Open Access

Recent approaches on Huntington's disease (Review)

  • Authors:
    • Anastasia Marina Palaiogeorgou
    • Eleni Papakonstantinou
    • Rebecca Golfinopoulou
    • Markezina Sigala
    • Thanasis Mitsis
    • Louis Papageorgiou
    • Io Diakou
    • Katerina Pierouli
    • Konstantina Dragoumani
    • Demetrios A. Spandidos
    • Flora Bacopoulou
    • George P. Chrousos
    • Elias Eliopoulos
    • Dimitrios Vlachakis
  • View Affiliations

  • Published online on: November 21, 2022     https://doi.org/10.3892/br.2022.1587
  • Article Number: 5
  • Copyright: © Palaiogeorgou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Huntington's disease (HD) is a neurodegenerative disorder characterized by severe motor, cognitive and psychiatric symptoms. Patients of all ages can present with a dysfunction of the nervous system, which leads to the progressive loss of movement control and disabilities in speech, swallowing, communications, etc. The molecular basis of the disease is well‑known, as HD is related to a mutated gene, a trinucleotide expansion, which encodes to the huntingtin protein. This protein is linked to neurogenesis and the loss of its function leads to neurodegenerative disorders. Although the genetic cause of the disorder has been known for decades, no effective treatment is yet available to prevent onset or to eliminate the progression of symptoms. Thus, the present review focused on the development of novel methods for the timely and accurate diagnosis of HD in an aim to aid the development of therapies which may reduce the severity of the symptoms and control their progression. The majority of the therapies include gene‑silencing mechanisms of the mutated huntingtin gene aiming to suppress its expression, and the use of various substances as drugs with highly promising results. In the present review, the latest approaches on the diagnosis of HD are discussed along with the need for genetic counseling and an up‑to‑date presentation of the applied treatments.

1. Introduction

Neurodegenerative disorders have a exhibited a marked increase in incidence worldwide, thus rendering them a primary concern for the scientific society. The genetic cause of numerous disorders has already been described (1-3) and, nowadays, research focuses on the timely diagnosis and effective therapy of the most common neurodegenerative disorders, such as Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis and Huntington's disease (HD). These disorders have diverse clinical manifestations; however, some of them demonstrate similarities among patients (4,5). Although in numerous cases, the onset of neurodegenerative disorders appears in middle to late adult life, there are patients who manifest symptoms of these disorders at a very early age (6,7). HD is one of the most common disorders with severe symptomatology, which affects individuals of all ages, progressively leading to severe disabilities. The genetic basis of this disorder has been established and has been known for a few decades now, and recent research has revealed promising mechanisms for eliminating HD symptoms (8). Furthermore, HD can be regarded as a model neurodegenerative disorder for the study of other cases with shared symptoms, and knowledge of other diseases may be useful for HD diagnosis and treatment.

2. Genetics and pathology of Huntington's disease

HD is a fatal, autosomal dominant, progressive neurodegenerative disorder characterized by severe symptoms, including motor, cognitive and psychiatric symptoms, atrophy of the basal ganglia and the cerebral cortex, and an inevitably progressive course, resulting in mortality 5-20 years following the manifestation of symptoms. Typically, the motor defects include chorea and loss of coordination, and patients also demonstrate difficulty with speech and swallowing (9). Cognitive symptoms can be detected up to a decade prior to diagnosis and cognitive ability declines as the disease progresses (10). Psychiatric symptoms, such as depression, psychosis and obsessive-compulsive disorder, are also common in HD and are particularly distressing for patients (11,12). Patients with HD eventually require a wheelchair and more severe symptoms may lead to them becoming bedridden, with all the complications that may derive from that form of immobility.

From a neuropathological point of view, in patients with HD, the dysfunction and death of specific neurons within their brains are observed. There is a wide range in the age of onset of HD, as both juveniles (13,14) and adults have been diagnosed with the disorder thus far. For instance, kindred members of families that revealed a history consistent with HD autosomal dominant inheritance, took part in a 20-year study, which was published in 2004(15). The researchers of that study found that the typical ages of disease onset were between 21 and 50 years of age (15). Although the disorder typically manifests in adulthood, juvenile HD (JHD) is also frequent among patients (16). A recent study, conducted in Argentina in 2015(17), reported that almost 20% of the patients diagnosed with HD, revealed their first symptoms of the disorder during their childhood. It should be noted that the overall estimated prevalence of JHD of that study was higher than that in any other population recorded to date (17). The brain structure in young patients was previously assessed by Tereshchenko et al (18) in 2019, proposing that the morphology differs among juveniles and adult patients, as young patients revealed proportional cerebellar enlargement (18). In the same year, another study suggested that the pathogenesis of HD begins with abnormal brain development in both child and adolescent patients (19).

The first attempt to discover the genetic cause for HD by Gusella et al (20) revealed that the HD gene is linked to a polymorphic DNA marker that maps to human chromosome 4, in particular 4p16.3. A decade later, a Huntington's Disease Collaborative Research Group discovered that mutations in the Huntington (HTT) gene encoding the huntingtin protein, a large protein of 3,144 amino acids, led to the neurodegenerative disorder (21). In particular, they suggested that the disorder is caused due to a cytosine-adenine-guanine (CAG) trinucleotide expansion in exon 1 that codes for polyglutamine (polyQ) in the N-terminal of the HTT gene (21). The CAG sequence is normally repeated 9 to 35 times, with an average median of between 17 and 20 repeats. However, patients with HD usually reveal a CAG expansion exceeding 35 repeats (22). Above a threshold of ~35 CAG repeats, the age of onset of HD is inversely associated with the length of the expansion. A recent study conducted by Schultz et al (23) demonstrated that the development of verbal skills appeared to plateau earlier as CAG repeat length increased. The repeats are usually between 36 and 39, depending on the age. Juveniles with HD exhibit high repeat lengths (24). In certain rare cases, patients exhibit less repeats in their genome, 27-35, demonstrating an endophenotype (25).

Of note, the huntingtin protein is expressed in all cell types of the body, both at the tissue and subcellular level, in all developmental stages. Recently, research has focused on the investigation of the HTT structure via cryo-electron microscopy contributing to a better comprehension of its morphology and form abnormalities (26). It has been described as a 350-kDa HEAT-repeat protein which interacts with hundreds of other proteins (27) and participates in numerous cellular processes. Although the cellular functions of HTT protein are not yet completely understood, it appears to play a crucial role during early embryonic development and neurogenesis. In particular, Saudou and Humbert (22) described the human huntingtin protein sequence and its post-translational modifications in detail. They suggested that it coordinates cell division, as it participates in the proper mitotic spindle positioning and it regulates ciliogenesis. They also noted that huntingtin mediates endocytosis, vesicle recycling and endosomal trafficking, as it interacts with other proteins which are related to these mechanisms. Other functions of the protein include autophagy and transcription (22). It should be underlined that the HTT gene physiological expression is essential for organism homeostasis as it plays a neuroprotective role as well, even against mutant HTT (mHtt) toxicity (28,29). Moreover, HTT protein plays a crucial role in mitochondrial structure and function in the embryogenesis and oxidative metabolism, and HTT mutations have been linked to mitochondrial abnormalities (30-32).

The role of both wild-type Htt and mHtt in gene silencing studies has been investigated for the development of an effective therapy. As regards mHtt molecules, they form toxic aggregates into the central nervous system, depending on the length of polyQ expansion. For instance, mHtt co-aggregates with other proteins which play a crucial role in the cell, leading to misfunctioned phenotypes. Numerous studies have focused on the effects of HTT gene knockouts and knockdowns in cellular function. For example, a study published in 2017 suggested that mutations in HTT protein are related to nucleocytoplasmic transport disruption, leading to the improper function of cells (33). During initial experiments performed on mice with HTT knockdown mutants, the mice succumbed after 8 days of gestation (34). Other studies have demonstrated that HTT deletion in the mouse central nervous system leads to a phenotype similar to that of HD (35,36). It is worth mentioning that a recent study suggested that HTT variants are also linked to another disorder with similar symptomatology with HD, the so-called Lopes-Maciel-Rodan syndrome (37). In addition, other studies have revealed the ability of various molecular chaperones, such as the heat shock family proteins, HSP40, HSP70, HSP90 and HSP105, to combine with misfolded mHtt and inhibit aggregate formation, leading to cell survival (38,39).

It is clear that the loss of HTT function contributes to HD pathology and for this reason, it is essential for survival. The reduction of mHtt levels should be accompanied by regular HTT expression.

3. Diagnosis and genetic counseling

It has already been mentioned that the age of onset of HD is inversely associated with the length of the expansion in the HTT gene. For instance, rare carriers of 36 to 39 CAG repeats have lower penetrance and a later onset of the disease than those with 40 or more CAG repeats. Additionally, Keum et al (40) found that, along with clinical onset, the age of patients with HD at the time of death was well determined by an expanded CAG-repeat length. However, they claimed that the overall duration of the disease was independent of the length of the mutation's (40). These data may be useful, not only for the molecular diagnosis of the disorder, but also for the prediction of the outset of HD symptomatology. For the molecular diagnosis of the disorder, various PCR methods have been demonstrated in order to detect CAG expansions (41). A recent study presented a novel triplet-primed PCR-based assay aiming to improve the test reliability and accuracy by detecting CAG expansions in samples with sequence variations in the HTT gene (42).

It is known that miRNAs are involved in the biological processes of development, proliferation, inflammation and apoptosis, and their expression has been linked to HD diagnosis and symptomatology. For instance, Langfelder et al (43) found that the abnormal expression of miRNAs played a critical role in HD pathogenesis. For this reason, apart from the direct quantification of mHTT itself, which is the main disease-related biomarker, other miRNAs may be useful tools as biomarkers for HD prognosis (44).

Furthermore, numerous diagnostic tests have been proposed thus far, based on criteria related to inheritance and the symptomatology of the individual; however, these methods need to be improved. Patients who experience certain cognitive and behavioral symptoms may have HD (45,46). A recent study proposed the Enroll-HD dataset for estimating disease onset and its diagnostic confidence level (47). The results of that study were not promising, suggesting that it is important to develop more reliable diagnostic criteria (47). Another diagnostic approach suggested that the concentration of trace elements in the blood of patients with HD differs from that of healthy individuals. Researchers found increased levels of the essential elements iron, chromium, selenium and zinc and of the non-essential element, arsenic, in the blood of patients with HD, suggesting that the blood metal profile may be used as an easy tool for the disorder's medical detection (48).

HD follows the Gregor Mendel's principles of inheritance, as it is inherited in an autosomal-dominant manner. The offspring of an individual with a pathogenic variant, heterozygote, have a 50% chance of inheriting the disease-causing allele. Genetic counseling includes predictive testing in asymptomatic adults and prenatal testing in order to reveal the mutated allele (49). The prevalence of HD is ~1 in 10,000 individuals in the USA, as well as in Europe (50,51). In the year 2000, Sobel and Cowan (52) conducted predictive testing on asymptomatic individuals at risk of developing HD in the context of genetic counseling. Family members were requested to describe their communication and interactions with the social environment, and provide concerns about their future care. Members in 50% of the families experienced changes in patterns of communication and 56% of the participants reported changes in current relationships. The researchers suggested that families may benefit in pretest sessions by examining their patterns of dealing with illness issues, both past and present (52).

Migliore et al (53) suggested different approaches of counseling, depending on the genetic condition of the individual. For instance, in the case of intermediate alleles (27-35 CAG repeats) the experts should explain the potential risk of mutations and other members of the family should also be tested. In the case of low penetrance alleles (36-39 CAG repeats), individuals should be informed about the risk of HD symptoms manifesting at any age. Counseling for all family members is also required when juveniles are diagnosed with JHD. When the HD mutation is detected in a prenatal genetic test, the parents should be informed for the risk of the newborn manifesting the disease and should be given the option of terminating the pregnancy (53). This approach utilizes the current knowledge of the molecular basis of HD with the inclusive genetic counseling of all relatives. Recently, MacLeod et al (54) proposed a family systems approach to genetic counseling, which uses the narrative model. With the narrative resources, the genetic counselor can contribute to generate new meanings that the person may give to their experience of the genetic condition and help the patient adapt to living with the disorder or its risks (54). Another interesting comparative study, that was performed over the past two decades, on how parents inform their children who are at risk about their genetic risk demonstrated that, although testing is performed more often, the overall attitude towards information and testing has not changed significantly (55). This ascertainment proposes that new methods for more comprehensible information and accessible genetic counseling need be developed.

4. Treatment

Research focusing on understanding the underlying molecular mechanisms leading to the HTT gene mutations is highly promising, aiming to find a cure for HD. However, current treatments for HD are still limited. The therapies applied focus on the treatment of symptoms, as neuroprotective therapies to prevent disease onset and to attenuate the progression of the disease are not yet available. For instance, it has been proven that HD is caused by toxic properties of mHTT, rather than merely the decrease of wild-type HTT; for this reason, approaches focusing on mHTT expression, such as lowering HTT mRNA and mutant huntingtin protein, appear to be promising (56). The main strategies which have been demonstrated thus far as treatments for HD are presented in Fig. 1.

A recent study proposed that targeting of CAG repeat-dependent mechanisms, through gene-silencing approaches, may affect the rate of functional, motor and cognitive impairment, but not weight loss, in manifest HD mutation carriers (57). The standard approaches to DNA targeting use some form of specific DNA-binding element combined with nucleases, epigenetic modulators, or transcription factors. Zinc-finger transcriptional repressor approaches may lower mHTT levels by targeting DNA without altering it, whereas zinc-finger nucleases can add to the repressive effect of Zinc-finger proteins that reduce the levels of gene expression by simply binding to DNA and preventing gene transcription by actually disrupting or correcting the mutant gene (58). Along with similar techniques to other direct genome editing strategies, such as CRISPR/Cas9, strategies that are targeted in lowering huntingtin and HTT genome editing have immense potential for the treatment of HD. The main advantage is the permanent correction of the disease-causing CAG expansion. An antisense mechanism targeting HTT RNA, using synthetic antisense oligonucleotides (ASOs) that bind to the specific sequence of ribonucleic acid, may reduce the mRNA translation to the HTT disease-causing protein (59). The ASOs are widely distributed throughout the central nervous system and they do not require a viral or lipid carrier, resulting to an effective and simple to develop treatment. Tabrizi et al (60) used the antisense oligonucleotide IONIS-HTTRx designed to inhibit HTT mRNA by triggering the RNase H1-mediated degradation of the target mRNA, in order to minimize the concentration of mutant huntingtin in cerebrospinal fluid. They conducted an extended research with 34 patients who were treated with increasing dose levels of 10 to 120 mg and the observations were compared with individuals who received the placebo. The results revealed that the reduction in concentrations of mutant huntingtin was dose-dependent (60). The strategy for post-transcriptional gene suppression using non-coding double-stranded RNA sequences is known as the RNA interference (RNAi) mechanism. The RNAi pathway has been used thus far to suppress specific genes of interest and the results are highly promising for numerous diseases. There are various molecules which can be used for this purpose, such as siRNAs, shRNAs and artificial miRNAs that have been used to eliminate the HD symptoms. The first trials were performed two decades ago in rodents. In 2005, Harper et al (61) used a shRNA molecule to target the HTT mutant gene and the results were satisfactory, as the reduction in mHTT synthesis, prevented inclusions, gait deficits and rotarod dysfunction. In another case, a siRNA molecule was injected into the mouse striatum, and the reduction in mHTT synthesis prolonged striatal neuron survival, reduced aggregates and prevented motor dysfunction (62). The application of siRNA approaches has been successful in multiple animal systems (63). In a recent study for example, a single-stranded siRNA (ss-siRNA) was used for RNAi, resulting in a selective decrease of CAG-expanded HTT protein in various regions throughout the mouse brain (64). Other ribonucleic acids, such as miRNAs have been used for the suppression of mHTT in genetically modified mice and the results have been promising; in one case, this strategy led to the prevention of regional cortical and striatal atrophy, and reduced weight loss (65). It is worth mentioning that the majority of the previous technics, apart from gene editing, effectively reduce, but do not completely eliminate the production of mHTT.

In addition, the therapeutic approach of overexpressing wild-type HTT has been investigated. Early trials of inserting the wild-type HTT into mammalian cells which expressed mHTT have led to reduced cell death (26).

Numerous research studies have used viral vectors, such as adeno-associated virus, which encapsulate the RNA molecules, and their genome is combined with enhancers and promoters, in order to deliver these agents by injection into the body (66).

Different compounds may be candidates for the treatment of neurodegenerative disorders, including HD. A recent study proposed the utilization of flavonoids, which may reduce cellular stress and play an anti-inflammatory and anti-apoptotic role in the cell (67). In another case, researchers proposed that marine compounds may be used for the treatment of various neurodegenerative diseases, as they also demonstrate antioxidant, anti-inflammatory and anti-apoptotic properties. Tetrabenazine (TBZ), which is an inhibitor that blocks dopamine uptake into vesicles, has been shown to exert antichorea effects in patients with HD and was the first approved drug for medication (68). Since then, studies on the optimization of drug delivery and bioavailability of TBZ in patients have been conducted based on latest nanotechnology technics (69). Several molecules have been suggested for the treatment of Parkinson's disease, such as fucoidan and xyloketal B, and fucoxanthin and cerebrosides for Alzheimer's disease, and have also been investigated for other disorders, such as HD for effective treatment (70). In 2020, Jabłońska et al (71) suggested that pridopidine, a dopamine stabilizer, may be a promising drug for HD symptoms. It is well-known that there is an association between the amount of dopamine in the central nervous system and the stage of the disease, as the causes of HD are dopaminergic conduction disorders, and experiments on animal models have demonstrated the protective effect of pridopidine on nerve cells (72,73). Τhe main advantage of drug treatment for HD is that the effectiveness and tolerance of each active compound is well-studied for other neurodegenerative diseases with similar symptomatology. Thus, it is easier to design a suitable medication personalized to patient diagnostics. Another study proposed that the application of monoclonal antibody, which targets the HTT protein may deplete its concentration in the cell, proving that monoclonal antibodies can interfere with the pathological processes of mHTT spreading in vivo (74).

Cell replacement therapy for HD using stem cells may be another opportunity to alleviate symptoms in patients (75). Furthermore, some case studies have indicated that exercise and physical activity may be beneficial for patients in terms of motor function, gait speed and balance, and social benefits have been also identified (76). Thus, exercise may play a complementary role in the treatment of the disorder.

5. Conclusions and future perspectives

HD is the first trinucleotide disease that was described and the first autosomal-dominant disease with a possible diagnosis prior to the manifestation of symptoms. Since 1983 and the localization of the gene, knowledge of the disorder has markedly increased, which is necessary in order to improve the quality of life of patients and improve therapeutic strategies by discovering novel molecular targets. The pathophysiology of HD is significant for designing and developing proper treatments (77). Science offers possibilities for attenuating the symptoms of the disease, and even the onset; however, it is also critical to identify effective biomarkers that may help prevent HD manifestation by early detection and blocking its course. Modern therapeutic trial design also vastly relies on identifying and examining biomarkers relevant to each disease.

The next step may be to evaluate and use data from genome-wide association studies and account for their clinical utility. Studies (as aforementioned) towards this direction have contributed to the existing knowledge concerning the association of genetic variations to the onset of symptoms and the progression of HD. The combination of early testing in order to predict the possible HD onset and new targeted and personalized medicine represents the future in preventing and hopefully, eliminating neurodegenerative diseases. The path for science ahead to help patients with HD is a long one. Until then, finding the optimal care for patients and caregivers is significant.

Acknowledgements

Not applicable.

Funding

Funding: The authors would like to acknowledge funding from the following organizations: i) AdjustEBOVGP-Dx (RIA2018EF-2081): Biochemical Adjustments of native EBOV Glycoprotein in Patient Sample to Unmask target Epitopes for Rapid Diagnostic Testing. A European and Developing Countries Clinical Trials Partnership (EDCTP2) under the Horizon 2020 ‘Research and Innovation Actions’ DESCA; ii) ‘MilkSafe: A novel pipeline to enrich formula milk using omics technologies’, a research co-financed by the European Regional Development Fund of the European Union and Greek national funds through the Operational Program Competitiveness, Entrepreneurship and Innovation, under the call RESEARCH-CREATE-INNOVATE (project code: T2EDK-02222); iii) ‘INSPIRED-The National Research Infrastructures on Integrated Structural Biology, Drug Screening Efforts and Drug Target Functional Characterization’ (Grant MIS 5002550) implemented under the Action ‘Reinforcement of the Research and Innovation Infrastructure’, funded by the Operational Program ‘Competitiveness, Entrepreneurship and Innovation’ (NSRF 2014-2020) and co-financed by Greece and the European Union (European Regional Development Fund), and iv) ‘OPENSCREENGR An Open-Access Research Infrastructure of Chemical Biology and Target-Based Screening Technologies for Human and Animal Health, Agriculture and the Environment’ (Grant MIS 5002691), implemented under the Action ‘Reinforcement of the Research and Innovation Infrastructure’, funded by the Operational Program ‘Competitiveness, Entrepreneurship and Innovation’ (NSRF 2014-2020) and co-financed by Greece and the European Union (European Regional Development Fund).

Availability of data and materials

Not applicable.

Authors' contributions

All authors (AMP, EP, RG, MS, TM, LP, ID, KP, KD, DAS, FB, GPC, EE and DV) contributed to the conceptualization, design, writing, drafting, revising, editing and reviewing of the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

DAS is the Editor-in-Chief for the journal, but had no personal involvement in the reviewing process, or any influence in terms of adjudicating on the final decision, for this article. The other authors declare that they have no competing interests.

References

1 

Recabarren D and Alarcon M: Gene networks in neurodegenerative disorders. Life Sci. 183:83–97. 2017.PubMed/NCBI View Article : Google Scholar

2 

Pihlstrøm L, Wiethoff S and Houlden H: Genetics of neurodegenerative diseases: An overview. Handb Clin Neurol. 145:309–323. 2017.PubMed/NCBI View Article : Google Scholar

3 

Shen T, You Y, Joseph C, Mirzaei M, Klistorner A, Graham SL and Gupta V: BDNF polymorphism: A review of its diagnostic and clinical relevance in neurodegenerative disorders. Aging Dis. 9(523)2018.PubMed/NCBI View Article : Google Scholar

4 

Guzman-Martinez L, Maccioni RB, Andrade V, Navarrete LP, Pastor MG and Ramos-Escobar N: Neuroinflammation as a common feature of neurodegenerative disorders. Front Pharmacol. 10(1008)2019.PubMed/NCBI View Article : Google Scholar

5 

Ma MW, Wang J, Zhang Q, Wang R, Dhandapani KM, Vadlamudi RK and Brann DW: NADPH oxidase in brain injury and neurodegenerative disorders. Mol Neurodegener. 12(7)2017.PubMed/NCBI View Article : Google Scholar

6 

Mendez MF: Early-onset Alzheimer disease and its variants. Continuum (Minneap Minn). 25:34–51. 2019.PubMed/NCBI View Article : Google Scholar

7 

Bakels HS, Roos RAC, van Roon-Mom WMC and de Bot ST: Juvenile-Onset huntington disease pathophysiology and neurodevelopment: A review. Mov Disord. 37:16–24. 2022.PubMed/NCBI View Article : Google Scholar

8 

Kumar A, Kumar V, Singh K, Kumar S, Kim YS, Lee YM and Kim JJ: Therapeutic advances for Huntington's disease. Brain Sci. 10(43)2020.PubMed/NCBI View Article : Google Scholar

9 

Yanagisawa N: The spectrum of motor disorders in Huntington's disease. Clin Neurol Neurosurg. 94 (Suppl):S182–S184. 1992.PubMed/NCBI View Article : Google Scholar

10 

Bonner-Jackson A, Long JD, Westervelt H, Tremont G, Aylward E and Paulsen JS: PREDICT-HD Investigators and Coordinators of the Huntington Study Group. Cognitive reserve and brain reserve in prodromal Huntington's disease. J Int Neuropsychol Soc. 19:739–750. 2013.PubMed/NCBI View Article : Google Scholar

11 

Harrington DL, Liu D, Smith MM, Mills JA, Long JD, Aylward EH and Paulsen JS: Neuroanatomical correlates of cognitive functioning in prodromal Huntington disease. Brain Behav. 4:29–40. 2014.PubMed/NCBI View Article : Google Scholar

12 

Epping EA, Mills JA, Beglinger LJ, Fiedorowicz JG, Craufurd D, Smith MM, Groves M, Bijanki KR, Downing N, Williams JK, et al: Characterization of depression in prodromal Huntington disease in the neurobiological predictors of HD (PREDICT-HD) study. J Psychiatr Res. 47:1423–1431. 2013.PubMed/NCBI View Article : Google Scholar

13 

van Dijk JG, van der Velde EA, Roos RA and Bruyn GW: Juvenile huntington disease. Hum Genet. 73:235–239. 1986.PubMed/NCBI View Article : Google Scholar

14 

Siesling S, Vegter-van der Vlis M and Roos RA: Juvenile Huntington disease in the Netherlands. Pediatr Neurol. 17:37–43. 1997.PubMed/NCBI View Article : Google Scholar

15 

Wexler NS, Lorimer J, Porter J, Gomez F, Moskowitz C, Shackell E, Marder K, Penchaszadeh G, Roberts SA, Gayán J, et al: Venezuelan kindreds reveal that genetic and environmental factors modulate Huntington's disease age of onset. Proc Natl Acad Sci USA. 101:3498–3503. 2004.PubMed/NCBI View Article : Google Scholar

16 

Quarrell OWJ, Brewer HM, Squitieri F, Barker R, Nance MA and Lanswehrmeyer BG: ‘Juvenile Huntington's Disease (and other trinucleotide repeat disorders)’. Oxford University Press, Oxford, 2009.

17 

Gatto EM, Parisi V, Etcheverry JL, Sanguinetti A, Cordi L, Binelli A, Persi G and Squitieri F: Juvenile Huntington disease in Argentina. Arq Neuropsiquiatr. 74:50–54. 2016.PubMed/NCBI View Article : Google Scholar

18 

Tereshchenko A, Magnotta V, Epping E, Mathews K, Espe-Pfeifer P, Martin E, Dawson J, Duan W and Nopoulos P: Brain structure in juvenile-onset Huntington disease. Neurology. 92:e1939–e1947. 2019.PubMed/NCBI View Article : Google Scholar

19 

van der Plas E, Langbehn DR, Conrad AL, Koscik TR, Tereshchenko A, Epping EA, Magnotta VA and Nopoulos PC: Abnormal brain development in child and adolescent carriers of mutant huntingtin. Neurology. 93:e1021–e1030. 2019.PubMed/NCBI View Article : Google Scholar

20 

Gusella JF, Wexler NS, Conneally PM, Naylor SL, Anderson MA, Tanzi RE, Watkins PC, Ottina K, Wallace MR, Sakaguchi AY, et al: A polymorphic DNA marker genetically linked to Huntington's disease. Nature. 306:234–238. 1983.PubMed/NCBI View Article : Google Scholar

21 

A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington's disease chromosomes. The Huntington's Disease Collaborative Research Group. Cell. 72:971–983. 1993.PubMed/NCBI View Article : Google Scholar

22 

Saudou F and Humbert S: The biology of Huntingtin. Neuron. 89:910–926. 2016.PubMed/NCBI View Article : Google Scholar

23 

Schultz JL, van der Plas E, Langbehn DR, Conrad AL and Nopoulos PC: Age-Related cognitive changes as a function of CAG repeat in child and adolescent carriers of mutant Huntingtin. Ann Neurol. 89:1036–1040. 2021.PubMed/NCBI View Article : Google Scholar

24 

Andrew SE, Goldberg YP, Kremer B, Telenius H, Theilmann J, Adam S, Starr E, Squitieri F, Lin B, Kalchman MA, et al: The relationship between trinucleotide (CAG) repeat length and clinical features of Huntington's disease. Nat Genet. 4:398–403. 1993.PubMed/NCBI View Article : Google Scholar

25 

Squitieri F and Jankovic J: Huntington's disease: How intermediate are intermediate repeat lengths? Mov Disord. 27:1714–1717. 2012.PubMed/NCBI View Article : Google Scholar

26 

Ho LW, Brown R, Maxwell M, Wyttenbach A and Rubinsztein DC: Wild type Huntingtin reduces the cellular toxicity of mutant Huntingtin in mammalian cell models of Huntington's disease. J Med Genet. 38:450–452. 2001.PubMed/NCBI View Article : Google Scholar

27 

Shirasaki DI, Greiner ER, Al-Ramahi I, Gray M, Boontheung P, Geschwind DH, Botas J, Coppola G, Horvath S, Loo JA and Yang XW: Network organization of the huntingtin proteomic interactome in mammalian brain. Neuron. 75:41–57. 2012.PubMed/NCBI View Article : Google Scholar

28 

Nuzzo MT and Marino M: Estrogen/Huntingtin: A novel pathway involved in neuroprotection. Neural Regen Res. 11:402–403. 2016.PubMed/NCBI View Article : Google Scholar

29 

Leavitt BR, van Raamsdonk JM, Shehadeh J, Fernandes H, Murphy Z, Graham RK, Wellington CL, Raymond LA and Hayden MR: Wild-type huntingtin protects neurons from excitotoxicity. J Neurochem. 96:1121–1129. 2006.PubMed/NCBI View Article : Google Scholar

30 

Ismailoglu I, Chen Q, Popowski M, Yang L, Gross SS and Brivanlou AH: Huntingtin protein is essential for mitochondrial metabolism, bioenergetics and structure in murine embryonic stem cells. Dev Biol. 391:230–240. 2014.PubMed/NCBI View Article : Google Scholar

31 

Guedes-Dias P, Pinho BR, Soares TR, de Proença J, Duchen MR and Oliveira JM: Mitochondrial dynamics and quality control in Huntington's disease. Neurobiol Dis. 90:51–57. 2016.PubMed/NCBI View Article : Google Scholar

32 

Brustovetsky N: Mutant huntingtin and elusive defects in oxidative metabolism and mitochondrial calcium handling. Mol Neurobiol. 53:2944–2953. 2016.PubMed/NCBI View Article : Google Scholar

33 

Grima JC, Daigle JG, Arbez N, Cunningham KC, Zhang K, Ochaba J, Geater C, Morozko E, Stocksdale J, Glatzer JC, et al: Mutant Huntingtin disrupts the nuclear pore complex. Neuron. 94:93–107.e6. 2017.PubMed/NCBI View Article : Google Scholar

34 

Zeitlin S, Liu JP, Chapman DL, Papaioannou VE and Efstratiadis A: Increased apoptosis and early embryonic lethality in mice nullizygous for the Huntington's disease gene homologue. Nat Genet. 11:155–163. 1995.PubMed/NCBI View Article : Google Scholar

35 

McKinstry SU, Karadeniz YB, Worthington AK, Hayrapetyan VY, Ozlu MI, Serafin-Molina K, Risher WC, Ustunkaya T, Dragatsis I, Zeitlin S, et al: Huntingtin is required for normal excitatory synapse development in cortical and striatal circuits. J Neurosci. 34:9455–9472. 2014.PubMed/NCBI View Article : Google Scholar

36 

Mehler MF, Petronglo JR, Arteaga-Bracho EE, Gulinello ME, Winchester ML, Pichamoorthy N, Young SK, DeJesus CD, Ishtiaq H, Gokhan S and Molero AE: Loss-of-Huntingtin in medial and lateral ganglionic lineages differentially disrupts regional interneuron and projection neuron subtypes and promotes Huntington's disease-associated behavioral, cellular, and pathological Hallmarks. J Neurosci. 39:1892–1909. 2019.PubMed/NCBI View Article : Google Scholar

37 

Jung R, Lee Y, Barker D, Correia K, Shin B, Loupe J, Collins RL, Lucente D, Ruliera J, Gillis T, et al: Mutations causing Lopes-Maciel-Rodan syndrome are huntingtin hypomorphs. Hum Mol Genet. 30:135–148. 2021.PubMed/NCBI View Article : Google Scholar

38 

Lackie RE, Maciejewski A, Ostapchenko VG, Marques-Lopes J, Choy WY, Duennwald ML, Prado VF and Prado MAM: The Hsp70/Hsp90 chaperone machinery in neurodegenerative diseases. Front Neurosci. 11(254)2017.PubMed/NCBI View Article : Google Scholar

39 

Qi L, Zhang XD, Wu JC, Lin F, Wang J, DiFiglia M and Qin ZH: The role of chaperone-mediated autophagy in huntingtin degradation. PLoS One. 7(e46834)2012.PubMed/NCBI View Article : Google Scholar

40 

Keum JW, Shin A, Gillis T, Mysore JS, Abu Elneel K, Lucente D, Hadzi T, Holmans P, Jones L, Orth M, et al: The HTT CAG-Expansion mutation determines age at death but not disease duration in huntington disease. Am J Hum Genet. 98:287–298. 2016.PubMed/NCBI View Article : Google Scholar

41 

Dulski J, Sulek A, Krygier M, Radziwonik W and Slawek J: False-negative tests in Huntington's disease: A new variant within primer hybridization site. Eur J Neurol. 28:2103–2105. 2021.PubMed/NCBI View Article : Google Scholar

42 

De Luca A, Morella A, Consoli F, Fanelli S, Thibert JR, Statt S, Latham GJ and Squitieri F: A Novel Triplet-Primed PCR assay to detect the full range of trinucleotide CAG repeats in the huntingtin gene (HTT). Int J Mol Sci. 22(1689)2021.PubMed/NCBI View Article : Google Scholar

43 

Langfelder P, Gao F, Wang N, Howland D, Kwak S, Vogt TF, Aaronson JS, Rosinski J, Coppola G, Horvath S and Yang XW: MicroRNA signatures of endogenous Huntingtin CAG repeat expansion in mice. PLoS One. 13(e0190550)2018.PubMed/NCBI View Article : Google Scholar

44 

Reed ER, Latourelle JC, Bockholt JH, Bregu J, Smock J, Paulsen JS and Myers RH: PREDICT-HD CSF Ancillary Study Investigators. MicroRNAs in CSF as prodromal biomarkers for Huntington disease in the PREDICT-HD study. Neurology. 90:e264–e272. 2018.PubMed/NCBI View Article : Google Scholar

45 

Glidden AM, Luebbe EA, Elson MJ, Goldenthal SB, Snyder CW, Zizzi CE, Dorsey ER and Heatwole CR: Patient-reported impact of symptoms in Huntington disease: PRISM-HD. Neurology. 94:e2045–e2053. 2020.PubMed/NCBI View Article : Google Scholar

46 

Paulsen JS, Miller AC, Hayes T and Shaw E: Cognitive and behavioral changes in Huntington disease before diagnosis. Handb Clin Neurol. 144:69–91. 2017.PubMed/NCBI View Article : Google Scholar

47 

Oosterloo M, de Greef BTA, Bijlsma EK, Durr A, Tabrizi SJ, Estevez-Fraga C, de Die-Smulders CEM and Roos RAC: Disease onset in Huntington's disease: When is the conversion? Mov Disord Clin Pract. 8:352–360. 2021.PubMed/NCBI View Article : Google Scholar

48 

Squadrone S, Brizio P, Abete MC and Brusco A: Trace elements profile in the blood of Huntington' disease patients. J Trace Elem Med Biol. 57:18–20. 2020.PubMed/NCBI View Article : Google Scholar

49 

Caron NS, Wright GEB and Hayden MR: Huntington Disease. 1998 Oct 23 (updated 2020 Jun 11). In: GeneReviews® (Internet). Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Mirzaa G and Amemiya A (eds). University of Washington, Seattle, WA, 1993-2021.

50 

Shoulson I and Young AB: Milestones in huntington disease. Mov Disord. 26:1127–1133. 2011.PubMed/NCBI View Article : Google Scholar

51 

Pringsheim T, Wiltshire K, Day L, Dykeman J, Steeves T and Jette N: The incidence and prevalence of Huntington's disease: A systematic review and meta-analysis. Mov Disord. 27:1083–1091. 2012.PubMed/NCBI View Article : Google Scholar

52 

Sobel SK and Cowan DB: Impact of genetic testing for Huntington disease on the family system. Am J Med Genet. 90:49–59. 2000.PubMed/NCBI View Article : Google Scholar

53 

Migliore S, Jankovic J and Squitieri F: Genetic Counseling in Huntington's disease: Potential new challenges on Horizon? Front Neurol. 10(453)2019.PubMed/NCBI View Article : Google Scholar

54 

MacLeod R, Metcalfe A and Ferrer-Duch M: A family systems approach to genetic counseling: Development of narrative inter-ventions. J Genet Couns. 30:22–29. 2021.PubMed/NCBI View Article : Google Scholar

55 

Pierron L, Hennessy J, Tezenas du Montcel S, Coarelli G, Heinzmann A, Schaerer E, Herson A, Petit E, Gargiulo M and Durr A: Informing about genetic risk in families with Huntington disease: Comparison of attitudes across two decades. Eur J Hum Genet. 29:672–679. 2021.PubMed/NCBI View Article : Google Scholar

56 

Barker RA, Fujimaki M, Rogers P and Rubinsztein DC: Huntingtin-lowering strategies for Huntington's disease. Expert Opin Investig Drugs. 29:1125–1132. 2020.PubMed/NCBI View Article : Google Scholar

57 

Aziz NA, van der Burg JMM, Tabrizi SJ and Landwehrmeyer GB: Overlap between age-at-onset and disease-progression determinants in Huntington disease. Neurology. 90:e2099–e2106. 2018.PubMed/NCBI View Article : Google Scholar

58 

Tabrizi SJ, Ghosh R and Leavitt BR: Huntingtin lowering strategies for disease modification in Huntington's disease,. Neuron. 101:801–819. 2019.PubMed/NCBI View Article : Google Scholar

59 

Lane RM, Smith A, Baumann T, Gleichmann M, Norris D, Bennett CF and Kordasiewicz H: Translating antisense technology into a treatment for Huntington's disease. Methods Mol Biol. 1780:497–523. 2018.PubMed/NCBI View Article : Google Scholar

60 

Tabrizi SJ, Leavitt BR, Landwehrmeyer GB, Wild EJ, Saft C, Barker RA, Blair NF, Craufurd D, Priller J, Rickards H, et al: Targeting Huntingtin expression in patients with Huntington's disease. N Engl J Med. 380:2307–2316. 2019.PubMed/NCBI View Article : Google Scholar

61 

Harper SQ, Staber PD, He X, Eliason SL, Martins IH, Mao Q, Yang L, Kotin RM, Paulson HL and Davidson BL: RNA interference improves motor and neuropathological abnormalities in a Huntington's disease mouse model. Proc Natl Acad Sci USA. 102:5820–5825. 2005.PubMed/NCBI View Article : Google Scholar

62 

DiFiglia M, Sena-Esteves M, Chase K, Sapp E, Pfister E, Sass M, Yoder J, Reeves P, Pandey RK, Rajeev KG, et al: Therapeutic silencing of mutant huntingtin with siRNA attenuates striatal and cortical neuropathology and behavioral deficits. Proc Natl Acad Sci USA. 104:17204–17209. 2007.PubMed/NCBI View Article : Google Scholar

63 

Keiser MS, Kordasiewicz HB and McBride JL: Gene suppression strategies for dominantly inherited neurodegenerative diseases: Lessons from Huntington's disease and spinocerebellar ataxia. Hum Mol Genet. 25(R1):R53–R64. 2016.PubMed/NCBI View Article : Google Scholar

64 

Yu D, Pendergraff H, Liu J, Kordasiewicz HB, Cleveland DW, Swayze EE, Lima WF, Crooke ST, Prakash TP and Corey DR: Single-stranded RNAs use RNAi to potently and allele-selectively inhibit mutant huntingtin expression. Cell. 150:895–908. 2012.PubMed/NCBI View Article : Google Scholar

65 

Dufour BD, Smith CA, Clark RL, Walker TR and McBride JL: Intrajugular vein delivery of AAV9-RNAi prevents neuropathological changes and weight loss in Huntington's disease mice. Mol Ther. 22:797–810. 2014.PubMed/NCBI View Article : Google Scholar

66 

Miniarikova J, Zanella I, Huseinovic A, van der Zon T, Hanemaaijer E, Martier R, Koornneef A, Southwell AL, Hayden MR, van Deventer SJ, et al: Design, Characterization, and lead selection of therapeutic miRNAs targeting huntingtin for development of gene therapy for Huntington's disease. Mol Ther Nucleic Acids. 5(e297)2016.PubMed/NCBI View Article : Google Scholar

67 

Devi S, Kumar V, Singh SK, Dubey AK and Kim JJ: Flavonoids: Potential candidates for the treatment of neurodegenerative disorders. Biomedicines. 9(99)2021.PubMed/NCBI View Article : Google Scholar

68 

Huntington Study Group. Tetrabenazine as antichorea therapy in Huntington disease: A randomized controlled trial. Neurology. 66:366–372. 2006.PubMed/NCBI View Article : Google Scholar

69 

Arora A, Kumar S, Ali J and Baboota S: Intranasal delivery of tetrabenazine nanoemulsion via olfactory region for better treatment of hyperkinetic movement associated with Huntington's disease: Pharmacokinetic and brain delivery study. Chem Phys Lipids. 230(104917)2020.PubMed/NCBI View Article : Google Scholar

70 

Catanesi M, Caioni G, Castelli V, Benedetti E, d'Angelo M and Cimini A: Benefits under the Sea: The role of marine compounds in neurodegenerative disorders. Mar Drugs. 19(24)2021.PubMed/NCBI View Article : Google Scholar

71 

Jabłońska M, Grzelakowska K, Wiśniewski B, Mazur E, Leis K and Gałązka P: Pridopidine in the treatment of Huntington's disease. Rev Neurosci. 31:441–451. 2020.PubMed/NCBI View Article : Google Scholar

72 

Squitieri F, Di Pardo A, Favellato M, Amico E, Maglione V and Frati L: Pridopidine, a dopamine stabilizer, improves motor performance and shows neuroprotective effects in Huntington disease R6/2 mouse model. J Cell Mol Med. 19:2540–2548. 2015.PubMed/NCBI View Article : Google Scholar

73 

Garcia-Miralles M, Geva M, Tan JY, Yusof NABM, Cha Y, Kusko R, Tan LJ, Xu X, Grossman I, Orbach A, et al: Early pridopidine treatment improves behavioral and transcriptional deficits in YAC128 Huntington disease mice. JCI insight. 2(e95665)2017.PubMed/NCBI View Article : Google Scholar

74 

Bartl S, Oueslati A, Southwell AL, Siddu A, Parth M, David LS, Maxan A, Salhat N, Burkert M, Mairhofer A, et al: Inhibiting cellular uptake of mutant huntingtin using a monoclonal antibody: Implications for the treatment of Huntington's disease. Neurobiol Dis. 141(104943)2020.PubMed/NCBI View Article : Google Scholar

75 

Connor B: Concise review: The use of stem cells for understanding and treating Huntington's disease. Stem Cells. 36:146–160. 2018.PubMed/NCBI View Article : Google Scholar

76 

Fritz NE, Rao AK, Kegelmeyer D, Kloos A, Busse M, Hartel L, Carrier J and Quinn L: Physical therapy and exercise interventions in Huntington's disease: A mixed methods systematic review. J Huntingtons Dis. 6:217–235. 2017.PubMed/NCBI View Article : Google Scholar

77 

Roos RA: Huntington's disease: A clinical review. Orphanet J Rare Dis. 5(40)2010.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

January-2023
Volume 18 Issue 1

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Palaiogeorgou AM, Papakonstantinou E, Golfinopoulou R, Sigala M, Mitsis T, Papageorgiou L, Diakou I, Pierouli K, Dragoumani K, Spandidos DA, Spandidos DA, et al: Recent approaches on Huntington's disease (Review). Biomed Rep 18: 5, 2023
APA
Palaiogeorgou, A.M., Papakonstantinou, E., Golfinopoulou, R., Sigala, M., Mitsis, T., Papageorgiou, L. ... Vlachakis, D. (2023). Recent approaches on Huntington's disease (Review). Biomedical Reports, 18, 5. https://doi.org/10.3892/br.2022.1587
MLA
Palaiogeorgou, A. M., Papakonstantinou, E., Golfinopoulou, R., Sigala, M., Mitsis, T., Papageorgiou, L., Diakou, I., Pierouli, K., Dragoumani, K., Spandidos, D. A., Bacopoulou, F., Chrousos, G. P., Eliopoulos, E., Vlachakis, D."Recent approaches on Huntington's disease (Review)". Biomedical Reports 18.1 (2023): 5.
Chicago
Palaiogeorgou, A. M., Papakonstantinou, E., Golfinopoulou, R., Sigala, M., Mitsis, T., Papageorgiou, L., Diakou, I., Pierouli, K., Dragoumani, K., Spandidos, D. A., Bacopoulou, F., Chrousos, G. P., Eliopoulos, E., Vlachakis, D."Recent approaches on Huntington's disease (Review)". Biomedical Reports 18, no. 1 (2023): 5. https://doi.org/10.3892/br.2022.1587