Next Article in Journal
The Association Between Indoor Air Pollutants and Brain Structure Indicators Using eTIV-Adjusted and Unadjusted Models: A Study in Seoul and Incheon
Previous Article in Journal
Exploring Links Between Lexical Representations and Cognitive Skills in School-Aged Children with High-Functioning Autism Spectrum Disorder
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

AI-Enhanced Transcriptomic Discovery of Druggable Targets and Repurposed Therapies for Huntington’s Disease

by
Rodrigo Pinheiro Araldi
1,2,
João Rafael Dias Pinto
1,2 and
Irina Kerkis
3,*
1
BioDecision Analytics Ltd., São Paulo 04021-001, Brazil
2
Structural and Functional Post-Graduation Program, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo 04021-001, Brazil
3
Genetics Laboratory, Butantan Institute, São Paulo 05503-900, Brazil
*
Author to whom correspondence should be addressed.
Brain Sci. 2025, 15(8), 865; https://doi.org/10.3390/brainsci15080865
Submission received: 7 July 2025 / Revised: 1 August 2025 / Accepted: 6 August 2025 / Published: 14 August 2025

Abstract

Huntington’s disease (HD) is an autosomal dominant neurodegenerative disorder characterized by progressive motor dysfunction, psychiatric disturbances, and cognitive decline. The pathophysiology of HD centers on a polyglutamine expansion in the huntingtin protein, which triggers widespread transcriptional dysregulation, impaired proteostasis, mitochondrial dysfunction, and excitotoxic neuronal loss—most prominently within the striatum and cortex. Despite decades of research, disease-modifying therapies remain elusive. This review synthesizes how the emerging integration of translational bioinformatics, spotlighting artificial intelligence-driven transcriptomic analyses, has identified transcriptional signatures correlating with disease progression and therapeutic response. These integrative approaches hold promise for accelerating the bench-to-bedside translation of HD therapeutics, positioning AI-powered discovery as a frontier for overcoming the complexity of neurodegeneration.

1. Huntington’s Disease: From the First Clinical Report to the Discovery of Its Etiology

Huntington’s disease (HD, OMIM 143100) is a rare and fatal monogenic autosomal-dominant neurodegenerative disorder characterized by progressive motor dysfunction, neuropsychiatric symptoms, and cognitive decline [1,2].
Although HD prevalence varies considerably across geographic regions, global estimates suggest it affects approximately 4.88 individuals per 100,000 population [1,2]. Notably the prevalence is higher in populations of European (6.37 per 100,000; 95% CI, 4.50–8.81) and North American ancestry (8.87 per 100,000; 95% CI, 4.69–16.78) [1]. The elevated prevalence is attributed to several factors, including the ability of individuals with HD gene mutation to pass it on to the next generation, improved diagnostic molecular tools, and the reclassification of previously misdiagnosed cases [3,4,5]. In contrast, much lower prevalence is reported in regions such as Africa (0.25 per 100,000) and East Asia (0.41 per 100,000), likely reflecting differences in genetic background, healthcare infrastructure, and diagnostic practices.
In Brazil, HD remains underdiagnosed, with limited nationwide epidemiological data available. The principal source of such data is the registry maintained by the Brazilian Huntington Association (ABH), founded in 1997. According to ABH estimates, approximately 14,000–20,000 individuals currently live with HD, while 73,000–100,000 are at risk of developing the disease. Interestingly, some regions in Brazil exhibit a disproportionately high prevalence of HD, a phenomenon attributed primarily to genetic clustering within isolated populations. Notably, cities such as Feira Grande (Alagoas), Senador Sá (Ceará), and Ervália (Minas Gerais) have been identified as HD hotspots, with Feira Grande reporting a prevalence of 10.4 per 10,000 inhabitants—significantly above the national average [4]. These elevated rates are likely the result of founder effects, where a small number of ancestors carrying the mutant HTT gene contributed disproportionately to the genetic makeup of subsequent generations. Limited genetic diversity and consanguinity in these communities may have amplified the transmission of the disease.
The clinical presentation of HD varies among individuals, but typically affects motor, neuropsychiatric and cognitive domains, with symptoms severity increasing over time [5,6,7].
In the early stage, patients may present subtle involuntary movements such as chorea, clumsiness, or impaired coordination (motor symptoms). Psychiatric symptoms often include depression, irritability, and anxiety, apathy, or obsessive–compulsive behaviors. Cognitive manifestation commonly involves difficulties with multitasking, slower processing speed, and impaired organizational or planning abilities [8].
As the disease progresses to the middle stage, choreic movements become more pronounced and may involve the limbs and facial muscles. Voluntary motor functions such as ambulation and speech are increasingly compromised. Psychiatric manifestation may worsen, with mood swings, impulsivity, and symptoms of mania or severe depression. Social behavior may be disrupted due to disinhibition or irritability, and cognitive deficits—particularly in memory, reasoning, and problem-solving—may become more evident [8].
In the late stage, chorea typically diminishes, while rigidity and bradykinesia (slowness of movements) predominate. Swallowing difficulties and severe motor impairment often necessitate full-time care. Profound apathy and social withdrawal are frequently observed. In some cases, psychosis and delusional thinking emerge. Dementia develops, severely impairing communication, memory, and comprehension [8].
HD was first described in 1872 by George Huntington in the seminal publication “On chorea”, where he characterized a hereditary disorder marked by involuntary movement, cognitive impairments, and psychiatric disturbances, as observed in families from Long Island, New York [9,10]. However, the genetic basis of the disease remained elusive for more than a century. In 1993, Huntington’s Disease Collaborative Research Group identified the casual mutation: an expansion of cytosine-adenine-guanine (CAG) trinucleotide (>36) repeats in the first exon of the HTT gene, located at chromosome 4p16.3. This gene encodes the huntingtin protein (HTT) [11]. Since Huntington’s initial report, over 28,000 scientific articles on HD have been published, with the vast majority in the past two decades (Figure 1), reflecting a growing global interest in understanding and treating this devastating disease.

2. How Is the CAG Expansion Involved in HD Physiopathology?

In the general population, the wild-type HTT allele (wtHTT) typically contains fewer than 36 CAG repeats, most commonly ranging from 10 to 26 [12]. About 6% of healthy people have intermediate alleles (27–35 CAG repeats), which do not cause disease but can be unstable during germline transmission and may expand to pathogenic ranges in future generations [13,14,15]. Interestingly, the frequency of IAs varies considerably across populations, from 0.45% in Thailand to 8.7% in Brazil [13].
Although IA carriers are not formally diagnosed with HD, a subset of individuals with intermediate repeat lengths have been reported to exhibit HD-compatible phenotype, including movement disorder, cognitive decline, and neuropsychiatric disturbances [13,15,16,17,18,19,20].
In contrast, individuals harboring mutated HTT allele (mHTT), characterized by more than 36 CAG repeats, are at risk of developing HD. The pathogenic threshold is stratified into reduced full penetrance categories: alleles with 36–39 CAG repeats are associated with reduced penetrance, meaning some carriers may remain asymptomatic throughout life, while alleles with ≥40 repeats confer full penetrance and are invariably associated with clinical manifestation of the disease [21], as summarized in Figure 2.
Huntingtin (HTT) is a soluble α-helical protein with a molecular weight 348 kDa, which is composed of 3144 amino acids and is encoded by the HTT gene [22]. Although it is ubiquitously expressed, HTT is found to have the highest concentration in the central nervous system (CNS), where it plays a crucial role in both CNS development and homeostatic maintenance [7,23]. This is supported by studies showing that HTT-null embryos (wtHTT-/-) exhibit developmental delay and disorganization, leading to death between embryonic days 8.5 and 10.5 [24,25,26]. In contrast, HTT-heterozygous mice (wtHTT+/-) develop motor and cognitive impairments, along with significant neuronal loss—up to 50% in the globus pallidus and the subthalamic nucleus [26,27,28], although no cell death is observed in the striatum, the primary brain area affected by HD [28]. These effects are attributed to the HTT protein’s interaction with over 200 other proteins, participating in numerous cellular and biological processes, including vesicular transport and recycling, endocytosis, endosomal trafficking, autophagy, and transcription regulation, the cell cycle, and energy metabolism—functions that are particularly important for neuronal synaptic activity [7,12,28].
Biochemically, the N-terminal region of HTT is the most extensively studied region, as it contains (polyQ) tract encoded by the CAG repeats implicated in HD [7].
The polyQ sequence beginning at residue 18 typically comprises up to 34 glutamines in unaffected individuals. It is preceded by 17 conserved residues (N17) essential for endoplasmic reticulum retentions [8,29,30,31], and it is followed by a proline-rich domain (PRD) [8], which mediates interaction with proteins that contain tryptophan in the Src homolog 3 (SH3) domains [29,32]. The remaining 97% of the protein, encoded by exon 2–67 (amino acid 69-3144), is less well characterized but includes HEAT repeats involved in scaffolding multiprotein complexes [33,34,35] and the nuclear export signal (NES), subject to post-translational modifications such as acetylation, sumoylation, and ubiquitination [36,37]. HTT also contains multiple proteolytic cleavage sites; both wild type and mHTT can be cleaved by caspases (e.g., caspase-3 at residues 469, 513 and 536, and 552; caspase-1 at 572) and the metalloproteinase (MMP)-10 at residue 402 [38], as shown in Figure 3.
The expansion of CAG trinucleotide in HTT leads to increased polyQ residues, reducing solubility of mHTT protein and promoting intracellular aggregates (inclusion bodies), particularly in GABAergic medium spiny neurons (MSNs) of the striatum that express dopamine D2 receptor (D2R), DARP32, and enkephalin [7,40]. MSNs constitute 85–95% of striatal neurons and are essential for motor control and higher-order cognitive functions, such as reward processing and decision-making [41,42].
Initially, insoluble mHTT proteins were thought to drive neurodegeneration [41,42,43,44,45]. However, evidence showing accumulation of toxic N-terminal mHTT fragments in various neuronal compartments suggested the cleavage products may be more pathogenic, while larger aggregates may serve a protective sequestration role [12,44,46,47,48,49]. The precise contribution of these aggregates remains debated. Notably, N-terminal mHTT fragments impair mitochondrial axonal transport even in the absence of inclusion bodies, indicating an early pathogenic mechanism [50].
Moreover, mHTT undergoes differential ubiquitination: K48-linked ubiquitination promotes degradation, while K63-linked ubiquitination promotes aggregation. The age-related decline of Ube3a, essential for K48-linked ubiquitination, may explain symptom onset in mid-life and IB accumulation in late-stage HD [51,52].
Since the discovery of mHTT in 1993, it has been implicated in a cascade of pathogenic events, including mitochondrial dysfunction [2,53], metal ion dyshomeostasis [54,55,56], cholesterol imbalance [57,58,59], oxidative stress [7,60,61], excitotoxicity [62,63], impaired proteostasis [64,65], and neuroinflammation [66,67]. This review summarizes the mHTT role in each of these interconnected processes.

2.1. mHTT Contributes to Mitochondrial Dysfunction and Increased Oxidative Stress

Neurons require a continuous energy supply to maintain their complex functions, with mitochondria playing a central role by generating adenosine triphosphate (ATP) through oxidative phosphorylation [68,69]. This process converts acetyl coenzyme A (Ac-CoA)—derived from carbohydrates, fatty acids, and amino acids—into ATP with high efficiency, yielding over 30 ATP per glucose, in contrast to only 2 from glycolysis [68,70,71]. Due to limited ATP storage capacity, sustained mitochondrial activity is critical for cellular homeostasis [68].
However, oxidative phosphorylation is also a primary endogenous source of reactive oxygen species (ROS). During electron transport, a fraction of electrons prematurely reacts with molecular oxygen, forming superoxide anion (O2) [72].
These reactive oxygen species can damage cellular components, contributing to oxidative stress—a key feature of mHTT-induced mitochondrial dysfunction in HD.
Superoxide is a highly reactive free radical containing an unpaired electron, enabling it to donate or accept electrons from biomolecules, such as proteins, lipids, metabolites and even nucleic acids, leading to structural damage or functional disruption. To limit this toxicity, superoxide is rapidly converted into less reactive hydrogen peroxide (H2O2) by superoxide dismutase SOD1 (cytosolic) or SOD2 (mitochondrial) through a reaction often linked to Fenton chemistry [73,74].
Although less reactive than superoxide, hydrogen peroxide can still damage biomolecules. To prevent this, glutathione peroxidase (Gpx) reduces H2O2 to water using reduced glutathione (GSH) as an electron donor [75]. The mHTT interacts with mitochondrial protein AIFM1, impairing complex I (NADH oxidase) activity and leading to OXOPHOS deficiency and increased reactive oxygen species production, as complex I is a major source of superoxide [76,77,78]. Additionally, mHTT selectively reduced complex II activity, particularly in striatal MSNs [74]. Restoration of complex II function via ectopic expression of its subunits rescue mitochondrial activity and cell viability [79,80]. Supporting this, exposure to 3-nitro propionic acid (3-NP), a selective complex II inhibitor, reproduces MSN loss and HD-like motor symptoms in rodents and humans—though chorea is typically absent in animal models [56,61,81,82,83,84].
Nuclear mHTT aggregates also sequester transcriptional factors, disrupting antioxidant gene expression [84,85], including genes regulated by peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), a master coactivator of mitochondrial biogenesis and antioxidant defense [77,86,87,88,89]. In HD, mHTT reduces PGC-1a levels directly and via transglutaminase (Tgase) activation [61,84,85], lowering the expression of antioxidant enzymes such as SOD-1, SOD-2, and Gpx-1 [77,84,86]. These alterations lead to mitochondrial dysfunction, elevated reactive oxygen species, and bioenergetic failure, contributing to neurodegeneration [87].
Although oxidative stress is consistently observed in HD [88,89], clinical trials using classical antioxidants have failed to show therapeutic benefits [90,91,92,93,94]. This may reflect uncertainly over whether oxidative stress is a cause or a downstream effect of primary HD mechanism such as transcriptional and mitochondrial dysfunction [72,95,96,97,98].

2.2. Mutant Huntingtin (HTT) Protein Leads to Dysregulation of Metal Homeostasis

Mutated HTT disrupts metal and calcium homeostasis, both contributing to mitochondrial dysfunction. It enhances mitochondrial Ca2+ uptake NMDA receptor-mediated influx, increasing mitochondrial ROS and mitochondrial DNA (mtDNA) damage [99,100]. Additionally, mHTT induces abnormal release of transition metals (Cu, Fe, Zn) from their storage sites, promoting oxidative stress [101,102,103,104].
Iron (Fe) and copper (Cu) are particularly implicated in HD, with elevated levels found in HD postmortem patient brains, R6/2 mice, and in a Drosophila model [54,99,100]. Magnetic resonance image (MRI) studies confirmed iron accumulation in the basal ganglia and cortex of HD patients, correlating with CAG repeat length and disease severity [54]. However, the regulation of metal transporters such as transferrin receptor, ferroportin, and metallothionein remains poorly characterized in HD [56,105].
Iron contributes to oxidative damage primarily via Fenton chemistry and activation of iron dependent enzymes like hypoxia inducible factor (HIF) prolyl hydroxylases [54,106]. Copper, in turn, binds directly to the N-terminal domain of HT, catalyzing cysteine oxidation and promoting toxic oligomer formation due to impaired autophagic clearance [107,108,109]. Copper can also exacerbate reactive oxygen species production through redox cycling.
Metal chelators have been explored as therapeutic agents, but most high-affinity compounds exhibit limited blood–brain barrier permeability due to high weight (>500) or hydrophilicity [110]. For example, intraventricular administration of deferoxamine (DFO) improved motor behavior in R6/2 mice [105], yet systemic DFO (MW = 657) is unlikely cross the BBB effectively, limiting clinical utility [111].

2.3. mHTT and Its Role in Excitotoxicity

Excitotoxicity, characterized by excessive glutamate-induced activation of NMDA receptors, contributes to neuronal death in HD [112,113,114]. NMDA receptors are formed by NR1, NR2 (A–D) and NR3 (A–B) subunits [62]. In vitro studies using HEK293 cells co-expressing mHTT and NMDA receptor subtypes revealed that mHTT selectively induces apoptosis in NR1/NR2B expressing cells [115], aligning with the vulnerability of neostriatal MSNs, where NR2B predominates [62,115,116].
Moreover, mHTT promotes aberrant extrasynaptic glutamatergic signaling and mislocalization of NMDARs, disrupting the CREB-PGC-1α pathway [116]. As PGC-1α regulates mitochondrial antioxidant defenses [117], its dysfunction contributes to oxidative stress and neuronal damage [112].

2.4. Mutated HTT Promotes Cholesterol Metabolism Deregulation

Despite representing only ~2% of total body mass, the CNS contains the highest cholesterol average concentration (15–20 mg/g) for other tissues, largely unesterified and concentrated in oligodendrocyte-derived myelin [113,114]. Cholesterol plays critical roles in membrane organization, receptor function, and synaptic vesicle dynamics [118,119,120,121,122,123,124,125,126]. Its depletion destabilizes lipid rafts, progressively impairing synaptic integrity and neuronal viability [125,126]. Although cholesterol turnover in the adult CNS is remarkably slow, disturbances in cholesterol homeostasis have been linked to neurodegenerative disorders, including HD [113]. In HD models and postmortem brains, the biosynthesis of cholesterol and fatty acids is consistently impaired [123,127,128] due to the reduced activity of sterol regulatory element-binding proteins—SREBP-2 for cholesterol and SREBP-1c—for fatty acid [113]. This dysregulation leads to diminished transcription of key biosynthetic enzymes, such as HMG-CoA reductase (for cholesterol synthesis) and fatty acid synthase (FASN) (for fatty acid synthesis), contributing to early pathogenic changes.
A downstream consequence is the decreased concentration of 24S-hydroxycholesterol (24-OH-Chol), a brain-derived metabolite that crosses the blood–brain barrier and may serve as a surrogate biomarker of cholesterol synthesis and disease progression [129,130]. Supporting this, HD patients exhibit a plasma cholesterol level ~40mç/dL lower than that of healthy individuals [113,131]. Although CNS-blood cholesterol correlation is limited, 24-OH-Chol accounts for ~10% of total plasma cholesterol [132].
Fatty acid metabolism is similarly affected due to shared transcriptional regulation via SREBPs [113,133]. HD patient-derived fibroblasts exhibit reduced growth under lipid-depleted conditions, reversible by linoleic and linolenic acids supplementation [113,132]. Moreover, altered membrane fluidity observed in HD cells is consistent with aberrant fatty acid elongation and desaturation [114,134].
Palmitoylation—a reversible lipid modification that facilitates membrane targeting and intracellular trafficking—is disrupted in HD. This is attributed to impaired interaction between huntingtin and its palmitoyltransferase, the huntingtin-interacting protein, particularly in striatal neurons [114,135,136]. Loss of palmitoylation comprises huntingtin’s subcellular localization and neuronal transport, contributing to dysfunction [137].
Despite increased appetite and caloric intake, HD patients frequently loss weight due to hypercatabolic state marked by elevated resting energy expenditure and altered fatty tissue and acid metabolism [40,138]. The mechanism underlying this energy imbalance remain poorly understood but are associated with disease progression.
Moreover, mHTT also suppress brain-derived neurotrophic factor (BDNF) transcription and vesicular transport [40,127,128,129,130,139,140,141,142,143], compounding metabolic deficit. BDNF plays a critical role in cholesterol biosynthesis and lipid exchange between neurons and glia [138]. Its deficiency in HD contributes to cholesterol scarcity, exacerbating neuronal vulnerability. Therapeutic strategies aimed at restoring BDNF signaling have shown promise, normalizing lipid metabolism and preserving neuronal function in preclinical HD models [40,114,130,135,136,140,144,145,146,147,148].

2.5. Systemic Effects of Mutated Huntingtin

While HD is primarily recognized as a neurodegenerative disorder, mounting evidence highlights its systemic nature, with pathological changes extending beyond the CNS [149,150,151]. The expression of both wild-type and mHTT has been confirmed in multiple peripheral tissues, including skeletal muscle, liver, heart, kidney, pancreas, testis, stomach, and immune cells (Figure 4). This finding supports the concept that HD is a multisystem disorder, with peripheral manifestation contributing to disease progression [150].
Furthermore, mHTT disrupts crosstalk between the brain and peripheral organs [149], contributing to widespread metabolic dysfunction. HD individuals frequently exhibit weight loss despite normal or increased caloric intake, driven by altered lipid and glucose metabolism and peripheral insulin resistance [152,153]. Mutant HTT also promotes oxidative stress and low-grade systemic inflammation, increasing cardiovascular risk [154].
Gastrointestinal disturbances are increasingly reported in HD, including changes in gut motility and microbiome composition [154], further supporting the role of mHTT in peripheral tissue homeostasis. In parallel, hormonal dysregulation, involving cortisol and thyroid hormone, reflects mHTT-induced disruption of endocrine signaling [153]. Moreover, mHTT impairs peripheral neuronal, leading to sensory abnormalities and autonomic dysfunctions such as altered thermoregulation and blood pressure control [154].
These systemic alterations highlight the need for therapeutic strategies that extend beyond CNS targeting. Addressing the peripheral consequences of mHTT may be essential for improving disease outcomes and quality of life in HD.

3. Therapeutic Opportunities Targeting HD Pathophysiology

HD has traditionally been viewed as a striatal disorder, primarily Gamma-aminobutyric acid (GABA)ergic neurons in the basal ganglia, particularly those expressing dopamine D1 and D2 receptors [155,156]. These neurons receive convergent glutamatergic inputs from the cortex and dopaminergic projections from substantia nigra, forming cortico-nigrostriatal circuits [157,158]. In addition to excitatory input, cortical glutamatergic neurons are also the principal source of BDNF, which is essential for the survival of MSNs.
Glutamate is the most abundant excitatory neurotransmitter in the CNS, mediating key processes such as synaptic plasticity and learning, including motor cognition [159,160,161]. Approximately 40% of all neurons are glutamatergic, and over 90% of neurons express glutamate receptors, with highest density found in the frontal cortex [162].
Synaptic plasticity—the ability of neurons to adapt their structure and function in response to activity and environmental cues—is often calcium-dependent and relies on dynamic remodeling of the dendritic actin cytoskeleton, as well as localized mRNA translation of synaptic proteins [163,164,165]. Dopamine modulates this plasticity by regulating glutamatergic transmission, dendritic integration, and the intrinsic excitability of MSNs [157,166], thereby contributing to motor control and emotional regulation.
The integrity of cortico-striatal circuits is critical for MSN survival [128], in part because cortical neurons provide BDNF, a pro-survival neurotrophin required for striatal homeostasis [167,168]. In HD, BDNF levels are markedly reduced in both patient’s brains and transgenic animal models [169,170]. This reduction is attributed to impaired transcription of the BDNF gene caused by mHTT [169], as well as disrupted axonal transport of BDNF from cortical neurons to the striatum [159].
In addition to cortical glutamatergic neurons, astrocytes also present an important source of BDNF [160,161,163]. However, mHTT disrupts the BDNF in astrocytes through multiple mechanisms, including interference with transcriptional regulation and vesicular trafficking. One key pathway involves the aggregation of mHTT, which impairs the normal function of transcription factors and intracellular signaling cascades essential for BDNF synthesis [160,161,163].
However, mHTT aggregation is rare in astrocytes from HD brains that express endogenous levels of mHTT [165], suggesting an alternative mechanism of BDNF deregulation. Notably, mHTT impairs BDNF secretion by disrupting the Rab3a-GTP/GDP cycle, which is essential for vesicle docking on the astrocyte membrane [171]. mHTT-expressing astrocytes also show a diminished ATP release and reduced docking of ATP-containing dense-core vesicles, potentially linked to mitochondrial dysfunction [172].
The reduction in BDNF is strongly implicated with HD neuropathology, and its restoration ameliorates phenotypes in HD models [173,174,175]. Nonetheless, clinical translation remains limited. Common HD models fail to recapture chorea, and BDNF’s inability to cross the blood–brain barrier [176] limits systemic delivery. Even when administrated directly into the striatum, BDNF improves neuronal function but not survival [173,177], underscoring its limited therapeutic effect as a monotherapy.
Although BDNF modulates MSNs morphology and connectivity [178,179,180], it does not induce formation of fully functional synapses alone [155]. Thus, BDNF-based therapies are unlikely to address the broad motor, cognitive. and neuropsychiatric impairments in HD. This limitation is consistent with the complex molecular landscape of the disease, which, as demonstrated in our transcriptomic analysis, involves the dysregulation of over 13,000 genes. Accordingly, interventions targeting isolated pathways or molecules are unlikely to modify the natural course of HD.
Tetrabenazine (Xenazine®), approved by the FDA in 2008 for chorea in HD, offers motor symptom relief but does not halt or prevent or delay neurodegeneration. It is a vesicular monoamine transporter 2 (VMAT2) inhibitor that depletes presynaptic monoamines, particularly dopamine in the brain, which, in turn, affects D2 receptors [181].
Despite its benefits, tetrabenazine is associated with increased risk of depression and suicidality; in a controlled trial, 19% of patients developed depression, and one suicide occurred in the treatment group, with no such event in the placebo group [182]. Common adverse effects include sedation (28%), akathisia (13%), parkinsonism (7%), depression (5.5%), anxiety (4%), fatigue (2%), and diarrhea (2%) [182].
A major barrier to advancing HD therapies remains the lack of robust molecular targets and reliable biomarkers. In the absence of such markers, most of the 256 clinical trials rely on functional scales—primarily the Unified Huntington’s Disease Rating Scale (UHDRS®)—as primary or secondary endpoints (Figure 5).
The UHDRS®, developed by the Huntington Study Group (HSG), is the most widely used clinical tool to track HD progression across multiple domains, including motor function, cognitive performance, behavioral symptoms, and functional capacity [183]. Given the centrality of motor symptoms in HD, the International Parkinson and Movement Disorder Society commissioned a clinimetric evaluation of six motor rating scales used in HD, including the Abnormal Involuntary Movement Scale, UHDRS-Total Motor Score (UHDRS-TMS), Quantified Neurological Examination, and the Marsden and Quinn Chorea Severity Scale. Among these, only UHDRS-TMS was classified as “recommended” for assessing motor severity based on standardized criteria [184]. Consequently, UHDRS-TMS is the most frequently used primary or secondary endpoint in HD clinical trials. The score (range 0–124) increases at an average rate of ~4.65 points per year (range: 2.9–6.4), although this progression varies according to individual genetic background, disease stage, and comorbid conditions [171].
Another commonly used scale is the UHDRS Total Functional Capacity (TFC), which assesses independence in daily activities, including employment, financial management, domestic chores, and care needs [174]. TFC also enables staging of disease severity: scores of 11–13 typically represent early-stage HD; 7–10 indicate mid-stage; and 0–6 correspond to advanced disease [174,175].
Despite their utility, these scales are limited by the heterogeneity and slow progression of HD symptoms, which reduce sensitivity to change over short observation periods [185]. As a result, there is an urgent need for more robust, objective, and sensitive biomarkers to reliably monitor disease progression and evaluate therapeutic responses in clinical trials.

4. Transcriptomic Studies of HD

Multiple approaches have been employed to identify biomarkers that can monitor HD progression and uncover potential therapeutic targets, particularly druggable genes [185]. Given that transcriptional dysregulation is a hallmark of HD, RNA sequencing (RNA-Seq) has emerged as a powerful tool to delineate disease-associated molecular alterations and to guide an attractive the development of biomarkers and therapies [185].
For biomarker discovery, peripheral blood is particularly attractive due to its accessibility and renewable nature [185]. In contrast, the identification of druggable targets required brain tissue. However, using striatal tissue poses challenges due to profound neuronal loss in this region—up to 90% of MSNs are lost in late-stage HD—complicating the acquisition of high-quality transcriptomic data from postmortem striatum. Given the integral role of the cortico-striatal circuity in striatal function physiology, cerebral cortex became an important brain area to be molecularly explored to identify potential drug targets for HD [186,187,188,189].
Given the integral role of the cortico-striatal circuity in maintaining striatal physiology, the cerebral cortex—particularly the Brodmann Area 9 (BA9, dorsolateral pre-frontal cortex)—has become a focal point of transcriptomic investigation [187,188,189,190,191]. Structural MRI studies have demonstrated that BA9 undergoes cortical degradation in HD, including the loss of projection neurons in layers III, V, and VI and increased glial density in deeper layers, particularly layer VI. These findings underscore BA9′s relevance as a surrogate region for molecular profiling and therapeutic target identification.
To date at least four independent transcriptomic studies have investigated brain regions implicated in HD pathology, with a focus on BA9, and the caudate nucleus (CAU) [192,193,194,195,196]. This is because approximately 90% of MSNs are lost in the late stages of the disease [42,192,193,197,198,199]. These efforts, supported by advances in bioinformatics, have enabled in-depth reanalysis and interpretation of gene expression patterns in HD. However, despite these considerable efforts, the differentially expressed genes (DEGs) identified so far have not been translated into clinically actionable biomarkers or viable therapeutic targets [189,191,199,200].
The lack of validated biomarkers and therapeutic targets remains a major bottleneck in HD drug development. In the absence of reliable tools to track disease progression or confirm the efficacy of candidate treatments, the successful translation of preclinical findings into effective therapies continues to be impeded. As a result, no approved disease-modifying therapy currently exists to halt or slow HD progression. Consequently, the burden of patients and their families continues to grow with disease advancement, alongside increasing healthcare and social costs [6].
Economic projections based on a compound annual growth rate (CAGR) of 16.5% estimate that global HD treatment will increase from USD 436 million in 2023 to USD 1728 million by 2032 [201]. This sharp rise underscores the urgent need for effective biomarkers and druggable targets to facilitate the development of therapies that can truly affect the clinical trajectory of HD [6].

5. Approved Drugs for HD Treatment

The pharmacological landscape for HD remains markedly limited. To date, three vesicular VMAT2 inhibitors were approved for the treatment of chorea in HD—tetrabenazine (TBZ, Xenazine®) and deutetrabenazine (DEU, Austedo®), approved by the U.S. Food and Drug Administration (FDA) on 15 August 2008 and on 3 April 2017, respectively; and Valbenazina (VBZ, Ingrezza®), approved on August 2023 by the U.S. FDA [194,195,196,202]. These agents act by reducing the presynaptic release of monoamines, particularly dopamine. This mechanism provides relief from chorea, one of the most characteristic motor manifestations of HD. However, these drugs do not influence the underlining neurodegenerative process and thus do not alter the natural course of the disease [194,202,203].
By depleting neuroactive monoamines postsynaptically, these treatments—particularly tetrabenazine and deutetrabenazine—are associated with a high incidence of both common and serious adverse events that may significantly compromise quality of life. Frequently reported adverse effects include sedation, somnolence, and fatigue, all of which may exacerbate the disease’s disabling features. More concerning are severe psychiatric adverse effects, such as depression, anxiety, and dyskinesia, which necessitate treatment discontinuation [181,182,196,202,203,204,205,206]. Of these, depression is particularly significant, occurring in ~19% of tetrabenazine-treated individuals in clinical trial [182], and is strongly associated with increased suicide rick [205]. Consequently, patients receiving these therapies require close psychiatric monitoring throughout the treatment course [207].
In addition to safety concerns, the pharmacokinetic of tetrabenazine presents further clinical challenges. The drug’s short half-life and rapid clearance of its active metabolites result in pronounced plasma concentration fluctuations, necessitating thrice-daily dosing to maintain symptomatic control [203]. These fluctuations are also thought to contribute to CNS-related adverse effects, such as drowsiness, insomnia, and anxiety [203].
Deutetrabenazine, a deuterated form of tetrabenazine, was developed to address these pharmacokinetic shortcomings. The incorporation of deuterium—non-toxic, stable isotope of hydrogen—slows the drug metabolism, extending its half-life and leading to more stable plasma concentration. As a result, deutetrabenazine is associated with a more consistent therapeutic effect and a potentially improved safety profile compared to tetrabenazine [202,203,204].
Although there are no head-to-head clinical trials directly compared tetrabenazine and deutetrabenazine, indirect cooperation through meta-analysis is possible due to the similarity of trial designs between the First-HD study (deutetrabenazine) and the TETRA-HD study (tetrabenazine) [205]. Both agents demonstrated modest reductions in chorea and total motor scores of the UHDRS-TMS relative to placebo (Table 1). Deutetrabenazine was associated with fewer adverse effects than tetrabenazine, though its benefits remained limited to the control of chorea, without broader impact on disease progression.
Similar outcomes were reported in the KINECKT-HD trial evaluating valbenazine, whose mechanism of action remains not completely elucidated [194]. Although the precise mechanism of action of valbenazine in HD remains incompletely understood, it is thought to act through VMAT2 inhibition. Like its predecessors, valbenazine offered only a modest benefit limited to chorea management [194].
Understanding the mechanisms of action and potential adverse effects of available therapies is crucial for optimizing patient outcomes in HD. Given the complexity of these neurodegenerative conditions, distinguishing between symptomatic treatments and those capable of modifying disease biology is essential for guiding clinical decisions. In the absence of disease-modifying therapies, HD management often extends beyond chorea control and induces the use of antipsychotics, antidepressants, and mood stabilizers to address the broader neuropsychiatric spectrum of the disease [208,209,210].
The development of novel therapeutic strategies, especially involving advanced cell-based products, offers a promising path toward altering disease progression. However, the clinical translation of such innovations has proven challenging, and effective disease-modifying treatments remain elusive [7,40,211,212,213,214,215,216,217].

6. Translational Bioinformatics and Pharma Intelligence Bring New Horizons for HD Treatment

Given the lack of disease-modifying therapies for HD and the limited efficacy of currently approved drugs, the clinical management of HD often requires the use of multiple medications. These treatments are typically guided by the published literature or clinical experience of practitioners. In this context, drug repositioning—particularly of already approved pharmaceuticals—represents a promising strategy to improve symptom management and overall outcomes in HD patients.
RNA-Seq has emerged as a useful technique for identifying potential druggable genes. These insights can guide rational drug repositioning or inform the development of novel therapeutics though computer-aided drug design (CADD) [218].
CADD is a transformative approach in pharmaceutical research that employs computational methods to streamline drug discovery and development. By integrating biological, chemical, and technological data, CADD helps predict drug behavior, evaluate interactions with biological targets, and optimize pharmacokinetic properties before a compound is synthesized and tested experimentally [218,219].
CADD includes two main strategies: (i) structure-based drug design (SBDD), which uses the 3D structure of a biological target (e.g., a protein) to design molecules capable of binding to it effectively; (ii) ligand-based drug discovery (LBDD), which relies on knowledge of existing molecules that interact with a target to predict new compounds with similar properties [218,219]. These approaches significantly accelerate drug discovery by reducing both time and cost compared with traditional experimental methods. However, the success of CADD depends critically on the prior identification of violable biological targets—namely, druggable genes—highlighting the importance of integrative bioinformatics tools like GNA-Seq in the HD research landscape.
In this regard, RNA-Seq (transcriptomic) analysis is a powerful tool that can significantly contribute to the development of new drugs, including those for HD [189,212,220,221,222]. This technique enables a comprehensive analysis of gene expression patterns, allowing the identification of differences between healthy and HD-affected cells or tissues. RNA-Seq can uncover genes and pathways involved in HD pathophysiology, such as those regulating mitochondrial function, iron metabolism, neuroinflammation, and neurodegeneration [189]. Understanding these mechanisms provides a foundation for targeting specific molecular pathways for therapeutic intervention. Additionally, RNA-Seq has the senility to detect low-abundance transcripts that may be missed by traditional methods, thereby revealing novel drug targets.
The RNA-Seq workflow begins with RNA isolation from samples, followed by either mRNA enrichment or rRNA depletion. For pharmaceutical applications, protein-coding RNAs are typically prioritized, making mRNA enrichment the preferred method. The enriched mRNAs are often then reverse-transcribed into complementary DNA (cDNA) and sequenced using high-throughput platforms, such as IlluminaTM, at a typical depth of 10 to 100 million reads per sample. The generated sequences undergo quality control assessment, including evaluation of Phred score (for base-calling accuracy), GC content (to detect contamination exogenous RNA), and fragment length distribution. After quality control, the reads are aligned to a reference genome to identify expressed genes. Gene expression levels are then quantified, normalized, and statistically compared to control samples to identify DEGs [189,223].
A key determinant of RNA-Seq data ratability is the number of biological replicates used per condition [223,224]. Empirical evidence suggests that a minimum of four replicates (samples) is required to ensure sufficient statistical power [224,225]. However, small sample sizes may limit the detection of druggable genes particularly in heterogeneous diseases such as HD. Phenotypic variability, both across individuals and disease stages, can obscure transcriptomic signals relevant to pathophysiology and therapeutic targeting.
Early transcriptomics studies in HD, based on limited postmortem material, suffered from small cohorts and incomplete clinical matching, which hampered reproducibility and consensus on therapeutic targets [187,188,191]. Notably, reanalysis by Dias-Pinto et al. [189] demonstrated that the lack of age matching between HD and control samples introduced systemic bias, resulting in false-negative and false-positive gene identification. These findings highlight the importance of careful cohort design and metadata control in transcriptomic studies of neurodegenerative disorders.
Beyond biological variation, technical aspects of differential expression analysis (DEA)—including statistical remodeling, normalization strategies, and multiple testing corrections—further influence target discovery [226,227,228]. To address these limitations and enhance the accuracy of therapeutic target identification, we developed BDASeq®, a tailored algorithm designed to reduce both type I and type II errors in RNA-Seq-based biomarker discovery [221,229].
BDASeq® is an integrative analytics-based platform designed to facilitate identification of therapeutic targets through transcriptomic profiling. Central to its architecture is the Recursive Method Combination (RMC) framework, a novel ensemble strategy that aggregates the results of eight most widely used DEA algorithms (DESeq2 [228], edgeR [230], limma-Voom [231], NOISeq [232], EBSeq [233], dearseq [234], Wilcoxon [235], and Firth’s logistic regression [236]) [190]. This ensemble approach mitigates method-specific biases and improves the robustness of gene detection across heterogeneous datasets. Additionally, BDASeq integrates artificial intelligence (AI) algorithms that combine transcriptomic signature with clinical metadata, enabling prioritization of targets for CADD.
Leveraging BDASeq®, we conducted the largest transcriptomic meta-analysis in HD to date, reanalyzing 353 samples from 12 independent BioProjects (PRJNA271929, PRJNA316625, PRJNA670925, PRJEB59710, PRJNA253002, PRJNA557205, PRJNA602538, PRJNA531456, PRJNA398545, PRJNA729761, PRJNA388174, and PRJNA755746) deposited in the NCBI Sequence Read Archive. This large-scale integrative analysis identified 394 candidate druggable genes associated with HD pathogenesis [190]. This finding, which earned recognition at the MENA Congress for Rare Disease (Abu Dhabi, UAE, 16–19 May 2024) [221,222], opened new avenues for therapeutic development—particularly through drug repositioning strategies [190].
By integrating LBDD and pharmacogenomics approaches, several of the top-ranked gene targets identified by BDASeq® have been linked to existing pharmacological agents with reported or putative activity in HD models. These include valproic acid, cyclosporine A, selenium, estradiol, acetaminophen, vorinostat, calcitriol, entinostat, vitamin E, troglitazone, progesterone, genistein, simvastatin, and Panobinostat and can offer new avenues for HD treatment (Figure 6). All these compounds have previously demonstrated potential neuroprotective, anti-inflammatory, or epigenetic effects relevant to HD, and their prioritization by BDASeq® underscores the promise of AI-driven transcriptomics in accelerating drug repurposing for neurodegenerative diseases.
The listed compounds (Table 2) were prioritized based on their mechanistic relevance to HD pathophysiology—particularly targeting mitochondrial dysfunction, oxidative stress, neuroinflammation, and transcriptional dysregulation—and their potential for drug repositioning. For each drug, the table outlines the primary mechanism of action, specific HD-relevant effects observed in preclinical or clinical studies, the level of supporting evidence (ranging from in vitro models to clinical observations), and the corresponding literature references. Notably, several agents (e.g., valproic acid, cyclosporine A, genistein, and simvastatin) have shown the ability to cross the blood–brain barrier and ameliorate HD-related phenotypes in animal models or clinical settings. Others, such as entinostat, are emerging candidates requiring further validation. This comprehensive overview underscores the utility of BDASeq® in uncovering actionable targets for HD and highlights both repurposed and novel compounds with translational potential.
Although these synthetic drugs target downstream symptoms or cellular stress responses, their clinical efficacy is often modest, inconsistent, or unproven in large-scale trials. While synthetic drugs remain essential for symptom management, their gene-specific action limits their ability to alter the natural history of HD. In contrast, cell-based therapies using human dental pulp stem-cell (hDPSC), such as NestaCell®, offer a multifaceted approach, as shown in Table 3. This is because the repertoire of biomolecules naturally produced by these cells, including BNDF, which confers neuroprotection, immunomodulation, and neuroregeneration, combined with their capability to cross the brain–blood barrier when intravenously administrated, positions NestaCell® as a promising disease-modifying agent in the evolving landscape of HD therapeutics [7,211,213,215,269,270].

7. Conclusions

The landscape of HD treatment remains a significant clinical challenge, marked by the absence of effective disease-modifying therapies despite decades of intense research. While current pharmacological approaches may alleviate certain symptoms, particularly chorea in early-to-mid-stage HD, the overarching goal continues to be the development of interventions capable of altering the disease course. In this context, our study demonstrates the power of integrative transcriptomic analysis to overcome longstanding obstacles in target discovery. By developing BDASeq®, an AI-enhanced, ensemble-based differential expression analysis platform, we reanalyzed 353 samples from 12 independent BioProjects and identified 394 putative druggable genes linked to key pathological mechanisms in HD. However, to ensure the translational relevance of these targets, it is imperative to validate them using complementary methodologies. Techniques such as quantitative PCR (qPCR) and immunoassays can confirm gene and protein expression changes in patient-derived tissues. Furthermore, functional assays in cellular and animal models, including CRISPR-based gene editing and pharmacological modulation, are essential to establish the causal role of these targets in disease pathology. This enabled a systematic drug repositioning effort, nominating several FDA-approved compounds with mechanistic relevance and prior preclinical or clinical support. Our findings highlight the urgent need for innovative methodological frameworks in therapeutic discovery and clinical trial design. As the field moves forward, combining such transcriptomic insights with refined, biomarker-driven trials may finally usher in a new era of disease-modifying therapies for HD. A concerted, multidisciplinary effort will be essential to translate these promising avenues into tangible benefits for patients.

Author Contributions

Conceptualization, methodology, investigation, writing—original draft preparation, review, and editing, R.P.A., J.R.D.P. and I.K.; visualization J.R.D.P.; funding acquisition, R.P.A. and J.R.D.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP), grant numbers 2023/06116-2, 2023/10353-0, 2024/07829-5, and 2024/17577-3.

Conflicts of Interest

R.P.A and J.R.D.P was employed by the BioDecision Analytics.

Abbreviations

The following abbreviations are used in this manuscript:
ABHBrazil Huntington Association
BDASeq®Biomarker Discovery Algorithm
CADDComputer-aided drug design
CAG Cytosine–adenine–guanine
DEADifferential expression analysis
DEGDifferentially expressed genes
HDHuntington’s disease
RMCRecursive Method Combination
TFCTotal Function Capacity
TMSTotal Motor Score
UHDRSUnified Huntington’s Disease Rate Scale

References

  1. Medina, A.; Mahjoub, Y.; Shaver, L.; Pringsheim, T. Prevalence and Incidence of Huntington’s Disease: An Updated Systematic Review and Meta-Analysis. Mov. Disord. 2022, 37, 2327–2335. [Google Scholar] [CrossRef]
  2. Jurcau, A.; Jurcau, C. Mitochondria in Huntington’s Disease: Implications in Pathogenesis and Mitochondrial-Targeted Therapeutic Strategies. Neural Regen. Res. 2023, 18, 1472–1477. [Google Scholar] [CrossRef]
  3. Kay, C.; Hayden, M.R.; Leavitt, B.R. Epidemiology of Huntington Disease. Handb. Clin. Neurol. 2017, 144, 31–46. [Google Scholar]
  4. Alencar, M.A.; Figueiredo, E.; Porciúncula, C.; Monlleó, I. I5 Huntington’s Disease and Prevalence in Brazil. J. Neurol. Neurosurg. Psychiatry 2016, 87, A60–A61. [Google Scholar] [CrossRef]
  5. Gatto, E.M.; Rojas, N.G.; Persi, G.; Etcheverry, J.L.; Cesarini, M.E.; Perandones, C. Huntington Disease: Advances in the Understanding of Its Mechanisms. Clin. Park. Relat. Disord. 2020, 3, 100056. [Google Scholar] [CrossRef]
  6. Rodríguez-Santana, I.; Mestre, T.; Squitieri, F.; Willock, R.; Arnesen, A.; Clarke, A.; D’Alessio, B.; Fisher, A.; Fuller, R.; Hamilton, J.L.; et al. Economic Burden of Huntington Disease in Europe and the USA: Results from the Huntington’s Disease Burden of Illness Study. Eur. J. Neurol. 2023, 30, 1109–1117. [Google Scholar] [CrossRef]
  7. Kerkis, I.; Araldi, R.; Wenceslau, C.; Mendes, T. Advances in Cellular and Cell-Free Therapy Medicinal Products for Huntigton’s Disease Treatment. In From Physiopatology to Treatment of Huntigton’s Disease; InTech: London, UK, 2022; pp. 1–27. [Google Scholar]
  8. Saudou, F.; Humbert, S. The Biology of Huntingtin. Neuron 2016, 89, 910–926. [Google Scholar] [CrossRef]
  9. Harper, P.S. The Epidemiology of Huntington’s Disease. Hum. Genet. 1992, 89, 365–376. [Google Scholar] [CrossRef]
  10. Bhattacharyya, K. The Story of George Huntington and His Disease. Ann. Indian Acad. Neurol. 2016, 19, 25–28. [Google Scholar] [CrossRef]
  11. MacDonald, M.E.; Ambrose, C.M.; Duyao, M.P.; Myers, R.H.; Lin, C.; Srinidhi, L.; Barnes, G.; Taylor, S.A.; James, M.; Groot, N.; et al. A Novel Gene Containing a Trinucleotide Repeat That Is Expanded and Unstable on Huntington’s Disease Chromosomes. Cell 1993, 72, 971–983. [Google Scholar] [CrossRef]
  12. Arning, L.; Nguyen, H.P. Huntington Disease Update: New Insights into the Role of Repeat Instability in Disease Pathogenesis. Med. Genet. 2022, 33, 293–300. [Google Scholar] [CrossRef]
  13. de Sabando, A.R.; Ciosi, M.; Galbete, A.; Cumming, S.A.; Álvarez, V.; Martinez-Descals, A.; Mila, M.; Trujillo-Tiebas, M.J.; López-Sendón, J.L.; Fenollar-Cortés, M.; et al. Somatic CAG Repeat Instability in Intermediate Alleles of the HTT Gene and Its Potential Association with a Clinical Phenotype. Eur. J. Hum. Genet. 2024, 32, 770–778. [Google Scholar] [CrossRef]
  14. Semaka, A.; Kay, C.; Doty, C.; Collins, J.A.; Bijlsma, E.K.; Richards, F.; Goldberg, Y.P.; Hayden, M.R. CAG Size-Specific Risk Estimates for Intermediate Allele Repeat Instability in Huntington Disease. J. Med. Genet. 2013, 50, 696–703. [Google Scholar] [CrossRef]
  15. Martí-Martínez, S.; Valor, L.M. A Glimpse of Molecular Biomarkers in Huntington’s Disease. Int. J. Mol. Sci. 2022, 23, 5411. [Google Scholar] [CrossRef]
  16. Kenney, C.; Powell, S.; Jankovic, J. Autopsy-proven Huntington’s Disease with 29 Trinucleotide Repeats. Mov. Disord. 2007, 22, 127–130. [Google Scholar] [CrossRef]
  17. Killoran, A.; Biglan, K.M.; Jankovic, J.; Eberly, S.; Kayson, E.; Oakes, D.; Young, A.B.; Shoulson, I. Characterization of the Huntington Intermediate CAG Repeat Expansion Phenotype in PHAROS. Neurology 2013, 80, 2022–2027. [Google Scholar] [CrossRef]
  18. Cubo, E.; Ramos-Arroyo, M.A.; Martinez-Horta, S.; Martínez-Descalls, A.; Calvo, S.; Gil-Polo, C.; Bachoud-Lévi, A.-C.; Bentivoglio, A.R.; Biunno, I.; Bonelli, R.M.; et al. Clinical Manifestations of Intermediate Allele Carriers in Huntington Disease. Neurology 2016, 87, 571–578. [Google Scholar] [CrossRef]
  19. Savitt, D.; Jankovic, J. Clinical Phenotype in Carriers of Intermediate Alleles in the Huntingtin Gene. J. Neurol. Sci. 2019, 402, 57–61. [Google Scholar] [CrossRef]
  20. Acuña, A.R.; Martín, E.S.S.; Fernández, C.G.; Menéndez, S.F.; Estrada, M.B.; Díaz, M.A.; González, M.M.; Martínez, V.Á. A Series of Cases with Huntington-like Phenotype and Intermediate Repeats in HTT. J. Neurol. Sci. 2021, 425, 117452. [Google Scholar] [CrossRef]
  21. Alcalá-Vida, R.; Seguin, J.; Lotz, C.; Molitor, A.M.; Irastorza-Azcarate, I.; Awada, A.; Karasu, N.; Bombardier, A.; Cosquer, B.; Skarmeta, J.L.G.; et al. Age-Related and Disease Locus-Specific Mechanisms Contribute to Early Remodelling of Chromatin Structure in Huntington’s Disease Mice. Nat. Commun. 2021, 12, 364. [Google Scholar] [CrossRef]
  22. Guo, Q.; Huang, B.; Cheng, J.; Seefelder, M.; Engler, T.; Pfeifer, G.; Oeckl, P.; Otto, M.; Moser, F.; Maurer, M.; et al. The Cryo-Electron Microscopy Structure of Huntingtin. Nature 2018, 555, 117–120. [Google Scholar] [CrossRef]
  23. Imarisio, S.; Carmichael, J.; Korolchuk, V.; Chen, C.-W.; Saiki, S.; Rose, C.; Krishna, G.; Davies, J.E.; Ttofi, E.; Underwood, B.R.; et al. Huntington’s Disease: From Pathology and Genetics to Potential Therapies. Biochem. J. 2008, 412, 191–209. [Google Scholar] [CrossRef]
  24. Duyao, M.P.; Auerbach, A.B.; Ryan, A.; Persichetti, F.; Barnes, G.T.; McNeil, S.M.; Ge, P.; Vonsattel, J.-P.; Gusella, J.F.; Joyner, A.L.; et al. Inactivation of the Mouse Huntington’s Disease Gene Homolog Hdh. Science 1995, 269, 407–410. [Google Scholar] [CrossRef]
  25. Nasir, J.; Floresco, S.B.; O’Kusky, J.R.; Diewert, V.M.; Richman, J.M.; Zeisler, J.; Borowski, A.; Marth, J.D.; Phillips, A.G.; Hayden, M.R. Targeted Disruption of the Huntington’s Disease Gene Results in Embryonic Lethality and Behavioral and Morphological Changes in Heterozygotes. Cell 1995, 81, 811–823. [Google Scholar] [CrossRef]
  26. Zeitlin, S.; Liu, J.-P.; Chapman, D.L.; Papaioannou, V.E.; Efstratiadis, A. Increased Apoptosis and Early Embryonic Lethality in Mice Nullizygous for the Huntington’s Disease Gene Homologue. Nat. Genet. 1995, 11, 155–163. [Google Scholar] [CrossRef]
  27. O’Kusky, J.R.; Nasir, J.; Cicchetti, F.; Parent, A.; Hayden, M.R. Neuronal Degeneration in the Basal Ganglia and Loss of Pallido-Subthalamic Synapses in Mice with Targeted Disruption of the Huntington’s Disease Gene. Brain Res. 1999, 818, 468–479. [Google Scholar] [CrossRef]
  28. Barron, J.C.; Hurley, E.P.; Parsons, M.P. Huntingtin and the Synapse. Front. Cell. Neurosci 2021, 15, 689332. [Google Scholar] [CrossRef]
  29. Tartari, M.; Gissi, C.; Lo Sardo, V.; Zuccato, C.; Picardi, E.; Pesole, G.; Cattaneo, E. Phylogenetic Comparison of Huntingtin Homologues Reveals the Appearance of a Primitive PolyQ in Sea Urchin. Mol. Biol. Evol. 2008, 25, 330–338. [Google Scholar] [CrossRef]
  30. Atwal, R.S.; Xia, J.; Pinchev, D.; Taylor, J.; Epand, R.M.; Truant, R. Huntingtin Has a Membrane Association Signal That Can Modulate Huntingtin Aggregation, Nuclear Entry and Toxicity. Hum. Mol. Genet. 2007, 16, 2600–2615. [Google Scholar] [CrossRef]
  31. Rockabrand, E.; Slepko, N.; Pantalone, A.; Nukala, V.N.; Kazantsev, A.; Marsh, J.L.; Sullivan, P.G.; Steffan, J.S.; Sensi, S.L.; Thompson, L.M. The First 17 Amino Acids of Huntingtin Modulate Its Sub-Cellular Localization, Aggregation and Effects on Calcium Homeostasis. Hum. Mol. Genet. 2007, 16, 61–77. [Google Scholar] [CrossRef]
  32. Harjes, P.; Wanker, E.E. The Hunt for Huntingtin Function: Interaction Partners Tell Many Different Stories. Trends Biochem. Sci. 2003, 28, 425–433. [Google Scholar] [CrossRef]
  33. Maiuri, T.; Mocle, A.J.; Hung, C.L.; Xia, J.; van Roon-Mom, W.M.C.; Truant, R. Huntingtin Is a Scaffolding Protein in the ATM Oxidative DNA Damage Response Complex. Hum. Mol. Genet. 2016, 26, 395–406. [Google Scholar] [CrossRef] [PubMed]
  34. Rui, Y.-N.; Xu, Z.; Patel, B.; Chen, Z.; Chen, D.; Tito, A.; David, G.; Sun, Y.; Stimming, E.F.; Bellen, H.J.; et al. Huntingtin Functions as a Scaffold for Selective Macroautophagy. Nat. Cell Biol. 2015, 17, 262–275. [Google Scholar] [CrossRef] [PubMed]
  35. Harding, R.J.; Deme, J.C.; Hevler, J.F.; Tamara, S.; Lemak, A.; Cantle, J.P.; Szewczyk, M.M.; Begeja, N.; Goss, S.; Zuo, X.; et al. Huntingtin Structure Is Orchestrated by HAP40 and Shows a Polyglutamine Expansion-Specific Interaction with Exon 1. Commun. Biol. 2021, 4, 1374. [Google Scholar] [CrossRef] [PubMed]
  36. Shimojo, M. Huntingtin Regulates RE1-Silencing Transcription Factor/Neuron-Restrictive Silencer Factor (REST/NRSF) Nuclear Trafficking Indirectly through a Complex with REST/NRSF-Interacting LIM Domain Protein (RILP) and Dynactin P150Glued. J. Biol. Chem. 2008, 283, 34880–34886. [Google Scholar] [CrossRef]
  37. Ravache, M.; Weber, C.; Mérienne, K.; Trottier, Y. Transcriptional Activation of REST by Sp1 in Huntington’s Disease Models. PLoS ONE 2010, 5, e14311. [Google Scholar] [CrossRef]
  38. Schulte, J.; Littleton, J.T. The Biological Function of the Huntingtin Protein and Its Relevance to Huntington’s Disease Pathology. Curr. Trends Neurol. 2011, 5, 65–78. [Google Scholar]
  39. Lorenzetti, V.; Cousijn, J. Cannabis Use Disorders and Brain Morphology. In Neuropathology of Drug Addictions and Substance Misuse; Elsevier: Amsterdam, The Netherlands, 2016; pp. 773–785. [Google Scholar]
  40. Wenceslau, C.V.; de Souza, D.M.; Mambelli-Lisboa, N.C.; Ynoue, L.H.; Araldi, R.P.; da Silva, J.M.; Pagani, E.; Haddad, M.S.; Kerkis, I. Restoration of BDNF, DARPP32, and D2R Expression Following Intravenous Infusion of Human Immature Dental Pulp Stem Cells in Huntington’s Disease 3-NP Rat Model. Cells 2022, 11, 1664. [Google Scholar] [CrossRef]
  41. Davies, S.W.; Turmaine, M.; Cozens, B.A.; DiFiglia, M.; Sharp, A.H.; Ross, C.A.; Scherzinger, E.; Wanker, E.E.; Mangiarini, L.; Bates, G.P. Formation of Neuronal Intranuclear Inclusions Underlies the Neurological Dysfunction in Mice Transgenic for the HD Mutation. Cell 1997, 90, 537–548. [Google Scholar] [CrossRef]
  42. DiFiglia, M.; Sapp, E.; Chase, K.O.; Davies, S.W.; Bates, G.P.; Vonsattel, J.P.; Aronin, N. Aggregation of Huntingtin in Neuronal Intranuclear Inclusions and Dystrophic Neurites in Brain. Science 1997, 277, 1990–1993. [Google Scholar] [CrossRef]
  43. Becher, M.W.; Kotzuk, J.A.; Sharp, A.H.; Davies, S.W.; Bates, G.P.; Price, D.L.; Ross, C.A. Intranuclear Neuronal Inclusions in Huntington’s Disease and Dentatorubral and Pallidoluysian Atrophy: Correlation between the Density of Inclusions andIT15CAG Triplet Repeat Length. Neurobiol. Dis. 1998, 4, 387–397. [Google Scholar] [CrossRef] [PubMed]
  44. Ordway, J.M.; Tallaksen-Greene, S.; Gutekunst, C.-A.; Bernstein, E.M.; Cearley, J.A.; Wiener, H.W.; Dure, L.S.; Lindsey, R.; Hersch, S.M.; Jope, R.S.; et al. Ectopically Expressed CAG Repeats Cause Intranuclear Inclusions and a Progressive Late Onset Neurological Phenotype in the Mouse. Cell 1997, 91, 753–763. [Google Scholar] [CrossRef] [PubMed]
  45. Freeman, T.B.; Cicchetti, F.; Hauser, R.A.; Deacon, T.W.; Li, X.-J.; Hersch, S.M.; Nauert, G.M.; Sanberg, P.R.; Kordower, J.H.; Saporta, S.; et al. Transplanted Fetal Striatum in Huntington’s Disease: Phenotypic Development and Lack of Pathology. Proc. Natl. Acad. Sci. USA 2000, 97, 13877–13882. [Google Scholar] [CrossRef] [PubMed]
  46. Arrasate, M.; Finkbeiner, S. Protein Aggregates in Huntington’s Disease. Exp. Neurol. 2012, 238, 1–11. [Google Scholar] [CrossRef]
  47. Ratovitski, T.; Gucek, M.; Jiang, H.; Chighladze, E.; Waldron, E.; D’Ambola, J.; Hou, Z.; Liang, Y.; Poirier, M.A.; Hirschhorn, R.R.; et al. Mutant Huntingtin N-Terminal Fragments of Specific Size Mediate Aggregation and Toxicity in Neuronal Cells. J. Biol. Chem. 2009, 284, 10855–10867. [Google Scholar] [CrossRef]
  48. Marcellin, D.; Abramowski, D.; Young, D.; Richter, J.; Weiss, A.; Marcel, A.; Maassen, J.; Kauffmann, M.; Bibel, M.; Shimshek, D.R.; et al. Fragments of HdhQ150 Mutant Huntingtin Form a Soluble Oligomer Pool That Declines with Aggregate Deposition upon Aging. PLoS ONE 2012, 7, e44457. [Google Scholar] [CrossRef]
  49. Lunkes, A.; Lindenberg, K.S.; Ben-Haïem, L.; Weber, C.; Devys, D.; Landwehrmeyer, G.B.; Mandel, J.-L.; Trottier, Y. Proteases Acting on Mutant Huntingtin Generate Cleaved Products That Differentially Build Up Cytoplasmic and Nuclear Inclusions. Mol. Cell 2002, 10, 259–269. [Google Scholar] [CrossRef]
  50. Tian, J.; Yan, Y.-P.; Zhou, R.; Lou, H.-F.; Rong, Y.; Zhang, B.-R. Soluble N-Terminal Fragment of Mutant Huntingtin Protein Impairs Mitochondrial Axonal Transport in Cultured Hippocampal Neurons. Neurosci. Bull. 2014, 30, 74–80. [Google Scholar] [CrossRef]
  51. Bhat, K.P.; Yan, S.; Wang, C.-E.; Li, S.; Li, X.-J. Differential Ubiquitination and Degradation of Huntingtin Fragments Modulated by Ubiquitin-Protein Ligase E3A. Proc. Natl. Acad. Sci. USA 2014, 111, 5706–5711. [Google Scholar] [CrossRef]
  52. Ross, C.A.; Tabrizi, S.J. Huntington’s Disease: From Molecular Pathogenesis to Clinical Treatment. Lancet Neurol. 2011, 10, 83–98. [Google Scholar] [CrossRef]
  53. Quintanilla, R.A.; Johnson, G.V.W. Role of Mitochondrial Dysfunction in the Pathogenesis of Huntington’s Disease. Brain Res. Bull. 2009, 80, 242–247. [Google Scholar] [CrossRef] [PubMed]
  54. Rosas, H.D.; Chen, Y.I.; Doros, G.; Salat, D.H.; Chen, N.; Kwong, K.K.; Bush, A.; Fox, J.; Hersch, S.M. Alterations in Brain Transition Metals in Huntington Disease. Arch. Neurol. 2012, 69, 887–893. [Google Scholar] [CrossRef] [PubMed]
  55. Pfalzer, A.C.; Yan, Y.; Kang, H.; Totten, M.; Silverman, J.; Bowman, A.B.; Erikson, K.; Claassen, D.O. Alterations in Metal Homeostasis Occur Prior to Canonical Markers in Huntington Disease. Sci. Rep. 2022, 12, 10373. [Google Scholar] [CrossRef] [PubMed]
  56. Hands, S.L.; Mason, R.; Sajjad, M.U.; Giorgini, F.; Wyttenbach, A. Metallothioneins and Copper Metabolism Are Candidate Therapeutic Targets in Huntington’s Disease. Biochem. Soc. Trans. 2010, 38, 552–558. [Google Scholar] [CrossRef]
  57. González-Guevara, E.; Cárdenas, G.; Pérez-Severiano, F.; Martínez-Lazcano, J.C. Dysregulated Brain Cholesterol Metabolism Is Linked to Neuroinflammation in Huntington’s Disease. Mov. Disord. 2020, 35, 1113–1127. [Google Scholar] [CrossRef]
  58. Kacher, R.; Mounier, C.; Caboche, J.; Betuing, S. Altered Cholesterol Homeostasis in Huntington’s Disease. Front. Aging Neurosci. 2022, 14, 797220. [Google Scholar] [CrossRef]
  59. Phillips, G.R.; Hancock, S.E.; Brown, S.H.J.; Jenner, A.M.; Kreilaus, F.; Newell, K.A.; Mitchell, T.W. Cholesteryl Ester Levels Are Elevated in the Caudate and Putamen of Huntington’s Disease Patients. Sci. Rep. 2020, 10, 20314. [Google Scholar] [CrossRef]
  60. Abramov, A.Y.; Potapova, E.V.; Dremin, V.V.; Dunaev, A.V. Interaction of Oxidative Stress and Misfolded Proteins in the Mechanism of Neurodegeneration. Life 2020, 10, 101. [Google Scholar] [CrossRef]
  61. Zheng, J.; Winderickx, J.; Franssens, V.; Liu, B. A Mitochondria-Associated Oxidative Stress Perspective on Huntington’s Disease. Front. Mol. Neurosci. 2018, 11, 329. [Google Scholar] [CrossRef]
  62. Sánchez, A.M.E.; Mejía-Toiber, J.; Massieu, L. Excitotoxic Neuronal Death and the Pathogenesis of Huntington’s Disease. Arch. Med. Res. 2008, 39, 265–276. [Google Scholar] [CrossRef]
  63. Glaser, T.; Silva, J.B.; Juvenal, G.; Maiolini, P.N.; Turrini, N.; Petiz, L.L.; Marques, L.B.; Ribeiro, D.E.; Ye, Q.; Tang, Y.; et al. Various Facets of Excitotoxicity. Explor. Neuroprot. Ther. 2022, 2, 36–64. [Google Scholar] [CrossRef]
  64. Yablonska, S.; Ganesan, V.; Ferrando, L.M.; Kim, J.; Pyzel, A.; Baranova, O.V.; Khattar, N.K.; Larkin, T.M.; Baranov, S.V.; Chen, N.; et al. Mutant Huntingtin Disrupts Mitochondrial Proteostasis by Interacting with TIM23. Proc. Natl. Acad. Sci. USA 2019, 116, 16593–16602. [Google Scholar] [CrossRef]
  65. Soares, T.R.; Reis, S.D.; Pinho, B.R.; Duchen, M.R.; Oliveira, J.M.A. Targeting the Proteostasis Network in Huntington’s Disease. Ageing Res. Rev. 2019, 49, 92–103. [Google Scholar] [CrossRef]
  66. Young, D.; Mayer, F.; Vidotto, N.; Schweizer, T.; Berth, R.; Abramowski, D.; Shimshek, D.R.; van der Putten, P.H.; Schmid, P. Mutant Huntingtin Gene-Dose Impacts on Aggregate Deposition, DARPP32 Expression and Neuroinflammation in HdhQ150 Mice. PLoS ONE 2013, 8, e75108. [Google Scholar] [CrossRef]
  67. Crotti, A.; Benner, C.; Kerman, B.E.; Gosselin, D.; Lagier-Tourenne, C.; Zuccato, C.; Cattaneo, E.; Gage, F.H.; Cleveland, D.W.; Glass, C.K. Mutant Huntingtin Promotes Autonomous Microglia Activation via Myeloid Lineage-Determining Factors. Nat. Neurosci. 2014, 17, 513–521. [Google Scholar] [CrossRef]
  68. Casanova, A.; Wevers, A.; Navarro-Ledesma, S.; Pruimboom, L. Mitochondria: It Is All about Energy. Front. Physiol. 2023, 14, 1114231. [Google Scholar] [CrossRef]
  69. Pinna, S.; Kunz, C.; Halpern, A.; Harrison, S.A.; Jordan, S.F.; Ward, J.; Werner, F.; Lane, N. A Prebiotic Basis for ATP as the Universal Energy Currency. PLoS Biol. 2022, 20, e3001437. [Google Scholar] [CrossRef]
  70. Herzig, S.; Shaw, R.J. AMPK: Guardian of Metabolism and Mitochondrial Homeostasis. Nat. Rev. Mol. Cell Biol. 2018, 19, 121–135. [Google Scholar] [CrossRef]
  71. Araldi, R.; Módolo, D.; De-Sá-Júnior, P.; Consonni, S.; Carvalho, R.; Roperto, F.; Beçak, W.; Stocco, R. Genetics and Metabolic Deregulation Following Cancer Initiation: A World to Explore. Biomed. Pharmacother. 2016, 82, 449–458. [Google Scholar] [CrossRef] [PubMed]
  72. Kumar, A.; Ratan, R.R. Oxidative Stress and Huntington’s Disease: The Good, The Bad, and The Ugly. J. Huntingt. Dis. 2016, 5, 217–237. [Google Scholar] [CrossRef] [PubMed]
  73. Ratovitski, T.; Chighladze, E.; Arbez, N.; Boronina, T.; Herbrich, S.; Cole, R.N.; Ross, C.A. Huntingtin Protein Interactions Altered by Polyglutamine Expansion as Determined by Quantitative Proteomic Analysis. Cell Cycle 2012, 11, 2006–2021. [Google Scholar] [CrossRef]
  74. Damiano, M.; Diguet, E.; Malgorn, C.; D’Aurelio, M.; Galvan, L.; Petit, F.; Benhaim, L.; Guillermier, M.; Houitte, D.; Dufour, N.; et al. A Role of Mitochondrial Complex II Defects in Genetic Models of Huntington’s Disease Expressing N-Terminal Fragments of Mutant Huntingtin. Hum. Mol. Genet. 2013, 22, 3869–3882. [Google Scholar] [CrossRef] [PubMed]
  75. De Marco, F. Oxidative Stress and HPV Carcinogenesis. Viruses 2013, 5, 708–731. [Google Scholar] [CrossRef] [PubMed]
  76. Damiano, M.; Galvan, L.; Déglon, N.; Brouillet, E. Mitochondria in Huntington’s Disease. Biochim. Biophys. Acta (BBA)—Mol. Basis Dis. 2010, 1802, 52–61. [Google Scholar] [CrossRef] [PubMed]
  77. Bossy-Wetzel, E.; Petrilli, A.; Knott, A.B. Mutant Huntingtin and Mitochondrial Dysfunction. Trends Neurosci. 2008, 31, 609–616. [Google Scholar] [CrossRef]
  78. Oliveira, J.M.A.; Jekabsons, M.B.; Chen, S.; Lin, A.; Rego, A.C.; Gonçalves, J.; Ellerby, L.M.; Nicholls, D.G. Mitochondrial Dysfunction in Huntington’s Disease: The Bioenergetics of Isolated and in Situ Mitochondria from Transgenic Mice. J. Neurochem. 2007, 101, 241–249. [Google Scholar] [CrossRef]
  79. Carmo, C.; Naia, L.; Lopes, C.; Rego, A.C. Mitochondrial Dysfunction in Huntington’s Disease. Adv. Exp. Med. Biol. 2018, 1049, 59–83. [Google Scholar]
  80. Paul, B.D.; Sbodio, J.I.; Xu, R.; Vandiver, M.S.; Cha, J.Y.; Snowman, A.M.; Snyder, S.H. Cystathionine γ-Lyase Deficiency Mediates Neurodegeneration in Huntington’s Disease. Nature 2014, 509, 96–100. [Google Scholar] [CrossRef]
  81. Paul, B.D.; Snyder, S.H. Neurodegeneration in Huntington’s Disease Involves Loss of Cystathionine γ-Lyase. Cell Cycle 2014, 13, 2491–2493. [Google Scholar] [CrossRef]
  82. Sugars, K.L.; Rubinsztein, D.C. Transcriptional Abnormalities in Huntington Disease. Trends Genet. 2003, 19, 233–238. [Google Scholar] [CrossRef]
  83. Cha, J.-H.J. Transcriptional Signatures in Huntington’s Disease. Prog. Neurobiol. 2007, 83, 228–248. [Google Scholar] [CrossRef]
  84. Chaturvedi, R.K.; Hennessey, T.; Johri, A.; Tiwari, S.K.; Mishra, D.; Agarwal, S.; Kim, Y.S.; Beal, M.F. Transducer of Regulated CREB-Binding Proteins (TORCs) Transcription and Function Is Impaired in Huntington’s Disease. Hum. Mol. Genet. 2012, 21, 3474–3488. [Google Scholar] [CrossRef] [PubMed]
  85. McConoughey, S.J.; Basso, M.; Niatsetskaya, Z.V.; Sleiman, S.F.; Smirnova, N.A.; Langley, B.C.; Mahishi, L.; Cooper, A.J.L.; Antonyak, M.A.; Cerione, R.A.; et al. Inhibition of Transglutaminase 2 Mitigates Transcriptional Dysregulation in Models of Huntington Disease. EMBO Mol. Med. 2010, 2, 349–370. [Google Scholar] [CrossRef] [PubMed]
  86. Jeong, H.; Cohen, D.E.; Cui, L.; Supinski, A.; Savas, J.N.; Mazzulli, J.R.; Yates, J.R.; Bordone, L.; Guarente, L.; Krainc, D. Sirt1 Mediates Neuroprotection from Mutant Huntingtin by Activation of the TORC1 and CREB Transcriptional Pathway. Nat. Med. 2012, 18, 159–165. [Google Scholar] [CrossRef] [PubMed]
  87. Cui, L.; Jeong, H.; Borovecki, F.; Parkhurst, C.N.; Tanese, N.; Krainc, D. Transcriptional Repression of PGC-1α by Mutant Huntingtin Leads to Mitochondrial Dysfunction and Neurodegeneration. Cell 2006, 127, 59–69. [Google Scholar] [CrossRef]
  88. Tobore, T.O. Towards a Comprehensive Understanding of the Contributions of Mitochondrial Dysfunction and Oxidative Stress in the Pathogenesis and Pathophysiology of Huntington’s Disease. J. Neurosci. Res. 2019, 97, 1455–1468. [Google Scholar] [CrossRef]
  89. Perluigi, M.; Poon, H.F.; Maragos, W.; Pierce, W.M.; Klein, J.B.; Calabrese, V.; Cini, C.; De Marco, C.; Butterfield, D.A. Proteomic Analysis of Protein Expression and Oxidative Modification in R6/2 Transgenic Mice. Mol. Cell. Proteom. 2005, 4, 1849–1861. [Google Scholar] [CrossRef]
  90. Feigin, A.; Kieburtz, K.; Como, P.; Hickey, C.; Abwender, D.; Zimmerman, C.; Steinberg, K.; Shoulson, I. Assessment of Coenzyme Q10 Tolerability in Huntington’s Disease. Mov. Disord. 1996, 11, 321–323. [Google Scholar] [CrossRef]
  91. The Huntington Study Group. A Randomized, Placebo-Controlled Trial of Coenzyme Q10 and Remacemide in Huntington’s Disease. Neurology 2001, 57, 397–404. [Google Scholar] [CrossRef]
  92. Hersch, S.M.; Gevorkian, S.; Marder, K.; Moskowitz, C.; Feigin, A.; Cox, M.; Como, P.; Zimmerman, C.; Lin, M.; Zhang, L.; et al. Creatine in Huntington Disease Is Safe, Tolerable, Bioavailable in Brain and Reduces Serum 8OH2′dG. Neurology 2006, 66, 250–252. [Google Scholar] [CrossRef]
  93. Huntington Study Group TREND-HD Investigators. Randomized Controlled Trial of Ethyl-Eicosapentaenoic Acid in Huntington Disease. Arch. Neurol. 2008, 65, 1582–1589. [Google Scholar] [CrossRef] [PubMed]
  94. Ranen, N.G.; Peyser, C.E.; Coyle, J.T.; Bylsma, F.W.; Sherr, M.; Day, L.; Folstein, M.F.; Brandt, J.; Ross, C.A.; Folstein, S.E. A Controlled Trial of Idebenone in Huntington’s Disease. Mov. Disord. 1996, 11, 549–554. [Google Scholar] [CrossRef] [PubMed]
  95. Luthi-Carter, R. Decreased Expression of Striatal Signaling Genes in a Mouse Model of Huntington’s Disease. Hum. Mol. Genet. 2000, 9, 1259–1271. [Google Scholar] [CrossRef]
  96. Achour, M.; Le Gras, S.; Keime, C.; Parmentier, F.; Lejeune, F.-X.; Boutillier, A.-L.; Neri, 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, 24, 3481–3496. [Google Scholar] [CrossRef]
  97. Iannicola, C.; Moreno, S.; Oliverio, S.; Nardacci, R.; Ciofi-Luzzatto, A.; Piacentini, M. Early Alterations in Gene Expression and Cell Morphology in a Mouse Model of Huntington’s Disease. J. Neurochem. 2000, 75, 830–839. [Google Scholar] [CrossRef]
  98. Kumar, A.; Vaish, M.; Ratan, R.R. Transcriptional Dysregulation in Huntington’s Disease: A Failure of Adaptive Transcriptional Homeostasis. Drug Discov. Today 2014, 19, 956–962. [Google Scholar] [CrossRef]
  99. Bartzokis, G.; Lu, P.H.; Geschwind, D.H.; Tingus, K.; Huang, D.; Mendez, M.F.; Edwards, N.; Mintz, J. Apolipoprotein E Affects Both Myelin Breakdown and Cognition: Implications for Age-Related Trajectories of Decline into Dementia. Biol. Psychiatry 2007, 62, 1380–1387. [Google Scholar] [CrossRef]
  100. Fox, J.H.; Kama, J.A.; Lieberman, G.; Chopra, R.; Dorsey, K.; Chopra, V.; Volitakis, I.; Cherny, R.A.; Bush, A.I.; Hersch, S. Mechanisms of Copper Ion Mediated Huntington’s Disease Progression. PLoS ONE 2007, 2, e334. [Google Scholar] [CrossRef]
  101. Brazier, M.; Wedd, A.; Collins, S. Antioxidant and Metal Chelation-Based Therapies in the Treatment of Prion Disease. Antioxidants 2014, 3, 288–308. [Google Scholar] [CrossRef]
  102. Dexter, D.T.; Jenner, P.; Schapira, A.H.V.; Marsden, C.D. Alterations in Levels of Iron, Ferritin, and Other Trace Metals in Neurodegenerative Diseases Affecting the Basal Ganglia. Ann. Neurol. 1992, 32, S94–S100. [Google Scholar] [CrossRef]
  103. Bartzokis, G.; Lu, P.H.; Tishler, T.A.; Fong, S.M.; Oluwadara, B.; Finn, J.P.; Huang, D.; Bordelon, Y.; Mintz, J.; Perlman, S. Myelin Breakdown and Iron Changes in Huntington’s Disease: Pathogenesis and Treatment Implications. Neurochem. Res. 2007, 32, 1655–1664. [Google Scholar] [CrossRef] [PubMed]
  104. Muller, M.; Leavitt, B.R. Iron Dysregulation in Huntington’s Disease. J. Neurochem. 2014, 130, 328–350. [Google Scholar] [CrossRef]
  105. Chen, J.; Marks, E.; Lai, B.; Zhang, Z.; Duce, J.A.; Lam, L.Q.; Volitakis, I.; Bush, A.I.; Hersch, S.; Fox, J.H. Iron Accumulates in Huntington’s Disease Neurons: Protection by Deferoxamine. PLoS ONE 2013, 8, e77023. [Google Scholar] [CrossRef]
  106. García-Bueno, B.; Caso, J.R. Cannabis, Cannabinoid Receptors, and Stress-Induced Excitotoxicity. In Neuropathology of Drug Addictions and Substance Misuse; Elsevier: Amsterdam, The Netherlands, 2016; pp. 731–737. [Google Scholar]
  107. Fox, J.H.; Connor, T.; Stiles, M.; Kama, J.; Lu, Z.; Dorsey, K.; Liebermann, G.; Sapp, E.; Cherny, R.A.; Banks, M.; et al. Cysteine Oxidation within N-Terminal Mutant Huntingtin Promotes Oligomerization and Delays Clearance of Soluble Protein. J. Biol. Chem. 2011, 286, 18320–18330. [Google Scholar] [CrossRef]
  108. Xiao, G.; Fan, Q.; Wang, X.; Zhou, B. Huntington Disease Arises from a Combinatory Toxicity of Polyglutamine and Copper Binding. Proc. Natl. Acad. Sci. USA 2013, 110, 14995–15000. [Google Scholar] [CrossRef]
  109. Mitomi, Y.; Nomura, T.; Kurosawa, M.; Nukina, N.; Furukawa, Y. Post-Aggregation Oxidation of Mutant Huntingtin Controls the Interactions between Aggregates. J. Biol. Chem. 2012, 287, 34764–34775. [Google Scholar] [CrossRef] [PubMed]
  110. Barnham, K.J.; Bush, A.I. Biological Metals and Metal-Targeting Compounds in Major Neurodegenerative Diseases. Chem. Soc. Rev. 2014, 43, 6727–6749. [Google Scholar] [CrossRef]
  111. Karuppagounder, S.S.; Alim, I.; Khim, S.J.; Bourassa, M.W.; Sleiman, S.F.; John, R.; Thinnes, C.C.; Yeh, T.-L.; Demetriades, M.; Neitemeier, S.; et al. Therapeutic Targeting of Oxygen-Sensing Prolyl Hydroxylases Abrogates ATF4-Dependent Neuronal Death and Improves Outcomes after Brain Hemorrhage in Several Rodent Models. Sci. Transl. Med. 2016, 8, 328ra29. [Google Scholar] [CrossRef] [PubMed]
  112. Raymond, L.A. Excitotoxicity in Huntington Disease. Clin. Neurosci. Res. 2003, 3, 121–128. [Google Scholar] [CrossRef]
  113. Block, R.C.; Dorsey, E.R.; Beck, C.A.; Brenna, J.T.; Shoulson, I. Altered Cholesterol and Fatty Acid Metabolism in Huntington Disease. J. Clin. Lipidol. 2010, 4, 17–23. [Google Scholar] [CrossRef]
  114. Valenza, M.; Cattaneo, E. Cholesterol Dysfunction in Neurodegenerative Diseases: Is Huntington’s Disease in the List? Prog. Neurobiol. 2006, 80, 165–176. [Google Scholar] [CrossRef] [PubMed]
  115. Zeron, M.M.; Chen, N.; Moshaver, A.; Ting-Chun Lee, A.; Wellington, C.L.; Hayden, M.R.; Raymond, L.A. Mutant Huntingtin Enhances Excitotoxic Cell Death. Mol. Cell. Neurosci. 2001, 17, 41–53. [Google Scholar] [CrossRef] [PubMed]
  116. Okamoto, S.; Pouladi, M.A.; Talantova, M.; Yao, D.; Xia, P.; Ehrnhoefer, D.E.; Zaidi, R.; Clemente, A.; Kaul, M.; Graham, R.K.; et al. Balance between Synaptic versus Extrasynaptic NMDA Receptor Activity Influences Inclusions and Neurotoxicity of Mutant Huntingtin. Nat. Med. 2009, 15, 1407–1413. [Google Scholar] [CrossRef]
  117. Valle, I.; Alvarez-Barrientos, A.; Arza, E.; Lamas, S.; Monsalve, M. PGC-1α Regulates the Mitochondrial Antioxidant Defense System in Vascular Endothelial Cells. Cardiovasc. Res. 2005, 66, 562–573. [Google Scholar] [CrossRef] [PubMed]
  118. Sankaram, M.B.; Thompson, T.E. Modulation of Phospholipid Acyl Chain Order by Cholesterol. A Solid-State Deuterium Nuclear Magnetic Resonance Study. Biochemistry 1990, 29, 10676–10684. [Google Scholar] [CrossRef]
  119. Rubenstein, J.L.; Smith, B.A.; McConnell, H.M. Lateral Diffusion in Binary Mixtures of Cholesterol and Phosphatidylcholines. Proc. Natl. Acad. Sci. USA 1979, 76, 15–18. [Google Scholar] [CrossRef]
  120. Gimpl, G.; Burger, K.; Fahrenholz, F. Cholesterol as Modulator of Receptor Function. Biochemistry 1997, 36, 10959–10974. [Google Scholar] [CrossRef]
  121. Takamori, S.; Holt, M.; Stenius, K.; Lemke, E.A.; Grønborg, M.; Riedel, D.; Urlaub, H.; Schenck, S.; Brügger, B.; Ringler, P.; et al. Molecular Anatomy of a Trafficking Organelle. Cell 2006, 127, 831–846. [Google Scholar] [CrossRef]
  122. van der Kant, R.; Fish, A.; Janssen, L.; Janssen, H.; Krom, S.; Ho, N.; Brummelkamp, T.; Carette, J.; Rocha, N.; Neefjes, J. Late Endosomal Transport and Tethering Are Coupled Processes Controlled by RILP and the Cholesterol Sensor ORP1L. J. Cell Sci. 2013, 126, 3462–3474. [Google Scholar] [CrossRef]
  123. Koseoglu, S.; Love, S.A.; Haynes, C.L. Cholesterol Effects on Vesicle Pools in Chromaffin Cells Revealed by Carbon-Fiber Microelectrode Amperometry. Anal. Bioanal. Chem. 2011, 400, 2963–2971. [Google Scholar] [CrossRef]
  124. Korinek, M.; Gonzalez-Gonzalez, I.M.; Smejkalova, T.; Hajdukovic, D.; Skrenkova, K.; Krusek, J.; Horak, M.; Vyklicky, L. Cholesterol Modulates Presynaptic and Postsynaptic Properties of Excitatory Synaptic Transmission. Sci. Rep. 2020, 10, 12651. [Google Scholar] [CrossRef] [PubMed]
  125. Hering, H.; Lin, C.-C.; Sheng, M. Lipid Rafts in the Maintenance of Synapses, Dendritic Spines, and Surface AMPA Receptor Stability. J. Neurosci. 2003, 23, 3262–3271. [Google Scholar] [CrossRef] [PubMed]
  126. Dejanovic, B.; Sheng, M.; Hanson, J.E. Targeting Synapse Function and Loss for Treatment of Neurodegenerative Diseases. Nat. Rev. Drug Discov. 2024, 23, 23–42. [Google Scholar] [CrossRef] [PubMed]
  127. Plinta, K.; Plewka, A.; Pawlicki, K.; Zmarzły, N.; Wójcik-Pędziwiatr, M.; Rudziński, M.; Krzak-Kubica, A.; Doręgowska-Stachera, M.; Rudzińska-Bar, M. The Utility of BDNF Detection in Assessing Severity of Huntington’s Disease. J. Clin. Med. 2021, 10, 5181. [Google Scholar] [CrossRef]
  128. Zuccato, C.; Ciammola, A.; Rigamonti, D.; Leavitt, B.R.; Goffredo, D.; Conti, L.; MacDonald, M.E.; Friedlander, R.M.; Silani, V.; Hayden, M.R.; et al. Loss of Huntingtin-Mediated BDNF Gene Transcription in Huntington’s Disease. Science 2001, 293, 493–498. [Google Scholar] [CrossRef]
  129. Giralt, A.; Carretón, O.; Lao-Peregrin, C.; Martín, E.D.; Alberch, J. Conditional BDNF Release under Pathological Conditions Improves Huntington’s Disease Pathology by Delaying Neuronal Dysfunction. Mol. Neurodegener. 2011, 6, 71. [Google Scholar] [CrossRef]
  130. Xie, Y.; Hayden, M.R.; Xu, B. BDNF Overexpression in the Forebrain Rescues Huntington’s Disease Phenotypes in YAC128 Mice. J. Neurosci. 2010, 30, 14708–14718. [Google Scholar] [CrossRef]
  131. Markianos, M.; Panas, M.; Kalfakis, N.; Vassilopoulos, D. Low Plasma Total Cholesterol in Patients with Huntington’s Disease and First-Degree Relatives. Mol. Genet. Metab. 2008, 93, 341–346. [Google Scholar] [CrossRef]
  132. Lütjohann, D.; Breuer, O.; Ahlborg, G.; Nennesmo, I.; Sidén, A.; Diczfalusy, U.; Björkhem, I. Cholesterol Homeostasis in Human Brain: Evidence for an Age-Dependent Flux of 24S-Hydroxycholesterol from the Brain into the Circulation. Proc. Natl. Acad. Sci. USA 1996, 93, 9799–9804. [Google Scholar] [CrossRef]
  133. Valenza, M.; Rigamonti, D.; Goffredo, D.; Zuccato, C.; Fenu, S.; Jamot, L.; Strand, A.; Tarditi, A.; Woodman, B.; Racchi, M.; et al. Dysfunction of the Cholesterol Biosynthetic Pathway in Huntington’s Disease. J. Neurosci. 2005, 25, 9932–9939. [Google Scholar] [CrossRef]
  134. Torrejon, C.; Jung, U.J.; Deckelbaum, R.J. N-3 Fatty Acids and Cardiovascular Disease: Actions and Molecular Mechanisms. Prostagland. Leukot. Essent. Fat. Acids 2007, 77, 319–326. [Google Scholar] [CrossRef]
  135. Yanai, A.; Huang, K.; Kang, R.; Singaraja, R.R.; Arstikaitis, P.; Gan, L.; Orban, P.C.; Mullard, A.; Cowan, C.M.; Raymond, L.A.; et al. Palmitoylation of Huntingtin by HIP14is Essential for Its Trafficking and Function. Nat. Neurosci. 2006, 9, 824–831. [Google Scholar] [CrossRef]
  136. Singaraja, R.R. HIP14, a Novel Ankyrin Domain-Containing Protein, Links Huntingtin to Intracellular Trafficking and Endocytosis. Hum. Mol. Genet. 2002, 11, 2815–2828. [Google Scholar] [CrossRef] [PubMed]
  137. Katsuno, M.; Adachi, H.; Sobue, G. Getting a Handle on Huntington’s Disease: The Case for Cholesterol. Nat. Med. 2009, 15, 253–254. [Google Scholar] [CrossRef] [PubMed]
  138. Suzuki, S.; Kiyosue, K.; Hazama, S.; Ogura, A.; Kashihara, M.; Hara, T.; Koshimizu, H.; Kojima, M. Brain-Derived Neurotrophic Factor Regulates Cholesterol Metabolism for Synapse Development. J. Neurosci. 2007, 27, 6417–6427. [Google Scholar] [CrossRef] [PubMed]
  139. Lenoir, S.; Lahaye, R.A.; Vitet, H.; Scaramuzzino, C.; Virlogeux, A.; Capellano, L.; Genoux, A.; Gershoni-Emek, N.; Geva, M.; Hayden, M.R.; et al. Pridopidine Rescues BDNF/TrkB Trafficking Dynamics and Synapse Homeostasis in a Huntington Disease Brain-on-a-Chip Model. Neurobiol. Dis. 2022, 173, 105857. [Google Scholar] [CrossRef]
  140. Kim, H.S.; Jeon, I.; Noh, J.-E.; Lee, H.; Hong, K.S.; Lee, N.; Pei, Z.; Song, J. Intracerebral Transplantation of BDNF-Overexpressing Human Neural Stem Cells (HB1.F3.BDNF) Promotes Migration, Differentiation and Functional Recovery in a Rodent Model of Huntington’s Disease. Exp. Neurobiol. 2020, 29, 130–137. [Google Scholar] [CrossRef]
  141. Zuccato, C.; Marullo, M.; Conforti, P.; MacDonald, M.E.; Tartari, M.; Cattaneo, E. Systematic Assessment of BDNF and Its Receptor Levels in Human Cortices Affected by Huntington’s Disease. Brain Pathol. 2007, 18, 225–238. [Google Scholar] [CrossRef]
  142. Yu, C.; Li, C.H.; Chen, S.; Yoo, H.; Qin, X.; Park, H. Decreased BDNF Release in Cortical Neurons of a Knock-in Mouse Model of Huntington’s Disease. Sci. Rep. 2018, 8, 16976. [Google Scholar] [CrossRef]
  143. Maloney, M.T.; Wang, W.; Bhowmick, S.; Millan, I.; Kapur, M.; Herrera, N.; Frost, E.; Zhang, E.Y.; Song, S.; Wang, M.; et al. Failure to Thrive: Impaired BDNF Transport along the Cortical–Striatal Axis in Mouse Q140 Neurons of Huntington’s Disease. Biology 2023, 12, 157. [Google Scholar] [CrossRef]
  144. Lejkowska, R.; Kawa, M.; Pius-Sadowska, E.; Rogińska, D.; Łuczkowska, K.; Machaliński, B.; Machalińska, A. Preclinical Evaluation of Long-Term Neuroprotective Effects of BDNF-Engineered Mesenchymal Stromal Cells as Intravitreal Therapy for Chronic Retinal Degeneration in Rd6 Mutant Mice. Int. J. Mol. Sci. 2019, 20, 777. [Google Scholar] [CrossRef] [PubMed]
  145. Scheper, V.; Schwieger, J.; Hamm, A.; Lenarz, T.; Hoffmann, A. BDNF-overexpressing Human Mesenchymal Stem Cells Mediate Increased Neuronal Protection in Vitro. J. Neurosci. Res. 2019, 97, 1414–1429. [Google Scholar] [CrossRef] [PubMed]
  146. Kowiański, P.; Lietzau, G.; Czuba, E.; Waśkow, M.; Steliga, A.; Moryś, J. BDNF: A Key Factor with Multipotent Impact on Brain Signaling and Synaptic Plasticity. Cell. Mol. Neurobiol. 2018, 38, 579–593. [Google Scholar] [CrossRef] [PubMed]
  147. Wu, S.-Y.; Pan, B.-S.; Tsai, S.-F.; Chiang, Y.-T.; Huang, B.-M.; Mo, F.-E.; Kuo, Y.-M. BDNF Reverses Aging-Related Microglial Activation. J. Neuroinflamm. 2020, 17, 210. [Google Scholar] [CrossRef]
  148. Deng, P.; Anderson, J.D.; Yu, A.S.; Annett, G.; Fink, K.D.; Nolta, J.A. Engineered BDNF Producing Cells as a Potential Treatment for Neurologic Disease. Expert Opin. Biol. Ther. 2016, 16, 1025–1033. [Google Scholar] [CrossRef]
  149. Valadão, P.A.C.; Santos, K.B.S.; Vieira, T.H.F.E.; Cordeiro, T.M.E.; Teixeira, A.L.; Guatimosim, C.; de Miranda, A.S. Inflammation in Huntington’s Disease: A Few New Twists on an Old Tale. J. Neuroimmunol. 2020, 348, 577380. [Google Scholar] [CrossRef]
  150. van der Burg, J.M.; Björkqvist, M.; Brundin, P. Beyond the Brain: Widespread Pathology in Huntington’s Disease. Lancet Neurol. 2009, 8, 765–774. [Google Scholar] [CrossRef]
  151. Zuccato, C.; Valenza, M.; Cattaneo, E. Molecular Mechanisms and Potential Therapeutical Targets in Huntington’s Disease. Physiol. Rev. 2010, 90, 905–981. [Google Scholar] [CrossRef]
  152. Gómez-Jaramillo, L.; Cano-Cano, F.; del Carmen González-Montelongo, M.; Campos-Caro, A.; Aguilar-Diosdado, M.; Arroba, A.I. A New Perspective on Huntington’s Disease: How a Neurological Disorder Influences the Peripheral Tissues. Int. J. Mol. Sci. 2022, 23, 6089. [Google Scholar] [CrossRef]
  153. Aziz, N.A.; Pijl, H.; Frolich, M.; Snel, M.; Streefland, T.C.M.; Roelfsema, F.; Roos, R.A.C. Systemic Energy Homeostasis in Huntington’s Disease Patients. J. Neurol. Neurosurg. Psychiatry 2010, 81, 1233–1237. [Google Scholar] [CrossRef]
  154. Burtscher, J.; Strasser, B.; Pepe, G.; Burtscher, M.; Kopp, M.; Di Pardo, A.; Maglione, V.; Khamoui, A.V. Brain–Periphery Interactions in Huntington’s Disease: Mediators and Lifestyle Interventions. Int. J. Mol. Sci. 2024, 25, 4696. [Google Scholar] [CrossRef]
  155. Paraskevopoulou, F.; Herman, M.A.; Rosenmund, C. Glutamatergic Innervation onto Striatal Neurons Potentiates GABAergic Synaptic Output. J. Neurosci. 2019, 39, 4448–4460. [Google Scholar] [CrossRef]
  156. Tunstall, M.J.; Oorschot, D.E.; Kean, A.; Wickens, J.R. Inhibitory Interactions Between Spiny Projection Neurons in the Rat Striatum. J. Neurophysiol. 2002, 88, 1263–1269. [Google Scholar] [CrossRef]
  157. Lassus, B.; Naudé, J.; Faure, P.; Guedin, D.; Von Boxberg, Y.; la Cour, C.M.; Millan, M.J.; Peyrin, J.-M. Glutamatergic and Dopaminergic Modulation of Cortico-Striatal Circuits Probed by Dynamic Calcium Imaging of Networks Reconstructed in Microfluidic Chips. Sci. Rep. 2018, 8, 17461. [Google Scholar] [CrossRef]
  158. Kubota, Y.; Inagaki, S.; Shimada, S.; Kito, S.; Eckenstein, F.; Tohyama, M. Neostriatal Cholinergic Neurons Receive Direct Synaptic Inputs from Dopaminergic Axons. Brain Res. 1987, 413, 179–184. [Google Scholar] [CrossRef] [PubMed]
  159. Gauthier, L.R.; Charrin, B.C.; Borrell-Pagès, M.; Dompierre, J.P.; Rangone, H.; Cordelières, F.P.; De Mey, J.; MacDonald, M.E.; Leßmann, V.; Humbert, S.; et al. Huntingtin Controls Neurotrophic Support and Survival of Neurons by Enhancing BDNF Vesicular Transport along Microtubules. Cell 2004, 118, 127–138. [Google Scholar] [CrossRef] [PubMed]
  160. Miyamoto, N.; Maki, T.; Shindo, A.; Liang, A.C.; Maeda, M.; Egawa, N.; Itoh, K.; Lo, E.K.; Lok, J.; Ihara, M.; et al. Astrocytes Promote Oligodendrogenesis after White Matter Damage via Brain-Derived Neurotrophic Factor. J. Neurosci. 2015, 35, 14002–14008. [Google Scholar] [CrossRef] [PubMed]
  161. Wu, X.; Chen, P.S.; Dallas, S.; Wilson, B.; Block, M.L.; Wang, C.-C.; Kinyamu, H.; Lu, N.; Gao, X.; Leng, Y.; et al. Histone Deacetylase Inhibitors Up-Regulate Astrocyte GDNF and BDNF Gene Transcription and Protect Dopaminergic Neurons. Int. J. Neuropsychopharmacol. 2008, 11, 1123–1134. [Google Scholar] [CrossRef]
  162. Gasiorowska, A.; Wydrych, M.; Drapich, P.; Zadrozny, M.; Steczkowska, M.; Niewiadomski, W.; Niewiadomska, G. The Biology and Pathobiology of Glutamatergic, Cholinergic, and Dopaminergic Signaling in the Aging Brain. Front. Aging Neurosci. 2021, 13, 654931. [Google Scholar] [CrossRef]
  163. Quesseveur, G.; David, D.J.; Gaillard, M.C.; Pla, P.; Wu, M.V.; Nguyen, H.T.; Nicolas, V.; Auregan, G.; David, I.; Dranovsky, A.; et al. BDNF Overexpression in Mouse Hippocampal Astrocytes Promotes Local Neurogenesis and Elicits Anxiolytic-like Activities. Transl. Psychiatry 2013, 3, e253. [Google Scholar] [CrossRef]
  164. Wang, L.; Lin, F.; Wang, J.; Wu, J.; Han, R.; Zhu, L.; Zhang, G.; DiFiglia, M.; Qin, Z. Truncated N-Terminal Huntingtin Fragment with Expanded-Polyglutamine (Htt552-100Q) Suppresses Brain-Derived Neurotrophic Factor Transcription in Astrocytes. Acta Biochim. Biophys. Sin. 2012, 44, 249–258. [Google Scholar] [CrossRef]
  165. Shin, J.-Y.; Fang, Z.-H.; Yu, Z.-X.; Wang, C.-E.; Li, S.-H.; Li, X.-J. Expression of Mutant Huntingtin in Glial Cells Contributes to Neuronal Excitotoxicity. J. Cell Biol. 2005, 171, 1001–1012. [Google Scholar] [CrossRef]
  166. Kreitzer, A.C.; Malenka, R.C. Striatal Plasticity and Basal Ganglia Circuit Function. Neuron 2008, 60, 543–554. [Google Scholar] [CrossRef]
  167. Park, H.; Poo, M. Neurotrophin Regulation of Neural Circuit Development and Function. Nat. Rev. Neurosci. 2013, 14, 7–23. [Google Scholar] [CrossRef] [PubMed]
  168. Baydyuk, M.; Xu, B. BDNF Signaling and Survival of Striatal Neurons. Front. Cell. Neurosci. 2014, 8, 254. [Google Scholar] [CrossRef] [PubMed]
  169. Zuccato, C.; Caettaneo, E. Role of Brain-Derived Neurotrophic Factor in Huntington’s Disease. Prog. Neurobiol. 2007, 81, 294–330. [Google Scholar] [CrossRef]
  170. Hermel, E.; Gafni, J.; Propp, S.S.; Leavitt, B.R.; Wellington, C.L.; Young, J.E.; Hackam, A.S.; Logvinova, A.V.; Peel, A.L.; Chen, S.F.; et al. Specific Caspase Interactions and Amplification Are Involved in Selective Neuronal Vulnerability in Huntington’s Disease. Cell Death Differ. 2004, 11, 424–438. [Google Scholar] [CrossRef]
  171. Shin, C.; Kim, R.; Yoo, D.; Oh, E.; Moon, J.; Kim, M.; Lee, J.-Y.; Kim, J.-M.; Koh, S.-B.; Kim, M.; et al. A Practical Guide for Clinical Approach to Patients with Huntington’s Disease in Korea. J. Mov. Disord. 2024, 17, 138–149. [Google Scholar] [CrossRef]
  172. Hong, Y.; Zhao, T.; Li, X.-J.; Li, S. Mutant Huntingtin Impairs BDNF Release from Astrocytes by Disrupting Conversion of Rab3a-GTP into Rab3a-GDP. J. Neurosci. 2016, 36, 8790–8801. [Google Scholar] [CrossRef]
  173. Sauer, H.; Fischer, W.; Nikkhah, G.; Wiegand, S.J.; Brundin, P.; Lindsay, R.M.; Björklund, A. Brain-Derived Neurotrophic Factor Enhances Function Rather than Survival of Intrastriatal Dopamine Cell-Rich Grafts. Brain Res. 1993, 626, 37–44. [Google Scholar] [CrossRef]
  174. van der Zwaan, K.F.; Feleus, S.; Dekkers, O.M.; Roos, R.A.C.; de Bot, S.T. Total Functioning Capacity Scale in Huntington’s Disease: Natural Course over Time. J. Neurol. 2025, 272, 140. [Google Scholar] [CrossRef]
  175. Shoulson, I. Huntington Disease: Functional Capacities in Patients Treated with Neuroleptic and Antidepressant Drugs. Neurology 1981, 31, 1333–1335. [Google Scholar] [CrossRef]
  176. Fujisawa, M.; Takeshita, Y.; Fujikawa, S.; Matsuo, K.; Okamoto, M.; Tamada, M.; Shimizu, F.; Sano, Y.; Koga, M.; Kanda, T. Exploring Lipophilic Compounds That Induce BDNF Secretion in Astrocytes beyond the BBB Using a New Multi-Cultured Human in Vitro BBB Model. J. Neuroimmunol. 2022, 362, 577783. [Google Scholar] [CrossRef]
  177. Saylor, A.J.; McGinty, J.F. An Intrastriatal Brain-Derived Neurotrophic Factor Infusion Restores Striatal Gene Expression in Bdnf Heterozygous Mice. Brain Struct. Funct. 2010, 215, 97–104. [Google Scholar] [CrossRef] [PubMed]
  178. Nakao, N.; Brundin, P.; Funa, K.; Lindvall, O.; Odin, P. Trophic and Protective Actions of Brain-Derived Neurotrophic Factor on Striatal DARPP-32-Containing Neurons in Vitro. Dev. Brain Res. 1995, 90, 92–101. [Google Scholar] [CrossRef] [PubMed]
  179. Palizvan, M.R.; Sohya, K.; Kohara, K.; Maruyama, A.; Yasuda, H.; Kimura, F.; Tsumoto, T. Brain-Derived Neurotrophic Factor Increases Inhibitory Synapses, Revealed in Solitary Neurons Cultured from Rat Visual Cortex. Neuroscience 2004, 126, 955–966. [Google Scholar] [CrossRef] [PubMed]
  180. Rauskolb, S.; Zagrebelsky, M.; Dreznjak, A.; Deogracias, R.; Matsumoto, T.; Wiese, S.; Erne, B.; Sendtner, M.; Schaeren-Wiemers, N.; Korte, M.; et al. Global Deprivation of Brain-Derived Neurotrophic Factor in the CNS Reveals an Area-Specific Requirement for Dendritic Growth. J. Neurosci. 2010, 30, 1739–1749. [Google Scholar] [CrossRef]
  181. Login, I.S.; Cronin, M.J.; MacLeod, R.M. Tetrabenazine Has Properties of a Dopamine Receptor Antagonist. Ann. Neurol. 1982, 12, 257–262. [Google Scholar] [CrossRef]
  182. Shahani, L. Tetrabenazine and Suicidal Ideation. J. Neuropsychiatry Clin. Neurosci. 2013, 25, E30. [Google Scholar] [CrossRef]
  183. Huntington Study Group. Unified Huntington’s Disease Rating Scale: Reliability and Consistency. Mov. Disord. 1996, 11, 136–142. [Google Scholar] [CrossRef]
  184. Mestre, T.A.; Forjaz, M.J.; Mahlknecht, P.; Cardoso, F.; Ferreira, J.J.; Reilmann, R.; Sampaio, C.; Goetz, C.G.; Cubo, E.; Martinez-Martin, P.; et al. Rating Scales for Motor Symptoms and Signs in Huntington’s Disease: Critique and Recommendations. Mov. Disord. Clin. Pract. 2018, 5, 111–117. [Google Scholar] [CrossRef] [PubMed]
  185. Mitchell, C.T.; Krier, I.; Arjomand, J.; Borowsky, B.; Tabrizi, S.J.; Leavitt, B.R.; Arran, N.; Axelson, E.; Bardinet, E.; Bechtel, N.; et al. Longitudinal Expression Changes Are Weak Correlates of Disease Progression in Huntington’s Disease. Brain Commun. 2020, 2, fcaa172. [Google Scholar] [CrossRef] [PubMed]
  186. Labadorf, A.; Choi, S.H.; Myers, R.H. Evidence for a Pan-Neurodegenerative Disease Response in Huntington’s and Parkinson’s Disease Expression Profiles. Front. Mol. Neurosci. 2018, 10, 430. [Google Scholar] [CrossRef]
  187. Agus, F.; Crespo, D.; Myers, R.H.; Labadorf, A. The Caudate Nucleus Undergoes Dramatic and Unique Transcriptional Changes in Human Prodromal Huntington’s Disease Brain. BMC Med. Genom. 2019, 12, 137. [Google Scholar] [CrossRef]
  188. Labadorf, A.; Hoss, A.G.; Lagomarsino, V.; Latourelle, J.C.; Hadzi, T.C.; Bregu, J.; MacDonald, M.E.; Gusella, J.F.; Chen, J.F.; Akbarian, S.; et al. RNA Sequence Analysis of Human Huntington Disease Brain Reveals an Extensive Increase in Inflammatory and Developmental Gene Expression. PLoS ONE 2015, 10, e0143563, Erratum in PLoS ONE 2016, 11, e0160295. [Google Scholar] [CrossRef]
  189. Dias-Pinto, J.R.; Faustinoni-Neto, B.; Sanches-Fernandes, J.M.; Kerkis, I.; Araldi, R.P. How Does the Age of Control Individuals Hinder the Identification of Target Genes for Huntington’s Disease? Front. Genet. 2024, 15, 1377237. [Google Scholar] [CrossRef]
  190. Dias-Pinto, J.R.; Faustinoni-Neto, B.; Munhoz, L.; Kerkis, I.; Araldi, R.P. From Brain to Blood: Uncovering Potential Therapeutical Targets and Biomarkers for Huntington’s Disease Using an Integrative RNA-Seq Analytical Platform (BDASeq®). Cells 2025, 14, 976. [Google Scholar] [CrossRef]
  191. Lin, L.; Park, J.W.; Ramachandran, S.; Zhang, Y.; Tseng, Y.T.; Shen, S.; Waldvogel, H.J.; Curtis, M.A.; Richard, R.L.; Troncoso, J.C.; et al. Transcriptome Sequencing Reveals Aberrant Alternative Splicing in Huntington’s Disease. Hum. Mol. Genet. 2016, 25, 3454–3466. [Google Scholar] [CrossRef]
  192. Selemon, L.D.; Rajkowska, G.; Goldman-Rakic, P.S. Evidence for Progression in Frontal Cortical Pathology in Late-stage Huntington’s Disease. J. Comp. Neurol. 2004, 468, 190–204. [Google Scholar] [CrossRef]
  193. Delmaire, C.; Dumas, E.M.; Sharman, M.A.; van den Bogaard, S.J.A.; Valabregue, R.; Jauffret, C.; Justo, D.; Reilmann, R.; Stout, J.C.; Craufurd, D.; et al. The Structural Correlates of Functional Deficits in Early Huntington’s Disease. Hum. Brain Mapp. 2013, 34, 2141–2153. [Google Scholar] [CrossRef] [PubMed]
  194. Furr Stimming, E.; Claassen, D.O.; Kayson, E.; Goldstein, J.; Mehanna, R.; Zhang, H.; Liang, G.S.; Haubenberger, D.; Adams, J.; Beck, C.; et al. Safety and Efficacy of Valbenazine for the Treatment of Chorea Associated with Huntington’s Disease (KINECT-HD): A Phase 3, Randomised, Double-Blind, Placebo-Controlled Trial. Lancet Neurol. 2023, 22, 494–504. [Google Scholar] [CrossRef] [PubMed]
  195. Heim, B.; Seppi, K. Valbenazine as Treatment for Huntington’s Disease Chorea. Lancet Neurol. 2023, 22, 459–460. [Google Scholar] [CrossRef] [PubMed]
  196. Kaur, N.; Kumar, P.; Jamwal, S.; Deshmukh, R.; Gauttam, V. Tetrabenazine: Spotlight on Drug Review. Ann. Neurosci. 2016, 23, 176–185. [Google Scholar] [CrossRef] [PubMed]
  197. Aylward, E.H.; Sparks, B.F.; Field, K.M.; Yallapragada, V.; Shpritz, B.D.; Rosenblatt, A.; Brandt, J.; Gourley, L.M.; Liang, K.; Zhou, H.; et al. Onset and Rate of Striatal Atrophy in Preclinical Huntington Disease. Neurology 2004, 63, 66–72. [Google Scholar] [CrossRef]
  198. Aylward, E.H.; Nopoulos, P.C.; Ross, C.A.; Langbehn, D.R.; Pierson, R.K.; Mills, J.A.; Johnson, H.J.; Magnotta, V.A.; Juhl, A.R.; Paulsen, J.S. Longitudinal Change in Regional Brain Volumes in Prodromal Huntington Disease. J. Neurol. Neurosurg. Psychiatry 2011, 82, 405–410. [Google Scholar] [CrossRef]
  199. Sneha, N.P.; Dharshini, S.A.P.; Taguchi, Y.-h.; Gromiha, M.M. Investigating Neuron Degeneration in Huntington’s Disease Using RNA-Seq Based Transcriptome Study. Genes 2023, 14, 1801. [Google Scholar] [CrossRef]
  200. Seefelder, M.; Kochanek, S. A Meta-Analysis of Transcriptomic Profiles of Huntington’s Disease Patients. PLoS ONE 2021, 16, e0253037. [Google Scholar] [CrossRef]
  201. Research and Markets. Global Huntington’s Disease Treatment Market Forecast Report by Drug Type (Approved Drugs, Off-Label Drugs) End User (Hospital Pharmacy, Drug Store & Retail Pharmacy, Online Pharmacy) Countries and Company Analysis, 2024–2032; Research and Markets: Dublin, Ireland, 2024; Available online: https://www.researchandmarkets.com/report/huntington-disease-drug?srsltid=AfmBOop1kZhsccNpDXkzXcUx9MvGnn1EBOuWOCFGtC96HHJEb7RSILHR (accessed on 1 August 2025).
  202. Claassen, D.O.; Ayyagari, R.; Garcia-Horton, V.; Zhang, S.; Alexander, J.; Leo, S. Real-World Adherence to Tetrabenazine or Deutetrabenazine Among Patients with Huntington’s Disease: A Retrospective Database Analysis. Neurol. Ther. 2022, 11, 435–448. [Google Scholar] [CrossRef]
  203. Frank, S.; Testa, C.; Edmondson, M.C.; Goldstein, J.; Kayson, E.; Leavitt, B.R.; Oakes, D.; O’Neill, C.; Vaughan, C.; Whaley, J.; et al. The Safety of Deutetrabenazine for Chorea in Huntington Disease: An Open-Label Extension Study. CNS Drugs 2022, 36, 1207–1216. [Google Scholar] [CrossRef]
  204. Rodrigues, F.B.; Duarte, G.S.; Costa, J.; Ferreira, J.J.; Wild, E.J. Tetrabenazine Versus Deutetrabenazine for Huntington’s Disease: Twins or Distant Cousins? Mov. Disord. Clin. Pract. 2017, 4, 582–585. [Google Scholar] [CrossRef]
  205. Claassen, D.O.; Carroll, B.; De Boer, L.M.; Wu, E.; Ayyagari, R.; Gandhi, S.; Stamler, D. Indirect Tolerability Comparison of Deutetrabenazine and Tetrabenazine for Huntington Disease. J. Clin. Mov. Disord. 2017, 4, 3. [Google Scholar] [CrossRef] [PubMed]
  206. Dalton, M.P.; Cheng, M.H.; Bahar, I.; Coleman, J.A. Structural Mechanisms for VMAT2 Inhibition by Tetrabenazine. eLife 2024, 12, RP91973. [Google Scholar] [CrossRef]
  207. Alothman, D.; Marshall, C.R.; Tyrrell, E.; Lewis, S.; Card, T.; Fogarty, A. Risk of Mortality from Suicide in Patients with Huntington’s Disease Is Increased Compared to the General Population in England. J. Neurol. 2022, 269, 4436–4439. [Google Scholar] [CrossRef] [PubMed]
  208. Molnar, M.J.; Molnar, V.; Fedor, M.; Csehi, R.; Acsai, K.; Borsos, B.; Grosz, Z. Improving Mood and Cognitive Symptoms in Huntington’s Disease with Cariprazine Treatment. Front. Psychiatry 2022, 12, 825532. [Google Scholar] [CrossRef] [PubMed]
  209. van Duijn, E. Treatment of Irritability in Huntington’s Disease. Curr. Treat. Options Neurol. 2010, 12, 424–433. [Google Scholar] [CrossRef]
  210. Andriessen, R.L.; Oosterloo, M.; Hollands, A.; Linden, D.E.J.; de Greef, B.T.A.; Leentjens, A.F.G. Psychotropic Medication Use in Huntington’s Disease: A Retrospective Cohort Study. Parkinsonism Relat. Disord. 2022, 105, 69–74. [Google Scholar] [CrossRef]
  211. Kerkis, I.; Wenceslau, C.V.; Araldi, R.P. Stem Cells from Dental Pulp of Deciduous Teeth: Twenty Years of Experience. In Recent Update on Mesenchymal Stem Cells; IntechOpen: London, UK, 2024. [Google Scholar]
  212. Araldi, R.P.; Dias-Pinto, J.; Kerkis, I. Pharma Intelligence Applied to the Research and Development of Cell and Cell-Free Therapy: A New Era of the Medicine. In Recent Update on Mesenchymal Stem Cells; IntechOpen: London, UK, 2024. [Google Scholar]
  213. Araldi, R.P.; Viana, M.; Colozza-Gama, G.A.; Dias-Pinto, J.R.; Ankol, L.; Valverde, C.W.; Perlson, E.; Kerkis, I. Unique Transcriptional Signatures Observed in Stem Cells from the Dental Pulp of Deciduous Teeth Produced on a Large Scale. Pharmacologia 2023, 14, 72–95. [Google Scholar] [CrossRef]
  214. Araldi, R.P.; D’Amelio, F.; Vigerelli, H.; de Melo, T.C.; Kerkis, I. Stem Cell-Derived Exosomes as Therapeutic Approach for Neurodegenerative Ddsorders: From Biology to Biotechnology. Cells 2020, 9, 2663. [Google Scholar] [CrossRef]
  215. Kerkis, I.; Wenceslau, C.V.; Souza, D.M.; Mambelli-Lisboa, N.C.; Ynoue, L.H.; Araldi, R.P.; Silva, J.M.; Pagani, E.; Haddad, M.S. Preclincal Assessment of NestaCell® in Huntigton’s Disease 3-NP Rat Model Demonstrates Restoration of BDNF, DARPP32, and D2R Expression Following Intravenous Single versus Multiple Injections. Cytotherapy 2022, 24, S5. [Google Scholar] [CrossRef]
  216. Araldi, R.P.; Ramos, A.T.; Alievi, A.L.; Policíquio, B.; Teixei, M.R.; Mendes, T.B.; Gongaza, V.; Costa, V.R.; Troncone, L.R.; Kerkis, I. NestaCell® Promotes Motor, Cognitive and Neuropsychiatric Functions Amelioration and Dopaminergic Neurons Restoration in a Preclinical Model of Parkinson’s Disease. Cytotherapy 2022, 24, S3. [Google Scholar] [CrossRef]
  217. da Silva, J.M.; Araldi, R.P.; Colozza-Gama, G.A.; Pagani, E.; Sid, A.; Valverde, C.W.; Kerkis, I. Human Immature Dental Pulp Stem Cells Did Not Graft into a Preexisting Human Lung Adenocarcinoma. Case Rep. Oncol. 2022, 15, 413–422. [Google Scholar] [CrossRef] [PubMed]
  218. Niazi, S.K.; Mariam, Z. Computer-Aided Drug Design and Drug Discovery: A Prospective Analysis. Pharmaceuticals 2023, 17, 22. [Google Scholar] [CrossRef] [PubMed]
  219. Mihai, D.P.; Nitulescu, G.M. Computer-Aided Drug Design and Drug Discovery. Pharmaceuticals 2025, 18, 436. [Google Scholar] [CrossRef]
  220. Faustinoni-Neto, B.; Dias-Pinto, J.R.; Araldi, R.P. Pharma Data Analytics (Pharma Intelligence): Opportunities and Challenges in the Multi-Omics Era of Drug Discovery and Development. Pharmacologia 2023, 14, 29–39. [Google Scholar] [CrossRef]
  221. Dias-Pinto, J.R.; Faustinoni-Neto, B.; Araldi, R.P. BDASeq: A Novel and Poweful Tool Able to Accurately Discover Theraeputic Targets for the Treatment of Rare Diseases. In Proceedings of the MENA Rare Disease Congress, Abu Dhabi, United Arab Emirates, 16–19 May 2024; p. 5. [Google Scholar]
  222. Araldi, R.P.; Faustinoni-Neto, B.; Dias-Pinto, J.R. Novel Pharmaceutical Tragets and Prognostic Biomarkers for Huntington’s Disease: New Perspectives for the Drug Development. In Proceedings of the MENA Rare Disease Congress, Abu Dhabi, United Arab Emirates, 16–19 May 2024; pp. 16–19. [Google Scholar]
  223. Stark, R.; Grzelak, M.; Hadfield, J. RNA Sequencing: The Teenage Years. Nat. Rev. Genet. 2019, 20, 631–656. [Google Scholar] [CrossRef]
  224. Conesa, A.; Madrigal, P.; Tarazona, S.; Gomez-Cabrero, D.; Cervera, A.; McPherson, A.; Szcześniak, M.W.; Gaffney, D.J.; Elo, L.L.; Zhang, X.; et al. A Survey of Best Practices for RNA-Seq Data Analysis. Genome Biol. 2016, 17, 1–19. [Google Scholar] [CrossRef]
  225. Lamarre, S.; Frasse, P.; Zouine, M.; Labourdette, D.; Sainderichin, E.; Hu, G.; Le Berre-Anton, V.; Bouzayen, M.; Maza, E. Optimization of an RNA-Seq Differential Gene Expression Analysis Depending on Biological Replicate Number and Library Size. Front. Plant Sci. 2018, 9, 108. [Google Scholar] [CrossRef]
  226. Maza, E. In Papyro Comparison of TMM (EdgeR), RLE (DESeq2), and MRN Normalization Methods for a Simple Two-Conditions-Without-Replicates RNA-Seq Experimental Design. Front. Genet. 2016, 7, 164. [Google Scholar] [CrossRef]
  227. Zhao, Y.; Li, M.-C.; Konaté, M.M.; Chen, L.; Das, B.; Karlovich, C.; Williams, P.M.; Evrard, Y.A.; Doroshow, J.H.; McShane, L.M. TPM, FPKM, or Normalized Counts? A Comparative Study of Quantification Measures for the Analysis of RNA-Seq Data from the NCI Patient-Derived Models Repository. J. Transl. Med. 2021, 19, 269. [Google Scholar] [CrossRef]
  228. Love, M.I.; Huber, W.; Anders, S. Moderated Estimation of Fold Change and Dispersion for RNA-Seq Data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef]
  229. Dias-Pinto, J.R.; Araldi, R.P.; Faustinoni-Neto, B. Biomarker Discovery Algorithm (BDASeq): An Integrative Analytics-Based Tool Designed to Identify Potential Diagnostic and Therapeutic Targets for Diseases. In Proceedings of the MENA Rare Disease Congress, Abu Dhabi, United Arab Emirates, 16–19 May 2024; p. 1. [Google Scholar]
  230. Robinson, M.D.; McCarthy, D.J.; Smyth, G.K. EdgeR: A Bioconductor Package for Differential Expression Analysis of Digital Gene Expression Data. Bioinformatics 2010, 26, 139–140. [Google Scholar] [CrossRef]
  231. Ritchie, M.E.; Phipson, B.; Wu, D.; Hu, Y.; Law, C.W.; Shi, W.; Smyth, G.K. Limma Powers Differential Expression Analyses for RNA-Sequencing and Microarray Studies. Nucleic Acids Res. 2015, 43, e47. [Google Scholar] [CrossRef]
  232. Tarazona, S.; García, F.; Ferrer, A.; Dopazo, J.; Conesa, A. NOIseq: A RNA-Seq Differential Expression Method Robust for Sequencing Depth Biases. EMBnet J. 2012, 17, 18. [Google Scholar] [CrossRef]
  233. Leng, N.; Dawson, J.A.; Thomson, J.A.; Ruotti, V.; Rissman, A.I.; Smits, B.M.G.; Haag, J.D.; Gould, M.N.; Stewart, R.M.; Kendziorski, C. EBSeq: An Empirical Bayes Hierarchical Model for Inference in RNA-Seq Experiments. Bioinformatics 2013, 29, 1035–1043. [Google Scholar] [CrossRef] [PubMed]
  234. Gauthier, M.; Agniel, D.; Thiébaut, R.; Hejblum, B.P. Dearseq: A Variance Component Score Test for RNA-Seq Differential Analysis That Effectively Controls the False Discovery Rate. NAR Genom. Bioinform. 2020, 2, lqaa093. [Google Scholar] [CrossRef] [PubMed]
  235. Wilcoxon, F. Individual Comparisons by Ranking Methods. Biom. Bull. 1945, 1, 80–83. [Google Scholar] [CrossRef]
  236. Firth, D. Bias Reduction of Maximum Likelihood Estimates. Biometrika 1993, 80, 27–38. [Google Scholar] [CrossRef]
  237. Unti, E.; Mazzucchi, S.; Kiferle, L.; Bonuccelli, U.; Ceravolo, R. Q09 Valproic Acid for the Treatment of Aggressiveness in Huntington’s Disease: 1-Year Follow-Up. J. Neurol. Neurosurg. Psychiatry 2012, 83, A57. [Google Scholar] [CrossRef]
  238. Saft, C.; Lauter, T.; Kraus, P.H.; Przuntek, H.; Andrich, J.E. Dose-Dependent Improvement of Myoclonic Hyperkinesia Due to Valproic Acid in Eight Huntington’s Disease Patients: A Case Series. BMC Neurol. 2006, 6, 11. [Google Scholar] [CrossRef]
  239. Pearce, I.; Heathfield, K.W.G.; Pearce, J.M.S. Valproate Sodium in Huntington Chorea. Arch. Neurol. 1977, 34, 308–309. [Google Scholar] [CrossRef]
  240. Scholefield, M.; Patassini, S.; Xu, J.; Cooper, G.J.S. Widespread Selenium Deficiency in the Brain of Cases with Huntington’s Disease Presents a New Potential Therapeutic Target. EBioMedicine 2023, 97, 104824. [Google Scholar] [CrossRef] [PubMed]
  241. Lu, Z.; Marks, E.; Chen, J.; Moline, J.; Barrows, L.; Raisbeck, M.; Volitakis, I.; Cherny, R.A.; Chopra, V.; Bush, A.I.; et al. Altered Selenium Status in Huntington’s Disease: Neuroprotection by Selenite in the N171-82Q Mouse Model. Neurobiol. Dis. 2014, 71, 34–42. [Google Scholar] [CrossRef] [PubMed]
  242. Kumar, P.; Kalonia, H.; Kumar, A. Cyclosporine A Attenuates 3-Nitropropionic Acid–Induced Huntington-Like Symptoms in Rats: Possible Nitric Oxide Mechanism. Int. J. Toxicol. 2010, 29, 318–325. [Google Scholar] [CrossRef] [PubMed]
  243. Quintanilla, R.A.; Jin, Y.N.; von Bernhardi, R.; Johnson, G.V. Mitochondrial Permeability Transition Pore Induces Mitochondria Injury in Huntington Disease. Mol. Neurodegener. 2013, 8, 45. [Google Scholar] [CrossRef]
  244. Cong, W.; Bai, R.; Li, Y.-F.; Wang, L.; Chen, C. Selenium Nanoparticles as an Efficient Nanomedicine for the Therapy of Huntington’s Disease. ACS Appl. Mater. Interfaces 2019, 11, 34725–34735. [Google Scholar] [CrossRef]
  245. Koller, W.C.; Barr, A.; Biary, N. Estrogen Treatment of Dyskinetic Disorders. Neurology 1982, 32, 547. [Google Scholar] [CrossRef]
  246. Nuzzo, M.; Marino, M. Estrogen/Huntingtin: A Novel Pathway Involved in Neuroprotection. Neural Regen. Res. 2016, 11, 402–403. [Google Scholar] [CrossRef]
  247. Nilsen, J. Estradiol and Neurodegenerative Oxidative Stress. Front. Neuroendocr. 2008, 29, 463–475. [Google Scholar] [CrossRef]
  248. Bassani, T.B.; Bartolomeo, C.S.; Oliveira, R.B.; Ureshino, R.P. Progestogen-Mediated Neuroprotection in Central Nervous System Disorders. Neuroendocrinology 2023, 113, 14–35. [Google Scholar] [CrossRef]
  249. Kumar, P.; Kumar, P.; Deshmukh, R.; Sharma, P. P2–044: Neuroprotective Role of Progesterone against 3-nitropropionic-acid–Induced Huntington’s Disease-like Symptoms in Rats. Alzheimer’s Dement. 2013, 9, P360. [Google Scholar] [CrossRef]
  250. Theis, V.; Theiss, C. Progesterone: A Universal Stimulus for Neuronal Cells? Neural Regen. Res. 2015, 10, 547. [Google Scholar] [CrossRef] [PubMed]
  251. Li, J.; Wang, Y.; Yang, R.; Ma, W.; Yan, J.; Li, Y.; Chen, G.; Pan, J. Pain in Huntington’s Disease and Its Potential Mechanisms. Front. Aging Neurosci. 2023, 15, 563. [Google Scholar] [CrossRef] [PubMed]
  252. Roos, R.A.C.; Steenvoorden, J.M.C.; Mulder, G.J.; Van Kempen, G.M.J. Acetaminophen Sulfation in Patients with Parkinson’s Disease or Huntington’s Disease Is Not Impaired. Neurology 1993, 43, 1373. [Google Scholar] [CrossRef] [PubMed]
  253. Siebzehnrübl, F.A.; Raber, K.A.; Urbach, Y.K.; Schulze-Krebs, A.; Canneva, F.; Moceri, S.; Habermeyer, J.; Achoui, D.; Gupta, B.; Steindler, D.A.; et al. Early Postnatal Behavioral, Cellular, and Molecular Changes in Models of Huntington Disease Are Reversible by HDAC Inhibition. Proc. Natl. Acad. Sci. USA 2018, 115, E8765–E8774. [Google Scholar] [CrossRef]
  254. Athira, K.V.; Sadanandan, P.; Chakravarty, S. Repurposing Vorinostat for the Treatment of Disorders Affecting Brain. Neuromol. Med. 2021, 23, 449–465. [Google Scholar] [CrossRef]
  255. Shukla, S.; Shariat-Madar, Z.; Walker, L.A.; Tekwani, B.L. Mechanism for Neurotropic Action of Vorinostat, a Pan Histone Deacetylase Inhibitor. Mol. Cell. Neurosci. 2016, 77, 11–20. [Google Scholar] [CrossRef]
  256. Gray, S.G. Targeting Huntington’s Disease through Histone Deacetylases. Clin. Epigenet. 2011, 2, 257–277. [Google Scholar] [CrossRef]
  257. Kang, J.; Park, M.; Lee, E.; Jung, J.; Kim, T. The Role of Vitamin D in Alzheimer’s Disease: A Transcriptional Regulator of Amyloidopathy and Gliopathy. Biomedicines 2022, 10, 1824. [Google Scholar] [CrossRef]
  258. Jeong, S.P.; Sharma, N.; An, S.S.A. Role of Calcitriol and Vitamin D Receptor (VDR) Gene Polymorphisms in Alzheimer’s Disease. Int. J. Mol. Sci. 2024, 25, 4806. [Google Scholar] [CrossRef]
  259. Fryer, M.J. Vitamin E Status and Neurodegenerative Disease. Nutr. Neurosci. 1998, 1, 327–351. [Google Scholar] [CrossRef]
  260. Kašparová, S.; Sumbalová, Z.; Bystrický, P.; Kucharská, J.; Liptaj, T.; Mlynárik, V.; Gvozdjáková, A. Effect of Coenzyme Q10 and Vitamin E on Brain Energy Metabolism in the Animal Model of Huntington’s Disease. Neurochem. Int. 2006, 48, 93–99. [Google Scholar] [CrossRef]
  261. Dickey, A.S.; Pineda, V.V.; Tsunemi, T.; Liu, P.P.; Miranda, H.C.; Gilmore-Hall, S.K.; Lomas, N.; Sampat, K.R.; Buttgereit, A.; Torres, M.-J.M.; et al. PPAR-δ Is Repressed in Huntington’s Disease, Is Required for Normal Neuronal Function and Can Be Targeted Therapeutically. Nat. Med. 2016, 22, 37–45. [Google Scholar] [CrossRef]
  262. Pierzynowska, K.; Gaffke, L.; Cyske, Z.; Węgrzyn, G. Genistein Induces Degradation of Mutant Huntingtin in Fibroblasts from Huntington’s Disease Patients. Metab. Brain Dis. 2019, 34, 715–720. [Google Scholar] [CrossRef]
  263. Sharma, D.; Singh, V.; Kumar, A.; Singh, T.G. Genistein: A Promising Ally in Combating Neurodegenerative Disorders. Eur. J. Pharmacol. 2025, 991, 177273. [Google Scholar] [CrossRef] [PubMed]
  264. Menze, E.T.; Esmat, A.; Tadros, M.G.; Abdel-Naim, A.B.; Khalifa, A.E. Genistein Improves 3-NPA-Induced Memory Impairment in Ovariectomized Rats: Impact of Its Antioxidant, Anti-Inflammatory and Acetylcholinesterase Modulatory Properties. PLoS ONE 2015, 10, e0117223. [Google Scholar] [CrossRef] [PubMed]
  265. Li, R.; Robinson, M.; Ding, X.; Geetha, T.; Al-Nakkash, L.; Broderick, T.L.; Babu, J.R. Genistein: A Focus on Several Neurodegenerative Diseases. J. Food Biochem. 2022, 46, e14155. [Google Scholar] [CrossRef] [PubMed]
  266. Vuu, Y.M.; Kadar Shahib, A.; Rastegar, M. The Potential Therapeutic Application of Simvastatin for Brain Complications and Mechanisms of Action. Pharmaceuticals 2023, 16, 914. [Google Scholar] [CrossRef]
  267. da Cruz, J.N.; Magro, D.D.D.; de Lima, D.D.; da Cruz, J.G.P. Simvastatin Treatment Reduces the Cholesterol Content of Membrane/Lipid Rafts, Implicating the N-Methyl-D-Aspartate Receptor in Anxiety: A Literature Review. Braz. J. Pharm. Sci. 2017, 53, e16102. [Google Scholar] [CrossRef]
  268. Maheshwari, M.; Bhutani, S.; Das, A.; Mukherjee, R.; Sharma, A.; Kino, Y.; Nukina, N.; Jana, N.R. Dexamethasone Induces Heat Shock Response and Slows down Disease Progression in Mouse and Fly Models of Huntington’s Disease. Hum. Mol. Genet. 2014, 23, 2737–2751. [Google Scholar] [CrossRef]
  269. Haddad, M.S.; Wenceslau, C.V.; Pompeia, C.; Kerkis, I. Cell-Based Technologies for Huntington’s Disease. Dement. Neuropsychol. 2016, 10, 287–295. [Google Scholar] [CrossRef]
  270. Macedo, J.; Pagani, E.; Wenceslau, C.; Ferrara, L.; Kerkis, I. A Pahse I Clinical Trial on Intravenous Administration of Immature Human Dental Pulp Stem Cells (NestaCell) to Huntington’s Disease Patients. Cytotherapy 2021, 23, 1. [Google Scholar] [CrossRef]
Figure 1. Growth in Huntington’s disease research publications over time. Number of publications related to Huntington’s disease indexed in PubMed from the initial clinical description in 1872 to 10 March 2025. A marked increase in publication volume is observed beginning in 1993, coinciding with the discovery of the genetic cause of HD—the CAG trinucleotide repeat expansion in the HTT gene—highlighting the pivotal impact of this breakthrough on research activity in the field [11].
Figure 1. Growth in Huntington’s disease research publications over time. Number of publications related to Huntington’s disease indexed in PubMed from the initial clinical description in 1872 to 10 March 2025. A marked increase in publication volume is observed beginning in 1993, coinciding with the discovery of the genetic cause of HD—the CAG trinucleotide repeat expansion in the HTT gene—highlighting the pivotal impact of this breakthrough on research activity in the field [11].
Brainsci 15 00865 g001
Figure 2. Genetic etiology and pathophysiological consequences of HD. Schematic representation of the genetic basis of HD. (a) HD is an autosomal dominant disorder caused by a CAG trinucleotide repeat expansion in the first exon of the HTT gene. (b) This expansion encodes an abnormally elongated polyglutamine (polyQ) tract in the huntingtin protein. Individuals with intermediate alleles (27–35 CAG repeats) are typically unaffected but may transmit expanded alleles into the pathogenic range during germline transmission. Some carriers of intermediate alleles can exhibit HD-like phenotypes, including motor dysfunction, cognitive decline, and behavioral disturbances. Individuals with ≥36 CAG repeats are considered mutation carriers: those with 36–39 repeats show reduced penetrance and may remain asymptomatic, whereas those with ≥40 repeats exhibit full penetrance and develop clinical HD. (c) The mutant huntingtin protein (mHTT) induces widespread cellular dysfunction, ultimately leading to neuronal degeneration, with prominent loss observed in the striatum.
Figure 2. Genetic etiology and pathophysiological consequences of HD. Schematic representation of the genetic basis of HD. (a) HD is an autosomal dominant disorder caused by a CAG trinucleotide repeat expansion in the first exon of the HTT gene. (b) This expansion encodes an abnormally elongated polyglutamine (polyQ) tract in the huntingtin protein. Individuals with intermediate alleles (27–35 CAG repeats) are typically unaffected but may transmit expanded alleles into the pathogenic range during germline transmission. Some carriers of intermediate alleles can exhibit HD-like phenotypes, including motor dysfunction, cognitive decline, and behavioral disturbances. Individuals with ≥36 CAG repeats are considered mutation carriers: those with 36–39 repeats show reduced penetrance and may remain asymptomatic, whereas those with ≥40 repeats exhibit full penetrance and develop clinical HD. (c) The mutant huntingtin protein (mHTT) induces widespread cellular dysfunction, ultimately leading to neuronal degeneration, with prominent loss observed in the striatum.
Brainsci 15 00865 g002
Figure 3. Structural features and proteolytic processing sites of the huntingtin protein. Schematic representation of the huntingtin amino acid sequence. The polyglutamine tract, denoted by (Q)ₙ, is followed by a proline-rich domain (P)ₙ. Red squares indicate the three main clusters of HEAT repeats, which contribute to the protein’s scaffold-like architecture. Arrows denote caspase cleavage sites and their corresponding amino acid positions, while blue arrowheads indicate calpain cleavage sites. Proteolytic regions preferentially cleaved in the cortex (B), striatum (C), or both regions (A) are labeled accordingly. Green and orange arrowheads mark additional protease-sensitive regions. The nuclear export signal (NES) is also indicated. Red and blue circles represent post-translational modifications: red denotes sites of ubiquitination (UBI) and/or sumoylation (SUMO); blue marks phosphorylation sites at serine 421 and serine 434. Glutamic acid (Glu)-, serine (Ser)-, and proline (Pro)-rich regions are shown, with serine-rich domains highlighted by green outlines. These structural and regulatory elements underscore the complex post-translational modulation and proteolytic fragmentation that contribute to huntingtin’s pathogenicity. Image adapted from Cattaneo et al. [39].
Figure 3. Structural features and proteolytic processing sites of the huntingtin protein. Schematic representation of the huntingtin amino acid sequence. The polyglutamine tract, denoted by (Q)ₙ, is followed by a proline-rich domain (P)ₙ. Red squares indicate the three main clusters of HEAT repeats, which contribute to the protein’s scaffold-like architecture. Arrows denote caspase cleavage sites and their corresponding amino acid positions, while blue arrowheads indicate calpain cleavage sites. Proteolytic regions preferentially cleaved in the cortex (B), striatum (C), or both regions (A) are labeled accordingly. Green and orange arrowheads mark additional protease-sensitive regions. The nuclear export signal (NES) is also indicated. Red and blue circles represent post-translational modifications: red denotes sites of ubiquitination (UBI) and/or sumoylation (SUMO); blue marks phosphorylation sites at serine 421 and serine 434. Glutamic acid (Glu)-, serine (Ser)-, and proline (Pro)-rich regions are shown, with serine-rich domains highlighted by green outlines. These structural and regulatory elements underscore the complex post-translational modulation and proteolytic fragmentation that contribute to huntingtin’s pathogenicity. Image adapted from Cattaneo et al. [39].
Brainsci 15 00865 g003
Figure 4. Peripheral organ involvement in HD. Schematic illustration highlighting extra-neuronal manifestations of HD. Mutant huntingtin expression perturbs multiple peripheral tissues, including striated skeletal muscle (myopathy and altered energy metabolism), striated cardiac muscle (cardiomyopathy and conduction abnormalities), pancreas (β-cell dysfunction and impaired glucose homeostasis), hypothalamus (disrupted energy balance and neuroendocrine signaling), hypophysis (pituitary hormone dysregulation), and adrenal glands (aberrant stress axis responses). Together, these systemic alterations underscore the multisystemic nature of Huntington’s disease and reinforce the need for therapeutic strategies that address both central and peripheral pathophysiology.
Figure 4. Peripheral organ involvement in HD. Schematic illustration highlighting extra-neuronal manifestations of HD. Mutant huntingtin expression perturbs multiple peripheral tissues, including striated skeletal muscle (myopathy and altered energy metabolism), striated cardiac muscle (cardiomyopathy and conduction abnormalities), pancreas (β-cell dysfunction and impaired glucose homeostasis), hypothalamus (disrupted energy balance and neuroendocrine signaling), hypophysis (pituitary hormone dysregulation), and adrenal glands (aberrant stress axis responses). Together, these systemic alterations underscore the multisystemic nature of Huntington’s disease and reinforce the need for therapeutic strategies that address both central and peripheral pathophysiology.
Brainsci 15 00865 g004
Figure 5. Primary and secondary outcome measures in Huntington’s disease clinical trials. Key clinical features used as outcome measures in trials for HD, categorized into primary (A) and secondary endpoints (B). Analysis of 256 clinical trials registered on ClinicalTrials.gov as of 1 April 2025 reveals that, in the absence of validated biomarkers, the Unified Huntington’s Disease Rating Scale (UHDRS) remains the most widely used tool to assess treatment efficacy. Data were generated using BioDecision AnalyticsTM and reflect the current landscape of evidence generation in HD clinical research.
Figure 5. Primary and secondary outcome measures in Huntington’s disease clinical trials. Key clinical features used as outcome measures in trials for HD, categorized into primary (A) and secondary endpoints (B). Analysis of 256 clinical trials registered on ClinicalTrials.gov as of 1 April 2025 reveals that, in the absence of validated biomarkers, the Unified Huntington’s Disease Rating Scale (UHDRS) remains the most widely used tool to assess treatment efficacy. Data were generated using BioDecision AnalyticsTM and reflect the current landscape of evidence generation in HD clinical research.
Brainsci 15 00865 g005
Figure 6. Candidate therapeutic compounds for HD identified through integrative AI-guided drug discovery. Drugs predicted using LBDD and pharmacogenomic approaches, based on 394 putative druggable genes identified by BDASeq®. Bar graph displays candidate compounds, with numbers within bars indicating the count of target genes modulated by each drug. This integrative strategy prioritizes compounds with potential to modulate multiple HD-relevant pathways, offering a platform for rational drug repurposing and multi-target intervention.
Figure 6. Candidate therapeutic compounds for HD identified through integrative AI-guided drug discovery. Drugs predicted using LBDD and pharmacogenomic approaches, based on 394 putative druggable genes identified by BDASeq®. Bar graph displays candidate compounds, with numbers within bars indicating the count of target genes modulated by each drug. This integrative strategy prioritizes compounds with potential to modulate multiple HD-relevant pathways, offering a platform for rational drug repurposing and multi-target intervention.
Brainsci 15 00865 g006
Table 1. Comparative clinical outcomes of tetrabenazine and deutetrabenazine in HD.
Table 1. Comparative clinical outcomes of tetrabenazine and deutetrabenazine in HD.
Direct Analyses 1Indirect Analyses 2
Outcomes/Adverse EffectsTBZ-PlaceboDEU-PlaceboTBZ-DEU
UHDRS (chorea)−3.20 3−2.50 4−1.00
UHDRS-TMS−3.30−4.00 40.70
Depression scale0.76 5−0.18 40.94
Insomnia scale1.80 5−0.30 42.10
Severe adverse effects5.441.005.44
Drowsiness13.32 32.694.95
Diarrhea0.729.870.07
Insomnia21.84 31.5414.18
Fatigue1.861.541.21
Fall1.300.482.71
Depression11.150.6517.15
1 For direct analyses, values presented show the difference in mean score between those treated with the drug under investigation and placebo. Positive values favor placebo and negative values favor treatment. 2 For indirect analyses, values represent differences in mean score between tetrabenazine and deutetrabenazine. Positive values favor deutetrabenazine and negative values favor tetrabenazine. 3 Significantly favorable for tetrabenazine. 4 Significantly favorable for deutetrabenazine. 5 Significantly favorable for placebo. Source: modified from Rodrigues et al. [204].
Table 2. Summary of candidate therapeutic agents for HD identified via BDASeq®.
Table 2. Summary of candidate therapeutic agents for HD identified via BDASeq®.
DrugMechanism of ActionHD-Relevant EffectsEvidence LevelReferences
Valproic acidGABAergic activity; HDAC inhibitionReduces aggression, improves motor symptoms (e.g., myoclonic hyperkinesia); potential mood stabilizerPreclinical 1 & clinical 2[237,238,239]
Cyclosporine ACalcineurin inhibition; mitochondrial protectionAttenuates mitochondrial dysfunction and oxidative stress; improves behavior in HD modelsPreclinical[240,241]
SeleniumAntioxidant; glutathione peroxidase cofactorAddresses selenium deficiency in HD brains; reduces oxidative damagePreclinical and Post-mortem Human 3[242,243,244]
EstradiolEstrogen receptor activation; mitochondrial regulationReduces oxidative stress and inflammation; modulates energy metabolismPreclinical[245,246,247]
ProgesteroneNeurosteroid; anti-inflammatoryReduces oxidative stress; neuroprotective in HD modelsPreclinical[247,248,249,250]
AcetaminophenCOX inhibition; analgesicPain relief for muscle aches in early-stage HDClinical Use (Non-HD-specific)[251,252]
PanobinostatHDAC inhibitionModulates gene expression; potential to reduce neurodegenerationPreclinical[253]
Vorinostat (SAHA)HDAC inhibitionImproves neuronal survival; reduces oxidative stressPreclinical[254,255,256]
EntinostatHDAC1/3 selective inhibitorPutative neuroprotective, but not yet studied in HD modelsTheoretical 4
CalcitriolVitamin D receptor activationReduces oxidative stress, inflammation; modulates calcium homeostasis and mitochondriaPreclinical[257,258]
Vitamin EAntioxidantProtects neurons from oxidative stress and lipid peroxidationPreclinical and Clinical (limited)[259,260]
TroglitazonePPAR-γ agonistReduces oxidative stress; improves mitochondrial and energy metabolismPreclinical[261]
GenisteinAutophagy induction; estrogenic activityClears mHTT aggregates; improves motor, cognitive and behavioral outcomesPreclinical[262,263,264,265]
SimvastatinCholesterol modulation; anti-inflammatoryCrosses BBB; reduces oxidative stress; potential neuroprotectionPreclinical and Observational[58,266,267]
DexamethasoneGlucocorticoid receptor agonist; heat shock responseReduces mHTT aggregation; improves behavioral phenotypePreclinical[268]
1 Preclinical: evidence derived from in vitro or animal models. 2 Clinical: evidence from human trials or patient treatment studies. 3 Postmortem human: observations from postmortem tissue. 4 Theoretical: hypothetical or inferred based on drug class, not yet experimentally tested in HD.
Table 3. Main differences between synthetic drugs and hDPSC-based treatments.
Table 3. Main differences between synthetic drugs and hDPSC-based treatments.
FeatureSynthetic DrugshDPSC-Based Treatments
TargetsNarrow (few genes or pathways)Broad (multi-pathway modulation)
NeuroprotectionLimitedStrong (via trophic support)
NeuroregenerationAbsentPotential for neuronal replacement
Immunomodulatory actionIndirectDirect immunomodulatory effects
Disease modificationUnlikelyPromising in preclinical models and Phase I/II clinical trial
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Araldi, R.P.; Dias Pinto, J.R.; Kerkis, I. AI-Enhanced Transcriptomic Discovery of Druggable Targets and Repurposed Therapies for Huntington’s Disease. Brain Sci. 2025, 15, 865. https://doi.org/10.3390/brainsci15080865

AMA Style

Araldi RP, Dias Pinto JR, Kerkis I. AI-Enhanced Transcriptomic Discovery of Druggable Targets and Repurposed Therapies for Huntington’s Disease. Brain Sciences. 2025; 15(8):865. https://doi.org/10.3390/brainsci15080865

Chicago/Turabian Style

Araldi, Rodrigo Pinheiro, João Rafael Dias Pinto, and Irina Kerkis. 2025. "AI-Enhanced Transcriptomic Discovery of Druggable Targets and Repurposed Therapies for Huntington’s Disease" Brain Sciences 15, no. 8: 865. https://doi.org/10.3390/brainsci15080865

APA Style

Araldi, R. P., Dias Pinto, J. R., & Kerkis, I. (2025). AI-Enhanced Transcriptomic Discovery of Druggable Targets and Repurposed Therapies for Huntington’s Disease. Brain Sciences, 15(8), 865. https://doi.org/10.3390/brainsci15080865

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop