Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Neurobiol Dis ; 153: 105318, 2021 06.
Article in English | MEDLINE | ID: mdl-33636386

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder characterized by accumulation of mutant huntingtin protein and significant loss of neurons in striatum and cortex. Along with motor difficulties, the HD patients also manifest anxiety and loss of cognition. Unfortunately, the clinically approved drugs only offer symptomatic relief and are not free from side effects. This study underlines the importance of glyceryl tribenzoate (GTB), an FDA-approved food flavoring ingredient, in alleviating HD pathology in transgenic N171-82Q mouse model. Oral administration of GTB significantly reduced mutant huntingtin level in striatum, motor cortex as well as hippocampus and increased the integrity of viable neurons. Furthermore, we found the presence of sodium benzoate (NaB), a FDA-approved drug for urea cycle disorders and glycine encephalopathy, in the brain of GTB-fed HD mice. Accordingly, NaB administration also markedly decreased huntingtin level in striatum and cortex. Glial activation is found to coincide with neuronal death in affected regions of HD brains. Interestingly, both GTB and NaB treatment suppressed activation of glial cells and inflammation in the brain. Finally, neuroprotective effect of GTB and NaB resulted in improved motor performance of HD mice. Collectively, these results suggest that GTB and NaB may be repurposed for HD.


Subject(s)
Benzoates/administration & dosage , Flavoring Agents/pharmacology , Food Preservatives/pharmacology , Huntingtin Protein/drug effects , Huntington Disease/metabolism , Motor Cortex/drug effects , Neostriatum/drug effects , Sodium Benzoate/pharmacology , Administration, Oral , Animals , Benzoates/pharmacology , Benzoic Acid/pharmacology , Gait Analysis , Hand Strength , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/physiopathology , Mice , Mice, Transgenic , Motor Cortex/metabolism , Neostriatum/metabolism , Open Field Test , Rotarod Performance Test , Sodium Benzoate/metabolism
2.
Brain ; 143(2): 407-429, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31738395

ABSTRACT

Polyglutamine (polyQ) disorders are a group of nine neurodegenerative diseases that share a common genetic cause, which is an expansion of CAG repeats in the coding region of the causative genes that are otherwise unrelated. The trinucleotide expansion encodes for an expanded polyQ tract in the respective proteins, resulting in toxic gain-of-function and eventually in neurodegeneration. Currently, no disease-modifying therapies are available for this group of disorders. Nevertheless, given their monogenic nature, polyQ disorders are ideal candidates for therapies that target specifically the gene transcripts. Antisense oligonucleotides (ASOs) have been under intense investigation over recent years as gene silencing tools. ASOs are small synthetic single-stranded chains of nucleic acids that target specific RNA transcripts through several mechanisms. ASOs can reduce the levels of mutant proteins by breaking down the targeted transcript, inhibit mRNA translation or alter the maturation of the pre-mRNA via splicing correction. Over the years, chemical optimization of ASO molecules has allowed significant improvement of their pharmacological properties, which has in turn made this class of therapeutics a very promising strategy to treat a variety of neurodegenerative diseases. Indeed, preclinical and clinical strategies have been developed in recent years for some polyQ disorders using ASO therapeutics. The success of ASOs in several animal models, as well as encouraging results in the clinic for Huntington's disease, points towards a promising future regarding the application of ASO-based therapies for polyQ disorders in humans, offering new opportunities to address unmet medical needs for this class of disorders. This review aims to present a brief overview of key chemical modifications, mechanisms of action and routes of administration that have been described for ASO-based therapies. Moreover, it presents a review of the most recent and relevant preclinical and clinical trials that have tested ASO therapeutics in polyQ disorders.


Subject(s)
Huntingtin Protein/drug effects , Huntington Disease/drug therapy , Neurodegenerative Diseases/drug therapy , Oligonucleotides, Antisense/pharmacology , Peptides/genetics , Animals , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Neurodegenerative Diseases/genetics , Trinucleotide Repeat Expansion/genetics
3.
Brain ; 143(1): 266-288, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31848580

ABSTRACT

Huntington's disease is associated with a reactive microglial response and consequent inflammation. To address the role of these cells in disease pathogenesis, we depleted microglia from R6/2 mice, a rapidly progressing model of Huntington's disease marked by behavioural impairment, mutant huntingtin (mHTT) accumulation, and early death, through colony-stimulating factor 1 receptor inhibition (CSF1Ri) with pexidartinib (PLX3397) for the duration of disease. Although we observed an interferon gene signature in addition to downregulated neuritogenic and synaptic gene pathways with disease, overt inflammation was not evident by microglial morphology or cytokine transcript levels in R6/2 mice. Nonetheless, CSF1Ri-induced microglial elimination reduced or prevented disease-related grip strength and object recognition deficits, mHTT accumulation, astrogliosis, and striatal volume loss, the latter of which was not associated with reductions in cell number but with the extracellular accumulation of chondroitin sulphate proteoglycans (CSPGs)-a primary component of glial scars. A concurrent loss of proteoglycan-containing perineuronal nets was also evident in R6/2 mice, and microglial elimination not only prevented this but also strikingly increased perineuronal nets in the brains of naïve littermates, suggesting a new role for microglia as homeostatic regulators of perineuronal net formation and integrity.


Subject(s)
Aminopyridines/pharmacology , Extracellular Matrix/drug effects , Huntingtin Protein/drug effects , Huntington Disease/metabolism , Microglia/drug effects , Neostriatum/drug effects , Pyrroles/pharmacology , Recognition, Psychology/drug effects , Animals , Astrocytes/drug effects , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Cytokines/drug effects , Cytokines/genetics , Disease Models, Animal , Down-Regulation , Extracellular Matrix/metabolism , Hand Strength , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/pathology , Huntington Disease/physiopathology , Inflammation , Mice , Mice, Transgenic , Neostriatum/pathology , Neurites/drug effects , RNA, Messenger/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Synapses/drug effects , Transcriptome
4.
J Biol Chem ; 294(6): 1915-1923, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30538129

ABSTRACT

Huntington's disease (HD) is a neurodegenerative, age-onset disorder caused by a CAG DNA expansion in exon 1 of the HTT gene, resulting in a polyglutamine expansion in the huntingtin protein. Nuclear accumulation of mutant huntingtin is a hallmark of HD, resulting in elevated mutant huntingtin levels in cell nuclei. Huntingtin is normally retained at the endoplasmic reticulum via its N17 amphipathic α-helix domain but is released by oxidation of Met-8 during reactive oxygen species (ROS) stress. Huntingtin enters the nucleus via an importin ß1- and 2-dependent proline-tyrosine nuclear localization signal (PY-NLS), which has a unique intervening sequence in huntingtin. Here, we have identified the high-mobility group box 1 (HMGB1) protein as an interactor of the intervening sequence within the PY-NLS. Nuclear levels of HMGB1 positively correlated with varying levels of nuclear huntingtin in both HD and normal human fibroblasts. We also found that HMGB1 interacts with the huntingtin N17 region and that this interaction is enhanced by the presence of ROS and phosphorylation of critical serine residues in the N17 region. We conclude that HMGB1 is a huntingtin N17/PY-NLS ROS-dependent interactor, and this protein bridging is essential for relaying ROS sensing by huntingtin to its nuclear entry during ROS stress. ROS may therefore be a critical age-onset stress that triggers nuclear accumulation of mutant huntington in Huntington's disease.


Subject(s)
Active Transport, Cell Nucleus , HMGB1 Protein/physiology , Huntingtin Protein/metabolism , Reactive Oxygen Species/pharmacology , Binding Sites , Cells, Cultured , Fibroblasts/metabolism , Humans , Huntingtin Protein/drug effects , Huntingtin Protein/physiology , Nuclear Localization Signals , Nuclear Proteins/metabolism , Phosphorylation , Protein Binding
5.
Metab Brain Dis ; 34(3): 715-720, 2019 06.
Article in English | MEDLINE | ID: mdl-30850940

ABSTRACT

Mutations in the HTT gene, consisting of expansion of CAG triplets, cause the Huntington's disease (HD), one of the major neurodegenerative disorders. Formation of aggregates of mutant huntingtin (mHTT, the product of the mutant HTT gene) leads to cellular dysfunctions, and subsequent neurodegeneration which manifest clinically as motor abnormalities and cognitive deficits. We recently used immortalized HEK-293 cells expressing the 1st exon of the mutant HTT gene as a cellular model of HD, and showed that the stimulation of autophagy by genistein corrected the mutant phenotype. However, effects of genistein on HD patient-derived cells remained unknown. In this report, we demonstrated that genistein also instigated degradation of mHTT in fibroblasts derived from HD patients. This was assessed as a significant decrease in the levels of HTT in HD fibroblasts measured by Western-blotting, and the disappearance of intracellular mHTT aggregates in cells observed by fluorescent microscopy. Fibroblasts derived from control persons were not affected by genistein treatment. These results indicate that genistein can improve HD phenotype in patient-derived cells, and substantiates the need for further studies of this isoflavone as a potential therapeutic agent.


Subject(s)
Autophagy/drug effects , Fibroblasts/drug effects , Genistein/pharmacology , Huntington Disease/chemically induced , Adult , Animals , Disease Models, Animal , Female , Fibroblasts/metabolism , Humans , Huntingtin Protein/drug effects , Huntington Disease/metabolism , Male , Middle Aged , Mutation/drug effects , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/chemically induced
6.
Curr Neurol Neurosci Rep ; 17(2): 18, 2017 02.
Article in English | MEDLINE | ID: mdl-28265888

ABSTRACT

Huntington disease (HD) is an autosomal dominant neurodegenerative condition caused by a CAG trinucleotide expansion in the huntingtin gene. At present, the HD field is experiencing exciting times with the assessment for the first time in human subjects of interventions aimed at core disease mechanisms. Out of a portfolio of interventions that claim a potential disease-modifying effect in HD, the target huntingtin has more robust validation. In this review, we discuss the spectrum of huntingtin-lowering therapies that are currently being considered. We provide a critical appraisal of the validation of huntingtin as a drug target, describing the advantages, challenges, and limitations of the proposed therapeutic interventions. The development of these new therapies relies strongly on the knowledge of HD pathogenesis and the ability to translate this knowledge into validated pharmacodynamic biomarkers. Altogether, the goal is to support a rational drug development that is ethical and cost-effective. Among the pharmacodynamic biomarkers under development, the quantification of mutant huntingtin in the cerebral spinal fluid and PET imaging targeting huntingtin or phosphodiesterase 10A deserve special attention. Huntingtin-lowering therapeutics are eagerly awaited as the first interventions that may be able to change the course of HD in a meaningful way.


Subject(s)
Huntingtin Protein/drug effects , Huntington Disease/genetics , Molecular Targeted Therapy/methods , Animals , Humans , Huntingtin Protein/cerebrospinal fluid , Huntington Disease/diagnostic imaging , Neuroimaging , Phosphoric Diester Hydrolases/metabolism , Positron-Emission Tomography
7.
J Huntingtons Dis ; 10(1): 203-220, 2021.
Article in English | MEDLINE | ID: mdl-32925081

ABSTRACT

DNA damage repair (DDR) mechanisms have been implicated in a number of neurodegenerative diseases (both genetically determined and sporadic). Consistent with this, recent genome-wide association studies in Huntington's disease (HD) and other trinucleotide repeat expansion diseases have highlighted genes involved in DDR mechanisms as modifiers for age of onset, rate of progression and somatic instability. At least some clinical genetic modifiers have been shown to have a role in modulating trinucleotide repeat expansion biology and could therefore provide new disease-modifying therapeutic targets. In this review, we focus on key considerations with respect to drug discovery and development using DDR mechanisms as a target for trinucleotide repeat expansion diseases. Six areas are covered with specific reference to DDR and HD: 1) Target identification and validation; 2) Candidate selection including therapeutic modality and delivery; 3) Target drug exposure with particular focus on blood-brain barrier penetration, engagement and expression of pharmacology; 4) Safety; 5) Preclinical models as predictors of therapeutic efficacy; 6) Clinical outcome measures including biomarkers.


Subject(s)
DNA Damage/genetics , DNA Mismatch Repair/genetics , Drug Development , Drug Discovery , Huntingtin Protein/genetics , Huntington Disease/drug therapy , Huntington Disease/genetics , Trinucleotide Repeat Expansion/genetics , Animals , DNA Damage/drug effects , DNA Mismatch Repair/drug effects , Humans , Huntingtin Protein/drug effects , Trinucleotide Repeat Expansion/drug effects
8.
Food Funct ; 11(2): 1334-1348, 2020 Feb 26.
Article in English | MEDLINE | ID: mdl-32043503

ABSTRACT

Huntington's disease (HD) is a genetic neurodegenerative disorder caused by a highly polymorphic CAG trinucleotide repeat expansion encoding an extended polyglutamine (polyQ) tract at the N-terminus of huntingtin protein (HTT). The polyQ tract promotes the formation of toxic oligomers and aggregates of HTT, which leads to neuronal dysfunction and death. Therapies to lower mutant HTT (mHTT) and its aggregates appear to be the most promising strategies. Ellagic acid (EA) has been marketed as a dietary supplement with various claimed benefits and neuroprotective effects on several neurodegenerative disorders, while its effect on mHTT pathology is still unknown. Here we reported that EA significantly attenuated motor and cognitive deficits in R6/2 mice. Moreover, EA significantly lowered mHTT levels, reduced neuroinflammation, rescued synapse loss, and decreased oxidative stress in R6/2 mouse brains. These findings indicated that EA has promising therapeutic potential for HD treatment.


Subject(s)
Cognitive Dysfunction/drug therapy , Ellagic Acid/pharmacology , Huntingtin Protein/drug effects , Huntington Disease/drug therapy , Neuroprotective Agents/pharmacology , Animals , Cognitive Dysfunction/metabolism , Disease Models, Animal , Female , Huntingtin Protein/genetics , Huntington Disease/metabolism , Mice , Mice, Transgenic , Motor Activity/drug effects
9.
Neuropharmacology ; 162: 107812, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31622602

ABSTRACT

One of the pathological hallmarks of Huntington disease (HD) is accumulation of the disease-causing mutant huntingtin (mHTT), which leads to the disruption of a variety of cellular functions, ultimately resulting in cell death. Induction of autophagy, for example by the inhibition of mechanistic target of rapamycin (mTOR) signaling, has been shown to reduce HTT levels and aggregates. While rapalogs like rapamycin allosterically inhibit the mTOR complex 1 (TORC1), ATP-competitive mTOR inhibitors suppress activities of TORC1 and TORC2 and have been shown to be more efficient in inducing autophagy and reducing protein levels and aggregates than rapalogs. The ability to cross the blood-brain barrier of first generation catalytic mTOR inhibitors has so far been limited, and therefore sufficient target coverage in the brain could not be reached. Two novel, brain penetrant compounds - the mTORC1/2 inhibitor PQR620, and the dual pan-phosphoinositide 3-kinase (PI3K) and mTORC1/2 kinase inhibitor PQR530 - were evaluated by assessing their potential to induce autophagy and reducing mHTT levels. For this purpose, expression levels of autophagic markers and well-defined mTOR targets were analyzed in STHdh cells and HEK293T cells and in mouse brains. Both compounds potently inhibited mTOR signaling in cell models as well as in mouse brain. As proof of principle, reduction of aggregates and levels of soluble mHTT were demonstrated upon treatment with both compounds. Originally developed for cancer treatment, these second generation mTORC1/2 and PI3K/mTOR inhibitors show brain penetrance and efficacy in cell models of HD, making them candidate molecules for further investigations in HD.


Subject(s)
Azabicyclo Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Huntingtin Protein/drug effects , Huntington Disease/metabolism , Morpholines/pharmacology , Neurons/drug effects , Protein Aggregates/drug effects , Pyridines/pharmacology , Triazines/pharmacology , Animals , Autophagy/drug effects , Blood-Brain Barrier , Cell Line , Corpus Striatum/cytology , HEK293 Cells , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mice , Neurons/metabolism , Phosphatidylinositol 3-Kinases , Phosphoinositide-3 Kinase Inhibitors/pharmacology
10.
Parkinsonism Relat Disord ; 59: 125-130, 2019 02.
Article in English | MEDLINE | ID: mdl-30616867

ABSTRACT

INTRODUCTION: Huntington disease (HD) is a rare genetic neurodegenerative condition. The availability of a genetic diagnosis makes HD an attractive model for the development of therapies that can delay or, at best, halt the progression of neurodegenerative conditions. Tetrabenazine and deutetrabenazine are the only treatment options with a formal indication (chorea) for this patient population. METHODS: Literature review on HD and clinical trials using the medical databases Pubmed, Web of Science, and clinical trial registries. Recent clinical trials conducted with the goal of disease-modification or new symptomatic treatment indications were included. Non-pharmacological interventions were excluded. RESULTS: Therapeutic approaches aiming at disease-modification include huntingtin-lowering strategies, the modulation of huntingtin homeostasis and neuroinflammation. Huntingtin-lowering strategies are of particular interest by targeting the mRNA of the huntingtin (HTT) gene at the core of HD biology. Antisense oligonucleotides (ASO) are the only huntingtin-lowering strategies in clinical development. The initial results suggest that the first non-allele specific ASO was safe and associated with a reduction in the levels of mutated huntingtin protein (mHTT). Other clinical trials for disease-modification in HD have generated negative results or are ongoing. Assays to measure CSF mHTT and brain nuclear imaging specific to HD can support the rational development of these therapies. Novel symptomatic treatment indications explored in clinical trials include motor disability, irritability and apathy. CONCLUSIONS: The years ahead are promising for novel and revolutionary therapies aimed at core disease mechanisms in HD. Clinical research platforms such as Enroll-HD are expected to potentiate the conduction of clinical trials in HD.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/therapeutic use , Dopamine Agents/therapeutic use , Huntingtin Protein/drug effects , Huntington Disease/drug therapy , Huntington Disease/physiopathology , Immunotherapy , Oligonucleotides, Antisense/therapeutic use , Phosphodiesterase Inhibitors/therapeutic use , Humans
11.
J Huntingtons Dis ; 8(1): 9-22, 2019.
Article in English | MEDLINE | ID: mdl-30636742

ABSTRACT

To date, no candidate intervention has demonstrated a disease-modifying effect in Huntington's disease, despite promising results in preclinical studies. In this commentary we discuss disease-modifying therapies that have been trialled in Huntington's disease and speculate that these failures may be attributed, in part, to the assumption that a single drug selectively targeting one aspect of disease pathology will be universally effective, regardless of disease stage or "subtype". We therefore propose an alternative approach for effective disease-modification that uses 1) a combination approach rather than monotherapy, and 2) targets the disease process early on - before it is clinically manifest. Finally, we will consider whether this change in approach that we propose will be relevant in the future given the recent shift to targeting more proximal disease processes-e.g., huntingtin gene expression; a timely question given Roche's recent decision to take on the clinical development of a promising new drug candidate in Huntington's disease, IONIS-HTTRx.


Subject(s)
Gene Expression , Huntingtin Protein/drug effects , Huntington Disease/drug therapy , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Treatment Outcome
12.
Neurosci Bull ; 35(6): 1024-1034, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31432317

ABSTRACT

Huntington's disease (HD) is a deadly neurodegenerative disease with abnormal expansion of CAG repeats in the huntingtin gene. Mutant Huntingtin protein (mHTT) forms abnormal aggregates and intranuclear inclusions in specific neurons, resulting in cell death. Here, we tested the ability of a natural heat-shock protein 90 inhibitor, Gedunin, to degrade transfected mHTT in Neuro-2a cells and endogenous mHTT aggregates and intranuclear inclusions in both fibroblasts from HD patients and neurons derived from induced pluripotent stem cells from patients. Our data showed that Gedunin treatment degraded transfected mHTT in Neuro-2a cells, endogenous mHTT aggregates and intranuclear inclusions in fibroblasts from HD patients, and in neurons derived from induced pluripotent stem cells from patients in a dose- and time-dependent manner, and its activity depended on the proteasomal pathway rather than the autophagy route. These findings also showed that although Gedunin degraded abnormal mHTT aggregates and intranuclear inclusions in cells from HD patient, it did not affect normal cells, thus providing a new perspective for using Gedunin to treat HD.


Subject(s)
Huntingtin Protein/drug effects , Huntington Disease/drug therapy , Intranuclear Inclusion Bodies/drug effects , Limonins/pharmacology , Mutant Proteins/drug effects , Protein Aggregates/drug effects , Animals , Cell Culture Techniques , Fibroblasts/drug effects , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/metabolism , Humans , Huntingtin Protein/genetics , Induced Pluripotent Stem Cells/drug effects , Leupeptins/pharmacology , Mice , Mutation , Neurons/drug effects , Proteasome Endopeptidase Complex , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL