Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Cardiol ; 345: 98-104, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34710491

RESUMO

Programmed cell death 5 (PDCD5) is a tumor suppressor gene that regulates the cell cycle, apoptosis and immune responses. However, the physiological function of Pdcd5 in cardiac aging remains unknown. We find that Pdcd5 mRNA and protein levels were significantly increased in the heart of mice with age. Therefore, we hypothesize that Pdcd5 regulates cardiac aging. To test the hypothesis, we generated muscle-specific Pdcd5-deficient mice. Mature adult Pdcd5-deficient mice had normal cardiac morphology and function. In naturally aged mice, Pdcd5 deficiency alleviated age-related cardiac phenotypes including reduced fibrosis and suppressed cardiomyocyte hypertrophy. Moreover, muscle-specific Pdcd5 deficiency attenuated cellular senescence in the heart as demonstrated by decreased number of senescence-associated ß-galactosidase-positive cells, diminished p53, p21 and p16 expression, and reduced the senescence-associated secretory phenotype. Apoptotic cell death was reduced by Pdcd5 deficiency in the heart as revealed by terminal deoxynucleotidyl transferase dUTP nick end labeling assay, which was coincident with diminished Bcl-2-associated X protein, and enhanced B-cell lymphoma 2 and X-linked inhibitor of apoptosis protein expression. Mitochondrial quality in cardiomyocytes was improved by Pdcd5 deficiency through increased Parkin-mediated mitophagy. In addition, Pdcd5 deficiency alleviated doxorubicin-induced premature cellular senescence and cardiac aging. Furthermore, Pdcd5 protein abundance was significantly correlated with p53 protein abundance, and Pdcd5 interacted with p53 in the heart. Taken together, our results reveal that Pdcd5 deficiency attenuates cardiac aging by reducing cellular senescence and apoptosis, and increasing Parkin-mediated mitophagy, likely through p53. Pdcd5 is a novel regulator of cardiac aging and a potential therapeutic target.


Assuntos
Envelhecimento , Senescência Celular , Envelhecimento/genética , Animais , Apoptose , Proteínas Reguladoras de Apoptose/genética , Camundongos , Mitofagia , Miócitos Cardíacos , Proteínas de Neoplasias
2.
Biomolecules ; 11(2)2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33567640

RESUMO

Chimeric antigen receptor (CAR) T therapy represents a form of immune cellular therapy with clinical efficacy and a specific target. A typical chimeric antigen receptor (CAR) construct consists of an antigen binding domain, a transmembrane domain, and a cytoplasmic domain. Nanobodies have been widely applied as the antigen binding domain of CAR-T due to their small size, optimal stability, high affinity, and manufacturing feasibility. The nanobody-based CAR structure has shown a proven function in more than ten different tumor-specific targets. After being transduced in Jurkat cells, natural killer cells, or primary T cells, the resulting nanobody-based CAR-T or CAR-NK cells demonstrate anti-tumor effects both in vitro and in vivo. Interestingly, anti-BCMA CAR-T modulated by a single nanobody or bi-valent nanobody displays comparable clinical effects with that of single-chain variable fragment (scFv)-modulated CAR-T. The application of nanobodies in CAR-T therapy has been well demonstrated from bench to bedside and displays great potential in forming advanced CAR-T for more challenging tasks.


Assuntos
Imunoterapia Adotiva/métodos , Anticorpos de Domínio Único , Humanos , Células Jurkat , Células Matadoras Naturais/imunologia , Anticorpos de Cadeia Única/imunologia
3.
Xenotransplantation ; 26(5): e12526, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31127671

RESUMO

BACKGROUND: Porcine vascular endothelial cells are a major participant in xenograft rejection. The Toll-like receptor 2 (TLR2) pathway plays an important role in both innate and adaptive immunity. The specific role of TLR2 in the response to a xenograft has not been reported. Whether the TLR2 pathway in pig vascular endothelial cells is involved in acute rejection needs to be investigated, and the mechanism is explored. METHODS: We used a modified antibody-dependent complement-mediated cytotoxicity (ADCC) assay to conduct in vitro experiments. In porcine iliac artery endothelial cells (PIECs), siRNA was used to knock down the expression of TLR2, CXCL8, and CCL2. The effect of human serum or inactivated human serum on the expression of TLR2 was analyzed by real-time PCR and Western blotting, and transwell assays were used to assess the chemotactic efficiency of PIECs on human monocyte-macrophages (THP-1 cells) and human neutrophils. The downstream signaling pathways activated by human serum were detected by Western blotting, and the regulation of proinflammatory chemokines and cytokines by TLR2 signaling was assessed by real-time PCR and ELISA. RESULTS: TLR2 was significantly upregulated in PIECs after exposure to human serum, and porcine proinflammatory chemokines, CXCL8 and CCL2, were induced, at least partially, in a TLR2-dependent pattern; the upregulated chemokines participated in the chemotaxis of human neutrophils and THP-1 cells across the species barrier. CONCLUSIONS: (i) TLR2 is significantly upregulated in PIECs by human serum, (ii) the elevated TLR2 participates in the chemotaxis of inflammatory cells through the secretion of chemokine CCL2 and CXCL8, and (iii) blockade of TLR2 would be beneficial for xenograft survival.


Assuntos
Células Endoteliais/imunologia , Rejeição de Enxerto/imunologia , Artéria Ilíaca/imunologia , Receptor 2 Toll-Like/imunologia , Transplante Heterólogo , Animais , Biomarcadores/metabolismo , Western Blotting , Células Endoteliais/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Artéria Ilíaca/metabolismo , Técnicas In Vitro , Reação em Cadeia da Polimerase em Tempo Real , Suínos , Receptor 2 Toll-Like/metabolismo
4.
Xenotransplantation ; 26(4): e12522, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31077480

RESUMO

Monitoring for immune rejection is crucial for long-term survival of pig xenografts. Circulating DNA is a promising non-invasive biomarker for either organ injury or response to therapy. In this study, circulating pig-specific DNA (cpsDNA) was monitored during xenograft rejection. Potential targets of cpsDNA were selected by in silico analysis, and species specificity of selected primers was confirmed by PCR. Subsequently, cpsDNA as a biomarker was evaluated using a complement-dependent cytotoxicity (CDC) assay in vitro. Then, early diagnosis and response to rapamycin were assessed by an in vivo imaging model of pig-to-mouse cell transplantation. Finally, cpsDNA was monitored in a pig-to-monkey artery patch transplantation model. The results showed that (a) a method of cpsDNA quantitation was established for application in mouse and nonhuman primate models; (b) cpsDNA reflected CDC in vitro; (c) cpsDNA in vivo mirrored xenograft rejection, and correlated with xenograft loss in pig-to-mouse cell transplantation; (d) cpsDNA was significantly reduced when rapamycin was administered; and (e) dynamic cpsDNA was detectable in pig-to-monkey artery patch transplantation. In conclusion, measurement of cpsDNA could prove to be a less invasive, but more specific and sensitive low-cost biomarker enabling monitoring of xenograft rejection and the response to immunosuppressive therapy.


Assuntos
DNA/sangue , Rejeição de Enxerto/sangue , Porco Miniatura/genética , Animais , Anticorpos Heterófilos/sangue , Biomarcadores , Células Cultivadas , Primers do DNA , Células Endoteliais/transplante , Feminino , Genes Reporter , Rejeição de Enxerto/tratamento farmacológico , Rejeição de Enxerto/patologia , Xenoenxertos , Humanos , Artéria Ilíaca/citologia , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sirolimo/uso terapêutico , Especificidade da Espécie , Suínos
5.
Cardiovasc Res ; 115(8): 1320-1331, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30428088

RESUMO

AIMS: WD40 repeat and FYVE domain containing 3 (WDFY3) is an adaptor protein involved in selective degradation of protein aggregates by autophagy. Recent studies have revealed that Wdfy3 is critical in the regulation of brain development and osteoclastogenesis in vivo. However, the function of Wdfy3 in cardiac development remains completely unknown. In this study, we explore the role of Wdfy3 in cardiac morphogenesis using Wdfy3-deficient mice. METHODS AND RESULTS: Wdfy3 was expressed in the developing heart in mice and peaked at embryonic day 12.5 (E12.5). Loss of Wdfy3 in mice led to embryonic and neonatal lethality. Wdfy3-deficient mice displayed various congenital heart defects including membranous ventricular septal defect (VSD), aortic overriding (AO), double outlet right ventricle (DORV), thinning of ventricular wall, ventricular dilation, and disorganized ventricular trabeculation at E14.5. Cell proliferation was reduced in the hearts from Wdfy3-deficient mice at E12.5 and E14.5, which was associated with enhanced p21 expression. Cardiomyocyte differentiation was diminished as demonstrated by reduced Myh6 and MLC2v in Wdfy3-deficient mice at E14.5. In addition, Nkx2-5 and Mef2c, two cardiac transcription factors regulating cardiomyocyte differentiation, were decreased in Wdfy3-deficient mice at E14.5. Apoptotic cell death remained unaltered. These data suggest that reduced cell proliferation and cardiomyocyte differentiation contribute to cardiac defects in Wdfy3-deficient mice. Mechanistically, loss of Wdfy3 led to a reduction in protein levels of Notch 1 intracellular domain and its downstream targets Hes1 and Hey1, which was accompanied with enhanced full-length Notch1 protein levels. In vitro luciferase assay showed that Wdfy3 deficiency induced activity of p21 promoter, while diminished activity of Hes1 promoter through modulation of Notch1 signalling. Moreover, Wdfy3 was co-localized with Notch1 in primary embryonic cardiomyocytes. Endogenous Wdfy3 physically interacted with full-length Notch1 in the developing heart. These results suggest that Notch1 signalling is perturbed in the hearts from Wdfy3-deficient mice. No alteration of autophagy was detected in the hearts from Wdfy3-deficient mice. CONCLUSION: Taken together, our data suggest that Wdfy3 plays an essential role in cardiac development, which may be mediated by modulation of Notch1 signalling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Relacionadas à Autofagia/metabolismo , Cardiopatias Congênitas/metabolismo , Coração/embriologia , Miócitos Cardíacos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose , Proteínas Relacionadas à Autofagia/deficiência , Proteínas Relacionadas à Autofagia/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Coração/fisiopatologia , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese , Miócitos Cardíacos/patologia , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transdução de Sinais , Fatores de Transcrição HES-1/genética , Fatores de Transcrição HES-1/metabolismo
6.
Biomed Res Int ; 2018: 2983908, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29750148

RESUMO

The pig is an important source of meat production and provides a valuable model for certain human diseases. MicroRNA (miRNA), which is noncoding RNA and regulates gene expression at the posttranscriptional level, plays a critical role in various biological processes. Studies on identification and function of mature miRNAs in multiple pig tissues are increasing, yet the literature is limited. Therefore, we reviewed current research to determine the miRNAs expressed in specific pig tissues that are involved in carcass values (including muscle and adipocytes), reproduction (including pituitary, testis, and ovary), and development of some solid organs (e.g., brain, lung, kidney, and liver). We also discuss the possible regulating mechanisms of miRNA. Finally, as pig organs are suitable candidates for xenotransplantation, biomarkers of their miRNA in xenotransplantation were evaluated.


Assuntos
MicroRNAs/genética , Animais , Biomarcadores/metabolismo , Regulação da Expressão Gênica/genética , Humanos , Reprodução/genética , Suínos , Transcriptoma/genética , Transplante Heterólogo/métodos
7.
Sci Rep ; 7: 43251, 2017 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-28225086

RESUMO

Doxorubicin (DOX) is an effective chemotherapeutic drug in the treatment of various types of cancers. However, its clinical application has been largely limited by potential development of cardiotoxicity. Previously we have shown that ultra-violet radiation resistance-associated gene (UVRAG), an autophagy-related protein, is essential for the maintenance of autophagic flux in the heart under physiological conditions. Here, we sought to determine the role of UVRAG-mediated autophagy in DOX-induced cardiotoxicity. Mouse models of acute or chronic DOX-induced cardiotoxicity were established. UVRAG deficiency exacerbated DOX-induced mortality and cardiotoxicity manifested by increased cytoplasmic vacuolization, enhanced collagen accumulation, elevated serum activities of lactate dehydrogenase and myocardial muscle creatine kinase, higher ROS levels, aggravated apoptosis and more depressed cardiac function. Autophagic flux was impaired in DOX-induced cardiotoxicity. UVRAG deficiency aggravated impaired autophagic flux in DOX-induced cardiotoxicity. Intermittent fasting restored autophagy and ameliorated pathological alterations of DOX-induced cardiotoxicity. Collectively, our data suggest that UVRAG deficiency exacerbates DOX-induced cardiotoxicity, at least in part, through aggravation of DOX-induced impaired autophagic flux. Intermittent fasting, which restores blunted autophagic flux and ameliorates pathology in the mouse models of DOX-induced cardiotoxicity, may be used as a potential preventive or therapeutic approach for DOX cardiotoxicity.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Cardiotoxicidade , Doxorrubicina/efeitos adversos , Proteínas Supressoras de Tumor/deficiência , Animais , Camundongos , Análise de Sobrevida
8.
J Cardiovasc Pharmacol ; 65(3): 252-61, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25502307

RESUMO

: Rubicon has been suggested to suppress autophagosome maturation by negatively regulating PI3KC3/Vps34 activity. However, the physiological function of Rubicon remains elusive. We hypothesized that Rubicon deficiency enhances autophagic flux in the heart and affects cardiac function. Rubicon knockout (KO) mice were generated by piggyBac transposition. Loss of Rubicon was demonstrated at both mRNA and protein levels. Rubicon KO mice were born in Mendelian ratios. Autophagic flux, assessed by bafilomycin A1-induced changes in LC3 II protein abundance, was enhanced in the heart of Rubicon KO mice compared with wild-type (WT) controls. Hematoxylin-eosin staining and picrosirius red staining showed that Rubicon KO mice exhibited normal baseline cardiac morphology. Echocardiography revealed that ejection fraction and fractional shortening, 2 indices of cardiac function, were comparable between Rubicon KO mice at 2, 8, and 12 months of age (n = 6-8 for each age group) and the corresponding WT controls (n = 6-8 for each age group). In a mouse model of lipopolysaccharide (LPS)-induced sepsis, the survival time of LPS-treated Rubicon KO mice (n = 10) was prolonged compared with LPS-treated WT controls (n = 11). Echocardiography revealed that Rubicon deficiency partially normalized LPS-induced reduction in stroke volume and cardiac output 12 hours after LPS administration compared with LPS-treated WT controls (n = 6 for each group). Autophagic flux was enhanced in Rubicon-deficient hearts 12 hours after LPS treatment compared with LPS-treated WT controls. Real-time quantitative polymerase chain reaction suggested that proinflammatory cytokine expression was not significantly different between LPS-treated Rubicon KO mice and WT controls (n = 3 for each group). Our data demonstrate for the first time that Rubicon deficiency enhances autophagic flux in the heart and protects mice from lethality and reduction in stroke volume induced by LPS.


Assuntos
Autofagia , Cardiopatias/prevenção & controle , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Lipopolissacarídeos , Contração Miocárdica , Miocárdio/metabolismo , Sepse/metabolismo , Volume Sistólico , Animais , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Genótipo , Cardiopatias/genética , Cardiopatias/metabolismo , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos Knockout , Miocárdio/patologia , Fenótipo , RNA Mensageiro/metabolismo , Sepse/induzido quimicamente , Sepse/genética , Sepse/patologia , Sepse/fisiopatologia , Fatores de Tempo
9.
Cardiovasc Res ; 101(1): 48-56, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24081163

RESUMO

AIMS: Ultraviolet irradiation resistance-associated gene (UVRAG) is a tumour suppressor candidate that regulates cell autophagy and endocytosis. However, the in vivo function of UVRAG remains poorly understood. We sought to determine the physiological role of UVRAG in the heart. METHODS AND RESULTS: We characterized mice with disruption of the UVRAG gene by piggyBac (PB) transposon insertion. PB construct was inserted into intron 14 of the UVRAG gene and disruption of UVRAG transcript was confirmed by reverse transcript-polymerase chain reaction. Immunoblotting revealed that UVRAG was deficient in multiple tissues. Autophagic flux was attenuated in UVRAG-deficient (UVRAG(PB/PB)) mouse embryonic fibroblasts. In UVRAG-deficient hearts, autophagosomes were accumulated and autophagic flux, assessed as the increased protein abundance of LC3 II in chloroquine-treated animals, was impaired. UVRAG-deficient mice were viable, fertile, and developmentally normal. However, they developed age-related cardiomyopathy associated with compromised cardiac function. In addition, inflammatory response was enhanced in UVRAG-deficient hearts. CONCLUSION: Collectively, our findings suggest that UVRAG is essential for the regulation of autophagy and maintenance of cardiac function.


Assuntos
Autofagia , Cardiomiopatias/etiologia , Coração/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Envelhecimento/fisiologia , Animais , Cardiomiopatias/metabolismo , Células Cultivadas , Feminino , Inflamação/etiologia , Inflamação/metabolismo , Masculino , Camundongos , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA