz R, Sjögren M, Stenkula KG, Björkqvist M. Altered Adipocyte Cell Size Distribution Prior to Weight Loss in the R6/2 Model of Huntington’s Disease. J Huntingtons Dis. 2023 Sep 12. doi: 10.3233/JHD-230587. Epub ahead of print. PMID: 37718850.
- Dusek P, Kopal A, Brichova M, Roth J, Ulmanova O, Klempir J, Preiningerova JL. Is retina affected in Huntington’s disease? Is optical coherence tomography a good biomarker? PLoS One. 2023 Feb 24;18(2):e0282175. doi: 10.1371/journal.pone.0282175. PMID: 36827300; PMCID: PMC9955964
- Ekkel MR, Veenhuizen RB, van Loon AM, Depla MFIA, Verschuur EML, Onwuteaka-Philipsen BD, Hertogh CMPM. Nursing home residents with Huntington’s disease: Heterogeneity in characteristics and functioning. Brain Cogn. 2023 Jul;169:106002. doi: 10.1016/j.bandc.2023.106002. Epub 2023 Jun 1. PMID: 37269816
- Espina M, Di Franco N, Brañas-Navarro M, Navarro IR, Brito V, Lopez-Molina L, Costas-Insua C, Guzmán M, Ginés S. The GRP78-PERK axis contributes to memory and synaptic impairments in Huntington’s disease R6/1 mice. Neurobiol Dis. 2023 Aug;184:106225. doi: 10.1016/j.nbd.2023.106225. Epub 2023 Jul 11.PMID: 37442396.
- Estevez-Fraga C, Altmann A, Parker CS, Scahill RI, Costa B, Chen Z, Manzoni C, Zarkali A, Durr A, Roos RAC, Landwehrmeyer B, Leavitt BR, Rees G, Tabrizi SJ, McColgan P. Genetic topography and cortical cell loss in Huntington’s disease link development and neurodegeneration. Brain. 2023 Nov 2;146(11):4532-4546. . PMID: 37587097; PMCID: PMC10629790.
- Estevez-Fraga C, Elmalem MS, Papoutsi M, Durr A, Rees EM, Hobbs NZ, Roos RAC, Landwehrmeyer B, Leavitt BR, Langbehn DR, Scahill RI, Rees G, Tabrizi SJ, Gregory S. Progressive alterations in white matter microstructure across the timecourse of Huntington’s disease. Brain Behav. 2023 Mar 14:e2940. doi: 10.1002/brb3.2940. Epub ahead of print. PMID: 36917716
- Fahy N, Rice C, Lahiri N, Desai R, Stott J. Genetic risk for Huntington Disease and reproductive decision-making: A systematic review. Clin Genet. 2023 Apr 24. doi: 10.1111/cge.14345. Epub ahead of print. PMID: 37095632
- Faquih TO, Aziz NA, Gardiner SL, Li-Gao R, de Mutsert R, Milaneschi Y, Trompet S, Jukema JW, Rosendaal FR, van Hylckama Vlieg A, van Dijk KW, MookKanamori DO. Normal range CAG repeat size variations in the HTT gene are associated with an adverse lipoprotein profile partially mediated by body mass index. Hum Mol Genet. 2023 May 5;32(10):1741-1752. doi: 10.1093/hmg/ddad020. PMID: 36715614
- Faquih TO, Aziz NA, Gardiner SL, Li-Gao R, de Mutsert R, Milaneschi Y, Trompet S, Jukema JW, Rosendaal FR, Hylckama Vlieg A, Dijk KW, Mook-Kanamori DO. Normal range CAG repeat size variations in the HTT gene are associated with an adverse lipoprotein profile partially mediated by body mass index. Hum Mol Genet. 2023 Jan 30:ddad020. doi: 10.1093/hmg/ddad020. Epub ahead of print. PMID: 36715614
- Feleus S, van der Lee M, Swen JJ, Roos RAC, de Bot ST. Study protocol of the HD-MED study aiming to personalize drug treatment in Huntington’s disease: a longitudinal, observational study to assess medication use and efficacy in relation to pharmacogenetics. Ther Adv Rare Dis. 2023 Nov 8;4:26330040231204643. doi: 10.1177/26330040231204643. PMID: 37955016; PMCID: PMC10634258.
- Ferguson R, Tabrizi SJ. Can MSH3 lowering stop HTT repeat expansion in its CAG tract? Mol Ther. 2023 Jun 7;31(6):1509-1511. doi: 10.1016/j.ymthe.2023.05.010. Epub 2023 May 25. PMID: 37236185; PMCID: PMC10277920
- Ferrari Bardile C, Radulescu CI, Pouladi MA. Oligodendrocyte pathology in Huntington’s disease: from mechanisms to therapeutics. Trends Mol Med. 2023 Oct;29(10):802-816. . Epub 2023 Aug 15. PMID: 37591764
- Festa BP, Siddiqi FH, Jimenez-Sanchez M, Won H, Rob M, Djajadikerta A, Stamatakou E, Rubinsztein DC. Microglial-to-neuronal CCR5 signaling regulates autophagy in neurodegeneration. Neuron. 2023 Apr 18:S0896-6273(23)00268-4. doi: 10.1016/j.neuron.2023.04.006. Epub ahead of print. PMID: 37105172
- Fisher A, Lavis A, Greenfield S, Rickards H. What does social cognition look like in everyday social functioning in Huntington’s disease? A protocol for a scoping review to explore and synthesise knowledge about social cognition alongside day-to-day social functioning of people with Huntington’s disease. BMJ Open. 2023 Jul 14;13(7):e073655. doi: 10.1136/bmjopen-2023-073655. PMID: 37451719; PMCID: PMC10351301.
- Garcia-Forn M, Castany-Pladevall C, Golbano A, Pérez-Pérez J, Brito V, Kulisevsky J, Pérez-Navarro E. Lamin B1 and nuclear morphology in peripheral cells as new potential biomarkers to follow treatment response in Huntington’s disease. Clin Transl Med. 2023 Feb;13(2):e1154. doi: 10.1002/ctm2.1154. PMID: 36781300; PMCID: PMC9925371
- Goldman JS, Uhlmann WR, Naini AB, Klitzman RL, Marder KS. Genetic Testing of HTT Modifiers for Huntington’s Disease: Considerations for Clinical Guidelines. Mov Disord. 2023 Dec;38(12):2151-2154. doi: 10.1002/mds.29650. Epub 2023 Nov 17. PMID: 37975739.
- Gunn S, Dale M, Ovaska-Stafford N, Maltby J. Mental health symptoms among those affected by Huntington’s disease: A cross-sectional study. Brain Behav. 2023 Mar 6:e2954. doi: 10.1002/brb3.2954. Epub ahead of print. PMID: 36880126
- Hamilton J, Farag M, Tabrizi SJ. Complementary insights into corticostriatal synapse loss and cognition in Huntington’s disease. Cell Rep Med. 2023 Dec 19;4(12):101314. doi: 10.1016/j.xcrm.2023.101314. PMID: 38118416; PMCID: PMC10772369.
- Hamilton JL, Mills JA, Stebbins GT, Long JD, Fuller RLM, Sathe S, Roché M, Sampaio C. Defining Clinical Meaningfulness in Huntington’s Disease. Mov Disord. 2023 May 5. doi: 10.1002/mds.29394. Epub ahead of print. PMID: 37147862.
- Heim B, Seppi K. Valbenazine as treatment for Huntington’s disease chorea. Lancet Neurol. 2023 Jun;22(6):459-460. doi: 10.1016/S1474-4422(23)00163-1. PMID: 37210087
- Heinzmann A, Sayah S, Lejeune FX, Hahn V, Teichmann M, Monin ML, Marchionni E, Gérard F, Charles P, Pariente J, Durr A. Huntington’s Disease with Small CAG Repeat Expansions. Mov Disord. 2023 Jun 8. doi: 10.1002/mds.29427. Epub ahead of print. PMID: 37288993
- Hett K, Eisma JJ, Hernandez AB, McKnight CD, Song A, Elenberger J, Considine C, Donahue MJ, Claassen DO. Cerebrospinal Fluid Flow in Patients with Huntington’s Disease. Ann Neurol. 2023 Nov;94(5):885-894. doi: 10.1002/ana.26749. Epub 2023 Aug 7. PMID: 37493342; PMCID: PMC10615133 (available on 2024-11-01).
- Holley SM, Reidling JC, Cepeda C, Wu J, Lim RG, Lau A, Moore C, Miramontes R, Fury B, Orellana I, Neel M, Coleal-Bergum D, Monuki ES, Bauer G, Meshul CK, Levine MS, Thompson LM. Transplanted human neural stem cells rescue phenotypes in zQ175 Huntington’s disease mice and innervate the striatum. Mol Ther. 2023 Dec 6;31(12):3545-3563. doi: 10.1016/j.ymthe.2023.10.003. Epub 2023 Oct 7. PMID: 37807512; PMCID: PMC10727970 (available on 2024-12-06) .
- Horta-Barba A, Martinez-Horta S, Pérez-Pérez J, Puig-Davi A, de Lucia N, de Michele G, Salvatore E, Kehrer S, Priller J, Migliore S, Squitieri F, Castaldo A, Mariotti C, Mañanes V, Lopez-Sendon JL, Rodriguez N, Martinez-Descals A, Júlio F, Januário C, Delussi M, de Tommaso M, Noguera S, Ruiz-Idiago J, Sitek EJ, Wallner R, Nuzzi A, Pagonabarraga J, Kulisevsky J; Cognitive Phenotype Working Group of the European Huntington’s Disease Network. Measuring cognitive impairment and monitoring cognitive decline in Huntington’s disease: a comparison of assessment instruments. J Neurol. 2023 Nov;270(11):5408-5417. doi: 10.1007/s00415-023-11804-0. Epub 2023 Jul 18. PMID: 37462754; PMCID: PMC10576674.
- Horta-Barba A, Martinez-Horta S, Sampedro F, Pérez-Pérez J, Pagonabarraga J, Kulisevsky J. Structural and metabolic brain correlates of arithmetic word-problem solving in Huntington’s disease. J Neurosci Res. 2023 Feb 19. doi:10.1002/jnr.25174. Epub ahead of print. PMID: 36807154
- Joachimiak P, Ciesiołka A, Kozłowska E, Świtoński PM, Figura G, Ciołak A, Adamek G, Surdyka M, Kalinowska-Pośka Ż, Figiel M, Caron NS, Hayden MR, Fiszer A. Allele-specific quantitation of ATXN3 and HTT transcripts in polyQ disease models. BMC Biol. 2023 Feb 1;21(1):17. doi: 10.1186/s12915- 023-01515-3. PMID: 36726088; PMCID: PMC9893648
- Kalia LV, Nimmo GAM, Mestre TA. Genetic Testing in Clinical Movement Disorders: A Case-Based Review. Semin Neurol. 2023 Feb 28. doi: 10.1055/s-0043-1763507. Epub ahead of print. PMID: 36854393
- Kasper J, Eickhoff SB, Caspers S, Peter J, Dogan I, Wolf RC, Reetz K, Dukart J, Orth M. Local synchronicity in dopamine-rich caudate nucleus influences Huntington’s disease motor phenotype. Brain. 2023 Feb 16:awad043. doi: 10.1093/brain/awad043. Epub ahead of print. PMID: 36795496
- Ketels MMA, Quarrell OW, Oosterloo M. Dystonia in Pediatric Huntington’s Disease; Prominent and Possibly Painful. Mov Disord Clin Pract. 2023 Aug 18;10(10):1552-1553. doi: 10.1002/mdc3.13850. PMID: 37868924; PMCID: PMC10585960.
- Kouba T, Frank W, Tykalova T, Mühlbäck A, Klempíř J, Lindenberg KS, Landwehrmeyer GB, Rusz J. Speech biomarkers in Huntington’s disease: A cross-sectional study in pre-symptomatic, prodromal and early manifest stages. Eur J Neurol. 2023 Feb 2. doi: 10.1111/ene.15726. Epub ahead of print. PMID: 36732902
- Kozel J, Školoudík D, Ressner P, Mikulčová P, Dušek P, Hanzlíková P, Dvořáčková N, Heryán T, Bártová P. Echogenicity of Brain Structures in Huntington’s Disease Patients Evaluated by Transcranial Sonography – Magnetic Resonance Fusion Imaging using Virtual Navigator and Digital Image Analysis. Ultraschall Med. 2023 May 24. English. doi: 10.1055/a-2081-1635. Epub ahead of print. PMID: 37224875
- Kozel J, Školoudík D, Ressner P, Michalčová P, Dušek P, Hanzlíková P, Dvořáčková N, Heryán T, Bártová P. Correction: Echogenicity of Brain Structures in Huntington’s Disease Patients Evaluated by Transcranial Sonography – Magnetic Resonance Fusion Imaging using Virtual Navigator and Digital Image Analysis. Ultraschall Med. 2023 Oct;44(5):e209. English. doi: 10.1055/a-2118-7343. Epub 2023 Jul 11. Erratum for: Ultraschall Med. 2023 Oct;44(5):495-502. PMID: 37494131.
- Lange J, Gillham O, Flower M, Ging H, Eaton S, Kapadia S, Neueder A, Duchen MR, Ferretti P, Tabrizi SJ. PolyQ length-dependent metabolic alterations and DNA damage drive human astrocyte dysfunction in Huntington’s disease. Prog Neurobiol. 2023 Jun;225:102448. doi: 10.1016/j.pneurobio.2023.102448. Epub 2023 Apr 5. PMID: 37023937
- Lunven M, Hernandez Dominguez K, Youssov K, Hamet Bagnou J, Fliss R, Vandendriessche H, Bapst B, Morgado G, Remy P, Schubert R, Reilmann R, Busse M, Craufurd D, Massart R, Rosser A, BachoudLévi AC. A new approach to digitized cognitive monitoring: validity of the SelfCog in Huntington’s disease. Brain Commun. 2023 Mar 6;5(2):fcad043. doi: 10.1093/braincomms/fcad043. PMID: 36938527; PMCID: PMC10018460
- Martinez-Horta S, Aracil-Bolaños I, Perez-Perez J, Perez-Carasol L, Garcia-Cornet J, Campolongo A, Aibar-Duran JA, Rodriguez-Rodriguez R, Pascual-Sedano B, Kulisevsky J. Theta/Alpha Band Suppression and Clinical Outcomes During Globus Pallidus Internus Deep Brain Stimulation in Huntington’s Disease. Mov Disord Clin Pract. 2023 Jan 10;10(3):518-520. doi: 10.1002/mdc3.13644. PMID: 36949795; PMCID: PMC10026271
- Martínez-Horta S, Perez-Perez J, Oltra-Cucarella J, Sampedro F, Horta-Barba A, Puig-Davi A, Pagonabarraga J, Kulisevsky J. Divergent cognitive trajectories in early stage Huntington’s disease: A 3-year longitudinal study. Eur J Neurol. 2023 Jul;30(7):1871-1879. doi: 10.1111/ene.15806. Epub 2023 Apr 14. PMID: 36994811
- McBride SD, Ober J, Dylak J, Schneider W, Morton AJ. Oculomotor Abnormalities in a Sheep (Ovis aries) Model of Huntington’s Disease: Towards a Biomarker for Assessing Therapeutic Efficacy. J Huntingtons Dis. 2023 Sep 11. doi: 10.3233/JHD-230584. Epub ahead of print. PMID: 37718849.
- McColgan P, Thobhani A, Boak L, Schobel SA, Nicotra A, Palermo G, Trundell D, Zhou J, Schlegel V, Sanwald Ducray P, Hawellek DJ, Dorn J, Simillion C, Lindemann M, Wheelock V, Durr A, Anderson KE, Long JD, Wild EJ, Landwehrmeyer GB, Leavitt BR, Tabrizi SJ, Doody R; GENERATION HD1 Investigators. Tominersen in Adults with Manifest Huntington’s Disease. N Engl J Med. 2023 Dec 7;389(23):2203-2205. doi: 10.1056/NEJMc2300400. PMID: 38055260.
- McLean ZL, Gao D, Correia K, Roy JCL, Shibata S, Farnum IN, Valdepenas-Mellor Z, Rapuru M, Morini E, Ruliera J, Gillis T, Lucente D, Kleinstiver BP, Lee JM, MacDonald ME, Wheeler VC, Pinto RM, Gusella JF. PMS1 as a target for splice modulation to prevent somatic CAG repeat expansion in Huntington’s disease. bioRxiv [Preprint]. 2023 Jul 27:2023.07.25.550489. doi: 10.1101/2023.07.25.550489. PMID: 37547003; PMCID: PMC10402039.
- Mendonça MCP, Sun Y, Cronin MF, Lindsay AJ, Cryan JF, O’Driscoll CM. Cyclodextrin-Based Nanoparticles for Delivery of Antisense Oligonucleotides Targeting Huntingtin. Pharmaceutics. 2023 Feb 3;15(2):520. doi: 10.3390/pharmaceutics15020520. PMID: 36839842; PMCID: PMC9965918.
- Migliore S, D’Aurizio G, Ceccarelli C, Casella M, Curcio G, Squitieri F. The validation of the Italian version of multiple sclerosis neuropsychological screening questionnaire in Huntington’s disease. Neurol Sci. 2023 Dec;44(12):4343-4348. doi: 10.1007/s10072-023-06950-2. Epub 2023 Jul 11. PMID: 37432564.
- Miguez A, Gomis C, Vila C, Monguió-Tortajada M, Fernández-García S, Bombau G, Galofré M, García-Bravo M, Sanders P, Fernández-Medina H, Poquet B, Salado-Manzano C, Roura S, Alberch J, Segovia JC, Allen ND, Borràs FE, Canals JM. Soluble mutant huntingtin drives early human pathogenesis in Huntington’s disease. Cell Mol Life Sci. 2023 Aug 3;80(8):238. doi: 10.1007/s00018-023-04882-w. PMID: 37535170; PMCID: PMC10400696.
- Miller DC, Lisowski P, Genehr C, Wanker EE, Priller J, Prigione A, Diecke S. Generation of an induced pluripotent stem cell line from a Huntington’s disease patient with a long HTT-PolyQ sequence. Stem Cell Res. 2023 Apr;68:103056. doi: 10.1016/j.scr.2023.103056. Epub 2023 Feb 26. PMID: 36863131.
- Morton AJ. Sleep and Circadian Rhythm Dysfunction in Animal Models of Huntington’s Disease. J Huntingtons Dis. 2023 Jun 9. doi: 10.3233/JHD-230574. Epub ahead of print. PMID: 37334613.
- Mühlbäck A, Mana J, Wallner M, Frank W, Lindenberg KS, Hoffmann R, Klempířová O, Klempíř J, Landwehrmeyer GB, Bezdicek O; REGISTRY investigators of the European Huntington’s Disease Network, the Enroll-HD investigators. Establishing normative data for the evaluation of cognitive performance in Huntington’s disease considering the impact of gender, age, language, and education. J Neurol. 2023 Oct;270(10):4903-4913. doi: 10.1007/s00415-023-11823-x. Epub 2023 Jun 22. PMID: 37347292; PMCID: PMC10511566.
- Mühlbӓck A, van Walsem M, Nance M, Arnesen A, Page K, Fisher A, van Kampen M, Nuzzi A, Limpert R, Fossmo HL, Cruickshank T, Veenhuizen R; Multidisciplinary Care and Treatment Working Group of the European Huntington’s Disease Network. What we don’t need to prove but need to do in multidisciplinary treatment and care in Huntington’s disease: a position paper. Orphanet J Rare Dis. 2023 Jan 30;18(1):19. doi: 10.1186/s13023-023-02622-8. PMID: 36717864; PMCID: PMC9887752.
- Mullari M, Fossat N, Skotte NH, Asenjo-Martinez A, Humphreys DT, Bukh J, Kirkeby A, Scheel TKH, Nielsen ML. Characterising the RNA-binding protein atlas of the mammalian brain uncovers RBM5 misregulation in mouse models of Huntington’s disease. Nat Commun. 2023 Jul 19;14(1):4348. doi: 10.1038/s41467-023-39936-x. PMID: 37468457; PMCID: PMC10356804.
- Murueta-Goyena A, Del Pino R, Acera M, Teijeira-Portas S, Romero D, Ayala U, Fernández-Valle T, Tijero B, Gabilondo I, Gómez Esteban JC. Retinal thickness as a biomarker of cognitive impairment in manifest Huntington’s disease. J Neurol. 2023 Apr 20. doi: 10.1007/s00415-023-11720-3. Epub ahead of print. PMID: 37079031.
- Nguyen TB, Miramontes R, Chillon-Marinas C, Maimon R, Vazquez-Sanchez S, Lau AL, McClure NR, England WE, Singha M, Stocksdale JT, Jang KH, Jung S, McKnight JI, Ho LN, Faull RLM, Steffan JS, Reidling JC, Jang C, Lee G, Cleveland DW, Lagier-Tourenne C, Spitale RC, Thompson LM. Aberrant splicing in Huntington’s disease via disrupted TDP-43 activity accompanied by altered m6A RNA modification. bioRxiv [Preprint]. 2023 Nov 2:2023.10.31.565004. doi: 10.1101/2023.10.31.565004. PMID: 37961595; PMCID: PMC10635028.
- Nopoulos S, Reasoner EE, Ogilvie AC, Killoran A, Schultz JL. Evaluating motor progression of juvenile-onset Huntington’s Disease: An Enroll-HD analysis. Parkinsonism Relat Disord. 2023 Dec 7;119:105954. doi: 10.1016/j.parkreldis.2023.105954. Epub ahead of print. PMID: 38142629; PMCID: PMC10903276.
- O’Reilly D, Belgrad J, Ferguson C, Summers A, Sapp E, McHugh C, Mathews E, Boudi A, Buchwald J, Ly S, Moreno D, Furgal R, Luu E, Kennedy Z, Hariharan V, Monopoli K, Yang XW, Carroll J, DiFiglia M, Aronin N, Khvorova A. Di-valent siRNA-mediated silencing of MSH3 blocks somatic repeat expansion in mouse models of Huntington’s disease. Mol Ther. 2023 Nov 1;31(11):3355-3356. doi: 10.1016/j.ymthe.2023.09.016. Epub 2023 Sep 25. Erratum for: Mol Ther. 2023 Jun 7;31(6):1661-1674. PMID: 37751745; PMCID: PMC10638064 (available on 2024-11-01) .
- Ouwerkerk J, Feleus S, van der Zwaan KF, Li Y, Roos M, van Roon-Mom WMC, de Bot ST, Wolstencroft KJ, Mina E. Machine learning in Huntington’s disease: exploring the Enroll-HD dataset for prognosis and driving capability prediction. Orphanet J Rare Dis. 2023 Jul 27;18(1):218. doi: 10.1186/s13023-023-02785-4. PMID: 37501188; PMCID: PMC10375780.
- Owen NE, Barker RA, Voysey ZJ. Sleep Dysfunction in Huntington’s Disease: Impacts of current medications and prospects for treatment. J Huntingtons Dis. 2023 May 23. doi: 10.3233/JHD-230567. Epub ahead of print. PMID: 37248911
- Piechota M, Latoszek E, Liszewska E, Hansíková H, Klempíř J, Mühlbäck A, Landwehrmeyer GB, Kuźnicki J, Czeredys M. Generation of two human iPSC lines from dermal fibroblasts of adult- and juvenile-onset Huntington’s disease patients and two healthy donors. Stem Cell Res. 2023 Sep;71:103194. doi: 10.1016/j.scr.2023.103194. Epub 2023 Aug 25. PMID: 37651831.
- Ponomareva NV, Klyushnikov SA, Abramycheva N, Konovalov RN, Krotenkova M, Kolesnikova E, Malina D, Urazgildeeva G, Kanavets E, Mitrofanov A, Fokin V, Rogaev E, Illarioshkin SN. Neurophysiological hallmarks of Huntington’s disease progression: an EEG and fMRI connectivity study. Front Aging Neurosci. 2023 Dec 15;15:1270226. doi: 10.3389/fnagi.2023.1270226. PMID: 38161585; PMCID: PMC10755012.
- Prange S, Laurencin C, Roche P, Quadrio I, Thobois S. PSP-Richardson’s Syndrome as a Rare Phenotypic Expression of Very Late-Onset Huntington’s Disease: A Case Report. Mov Disord Clin Pract. 2023 Dec 21. doi: 10.1002/mdc3.13943. Epub ahead of print. PMCID: PMC10928328; PMID: 38173343.
- Regio S, Vachey G, Goñi E, Duarte F, Rybarikova M, Sipion M, Rey M, Huarte M, Déglon N. Revisiting the outcome of adult wild-type Htt inactivation in the context of HTT-lowering strategies for Huntington’s disease. Brain Commun. 2023 Dec 7;5(6):fcad344. doi: 10.1093/braincomms/fcad344. PMID: 38116140; PMCID: PMC10729863.
- Rodríguez-Urgellés E, Casas-Torremocha D, Sancho-Balsells A, Ballasch I, García-García E, Miquel-Rio L, Manasanch A, Del Castillo I, Chen W, Pupak A, Brito V, Tornero D, Rodríguez MJ, Bortolozzi A, Sanchez-Vives MV, Giralt A, Alberch J. Thalamic Foxp2 regulates output connectivity and sensory-motor impairments in a model of Huntington’s Disease. Cell Mol Life Sci. 2023 Nov 21;80(12):367. doi: 10.1007/s00018-023-05015-z. PMID: 37987826; PMCID: PMC10663254.
- Roussakis AA, Gennaro M, Gordon MF, Reilmann R, Borowsky B, Rynkowski G, Lao-Kaim NP, Papoutsou Z, Savola JM, Hayden MR, Owen DR, Kalk N, Lingford-Hughes A, Gunn RN, Searle G, Tabrizi SJ, Piccini P. A PET-CT study on neuroinflammation in Huntington’s disease patients participating in a randomized trial with laquinimod. Brain Commun. 2023 Apr 3;5(2):fcad084. doi: 10.1093/braincomms/fcad084. PMID: 37020532; PMCID: PMC10069663
- Ruiz de Sabando A, Urrutia Lafuente E, Galbete A, Ciosi M, García Amigot F, García Solaesa V; Spanish HD Collaborative group; Monckton DG, Ramos-Arroyo MA. Spanish HTT gene study reveals haplotype and allelic diversity with possible implications for germline expansion dynamics in Huntington disease. Hum Mol Genet. 2023 Mar 6;32(6):897-906. doi: 10.1093/hmg/ddac224. PMID: 36130218; PMCID: PMC9990985
- Saft C, Burgunder JM, Dose M, Jung HH, Katzenschlager R, Priller J, Nguyen HP, Reetz K, Reilmann R, Seppi K, Landwehrmeyer GB. Symptomatic treatment options for Huntington’s disease (guidelines of the German Neurological Society). Neurol Res Pract. 2023 Nov 16;5(1):61. doi: 10.1186/s42466-023-00285-1. PMID: 37968732; PMCID: PMC10652593.
- Saher O, Zaghloul EM, Umek T, Hagey DW, Mozafari N, Danielsen MB, Gouda AS, Lundin KE, Jørgensen PT, Wengel J, Smith CIE, Zain R. Chemical Modifications and Design Influence the Potency of Huntingtin Anti-Gene Oligonucleotides. Nucleic Acid Ther. 2023 Feb 3. doi: 10.1089/nat.2022.0046. Epub ahead of print. PMID: 36735581
- Santarelli S, Londero C, Soldano A, Candelaresi C, Todeschini L, Vernizzi L, Bellosta P. Drosophila melanogaster as a model to study autophagy in neurodegenerative diseases induced by proteinopathies. Front Neurosci. 2023 May 18;17:1082047. doi: 10.3389/fnins.2023.1082047. PMID: 37274187; PMCID: PMC10232775
- Sap KA, Geijtenbeek KW, Schipper-Krom S, Guler AT, Reits EA. Ubiquitin-modifying enzymes in Huntington’s disease. Front Mol Biosci. 2023 Feb 8;10:1107323. doi: 10.3389/fmolb.2023.1107323. PMID: 36926679; PMCID: PMC10013475
- Schellino R, Besusso D, Parolisi R, Gómez-González GB, Dallere S, Scaramuzza L, Ribodino M, Campus I, Conforti P, Parmar M, Boido M, Cattaneo E, Buffo A. hESC-derived striatal progenitors grafted into a Huntington’s disease rat model support long-term functional motor recovery by differentiating, self-organizing and connecting into the lesioned striatum. Stem Cell Res Ther. 2023 Jul 28;14(1):189. doi: 10.1186/s13287-023-03422-4. PMID: 37507794; PMCID: PMC10386300.
- Schültke E, Pinzer BR, Stampanoni M, Harsan L, Döbrössy M. 3D Imaging of Striatal Transplants in a Small Animal Model of Huntington’s Disease. Neurol Int. 2023 Jul 24;15(3):896-907. doi: 10.3390/neurolint15030057. PMID: 37489363; PMCID: PMC10366744.
- Sierra LA, Ullman CJ, Baselga-Garriga C, Pandeya SR, Frank SA, Laganiere S. Prevalence of neurocognitive disorder in Huntington’s disease using the Enroll- HD dataset. Front Neurol. 2023 Jul 14;14:1198145. doi: 10.3389/fneur.2023.1198145. PMID: 37521291; PMCID: PMC10375015.
- Sipilä JOT, Majamaa K. Stable low prevalence of Huntington’s disease in Finland. Clin Park Relat Disord. 2023 Apr 28;8:100198. doi: 10.1016/j.prdoa.2023.100198. PMID: 37152417; PMCID: PMC10154769
- Smith EJ, Sathasivam K, Landles C, Osborne GF, Mason MA, Gomez-Paredes C, Taxy BA, Milton RE, Ast A, Schindler F, Zhang C, Duan W, Wanker EE, Bates GP. Early detection of exon 1 huntingtin aggregation in zQ175 brains by molecular and histological approaches. Brain Commun. 2023 Jan 20;5(1):fcad010. doi: 10.1093/braincomms/fcad010. PMID: 36756307; PMCID: PMC9901570
- Spick M, Hancox TPM, Chowdhury NR, Middleton B, Skene DJ, Morton AJ. Metabolomic Analysis of Plasma in Huntington’s Disease Transgenic Sheep (Ovisaries) Reveals Progressive Circadian Rhythm Dysregulation. J Huntingtons Dis. 2023 Jan 4. doi: 10.3233/JHD-220552. Epub ahead of print. PMID: 36617787
- Stöberl N, Donaldson J, Binda CS, McAllister B, Hall-Roberts H, Jones L, Massey TH, Allen ND. Mutant huntingtin confers cell-autonomous phenotypes on Huntington’s disease iPSC-derived microglia. Sci Rep. 2023 Nov 22;13(1):20477. doi: 10.1038/s41598-023-46852-z. PMID: 37993517; PMCID: PMC10665390.
- Strong M, Quarrell OW. Prevalence and Incidence of Huntington’s Disease. Mov Disord. 2023 Aug;38(8):1570-1572. doi: 10.1002/mds.29532. PMID: 37565397.
- Tan K, Alpaugh M, Ashton NJ, Chouinard S, Barker RA, Blennow K, Zetterberg H, Cicchetti F, Benedet AL. Plasma GFAP and its association with disease severity in Huntington’s disease. J Neurol. 2023 Nov 18. doi: 10.1007/s00415-023-12109-y. Epub ahead of print. PMID: 37979092.
- Thomson SB, Stam A, Brouwers C, Fodale V, Bresciani A, Vermeulen M, Mostafavi S, Petkau TL, Hill A, Yung A, Russell-Schulz B, Kozlowski P, MacKay A, Ma D, Beg MF, Evers MM, Vallès A, Leavitt BR. AAV5-miHTT-mediated huntingtin lowering improves brain health in a Huntington’s disease mouse model. Brain. 2023 Jun 1;146(6):2298-2315. doi: 10.1093/brain/awac458. PMID: 36508327; PMCID: PMC10232253
- Valor LM. Molecular Research on Huntington’s Disease. Int J Mol Sci. 2023 Feb 21;24(5):4310. doi: 10.3390/ijms24054310.
PMID: 36901739; PMCID: PMC10002123
- van Lonkhuizen PJC, Frank W, Heemskerk AW, van Duijn E, de Bot ST, Mühlbäck A, Landwehrmeyer GB, Chavannes NH, Meijer E; HEALTHE-RND consortium. Quality of life, health-related quality of life, and associated factors in Huntington’s disease: a systematic review. J Neurol. 2023 Jan 30. doi: 10.1007/s00415-022-11551-8. Epub ahead of print. PMID: 36715747
- van de Zande NA, Bulk M, Najac C, van der Weerd L, de Bresser J, Lewerenz J, Ronen I, de Bot ST. Study protocol of IMAGINE-HD: Imaging iron accumulation and neuroinflammation with 7T-MRI + CSF in Huntington’s disease. Neuroimage Clin. 2023 Jun 8;39:103450. doi: 10.1016/j.nicl.2023.103450. Epub ahead of print. PMID: 37327706
- Velissaris S, Davis MC, Fisher F, Gluyas C, Stout JC. A pilot evaluation of an 8-week mindfulness-based stress reduction program for people with pre- symptomatic Huntington’s disease. J Community Genet. 2023 Aug;14(4):395-405. doi: 10.1007/s12687-023-00651-1. Epub 2023 Jul 17. PMID: 37458974; PMCID: PMC10444936.
- Wennagel D, Braz BY, Humbert S. Traiter des défauts transitoires précoces des neurones dans un modèle murin de la maladie de Huntington empêche l’apparition de symptômes à l’âge adulte [Treating early transient neuronal defects in a mouse model of Huntington’s disease delays the signs of the disease in adulthood]. Med Sci (Paris). 2023 Apr;39(4):313-316. French. doi: 10.1051/medsci/2023036. Epub 2023 Apr 24. PMID: 37094259
- Wilkes FA, Jakabek D, Walterfang M, Velakoulis D, Poudel GR, Stout JC, Chua P, Egan GF, Looi JCL, Georgiou-Karistianis N. Hippocampal morphology in Huntington’s disease, implications for plasticity and pathogenesis: The IMAGE-HD study. Psychiatry Res Neuroimaging. 2023 Aug 2;335:111694. doi: 10.1016/j.pscychresns.2023.111694. Epub ahead of print. PMID: 37598529.
- Wu C, Yin H, Fu S, Yoo H, Zhang M, Park H. Altered anterograde axonal transport of mitochondria in cultured striatal neurons of a knock-in mouse model of Huntington’s disease. Biochem Biophys Res Commun. 2024 Jan 8;691:149246. doi: 10.1016/j.bbrc.2023.149246. Epub 2023 Nov 18. PMID: 38029540.
- Zsindely N, Nagy G, Siági F, Farkas A, Bodai L. Dysregulated miRNA and mRNA Expression Affect Overlapping Pathways in a Huntington’s Disease Model. Int J Mol Sci. 2023 Jul 26;24(15):11942. doi: 10.3390/ijms241511942. PMID: 37569316; PMCID: PMC10419151.
2022
- Achenbach J, Saft C, Faissner S, Ellrichmann G. Positive effect of immunomodulatory therapies on disease progression in Huntington’s disease? Data from a real-world cohort. Ther Adv Neurol Disord. 2022 Jul 23;15:17562864221109750. doi: 10.1177/17562864221109750. eCollection 2022. PMID: 35899100
- Almeida LM, Pinho BR, Duchen MR, Oliveira JMA. The PERKs of mitochondria protection during stress: insights for PERK modulation in neurodegenerative and metabolic diseases. Biol Rev Camb Philos Soc. 2022 Oct;97(5):1737-1748. doi: 10.1111/brv.12860. Epub 2022 Apr 26. PMID: 35475315
- Ananbeh H, Novak J, Juhas S, Juhasova J, Klempir J, Doleckova K, Rysankova I, Turnovcova K, Hanus J, Hansikova H, Vodicka P, Kupcova Skalnikova H. Huntingtin Co-Isolates with Small Extracellular Vesicles from Blood Plasma of TgHD and KI-HD Pig Models of Huntington’s Disease and Human Blood Plasma. Int J Mol Sci. 2022 May 17;23(10):5598. doi: 10.3390/ijms23105598. PMID: 35628406; PMCID: PMC9147436
- Andriessen RL, Oosterloo M, Hollands A, Linden DEJ, de Greef BTA, Leentjens AFG. Psychotropic medication use in Huntington’s disease: A retrospective cohort study. Parkinsonism Relat Disord. 2022 Dec;105:69-74. doi: 10.1016/j.parkreldis.2022.11.004. Epub 2022 Nov 7. PMID: 36379156
- Bakels HS, Feleus S, van Dis V, de Bot ST. More than a co-incidence? Comment on: Amyotrophic lateral sclerosis is over-represented in two Huntington’s disease brain bank cohorts: further evidence to support genetic pleiotropy of pathogenic HTT gene expansion. Acta Neuropathol. 2023 Feb;145(2):257-258. doi: 10.1007/s00401-022-02517-1. Epub 2022 Nov 6. PMID: 36335527; PMCID: PMC9849160
- Bayen E, de Langavant LC, Youssov K, Bachoud-Lévi AC. Informal care in Huntington’s disease: Assessment of objective-subjective burden and its associated risk and protective factors. Ann Phys Rehabil Med. 2022 Dec 2;66(4):101703. doi: 10.1016/j.rehab.2022.101703. Epub ahead of print. PMID: 36055643
- Bayen E, de Langavant LC, Youssov K, Lévi AB. Informal care in Huntington’s disease: assessment of objective-subjective burden and its associated risk and protective factors. Ann Phys Rehabil Med. 2022 Aug 30:101703. doi: 10.1016/j.rehab.2022.101703. Online ahead of print. PMID: 36055643
- Capizzi M, Carpentier R, Denarier E, Adrait A, Kassem R, Mapelli M, Couté Y, Humbert S. Developmental defects in Huntington’s disease show that axonal growth and microtubule reorganization require NUMA1. Neuron. 2022 Jan 5;110(1):36-50.e5. doi: 10.1016/j.neuron.2021.10.033. Epub 2021 Nov 17. PMID: 34793694
- Cheng Y, Gu X, Liu K, Yang T, Xiao Y, Jiang Q, Huang J, Lin J, Wei Q, Ou R, Hou Y, Zhang L, Li C, Burgunder JM, Shang H. The Comprehensive Analysis of Motor and Neuropsychiatric Symptoms in Patients with Huntington’s Disease from China: A Cross-Sectional Study. J Clin Med. 2022 Dec 27;12(1):206. doi: 10.3390/jcm12010206. PMID: 36615008; PMCID: PMC9821667
- Dale M, Wood A, Zarotti N, Eccles F, Gunn S, Kiani R, Mobley A, Robertson N, Simpson J. Using a Clinical Formulation to Understand Psychological Distress in People Affected by Huntington’s Disease: A Descriptive, Evidence-Based Model. J Pers Med. 2022 Jul 27;12(8):1222. doi: 10.3390/jpm12081222. PMID: 35893316
- De Paepe AE, Garcia-Gorro C, Martinez-Horta S, Perez JP, Kulisevsky J, Rodriguez-Dechicha N, Vaquer I, Subira S, Calopa M, Santacruz P, Muñoz E, Mareca C, Ruiz-Idiago J, de Diego-Balaguer R, Camara E. Delineating apathy profiles in Huntington’s disease with the short-Lille Apathy Rating Scale. Parkinsonism Relat Disord. 2022 Dec;105:83-89. doi: 10.1016/j.parkreldis.2022.10.025. Epub 2022 Oct 28. PMID: 36395542
- Dickson E, Soylu-Kucharz R, Petersén Å, Björkqvist M. Hypothalamic expression of huntingtin causes distinct metabolic changes in Huntington’s disease mice. Mol Metab. 2022 Mar;57:101439. doi: 10.1016/j.molmet.2022.101439. Epub 2022 Jan 7. PMID: 35007790
- Di Tella S, Lo Monaco MR, Petracca M, Zinzi P, Solito M, Piano C, Calabresi P, Silveri MC, Bentivoglio AR. Beyond the CAG triplet number: exploring potential predictors of delayed age of onset in Huntington’s disease. J Neurol. 2022 Aug 1. doi: 10.1007/s00415-022-11297-3. Online ahead of print. PMID: 35915275
- Dickson E, Dwijesha AS, Andersson N, Lundh S, Björkqvist M, Petersén Å, Soylu-Kucharz R. Microarray profiling of hypothalamic gene expression changes in Huntington’s disease mouse models. Front Neurosci. 2022 Nov 3;16:1027269. doi: 10.3389/fnins.2022.1027269. PMID: 36408416; PMCID: PMC9671106
- Eddy CM, Rickards H. Social cognition and quality of life in Huntington’s disease. Front Psychiatry. 2022 Aug 24;13:963457. doi: 10.3389/fpsyt.2022.963457. eCollection 2022. PMID: 36090376
- Ekkel MR, Depla MFIA, Verschuur EML, Veenhuizen RB, Hertogh CMPM, Onwuteaka- Philipsen BD. Patient perspectives on advance euthanasia directives in Huntington’s disease. A qualitative interview study. BMC Med Ethics. 2022 Oct 10;23(1):101. doi: 10.1186/s12910-022-00838-0. PMCID: PMC9552411; PMID: 36217136
- Estevez-Fraga C, Tabrizi SJ. Disentangling the Connection Between Neurodevelopment and Neurodegeneration in Huntington’s Disease. Mov Disord. 2022 Dec;37(12):2343-2344. doi: 10.1002/mds.29267. Epub 2022 Oct 29. PMID: 36308728
- Fão L, Coelho P, Duarte L, Vilaça R, Hayden MR, Mota SI, Rego AC. Restoration of c-Src/Fyn Proteins Rescues Mitochondrial Dysfunction in Huntington’s Disease. Antioxid Redox Signal. 2022 Aug 5. doi: 10.1089/ars.2022.0001. Online ahead of print. PMID: 35651273
- Feleus S, van Schaijk M, Roos RAC, de Bot ST. The Many Faces of Huntington’s Chorea Treatment: The Impact of Sudden Withdrawal of Tiapride after 40 Years of Use and a Systematic Review. J Pers Med. 2022 Apr 6;12(4):589. doi: 10.3390/jpm12040589. PMID: 35455705; PMID: PMC9025785
- Fienko S, Landles C, Sathasivam K, McAteer SJ, Milton RE, Osborne GF, Smith EJ, Jones ST, Bondulich MK, Danby ECE, Phillips J, Taxy BA, Kordasiewicz HB, Bates GP. Alternative processing of human HTT mRNA with implications for Huntington’s disease therapeutics. Brain. 2022 Jul 6:awac241. doi: 10.1093/brain/awac241. Online ahead of print. PMID: 35793238
- Fienko S, Landles C, Sathasivam K, McAteer SJ, Milton RE, Osborne GF, Smith EJ, Jones ST, Bondulich MK, Danby ECE, Phillips J, Taxy BA, Kordasiewicz HB, Bates GP. Alternative processing of human HTT mRNA with implications for Huntington’s disease therapeutics. Brain. 2022 Dec 19;145(12):4409-4424. doi: 10.1093/brain/awac241. PMID: 35793238; PMCID: PMC9762945
- Fritz NE, Busse M, Muratori LM, Rao AK, Kloos A, Kegelmeyer D, Quinn L. An MDS Evidence-Based Review on Treatments for Huntington’s Disease. Mov Disord. 2022 Jul;37(7):1566-1567. doi: 10.1002/mds.29059. PMID: 35856726 Review.
- Furby H, Moore S, Nordstroem AL, Houghton R, Lambrelli D, Graham S, Svenningsson P, Petersén Å. Comorbidities and clinical outcomes in adult- and juvenile-onset Huntington’s disease: a study of linked Swedish National Registries (2002-2019). J Neurol. 2022 Oct 18. doi: 10.1007/s00415-022-11418-y. Epub ahead of print. PMID: 36253622
- Gallezot C, Riad R, Titeux H, Lemoine L, Montillot J, Sliwinski A, Bagnou JH, Cao XN, Youssov K, Dupoux E, Bachoud Levi AC. Emotion expression through spoken language in Huntington disease. Cortex. 2022 Jul 19;155:150-161. doi: 10.1016/j.cortex.2022.05.024. Online ahead of print. PMID: 35986957
- Galyan SM, Ewald CY, Jalencas X, Masrani S, Meral S, Mestres J. Fragment- based virtual screening identifies a first-in-class preclinical drug candidate for Huntington’s disease. Sci Rep. 2022 Nov 16;12(1):19642. doi: 10.1038/s41598-022-21900-2. PMID: 36385140; PMCID: PMC9668931
- Gamez J, Calopa M, Muñoz E, Ferré A, Huertas O, McAllister K, Reig N, Scart- Grès C, Insa R, Kulisevsky J. A proof-of-concept study with SOM3355 (bevantolol hydrochloride) for reducing chorea in Huntington’s disease. Br J Clin Pharmacol. 2022 Dec 9. doi: 10.1111/bcp.15635. Epub ahead of print. PMID: 36494329
- Grigor’eva EV, Malakhova AA, Sorogina DA, Pavlova SV, Malankhanova TB, Abramycheva NY, Klyushnikov SA, Illarioshkin SN, Zakian SM. Generation of induced pluripotent stem cell line, ICGi033-A, by reprogramming peripheral blood mononuclear cells from a patient with Huntington’s disease. Stem Cell Res. 2022 Aug;63:102868. doi: 10.1016/j.scr.2022.102868. Epub 2022 Jul 13. PMID: 35872525
- Hamilton MJ, Atalaia A, McLean J, Cumming SA, Evans JJ, Ballantyne B, Jampana R, The Scottish Myotonic Dystrophy Consortium, Longman C, Livingston E, van der Plas E, Koscik T, Nopoulos P, Farrugia ME, Monckton DG. Clinical and neuroradiological correlates of sleep in myotonic dystrophy type 1. Neuromuscul Disord. 2022 May;32(5):377-389. doi: 10.1016/j.nmd.2022.02.003. Epub 2022 Feb 14. PMID: 35361525
- Hare E, Bachoud-Lévi AC, Reilmann R, Craufurd D, Busse M, Rosser A, McLauchlan D. Cognitive processes of apathy in Huntington’s disease show high sensitivity to disease progression. Clin Park Relat Disord. 2022 Oct 26;7:100168. doi: 10.1016/j.prdoa.2022.100168. PMID: 36405870; PMCID: PMC9673112
- Harris KL, Mason SL, Barker RA. Exploring the predictors of financial impairment in Huntington’s disease using the Enroll-HD dataset. J Neurol. 2022 May;32(5):377-389. doi: 10.1016/j.nmd.2022.02.003. Epub 2022 Feb 14. PMID: 35165768
- Hassan YR, Brogueira Rodrigues F, Zeun P, Byrne LM, Estevez-Fraga C, Tortelli R, Scahill RI, Wild EJ, Tabrizi SJ. Lumbar puncture safety and tolerability in premanifest and manifest Huntington’s disease: a multi-analysis cross-sectional study. Sci Rep. 2022 Nov 1;12(1):18377. doi: 10.1038/s41598-022-21934-6. PMID: 36319718; PMCID: PMC9626630
- Hendel RK, Hellem MNN, Hjermind LE, Nielsen JE, Vogel A. On the association between apathy and deficits of social cognition and executive functions in Huntington’s disease. J Int Neuropsychol Soc. 2022 Oct 3:1-8. doi: 10.1017/S1355617722000364. Epub ahead of print. PMID: 36189712
- Hendel RK, Hellem MNN, Hjermind LE, Nielsen JE, Vogel A. An Exploratory Study Investigating Autonomy in Huntington’s Disease Gene Expansion Carriers. J Huntingtons Dis. 2022 Aug 6. doi: 10.3233/JHD-220540. PMID: 35964199
- Horta-Barba A, Martínez-Horta S, Pérez-Pérez J, Sampedro F, Puig-Davi A, Pagonabarraga J, Kulisevsky J. Measuring the functional impact of cognitive impairment in Huntington’s disease. J Neurol. 2022 Jul;269(7):3541-3549. doi: 10.1007/s00415-021-10955-2. Epub 2022 Jan 21. PMID: 35061089
- Hubčíková K, Rakús T, Mühlbäck A, Benetin J, Bruncvik L, Petrášová Z, Bušková J, Brunovský M. Psychosocial Impact of Huntington’s Disease and Incentives to Improve Care for Affected Families in the Underserved Region of the Slovak Republic. J Pers Med. 2022 Nov 22;12(12):1941. doi: 10.3390/jpm12121941. PMID: 36556162; PMCID: PMC9783383
- Kim H, Lenoir S, Helfricht A, Jung T, Karneva ZK, Lee Y, Beumer W, van der Horst GB, Anthonijsz H, Buil LC, van der Ham F, Platenburg GJ, Purhonen P, Hebert H, Humbert S, Saudou F, Klein P, Song JJ. A pathogenic proteolysis-resistant huntingtin isoform induced by an antisense oligonucleotide maintains huntingtin function. JCI Insight. 2022 Sep 8;7(17):e154108. doi: 10.1172/jci.insight.154108. PMID: 35943803
- Kjoelaas S, Feragen KB, Jensen TK. Social support experiences when growing up with a parent with Huntington’s disease. Health Psychol Behav Med. 2022 Jul 29;10(1):655-675. doi: 10.1080/21642850.2022.2104286. eCollection 2022. PMID: 35923579
- Kjoelaas S, Jensen TK, Feragen KB. Dilemmas when talking about Huntington’s disease: A qualitative study of offspring and caregiver experiences in Norway. J Genet Couns. 2022 Jul 29. doi: 10.1002/jgc4.1610. Online ahead of print. PMID: 35903951
- Koval I, Dighiero-Brecht T, Tobin AJ, Tabrizi SJ, Scahill RI, Tezenas du Montcel S, Durrleman S, Durr A. Forecasting individual progression trajectories in Huntington disease enables more powered clinical trials. Sci Rep. 2022 Nov 7;12(1):18928. doi: 10.1038/s41598-022-18848-8. PMID: 36344508; PMCID: PMC9640581
- Krach F, Stemick J, Boerstler T, Weiss A, Lingos I, Reischl S, Meixner H, Ploetz S, Farrell M, Hehr U, Kohl Z, Winner B, Winkler J. An alternative splicing modulator decreases mutant HTT and improves the molecular fingerprint in Huntington’s disease patient neurons. Nat Commun. 2022 Nov 10;13(1):6797. doi: 10.1038/s41467-022-34419-x. PMID: 36357392; PMCID: PMC9649613
- Krysewski LM, Power Guerra N, Glatzel A, Holzmann C, Antipova V, Schmitt O, Yu-Taeger L, Nguyen HP, Wree A, Witt M. Differential Cellular Balance of Olfactory and Vomeronasal Epithelia in a Transgenic BACHD Rat Model of Huntington’s Disease. Int J Mol Sci. 2022 Jul 10;23(14):7625. doi: 10.3390/ijms23147625. PMID: 35886975
- Langbehn DR; Registry Investigators of the European Huntington Disease Network. Longer CAG repeat length is associated with shorter survival after disease onset in Huntington disease. Am J Hum Genet. 2022 Jan 6;109(1):172-179. doi: 10.1016/j.ajhg.2021.12.002. Epub 2021 Dec 22. PMID: 34942093
- Latoszek E, Piechota M, Liszewska E, Hansíková H, Klempíř J, Mühlbäck A, Landwehrmeyer GB, Kuznicki J, Czeredys M. Generation of three human iPSC lines from patients with Huntington’s disease with different CAG lengths and human control iPSC line from a healthy donor. Stem Cell Res. 2022 Oct;64:102931. doi: 10.1016/j.scr.2022.102931. Epub 2022 Oct 3. PMID: 36228511
- Layburn FE, Tan AYS, Mehrabi NF, Curtis MA, Tippett LJ, Turner CP, Riguet N, Aeschbach L, Lashuel HA, Dragunow M, Faull RLM, Singh-Bains MK. N-terminal mutant huntingtin deposition correlates with CAG repeat length and symptom onset, but not neuronal loss in Huntington’s disease. Neurobiol Dis. 2022 Nov;174:105884. doi: 10.1016/j.nbd.2022.105884. Epub 2022 Oct 8. PMID: 36220612
- Lee JM, Huang Y, Orth M, Gillis T, Siciliano J, Hong E, Mysore JS, Lucente D, Wheeler VC, Seong IS, McLean ZL, Mills JA, McAllister B, Lobanov SV, Massey TH, Ciosi M, Landwehrmeyer GB, Paulsen JS, Dorsey ER, Shoulson I, Sampaio C, Monckton DG, Kwak S, Holmans P, Jones L, MacDonald ME, Long JD, Gusella JF. Genetic modifiers of Huntington disease differentially influence motor and cognitive domains. Am J Hum Genet. 2022 May 5;109(5):885-899. doi: 10.1016/j.ajhg.2022.03.004. Epub 2022 Mar 23. PMID: 35325614; PMCID: PMC9118113
- Lemercier P, Cleret de Langavant L, Hamet Bagnou J, Youssov K, Lemoine L, Audureau E, Massart R, Bachoud-Lévi AC. Self-Reported Social Relationship Capacities Predict Motor, Functional and Cognitive Decline in Huntington’s Disease. J Pers Med. 2022 Jan 27;12(2):174. doi: 10.3390/jpm12020174. PMID: 35207662; PMCID: PMC8879028
- Lemoine L, Lunven M, Fraisse N, Youssov K, Bapst B, Morgado G, Reilmann R, Busse M, Craufurd D, Rosser A, de Gardelle V, Bachoud-Lévi AC. The striatum in time production: The model of Huntington’s disease in longitudinal study. Neuropsychologia. 2023 Jan 28;179:108459. doi: 10.1016/j.neuropsychologia.2022.108459. Epub 2022 Dec 22. PMID: 36567007
- Lobanov SV, McAllister B, McDade-Kumar M, Landwehrmeyer GB, Orth M, Rosser AE; REGISTRY Investigators of the European Huntington’s disease network, Paulsen JS; PREDICT-HD Investigators of the Huntington Study Group, Lee JM, MacDonald ME, Gusella JF, Long JD, Ryten M, Williams NM, Holmans P, Massey TH, Jones L. Huntington’s disease age at motor onset is modified by the tandem hexamer repeat in TCERG1. NPJ Genom Med. 2022 Sep 5;7(1):53. doi: 10.1038/s41525-022-00317-w. PMID: 36064847
- Lopes C, Ferreira IL, Maranga C, Beatriz M, Mota SI, Sereno J, Castelhano J, Abrunhosa A, Oliveira F, De Rosa M, Hayden M, Laço MN, Januário C, Castelo Branco M, Rego AC. Mitochondrial and redox modifications in early stages of Huntington’s disease. Redox Biol. 2022 Aug 10;56:102424. doi: 10.1016/j.redox.2022.102424. Online ahead of print. PMID: 35988447; PMCID: PMC9420526
- Lowe AJ, Rodrigues FB, Arridge M, De Vita E, Johnson EB, Scahill RI, Byrne LM, Tortelli R, Heslegrave A, Zetterberg H, Wild EJ. Longitudinal evaluation of proton magnetic resonance spectroscopy metabolites as biomarkers in Huntington’s disease. Brain Commun. 2022 Oct 12;4(6):fcac258. doi: 10.1093/braincomms/fcac258. PMID: 36382217; PMCID: PMC9665272
- McAllister B, Donaldson J, Binda CS, Powell S, Chughtai U, Edwards G, Stone J, Lobanov S, Elliston L, Schuhmacher LN, Rees E, Menzies G, Ciosi M, Maxwell A, Chao MJ, Hong EP, Lucente D, Wheeler V, Lee JM, MacDonald ME, Long JD, Aylward EH, Landwehrmeyer GB, Rosser AE; REGISTRY Investigators of the European Huntington’s disease network, Paulsen JS; PREDICT-HD Investigators of the Huntington Study Group, Williams NM, Gusella JF, Monckton DG, Allen ND, Holmans P, Jones L, Massey TH. Exome sequencing of individuals with Huntington’s disease implicates FAN1 nuclease activity in slowing CAG expansion and disease onset. Nat Neurosci. 2022 Apr;25(4):446-457. doi: 10.1038/s41593-022-01033-5. Epub 2022 Apr 4. PMID: 35379994; PMCID: PMC8986535
- McColgan P, Gregory S, Zeun P, Zarkali A, Johnson EB, Parker C, Fayer K, Lowe J, Nair A, Estevez-Fraga C, Papoutsi M, Zhang H, Scahill RI, Tabrizi SJ, Rees G. Neurofilament light-associated connectivity in young-adult Huntington’s disease is related to neuronal genes. Brain. 2022 Nov 21;145(11):3953-3967. doi: 10.1093/brain/awac227. PMID: 35758263; PMCID: PMC9679168
- McLauchlan DJ, Lancaster T, Craufurd D, Linden DEJ, Rosser AE. Different depression: motivational anhedonia governs antidepressant efficacy in Huntington’s disease. Brain Commun. 2022 Nov 9;4(6):fcac278. doi: 10.1093/braincomms/fcac278. PMID: 36440100; PMCID: PMC9683390
- McLauchlan DJ, Linden DEJ, Rosser AE. Excessive response to provocation rather than disinhibition mediates irritable behaviour in Huntington’s disease. Front Neurosci. 2022 Dec 29;16:993357. doi: 10.3389/fnins.2022.993357. PMID: 36643017; PMCID: PMC9836783
- Merino M, Sequedo MD, Sánchez-Sánchez AV, Clares MP, García-España E, Vázquez-Manrique RP, Mullor JL. Mn(II) Quinoline Complex (4QMn) Restores Proteostasis and Reduces Toxicity in Experimental Models of Huntington’s Disease. Int J Mol Sci. 2022 Aug 11;23(16):8936. doi: 10.3390/ijms23168936. PMID: 36012207
- Migliore S, D’Aurizio G, Scaricamazza E, Maffi S, Ceccarelli C, Ristori G, Romano S, Castaldo A, Fichera M, Curcio G, Squitieri F. Cognitive Reserve in Early Manifest Huntington Disease Patients: Leisure Time Is Associated with Lower Cognitive and Functional Impairment. J Pers Med. 2022 Jan 3;12(1):36. doi: 10.3390/jpm12010036. PMID: 35055351; PMCID: PMC8777615
- Miller DC, Lisowski P, Lickfett S, Mlody B, Bünning M, Genehr C, Ulrich C, Wanker EE, Diecke S, Priller J, Prigione A. Generation of induced pluripotent stem cells from three individuals with Huntington’s disease. Stem Cell Res. 2022 Dec;65:102976. doi: 10.1016/j.scr.2022.102976. Epub 2022 Nov 17. PMID: 36434993
- Nguyen QTR, Ortigoza Escobar JD, Burgunder JM, Mariotti C, Saft C, Hjermind LE, Youssov K, Landwehrmeyer GB, Bachoud-Lévi AC. Combining Literature Review With a Ground Truth Approach for Diagnosing Huntington’s Disease Phenocopy. Front Neurol. 2022 Feb 10;13:817753. doi: 10.3389/fneur.2022.817753. eCollection 2022. PMID: 35222250, PMCID: PMC8866848
- Palermo G, Di Fonzo A, Francesconi A, Unti E, Ceravolo R. Two cases of Huntington’s disease unmasked by the COVID-19 pandemic. Neurol Sci. 2022 Dec 29:1–3. doi: 10.1007/s10072-022-06564-0. Epub ahead of print. PMID: 36580215; PMCID: PMC9797887
- Papoutsi M, Flower M, Hensman Moss DJ, Holmans P, Estevez-Fraga C, Johnson EB, Scahill RI, Rees G, Langbehn D, Tabrizi SJ; Track-HD Investigators. Intellectual enrichment and genetic modifiers of cognition and brain volume in Huntington’s disease. Brain Commun. 2022 Oct 31;4(6):fcac279. doi: 10.1093/braincomms/fcac279. PMID: 36519153; PMCID: PMC9732861
- Peikert K, Storch A, Hermann A, Landwehrmeyer GB, Walker RH, Simionato G, Kaestner L, Danek A. Commentary: Acanthocytes identified in Huntington’s disease. Front Neurosci. 2022 Nov 4;16:1049676. doi: 10.3389/fnins.2022.1049676. PMID: 36408380; PMCID: PMC9673475
- Pellegrini M, Bergonzoni G, Perrone F, Squitieri F, Biagioli M. Current Diagnostic Methods and Non-Coding RNAs as Possible Biomarkers in Huntington’s Disease. Genes (Basel). 2022 Nov 3;13(11):2017. doi: 10.3390/genes13112017. PMID: 36360254; PMCID: PMC9689996
- Pérot JB, Célestine M, Palombo M, Dhenain M, Humbert S, Brouillet E, Flament J. Longitudinal multimodal MRI characterization of a knock-in mouse model of Huntington’s disease reveals early gray and white matter alterations. Hum Mol Genet. 2022 Oct 28;31(21):3581-3596. doi: 10.1093/hmg/ddac036. PMID: 35147158; PMCID: PMC9616570
- Petry S, Keraudren R, Nateghi B, Loiselle A, Pircs K, Jakobsson J, Sephton C, Langlois M, St-Amour I, Hébert SS. Widespread alterations in microRNA biogenesis in human Huntington’s disease putamen. Acta Neuropathol Commun. 2022 Jul 22;10(1):106. doi: 10.1186/s40478-022-01407-7. PMID: 35869509; PMCID: PMC9308264
- Pircs K, Drouin-Ouellet J, Horváth V, Gil J, Rezeli M, Garza R, Grassi DA, Sharma Y, St-Amour I, Harris K, Jönsson ME, Johansson PA, Vuono R, Fazal SV, Stoker T, Hersbach BA, Sharma K, Lagerwall J, Lagerström S, Storm P, Hébert SS, Marko-Varga G, Parmar M, Barker RA, Jakobsson J. Distinct subcellular autophagy impairments in induced neurons from patients with Huntington’s disease. Brain. 2022 Sep 14;145(9):3035-3057. doi: 10.1093/brain/awab473. PMID: 34936701; PMC ID: PMC9473361
- Poletti B, Solca F, Maffi S, Torre S, Carelli L, Aiello EN, Ferrucci R, Priori A, Monti A, Verde F, Ticozzi N, Migliore S, Scaricamazza E, Casella M, Squitieri F, Ciammola A, Silani V. Semiology and determinants of apathy across neurodegenerative motor disorders: A comparison between amyotrophic lateral sclerosis, Parkinson’s and Huntington’s disease. Front Aging Neurosci. 2022 Nov 2;14:1031908. doi: 10.3389/fnagi.2022.1031908.
PMID: 36408105; PMCID: PMC9667083 - Pupak A, Singh A, Sancho-Balsells A, Alcalá-Vida R, Espina M, Giralt A, Martí E, Ørom UAV, Ginés S, Brito V. Altered m6A RNA methylation contributes to hippocampal memory deficits in Huntington’s disease mice. Cell Mol Life Sci. 2022 Jul 11;79(8):416. doi: 10.1007/s00018-022-04444-6. PMID: 35819730; PMCID: PMC9276730
- Rodrigues FB, Owen G, Sathe S, Pak E, Kaur D, Ehrhardt AG, Lifer S, Townhill J, Schubert K, Leavitt BR, Guttman M, Bang J, Lewerenz J, Levey J; HDClarity Investigators, Sampaio C, Wild EJ. Safety and Feasibility of Research Lumbar Puncture in Huntington’s Disease: The HDClarity Cohort and Bioresource. J Huntingtons Dis. 2022;11(1):59-69. doi: 10.3233/JHD-210508. PMID: 35253773
- Rodríguez-Santana I, Mestre T, Squitieri F, Willock R, Arnesen A, Clarke A, D’Alessio B, Fisher A, Fuller R, Hamilton JL, Hubberstey H, Stanley C, Vetter L, Winkelmann M, Doherty M, Wu Y, Finnegan A, Frank S. Economic burden of Huntington disease in Europe and the USA: Results from the Huntington’s Disease Burden of Illness study. Eur J Neurol. 2022 Nov 24. doi: 10.1111/ene.15645. Epub ahead of print. PMID: 36421029
- Rodríguez-Urgellés E, Rodríguez-Navarro I, Ballasch I, Del Toro D, Del Castillo I, Brito V, Alberch J, Giralt A. Postnatal Foxp2 regulates early psychiatric-like phenotypes and associated molecular alterations in the R6/1 transgenic mouse model of Huntington’s disease. Neurobiol Dis. 2022 Aug 24;173:105854. doi: 10.1016/j.nbd.2022.105854. Online ahead of print. PMID: 36029989
- Romano S, Romano C, Peconi M, Fiore A, Bellucci G, Morena E, Troili F, Cipollini V, Annibali V, Giglio S, Mechelli R, Ferraldeschi M, Veneziano L, Mantuano E, Sani G, Vecchione A, Umeton R, Giubilei F, Salvetti M, Corbo RM, Scarabino D, Ristori G. Circulating U13 Small Nucleolar RNA as a Potential Biomarker in Huntington’s Disease: A Pilot Study. Int J Mol Sci. 2022 Oct 18;23(20):12440. doi: 10.3390/ijms232012440. PMID: 36293304; PMCID: PMC9604297
- Rosser AE, Busse ME, Gray WP, Aron Badin R, Perrier AL, Wheelock V, Cozzi E, Perpiña Martin U, Salado-Manzano C, Mills LJ, Drew C, Goldman SA, Canals JM, Thompson LM. Translating cell therapies for neurodegenerative diseases: Huntington’s disease as a model disorder. Brain. Jun 3;145(5):1584-1597. doi: 10.1093/brain/awac086. PMID: 35262656; PMCID: PMC9166564
- Saft C. Huntington’s disease: disappointments and new beginnings. Lancet Neurol. 2022 Jul;21(7):582-584. doi: 10.1016/S1474-4422(22)00189-2. PMID: 35716683
- Sampedro F, Martínez-Horta S, Pérez-Pérez J, Pérez-González R, Horta-Barba A, Campolongo A, Izquierdo C, Aracil-Bolaños I, Rivas E, Puig-Davi A, Pagonabarraga J, Gómez-Ansón B, Kulisevsky J. Plasma TDP-43 Reflects Cortical Neurodegeneration and Correlates with Neuropsychiatric Symptoms in Huntington’s Disease. Clin Neuroradiol. 2022 Mar 3. doi: 10.1007/s00062-022-01150-5. Online ahead of print. PMID: 35238950
- Scarabino D, Veneziano L, Mantuano E, Arisi I, Fiore A, Frontali M, Corbo RM. Leukocyte Telomere Length as Potential Biomarker of HD Progression: A Follow-Up Study. Int J Mol Sci. 2022 Nov 3;23(21):13449. doi: 10.3390/ijms232113449.
PMID: 36362235;PMCID: PMC9654348 - Simón-Vicente L, Rivadeneyra-Posadas J, Soto-Célix M, Raya-González J, Castillo D, Calvo S, Collazo C, Rodríguez-Fernández A, Fahed VS, Mariscal N, García-Bustillo Á, Aguado L, Cubo E. Accelerometer Cut-Points for Physical Activity Assessment in Adults with Mild to Moderate Huntington’s Disease: A Cross-Sectional Multicentre Study. Int J Environ Res Public Health. 2022 Nov 11;19(22):14834. doi: 10.3390/ijerph192214834. PMID: 36429552; PMCID: PMC9690573
- Solca F, Aiello EN, Migliore S, Torre S, Carelli L, Ferrucci R, Priori A, Verde F, Ticozzi N, Maffi S, Ceccarelli C, Squitieri F, Silani V, Ciammola A, Poletti B. Diagnostic properties of the Frontal Assessment Battery (FAB) in Huntington’s disease. Front Psychol. 2022 Nov 30;13:1031871. doi: 10.3389/fpsyg.2022.1031871. PMID: 36533005; PMCID: PMC9748548
- Tabrizi SJ, Estevez-Fraga C, van Roon-Mom WMC, Flower MD, Scahill RI, Wild EJ, Muñoz-Sanjuan I, Sampaio C, Rosser AE, Leavitt BR. Potential disease-modifying therapies for Huntington’s disease: lessons learned and future opportunities. Lancet Neurol. 2022 Jul;21(7):645-658. doi: 10.1016/S1474-4422(22)00121-1. PMID: 35716694 Review
- Tomczyk M, Braczko A, Mierzejewska P, Podlacha M, Krol O, Jablonska P, Jedrzejewska A, Pierzynowska K, Wegrzyn G, Slominska EM, Smolenski RT. Rosiglitazone Ameliorates Cardiac and Skeletal Muscle Dysfunction by Correction of Energetics in Huntington’s Disease. Cells. 2022 Aug 27;11(17):2662. doi: 10.3390/cells11172662. PMID: 36078070; PMCID: PMC9454785
- van Walsem MR, Frich JC, Gómez Castañeda M, Howe EI, Pihlstrøm L, Andelic N, Aas E. Health related quality of life, service utilization and costs for patients with Huntington’s disease in Norway. BMC Health Serv Res. 2022 Dec 14;22(1):1527. doi: 10.1186/s12913-022-08881-8.
PMID: 36517848; PMCID: PMC9753307 - Vanisova M, Stufkova H, Kohoutova M, Rakosnikova T, Krizova J, Klempir J, Rysankova I, Roth J, Zeman J, Hansikova H. Mitochondrial organization and structure are compromised in fibroblasts from patients with Huntington’s disease. Ultrastruct Pathol. 2022 Aug 10:1-14. doi: 10.1080/01913123.2022.2100951. Online ahead of print. PMID: 35946926
- Wennagel D, Braz BY, Capizzi M, Barnat M, Humbert S. Huntingtin coordinates dendritic spine morphology and function through cofilin-mediated control of the actin cytoskeleton. Cell Rep. 2022 Aug 30;40(9):111261. doi: 10.1016/j.celrep.2022.111261. PMID: 36044862
- Wu J, Möhle L, Brüning T, Eiriz I, Rafehi M, Stefan K, Stefan SM, Pahnke J. A Novel Huntington’s Disease Assessment Platform to Support Future Drug Discovery and Development. Int J Mol Sci. 2022 Nov 25;23(23):14763. doi: 10.3390/ijms232314763. PMID: 36499090; PMCID: PMC9740291
- Youssov K, Audureau E, Vandendriessche H, Morgado G, Layese R, Goizet C, Verny C, Bourhis ML, Bachoud-Lévi AC. The burden of Huntington’s disease: A prospective longitudinal study of patient/caregiver pairs. Parkinsonism Relat Disord. 2022 Aug 24;103:77-84. doi: 10.1016/j.parkreldis.2022.08.023. PMID: 36084356
- Zarotti N, Dale M, Eccles FJR, Simpson J. More than Just a Brain Disorder: A Five-Point Manifesto for Psychological Care for People with Huntington’s Disease. J Pers Med. 2022 Jan 7;12(1):64. doi: 10.3390/jpm12010064. PMID: 35055379; PMCID: PMC8780585
- EHDN 2022 Plenary Meeting (September 16-18, 2022): J Neurol Neurosurg Psychiatry 2022 93(Suppl 1)
2021
- Achenbach J, Saft C. Another Perspective on Huntington’s Disease: Disease Burden in Family Members and Pre-Manifest HD When Compared to Genotype-Negative Participants from ENROLL-HD. Brain Sci. 2021 Dec 8;11(12):1621. doi: 10.3390/brainsci11121621. PMID: 34942923
- Achenbach J., Faissner S., Saft C. Differential Diagnosis of Chorea-HIV Infection Delays Diagnosis of Huntington’s Disease by Years. Brain Sci. 2021 May 27;11(6):710. doi: 10.3390/brainsci11060710. PMID: 34071882
- Achenbach J, Saft C. Data from ENROLL-HD: Is the prevalence of juvenile and pediatric Huntington’s disease overestimated? Parkinsonism Relat Disord. 2021 Jul;88:1-2. doi: 10.1016/j.parkreldis.2021.05.012. Epub 2021 May 20. PMID: 34049236
- Bergonzoni G, Döring J, Biagioli M. D1R- and D2R-Medium-Sized Spiny Neurons Diversity: Insights Into Striatal Vulnerability to Huntington’s Disease Mutation. Front Cell Neurosci. 2021 Feb 10;15:628010. doi: 10.3389/fncel.2021.628010. eCollection 2021. PMID: 33642998
- Cain KK, Flanigan JL, Dalrymple WA, Patrie J, Harrison MB, Barrett MJ. The Effect of Education on Symptom Onset and Severity of Huntington’s Disease. Mov Disord Clin Pract. 2021 Mar 30;8(4):555-562. doi: 10.1002/mdc3.13195. eCollection 2021 May. PMID: 33981788
- Crowell V, Houghton R, Tomar A, Fernandes T, Squitieri F. Modeling Manifest Huntington’s Disease Prevalence Using Diagnosed Incidence and Survival Time. Neuroepidemiology. 2021;55(5):361-368. doi: 10.1159/000516767. Epub 2021 Jul 15. PMID: 34350853
- van Duijn E, Fernandes AR, Abreu D, Ware JJ, Neacy E, Sampaio C. Incidence of completed suicide and suicide attempts in a global prospective study of Huntington’s disease. BJPsych Open. 2021 Aug 31;7(5):e158. doi: 10.1192/bjo.2021.969. PMID: 34462049
- Ghazaleh N, Houghton R, Palermo G, Schobel SA, Wijeratne PA, Long JD. Ranking the Predictive Power of Clinical and Biological Features Associated With Disease Progression in Huntington’s Disease. Front Neurol. 2021 May 20;12:678484. doi: 10.3389/fneur.2021.678484. eCollection 2021. PMID: 34093422
- Griffin BA, Booth MS, Busse M, Wild EJ, Setodji C, Warner JH, Sampaio C, Mohan A. Estimating the causal effects of modifiable, non-genetic factors on Huntington disease progression using propensity score weighting. Parkinsonism Relat Disord. 2021 Feb;83:56-62. doi: 10.1016/j.parkreldis.2021.01.010. Epub 2021 Jan 13. PMID: 33476879
- Hentosh S, Zhu L, Patino J, Furr JW, Rocha NP, Furr Stimming E. Sex Differences in Huntington’s Disease: Evaluating the Enroll-HD Database. Mov Disord Clin Pract. 2021 Mar 8;8(3):420-426. doi: 10.1002/mdc3.13178. eCollection 2021 Apr. PMID: 33816672
- Horta-Barba A, Martinez-Horta S, Perez-Perez J, Sampedro F, de Lucia N, De Michele G, Salvatore E, Kehrer S, Priller J, Migliore S, Squitieri F, Castaldo A, Mariotti C, Mañanes V, Lopez-Sendon JL, Rodriguez N, Martinez-Descals A, Júlio F, Janurio C, Delussi M, de Tommaso M, Noguera S, Ruiz-Idiago J, Sitek EJ, Wallner R, Nuzzi A, Pagonabarraga J, Kulisevsky J; on behalf the Cognitive Phenotype Working Group of the European Huntington’s Disease Network. Arithmetic Word-Problem Solving as Cognitive Marker of Progression in Pre-Manifest and Manifest Huntington’s Disease. J Huntingtons Dis. 2021;10(4):459-468. doi: 10.3233/JHD-210480. PMID: 34602494
- Jones L, Wheeler V, Pearson CE. Special Issue: DNA Repair and Somatic Repeat Expansion in Huntington’s Disease. J Huntingtons Dis. 2021;10(1):3-5. doi: 10.3233/JHD-219001.PMID: 33554921
- McAllister B, Gusella JF, Landwehrmeyer GB, Lee JM, MacDonald ME, Orth M, Rosser AE, Williams NM, Holmans P, Jones L, Massey TH; REGISTRY investigators of the European Huntington’s disease network. Timing and Impact of Psychiatric, Cognitive, and Motor Abnormalities in Huntington Disease. Neurology. 2021 May 11;96(19):e2395-e2406. doi: 10.1212/WNL.0000000000011893. Epub 2021 Mar 25. PMID: 33766994
- McDonnell EI, Wang Y, Goldman J, Marder K. Age of Onset of Huntington’s Disease in Carriers of Reduced Penetrance Alleles. Mov Disord. 2021 Dec;36(12):2958-2961. doi: 10.1002/mds.28789. Epub 2021 Sep 18. PMID: 34536046
- Migliore S, D’Aurizio G, Maffi S, Ceccarelli C, Ristori G, Romano S, Castaldo A, Mariotti C, Curcio G, Squitieri F. Cognitive and behavioral associated changes in manifest Huntington disease: A retrospective cross-sectional study. Brain Behav. 2021 Jul;11(7):e02151. doi: 10.1002/brb3.2151. Epub 2021 Jun 10. PMID: 34110097
- Mohan A, Sun Z, Ghosh S, Li Y, Sathe S, Hu J, Sampaio C. A Machine-Learning Derived Huntington’s Disease Progression Model: Insights for Clinical Trial Design. Mov Disord. 2022 Mar;37(3):553-562. doi: 10.1002/mds.28866. Epub 2021 Dec 6.PMID: 34870344
- Ogilvie AC, Nopoulos PC, Schultz JL. Quantifying the Onset of Unintended Weight Loss in Huntington’s Disease: A Retrospective Analysis of Enroll-HD. J Huntingtons Dis. 2021;10(4):485-492. doi: 10.3233/JHD-210488. PMID: 34633327
- Ogilvie AC, Nopoulos PC, Schultz JL. Sleep disturbances by disease type and stage in Huntington’s disease. Parkinsonism Relat Disord. 2021 Oct;91:13-18. doi: 10.1016/j.parkreldis.2021.08.011. Epub 2021 Aug 21.PMID: 34450461
- Panegyres PK, Chen HY. Alzheimer’s disease, Huntington’s disease and cancer. J Clin Neurosci. 2021 Nov;93:103-105. doi: 10.1016/j.jocn.2021.09.012. Epub 2021 Sep 20. PMID: 34656231
- Puig-Davi A, Martinez-Horta S, Sampedro F, Horta-Barba A, Perez-Perez J, Campolongo A, Izquierdo-Barrionuevo C, Pagonabarraga J, Gomez-Anson B, Kulisevsky J. Cognitive and Affective Empathy in Huntington’s Disease. J Huntingtons Dis. 2021;10(3):323-334. doi: 10.3233/JHD-210469. PMID: 34486985
- Ranganathan M, Kostyk SK, Allain DC, Race JA, Daley AM. Age of onset and behavioral manifestations in Huntington’s disease: An Enroll-HD cohort analysis. Clin Genet. 2021 Jan;99(1):133-142. doi: 10.1111/cge.13857. Epub 2020 Oct 16. PMID: 33020896
- Schultz JL, Saft C, Nopoulos PC. Association of CAG Repeat Length in the Huntington Gene With Cognitive Performance in Young Adults. Neurology. 2021 May 11;96(19):e2407-e2413. doi: 10.1212/WNL.0000000000011823. Epub 2021 Mar 10.PMID: 33692166
- Sprenger GP, Roos RAC, van Zwet E, Reijntjes RH, Achterberg WP, de Bot ST. The prevalence of pain in Huntington’s disease in a large worldwide cohort. Parkinsonism Relat Disord. 2021 Aug;89:73-78. doi: 10.1016/j.parkreldis.2021.06.015. Epub 2021 Jun 19. PMID: 34243026
- Symonds AL, Macerollo A, Foy K, Alusi SH, Davies R. Genetic and Environmental Contributors to Neurodegeneration: An Exploration of the Effects of Alcohol on Clinical Features of Huntington’s Disease Using the Enroll-HD Global Platform. Int J Environ Res Public Health. 2021 May 12;18(10):5113. doi: 10.3390/ijerph18105113. PMID: 34065918
- van der Zwaan KF, Jacobs M, van Zwet EW, Roos RAC, de Bot ST. Predictors of Working Capacity Changes Related to Huntington’s Disease: A Longitudinal Study. J Huntingtons Dis. 2021;10(2):269-276. doi: 10.3233/JHD-200446.PMID: 33523014
- EHDN remote Plenary Meeting (September 9-11, 2021): J Neurol Neurosurg Psychiatry 2021 92(Suppl 1)
2020
- Eccles FJR, Craufurd D, Smith A, Davies R, Glenny K, Homberger M, Peeren S, Rogers D, Rose L, Skitt Z, Theed R, Simpson J. A feasibility investigation of mindfulness-based cognitive therapy for people with Huntington’s disease. Pilot Feasibility Stud. 2020 Jun 24;6:90. doi: 10.1186/s40814-020-00631-z. eCollection 2020. PMID: 32595978
- Fazio P, Fitzer-Attas CJ, Mrzljak L, Bronzova J, Nag S, Warner JH, Landwehrmeyer B, Al-Tawil N, Halldin C, Forsberg A, Ware J, Dilda V, Wood A, Sampaio C, Varrone A; PEARL-HD and LONGPDE10 study collaborators. PET molecular imaging of phosphodiesterase 10A: An early biomarker of Huntington’s disease progression. Mov Disord. 2020 Jan 22. doi: 10.1002/mds.27963. [Epub ahead of print] PMID: 31967355
- Martinez-Horta S, Horta-Barba A, Perez-Perez J, Sampedro F, de Lucia N, De Michele G, Kehrer S, Priller J, Migliore S, Squitieri F, Castaldo A, Mariotti C, Mañanes V, Lopez-Sendon JL, Rodriguez N, Martinez-Descals A, Garcia-Ruiz P, Júlio F, Januário C, Delussi M, de Tommaso M, Noguera S, Ruiz-Idiago J, Sitek EJ, Nuzzi A, Pagonabarraga J, Kulisevsky J; Cognitive Phenotype Working Group of the European Huntington’s Disease Network. Utility of the Parkinson’s disease-Cognitive Rating Scale for the screening of global cognitive status in Huntington’s disease. J Neurol. 2020 Feb 7. doi: 10.1007/s00415-020-09730-6. [Epub ahead of print] PMID: 32030521
- Mégret L, Nair SS, Dancourt J, Aaronson J, Rosinski J, Neri C. Combining feature selection and shape analysis uncovers precise rules for miRNA regulation in Huntington’s disease mice. BMC Bioinformatics. 2020 Feb 24;21(1):75. doi: 10.1186/s12859-020-3418-9. PMID: 32093602
- Mills JA, Long JD, Mohan A, Ware JJ, Sampaio C. Cognitive and Motor Norms for Huntington’s Disease. Arch Clin Neuropsychol. 2020 Aug 28;35(6):671-682. doi: 10.1093/arclin/acaa026. PMID: 32407458
- Rohiwal SS, Dvorakova N, Klima J, Vaskovicova M, Senigl F, Slouf M, Pavlova E, Stepanek P, Babuka D, Benes H, Ellederova Z, Stieger K. Polyethylenimine based magnetic nanoparticles mediated non-viral CRISPR/Cas9 system for genome editing. Sci Rep. 2020 Mar 12;10(1):4619. doi: 10.1038/s41598-020-61465-6. PMID: 32165679
- Spiers J, Smith JA, Ferrer-Duch M, Moldovan R, Roche J, MacLeod R. Evaluating a Genetic Counseling Narrative Group Session for People Who Have Tested Positive for the Huntington’s Disease Expansion: An Interpretative Phenomenolog-ical Analysis. J Genet Couns. 2020 Feb 19. doi: 10.1002/jgc4.1229. Online ahead of print. PMID: 32077165
- Steventon JJ, Rosser AE, Hart E, Murphy K. Hypertension, Antihypertensive Use and the Delayed-Onset of Huntington’s Disease. Mov Disord. 2020 Jun;35(6):937-946. doi: 10.1002/mds.27976. Epub 2020 Feb 4. PMID: 32017180
- Sun W, Zhou D, Warner JH, Langbehn DR, Hochhaus G, Wang Y. Huntington’s Disease Progression: A Population Modeling Approach to Characterization Using Clinical Rating Scales. J Clin Pharmacol. 2020 May 16. doi: 10.1002/jcph.1598. Online ahead of print. PMID: 32416008
- Vernizzi L, Paiardi C, Licata G, Vitali T, Santarelli S, Raneli M, Manelli V, Rizzetto M, Gioria MR, Pasini ME, Grifoni D, Vanoni MA, Gellera C, Taroni F, Bellosta B. Glutamine Synthetase 1 Increases Autophagy Lysosomal Degradation of Mutant Huntingtin Aggregates in Neurons, Ameliorating Motility in a Drosophila Model for Huntington’s Disease. Cells 2020 Jan 13;9(1):196. doi: 10.3390/cells9010196. PMID: 31941072
- Vuono R, Kouli A, Legault EM, Chagnon L, Allinson KS, La Spada A; REGISTRY Investigators of the European Huntington’s Disease Network, Biunno I, Barker RA, Drouin-Ouellet J. Association Between Toll-Like Receptor 4 (TLR4) and Triggering Receptor Expressed on Myeloid Cells 2 (TREM2) Genetic Variants and Clinical Progression of Huntington’s Disease. Mov Disord. 2020 Mar;35(3):401-408. doi: 10.1002/mds.27911. Epub 2019 Nov 14 PMID: 31724242
- Witkowski G, Jachinska K, Stepniak I, Ziora-Jakutowicz K, Sienkiewicz-Jarosz H. Alterations in transcranial sonography among Huntington’s disease patients with psychiatric symptoms. J Neural Transm (Vienna). 2020 Apr 13. doi: 10.1007/s00702-020-02187-x. Online ahead of print. PMID: 32285254
- Zielonka D, Witkowski G, Puch EA, Lesniczak M, Mazur-Michalek I, Isalan M, Mielcarek M. Prevalence of Non-psychiatric Comorbidities in Pre-symptomatic and Symptomatic Huntington’s Disease Gene Carriers in Poland. Front Med (Lausanne). 2020 Mar 11;7:79. doi: 10.3389/fmed.2020.00079. eCollection 2020. PMID: 32219094
2019
- Aldaz T, Nigro P, Sánchez-Gómez A, et al. Non-motor symptoms in Huntington’s disease: a comparative study with Parkinson’s disease. J Neurol. 2019 Jun;266(6):1340-1350. doi: 10.1007/s00415-019-09263-7. Epub 2019 Mar 5. PMID: 30834978
- Aubeeluck A, Stupple EJN, Schofield MB, Hughes AC, van der Meer L, Landwehrmeyer B, Ho AK. An International Validation of a Clinical Tool to Assess Carers’ Quality of Life in Huntington’s Disease. Front Psychol. 2019 Jul 23;10:1658. doi: 10.3389/fpsyg.2019.01658. eCollection 2019. PMID: 31402885
- Bartoszek A, Aubeeluck A, Stupple E, et al. Exploring the Reliability and Validity of the Huntington’s Disease Quality of Life Battery for Carers (HDQoL-C) within A Polish Population. Int J Environ Res Public Health. 2019 Jun 30;16(13). pii: E2323. doi: 10.3390/ijerph16132323. PMID: 31262100
- Bigan E, Sasidharan Nair S, Lejeune FX, Fragnaud H, Parmentier F, Mégret L, Verny M, Aaronson J, Rosinski J, Neri C. Genetic cooperativity in multi-layer networks implicates cell survival and senescence in the striatum of Huntington’s disease mice synchronous to symptoms. Bioinformatics. 2019 Jun 22. pii: btz514. doi: 10.1093/bioinformatics/btz514. [Epub ahead of print] PMID: 31228193
- Braisch U, Muche R, Rothenbacher D, Landwehrmeyer GB, Long JD, Orth M; Registry investigators of the European Huntington’s Disease Network and COHORT investigators of the Huntington Study Group. Identification of symbol digit modality test score extremes in Huntington’s disease. Am J Med Genet B Neuropsychiatr Genet. 2019 Apr;180(3):232-245. doi: 10.1002/ajmg.b.32719. Epub 2019 Feb 20 PMID: 30788902
- Cubo E, Martinez-Horta SI, Santalo FS et al. Clinical manifestations of homozygote allele carriers in Huntington disease. Neurology. 2019 Apr 30;92(18):e2101-8. doi: 10.1212/WNL.0000000000007147. Epub 2019 Mar 13. PMID: 30867264
- Gusella J, Genetic Modifiers of Huntington’s Disease (GeM-HD) Consortium. CAG Repeat Not Polyglutamine Length Determines Timing of Huntington’s Disease Onset. Cell. 2019 Aug 8;178(4):887-900.e14. doi:10.1016/j.cell.2019.06.036. PMID: 31398342
- Hawton A, Green C, Goodwin E, Harrower T. Health state utility values (QALY weights) for Huntington’s disease: an analysis of data from the European Huntington’s Disease Network (EHDN). Eur J Health Econ. 2019 Aug 13. doi: 10.1007/s10198-019-01092-9. [Epub ahead of print] PMID: 31410669
- Ho AK, Horton MH, GB Landwehrmeyer, Burgunder J-M & Tennant A. (2019). Meaningful and measurable health domains in Huntington’s: Large-scale validation of the Huntington’s Disease health-related Quality of Life questionnaire (HDQoL) across severity stages. Value in Health, 22(6), 712-720 PMID: 31198189
- Horton MC, Nopoulos P, Nance M, Landwehrmyer GB, Barker RA, Squitieri F; REGISTRY Investigators of the European Huntington’s Disease Network, Burgunder JM, Quarrell O. Assessment of the Performance of a Modified Motor Scale as Applied to Juvenile Onset Huntington’s Disease. J Huntingtons Dis. 2019;8(2):181-193. doi: 10.3233/JHD-180306. PMID: 30856116
- Mantuano E, Peconi M, Scarabino D. Can leukocyte telomere shortening be a possible biomarker to track Huntington’s disease progression? Neural Regen Res. 2019 Oct;14(10):1709-1710. doi: 10.4103/1673-5374.257522. PMID: 31169183
- Merienne N, Meunier C, Schneider A, JSeguin J, Nair SS, Rocher AB, Le Gras S, Keime C, Faull R, Pellerin L, Chatton JY, Neri C, Merienne K, Déglon N. Cell-Type-Specific Gene Expression Profiling in Adult Mouse Brain Reveals Normal and Disease-State Signatures. Cell Rep. 2019 Feb 26;26(9):2477-2493.e9. doi: 10.1016/j.celrep.2019.02.003. PMID: 30811995
- Mustafa R , Kreiner G, Kamińska K, Wood AEJ, Kirsch J, Tucker KL, Parlato R. Targeted Depletion of Primary Cilia in Dopaminoceptive Neurons in a Preclinical Mouse Model of Huntington’s Disease. Front Cell Neurosci 2019 Dec 20;13:565. doi: 10.3389/fncel.2019.00565. eCollection 2019. PMID: 31920562
- Oosterloo M, Bijlsma EK, van Kuijk SM, Minkels F, de Die-Smulders CE; REGISTRY Investigators of the European Huntington’s Disease Network; Registry Steering committee; Language coordinators; EHDN’s associate site in Singapore. Clinical and genetic characteristics of late-onset Huntington’s disease. Parkinsonism Relat Disord. 2019 Apr;61:101-105. doi: 10.1016/j.parkreldis.2018.11.009. Epub 2018 Nov 29. PMID: 30528461
- Quarrell OWJ, Nance MA, Nopoulos P, Reilmann R, Oosterloo M, Tabrizi SJ, Furby H, Saft C, Roos RAC, Squitieri F, Landwehrmeyer GB, Burgunder JM; Juvenile Huntington Disease Working Group of the European Huntington Disease Network. Defining paediatric Huntington disease: time to abandon the term Juvenile Huntington Disease? Mov Disord. 2019 Feb 20. doi: 10.1002/mds.27640. Epub ahead of print. PMID: 30788860
- Sahl SJ, Vonk WIM. Superresolution Fluorescence Imaging of Mutant Huntingtin Aggregation in Cells. Methods Mol Biol. 2019;1873:241-251. doi: 10.1007/978-1-4939-8820-4_15. PMID: 30341614
- Scarabino D, Veneziano L, Peconi M, Frontali M, Mantuano E, Corbo RM. Leukocyte telomere shortening in Huntington’s disease. J Neurol Sci. 2019 Jan 15;396:25-29. doi: 10.1016/j.jns.2018.10.024. Epub 2018 Oct 28. PMID: 30396032
2018
- Arnoux I, Willam M, Griesche N, Krummeich J, Watari H, Offermann N, Weber S, Narayan Dey P, Chen C, Monteiro O, Buettner S, Meyer K, Bano D, Radyushkin K, Langston R, Lambert JJ, Wanker E, Methner A, Krauss S, Schweiger S, Stroh A. Metformin reverses early cortical network dysfunction and behavior changes in Huntington’s disease. Elife. 2018 Sep 4;7. pii: e38744. doi: 10.7554/eLife.38744. PMID: 30179155
- Baake V, van Duijn E, Roos RAC. Huntington’s Disease Gene Expansion Carriers Are Aware of Their Degree of Apathy. J Neuropsychiatry Clin Neurosci. 2018 Summer;30(3):183-187. doi: 10.1176/appi.neuropsych.18020031. Epub 2018 May 30. PMID: 29843585
- Barkhuizen M, Rodrigues FB, Anderson DG, Winkens B; REGISTRY Investigators of the European Huntington’s Disease Network, Wild EJ, Kramer BW, Gavilanes AWD. Perinatal insults and neurodevelopmental disorders may impact Huntington’s disease age of diagnosis. Parkinsonism Relat Disord. 2018 Oct;55:55-60. doi: 10.1016/j.parkreldis.2018.05.016. Epub 2018 May 18. PMID: 29804730
- Chao MJ, Kim KH, Shin JW, Lucente D, Wheeler VC, Li H, Roach JC, Hood L, Wexler NS, Jardim LB, Holmans P, Jones L, Orth M, Kwak S, MacDonald ME, Gusella JF, Lee JM. Population-specific genetic modification of Huntington’s disease in Venezuela. PLoS Genet. 2018 May 11;14(5):e1007274. doi: 10.1371/journal.pgen.1007274. eCollection 2018 May. PMID: 29750799
- Di Pardo A, Pepe G, Castaldo S, Marracino F, Capocci L, Amico E, Madonna M, Giova S, Jeong SK, Park BM, Park BD, Maglione V. Stimulation of Sphingosine Kinase 1 (SPHK1) Is Beneficial in a Huntington’s Disease Pre-clinical Model. Front Mol Neurosci. 2019 Apr 24;12:100. doi: 10.3389/fnmol.2019.00100. eCollection 2019. PMID: 31068790
- Duarte AI, Sjögren M, Santos MS, Oliveira CR, Moreira PI, Björkqvist M. Dual Therapy with Liraglutide and Ghrelin Promotes Brain and Peripheral Energy Metabolism in the R6/2 Mouse Model of Huntington’s Disease. Sci Rep. 2018 Jun 12;8(1):8961. doi: 10.1038/s41598-018-27121-w. PMID: 29895889
- van Duijn E, Vrijmoeth EM, Giltay EJ, Bernhard Landwehrmeyer G; REGISTRY investigators of the European Huntington’s Disease Network. Suicidal ideation and suicidal behavior according to the C-SSRS in a European cohort of Huntington’s disease gene expansion carriers. J Affect Disord. 2018 Mar 1;228:194-204. doi: 10.1016/j.jad.2017.11.074. Epub 2017 Nov 15. PMID: 29253686
- Eddy CM, Rickards HE, Hansen PC. Through your eyes or mine? The neural correlates of mental state recognition in Huntington’s disease. Hum Brain Mapp. 2018 Mar;39(3):1354-1366. doi: 10.1002/hbm.23923. Epub 2017 Dec 17. PMID: 29250867
- Fusilli C, Migliore S, Mazza T, Consoli F, De Luca A, Barbagallo G, Ciammola A, Gatto EM, Cesarini M, Etcheverry JL, Parisi V, Al-Oraimi M, Al-Harrasi S, Al-Salmi Q, Marano M, Vonsattel JG, Sabatini U, Landwehrmeyer GB, Squitieri F. Biological and clinical manifestations of juvenile Huntington’s disease: a retrospective analysis. Lancet Neurol. 2018 Nov;17(11):986-993. doi: 10.1016/S1474-4422(18)30294-1. Epub 2018 Sep 19. PMID: 30243861
- Massey T, McAllister B, Jones L. Methods for Assessing DNA Repair and Repeat Expansion in Huntington’s Disease. Methods Mol Biol. 2018;1780:483-495. doi: 10.1007/978-1-4939-7825-0_22. PMID: 29856032
- McNulty P, Pilcher R, Ramesh R, Necuiniate R, Hughes A, Farewell D, Holmans P, Jones L; REGISTRY Investigators of the European Huntington’s Disease Network . Reduced Cancer Incidence in Huntington’s Disease: Analysis in the Registry Study. J Huntingtons Dis. 2018;7(3):209-222. doi: 10.3233/JHD-170263. PMID: 30103338
- Sun Z, Li Y, Ghosh S, Cheng Y, Mohan A, Sampaio C, Hu J. A Data-Driven Method for Generating Robust Symptom Onset Indicators in Huntington’s Disease Registry Data. AMIA Annu Symp Proc. 2018 Apr 16;2017:1635-1644. eCollection 2017. PMID: 29854234
- Valcárcel-Ocete L, Fullaondo A, Alkorta-Aranburu G, García-Barcina M, Roos RAC, Hjermind LE, Saft C, Frontali M, Reilmann R, Rickards H; REGISTRY investigators of the European Huntington’s Disease Network (EHDN), Zubiaga AM, Aguirre A. Does arterial hypertension influence the onset of Huntington’s disease? PLoS One. 2018 May 23;13(5):e0197975. doi: 10.1371/journal.pone.0197975. eCollection 2018. PMID: 29791508
- Zielonka D, Ren M, De Michele G, Roos RAC, Squitieri F, Bentivoglio AR, Marcinkowski JT, Landwehrmeyer GB. The contribution of gender differences in motor, behavioral and cognitive features to functional capacity, independence and quality of life in patients with Huntington’s disease. Parkinsonism Relat Disord. 2018 Apr;49:42-47. doi: 10.1016/j.parkreldis.2018.01.006. Epub 2018 Jan 5. PMID: 29326033
- EHDN 10th Plenary Meeting (September 14-16, 2018): J Neurol Neurosurg Psychiatry 2018 89(Suppl 1)
2017
- Baake V, Reijntjes RHAM, Dumas EM, Thompson JC; REGISTRY Investigators of the European Huntington’s Disease Network, Roos RAC. Cognitive decline in Huntington’s disease expansion gene carriers. Cortex. 2017 Aug 3;95:51-62. doi: 10.1016/j.cortex.2017.07.017. [Epub ahead of print] PMID: 28843844
- Braisch U, Hay B, Muche R, Rothenbacher D, Landwehrmeyer GB, Long JD, Orth M; REGISTRY Investigators of the European Huntington’s Disease Network and COHORT Investigators of the Huntington Study Group. Identification of extreme motor phenotypes in Huntington’s disease. Am J Med Genet B Neuropsychiatr Genet. 2017 Apr;174(3):283-294. doi: 10.1002/ajmg.b.32514. Epub 2016 Nov 21. PMID: 27868347
- Branco-Santos J, Herrera F, Poças GM, Pires-Afonso Y, Giorgini F, Domingos PM, Outeiro TF. Protein phosphatase 1 regulates huntingtin exon 1 aggregation and toxicity. Hum Mol Genet. 2017 Oct 1;26(19):3763-3775. doi: 10.1093/hmg/ddx260. PMID: 28934390
- Bowles KR, Stone T, Holmans P, Allen ND, Dunnett SB, Jones L. SMAD transcription factors are altered in cell models of HD and regulate HTT expression. Cell Signal. 2017 Feb;31:1-14. doi: 10.1016/j.cellsig.2016.12.005. PMID: 27988204
- Buck E, Zügel M, Schumann U, Merz T, Gumpp AM, Witting A, Steinacker JM, Landwehrmeyer GB, Weydt P, Calzia E, Lindenberg KS. High-resolution respirometry of fine-needle muscle biopsies in pre-manifest Huntington’s disease expansion mutation carriers shows normal mitochondrial respiratory function. PLoS One. 2017 Apr 13;12(4):e0175248. doi: 10.1371/journal.pone.0175248. eCollection 2017. PMID: 28406926
- Busse M, Quinn L, Drew C, Kelson M, Trubey R, McEwan K, Jones C, Townson J, Dawes H, Tudor-Edwards R, Rosser A, Hood K. Physical Activity Self-Management and Coaching Compared to Social Interaction in Huntington Disease: Results From the ENGAGE-HD Randomized, Controlled, Pilot Feasibility Trial.
Phys Ther. 2017 Mar 24. doi: 10.1093/ptj/pzx031. [Epub ahead of print]. PMID: 28371942
- Fritz NE, Busse M, Jones K, Khalil H, Quinn L; Members of the Physiotherapy Working Group of the European Huntingtonʼs Disease Network. A Classification System to Guide Physical Therapy Management in Huntington Disease: A Case Series. J Neurol Phys Ther. 2017 Jul;41(3):156-163. doi: 10.1097/NPT.0000000000000188. PMID: 28628549
- Gardiner SL, van Belzen MJ, Boogaard MW, van Roon-Mom WMC, Rozing MP, van Hemert AM, Smit JH, Beekman ATF, van Grootheest G, Schoevers RA, Oude Voshaar RC, Roos RAC, Comijs HC, Penninx BWJH, van der Mast RC, Aziz NA. Huntingtin gene repeat size variations affect risk of lifetime depression. Transl Psychiatry. 2017 Dec 11;7(12):1277. doi: 10.1038/s41398-017-0042-1. Review. PMID: 29225330
- Gilling M, Budtz-Jørgensen E, Boonen SE, Lildballe D, Bojesen A, Hertz JM, Sørensen SA. The Danish HD Registry (DHR) – a nationwide family registry of HD families in Denmark. Clin Genet. 2017 Sep;92(3):338-341. doi: 10.1111/cge.12984. Epub 2017 Mar 28. PMID: 28155235
- Jones L, Houlden H, Tabrizi SJ. DNA repair in the trinucleotide repeat disorders. Lancet Neurol. 2017 Jan;16(1):88-96. doi: 10.1016/S1474-4422(16)30350-7. Review. PMID: 27979358
- Jussi O.Sipilä PhD thesis: Huntington’s disease in Finland. Epidemiologic, genetic and clinical studies. (2017-04-21)
- Lee JM, Chao MJ, Harold D, Abu Elneel K, Gillis T, Holmans P, Jones L, Orth M, Myers RH, Kwak S, Wheeler VC, MacDonald ME, Gusella JF. A modifier of Huntington’s disease onset at the MLH1 locus. Hum Mol Genet. 2017 Oct 1;26(19):3859-3867. doi: 10.1093/hmg/ddx286. PMID: 28934397
- Long JD, Langbehn DR, Tabrizi SJ, Landwehrmeyer BG, Paulsen JS, Warner J, Sampaio C. Validation of a prognostic index for Huntington’s disease. Mov Disord. 2017 Feb;32(2):256-263. doi: 10.1002/mds.26838. Epub 2016 Nov 28. PMID: 27892614
- Maltby J, Dale M, Underwood M, Simpson J and the REGISTRY investigators of the European Huntington’s Disease Network. Irritability in Huntington’s Disease: Factor Analysis of Snaith’s Irritability Scale. Mov Dis Clinical Practice. 2017 May/June 4(3):342–348.
- Maurage P, Heeren A, Lahaye M, Jeanjean A, Guettat L, Verellen-Dumoulin C, Halkin S, Billieux J, Constant E. Attentional Impairments in Huntington’s Disease: A Specific Deficit for the Executive Conflict. Neuropsychology. 2017 May;31(4):424-436. doi: 10.1037/neu0000321. Epub 2017 Feb 27. PMID: 28240935
- Moss DJH, Pardiñas AF, Langbehn D, Lo K, Leavitt BR, Roos R, Durr A, Mead S; TRACK-HD investigators; REGISTRY investigators, Holmans P, Jones L, Tabrizi SJ. Identification of genetic variants associated with Huntington’s disease progression: a genome-wide association study. Lancet Neurol. 2017 Sep;16(9):701-711. doi: 10.1016/S1474-4422(17)30161-8. Epub 2017 Jun 20. PMID: 28642124
- Orth M, Bronzova J, Tritsch C, Dorsey ER, Ferreira JJ, Gemperli A and the EHDN REGISTRY and HSG COHORT Investigators. Comparison of Huntington’s Disease in Europe and North America. Mov Dis Clinical Practice. 2017 May/June 4(3):358-367.
- Reynolds RH, Petersen MH, Willert CW, Heinrich M, Nymann N, Dall M, Treebak JT, Björkqvist M, Silahtaroglu A, Hasholt L, Nørremølle A. Perturbations in the p53/miR-34a/SIRT1 pathway in the R6/2 Huntington’s disease model. Mol Cell Neurosci. 2017 Dec 28;88:118-129. doi: 10.1016/j.mcn.2017.12.009. [Epub ahead of print] PMID: 29289683
- Rodrigues FB, Abreu D, Damásio J, Gonçalves N, Correia Guedes L, Coelho M, Ferreira JJ, REGISTRY Investigators of the European Huntington’s Disease Network. Survival, Mortality, Causes and Places of Death in a European Huntington’s Disease Prospective Cohort. Mov Dis Clinical Practice. 2017 September/October 4(5):737-742.
- Ruiz-Idiago JM, Floriach M, Mareca C, Salvador R, López-Sendón JL, Mañanés V, Cubo E, Mariscal N, Muñoz E, Santacruz P, Noguera MF, Vivancos L, Roy P, Pomarol-Clotet E, Sarró S; Spanish Huntington Disease Network. Spanish Validation of the Problem Behaviors Assessment-Short (PBA-s) for Huntington’s Disease. J Neuropsychiatry Clin Neurosci. 2017 Winter;29(1):31-38. doi: 10.1176/appi.neuropsych.16020025. Epub 2016 Jul 15. PMID: 27417071
- Sjögren M, Duarte AI, McCourt AC, Shcherbina L, Wierup N, Björkqvist M. Ghrelin rescues skeletal muscle catabolic profile in the R6/2 mouse model of Huntington’s disease. Sci Rep. 2017 Oct 24;7(1):13896. doi: 10.1038/s41598-017-13713-5. PMID: 29066728
- Stuitje G, van Belzen MJ, Gardiner SL, van Roon-Mom WMC, Boogaard MW; REGISTRY Investigators of the European Huntington Disease Network, Tabrizi SJ, Roos RAC, Aziz NA. Age of onset in Huntington’s disease is influenced by CAG repeat variations in other polyglutamine disease-associated genes. Brain. 2017 Jul 1;140(7):e42. doi: 10.1093/brain/awx122. PMID: 28549075
- Suelves N, Kirkham-McCarthy L, Lahue RS, Ginés S. A selective inhibitor of histone deacetylase 3 prevents cognitive deficits and suppresses striatal CAG repeat expansions in Huntington’s disease mice. Sci Rep. 2017, 7:6082. doi:10.1038/s41598-017-05125-2. PMID: 28729730
- Underwood M, Bonas S, Dale M, REGISTRY Investigators of the European Huntington’s Disease Network. Huntington’s Disease: Prevalence and Psychological Indicators of Pain. Mov Dis Clin Pract 2017 March/April 4(2):198-204.
2016
- Baake V, Hart EP, Bos R, Roos RA. Participants at the Leiden Site of the REGISTRY Study: A Demographic Approach. J Huntingtons Dis. 2016 Mar 15; 5(1):83-90. doi: 10.3233/JHD-150157. PMID: 27003663
- Braisch U, Hay B, Muche R, Rothenbacher D, Landwehrmeyer GB, Long JD, Orth M; REGISTRY Investigators of the European Huntington’s Disease Network and COHORT Investigators of the Huntington Study Group. Identification of extreme motor phenotypes in Huntington’s disease. Am J Med Genet B Neuropsychiatr Genet. 2017 Apr;174(3):283-294. doi: 10.1002/ajmg.b.32514. Epub 2016 Nov 21. PMID: 27868347
- Bettencourt C, Hensman-Moss D, Flower M, Wiethoff S, Brice A, Goizet C, Stevanin G, Koutsis G, Karadima G, Panas M, Yescas-Gómez P, García-Velázquez LE, Alonso-Vilatela ME, Lima M, Raposo M, Traynor B, Sweeney M, Wood N, Giunti P; SPATAX Network, Durr A, Holmans P, Houlden H, Tabrizi SJ, Jones L. DNA repair pathways underlie a common genetic mechanism modulating onset in polyglutamine diseases. Ann Neurol. 2016 Jun;79(6):983-90. doi: 10.1002/ana.24656. Epub 2016 May 6. PMID: 27044000
- Cubo E, Rivadeneyra J, Mariscal N, Martinez A, Armesto D, Camara RJ and on behalf of the Spanish Members of the European Huntington’s Disease Registry. Factors Associated with Low Body Mass Index in Huntington’s Disease: A Spanish Multicenter Study of the European Huntington’s Disease Registry. Mov Dis Clinical Practice. 2016 September/October 3(5):452-459.
- Cubo E, Ramos-Arroyo MA, Martinez-Horta S, Martínez-Descalls A, Calvo S, Gil-Polo C; European HD Network. Clinical manifestations of intermediate allele carriers in Huntington disease. Neurology. 2016 Aug 9;87(6):571-8. doi: 10.1212/WNL.0000000000002944. Epub 2016 Jul 8. PMID: 27402890
- Dale M, Maltby J, Shimozaki S, Cramp R, Rickards H; REGISTRY Investigators of the European Huntington’s Disease Network. Disease stage, but not sex, predicts depression and psychological distress in Huntington’s disease: A European population study. J Psychosom Res. 2016 Jan; 80:17-22. doi: 10.1016/j.jpsychores.2015.11.003. Epub 2015 Nov 12. PMID: 26721543
- Jacobs M, Hart EP, van Zwet EW, Bentivoglio AR, Burgunder JM, Craufurd D, Reilmann R, Saft C, Roos RA; REGISTRY investigators of the European Huntington’s Disease Network. Progression of motor subtypes in Huntington’s disease: a 6-year follow-up study. J Neurol. 2016 Oct;263(10):2080-5. doi: 10.1007/s00415-016-8233-x. Epub 2016 Jul 19. PMID: 27435968
- Jones C, Busse M, Quinn L, Dawes H, Drew C, Kelson M, Hood K, Rosser A, Edwards RT. The societal cost of Huntington’s disease: are we underestimating the burden? Eur J Neurol. 2016 Oct;23(10):1588-90. doi: 10.1111/ene.13107. Epub 2016 Jul 27. PMID: 27461550
- Jones U, Busse M, Enright S, Rosser AE. Respiratory decline is integral to disease progression in Huntington’s disease. Eur Respir J. 2016 Aug;48(2):585-8. doi: 10.1183/13993003.02215-2015. PMID: 27338194
- Keum JW, Shin A, Gillis T, Mysore JS, Abu Elneel K, Lucente D, Hadzi T, Holmans P, Jones L, Orth M, Kwak S, MacDonald ME, Gusella JF, Lee JM. The HTT CAG-Expansion Mutation Determines Age at Death but Not Disease Duration in Huntington Disease. Am J Hum Genet. 2016 2016 Feb 4;98(2):287-98. doi: 10.1016/j.ajhg.2015.12.018. PMID: 26849111
- Maltby J, Dale M, Underwood M, Rickards H, Callaghan J; and REGISTRY Investigators of the European Huntington’s Disease Network. Exploring the Structural Relationship Between Interviewer and Self-Rated Affective Symptoms in Huntington’s Disease. J Neuropsychiatry Clin Neurosci. 2016 Summer;28(3):236-8. doi: 10.1176/appi.neuropsych.15090237. Epub 2016 Feb 22. PMID: 26900736
- Martinez-Horta S, Perez-Perez J, van Duijn E, Fernandez-Bobadilla R, Carceller M, Pagonabarraga J, Pascual-Sedano B, Campolongo A, Ruiz-Idiago J, Sampedro F, Landwehrmeyer GB; Spanish REGISTRY investigators of the European Huntington’s Disease Network, Kulisevsky J. Neuropsychiatric symptoms are very common in premanifest and early stage Huntington’s Disease. Parkinsonism Relat Disord. 2016 Apr;25:58-64. doi: 10.1016/j.parkreldis.2016.02.008. PMID: 26898966
- Maurage P, Lahaye M, Grynberg D, Jeanjean A, Guettat L, Verellen-Dumoulin C, Halkin S, Heeren A, Billieux J, Constant E. Dissociating emotional and cognitive empathy in pre-clinical and clinical Huntington’s disease. Psychiatry Res. 2016 Mar 30;237:103-8. doi: 10.1016/j.psychres.2016.01.070. PMID: 26869362
- Naseri NN, Bonica J, Xu H, Park LC, Arjomand J, Chen Z, Gibson GE. Novel Metabolic Abnormalities in the Tricarboxylic Acid Cycle in Peripheral Cells From Huntington’s Disease Patients. PLoS One 2016 Sep 9 ;11(9):e0160384. doi: 10.1371. PMID: 27611087
- Quinn L, Hamana K, Kelson M, Dawes H, Collett J, Townson J, Roos R, van der Plas AA, Reilmann R, Frich JC, Rickards H, Rosser A, Busse M. A randomized, controlled trial of a multi-modal exercise intervention in Huntington’s disease. Parkinsonism Relat Disord. 2016 Oct;31:46-52. doi: 10.1016/j.parkreldis.2016.06.023. Epub 2016 Jul 1. PMID: 27423921
- Reynolds Regina Master Thesis: Changes in the miR-34a – SIRT1 axis in Huntington’s disease
- Rivadeneyra J, Cubo E, Gil C, Calvo S, Mariscal N, Martínez A. Factors associated with Mediterranean diet adherence in Huntington’s disease. Clin Nutr ESPEN. 2016 Apr;12:e7-e13. doi: 10.1016/j.clnesp.2016.01.001. Epub 2016 Mar 5. PMID: 28531758
- Warner JH, Sampaio C. Modeling Variability in the Progression of Huntington’s Disease A Novel Modeling Approach Applied to Structural Imaging Markers from TRACK-HD. CPT Pharmacometrics Syst Pharmacol. 2016 Aug;5(8):437-45. doi: 10.1002/psp4.12097. PMID: 27481337
- EHDN 9th Plenary Meeting (September 16-18, 2016): J Neurol Neurosurg Psychiatry 2016 87(Suppl 1)
2015
- Genetic Modifiers of Huntington’s Disease (GeM-HD) Consortium. Identification of Genetic Factors that Modify Clinical Onset of Huntington’s Disease. Cell. 2015 Jul 30; 162(3):516-26. doi: 10.1016/j.cell.2015.07.003. PMID: 26232222
- Achour M, Le Gras S, Keime C, Parmentier F, Lejeune FX, Boutillier AL, Néri C, Davidson I, Merienne K. Neuronal identity genes regulated by super-enhancers are preferentially down-regulated in the striatum of Huntington’s disease mice. Hum Mol Genet. 2015 Jun 15;24(12):3481-96. doi: 10.1093/hmg/ddv099. Epub 2015 Mar 17. PMID: 25784504
- Bečanović K, Nørremølle A, Neal SJ, Kay C, Collins JA, Arenillas D, Lilja T, Gaudenzi G, Manoharan S, Doty CN, Beck J, Lahiri N, Portales-Casamar E, Warby SC, Connolly C, De Souza RA; REGISTRY Investigators of the European Huntington’s Disease Network, Tabrizi SJ, Hermanson O, Langbehn DR, Hayden MR, Wasserman WW, Leavitt BR. A SNP in the HTT promoter alters NF-κB binding and is a bidirectional genetic modifier of Huntington disease. Nat Neurosci 2015 Jun; 18(6):807-16. doi: 10.1038/nn.4014. Epub 2015 May 4. PMID: 25938884
- Correia K, Harold D, Kim KH, Holmans P, Jones L, Orth M, Myers RH, Kwak S, Wheeler VC, MacDonald ME, Gusella JF, Lee JM. The Genetic Modifiers of Motor OnsetAge (GeM MOA) Website: Genome-wide Association Analysis for Genetic Modifiers of Huntington’s Disease. J Huntingtons Dis 2015; 4(3):279-84. doi: 10.3233/JHD-150169. PMID: 26444025
- Cubo E, Rivadeneyra J, Armesto D, Mariscal N, Martinez A, Camara RJ; Spanish members of the European Huntington Disease Network. Relationship between Nutritional Status and the Severity of Huntington’s Disease. A Spanish Multicenter Dietary Intake Study. J Huntingtons Dis 2015; 4(1):78-85. PMID: 26333259
- Dale M, Maltby J, Martucci R, Shimozaki S; REGISTRY investigators of the European Huntington’s Disease Network. Factor analysis of the hospital anxiety and depression scale among a Huntington’s disease population. Mov Disord. 2015 Dec; 30(14):1954-60. doi: 10.1002/mds.26419. Epub 2015 Oct 7. PMID: 26443751
- Eddy CM, Rickards HE. Theory of mind can be impaired prior to motor onset in Huntington’s disease. Neuropsychology. 2015 Sep;29(5):792-8. doi: 10.1037/neu0000190. Epub 2015 Feb 9. PMID: 25664466
- Eddy CM, Rickards HE. Interaction without intent: the shape of the social world in Huntington’s disease. Soc Cogn Affect Neurosci. 2015 Sep;10(9):1228-35. doi: 10.1093/scan/nsv012. Epub 2015 Feb 12. PMID: 25680992
- Eddy CM, Rickards HE. Cognitive deficits predict poorer functional capacity in Huntington’s disease: but what is being measured? Neuropsychology. 2015 Mar;29(2):268-73. doi: 10.1037/neu0000134. Epub 2014 Aug 11. PMID: 25110931
- McCourt AC, O’Donovan KL, Ekblad E, Sand E, Craufurd D, Rosser A, Sanders D, Stoy N, Rickards H, Wierup N, Bates GP, Björkqvist M, Quarrell O. Characterization of Gastric Mucosa Biopsies Reveals Alterations in Huntington’s Disease. PLoS Curr. 2015 Jun 26;7. pii: ecurrents.hd.858b4cc7f235df068387e9c20c436a79. doi: 10.1371/currents.hd.858b4cc7f235df068387e9c20c436a79. PMID: 26581667
- Mort M, Carlisle FA, Waite AJ, Elliston L, Allen ND, Jones L, Hughes AC. Huntingtin Exists as Multiple Splice Forms in Human Brain. J Huntingtons Dis. 2015;4(2):161-71. doi: 10.3233/JHD-150151. PMID: 26397897
- Steventon JJ, Harrison DJ, Trueman RC, Rosser AE, Jones DK, Brooks SP. In Vivo MRI Evidence that Neuropathology is Attenuated by Cognitive Enrichment in the Yac128 Huntington’s Disease Mouse Model. J Huntingtons Dis. 2015;4(2):149-60. doi: 10.3233/JHD-150147. PMID: 26397896
- Tedroff J, Waters S, Barker RA, Roos R, Squitieri F; EHDN Registry Study Group. Antidopaminergic Medication is Associated with More Rapidly Progressive Huntington’s Disease. J Huntingtons Dis 2015; 4(2):131-40. doi: 10.3233/JHD-150143. PMID: 26397894
- Valcárcel-Ocete L, Alkorta-Aranburu G, Iriondo M, Fullaondo A, García-Barcina M, Fernández-García JM, Lezcano-García E, Losada-Domingo JM, Ruiz-Ojeda J, Álvarez de Arcaya A, Pérez-Ramos JM, Roos RA, Nielsen JE, Saft C; REGISTRY investigators of the European Huntington’s Disease Network, Zubiaga AM, Aguirre A. Exploring Genetic Factors Involved in Huntington Disease Age of Onset: E2F2 as a New Potential Modifier Gene. PLoS One. 2015 Jul 6; 10(7):e0131573. doi: 10.1371/journal.pone.0131573. eCollection 2015. PMID: 26148071
- Vuono R, Winder-Rhodes S, de Silva R, Cisbani G, Drouin-Ouellet J; REGISTRY Investigators of the European Huntington’s Disease Network, Spillantini MG, Cicchetti F, Barker RA. The role of tau in the pathological process and clinical expression of Huntington’s disease. Brain. 2015 Jul; 138(Pt 7):1907-18. doi: 10.1093/brain/awv107. Epub 2015 May 6. PMID: 25953777
- Wojtecki L, Groiss SJ, Ferrea S, Elben S, Hartmann CJ, Dunnett SB, Rosser A, Saft C, Südmeyer M, Ohmann C, Schnitzler A, Vesper J; Surgical Approaches Working Group of the European Huntington’s Disease Network (EHDN). A Prospective Pilot Trial for Pallidal Deep Brain Stimulation in Huntington’s Disease. Front Neurol. 2015 Aug 18;6:177. doi: 10.3389/fneur.2015.00177. eCollection 2015. PMID: 26347707
- Zügel M, Weydt P. Sports and Physical Activity in Patients Suffering from Rare Neurodegenerative Diseases: How Much is too Much, how Much is too Little? Deutsche Zeitschrift für Sportmedizin. 2015; 66(11):300-7.
2014
- Busse M, Quinn L, Khalil H, McEwan K. Optimising mobility outcome measures in Huntington’s disease. J Huntingtons Dis. 2014;3(2):175-88. doi: 10.3233/JHD-140091. PMID: 25062860
- Esther Cubo, Christopher G. Goetz, Glenn T. Stebbins, Nancy R. LaPelle, Barbara C. Tilley, Lu Wang, Sheng Luo and on behalf of the Spanish UDysRS Program Members. Independent Spanish Validation of the Unified Dyskinesia Rating Scale. Movement Disorders – Clinical Practice. 2014 Sep; 1(3):213-8. doi: 10.1002/mdc3.12065.
- van Duijn E, Craufurd D, Hubers AA, Giltay EJ, Bonelli R, Rickards H, Anderson KE, van Walsem MR, van der Mast RC, Orth M, Landwehrmeyer GB; the European Huntington’s Disease Network Behavioural Phenotype Working Group. Neuropsychiatric symptoms in a European Huntington’s disease cohort (REGISTRY). J Neurol Neurosurg Psychiatry. 2014 Dec; 85(12):1411-18. doi: 10.1136/jnnp-2013-307343. Epub 2014 May 14. PMID: 24828898
- Eddy CM, Sira Mahalingappa S, Rickards HE. Putting things into perspective: the nature and impact of theory of mind impairment in Huntington’s disease. Eur Arch Psychiatry Clin Neurosci. 2014 Dec;264(8):697-705. doi: 10.1007/s00406-014-0498-4. Epub 2014 Mar 20. PMID: 24647535
- Lindenberg KS, Weydt P, Müller HP, Bornstedt A, Ludolph AC, Landwehrmeyer GB, Rottbauer W, Kassubek J, Rasche V. Two-point magnitude MRI for rapid mapping of brown adipose tissue and its application to the R6/2 mouse model of Huntington disease. PLoS One. 2014 Aug 21; 9(8):e105556. doi: 10.1371/journal.pone.0105556. eCollection 2014.PMID: 25144457
- Magnusson-Lind A, Davidsson M, Silajdžić E, Hansen C, McCourt AC, Tabrizi SJ, Björkqvist M.Skeletal muscle atrophy in R6/2 mice – altered circulating skeletal muscle markers and gene expression profile changes. J Huntingtons Dis. 2014;3(1):13-24. doi: 10.3233/JHD-130075. PMID: 25062762
- Quinn L, Debono K, Dawes H, Rosser AE, Nemeth AH, Rickards H, Tabrizi SJ, Quarrell O, Trender-Gerhard I, Kelson J, Townson J, Busse M; members of the TRAIN-HD project group. Task-specific training in Huntington disease: a randomized controlled feasibility trial. Phys Ther. 2014 Nov;94(11):1555-68. doi: 10.2522/ptj.20140123. Epub 2014 Jul 10. PMID: 25012999
- Reilmann R, Squitieri F, Priller J, Saft C, Mariotti C, Suessmuth SD, Nemeth AH, Tabrizi SJ, Quarrell O, Craufurd D, Rickards H, Rosser A, Borje D, Michaela T, Angieszka S, Fischer DF, Macdonald D, Munoz-Sanjuan I, Pacifici R, Frost C, Farmer R, Landwehrmeyer B and Westerberg G. Safety and Tolerability of Selisistat for the Treatment of Huntington’s Disease: Results from a Randomized, Double-Blind, Placebo-Controlled Phase II Trial (S47.004). Neurology April 8, 2014 vol. 82 no. 10 Supplement S47.004.
- Ross CA, Aylward EH, Wild EJ, Langbehn DR, Long JD, Warner JH, Scahill RI, Leavitt BR, Stout JC, Paulsen JS, Reilmann R, Unschuld PG, Wexler A, Margolis RL, Tabrizi SJ. Huntington disease: natural history, biomarkers and prospects for therapeutics. Nat Rev Neurol. 2014 Apr; 10(4):204-16. doi: 10.1038/nrneurol.2014.24. Epub 2014 Mar 11. PMID: 24614516
- Tourette C, Farina F, Vazquez-Manrique RP, Orfila AM, Voisin J, Hernandez S, Offner N, Parker JA, Menet S, Kim J, Lyu J, Choi SH, Cormier K, Edgerly CK, Bordiuk OL, Smith K, Louise A, Halford M, Stacker S, Vert JP, Ferrante RJ, Lu W, Neri C. The Wnt receptor Ryk reduces neuronal and cell survival capacity by repressing FOXO activity during the early phases of mutant huntingtin pathogenicity. PLoS Biol. 2014 Jun 24;12(6):e1001895. doi: 10.1371/journal.pbio.1001895. eCollection 2014 Jun. PMID: 24960609
- Vittori A, Breda C, Repici M, Orth M, Roos RA, Outeiro TF, Giorgini F, Hollox EJ; REGISTRY investigators of the European Huntington’s Disease Network. Copy-number variation of the neuronal glucose transporter gene SLC2A3 and age of onset in Huntington’s disease. Hum Mol Genet 2014 Jun 15; 23(12):3129-37. doi: 10.1093/hmg/ddu022. Epub 2014 Jan 22. PMID: 24452335
- Weydt P, Soyal SM, Landwehrmeyer GB, Patsch W; European Huntington Disease Network. A single nucleotide polymorphism in the coding region of PGC-1α is a male-specific modifier of Huntington disease age-at-onset in a large European cohort. BMC Neurol. 2014 Jan 2; 14:1. doi: 10.1186/1471-2377-14-1. PMID: 24383721
- EHDN 8th Plenary Meeting (September 19-21, 2014): J Neurol Neurosurg Psychiatry 2014 85(Suppl 1)
2013
- HORIZON Investigators of the Huntington Study Group and European Huntington’s Disease Network. A randomized, double-blind, placebo-controlled study of latrepirdine in patients with mild to moderate Huntington disease. JAMA Neurol. 2013 Jan; 70(1):25-33. doi: 10.1001/2013.jamaneurol.382. PMID: 23108692
- Bohlen S, Ekwall C, Hellström K, Vesterlin H, Björnefur M, Wiklund L, Reilmann R. Physical therapy in Huntington’s disease-toward objective assessments? Eur J Neurol. 2013 Feb;20(2):389-93. doi: 10.1111/j.1468-1331.2012.03760.x. Epub 2012 Jun 4. PMID: 22672573
- Delmaire C, Dumas EM, Sharman MA, van den Bogaard SJ, Valabregue R, Jauffret C, Justo D, Reilmann R, Stout JC, Craufurd D, Tabrizi SJ, Roos RA, Durr A, Lehéricy S. The structural correlates of functional deficits in early huntington’s disease. Hum Brain Mapp. 2013 Sep;34(9):2141-53. doi: 10.1002/hbm.22055. Epub 2012 Mar 22. PMID: 22438242
- Eatough V, Santini H, Eiser C, Goller ML, Krysa W, de Nicola ‚, Paduanello M, Petrollini M, Rakowicz M, Squitieri F, Tibben A, Weille KL, Landwehrmeyer B, Quarrell O, Smith JA. The personal experience of parenting a child with juvenile Huntington’s disease: perceptions across Europe. Eur J Hum Genet. 2013 Oct; 21(10):1042-8. doi: 10.1038/ejhg.2013.15. Epub 2013 Feb 27. PMID: 23443023
- Eddy CM, Rickards HE. Impact of cognitive and behavioural changes on quality of life in Huntington’s disease. Basal Ganglia 2013 Jul; 3(2):123-6.
- Harrison DJ, Busse M, Openshaw R, Rosser AE, Dunnett SB, Brooks SP. Exercise attenuates neuropathology and has greater benefit on cognitive than motor deficits in the R6/1 Huntington’s disease mouse model. Exp Neurol. 2013 Oct;248:457-69. doi: 10.1016/j.expneurol.2013.07.014. Epub 2013 Jul 30. PMID: 23911978
- Hart EP, Marinus J, Burgunder JM, Bentivoglio AR, Craufurd D, Reilmann R, Saft C, Roos RA; REGISTRY Investigators of the European Huntington’s Disease Network. Better global and cognitive functioning in choreatic versus hypokinetic-rigid Huntington’s disease. Mov Disord. 2013 Jul; 28(8):1142-5. doi: 10.1002/mds.25422. Epub 2013 Mar 14. PMID: 23495076
- Hubers AA, van Duijn E, Roos RA, Craufurd D, Rickards H, Bernhard Landwehrmeyer G, van der Mast RC, Giltay EJ; The REGISTRY investigators of the European Huntington’s Disease Network. Suicidal ideation in a European Huntington’s disease population. J Affect Disord. 2013 Oct; 151(1):248-58. doi: 10.1016/j.jad.2013.06.001. Epub 2013 Jul 20. PMID: 23876196
- Krzysztoń-Russjan J, Zielonka D, Jackiewicz J, Kuśmirek S, Bubko I, Klimberg A, Marcinkowski JT, Anuszewska EL. A study of molecular changes relating to energy metabolism and cellular stress in people with Huntington’s disease: looking for biomarkers. J Bioenerg Biomembr. 2013 Feb;45(1-2):71-85. doi: 10.1007/s10863-012-9479-3. Epub 2012 Oct 16. PMID: 23070563
- Metzger S, Walter C, Riess O, Roos RA, Nielsen JE, Craufurd D; REGISTRY Investigators of the European Huntington’s Disease Network, Nguyen HP. The V471A polymorphism in autophagy-related gene ATG7 modifies age at onset specifically in Italian Huntington disease patients. PLoS One. 2013 Jul 22; 8(7):e68951. doi: 10.1371/journal.pone.0068951. Print 2013. PMID: 23894380
- Parmentier F, Lejeune FX, Neri C. Pathways to decoding the clinical potential of stress response FOXO-interaction networks for Huntington’s disease: of gene prioritization and context dependence. Front Aging Neurosci. 2013 Jun 13; 5:22. doi: 10.3389/fnagi.2013.00022. eCollection 2013. PMID: 23781200
- Quinn L, Khalil H, Dawes H, Fritz NE, Kegelmeyer D, Kloos AD, Gillard JW, Busse M; Outcome Measures Subgroup of the European Huntington’s Disease Network. Reliability and minimal detectable change of physical performance measures in individuals with pre-manifest and manifest Huntington disease. Phys Ther. 2013 Jul;93(7):942-56. doi: 10.2522/ptj.20130032. Epub 2013 Mar 21. PMID: 23520147
- Simonin C, Duru C, Salleron J, Hincker P, Charles P, Delval A, Youssov K, Burnouf S, Azulay JP, Verny C, Scherer C, Tranchant C, Goizet C, Debruxelles S, Defebvre L, Sablonnière B, Romon-Rousseaux M, Buée L, Destée A, Godefroy O, Dürr A, Landwehrmeyer B; REGISTRY Study of the European Huntington’s Disease Network, Bachoud-Levi AC, Richard F, Blum D, Krystkowiak P; Huntington French Speaking Network. Association between caffeine intake and age at onset in Huntington’s disease. Neurobiol Dis. 2013 Oct;58:179-82. doi: 10.1016/j.nbd.2013.05.013. Epub 2013 May 31. PMID: 23732677
- Vittori A, Orth M, Roos RA, Outeiro TF, Giorgini F, Hollox EJ; REGISTRY investigators of the European Huntington’s Disease Network. β-Defensin Genomic Copy Number Does Not Influence the Age of Onset in Huntington’s Disease. J Huntingt Dis 2013 Mar 27; 2(1):107-24. doi: 10.3233/JHD-130002. PMID: 25057107
- Youssov K, Dolbeau G, Maison P, Boissé MF, Cleret de Langavant L, Roos RA, Bachoud-Lévi AC. Unified Huntington’s disease rating scale for advanced patients: validation and follow-up study. Mov Disord. 2013 Oct;28(12):1717-23. doi: 10.1002/mds.25654. PMID: 24166899
- Zielonka D, Marinus J, Roos RA, De Michele G, Di Donato S, Putter H, Marcinkowski J, Squitieri F, Bentivoglio AR, Landwehrmeyer GB. The influence of gender on phenotype and disease progression in patients with Huntington’s disease. Parkinsonism Relat Disord. 2013 Feb; 19(2):192-7. doi: 10.1016/j.parkreldis.2012.09.012. Epub 2012 Oct 25. PMID: 23102616
2012
- Lee JH, Lee JM, Ramos EM, Gillis T, Mysore JS, Kishikawa S, Hadzi T, Hendricks AE, Hayden MR, Morrison PJ, Nance M, Ross CA, Margolis RL, Squitieri F, Gellera C, Gomez-Tortosa E, Ayuso C, Suchowersky O, Trent RJ, McCusker E, Novelletto A, Frontali M, Jones R, Ashizawa T, Frank S, Saint-Hilaire MH, Hersch SM, Rosas HD, Lucente D, Harrison MB, Zanko A, Abramson RK, Marder K, Sequeiros J, Landwehrmeyer GB; Registry Study of the European Huntington’s Disease Network, Shoulson I; Huntington Study Group COHORT project, Myers RH, MacDonald ME, Gusella JF. TAA repeat variation in the GRIK2 gene does not influence age at onset in Huntington’s disease. Biochem Biophys Res Commun. 2012 Aug 3; 424(3):404-8. Epub 2012 Jul 3. PMID: 22771793
- Lee JM, Ramos EM, Lee JH, Gillis T, Mysore JS, Hayden MR, Warby SC, Morrison P, Nance M, Ross CA, Margolis RL, Squitieri F, Orobello S, Di Donato S, Gomez-Tortosa E, Ayuso C, Suchowersky O, Trent RJ, McCusker E, Novelletto A, Frontali M, Jones R, Ashizawa T, Frank S, Saint-Hilaire MH, Hersch SM, Rosas HD, Lucente D, Harrison MB, Zanko A, Abramson RK, Marder K, Sequeiros J, Paulsen JS; PREDICT-HD study of the Huntington Study Group (HSG), Landwehrmeyer GB; REGISTRY study of the European Huntington’s Disease Network, Myers RH; HD-MAPS Study Group, MacDonald ME, Gusella JF; COHORT study of the HSG. CAG repeat expansion in Huntington disease determines age at onset in a fully dominant fashion. Neurology. 2012 Mar 6; 78(10):690-5. doi: 10.1212/WNL.0b013e318249f683. Epub 2012 Feb 8. PMID: 22323755
- Lejeune FX, Mesrob L, Parmentier F, Bicep C, Vazquez-Manrique RP, Parker JA, Vert JP, Tourette C, Neri C. Large-scale functional RNAi screen in C. elegans identifies genes that regulate the dysfunction of mutant polyglutamine neurons. BMC Genomics. 2012 Mar 13;13:91. doi: 10.1186/1471-2164-13-91. PMID: 22413862
- Quarrell OW, Handley O, O’Donovan K, Dumoulin C, Ramos-Arroyo M, Biunno I, Bauer P, Kline M, Landwehrmeyer GB; European Huntington’s Disease Network. Discrepancies in reporting the CAG repeat lengths for Huntington’s disease. Eur J Hum Genet. 2012 Jan; 20(1):20-6. doi: 10.1038/ejhg.2011.136. Epub 2011 Aug 3. PMID: 21811303
- Rinaldi C, Salvatore E, Giordano I, De Matteis S, Tucci T, Cinzia VR, Rossi F, Castaldo I, Morra VB, Di Maio L, Filla A, De Michele G. Predictors of survival in a Huntington’s disease population from southern Italy. Can J Neurol Sci. 2012 Jan;39(1):48-51. PMID: 22384495
- Robertson L, Santini H, O’Donovan KL, Squitieri F, Barker RA, Rakowicz M, Landwehrmeyer GB, Quarrell O. Current Pharmacological Management in Juvenile Huntington’s Disease. PLoS Curr 2012 Feb 15; 4:RRN1304. PMID: 22474619
- Soyal SM, Felder TK, Auer S, Hahne P, Oberkofler H, Witting A, Paulmichl M, Landwehrmeyer GB, Weydt P, Patsch W; European Huntington Disease Network. A greatly extended PPARGC1A genomic locus encodes several new brain-specific isoforms and influences Huntington disease age of onset. Hum Mol Genet. 2012 Aug 1; 21(15):3461-73. doi: 10.1093/hmg/dds177. Epub 2012 May 15. PMID: 22589246
- EHDN 7th Plenary Meeting (September 14-16, 2012): J Neurol Neurosurg Psychiatry 83(Suppl 1)
2011
- Busse M, Al-Madfai DH, Kenkre J, Landwehrmeyer GB, Bentivoglio A, Rosser A; European Huntington’s Disease Network. Utilisation of Healthcare and Associated Services in Huntington’s disease: a data mining study. PLoS Curr. 2011 Jan 21; 3:RRN1206. doi: 10.1371/currents.RRN1206. PMID: 21304753
- López-Sendón JL, Royuela A, Trigo P, Orth M, Lange H, Reilmann R, Keylock J, Rickards H, Piacentini S, Squitieri F, Landwehrmeyer B, Witjes-Ane MN, Jurgens CK, Roos RA, Abraira V, de Yébenes JG; European HD Network. What is the impact of education on Huntington’s disease? Mov Disord. 2011 Jul; 26(8):1489-95. doi: 10.1002/mds.23385. Epub 2011 Mar 22. PMID: 21432905
- Orth M; European Huntington’s Disease Network, Handley OJ, Schwenke C, Dunnett S, Wild EJ, Tabrizi SJ, Landwehrmeyer GB. Observing Huntington’s disease: the European Huntington’s Disease Network’s REGISTRY. J Neurol Neurosurg Psychiatry. 2011 Dec; 82(12):1409-12. doi: 10.1136/jnnp.2010.209668. Epub 2010 Nov 19. PMID: 21097549
- Orth M, Schwenke C. Age-at-onset in Huntington disease. PLoS Curr. 2011 Jul 29; 3:RRN1258. doi: 10.1371/currents.RRN1258. PMID: 22453877
- Reilmann R, Bohlen S, Kirsten F, Ringelstein EB, Lange HW. Assessment of involuntary choreatic movements in Huntington’s disease-toward objective and quantitative measures. Mov Disord. 2011 Oct;26(12):2267-73. doi: 10.1002/mds.23816. Epub 2011 Jun 9. PMID: 21661053
- Rickards H, De Souza J, Crooks J, van Walsem MR, van Duijn E, Landwehrmeyer B, Squitieri F, Simpson SA; European Huntington’s Disease Network. Discriminant analysis of Beck Depression Inventory and Hamilton Rating Scale for Depression in Huntington’s disease. J Neuropsychiatry Clin Neurosci. 2011 Fall; 23(4):399-402. doi: 10.1176/appi.neuropsych.23.4.399. PMID: 22231310
- Rickards H, De Souza J, van Walsem M, van Duijn E, Simpson SA, Squitieri F, Landwehrmeyer B; European Huntington’s Disease Network. Factor analysis of behavioural symptoms in Huntington’s disease. J Neurol Neurosurg Psychiatry. 2011 Apr; 82(4):411-2. doi: 10.1136/jnnp.2009.181149. Epub 2010 Apr 14. PMID: 20392980
- Saft C, Epplen JT, Wieczorek S, Landwehrmeyer GB, Roos RA, de Yebenes JG, Dose M, Tabrizi SJ, Craufurd D; REGISTRY Investigators of the European Huntington’s Disease Network, Arning L. NMDA receptor gene variations as modifiers in Huntington disease: a replication study. PLoS Curr. 2011 Oct 4; 3:RRN1247. doi: 10.1371/currents.RRN1247. PMID: 21989477
- Vaccarino AL, Anderson K, Borowsky B, Duff K, Giuliano J, Guttman M, Ho AK, Orth M, Paulsen JS, Sills T, van Kammen DP, Evans KR; PREDICT-HD and REGISTRY Investigators Coordinators. An item response analysis of the motor and behavioral subscales of the unified Huntington’s disease rating scale in huntington disease gene expansion carriers. Mov Disord. 2011 Apr; 26(5):877-84. doi: 10.1002/mds.23574. Epub 2011 Mar 2. PMID: 21370269
2010
- Henley SM, Ridgway GR, Scahill RI, Klöppel S, Tabrizi SJ, Fox NC, Kassubek J; EHDN Imaging Working Group. Pitfalls in the use of voxel-based morphometry as a biomarker: examples from huntington disease. AJNR Am J Neuroradiol. 2010 Apr; 31(4):711-9. doi: 10.3174/ajnr.A1939. Epub 2009 Dec 24. PMID: 20037137
- Orth M, Handley OJ, Schwenke C, Dunnett SB, Craufurd D, Ho AK, Wild E, Tabrizi SJ, Landwehrmeyer GB; Investigators of the European Huntington’s Disease Network. Observing Huntington’s Disease: the European Huntington’s Disease Network’s REGISTRY. PLoS Curr. 2010 Sep 28 [revised 2011 Apr 13]; 2:RRN1184. PMID: 20890398
- De Souza J, Jones LA, Rickards H. Validation of self-report depression rating scales in Huntington’s disease. Mov Disord. 2010 Jan 15; 25(1):91-6. doi: 10.1002/mds.22837. PMID: 19908314
- EHDN 6th Plenary Meeting (September 2010): J Neurol Neurosurg Psychiatry 81(Suppl 1)
2009
- Aziz NA, Jurgens CK, Landwehrmeyer GB; EHDN Registry Study Group, van Roon-Mom WM, van Ommen GJ, Stijnen T, Roos RA. Normal and mutant HTT interact to affect clinical severity and progression in Huntington disease. Neurology. 2009 Oct 20; 73(16):1280-5. doi: 10.1212/WNL.0b013e3181bd1121. Epub 2009 Sep 23. Erratum in: Neurology. 2009 Nov 10;73(19):1608. Neurology. 2011 Jan 11;76(2):202. Ciarmielo, Andrea [corrected to Ciarmiello, Andrea]. PMID: 19776381
- Tabrizi SJ, Langbehn DR, Leavitt BR, Roos RA, Durr A, Craufurd D, Kennard C, Hicks SL, Fox NC, Scahill RI, Borowsky B, Tobin AJ, Rosas HD, Johnson H, Reilmann R, Landwehrmeyer B, Stout JC; TRACK-HD investigators. Biological and clinical manifestations of Huntington’s disease in the longitudinal TRACK-HD study: cross-sectional analysis of baseline data. Lancet Neurol. 2009 Sep; 8(9):791-801. doi: 10.1016/S1474-4422(09)70170-X. Epub 2009 Jul 29. PMID: 19646924
- Weydt P, Soyal SM, Gellera C, Didonato S, Weidinger C, Oberkofler H, Landwehrmeyer GB, Patsch W. The gene coding for PGC-1alpha modifies age at onset in Huntington’s Disease. Mol Neurodegener. 2009 Jan 8;4:3. doi: 10.1186/1750-1326-4-3. PMID: 19133136
2008
- Klöppel S, Draganski B, Golding CV, Chu C, Nagy Z, Cook PA, Hicks SL, Kennard C, Alexander DC, Parker GJ, Tabrizi SJ, Frackowiak RS. White matter connections reflect changes in voluntary-guided saccades in pre-symptomatic Huntington’s disease. Brain. 2008 Jan;131(Pt 1):196-204. Epub 2007 Dec 3. PMID: 18056161
- Priller J, Ecker D, Landwehrmeyer B, Craufurd D. A Europe-wide assessment of current medication choices in Huntington’s disease. Mov Disord. 2008 Sep 15;23(12):1788. (Letters to the editor) doi: 10.1002/mds.22188. PMID: 18649399
- EHDN 5th Plenary Meeting (September 4-7, 2010): J Neurol Neurosurg Psychiatry 79(Suppl 1)
last update October 2024