Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Transl Pediatr ; 13(6): 1001-1006, 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38984033

RESUMO

Background: Laminin-α2 (LAMA2) chain-deficient muscular dystrophy (LAMA2-MD) is the most common congenital muscular dystrophy (CMD) in the world. Its main manifestations are muscle weakness and hypotonia that occur after birth or at early infancy. Case Description: We reported a case of a 3-year-old and 6-month-old boy presented with delayed motor development, elevated creatine kinase (CK) levels, and abnormal white matter in the brain. Whole exome sequencing (WES) showed compound heterozygous variants of the LAMA2 gene. This case reports for the first time the compound heterozygous LAMA2 variants c.5476C>T (p.R1826*) (paternal inheritance) with c.2749 + 2dup (maternal inheritance), as both variants are interpreted as pathogenic/potentially pathogenic variants. Conclusions: This study reports a novel heterozygous variant, including two pathogenic variants in the LAMA2 gene, and highlights the effectiveness of highly efficient exome sequencing applying in patients with undefined CMDs.

2.
Mol Neurobiol ; 59(11): 6918-6933, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36053438

RESUMO

Epilepsy is a chronic brain disease that makes serious cognitive and motor retardation. Ion channels affect the occurrence of epilepsy in various ways, but the mechanisms have not yet been fully elucidated. Transient receptor potential melastain2 (TRPM2) ion channel is a non-selective cationic channel that can permeate Ca2+ and critical for epilepsy. Here, TRPM2 gene knockout mice were used to generate a chronic kindling epilepsy model by PTZ administration in mice. We found that TRPM2 knockout mice were more susceptible to epilepsy than WT mice. Furthermore, the neuronal excitability in the hippocampal CA1 region of TRPM2 knockout mice was significantly increased. Compared with WT group, there were no significant differences in the input resistance and after hyperpolarization of CA1 neurons in TRPM2 knockout mice. Firing adaptation rate of hippocampal CA1 pyramidal neurons of TRPM2 knockout mice was lower than that of WT mice. We also found that activation of Kv7 channel by retigabine reduced the firing frequency of action potential in the hippocampal pyramidal neurons of TRPM2 knockout mice. However, inhibiting Kv7 channel increased the firing frequency of action potential in hippocampal pyramidal neurons of WT mice. The data suggest that activation of Kv7 channel can effectively reduce epileptic seizures in TRPM2 knockout mice. We conclude that genetic knockout of TRPM2 in hippocampal CA1 pyramidal neurons may increase neuronal excitability by inhibiting Kv7 channel, affecting the susceptibility to epilepsy. These findings may provide a potential therapeutic target for epilepsy.


Assuntos
Região CA1 Hipocampal , Epilepsia , Células Piramidais , Canais de Cátion TRPM , Potenciais de Ação , Animais , Região CA1 Hipocampal/citologia , Epilepsia/genética , Técnicas de Inativação de Genes , Camundongos , Camundongos Knockout , Células Piramidais/fisiologia , Canais de Cátion TRPM/genética
3.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 50(2): 267-276, 2021 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-34137233

RESUMO

Transient receptor potential M2 (TRPM2) ion channel is a non-selective cationic channel that can permeate calcium ions, and plays an important role in neuroinflammation, ischemic reperfusion brain injury, neurodegenerative disease, neuropathic pain, epilepsy and other neurological diseases. In ischemic reperfusion brain injury, TRPM2 mediates neuronal death by modulating the different subunits of glutamate N-methyl-D-aspartic acid receptor in response to calcium/zinc signal. In Alzheimer's disease, TRPM2 is activated by reactive oxygen species generated by ß-amyloid peptide to form a malignant positive feedback loop that induces neuronal death and is involved in the pathological process of glial cells by promoting inflammatory response and oxidative stress. In epilepsy, the TRPM2-knockout alleviates epilepsy induced neuronal degeneration by inhibiting autophagy and apoptosis related proteins. The roles of TRPM2 channel in the pathogenesis of various central nervous system diseases and its potential drug development and clinical application prospects are summarized in this review.


Assuntos
Doenças Neurodegenerativas , Canais de Cátion TRPM , Peptídeos beta-Amiloides/metabolismo , Animais , Humanos , Neuroglia , Canais de Cátion TRPM/genética
4.
Int Immunopharmacol ; 87: 106824, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32731181

RESUMO

Neuroinflammation contributes to the generation of epileptic seizures and is associate with neuropathology and comorbidities. Transient receptor potential melastatin 2 (TRPM2) expresses in various cell types in the brain. It plays a pathological role in a wide range of neuroinflammatory diseases, but has yet been studied in epilepsy. Here, a temporal lobe epilepsy model was generated by pilocarpine administration in mice. At 24 h, knockout (KO) TRPM2 alleviated the level of neuroinflammation, showing a reduction of IL-1ß, TNF-α, CXCL2 and IL-6 mRNA production, NLRP3, ASC, and Caspase-1 protein expression and glial activation. Moreover, KO TRPM2 alleviated neurodegeneration, concurrent with reduced Beclin-1 and ATG5 protein expression. Later, KO TRPM2 ameliorated the epilepsy-induced psychological disorders, with improved performance in the open-field, Y maze and novel object recognition test. Together, these results suggest that TRPM2 facilitates epilepsy-related brain injury and may shed light on its potential as a therapeutic target for epilepsy-associated neuropathology and comorbidities.


Assuntos
Cognição , Epilepsia , Canais de Cátion TRPM , Animais , Comportamento Animal , Citocinas/genética , Modelos Animais de Doenças , Epilepsia/induzido quimicamente , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/patologia , Pilocarpina , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo
5.
Brain Res Bull ; 155: 48-60, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31794795

RESUMO

Epilepsy is one of the most common neurological conditions. Recent findings suggest that one of the mechanisms promoting its existence is calcium influx. The transient receptor potential melastatin type 2 channel (TRPM2) is a Ca2+-permeable cation channel that contributes to cell apoptosis; its possible signaling pathway is the PARP1/BNIP3/AIF/Endo G pathway that may be related to epilepsy. The aim of this study was to investigate the TRPM2 channel's involvement in epilepsy and how it works. We also explored the possible role of the TRPM2 channel on cognitive ability and emotion in epilepsy. To accomplish our goals, we used different animal epilepsy models to study the effect of the TRPM2 channel on epilepsy. The results showed that the knockout (KO) of the TRPM2 gene might play a protective role in epilepsy. Considering the advantages attributed to pentylenetetrazole (PTZ)-induced kindling mouse model, we used the model for the following assessments: 1. to observe changes in cognition and anxiety between wild type (WT) mice and TRPM2-KO mice with the recognition of new things trial and elevated plus-maze; 2. to determine the expression of apoptosis-associated proteins (PARP1, BNIP3, AIF, and Endo G) using Reverse transcription-polymerase chain reaction (RT-PCR) and Western blot; 3. to observe neurons pathologic damages and astrocyte activation in each group. The main findings of our study were: (a) TRPM2-KO had a protective effect on epilepsy; (b) TRPM2-KO improved spatial memory deficits overtime during epilepsy, but it did not improve anxiety; (c) the protective effect probably occurred via the PARP1 downstream signaling pathway; (d) TRPM2-KO could ameliorate epilepsy-induced hippocampal pathological damages and weaken astrocyte activation. These findings may provide a new approach for the treatment of epilepsy and early intervention.


Assuntos
Disfunção Cognitiva/fisiopatologia , Epilepsia/fisiopatologia , Canais de Cátion TRPM/fisiologia , Animais , Apoptose , Astrócitos/fisiologia , Modelos Animais de Doenças , Epilepsia/induzido quimicamente , Epilepsia/patologia , Epilepsia/psicologia , Hipocampo/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/patologia , Pentilenotetrazol/administração & dosagem , Canais de Cátion TRPM/genética
6.
Int Immunopharmacol ; 75: 105836, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31450153

RESUMO

Sepsis is one of the most significant challenges in intensive care units, which is associated with increased morbidity and mortality. Sepsis-associated encephalopathy (SAE) is a severe complication which can cause death and serious disabilities. Calcium signaling in astrocyte is essential for cellular activation and the potential resolution of infection or inflammation in SAE patients. The transient receptor potential melastatin 2 (TRPM2) channel has been identified as a unique fusion of a Ca2+-permeable nonselective cation channel, which plays an important role in inflammation and immune response. Because of its role as an oxidative stress sensor in astrocytes, we investigated the function of TRPM2 in inflammation mediators (interleukin (IL)-1ß, IL-6 and tumor necrosis factor (TNF)-α) release, Bcl-2/E1B-19 K-interacting protein 3 (BNIP3), apoptosis inducing factor (AIF) and Endonuclease G (Endo G) expression. We showed that TRPM2-KO mice, when intraperitoneally (i.p) injected with LPS, exhibited better neurologic assessment scores and decreased inflammatory injury in hippocampal neurons compared with wild-type (WT) mice. The absence of TRPM2 triggered less production of inflammatory mediators (IL-1ß, IL-6, TNF-α) and decreased apoptosis related proteins (BNIP3, AIF, Endo G) expressions in response to LPS induced sepsis. Furthermore, TRPM2-deficient astrocytes (transfected with TRPM2 siRNA) upon LPS stimulation also induced decreased IL-1ß, IL-6 and TNF-α level. Our data suggested that decreased production of inflammatory cytokines and apoptosis related proteins with TRPM2 deletion could regulate inflammatory stress and decrease inflammatory injury in hippocampal neurons, and consequently, ameliorate brain disorder.


Assuntos
Astrócitos/imunologia , Encefalopatias/imunologia , Citocinas/imunologia , Sepse/imunologia , Canais de Cátion TRPM/imunologia , Animais , Apoptose , Fator de Indução de Apoptose/imunologia , Encefalopatias/etiologia , Encefalopatias/patologia , Citocinas/genética , Endodesoxirribonucleases/imunologia , Hipocampo/imunologia , Hipocampo/patologia , Lipopolissacarídeos , Masculino , Proteínas de Membrana/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mitocondriais/imunologia , Sepse/induzido quimicamente , Sepse/complicações , Sepse/patologia , Canais de Cátion TRPM/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...