Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
1.
Expert Opin Investig Drugs ; 33(5): 451-467, 2024 May.
Article in English | MEDLINE | ID: mdl-38758356

ABSTRACT

INTRODUCTION: Huntington's Disease (HD) is a genetic neurodegenerative disease for which there is currently no disease-modifying treatment. One of several underlying mechanisms proposed to be involved in HD pathogenesis is inflammation; there is now accumulating evidence that the immune system may play an integral role in disease pathology and progression. As such, modulation of the immune system could be a potential therapeutic target for HD. AREAS COVERED: To date, the number of trials targeting immune aspects of HD has been limited. However, targeting it, may have great advantages over other therapeutic areas, given that many drugs already exist that have actions in this system coupled to the fact that inflammation can be measured both peripherally and, to some extent, centrally using CSF and PET imaging. In this review, we look at evidence that the immune system and the newly emerging area of the microbiome are altered in HD patients, and then present and discuss clinical trials that have targeted different parts of the immune system. EXPERT OPINION: We then conclude by discussing how this field might develop going forward, focusing on the role of imaging and other biomarkers to monitor central immune activation and response to novel treatments in HD.


Subject(s)
Biomarkers , Huntington Disease , Inflammation , Huntington Disease/drug therapy , Huntington Disease/physiopathology , Huntington Disease/immunology , Humans , Animals , Inflammation/drug therapy , Inflammation/immunology , Biomarkers/metabolism , Molecular Targeted Therapy , Disease Progression , Drug Development , Immune System/drug effects , Microbiota
2.
J Neuroimmunol ; 363: 577801, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34973473

ABSTRACT

Huntington's disease (HD) is a late-onset; progressive, dominantly inherited neurological disorder marked by an abnormal expansion of polyglutamine (poly Q) repeats in Huntingtin (HTT) protein. The pathological effects of mutant Huntingtin (mHTT) are not restricted to the nervous system but systemic abnormalities including immune dysregulation have been evidenced in clinical and experimental settings of HD. Indeed, mHTT is ubiquitously expressed and could induce cellular toxicity by directly acting on immune cells. However, it is still unclear if selective expression of mHTT exon1 in neurons could induce immune responses and hemocytes' function. In the present study, we intended to monitor perturbations in the hemocytes' population and their physiological functions in Drosophila, caused by pan-neuronal expression of mHTT protein. A measure of hemocyte count and their physiological activities caused by pan-neuronal expression of mHTT protein highlighted the extent of immune dysregulation occurring with disease progression. We found that pan-neuronal expression of mHTT significantly alters crystal cells and plasmatocyte count in larvae and adults with disease progression. Interestingly, plasmatocytes isolated from diseased conditions exhibit a gradual decline in phagocytic activity ex vivo at progressive stages of the disease as compared to age-matched control groups. In addition, diseased flies displayed elevated reactive oxygen species (ROS) in circulating plasmatocytes at the larval stage and in sessile plasmatocytes of hematopoietic pockets at terminal stages of disease. These findings strongly implicate that neuronal expression of mHTT alone is sufficient to induce non-cell-autonomous immune dysregulation in vivo.


Subject(s)
Hemocytes/immunology , Huntingtin Protein/genetics , Huntington Disease/immunology , Phagocytosis/immunology , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila melanogaster , Humans , Mutation , Neurons/metabolism
3.
J Neurochem ; 160(2): 256-270, 2022 01.
Article in English | MEDLINE | ID: mdl-34665461

ABSTRACT

Huntington´s disease (HD) is a pathological condition that can be studied in mice by the administration of quinolinic acid (QUIN), an agonist of the N-methyl-d-aspartate receptor (NMDAR) that induces NMDAR-mediated cytotoxicity and neuroinflammation. Mast cells (MCs) participate in numerous inflammatory processes through the release of important amounts of histamine (HA). In this study, we aimed to characterize the participation of MCs and HA in the establishment of neural and oxidative damage in the QUIN-induced model of HD. C57BL6/J mice (WT), MC-deficient c-KitW-sh/W-sh (Wsh) mice and Wsh mice reconstituted by intracerebroventricular (i.c.v.) injection of 5 × 105 bone marrow-derived mast cells (BMMCs), or i.c.v. administered with HA (5 µg) were used. All groups of animals were intrastriatally injected with 1 µL QUIN (30 nmol/µL) and 3 days later, apomorphine-induced circling behavior, striatal GABA levels and the number of Fluoro-Jade positive cells, as indicators of neuronal damage, were determined. Also, lipid peroxidation (LP) and reactive oxygen species production (ROS), as markers of oxidative damage, were analyzed. Wsh mice showed less QUIN-induced neuronal and oxidative damage than WT and Wsh-MC reconstituted animals. Histamine administration restored the QUIN-induced neuronal and oxidative damage in the non-reconstituted Wsh mice to levels equivalent or superior to those observed in WT mice. Our results demonstrate that MCs and HA participate in the neuronal and oxidative damages observed in mice subjected to the QUIN -induced model of Huntington's disease.


Subject(s)
Histamine/immunology , Huntington Disease/immunology , Huntington Disease/pathology , Mast Cells/immunology , Neurons/pathology , Animals , Disease Models, Animal , Female , Histamine/metabolism , Huntington Disease/chemically induced , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Quinolinic Acid/toxicity
5.
J Neuroimmunol ; 348: 577380, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32896821

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disease characterized by prominent loss of neurons in the striatum and cortex. Traditionally research in HD has focused on brain changes as they cause progressive motor dysfunction, cognitive decline and psychiatric disorders. The discovery that huntingtin protein (HTT) and its mutated form (mHTT) are expressed not only in the brain but also in different organs and tissues paved the way for the hypothesis that HD might affect regions beyond the central nervous system (CNS). Besides pathological deposition of mHTT, other mechanisms, including inflammation, seem to underlie HD pathogenesis and progression. Altered inflammation can be evidenced even before the onset of classical symptoms of HD. Herein, we will discuss current pre-clinical and clinical evidence on immune/inflammatory changes in peripheral organs during HD development and progression. The understanding of the impact of inflammation on peripheral organs may open new venues for the development of novel therapeutic targets in HD.


Subject(s)
Huntington Disease , Inflammation , Animals , Humans , Huntington Disease/immunology , Huntington Disease/pathology , Inflammation/immunology , Inflammation/pathology
6.
Int J Mol Sci ; 21(17)2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32887270

ABSTRACT

Growing evidence suggests that inflammatory responses, in both the brain and peripheral tissues, contribute to disease pathology in Huntington's disease (HD), an inherited, progressive neurodegenerative disorder typically affecting adults in their 30-40 s. Hence, studies of inflammation-related markers in peripheral fluids might be useful to better characterize disease features. In this study, we measured levels of C-reactive protein (CRP), Interleukin-6 (IL-6), interleukin 1 beta (IL-1B), and alpha-amylase (AA) in saliva and plasma from n = 125 subjects, including n = 37 manifest HD patients, n = 36 premanifest patients, and n = 52 healthy controls, using immunoassays. We found increases in salivary levels of IL-6, IL-1B and CRP across different disease groups and increased levels of IL-6 in the plasma of HD patients as compared to premanifest patients and controls. The levels of salivary IL-6 were significantly correlated with each of the other salivary markers, as well as with IL-6 levels measured in plasma. Further, salivary IL-6 and IL-1B levels were significantly positively correlated with Total Motor Score (TMS) and chorea scores and negatively correlated with Total Functional Capacity (TFC) in HD patients, whereby in healthy control subjects, IL-6 was significantly negatively correlated with Montreal Cognitive Assessment (MoCA) and the Symbol Digit Modalities test (SDM). Interestingly, the plasma levels of IL-6 did not show similar correlations to any clinical measures in either HD or control subjects. These findings suggest that salivary IL-6 is particularly relevant as a potential non-invasive biomarker for HD symptoms. The advent of an effective, dependable salivary biomarker would meet the urgent need for a less invasive means of identifying and monitoring HD disease progression.


Subject(s)
Biomarkers/metabolism , Huntington Disease/pathology , Inflammation/pathology , Interleukin-6/metabolism , Plasma/metabolism , Saliva/metabolism , Adult , Aged , Case-Control Studies , Disease Progression , Female , Humans , Huntington Disease/immunology , Huntington Disease/metabolism , Inflammation/immunology , Inflammation/metabolism , Male , Middle Aged , Young Adult
7.
Neuron ; 107(5): 891-908.e8, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32681824

ABSTRACT

The mechanisms by which mutant huntingtin (mHTT) leads to neuronal cell death in Huntington's disease (HD) are not fully understood. To gain new molecular insights, we used single nuclear RNA sequencing (snRNA-seq) and translating ribosome affinity purification (TRAP) to conduct transcriptomic analyses of caudate/putamen (striatal) cell type-specific gene expression changes in human HD and mouse models of HD. In striatal spiny projection neurons, the most vulnerable cell type in HD, we observe a release of mitochondrial RNA (mtRNA) (a potent mitochondrial-derived innate immunogen) and a concomitant upregulation of innate immune signaling in spiny projection neurons. Further, we observe that the released mtRNAs can directly bind to the innate immune sensor protein kinase R (PKR). We highlight the importance of studying cell type-specific gene expression dysregulation in HD pathogenesis and reveal that the activation of innate immune signaling in the most vulnerable HD neurons provides a novel framework to understand the basis of mHTT toxicity and raises new therapeutic opportunities.


Subject(s)
Huntingtin Protein/immunology , Huntington Disease/immunology , Immunity, Innate/immunology , Neurons/immunology , RNA, Mitochondrial/immunology , Animals , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Mutation , Neurons/pathology , Transcriptome
8.
Neurobiol Dis ; 141: 104943, 2020 07.
Article in English | MEDLINE | ID: mdl-32407769

ABSTRACT

Huntington's disease (HD) is caused by a highly polymorphic CAG trinucleotide expansion in the gene encoding for the huntingtin protein (HTT). The resulting mutant huntingtin protein (mutHTT) is ubiquitously expressed but also exhibits the ability to propagate from cell-to-cell to disseminate pathology; a property which may serve as a new therapeutic focus. Accordingly, we set out to develop a monoclonal antibody (mAB) targeting a particularly exposed region close to the aa586 caspase-6 cleavage site of the HTT protein. This monoclonal antibody, designated C6-17, effectively binds mutHTT and is able to deplete the protein from cell culture supernatants. Using cell-based assays, we demonstrate that extracellular secretion of mutHTT into cell culture media and its subsequent uptake in recipient HeLa cells can be almost entirely blocked by mAB C6-17. Immunohistochemical stainings of post-mortem HD brain tissue confirmed the specificity of mAB C6-17 to human mutHTT aggregates. These findings demonstrate that mAB C6-17 not only successfully engages with its target, mutHTT, but also inhibits cell uptake suggesting that this antibody could interfere with the pathological processes of mutHTT spreading in vivo.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/immunology , Huntington Disease/metabolism , Animals , Biological Transport , Female , HEK293 Cells , HeLa Cells , Humans , Huntington Disease/prevention & control , Mice, Inbred BALB C , Mutation , Protein Aggregation, Pathological/immunology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/prevention & control
9.
J Huntingtons Dis ; 9(2): 115-128, 2020.
Article in English | MEDLINE | ID: mdl-32417788

ABSTRACT

Huntington's disease (HD) is a fatal, inherited neurodegenerative disorder caused by a mutation in the huntingtin gene (HTT). While mutant HTT is present ubiquitously throughout life, HD onset typically occurs in mid-life, suggesting that aging may play an active role in pathogenesis. Cellular aging is defined as the slow decline in stress resistance and accumulation of damage over time. While different cells and tissues can age at different rates, 9 hallmarks of aging have emerged to better define the cellular aging process. Strikingly, many of the hallmarks of aging are also hallmarks of HD pathology. Models of HD and HD patients possess markers of accelerated aging, and processes that decline during aging also decline at a more rapid rate in HD, further implicating the role of aging in HD pathogenesis. Furthermore, accelerating aging in HD mouse and patient-derived neurons unmasks HD-specific phenotypes, suggesting an active role for the aging process in the onset and progression of HD. Here, we review the overlap between the hallmarks of aging and HD and discuss how aging may contribute to pathogenesis in HD.


Subject(s)
Aging, Premature , Aging , Cellular Senescence , Huntington Disease , Aging/genetics , Aging/immunology , Aging/metabolism , Aging, Premature/genetics , Aging, Premature/immunology , Aging, Premature/metabolism , Animals , Cellular Senescence/genetics , Cellular Senescence/immunology , Humans , Huntington Disease/genetics , Huntington Disease/immunology , Huntington Disease/metabolism
10.
Front Immunol ; 11: 337, 2020.
Article in English | MEDLINE | ID: mdl-32161599

ABSTRACT

Modulation of immune activation using immunotherapy has attracted considerable attention for many years as a potential therapeutic intervention for several inflammation-associated neurodegenerative diseases. However, the efficacy of single-target immunotherapy intervention has shown limited or no efficacy in alleviating disease burden and restoring functional capacity. Marked immune system activation and neuroinflammation are important features and prodromal signs in polyQ repeat disorders and α-synucleinopathies. This review describes the current status and future directions of immunotherapies in proteinopathy-induced neurodegeneration with emphasis on preclinical and clinical efficacies of several anti-inflammatory compounds and antibody-based therapies for the treatment of Huntington's disease and α-synucleinopathies. The review concludes with how disease modification and functional restoration could be achieved by using targeted multimodality therapy to target multiple factors.


Subject(s)
Huntington Disease/drug therapy , Huntington Disease/immunology , Immunologic Factors/therapeutic use , Synucleinopathies/drug therapy , Synucleinopathies/immunology , Humans , Immunotherapy , Inflammation , Neurodegenerative Diseases
11.
Front Immunol ; 11: 603594, 2020.
Article in English | MEDLINE | ID: mdl-33679692

ABSTRACT

Emerging evidence indicates that gut dysbiosis may play a regulatory role in the onset and progression of Huntington's disease (HD). However, any alterations in the fecal microbiome of HD patients and its relation to the host cytokine response remain unknown. The present study investigated alterations and host cytokine responses in patients with HD. We enrolled 33 HD patients and 33 sex- and age- matched healthy controls. Fecal microbiota communities were determined through 16S ribosomal DNA gene sequencing, from which we analyzed fecal microbial richness, evenness, structure, and differential abundance of individual taxa between HD patients and healthy controls. HD patients were evaluated for their clinical characteristics, and the relationships of fecal microbiota with these clinical characteristics were analyzed. Plasma concentrations of interferon gamma (IFN-γ), interleukin 1 beta (IL-1ß), IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13, and tumor necrosis factor alpha were measured by Meso Scale Discovery (MSD) assays, and relationships between microbiota and cytokine levels were analyzed in the HD group. HD patients showed increased α-diversity (richness), ß-diversity (structure), and altered relative abundances of several taxa compared to those in healthy controls. HD-associated clinical characteristics correlated with the abundances of components of fecal microbiota at the genus level. Genus Intestinimonas was correlated with total functional capacity scores and IL-4 levels. Our present study also revealed that genus Bilophila were negatively correlated with proinflammatory IL-6 levels. Taken together, our present study represents the first to demonstrate alterations in fecal microbiota and inflammatory cytokine responses in HD patients. Further elucidation of interactions between microbial and host immune responses may help to better understand the pathogenesis of HD.


Subject(s)
Bacteria/immunology , Cytokines/blood , Gastrointestinal Microbiome , Huntington Disease/microbiology , Inflammation Mediators/blood , Intestines/microbiology , Adult , Bacteria/genetics , Case-Control Studies , China , Dysbiosis , Feces/microbiology , Female , Host-Pathogen Interactions , Humans , Huntington Disease/blood , Huntington Disease/diagnosis , Huntington Disease/immunology , Male , Middle Aged , Phylogeny , Ribotyping
12.
Ann Neurol ; 87(2): 246-255, 2020 02.
Article in English | MEDLINE | ID: mdl-31725947

ABSTRACT

OBJECTIVE: Huntington disease (HD) is an autosomal dominantly inherited neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin (HTT) gene. No disease-modifying therapy exists for the treatment of patients with HD. The purpose of this study was therefore to investigate early disease mechanisms that potentially could be used as a target therapeutically. METHODS: Lymphocyte activity in cerebrospinal fluid (CSF) from 4 cohorts of HTT gene expansion carriers (n = 121 in total) and controls was analyzed by techniques based on flow cytometry and enzyme-linked immunosorbent assays. RESULTS: The data of this study provide evidence of immune abnormalities before motor onset of disease. In CSF of HTT gene expansion carriers, we found increased levels of proinflammatory cytokines, including IL-17, and increased consumption of the lymphocyte growth factor IL-7 before motor onset of HD. In concordance, we observed an increased prevalence of IL-17-producing Th17.1 cells in the CSF of HTT gene expansion carriers, predominantly in pre-motor manifest individuals. The frequency of intrathecal Th17.1 cells correlated negatively with progression of HD and the level of neurodegeneration, suggesting a role of Th17.1 cells in the early disease stage. We also observed a skewing in the balance between proinflammatory and regulatory T cells potentially favoring a proinflammatory intrathecal environment in HTT gene expansion carriers. INTERPRETATION: These data suggest that Th17.1 cells are implicated in the earliest pathogenic phases of HD and suggest that treatment to dampen T -cell-driven inflammation before motor onset might be of benefit in HTT gene expansion carriers. ANN NEUROL 2020;87:246-255.


Subject(s)
Huntington Disease/immunology , Huntington Disease/physiopathology , Lymphocyte Activation/immunology , Th17 Cells/immunology , Adult , Aged , Cell Proliferation , Cytokines/cerebrospinal fluid , Cytokines/metabolism , Female , Heterozygote , Humans , Huntingtin Protein/genetics , Huntington Disease/cerebrospinal fluid , Huntington Disease/genetics , Male , Middle Aged , T-Lymphocyte Subsets/immunology , Th17 Cells/metabolism , Trinucleotide Repeat Expansion/genetics
13.
Nat Commun ; 10(1): 4357, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31554807

ABSTRACT

Cell therapy products (CTP) derived from pluripotent stem cells (iPSCs) may constitute a renewable, specifically differentiated source of cells to potentially cure patients with neurodegenerative disorders. However, the immunogenicity of CTP remains a major issue for therapeutic approaches based on transplantation of non-autologous stem cell-derived neural grafts. Despite its considerable side-effects, long-term immunosuppression, appears indispensable to mitigate neuro-inflammation and prevent rejection of allogeneic CTP. Matching iPSC donors' and patients' HLA haplotypes has been proposed as a way to access CTP with enhanced immunological compatibility, ultimately reducing the need for immunosuppression. In the present work, we challenge this paradigm by grafting autologous, MHC-matched and mis-matched neuronal grafts in a primate model of Huntington's disease. Unlike previous reports in unlesioned hosts, we show that in the absence of immunosuppression MHC matching alone is insufficient to grant long-term survival of neuronal grafts in the lesioned brain.


Subject(s)
Graft Rejection/immunology , Huntington Disease/therapy , Induced Pluripotent Stem Cells/transplantation , Major Histocompatibility Complex/immunology , Neurons/transplantation , Animals , Cell Differentiation/immunology , Cytotoxicity, Immunologic/immunology , Disease Models, Animal , Histocompatibility Testing , Humans , Huntington Disease/immunology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/immunology , Neurons/cytology , Neurons/immunology , Primates , Rats, Nude , Transplantation, Autologous
14.
Expert Rev Neurother ; 19(10): 983-995, 2019 10.
Article in English | MEDLINE | ID: mdl-31181964

ABSTRACT

Introduction: Huntington's disease (HD) is an inherited neurodegenerative condition for which there are no disease-modifying treatments. The availability of early genetic diagnosis makes HD an ideal candidate for early intervention. Growing understanding of pathogenesis has led to the identification of new therapeutic targets for which some compounds are now in clinical trials. Areas covered: A detailed review of medical databases and clinical trial registries was performed. Recent clinical trials aimed to establish disease-modification were included. Focus was assigned to RNA and DNA-based therapies aimed at lowering mutant huntingtin (mHTT) including antisense oligonucleotides (ASOs), RNA interference (RNAi), zinc finger proteins (ZFPs) and the CRISPR-Cas9 system. Modulation of mHTT and immunotherapies is also covered. Expert opinion: Targeting HD pathogenesis at its most proximal level is under intense investigation. ASOs are the only HTT-lowering strategy in clinical trials of manifest HD. Safety and efficacy of an allele specific vs. allele non-specific approach has yet to be established. Success will extend to premanifest carriers for which development of clinical and imaging biomarkers will be necessary. Scientific and technological advancement will bolster new methods of treatment delivery. Cumulative experience, collaborative research, and platforms such as ENROLL-HD will facilitate efficient and effective clinical trials.


Subject(s)
Huntington Disease , Humans , Huntington Disease/drug therapy , Huntington Disease/genetics , Huntington Disease/immunology
15.
Nucleic Acid Ther ; 29(5): 256-265, 2019 10.
Article in English | MEDLINE | ID: mdl-31184975

ABSTRACT

Huntington's disease is a neurodegenerative disorder caused by a CAG repeat expansion in the first exon of huntingtin gene (HTT) encoding for a toxic polyglutamine protein. This disease is characterized by motor, psychiatric, and cognitive impairments. Currently, there is no disease modifying treatment. However, reducing the expression of the huntingtin protein (HTT) using antisense oligonucleotides (ASOs) has been shown as a promising therapeutic strategy. In this study, we explore the therapeutic potential of ASO made of tricyclo-DNA (tcDNA), a conformationally constrained DNA analog, to silence HTT. We used a gapmer ASO, containing central DNA nucleotides flanked by tcDNA modifications on 5' and 3' ends, allowing the recruitment of RNAse H and subsequent degradation of the messenger RNA. After transfection of tcDNA-ASO in patient-derived fibroblast cell lines, we show a strong decrease of HTT mRNA and protein levels. As a control, 2'O-methyl-RNA targeting the same region of HTT was also tested and did not induce a significant effect. tcDNA-ASO were also evaluated in vivo in the YAC128 mice, containing the full-length human HTT gene with 128 CAG repeat expansion. Single intracerebroventricular (ICV) injections of tcDNA induce a significant decrease of HTT messenger and protein levels in the cortex, hippocampus, striatum, and cerebellum of treated mice. tcDNA-ASO were found well distributed in the central nervous system (CNS) and show long lasting effect with protein levels still low, 12 weeks after a single ICV injection. This proof of concept study suggests the therapeutic potential of gapmer tcDNA ASO to downregulate huntingtin in vitro and in vivo.


Subject(s)
Huntingtin Protein/genetics , Huntington Disease/therapy , Mutant Proteins/genetics , Oligonucleotides, Antisense/pharmacology , Animals , DNA, Antisense/pharmacology , Disease Models, Animal , Exons/genetics , Humans , Huntingtin Protein/antagonists & inhibitors , Huntington Disease/genetics , Huntington Disease/immunology , Huntington Disease/pathology , Mice , Mutant Proteins/antagonists & inhibitors , Oligonucleotides, Antisense/genetics , Ribonuclease H/genetics , Trinucleotide Repeat Expansion/genetics
16.
Neurosci Biobehav Rev ; 102: 56-84, 2019 07.
Article in English | MEDLINE | ID: mdl-30995512

ABSTRACT

Major depressive disorder (MDD) is a highly prevalent psychiatric disorder and a major cause of disability worldwide. This neurological condition is commonly associated with neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD), and has a significant impact on the increasing burden of these neuropathologies. Over the past decades, some of the pathophysiological and molecular mechanisms that contribute to these diseases have been elucidated and these findings indicate that, despite presenting distinct features, there are several similarities between the neurobiological alterations that lead to MDD and neurodegeneration in AD, PD, and HD. For instance, disturbances in monoaminergic transmission and the hypothalamic-pituitary-adrenal (HPA) axis, increased oxidative and neuroinflammatory events, and impaired trophic support are thought to contribute to neuronal atrophy and death in all these diseases. In addition, neuroimaging findings have helped elucidate the structural and functional changes implicated in the relationship between depression and neurodegeneration, thus establishing a neuroanatomical signature to explain, at least in part, the comorbidity between MDD and AD, PD, and HD. The present review summarizes these findings and the current evidence regarding the effectiveness of common antidepressant therapies for the treatment of MDD in patients with these neurodegenerative diseases. This population is particularly vulnerable to the drawdowns of conventional antidepressant therapy (namely inadequate response and high risk of side effects), and the development of emerging therapeutic approaches to treat MDD in patients with AD, PD, and HD is thus of paramount importance to improve the quality of life of these individuals.


Subject(s)
Alzheimer Disease , Comorbidity , Depressive Disorder, Major , Huntington Disease , Parkinson Disease , Alzheimer Disease/epidemiology , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Depressive Disorder, Major/epidemiology , Depressive Disorder, Major/immunology , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/physiopathology , Humans , Huntington Disease/epidemiology , Huntington Disease/immunology , Huntington Disease/metabolism , Huntington Disease/physiopathology , Parkinson Disease/epidemiology , Parkinson Disease/immunology , Parkinson Disease/metabolism , Parkinson Disease/physiopathology
17.
Mediators Inflamm ; 2019: 3540974, 2019.
Article in English | MEDLINE | ID: mdl-30766446

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder which is caused by a mutation of the huntingtin (HTT) gene. Although the pathogenesis of HD has been associated with inflammatory responses, if and how the immune system contributes to the onset of HD is largely unknown. Invariant natural killer T (iNKT) cells are a group of innate-like regulatory T lymphocytes that can rapidly produce various cytokines such as IFNγ and IL4 upon stimulation with the glycolipid α-galactosylceramide (α-GalCer). By employing both R6/2 Tg mice (murine HD model) and Jα18 KO mice (deficient in iNKT cells), we investigated whether alterations of iNKT cells affect the development of HD in R6/2 Tg mice. We found that Jα18 KO R6/2 Tg mice showed disease progression comparable to R6/2 Tg mice, indicating that the absence of iNKT cells did not have any significant effects on HD development. However, repeated activation of iNKT cells with α-GalCer facilitated HD progression in R6/2 Tg mice, and this was associated with increased infiltration of iNKT cells in the brain. Taken together, our results demonstrate that repeated α-GalCer treatment of R6/2 Tg mice accelerates HD progression, suggesting that immune activation can affect the severity of HD pathogenesis.


Subject(s)
Huntington Disease/immunology , Lymphocyte Activation , Natural Killer T-Cells/immunology , Animals , Brain/metabolism , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Galactosylceramides/chemistry , Genotype , Leukocytes/metabolism , Mice , Mice, Knockout
18.
Mol Psychiatry ; 24(3): 364-377, 2019 03.
Article in English | MEDLINE | ID: mdl-29487401

ABSTRACT

There is compelling evidence that the pathophysiology of many neurodegenerative diseases includes dysregulation of the immune system, with some elements that precede disease onset. However, if these alterations are prominent, why have clinical trials targeting this system failed to translate into long-lasting meaningful benefits for patients? This review focuses on Huntington's disease, a genetic disorder marked by notable cerebral and peripheral inflammation. We summarize ongoing and completed clinical trials that have involved pharmacological approaches to inhibit various components of the immune system and their pre-clinical correlates. We then discuss new putative treatment strategies using more targeted immunotherapies such as vaccination and intrabodies and how these may offer new hope in the treatment of Huntington's disease as well as other neurodegenerative diseases.


Subject(s)
Huntington Disease/immunology , Huntington Disease/therapy , Immunotherapy/methods , Humans , Huntington Disease/genetics
19.
Int Rev Neurobiol ; 142: 289-333, 2018.
Article in English | MEDLINE | ID: mdl-30409256

ABSTRACT

Huntington's disease (HD) is a rare monogenic neurodegenerative disorder caused by a trinucleotide CAG repeat expansion in the huntingtin gene resulting in the formation of intranuclear inclusions of mutated huntingtin. The accumulation of mutated huntingtin leads to loss of GABAergic medium spiny neurons (MSNs); subsequently resulting in the development of chorea, cognitive dysfunction and psychiatric symptoms. Premanifest HD gene expansion carriers, provide a unique cohort to examine very early molecular changes, occurring before the development of overt symptoms, to elucidate disease pathophysiology and identify reliable biomarkers of HD progression. Positron emission tomography (PET) is a non-invasive molecular imaging technique allowing the evaluation of specific molecular targets in vivo. Selective PET radioligands provide invaluable tools to investigate the role of the dopaminergic system, brain metabolism, microglial activation, phosphodiesterase 10A, and cannabinoid, GABA, adenosine and opioid receptors in HD. PET has been employed to monitor disease progression aiming to identify a reliable biomarker to predict phenoconversion from premanifest to manifest HD.


Subject(s)
Huntington Disease/diagnostic imaging , Huntington Disease/metabolism , Molecular Imaging/methods , Positron-Emission Tomography/methods , Humans , Huntington Disease/immunology
20.
Biomed Res Int ; 2018: 1012789, 2018.
Article in English | MEDLINE | ID: mdl-30345294

ABSTRACT

Autophagy begins with the nucleation of phagophores, which then expand to give rise to the double-membrane autophagosomes. Autophagosomes ultimately fuse with lysosomes, where the cytosolic cargoes are degraded. Accumulation of autophagosomes is a hallmark of autophagy and neurodegenerative disorders including Alzheimer's and Huntington's disease. In recent years, the sources of autophagosome membrane have attracted a great deal of interests, even so, the membrane donors for autophagosomes are still under debate. In this review, we describe the probable sources of autophagosome membrane.


Subject(s)
Alzheimer Disease/immunology , Autophagosomes/immunology , Autophagy/immunology , Huntington Disease/immunology , Lysosomes/immunology , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL