Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
PLoS Biol ; 20(3): e3001578, 2022 03.
Article in English | MEDLINE | ID: mdl-35263320

ABSTRACT

Neurodegenerative disorders refer to a group of diseases commonly associated with abnormal protein accumulation and aggregation in the central nervous system. However, the exact role of protein aggregation in the pathophysiology of these disorders remains unclear. This gap in knowledge is due to the lack of experimental models that allow for the spatiotemporal control of protein aggregation, and the investigation of early dynamic events associated with inclusion formation. Here, we report on the development of a light-inducible protein aggregation (LIPA) system that enables spatiotemporal control of α-synuclein (α-syn) aggregation into insoluble deposits called Lewy bodies (LBs), the pathological hallmark of Parkinson disease (PD) and other proteinopathies. We demonstrate that LIPA-α-syn inclusions mimic key biochemical, biophysical, and ultrastructural features of authentic LBs observed in PD-diseased brains. In vivo, LIPA-α-syn aggregates compromise nigrostriatal transmission, induce neurodegeneration and PD-like motor impairments. Collectively, our findings provide a new tool for the generation, visualization, and dissection of the role of α-syn aggregation in PD.


Subject(s)
Parkinson Disease , alpha-Synuclein , Cluster Analysis , Humans , Lewy Bodies/metabolism , Lewy Bodies/pathology , Parkinson Disease/metabolism , Protein Aggregates , alpha-Synuclein/metabolism
2.
Cell Mol Life Sci ; 80(2): 45, 2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36651994

ABSTRACT

Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder resulting from a CAG expansion in the huntingtin (HTT) gene, which leads to the production and accumulation of mutant huntingtin (mHTT). While primarily considered a disorder of the central nervous system, multiple changes have been described to occur throughout the body, including activation of the immune system. In other neurodegenerative disorders, activation of the immune system has been shown to include the production of antibodies against disease-associated pathological proteins. However, the existence of mHTT-targeted antibodies has never been reported. In this study, we assessed the presence and titer of antibodies recognizing HTT/mHTT in patients with HD (n = 66) and age- and gender-matched healthy controls (n = 66) using a combination of Western blotting and ELISA. Together, these analyses revealed that antibodies capable of recognizing HTT/mHTT were detectable in the plasma samples of all participants, including healthy controls. When antibody levels were monitored at different disease stages, it was observed that antibodies against full-length mHTT were highest in patients with severe disease while antibodies against HTTExon1 were elevated in patients with mild disease. Combined, these results suggest that antibodies detecting different forms of mHTT peak at different disease stages.


Subject(s)
Huntington Disease , Neurodegenerative Diseases , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Antibodies
3.
Neurobiol Dis ; 180: 106091, 2023 05.
Article in English | MEDLINE | ID: mdl-36967065

ABSTRACT

In a previous study, we have shown that parabiotic coupling of a knock-in mouse model (zQ175) of Huntington's disease (HD) to wild-type (WT) littermates resulted in a worsening of the normal phenotype as seen by detection of mutant huntingtin protein (mHTT) aggregates within peripheral organs and the cerebral cortex as well as vascular abnormalities in WT mice. In contrast, parabiosis improved disease features in the zQ175 mice such as reduction of mHTT aggregate number in the liver and cortex, decrease in blood-brain barrier (BBB) permeability and attenuation of mitochondrial impairments. While the shared circulation mediated these effects, no specific factor was identified. To better understand which blood elements were involved in the aforementioned changes, WT and zQ175 mice underwent parabiotic surgery prior to exposing one of the paired animals to irradiation. The irradiation procedure successfully eliminated the hematopoietic niche followed by repopulation with cells originating from the non-irradiated parabiont, as measured by the quantification of mHTT levels in peripheral blood mononuclear cells. Although irradiation of the WT parabiont, causing the loss of healthy hematopoietic cells, did lead to a few alterations in mitochondrial function in the muscle (TOM40 levels), and increased neuroinflammation in the striatum (GFAP levels), most of the changes observed were likely attributable to the irradiation procedure itself (e.g. mHTT aggregates in cortex and liver; cellular stress in peripheral organs). However, factors such as mHTT aggregation in the brain and periphery, and BBB leakage, which were improved in zQ175 mice when paired to WT littermates in the previous parabiosis experiment, were unaffected by perturbation of the hematopoietic niche. It would therefore appear that cells of the hematopoietic stem cell niche are largely uninvolved in the beneficial effects of parabiosis.


Subject(s)
Huntington Disease , Mice , Animals , Mice, Transgenic , Huntington Disease/genetics , Leukocytes, Mononuclear/metabolism , Disease Models, Animal , Phenotype , Huntingtin Protein/genetics , Huntingtin Protein/metabolism
4.
Mol Psychiatry ; 27(1): 269-280, 2022 01.
Article in English | MEDLINE | ID: mdl-34711942

ABSTRACT

If theories postulating that pathological proteins associated with neurodegenerative disorders behave similarly to prions were initially viewed with reluctance, it is now well-accepted that this occurs in several disease contexts. Notably, it has been reported that protein misfolding and subsequent prion-like properties can actively participate in neurodegenerative disorders. While this has been demonstrated in multiple cellular and animal model systems related to Alzheimer's and Parkinson's diseases, the prion-like properties of the mutant huntingtin protein (mHTT), associated with Huntington's disease (HD), have only recently been considered to play a role in this pathology, a concept our research group has contributed to extensively. In this review, we summarize the last few years of in vivo research in the field and speculate on the relationship between prion-like events and human HD. By interpreting observations primarily collected in in vivo models, our discussion will aim to discriminate which experimental factors contribute to the most efficient types of prion-like activities of mHTT and which routes of propagation may be more relevant to the human condition. A look back at nearly a decade of experimentation will inform future research and whether therapeutic strategies may emerge from this new knowledge.


Subject(s)
Huntington Disease , Neurodegenerative Diseases , Prions , Animals , Disease Models, Animal , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Neurodegenerative Diseases/metabolism , Prions/genetics , Prions/metabolism
5.
Mol Ther ; 30(4): 1500-1522, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35051614

ABSTRACT

Huntington's disease is classically described as a neurodegenerative disorder of monogenic aetiology. The disease is characterized by an abnormal polyglutamine expansion in the huntingtin gene, which drives the toxicity of the mutated form of the protein. However, accumulation of the microtubule-associated protein tau, which is involved in a number of neurological disorders, has also been observed in patients with Huntington's disease. In order to unravel the contribution of tau hyperphosphorylation to hallmark features of Huntington's disease, we administered weekly intraperitoneal injections of the anti-tau pS202 CP13 monoclonal antibody to zQ175 mice and characterized the resulting behavioral and biochemical changes. After 12 weeks of treatment, motor impairments, cognitive performance and general health were improved in zQ175 mice along with a significant reduction in hippocampal pS202 tau levels. Despite the lack of effect of CP13 on neuronal markers associated with Huntington's disease pathology, tau-targeting enzymes and gliosis, CP13 was shown to directly impact mutant huntingtin aggregation such that brain levels of amyloid fibrils and huntingtin oligomers were decreased, while larger huntingtin protein aggregates were increased. Investigation of CP13 treatment of Huntington's disease patient-derived induced pluripotent stem cells (iPSCs) revealed a reduction in pS202 levels in differentiated cortical neurons and a rescue of neurite length. Collectively, these findings suggest that attenuating tau pathology could mitigate behavioral and molecular hallmarks associated with Huntington's disease.


Subject(s)
Huntington Disease , Induced Pluripotent Stem Cells , Animals , Brain/metabolism , Disease Models, Animal , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/therapy , Immunization, Passive , Induced Pluripotent Stem Cells/metabolism , Mice , Neurons/metabolism
6.
Mol Psychiatry ; 26(7): 2685-2706, 2021 07.
Article in English | MEDLINE | ID: mdl-33495544

ABSTRACT

Neurodegenerative disorders emerge from the failure of intricate cellular mechanisms, which ultimately lead to the loss of vulnerable neuronal populations. Research conducted across several laboratories has now provided compelling evidence that pathogenic proteins can also contribute to non-cell autonomous toxicity in several neurodegenerative contexts, including Alzheimer's, Parkinson's, and Huntington's diseases as well as Amyotrophic Lateral Sclerosis. Given the nearly ubiquitous nature of abnormal protein accumulation in such disorders, elucidating the mechanisms and routes underlying these processes is essential to the development of effective treatments. To this end, physiologically relevant human in vitro models are critical to understand the processes surrounding uptake, release and nucleation under physiological or pathological conditions. This review explores the use of human-induced pluripotent stem cells (iPSCs) to study prion-like protein propagation in neurodegenerative diseases, discusses advantages and limitations of this model, and presents emerging technologies that, combined with the use of iPSC-based models, will provide powerful model systems to propel fundamental research forward.


Subject(s)
Huntington Disease , Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Prions , Humans , Neurons
7.
Mol Psychiatry ; 26(9): 5441-5463, 2021 09.
Article in English | MEDLINE | ID: mdl-32514103

ABSTRACT

Huntington's disease (HD) is a monogenic neurodegenerative disorder resulting from a mutation in the huntingtin gene. This leads to the expression of the mutant huntingtin protein (mHTT) which provokes pathological changes in both the central nervous system (CNS) and periphery. Accumulating evidence suggests that mHTT can spread between cells of the CNS but here, we explored the possibility that mHTT could also propagate and cause pathology via the bloodstream. For this, we used a parabiosis approach to join the circulatory systems of wild-type (WT) and zQ175 mice. After surgery, we observed mHTT in the plasma and circulating blood cells of WT mice and post-mortem analyses revealed the presence of mHTT aggregates in several organs including the liver, kidney, muscle and brain. The presence of mHTT in the brain was accompanied by vascular abnormalities, such as a reduction of Collagen IV signal intensity and altered vessel diameter in the striatum, and changes in expression of Glutamic acid decarboxylase 65/67 (GAD65-67) in the cortex. Conversely, we measured reduced pathology in zQ175 mice by decreased mitochondrial impairments in peripheral organs, restored vessel diameter in the cortex and improved expression of Dopamine- and cAMP-regulated phosphoprotein 32 (DARPP32) in striatal neurons. Collectively, these results demonstrate that circulating mHTT can disseminate disease, but importantly, that healthy blood can dilute pathology. These findings have significant implications for the development of therapies in HD.


Subject(s)
Huntington Disease , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32 , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Mice , Mice, Transgenic , Neurons/metabolism
8.
Proc Natl Acad Sci U S A ; 115(30): E7081-E7090, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29987005

ABSTRACT

The huntingtin N17 domain is a modulator of mutant huntingtin toxicity and is hypophosphorylated in Huntington's disease (HD). We conducted high-content analysis to find compounds that could restore N17 phosphorylation. One lead compound from this screen was N6-furfuryladenine (N6FFA). N6FFA was protective in HD model neurons, and N6FFA treatment of an HD mouse model corrects HD phenotypes and eliminates cortical mutant huntingtin inclusions. We show that N6FFA restores N17 phosphorylation levels by being salvaged to a triphosphate form by adenine phosphoribosyltransferase (APRT) and used as a phosphate donor by casein kinase 2 (CK2). N6FFA is a naturally occurring product of oxidative DNA damage. Phosphorylated huntingtin functionally redistributes and colocalizes with CK2, APRT, and N6FFA DNA adducts at sites of induced DNA damage. We present a model in which this natural product compound is salvaged to provide a triphosphate substrate to signal huntingtin phosphorylation via CK2 during low-ATP stress under conditions of DNA damage, with protective effects in HD model systems.


Subject(s)
Adenine , DNA Adducts/metabolism , DNA Damage , Huntington Disease/drug therapy , Neurons/metabolism , Signal Transduction/drug effects , Adenine/analogs & derivatives , Adenine/pharmacokinetics , Adenine/pharmacology , Adenine Phosphoribosyltransferase/genetics , Adenine Phosphoribosyltransferase/metabolism , Animals , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Line, Transformed , DNA Adducts/genetics , Disease Models, Animal , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Mice, Transgenic , Neurons/pathology , Phosphorylation/drug effects , Phosphorylation/genetics , Signal Transduction/genetics
9.
Neurobiol Dis ; 145: 105042, 2020 11.
Article in English | MEDLINE | ID: mdl-32798729

ABSTRACT

A number of publications have reported that cysteamine has significant therapeutic effects on several aspects of Parkinson's disease (PD)-related pathology but none of these studies have evaluated its impact on pathological forms of α-Synuclein (α-Syn), one of the main hallmarks of PD. We therefore tested the efficacy of cysteamine on the Thy1-α-Syn mouse model which over-expresses full-length human wild-type α-Syn. Two-month (early stage disease) and 6-month old (late stage disease) mice and littermate controls were treated daily with cysteamine (20 mg/kg, i.p.) to assess the protective and restorative properties of this compound. After 6 weeks of treatment, animals were tested using a battery of motor tests. Cysteamine-treated transgenic mice displayed significant improvements in motor performance as compared to saline-treated transgenic littermates. Post-mortem readouts revealed a reduction in fibrillation, phosphorylation and total levels of overexpresed human α-Syn. To determine if such outcomes extended to human cells, the benefits of cysteamine were additionally tested using 6-hydroxydopamine (6-OHDA) treated neurons differentiated from induced pluripotent stem cells (iPSCs) derived from a PD patient harbouring a triplication of the SNCA gene. SNCA neurons treated with cysteamine exhibited significantly more intact/healthy neurites than cells treated with 6-OHDA alone. Additionally, SNCA neurons treated with cysteamine in the absence of 6-OHDA showed a trend towards lower total α-Syn levels. Overall, our in vivo and in vitro findings suggest that cysteamine can act as a disease-modifying molecule by enhancing -the survival of dopaminergic neurons and reducing pathological forms of α-Syn.


Subject(s)
Cysteamine/pharmacology , Dopaminergic Neurons/drug effects , Induced Pluripotent Stem Cells/drug effects , Parkinsonian Disorders/pathology , alpha-Synuclein/genetics , Animals , Dopaminergic Neurons/pathology , Humans , Locomotion/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic
10.
Neurobiol Dis ; 141: 104941, 2020 07.
Article in English | MEDLINE | ID: mdl-32422281

ABSTRACT

In recent years, substantial evidence has emerged to suggest that spreading of pathological proteins contributes to disease pathology in numerous neurodegenerative disorders. Work from our laboratory and others have shown that, despite its strictly genetic nature, Huntington's disease (HD) may be another condition in which this mechanism contributes to pathology. In this study, we set out to determine if the mutant huntingtin protein (mHTT) present in post-mortem brain tissue derived from HD patients can induce pathology in mice and/or non-human primates. For this, we performed three distinct sets of experiments where homogenates were injected into the brains of adult a) Wild-type (WT) and b) BACHD mice or c) non-human primates. Neuropathological assessments revealed that, while changes in the endogenous huntingtin were not apparent, mHTT could spread between cellular elements and brain structures. Furthermore, behavioural differences only occurred in the animal model of HD which already overexpressed mHTT. Taken together, our results indicate that mHTT derived from human brains has only a limited capacity to propagate between cells and does not depict prion-like characteristics. This contrasts with recent work demonstrating that other forms of mHTT - such as fibrils of a pathological polyQ length or fibroblasts and induced pluripotent stem cells derived from HD cases - can indeed disseminate disease throughout the brain in a prion-like fashion.


Subject(s)
Brain/pathology , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Protein Aggregation, Pathological , Animals , Behavior, Animal , Brain/metabolism , Child , Female , Humans , Huntingtin Protein/administration & dosage , Macaca mulatta , Mice, Inbred C57BL , Mutation , Neurons/pathology
11.
Neurobiol Dis ; 141: 104951, 2020 07.
Article in English | MEDLINE | ID: mdl-32439599

ABSTRACT

In order to model various aspects of Huntington's disease (HD) pathology, in particular protein spread, we administered adeno-associated virus (AAV) expressing green fluorescent protein (GFP) or GFP coupled to HTT-Exon1 (19Q or 103Q) to the central nervous system of adult wild-type (WT) mice and non-human primates. All animals underwent behavioral testing and post-mortem analyses to determine the long-term consequences of AAV injection. Both mice and non-human primates demonstrated behavioral changes at 2-3 weeks post-surgery. In mice, these changes were absent after 3 months while in non-human primates, they persisted in the majority of tested animals. Post-mortem analysis revealed that spreading of the aggregates was limited, although the virus did spread between synaptically-connected brain regions. Despite circumscribed spreading, the presence of mHTT generated changes in endogenous huntingtin (HTT) levels in both models. Together, these results suggest that viral expression of mHTTExon1 can induce spreading and seeding of HTT in both mice and non-human primates.


Subject(s)
Dependovirus/genetics , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Protein Aggregation, Pathological , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Genetic Vectors , Green Fluorescent Proteins/genetics , Humans , Macaca mulatta , Male , Mice, Inbred C57BL
12.
Acta Neuropathol ; 137(6): 981-1001, 2019 06.
Article in English | MEDLINE | ID: mdl-30788585

ABSTRACT

In recent years, evidence has accumulated to suggest that mutant huntingtin protein (mHTT) can spread into healthy tissue in a prion-like fashion. This theory, however, remains controversial. To fully address this concept and to understand the possible consequences of mHTT spreading to Huntington's disease pathology, we investigated the effects of exogenous human fibrillar mHTT (Q48) and huntingtin (HTT) (Q25) N-terminal fragments in three cellular models and three distinct animal paradigms. For in vitro experiments, human neuronal cells [induced pluripotent stem cell-derived GABA neurons (iGABA) and (SH-SY5Y)] as well as human THP1-derived macrophages, were incubated with recombinant mHTT fibrils. Recombinant mHTT and HTT fibrils were taken up by all cell types, inducing cell morphology changes and death. Variations in HTT aggregation were further observed following incubation with fibrils in both THP1 and SH-SY5Y cells. For in vivo experiments, adult wild-type (WT) mice received a unilateral intracerebral cortical injection and R6/2 and WT pups were administered fibrils via bilateral intraventricular injections. In both protocols, the injection of Q48 fibrils resulted in cognitive deficits and increased anxiety-like behavior. Post-mortem analysis of adult WT mice indicated that most fibrils had been degraded/cleared from the brain by 14 months post-surgery. Despite the absence of fibrils at these later time points, a change in the staining pattern of endogenous HTT was detected. A similar change was revealed in post-mortem analysis of the R6/2 mice. These effects were specific to central administration of fibrils, as mice receiving intravenous injections were not characterized by behavioral changes. In fact, peripheral administration resulted in an immune response mounting against the fibrils. Together, the in vitro and in vivo data indicate that exogenously administered mHTT is capable of both causing and exacerbating disease pathology.


Subject(s)
GABAergic Neurons/metabolism , Huntingtin Protein/genetics , Protein Aggregates , Animals , Anxiety/etiology , Brain/pathology , Cell Line, Tumor , Cognition Disorders/chemically induced , Cognition Disorders/pathology , Exons , Exploratory Behavior , Female , GABAergic Neurons/ultrastructure , Humans , Huntingtin Protein/administration & dosage , Huntingtin Protein/chemistry , Huntingtin Protein/toxicity , Induced Pluripotent Stem Cells/cytology , Injections , Injections, Intraventricular , Male , Maze Learning , Mice , Mice, Inbred C57BL , Monocytes , Motor Activity , Neuroblastoma/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/toxicity
17.
Proc Natl Acad Sci U S A ; 109(9): 3528-33, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22331905

ABSTRACT

Huntington disease (HD) is a progressive neurodegenerative monogenic disorder caused by expansion of a polyglutamine stretch in the huntingtin (Htt) protein. Mutant huntingtin triggers neural dysfunction and death, mainly in the corpus striatum and cerebral cortex, resulting in pathognomonic motor symptoms, as well as cognitive and psychiatric decline. Currently, there is no effective treatment for HD. We report that intraventricular infusion of ganglioside GM1 induces phosphorylation of mutant huntingtin at specific serine amino acid residues that attenuate huntingtin toxicity, and restores normal motor function in already symptomatic HD mice. Thus, our studies have identified a potential therapy for HD that targets a posttranslational modification of mutant huntingtin with critical effects on disease pathogenesis.


Subject(s)
G(M1) Ganglioside/therapeutic use , Motor Activity/drug effects , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Processing, Post-Translational/drug effects , Animals , Codon/drug effects , Corpus Striatum/metabolism , Dimerization , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/biosynthesis , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Drug Evaluation, Preclinical , G(M1) Ganglioside/administration & dosage , Huntingtin Protein , Infusion Pumps, Implantable , Infusions, Parenteral , Mice , Mice, Neurologic Mutants , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/metabolism , Phosphorylation/drug effects , Phosphoserine/analysis , Psychomotor Performance/drug effects
18.
J Neurol ; 268(9): 3493-3504, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33625583

ABSTRACT

Decades of research on the prion protein and its associated diseases have caused a paradigm shift in our understanding of infectious agents. More recent years have been marked by a surge of studies supporting the application of these findings to a broad array of neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Here, we present evidence to suggest that Huntington's disease, a monogenic disorder of the central nervous system, shares features with prion disorders and that, it too, may be governed by similar mechanisms. We further posit that these similarities could suggest that, like other common neurodegenerative disorders, sporadic forms of Huntington's disease may exist.


Subject(s)
Huntington Disease , Neurodegenerative Diseases , Prion Diseases , Prions , Humans , Huntington Disease/genetics , Prion Proteins/genetics , Prions/genetics
19.
Cell Rep ; 34(10): 108833, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33691111

ABSTRACT

Sex differences in multiple sclerosis (MS) incidence and severity have long been recognized. However, the underlying cellular and molecular mechanisms for why male sex is associated with more aggressive disease remain poorly defined. Using a T cell adoptive transfer model of chronic experimental autoimmune encephalomyelitis (EAE), we find that male Th17 cells induce disease of increased severity relative to female Th17 cells, irrespective of whether transferred to male or female recipients. Throughout the disease course, a greater frequency of male Th17 cells produce IFNγ, a hallmark of pathogenic Th17 responses. Intriguingly, XY chromosomal complement increases the pathogenicity of male Th17 cells. An X-linked immune regulator, Jarid1c, is downregulated in pathogenic male murine Th17 cells, and functional experiments reveal that it represses the severity of Th17-mediated EAE. Furthermore, Jarid1c expression is downregulated in CD4+ T cells from MS-affected individuals. Our data indicate that male sex chromosomal complement critically regulates Th17 cell pathogenicity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Sex Chromosomes/genetics , Th17 Cells/immunology , Animals , Autoimmunity , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Down-Regulation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Severity of Illness Index , Th17 Cells/cytology , Th17 Cells/metabolism
20.
Sci Rep ; 9(1): 6444, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015544

ABSTRACT

Deep brain stimulation (DBS) has been used in clinical settings for many years despite a paucity of knowledge related to the anatomical and functional substrates that lead to benefits and/or side-effects in various disease contexts. In order to maximize the potential of this approach in humans, a better understanding of its mechanisms of action is absolutely necessary. However, the existing micro-stimulators available for pre-clinical models, are limited by the lack of relevant small size devices. This absence prevents sustained chronic stimulation and real time monitoring of animals during stimulation, parameters that are critical for comparison to clinical findings. We therefore sought to develop and refine a novel small wireless micro-stimulator as a means by which to study consequent behavioural to molecular changes in experimental animals. Building on previous work from our group, we refined our implantable micro-stimulator prototype, to be easily combined with intravital 2-photon imaging. Using our prototype we were able to replicate the well described clinical benefits on motor impairment in a mouse model of Parkinson's disease in addition to capturing microglia dynamics live during stimulation. We believe this new device represents a useful tool for performing pre-clinical studies as well as dissecting brain circuitry and function.


Subject(s)
Deep Brain Stimulation , Parkinson Disease , Wireless Technology , Animals , Disease Models, Animal , Humans , Male , Mice , Parkinson Disease/physiopathology , Parkinson Disease/therapy
SELECTION OF CITATIONS
SEARCH DETAIL