Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(37): e2402817121, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39236246

RESUMO

Autophagy of glycogen (glycophagy) is crucial for the maintenance of cellular glucose homeostasis and physiology in mammals. STBD1 can serve as an autophagy receptor to mediate glycophagy by specifically recognizing glycogen and relevant key autophagic factors, but with poorly understood mechanisms. Here, we systematically characterize the interactions of STBD1 with glycogen and related saccharides, and determine the crystal structure of the STBD1 CBM20 domain with maltotetraose, uncovering a unique binding mode involving two different oligosaccharide-binding sites adopted by STBD1 CBM20 for recognizing glycogen. In addition, we demonstrate that the LC3-interacting region (LIR) motif of STBD1 can selectively bind to six mammalian ATG8 family members. We elucidate the detailed molecular mechanism underlying the selective interactions of STBD1 with ATG8 family proteins by solving the STBD1 LIR/GABARAPL1 complex structure. Importantly, our cell-based assays reveal that both the STBD1 LIR/GABARAPL1 interaction and the intact two oligosaccharide binding sites of STBD1 CBM20 are essential for the effective association of STBD1, GABARAPL1, and glycogen in cells. Finally, through mass spectrometry, biochemical, and structural modeling analyses, we unveil that STBD1 can directly bind to the Claw domain of RB1CC1 through its LIR, thereby recruiting the key autophagy initiation factor RB1CC1. In all, our findings provide mechanistic insights into the recognitions of glycogen, ATG8 family proteins, and RB1CC1 by STBD1 and shed light on the potential working mechanism of STBD1-mediated glycophagy.


Assuntos
Família da Proteína 8 Relacionada à Autofagia , Autofagia , Glicogênio , Animais , Humanos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Autofagia/fisiologia , Família da Proteína 8 Relacionada à Autofagia/metabolismo , Família da Proteína 8 Relacionada à Autofagia/genética , Família da Proteína 8 Relacionada à Autofagia/química , Sítios de Ligação , Cristalografia por Raios X , Glicogênio/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/genética , Modelos Moleculares , Ligação Proteica
2.
Hum Mol Genet ; 30(23): 2255-2262, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34240207

RESUMO

Genetic mutations in the lamin A/C gene (LMNA) have been linked to cardiomyopathy. Different mutational sites exhibit different clinical manifestations and prognoses. Herein, we identified a novel LMNA frameshift mutation, p.P485Tfs*67, from a patient with early-onset atrial disease. To verify the pathogenicity of this variation, a transgenic zebrafish model was constructed, which demonstrated that adult zebrafish with the LMNA mutation showed an abnormal ECG and impaired myocardial structure. Our study suggests the atrial pathogenicity of the LMNA-P485Tfs mutation, which is helpful to understand the function of the Ig-like domain of lamin A/C.


Assuntos
Mutação da Fase de Leitura , Átrios do Coração/metabolismo , Átrios do Coração/fisiopatologia , Cardiopatias/diagnóstico , Cardiopatias/etiologia , Lamina Tipo A/genética , Adulto , Animais , Análise Mutacional de DNA , Modelos Animais de Doenças , Ecocardiografia , Eletrocardiografia , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Linhagem , Sequenciamento do Exoma , Peixe-Zebra
3.
Front Biosci (Landmark Ed) ; 29(5): 201, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38812314

RESUMO

BACKGROUND: Ibrutinib could increase the risk of atrial fibrillation (AF) in chronic lymphocytic leukemia (CLL) patients. However, the precise mechanism underlying ibrutinib-induced AF remains incompletely elucidated. METHODS: We investigated the proportion of ibrutinib-treated CLL patients with new-onset AF. Optical mapping was conducted to reveal the proarrhythmic effect of ibrutinib on HL-1 cells. Fluorescence staining and western blot were used to compare connexins 43 and 40 expression in ibrutinib-treated and control groups. To identify autophagy phenotypes, we used western blot to detect autophagy-related proteins, transmission electron microscopy to picture autophagosomes, and transfected mCherry-GFP-LC3 virus to label autophagosomes and lysosomes. Hydroxychloroquine as an autophagy inhibitor was administered to rescue ibrutinib-induced Cx43 and Cx40 degradation. RESULTS: About 2.67% of patients developed atrial arrhythmias after ibrutinib administration. HL-1 cells treated with ibrutinib exhibited diminished conduction velocity and a higher incidence of reentry-like arrhythmias compared to controls. Cx43 and Cx40 expression reduced along with autophagy markers increased in HL-1 cells treated with ibrutinib. Inhibiting autophagy upregulated Cx43 and Cx40. CONCLUSIONS: The off-target effect of ibrutinib on the PI3K-AKT-mTOR signaling pathway caused connexin degradation and atrial arrhythmia via promoting autophagy. CLINICAL TRIAL REGISTRATION: ChiCTR2100046062, https://clin.larvol.com/trial-detail/ChiCTR2100046062.


Assuntos
Adenina , Fibrilação Atrial , Autofagia , Conexina 43 , Conexinas , Fosfatidilinositol 3-Quinases , Piperidinas , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Serina-Treonina Quinases TOR , Humanos , Adenina/análogos & derivados , Adenina/farmacologia , Adenina/efeitos adversos , Serina-Treonina Quinases TOR/metabolismo , Autofagia/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piperidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Conexina 43/metabolismo , Conexina 43/genética , Feminino , Fibrilação Atrial/metabolismo , Fibrilação Atrial/induzido quimicamente , Conexinas/metabolismo , Conexinas/genética , Masculino , Idoso , Pessoa de Meia-Idade , Proteína alfa-5 de Junções Comunicantes , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/induzido quimicamente
4.
Heart Rhythm ; 2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39284398

RESUMO

BACKGROUND: Postmenopausal women with atrial fibrillation (AF) exhibit a higher level of atrial fibrosis and a higher recurrence rate after ablation compared to men. However, the underlying mechanism remains unclear. OBJECTIVE: To investigate the mechanism through which menopause promotes atrial fibrosis. METHODS: In a prospective cohort of women with AF, regression analyses were conducted to assess the relationship between low-voltage area (LVA) and sex hormone levels. CREM-IbΔC-X mice, a spontaneous AF model, underwent bilateral ovariectomy (OVX). Electrocardiograms, echocardiograms, and Masson staining were performed. FSH stimulation was applied in male mice for three months. OVX was also applied in an angiotensin II (Ang II) induced pressure overload mouse model, following programmed electrical stimulation and structural analyses. Bulk RNA sequencing (RNA-seq) was performed to elucidate potential mechanisms. RESULTS: Women demonstrated a significantly higher LVA burden than men (P<0.001). A positive correlation was observed between LVA burden and FSH level (P=0.002). Mice in the OVX group exhibited a significantly higher incidence of AF (P=0.040) and atrial fibrosis (P=0.021) compared to the Sham group, which could be attenuated by AAV-siFshr. In male CREM-IbΔC-X mice, FSH stimulation promoted the occurrence of AF (P=0.035) and atrial fibrosis (P=0.002). In Ang II-induced female mice, OVX prompted atrial fibrosis, increased AF inducibility, and shortened atrial effective refractory period, which could be attenuated with knockdown of Fshr. RNA-seq indicated mitochondrial dysfunction. CONCLUSION: Postmenopausal women exhibited a higher LVA burden than men, which was positively correlated with FSH level. FSH promoted atrial fibrosis through oxidative stress.

5.
Ann Clin Transl Neurol ; 10(7): 1136-1145, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37218344

RESUMO

OBJECTIVE: A rapid and accurate forecast for the early prognosis of ICH patients is challenging. This study investigated whether heart rate variability (HRV) and skin sympathetic nerve activity (SKNA) could prognosticate poor neurological outcomes in ICH patients. METHODS: Between November 2020 and November 2021, we studied 92 spontaneous ICH patients in the First Affiliated Hospital of Nanjing Medical University. Glasgow Outcome Scale (GOS) score at 2 weeks after the ICH was used to categorize patients into good and poor outcome groups. The modified Rankin Scale (mRS) assessed patients' ability to live independently for 1 year. We utilized a portable high-frequency electrocardiogram (ECG) recording system to record the HRV and SKNA information in ICH patients and control participants. RESULTS: 77 patients were eligible for the prediction of neurological outcome and were allocated into the good (n = 22) or poor (n = 55) outcome groups based on the GOS grade. In univariate logistic regression analysis, significant variables that could differentiate the outcomes were age, hypertension, tracheal intubation, Glasgow Coma Scale (GCS) score, existing intraventricular hemorrhage, white blood cells, neutrophil, lnVLF, lnTP, and aSKNA. Variables in the best fit multivariable logistic regression model were age, hypertension, GCS score, neutrophils, and aSKNA. The GCS score was the only independent risk factor for poor outcomes. At 30 days and 1 year of follow-up, patients with lower aSKNA had poor outcomes. INTERPRETATION: ICH patients had reduced aSKNA, which could be a prognostic indicator. A lower aSKNA suggested a worse prognosis. The present data indicate that ECG signals could be helpful for prognosticating ICH patients.


Assuntos
Hemorragia Cerebral , Hipertensão , Humanos , Hemorragia Cerebral/diagnóstico , Prognóstico , Biomarcadores , Escala de Coma de Glasgow
6.
Toxicol Sci ; 189(2): 216-224, 2022 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-35866629

RESUMO

Febuxostat is a xanthine oxidase inhibitor used to reduce the formation of uric acid and prevent gout attacks. Previous studies have suggested that febuxostat was associated with a higher risk of cardiovascular events, including atrial fibrillation, compared with allopurinol, another anti-hyperuricemia drug. Whereas in our clinical practice, we identified 2 cases of febuxostat-associated ventricular tachycardia (VT) events. The proarrhythmogenic effects of febuxostat on human cardiomyocytes and underlined mechanisms remain poorly understood. In this study, we employed real-time cell analysis and calcium transient to investigate the effects of febuxostat on the cytotoxicity and electrophysiology properties of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Up to 10 µM febuxostat treatment did not show toxicity to cell viability. However, 48-h febuxostat exposure generated dose-dependent increased irregular calcium transients and decreased calcium transient amplitude. Furthermore, RNA-seq analysis indicated that the MAPK signaling pathway was enriched in the febuxostat-treated group, especially the protein kinases c-Jun N-terminal kinase (JNK). Western blotting of 3 main protein kinases demonstrated that JNK activation is related to febuxostat-induced arrhythmia rather than extracellular signal regulated kinases (ERK) or p38. The dysfunctional calcium dynamics of febuxostat-treated hiPSC-CMs could be ameliorated by SP600125, the inhibitor of JNK. In conclusion, our study demonstrated that febuxostat increases the predisposition to ventricular arrhythmia by dysregulating calcium dynamics.


Assuntos
Febuxostat , Células-Tronco Pluripotentes Induzidas , Alopurinol/metabolismo , Alopurinol/toxicidade , Arritmias Cardíacas/metabolismo , Cálcio/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Febuxostat/metabolismo , Febuxostat/toxicidade , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Miócitos Cardíacos , Ácido Úrico/metabolismo , Ácido Úrico/farmacologia , Xantina Oxidase/metabolismo , Xantina Oxidase/farmacologia
7.
Stem Cell Res ; 64: 102909, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36095892

RESUMO

The iPSC line was generated from the peripheral blood mononuclear cells (PBMCs) from a 53-year-old female patient carrying the LMNA gene mutation (c.1304_1307dup) diagnosed with atrial fibrillation and paroxysmal ventricular tachycardia. Through comprehensive detection, it was verified that the cell line had the LMNA gene mutation, normal karyotype, and the potential to differentiate into the three germ layers. This cell line may reveal potential therapeutic targets for atrial and ventricular arrhythmias caused by LMNA mutations.


Assuntos
Fibrilação Atrial , Células-Tronco Pluripotentes Induzidas , Taquicardia Ventricular , Feminino , Humanos , Pessoa de Meia-Idade , Células-Tronco Pluripotentes Induzidas/metabolismo , Fibrilação Atrial/genética , Mutação da Fase de Leitura , Leucócitos Mononucleares/metabolismo , Mutação/genética , Taquicardia Ventricular/genética , Lamina Tipo A/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA