Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Neurobiol Dis ; 197: 106530, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38750673

RESUMO

Heterogeneity is one of the key features of the healthy brain and selective vulnerability characterizes many, if not all, neurodegenerative diseases. While cerebellum contains majority of brain cells, neither its heterogeneity nor selective vulnerability in disease are well understood. Here we describe molecular, cellular and functional heterogeneity in the context of healthy cerebellum as well as in cerebellar disease Spinocerebellar Ataxia Type 1 (SCA1). We first compared disease pathology in cerebellar vermis and hemispheres across anterior to posterior axis in a knock-in SCA1 mouse model. Using immunohistochemistry, we demonstrated earlier and more severe pathology of PCs and glia in the posterior cerebellar vermis of SCA1 mice. We also demonstrate heterogeneity of Bergmann glia in the unaffected, wild-type mice. Then, using RNA sequencing, we found both shared, as well as, posterior cerebellum-specific molecular mechanisms of pathogenesis that include exacerbated gene dysregulation, increased number of altered signaling pathways, and decreased pathway activity scores in the posterior cerebellum of SCA1 mice. We demonstrated unexpectedly large differences in the gene expression between posterior and anterior cerebellar vermis of wild-type mice, indicative of robust intraregional heterogeneity of gene expression in the healthy cerebellum. Additionally, we found that SCA1 disease profoundly reduces intracerebellar heterogeneity of gene expression. Further, using fiber photometry, we found that population level PC calcium activity was altered in the posterior lobules in SCA1 mice during walking. We also identified regional differences in the population level activity of Purkinje cells (PCs) in unrestrained wild-type mice that were diminished in SCA1 mice.


Assuntos
Cerebelo , Ataxias Espinocerebelares , Animais , Cerebelo/metabolismo , Cerebelo/patologia , Ataxias Espinocerebelares/patologia , Ataxias Espinocerebelares/metabolismo , Ataxias Espinocerebelares/genética , Camundongos , Ataxina-1/metabolismo , Ataxina-1/genética , Células de Purkinje/patologia , Células de Purkinje/metabolismo , Neuroglia/metabolismo , Neuroglia/patologia , Modelos Animais de Doenças , Camundongos Transgênicos , Camundongos Endogâmicos C57BL , Masculino
2.
Bratisl Lek Listy ; 125(6): 354-359, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38757591

RESUMO

BACKGROUND: In the present study, two structurally similar alkaloids from trees of Cinchona genus, chloroquine and cinchonine, were examined for their vasorelaxant effects in a model of phenylephrine-induced smooth muscle contractions. METHODS: Potential mechanisms of action associated with endothelial vasorelaxant compounds, voltage-gated Ca2+ channels (LTCCs), and inositol triphosphate receptors were examined in isolated rat aortic rings. Also, an in silico approach was used to predict the activity of the two test compounds. RESULTS: Experimental results revealed that both chloroquine and cinchonine significantly decrease phenylephrine-induced smooth muscle contractions, although to a different extent. Evaluated mechanisms of action indicate that endothelium is not involved in the vasorelaxant action of the two tested alkaloids. On the other hand, voltage-gated Ca2+ channels were found to be the dominant way of action associated with the vasorelaxant action of chloroquine and cinchonine. Finally, IP3R is found to have only a small impact on the observed activity of the tested compounds. CONCLUSION: Molecular docking studies predicted that chloroquine possesses a significant activity toward a suitable model of LTCCs, while cinchonine does not. The results of the present study point to the fact that great caution should be paid while administering chloroquine to vulnerable patients, especially those with cardiovascular disorders (Tab. 3, Fig. 3, Ref. 28).


Assuntos
Canais de Cálcio , Cloroquina , Simulação de Acoplamento Molecular , Músculo Liso Vascular , Animais , Cloroquina/farmacologia , Ratos , Músculo Liso Vascular/efeitos dos fármacos , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/metabolismo , Vasodilatadores/farmacologia , Tono Muscular/efeitos dos fármacos , Masculino , Ratos Wistar , Simulação por Computador , Fenilefrina/farmacologia
3.
Neurobiol Dis ; 187: 106318, 2023 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-37802154

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a neurodegenerative disease caused by an abnormal expansion of glutamine (Q) encoding CAG repeats in the ATAXIN1 (ATXN1) gene and characterized by progressive cerebellar ataxia, dysarthria, and eventual deterioration of bulbar functions. SCA1 shows severe degeneration of cerebellar Purkinje cells (PCs) and activation of Bergmann glia (BG), a type of cerebellar astroglia closely associated with PCs. Combining electrophysiological recordings, calcium imaging techniques, and chemogenetic approaches, we have investigated the electrical intrinsic and synaptic properties of PCs and the physiological properties of BG in SCA1 mouse model expressing mutant ATXN1 only in PCs. PCs of SCA1 mice displayed lower spontaneous firing rate and larger slow afterhyperpolarization currents (sIAHP) than wildtype mice, whereas the properties of the synaptic inputs were unaffected. BG of SCA1 mice showed higher calcium hyperactivity and gliotransmission, manifested by higher frequency of NMDAR-mediated slow inward currents (SICs) in PC. Preventing the BG calcium hyperexcitability of SCA1 mice by loading BG with the calcium chelator BAPTA restored sIAHP and spontaneous firing rate of PCs to similar levels of wildtype mice. Moreover, mimicking the BG hyperactivity by activating BG expressing Gq-DREADDs in wildtype mice reproduced the SCA1 pathological phenotype of PCs, i.e., enhancement of sIAHP and decrease of spontaneous firing rate. These results indicate that the intrinsic electrical properties of PCs, but not their synaptic properties, were altered in SCA1 mice and that these alterations were associated with the hyperexcitability of BG. Moreover, preventing BG hyperexcitability in SCA1 mice and promoting BG hyperexcitability in wildtype mice prevented and mimicked, respectively, the pathological electrophysiological phenotype of PCs. Therefore, BG plays a relevant role in the dysfunction of the electrical intrinsic properties of PCs in SCA1 mice, suggesting that they may serve as potential targets for therapeutic approaches to treat the spinocerebellar ataxia type 1.


Assuntos
Cálcio , Ataxias Espinocerebelares , Camundongos , Animais , Cálcio/fisiologia , Sinalização do Cálcio , Camundongos Transgênicos , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia , Cerebelo/patologia , Células de Purkinje/patologia , Neuroglia/patologia , Ataxina-1/genética
4.
Neurobiol Dis ; 178: 106023, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36724861

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is an adult-onset, dominantly inherited neurodegenerative disease caused by the expanded polyQ tract in the protein ATAXIN1 (ATXN1) and characterized by progressive motor and cognitive impairments. There are no disease-modifying treatments or cures for SCA1. Brain-derived neurotrophic factor (BDNF) plays important role in cerebellar physiology and has shown therapeutic potential for cerebellar pathology in the transgenic mouse model of SCA1, ATXN1[82Q] line that overexpress mutant ATXN1 under a cerebellar Purkinje-cell-specific promoter. Here we demonstrate decreased expression of brain derived neurotrophic factor (BDNF) in the cerebellum and medulla of patients with SCA1. Early stages of disease seem most amenable to therapy. Thus, we next quantified Bdnf expression in Atxn1154Q/2Q mice, a knock-in mouse model of SCA1, during the early symptomatic disease stage in four clinically relevant brain regions: cerebellum, medulla, hippocampus and motor cortex. We found that during the early stages of disease, Bdnf mRNA expression is reduced in the hippocampus and cerebellum, while it is increased in the cortex and brainstem. Importantly, we observed that pharmacological delivery of recombinant BDNF improved motor and cognitive performance, and mitigated pathology in the cerebellum and hippocampus of Atxn1154Q/2Q mice. Our findings demonstrate brain-region specific deficiency of BDNF in SCA1 and show that reversal of low BDNF levels offers the potential for meaningful treatment of motor and cognitive deficits in SCA1.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Ataxias Espinocerebelares , Camundongos , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Ataxina-1/genética , Ataxina-1/metabolismo , Ataxias Espinocerebelares/metabolismo , Cerebelo/patologia , Camundongos Transgênicos , Células de Purkinje/metabolismo , Modelos Animais de Doenças
5.
Int J Mol Sci ; 24(8)2023 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-37108212

RESUMO

The extracellular matrix (ECM) surrounds cells in the brain, providing structural and functional support. Emerging studies demonstrate that the ECM plays important roles during development, in the healthy adult brain, and in brain diseases. The aim of this review is to briefly discuss the physiological roles of the ECM and its contribution to the pathogenesis of brain disease, highlighting the gene expression changes, transcriptional factors involved, and a role for microglia in ECM regulation. Much of the research conducted thus far on disease states has focused on "omic" approaches that reveal differences in gene expression related to the ECM. Here, we review recent findings on alterations in the expression of ECM-associated genes in seizure, neuropathic pain, cerebellar ataxia, and age-related neurodegenerative disorders. Next, we discuss evidence implicating the transcription factor hypoxia-inducible factor 1 (HIF-1) in regulating the expression of ECM genes. HIF-1 is induced in response to hypoxia, and also targets genes involved in ECM remodeling, suggesting that hypoxia could contribute to ECM remodeling in disease conditions. We conclude by discussing the role microglia play in the regulation of the perineuronal nets (PNNs), a specialized form of ECM in the central nervous system. We show evidence that microglia can modulate PNNs in healthy and diseased brain states. Altogether, these findings suggest that ECM regulation is altered in brain disease, and highlight the role of HIF-1 and microglia in ECM remodeling.


Assuntos
Encefalopatias , Matriz Extracelular , Humanos , Matriz Extracelular/metabolismo , Encéfalo/metabolismo , Encefalopatias/genética , Encefalopatias/metabolismo
6.
Hum Mol Genet ; 29(1): 117-131, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31696233

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by abnormal expansion of glutamine (Q) encoding CAG repeats in the gene Ataxin-1 (ATXN1). Although motor and balance deficits are the core symptoms of SCA1, cognitive decline is also commonly observed in patients. While mutant ATXN1 is expressed throughout the brain, pathological findings reveal severe atrophy of cerebellar cortex in SCA1 patients. The cerebellum has recently been implicated in diverse cognitive functions, yet to what extent cerebellar neurodegeneration contributes to cognitive alterations in SCA1 remains poorly understood. Much of our understanding of the mechanisms underlying pathogenesis of motor symptoms in SCA1 comes from mouse models. Reasoning that mouse models could similarly offer important insights into the mechanisms of cognitive alterations in SCA1, we tested cognition in several mouse lines using Barnes maze and fear conditioning. We confirmed cognitive deficits in Atxn1154Q/2Q knock-in mice with brain-wide expression of mutant ATXN1 and in ATXN1 null mice. We found that shorter polyQ length and haploinsufficiency of ATXN1 do not cause significant cognitive deficits. Finally, ATXN1[82Q ] transgenic mice-with cerebellum limited expression of mutant ATXN1-demonstrated milder impairment in most aspects of cognition compared to Atxn1154Q/2Q mice, supporting the concept that cognitive deficits in SCA1 arise from a combination of cerebellar and extra-cerebellar dysfunctions.


Assuntos
Ataxina-1/metabolismo , Cerebelo/metabolismo , Disfunção Cognitiva/metabolismo , Animais , Ataxina-1/genética , Ataxina-3/genética , Ataxina-3/metabolismo , Cognição/fisiologia , Disfunção Cognitiva/genética , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/metabolismo
7.
Cerebellum ; 21(3): 452-481, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34378174

RESUMO

Spinocerebellar ataxias (SCAs) represent a large group of hereditary degenerative diseases of the nervous system, in particular the cerebellum, and other systems that manifest with a variety of progressive motor, cognitive, and behavioral deficits with the leading symptom of cerebellar ataxia. SCAs often lead to severe impairments of the patient's functioning, quality of life, and life expectancy. For SCAs, there are no proven effective pharmacotherapies that improve the symptoms or substantially delay disease progress, i.e., disease-modifying therapies. To study SCA pathogenesis and potential therapies, animal models have been widely used and are an essential part of pre-clinical research. They mainly include mice, but also other vertebrates and invertebrates. Each animal model has its strengths and weaknesses arising from model animal species, type of genetic manipulation, and similarity to human diseases. The types of murine and non-murine models of SCAs, their contribution to the investigation of SCA pathogenesis, pathological phenotype, and therapeutic approaches including their advantages and disadvantages are reviewed in this paper. There is a consensus among the panel of experts that (1) animal models represent valuable tools to improve our understanding of SCAs and discover and assess novel therapies for this group of neurological disorders characterized by diverse mechanisms and differential degenerative progressions, (2) thorough phenotypic assessment of individual animal models is required for studies addressing therapeutic approaches, (3) comparative studies are needed to bring pre-clinical research closer to clinical trials, and (4) mouse models complement cellular and invertebrate models which remain limited in terms of clinical translation for complex neurological disorders such as SCAs.


Assuntos
Qualidade de Vida , Ataxias Espinocerebelares , Animais , Cerebelo/patologia , Consenso , Camundongos , Modelos Animais , Ataxias Espinocerebelares/diagnóstico , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/terapia
8.
Cerebellum ; 20(3): 420-429, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33394333

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by an abnormal expansion of CAG repeats in the Ataxin1 (ATXN1) gene. SCA1 is characterized by motor deficits, cerebellar neurodegeneration, and gliosis and gene expression changes. Expression of brain-derived neurotrophic factor (BDNF), growth factor important for the survival and function of cerebellar neurons, is decreased in ATXN1[82Q] mice, the Purkinje neuron specific transgenic mouse model of SCA1. As this decrease in BDNF expression may contribute to cerebellar neurodegeneration, we tested whether delivery of extrinsic human BDNF via osmotic ALZET pumps has a beneficial effect on disease severity in this mouse model of SCA1. Additionally, to test the effects of BDNF on established and progressing cerebellar pathogenesis and motor deficits, we delivered BDNF post-symptomatically. We have found that post-symptomatic delivery of extrinsic BDNF ameliorated motor deficits and cerebellar pathology (i.e., dendritic atrophy of Purkinje cells, and astrogliosis) indicating therapeutic potential of BDNF even after the onset of symptoms in SCA1. However, BDNF did not alter Purkinje cell gene expression changes indicating that certain aspects of disease pathogenesis cannot be ameliorated/slowed down with BDNF and that combinational therapies may be needed.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/uso terapêutico , Ataxias Espinocerebelares/terapia , Animais , Cerebelo/patologia , Dendritos/patologia , Feminino , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Gliose/patologia , Gliose/terapia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Células de Purkinje/patologia , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia
9.
Int J Mol Sci ; 21(23)2020 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-33276471

RESUMO

Over the past decade, research has unveiled the intimate relationship between neuroinflammation and neurodegeneration. Microglia and astrocytes react to brain insult by setting up a multimodal inflammatory state and act as the primary defenders and executioners of neuroinflammatory structural and functional changes. Microglia and astrocytes also play critical roles in the maintenance of normal brain function. This intricate balance of homeostatic and neuroinflammatory functions can influence the onset and the course of neurodegenerative diseases. The emergent role of the microglial-astrocytic axis in neurodegenerative disease presents many druggable targets that may have broad therapeutic benefits across neurodegenerative disease. Here, we provide a brief review of the basal function of both microglia and astrocytes, how they are changed in disease states, the significant differences between mouse and human glia, and use of human induced pluripotent stem cells derived from patients to study cell autonomous changes in human astrocytes and microglia.


Assuntos
Suscetibilidade a Doenças , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/metabolismo , Neuroglia/metabolismo , Animais , Astrócitos/metabolismo , Diferenciação Celular , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Microglia/imunologia , Microglia/metabolismo , Doenças Neurodegenerativas/patologia , Especificidade da Espécie
10.
Glia ; 66(9): 1972-1987, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30043530

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a fatal, dominantly inherited neurodegenerative disease caused by the expansion of CAG repeats in the Ataxin-1 (ATXN1) gene. SCA1 is characterized by balance and coordination deficits due to the predominant loss of Purkinje neurons in the cerebellum. We previously demonstrated that cerebellar astrogliosis beings during the early stages of SCA1, prior to onset of motor deficits and loss of Purkinje neurons. We communicate here that cerebellar astrogliosis contributes to SCA1 pathogenesis in a biphasic, stage of disease dependent manner. We modulated astrogliosis by selectively reducing pro-inflammatory transcriptional regulator nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) signaling in astroglia via a Cre-lox mouse genetic approach. Our results indicate that inhibition of astroglial NF-κB signaling, prior to motor deficit onset, exacerbates disease severity. This is suggestive of a neuroprotective role mediated by astroglia during early stage SCA1. In contrast, inhibition of astroglial NF-κB signaling during late stage of disease ameliorated motor deficits, indicating a potentially harmful role of astroglia late in SCA1. These results indicate that astrogliosis may have a critical and dual role in disease. If so, our results imply that anti-inflammatory astroglia-based therapeutic approaches may need to consider disease progression to achieve therapeutic efficacy.


Assuntos
Astrócitos/fisiologia , Gliose/fisiopatologia , Ataxias Espinocerebelares/fisiopatologia , Animais , Astrócitos/patologia , Ataxina-1/genética , Ataxina-1/metabolismo , Cerebelo/patologia , Cerebelo/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/patologia , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/patologia , Microglia/fisiologia , Atividade Motora/fisiologia , NF-kappa B/metabolismo , Neurônios/patologia , Neurônios/fisiologia , Neuroproteção/fisiologia , Distribuição Aleatória , Ataxias Espinocerebelares/patologia
11.
J Neuroinflammation ; 14(1): 107, 2017 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-28545543

RESUMO

BACKGROUND: Polyglutamine (polyQ) expansion in the protein Ataxin-1 (ATXN1) causes spinocerebellar ataxia type 1 (SCA1), a fatal dominantly inherited neurodegenerative disease characterized by motor deficits, cerebellar neurodegeneration, and gliosis. Currently, there are no treatments available to delay or ameliorate SCA1. We have examined the effect of depleting microglia during the early stage of disease by using PLX, an inhibitor of colony-stimulating factor 1 receptor (CSFR1), on disease severity in a mouse model of SCA1. METHODS: Transgenic mouse model of SCA1, ATXN1[82Q] mice, and wild-type littermate controls were treated with PLX from 3 weeks of age. The effects of PLX on microglial density, astrogliosis, motor behavior, atrophy, and gene expression of Purkinje neurons were examined at 3 months of age. RESULTS: PLX treatment resulted in the elimination of 70-80% of microglia from the cerebellum of both wild-type and ATXN1[82Q] mice. Importantly, PLX ameliorated motor deficits in SCA1 mice. While we have not observed significant improvement in the atrophy or disease-associated gene expression changes in Purkinje neurons upon PLX treatment, we have detected reduced expression of pro-inflammatory cytokine tumor necrosis factor alpha (TNFα) and increase in the protein levels of wild-type ataxin-1 and post-synaptic density protein 95 (PSD95) that may help improve PN function. CONCLUSIONS: A decrease in the number of microglia during an early stage of disease resulted in the amelioration of motor deficits in SCA1 mice.


Assuntos
Fator Estimulador de Colônias de Macrófagos/metabolismo , Transtornos Motores/etiologia , Transtornos Motores/terapia , Ataxias Espinocerebelares/complicações , Aminopiridinas/uso terapêutico , Animais , Ataxina-1/genética , Ataxina-1/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Cerebelo/patologia , Proteína 4 Homóloga a Disks-Large/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Mutação/genética , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Equilíbrio Postural/efeitos dos fármacos , Equilíbrio Postural/genética , Pirróis/uso terapêutico , Ataxias Espinocerebelares/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
12.
Cerebellum ; 16(2): 340-347, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27306906

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited neurodegenerative disease caused by the expansion of a polyglutamine (Q) repeat tract in the protein ataxin-1 (ATXN1). Beginning as a cerebellar ataxic disorder, SCA1 progresses to involve the cerebral cortex, hippocampus, and brainstem. Using SCA1 knock-in mice that mirror the complexity of the human disease, we report a significant decrease in the capacity of adult neuronal progenitor cells (NPCs) to proliferate. Remarkably, a decrease in NPCs proliferation can be observed in vitro, outside the degenerative milieu of surrounding neurons or glia, demonstrating that mutant ATXN1 acting cell autonomously within progenitor cells interferes with their ability to proliferate. Our findings suggest that compromised adult neurogenesis contributes to the progressive pathology of the disease particularly in areas such as the hippocampus and cerebral cortex where stem cells provide neurotropic factors and participate in adult neurogenesis. These findings not only shed light on the biology of the disease but also have therapeutic implications in any future stem cell-based clinical trials.


Assuntos
Células-Tronco Adultas/metabolismo , Ataxina-1/metabolismo , Proliferação de Células/fisiologia , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Ataxias Espinocerebelares/metabolismo , Células-Tronco Adultas/patologia , Animais , Ataxina-1/genética , Western Blotting , Bromodesoxiuridina , Células Cultivadas , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Imuno-Histoquímica , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Células-Tronco Neurais/patologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Nicho de Células-Tronco/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Hum Mol Genet ; 23(14): 3733-45, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24594842

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is an incurable neurodegenerative disease caused by a pathogenic glutamine repeat expansion in the protein ataxin-1 (ATXN1). One likely mechanism mediating pathogenesis is excessive transcriptional repression induced by the expanded ATXN-1. Because ATXN1 binds HDAC3, a Class I histone deacetylase (HDAC) that we have found to be required for ATXN1-induced transcriptional repression, we tested whether genetically depleting HDAC3 improves the phenotype of the SCA1 knock-in mouse (SCA1(154Q/2Q)), the most physiologically relevant model of SCA1. Given that HDAC3 null mice are embryonic lethal, we used for our analyses a combination of HDAC3 haploinsufficient and Purkinje cell (PC)-specific HDAC3 null mice. Although deleting a single allele of HDAC3 in the context of SCA1 was insufficient to improve cerebellar and cognitive deficits of the disease, a complete loss of PC HDAC3 was highly deleterious both behaviorally, with mice showing early onset ataxia, and pathologically, with progressive histologic evidence of degeneration. Inhibition of HDAC3 may yet have a role in SCA1 therapy, but our study provides cautionary evidence that this approach could produce untoward effects. Indeed, the neurotoxic consequences of HDAC3 depletion could prove relevant, wherever pharmacologic inhibition of HDAC3 is being contemplated, in disorders ranging from cancer to neurodegeneration.


Assuntos
Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Células de Purkinje/metabolismo , Ataxias Espinocerebelares/patologia , Animais , Ataxina-1 , Ataxinas , Peso Corporal , Linhagem Celular Tumoral , Cerebelo/patologia , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Células HEK293 , Haploinsuficiência , Inibidores de Histona Desacetilases/farmacologia , Humanos , Camundongos , Atividade Motora , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Células de Purkinje/patologia , Ataxias Espinocerebelares/tratamento farmacológico , Ataxias Espinocerebelares/genética
14.
Cerebellum ; 14(1): 8-11, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25255716

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited neurodegenerative disease of the cerebellum caused by a polyglutamine-repeat expansion in the protein ATXN1. We have previously demonstrated that astrocytic activation occurs early in pathogenesis, correlates with disease progression, and can occur when mutant ATXN1 expression is limited to Purkinje neurons. We now show that expression of glutamate and aspartate transporter, GLAST, is decreased in cerebellar astrocytes in a mouse model of SCA1. This decrease occurs in non-cell autonomous manner late in disease and correlates well with the loss of Purkinje neurons. Astrogliosis or decreased neuronal activity does not correlate with diminished GLAST expression. In addition, Bergmann glia remain capable of transcriptional upregulation of GLAST in response to improvement in Purkinje neurons supporting the notion of active neuron-glia crosstalk in disease.


Assuntos
Astrócitos/metabolismo , Transportador 1 de Aminoácido Excitatório/deficiência , Células de Purkinje/patologia , Ataxias Espinocerebelares/metabolismo , Ataxias Espinocerebelares/patologia , Animais , Astrócitos/patologia , Ataxina-1 , Ataxinas , Gliose/metabolismo , Gliose/patologia , Humanos , Imuno-Histoquímica , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Células de Purkinje/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
15.
bioRxiv ; 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-39026827

RESUMO

The cerebellum role in cognition and its functional bi-directional connectivity with prefrontal cortex (PFC) is well recognized. However, how chronic cerebellar dysfunction affects PFC function and cognition remains less understood. Spinocerebellar ataxia type 1 (SCA1), is an inherited, fatal neurodegenerative disease caused by an abnormal expansion of glutamine (Q) encoding CAG repeats in the gene Ataxin-1 (ATXN1) and characterized by severe loss of Purkinje cells (PCs) in the cerebellum. Patients with SCA1 suffer from movement and balance deficits, cognitive decline and premature lethality. Cognitive deficits significantly impact patients quality of life, yet how exactly cerebellar degeneration contributes to cognitive deficits and PFC dysfunction in SCA1 is unknown. We have previously demonstrated that expression of mutant ATXN1 only in cerebellar Purkinje cells (PCs) is sufficient to cause cognitive deficits in a transgenic ATXN1[82Q] mouse line. To understand how cerebellar dysfunction impacts the PFC, we examined neuronal activity, synaptic density, and gene expression changes in the PFC of ATXN1[82Q] mice. Remarkably, we found decreased neuronal activity, reduced synaptic density, and altered expression of immediate early genes and pathways involved in glucose metabolism, inflammation and amphetamine in the PFC of ATXN1[82Q] mice. Furthermore, we characterized cellular and molecular PFC dysfunction in a novel conditional knock-in SCA1 line, f-ATXN1146Q mice, expressing floxed human expanded ATXN1 throughout the brain. Intriguingly, we found an increased number of neurons, increased synaptic density and large gene expression alterations in the PFC of f-ATXN1146Q mice. Finally, to precisely determine the role of cerebellar dysfunction in cognitive deficits and PFC dysfunction in SCA1, we crossed f-ATXN1146Q mice with Pcp2-Cre mice expressing Cre recombinase in PCs to delete expanded ATXN1 only in PCs. Surprisingly, we have found that deleting expanded ATXN1 in PCs exacerbated cognitive deficits and PFC dysfunction in these mice. Our findings demonstrate that circumscribed cerebellar dysfunction is sufficient to impact PFC activity and synaptic connectivity impairing cognition. However, when multiple brain regions are impacted in disease, cerebellar dysfunction may ameliorate PFC pathology and cognitive performance.

16.
JCI Insight ; 9(9)2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38512434

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by an expanded polyglutamine tract in the widely expressed ataxin-1 (ATXN1) protein. To elucidate anatomical regions and cell types that underlie mutant ATXN1-induced disease phenotypes, we developed a floxed conditional knockin mouse (f-ATXN1146Q/2Q) with mouse Atxn1 coding exons replaced by human ATXN1 exons encoding 146 glutamines. f-ATXN1146Q/2Q mice manifested SCA1-like phenotypes including motor and cognitive deficits, wasting, and decreased survival. Central nervous system (CNS) contributions to disease were revealed using f-ATXN1146Q/2Q;Nestin-Cre mice, which showed improved rotarod, open field, and Barnes maze performance by 6-12 weeks of age. In contrast, striatal contributions to motor deficits using f-ATXN1146Q/2Q;Rgs9-Cre mice revealed that mice lacking ATXN1146Q/2Q in striatal medium-spiny neurons showed a trending improvement in rotarod performance at 30 weeks of age. Surprisingly, a prominent role for muscle contributions to disease was revealed in f-ATXN1146Q/2Q;ACTA1-Cre mice based on their recovery from kyphosis and absence of muscle pathology. Collectively, data from the targeted conditional deletion of the expanded allele demonstrated CNS and peripheral contributions to disease and highlighted the need to consider muscle in addition to the brain for optimal SCA1 therapeutics.


Assuntos
Ataxina-1 , Modelos Animais de Doenças , Músculo Esquelético , Ataxias Espinocerebelares , Animais , Ataxina-1/genética , Ataxina-1/metabolismo , Camundongos , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia , Músculo Esquelético/patologia , Músculo Esquelético/metabolismo , Humanos , Masculino , Camundongos Transgênicos , Técnicas de Introdução de Genes , Feminino , Fenótipo , Neurônios/metabolismo , Neurônios/patologia
17.
Neurotherapeutics ; 20(1): 48-66, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-37020152

RESUMO

Neurodegenerative diseases are broadly characterized neuropathologically by the degeneration of vulnerable neuronal cell types in a specific brain region. The degeneration of specific cell types has informed on the various phenotypes/clinical presentations in someone suffering from these diseases. Prominent neurodegeneration of specific neurons is seen in polyglutamine expansion diseases including Huntington's disease (HD) and spinocerebellar ataxias (SCA). The clinical manifestations observed in these diseases could be as varied as the abnormalities in motor function observed in those who have Huntington's disease (HD) as demonstrated by a chorea with substantial degeneration of striatal medium spiny neurons (MSNs) or those with various forms of spinocerebellar ataxia (SCA) with an ataxic motor presentation primarily due to degeneration of cerebellar Purkinje cells. Due to the very significant nature of the degeneration of MSNs in HD and Purkinje cells in SCAs, much of the research has centered around understanding the cell autonomous mechanisms dysregulated in these neuronal cell types. However, an increasing number of studies have revealed that dysfunction in non-neuronal glial cell types contributes to the pathogenesis of these diseases. Here we explore these non-neuronal glial cell types with a focus on how each may contribute to the pathogenesis of HD and SCA and the tools used to evaluate glial cells in the context of these diseases. Understanding the regulation of supportive and harmful phenotypes of glia in disease could lead to development of novel glia-focused neurotherapeutics.


Assuntos
Doença de Huntington , Ataxias Espinocerebelares , Camundongos , Animais , Doença de Huntington/metabolismo , Neurônios/metabolismo , Ataxias Espinocerebelares/metabolismo , Neuroglia/patologia , Modelos Animais de Doenças , Camundongos Transgênicos
18.
Neuron ; 111(4): 493-507.e6, 2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-36577403

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a dominant trinucleotide repeat neurodegenerative disease characterized by motor dysfunction, cognitive impairment, and premature death. Degeneration of cerebellar Purkinje cells is a frequent and prominent pathological feature of SCA1. We previously showed that transport of ATXN1 to Purkinje cell nuclei is required for pathology, where mutant ATXN1 alters transcription. To examine the role of ATXN1 nuclear localization broadly in SCA1-like disease pathogenesis, CRISPR-Cas9 was used to develop a mouse with an amino acid alteration (K772T) in the nuclear localization sequence of the expanded ATXN1 protein. Characterization of these mice indicates that proper nuclear localization of mutant ATXN1 contributes to many disease-like phenotypes including motor dysfunction, cognitive deficits, and premature lethality. RNA sequencing analysis of genes with expression corrected to WT levels in Atxn1175QK772T/2Q mice indicates that transcriptomic aspects of SCA1 pathogenesis differ between the cerebellum, brainstem, cerebral cortex, hippocampus, and striatum.


Assuntos
Ataxina-1 , Ataxias Espinocerebelares , Transcriptoma , Animais , Camundongos , Ataxina-1/genética , Ataxina-1/metabolismo , Encéfalo/metabolismo , Cerebelo/metabolismo , Modelos Animais de Doenças , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Fenótipo , Transporte Proteico/genética , Células de Purkinje/metabolismo , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/metabolismo
19.
bioRxiv ; 2023 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-36798410

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by an expanded polyglutamine tract in the widely expressed ATXN1 protein. To elucidate anatomical regions and cell types that underlie mutant ATXN1-induced disease phenotypes, we developed a floxed conditional knockout mouse model ( f-ATXN1 146Q/2Q ) having mouse Atxn1 coding exons replaced by human exons encoding 146 glutamines. F-ATXN1 146Q/2Q mice manifest SCA1-like phenotypes including motor and cognitive deficits, wasting, and decreased survival. CNS contributions to disease were revealed using ATXN1 146Q/2Q ; Nestin-Cre mice, that showed improved rotarod, open field and Barnes maze performances. Striatal contributions to motor deficits were examined using f-ATXN1 146Q/2Q ; Rgs9-Cre mice. Mice lacking striatal ATXN1 146Q/2Q had improved rotarod performance late in disease. Muscle contributions to disease were revealed in f-ATXN1 146Q/2Q ; ACTA1-Cre mice which lacked muscle pathology and kyphosis seen in f-ATXN1 146Q/2Q mice. Kyphosis was not improved in f-ATXN1 146Q/2Q ;Nestin - Cre mice. Thus, optimal SCA1 therapeutics will require targeting mutant ATXN1 toxic actions in multiple brain regions and muscle.

20.
Neurobiol Dis ; 48(3): 526-32, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22884877

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is an autosomal dominant neurodegenerative disease that results from a pathogenic glutamine-repeat expansion in the protein ataxin-1 (ATXN1). Although the functions of ATXN1 are still largely unknown, there is evidence to suggest that ATXN1 plays a role in regulating gene expression, the earliest process known to go awry in SCA1 mouse models. In this study, we show that ATXN1 reduces histone acetylation, a post-translational modification of histones associated with enhanced transcription, and represses histone acetyl transferase-mediated transcription. In addition, we find that depleting the Leucine-rich Acidic Nuclear Protein (LANP)-an ATXN1 binding inhibitor of histone acetylation-reverses aspects of SCA1 neuritic pathology.


Assuntos
Histona Acetiltransferases/biossíntese , Histonas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/patologia , Proteínas Nucleares/metabolismo , Acetilação , Animais , Ataxina-1 , Ataxinas , Regulação da Expressão Gênica/fisiologia , Técnicas de Introdução de Genes , Células HeLa , Humanos , Camundongos , Neurônios/metabolismo , Células PC12 , Processamento de Proteína Pós-Traducional , Ratos , Ataxias Espinocerebelares/metabolismo , Ataxias Espinocerebelares/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA