Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Nucleic Acids Res ; 49(4): 1972-1986, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33503260

RESUMO

Maintenance of stem-cell identity requires proper regulation of enhancer activity. Both transcription factors OCT4/SOX2/NANOG and histone methyltransferase complexes MLL/SET1 were shown to regulate enhancer activity, but how they are regulated in embryonic stem cells (ESCs) remains further studies. Here, we report a transcription factor BACH1, which directly interacts with OCT4/SOX2/NANOG (OSN) and MLL/SET1 methyltransferase complexes and maintains pluripotency in mouse ESCs (mESCs). BTB domain and bZIP domain of BACH1 are required for these interactions and pluripotency maintenance. Loss of BACH1 reduced the interaction between NANOG and MLL1/SET1 complexes, and decreased their occupancy on chromatin, and further decreased H3 lysine 4 trimethylation (H3K4me3) level on gene promoters and (super-) enhancers, leading to decreased enhancer activity and transcription activity, especially on stemness-related genes. Moreover, BACH1 recruited NANOG through chromatin looping and regulated remote NANOG binding, fine-tuning enhancer-promoter activity and gene expression. Collectively, these observations suggest that BACH1 maintains pluripotency in ESCs by recruiting NANOG and MLL/SET1 complexes to chromatin and maintaining the trimethylated state of H3K4 and enhancer-promoter activity, especially on stemness-related genes.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Células-Tronco Embrionárias/metabolismo , Elementos Facilitadores Genéticos , Histona-Lisina N-Metiltransferase/metabolismo , Proteína Homeobox Nanog/metabolismo , Regiões Promotoras Genéticas , Animais , Fatores de Transcrição de Zíper de Leucina Básica/química , Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Linhagem Celular , Células Cultivadas , Cromatina/metabolismo , Histonas/metabolismo , Camundongos , Fator 3 de Transcrição de Octâmero/metabolismo , Domínios Proteicos , Fatores de Transcrição SOXB1/metabolismo
2.
Inorg Chem ; 61(21): 8135-8143, 2022 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-35588219

RESUMO

A series of theoretical methods, including density functional theory, multiconfiguration molecular orbital theory, and ab initio valence bond theory, are devoted to understanding the metal-ligand bonds in M-BP (BP = biphenyl; M = Sc, Y, or La) complexes. Different from most transition metal-BP complexes, the most stable metal-biphenyl conformers are not half-sandwich but clamshell. Energy decomposition analysis results reveal that the M-BP bonds in the clamshell conformers possess extra-large orbital relaxation. According to the wave function analysis, 2-fold donations and 2-fold back-donations exist in the clamshell M-BP bonds. The back-donations from M to BP are quite strong, while donations from BP to M are quite weak. Our work improves our understanding of the metal-ligand bonds, which can be considered as the "reversed" Dewar-Chatt-Duncanson model.

3.
Am J Transplant ; 20(10): 2755-2767, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32277602

RESUMO

This study aimed to determine the mechanism of isogeneic-induced pluripotent stem cells (iPSCs) homing to vascular transplants and their therapeutic effect on chronic allogeneic vasculopathy. We found that integrin ß1 (Intgß1) was the dominant integrin ß unit in iPSCs that mediates the adhesion of circulatory and endothelial cells (ECs). Intgß1 knockout or Intgß1-siRNAs inhibit iPSC adhesion and migration across activated endothelial monolayers. The therapeutic effects of the following were examined: iPSCs, Intgß1-knockout iPSCs, iPSCs transfected with Intgß1-siRNAs or nontargeting siRNAs, iPSC-derived ECs, iPSC-derived ECs simultaneously overexpressing Intgα4 and Intgß1, iPSCs precultured in endothelial medium for 3 days (endothelial-prone stem cells), primary aortic ECs, mouse embryonic fibroblasts, and phosphate-buffered saline (control). The cells were administered every 3 days for a period of 8 weeks. iPSCs, iPSCs transfected with nontargeting siRNAs, and endothelial-prone stem cells selectively homed on the luminal surface of the allografts, differentiated into ECs, and decreased neointimal proliferation. Through a single administration, we found that iPSCs trafficked to allograft lesions, differentiated into ECs within 1 week, and survived for 4-8 weeks. The therapeutic effect of a single administration was moderate. Thus, Intgß1 and pluripotency are essential for iPSCs to treat allogeneic vasculopathy.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Pluripotentes Induzidas , Animais , Diferenciação Celular , Células Endoteliais , Fibroblastos , Integrina beta1 , Camundongos
4.
Circ Res ; 122(1): 88-96, 2018 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-29018036

RESUMO

RATIONALE: The effectiveness of transplanted, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) for treatment of ischemic myocardial injury is limited by the exceptionally low engraftment rate. OBJECTIVE: To determine whether overexpression of the cell cycle activator CCND2 (cyclin D2) in hiPSC-CMs can increase the graft size and improve myocardial recovery in a mouse model of myocardial infarction by increasing the proliferation of grafted cells. METHODS AND RESULTS: Human CCND2 was delivered to hiPSCs via lentiviral-mediated gene transfection. In cultured cells, markers for cell cycle activation and proliferation were ≈3- to 7-folds higher in CCND2-overexpressing hiPSC-CMs (hiPSC-CCND2OECMs) than in hiPSC-CMs with normal levels of CCND2 (hiPSC-CCND2WTCMs; P<0.01). The pluripotent genes (Oct 4, Sox2, and Nanog) decrease to minimal levels and undetectable levels at day 1 and 10 after differentiating to CMs. In the mouse myocardial infarction model, cardiac function, infarct size, and the number of engrafted cells were similar at week 1 after treatment with hiPSC-CCND2OECMs or hiPSC-CCND2WTCMs but was about tripled in hiPSC-CCND2OECM-treated than in hiPSC-CCND2WTCM-treated animals at week 4 (P<0.01). The cardiac function and infarct size were significantly better in both cell treatment groups' hearts than in control hearts, which was most prominent in hiPSC-CCND2OECM-treated animals (P<0.05, each). No tumor formation was observed in any hearts. CONCLUSIONS: CCND2 overexpression activates cell cycle progression in hiPSC-CMs that results in a significant enhanced potency for myocardial repair as evidenced by remuscularization of injured myocardium. This left ventricular muscle regeneration and increased angiogenesis in border zone are accompanied by a significant improvement of left ventricular chamber function.


Assuntos
Ciclina D2/biossíntese , Ventrículos do Coração/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Miocárdio/metabolismo , Miócitos Cardíacos/fisiologia , Regeneração/fisiologia , Animais , Células Cultivadas , Ciclina D2/genética , Expressão Gênica , Células HEK293 , Ventrículos do Coração/lesões , Ventrículos do Coração/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Camundongos , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Isquemia Miocárdica/terapia , Miócitos Cardíacos/transplante
5.
J Mol Cell Cardiol ; 117: 49-61, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29452156

RESUMO

PRKAG2 cardiac syndrome is a distinct form of human cardiomyopathy characterized by cardiac hypertrophy, ventricular pre-excitation and progressive cardiac conduction disorder. However, it remains unclear how mutations in the PRKAG2 gene give rise to such a complicated disease. To investigate the underlying molecular mechanisms, we generated disease-specific hiPSC-derived cardiomyocytes from two brothers both carrying a heterozygous missense mutation c.905G>A (R302Q) in the PRKAG2 gene and further corrected the R302Q mutation with CRISPR-Cas9 mediated genome editing. Disease-specific hiPSC-cardiomyocytes recapitulated many phenotypes of PRKAG2 cardiac syndrome including cellular enlargement, electrophysiological irregularities and glycogen storage. In addition, we found that the PRKAG2-R302Q mutation led to increased AMPK activities, resulting in extensive glycogen deposition and cardiomyocyte hypertrophy. Finally we confirmed that disrupted phenotypes of PRKAG2 cardiac syndrome caused by the specific PRKAG2-R302Q mutation can be alleviated by small molecules inhibiting AMPK activity and be rescued with CRISPR-Cas9 mediated genome correction. Our results showed that disease-specific hiPSC-CMs and genetically-corrected hiPSC-cardiomyocytes would be a very useful platform for understanding the pathogenesis of, and testing autologous cell-based therapies for, PRKAG2 cardiac syndrome.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Cardiopatias/enzimologia , Cardiopatias/patologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Adulto , Sequência de Bases , Cálcio/metabolismo , Cardiomegalia/enzimologia , Cardiomegalia/patologia , Diferenciação Celular , Fenômenos Eletrofisiológicos , Glicogênio/metabolismo , Cardiopatias/fisiopatologia , Humanos , Masculino , Mitocôndrias/metabolismo , Mutação/genética , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Miócitos Cardíacos/ultraestrutura , Oxirredução , Fenótipo , Reprodutibilidade dos Testes , Síndrome
6.
Phys Chem Chem Phys ; 20(47): 29601-29609, 2018 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-30307452

RESUMO

New gas phase infrared spectroscopy is reported for an aromatic alpha hydroxy carboxylic acid homo dimer of 9-hydroxy-9-fluorene carboxylic acid (9HFCA)2, and the hetero dimer of 9HFCA with glycolic acid. In terms of the 9-hydroxy stretching frequency, the 16 cm-1 blue-shift in the homo dimer and the 17 cm-1 blue-shift in the hetero dimer, relative to that in 9HFCA monomer, are attributed to collective effects with anti-cooperativity stronger than cooperativity. Furthermore, for the hetero dimer, the two alpha hydroxy groups' stretching frequencies are clearly resolved, and differ by 30 cm-1. This difference represents a modest, quantitative enhancement of the intramolecular H-bond by the fluorene moiety in 9HFCA monomer, as opposed to that in glycolic acid. Accurate vibrational frequencies of the alpha OH, 3568 cm-1 in the bare glycolic acid, and 3584 cm-1 in the glycolic acid homo dimer are determined for the first time by comparison to 9HFCA monomer, homo and hetero dimers. The quantitative studies by infrared spectroscopy reveal subtle interactions among intra- and intermolecular H-bonds in the alpha hydroxyl acid dimers, which are also uniquely extended to probe each monomer's subtle intramolecular interactions.

7.
J Phys Chem A ; 122(27): 5886-5894, 2018 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-29901393

RESUMO

A new energy decomposition analysis (EDA) scheme based on valence bond (VB) wave function, called VB-EDA, is presented. In VB-EDA, the total interaction energy is decomposed into frozen, charge transfer, polarization and dynamic correlation terms based on valence bond calculations. The frozen term is the energy variation of the unrelaxed VB wave function according to the change of an interaction distance. The charge transfer term is the contribution of the additional VB structures while the polarization term is due to the relaxation of VB orbitals. Dynamic correlation term is computed by post-VBSCF methods. Different from other existing VB based EDA schemes, which were used to analyze noncovalent interactions for some specific complexes, the newly developed VB-EDA is designed for the general use. Using VB-EDA, the bonding nature of cation-π interactions in a series of cation-π complexes (cations = Li+, Na+, K+, Mg2+, and Ca2+; π systems = ethylene and benzene) is explored. Furthermore, a new covalency index, which demonstrates the covalency of cation-π interactions, is presented based on the VB-EDA results. The VB-EDA analysis reveals that the cation-π interactions in the Li+, Na+, and K+ complexes belong to the typical ionic bonds while the Mg2+ and Ca2+ complexes have the relatively large covalent characteristics. However, only the C2H4-Mg2+ complex can be regarded as a covalent bonding complex while the other complexes belong to the typical ionic complexes. Thereupon, it must be careful in the cognition for the covalency of intermolecular interaction. Large nonelectrostatic interaction component does not always correspond to a covalent bond.

8.
Circ Res ; 117(4): 364-375, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26123998

RESUMO

RATIONALE: Wnt/ß-catenin signaling has an important role in the angiogenic activity of endothelial cells (ECs). Bach1 is a transcription factor and is expressed in ECs, but whether Bach1 regulates angiogenesis is unknown. OBJECTIVE: This study evaluated the role of Bach1 in angiogenesis and Wnt/ß-catenin signaling. METHODS AND RESULTS: Hind-limb ischemia was surgically induced in Bach1(-/-) mice and their wild-type littermates and in C57BL/6J mice treated with adenoviruses coding for Bach1 or GFP. Lack of Bach1 expression was associated with significant increases in perfusion and vascular density and in the expression of proangiogenic cytokines in the ischemic hindlimb of mice, with enhancement of the angiogenic activity of ECs (eg, tube formation, migration, and proliferation). Bach1 overexpression impaired angiogenesis in mice with hind-limb ischemia and inhibited Wnt3a-stimulated angiogenic response and the expression of Wnt/ß-catenin target genes, such as interleukin-8 and vascular endothelial growth factor, in human umbilical vein ECs. Interleukin-8 and vascular endothelial growth factor were responsible for the antiangiogenic response of Bach1. Immunoprecipitation and GST pull-down assessments indicated that Bach1 binds directly to TCF4 and reduces the interaction of ß-catenin with TCF4. Bach1 overexpression reduces the interaction between p300/CBP and ß-catenin, as well as ß-catenin acetylation, and chromatin immunoprecipitation experiments confirmed that Bach1 occupies the TCF4-binding site of the interleukin-8 promoter and recruits histone deacetylase 1 to the interleukin-8 promoter in human umbilical vein ECs. CONCLUSIONS: Bach1 suppresses angiogenesis after ischemic injury and impairs Wnt/ß-catenin signaling by disrupting the interaction between ß-catenin and TCF4 and by recruiting histone deacetylase 1 to the promoter of TCF4-targeted genes.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Células Endoteliais/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Isquemia/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Via de Sinalização Wnt , beta Catenina/metabolismo , Acetilação , Animais , Apoptose , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/deficiência , Fatores de Transcrição de Zíper de Leucina Básica/genética , Sítios de Ligação , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Regulação para Baixo , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Feminino , Células HEK293 , Membro Posterior , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Isquemia/genética , Isquemia/fisiopatologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , Fator de Transcrição 4 , Fatores de Transcrição/metabolismo , Transfecção , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteína Wnt3A/metabolismo , beta Catenina/genética , Fatores de Transcrição de p300-CBP/metabolismo
9.
Arch Toxicol ; 91(1): 271-287, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27052460

RESUMO

Hydroxysteroid sulfotransferase 2B1b (SULT2B1b) sulfates cholesterol and oxysterols. Hepatic oval cells (HOCs), thought to be progenitor cells, can be triggered in chemically injured livers. The present study focused on the role of SULT2B1b in HOC proliferation after liver injury. Our experiments revealed that the expression of SULT2B1b was increased dramatically in a chemical-induced liver injury model, mainly in HOCs. Upon challenge with a hepatotoxic diet containing 0.1 % 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC), SULT2B1-/- mice presented alleviated liver injury and less HOC proliferation compared with wild-type (WT) mice, and these findings were verified by serum analysis, histopathology, immunofluorescence staining, RNA-seq, and Western blotting. HOCs derived from SULT2B1-/- mice showed lower proliferative capability than those from WT mice. SULT2B1b overexpression promoted growth of the WB-F344 hepatic oval cell line, whereas SULT2B1b knockdown inhibited growth of these cells. The IL-6/STAT3 signaling pathway also was promoted by SULT2B1b. Liquid chromatography and mass spectrometry indicated that the levels of 22-hydroxycholesterol, 25-hydroxycholesterol, and 24,25-epoxycholesterol were higher in the DDC-injured livers of SULT2B1-/- mice than in livers of WT mice. The above oxysterols are physiological ligands of liver X receptors (LXRs), and SULT2B1b suppressed oxysterol-induced LXR activation. Additional in vivo and in vitro experiments demonstrated that LXR activation could inhibit HOC proliferation and the IL-6/STAT3 signaling pathway, and these effects could be reversed by SULT2B1b. Our data indicate that upregulation of SULT2B1b might promote HOC proliferation and aggravate liver injury via the suppression of oxysterol-induced LXR activation in chemically induced mouse liver injury.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Modelos Animais de Doenças , Receptores X do Fígado/agonistas , Fígado/efeitos dos fármacos , Oxisteróis/farmacologia , Sulfotransferases/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Carcinógenos/toxicidade , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/patologia , Doença Hepática Induzida por Substâncias e Drogas/fisiopatologia , Progressão da Doença , Feminino , Fígado/metabolismo , Fígado/patologia , Fígado/fisiopatologia , Neoplasias Hepáticas/etiologia , Receptores X do Fígado/antagonistas & inibidores , Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxisteróis/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/patologia , Piridinas/toxicidade , Interferência de RNA , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/química , Sulfotransferases/genética
10.
IUBMB Life ; 68(12): 963-970, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27797149

RESUMO

Reactive oxygen species (ROS) and redox homeostasis have a pivotal role in the maintenance of stem cell pluripotency and in stem cell self-renewal; however, the mechanisms by which ROS regulate the self-renewal of stem cells have not been thoroughly studied. Here, we evaluated the role of the ROS produced by NADPH oxidase 2 (Nox2) and NADPH oxidase 4 (Nox4) in the self-renewal and stemness of murine induced-pluripotent stem cells (miPSCs). Targeted silencing of Nox2 or Nox4 reduced both NADPH oxidase activity and intracellular ROS levels, as well as alkaline phosphatase activity, the total number of miPSCs, the expression of insulin-like growth factor-1 (IGF-1), IGF-1 receptor, and the phosphorylation of extracellular signal regulated kinase (ERK) 1/2. Nox2/Nox4 overexpression or low, nontoxic concentration of H2 O2 increased cell proliferation in miPSCs. Furthermore, expression of the stemness genes Sox2 and Oct4 was lower in Nox2/Nox4-deficient miPSCs, and higher in Nox2/Nox4-overexpressing miPSCs, than in miPSCs with normal levels of Nox2/Nox4 expression. Collectively, these results suggest that Nox2- and Nox4-derived ROS contribute to stem cell pluripotency maintenance and self-renewal. © 2016 IUBMB Life, 68(12):963-970, 2016.


Assuntos
Autorrenovação Celular , Células-Tronco Pluripotentes Induzidas/fisiologia , Glicoproteínas de Membrana/fisiologia , NADPH Oxidases/fisiologia , Animais , Diferenciação Celular , Células Cultivadas , Técnicas de Silenciamento de Genes , Camundongos , NADPH Oxidase 2 , NADPH Oxidase 4 , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
11.
IUBMB Life ; 67(9): 710-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26337686

RESUMO

Recurrent stroke is difficult to treat and life threatening. Transfer of anti-inflammatory gene is a potential gene therapy strategy for ischemic stroke. Using recombinant adeno-associated viral vector 1 (rAAV1)-mediated interleukin 10 (IL-10), we investigated whether transfer of beneficial gene into the rat cerebral vessels during interventional treatment for initial stroke could attenuate brain injury caused by recurrent stroke. Male Wistar rats were administered rAAV1-IL-10, rAAV1-YFP, or saline into the left cerebral artery. Three weeks after gene transfer, rats were subjected to occlusion of the left middle cerebral artery (MCAO) for 45 min followed by reperfusion for 24 h. IL-10 levels in serum were significantly elevated 3 weeks after rAAV1-IL-10 injection, and virus in the cerebral vessels was confirmed by in situ hybridization. Pre-existing IL-10 but not YFP decreased the neurological dysfunction scores, brain infarction volume, and the number of injured neuronal cells. AAV1-IL-10 transduction increased heme oxygenase (HO-1) mRNA and protein levels in the infarct boundary zone of the brain. Thus, transduction of the IL-10 gene in the cerebral artery prior to ischemia attenuates brain injury caused by ischemia/reperfusion in rats. This preventive approach for recurrent stroke can be achieved during interventional treatment for initial stroke.


Assuntos
Lesões Encefálicas/prevenção & controle , Artérias Cerebrais/metabolismo , Terapia Genética , Interleucina-10/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Traumatismo por Reperfusão/complicações , Animais , Western Blotting , Lesões Encefálicas/etiologia , Artérias Cerebrais/patologia , Dependovirus/genética , Técnicas Imunoenzimáticas , Interleucina-10/genética , Masculino , Estresse Oxidativo , RNA Mensageiro/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Biochem Biophys Res Commun ; 443(2): 586-91, 2014 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-24333415

RESUMO

Clock is a basic helix-loop-helix (bHLH) transcription factor that plays important role in circadian rhythms of various physiological functions. Previous study showed that the expression of intercellular adhesion molecule-1 (ICAM-1) was reduced in the liver tissues of Clock mutant mice. However, how Clock regulates ICAM-1 expression and whether Clock affects cell adhesion function remain unknown. In the present study, we found that exogenous expression of Clock upregulated the gene expressions of ICAM-1 and other adhesion-related genes including VCAM1 and CCL-2, and increased the transcriptional activity of ICAM-1 in mouse brain microvascular endothelial cell lines. In contrast, loss of Clock decreased these gene expressions and ICAM-1 transcriptional activity. Chromatin immunoprecipitation (ChIP) assay revealed that Clock binds to the E-box-like enhancer of ICAM-1 gene. ICAM-1 gene showed rhythmic expression in endothelial cells after serum shock in vitro, suggesting ICAM-1 may be a Clock-controlled gene. Clock regulates the adhesion of mononuclear cells to endothelial cells via ICAM-1. Together, our findings show that Clock is a positive regulator of ICAM-1, and promotes the adhesion of mononuclear cells to endothelial cells.


Assuntos
Proteínas CLOCK/metabolismo , Adesão Celular/fisiologia , Células Endoteliais/fisiologia , Molécula 1 de Adesão Intercelular/metabolismo , Leucócitos Mononucleares/fisiologia , Regulação para Cima/fisiologia , Animais , Comunicação Celular/fisiologia , Linhagem Celular , Camundongos
13.
J Exp Med ; 204(3): 605-18, 2007 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-17339405

RESUMO

Stromal cell-derived factor 1 (SDF-1) plays a major role in the migration, recruitment, and retention of endothelial progenitor cells to sites of ischemic injury and contributes to neovascularization. We provide direct evidence demonstrating an important role for heme oxygenase 1 (HO-1) in mediating the proangiogenic effects of SDF-1. Nanomolar concentrations of SDF-1 induced HO-1 in endothelial cells through a protein kinase C zeta-dependent and vascular endothelial growth factor-independent mechanism. SDF-1-induced endothelial tube formation and migration was impaired in HO-1-deficient cells. Aortic rings from HO-1(-/-) mice were unable to form capillary sprouts in response to SDF-1, a defect reversed by CO, a byproduct of the HO-1 reaction. Phosphorylation of vasodilator-stimulated phosphoprotein was impaired in HO-1(-/-) cells, an event that was restored by CO. The functional significance of HO-1 in the proangiogenic effects of SDF-1 was confirmed in Matrigel plug, wound healing, and retinal ischemia models in vivo. The absence of HO-1 was associated with impaired wound healing. Intravitreal adoptive transfer of HO-1-deficient endothelial precursors showed defective homing and reendothelialization of the retinal vasculature compared with HO-1 wild-type cells following ischemia. These findings demonstrate a mechanistic role for HO-1 in SDF-1-mediated angiogenesis and provide new avenues for therapeutic approaches in vascular repair.


Assuntos
Quimiocinas CXC/fisiologia , Heme Oxigenase-1/fisiologia , Neovascularização Fisiológica , Animais , Células Cultivadas , Quimiocina CXCL12 , Células Endoteliais/enzimologia , Células Endoteliais/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/enzimologia , Endotélio Vascular/fisiologia , Heme Oxigenase-1/deficiência , Heme Oxigenase-1/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Estromais/enzimologia , Cicatrização/genética , Cicatrização/fisiologia
14.
Clin Exp Pharmacol Physiol ; 40(9): 626-34, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23763475

RESUMO

Oxidative stress caused by cellular accumulation of reactive oxygen species (ROS) is a major contributor to disease and cell death. However, how induced pluripotent stem cells (iPSC) respond to different levels of oxidative stress is largely unknown. Here, we investigated the effect of H2 O2 -induced oxidative stress on iPSC function in vitro. Mouse iPSC were treated with H2 O2 (25-100 µmol/L). IPSC adhesion, migration, viability, apoptosis and senescence were analysed. Expression of adhesion-related genes, stress defence genes, and osteoblast- and adipocyte-associated genes were determined by reverse transcription polymerase chain reaction. The present study found that H2 O2 (25-100 µmol/L) decreased iPSC adhesion to matrix proteins and endothelial cells, and downregulated gene expression levels of adhesion-related molecules, such as integrin alpha 7, cadherin 1 and 5, melanoma cell adhesion molecule, vascular cell adhesion molecule 1, and monocyte chemoattractant protein-1. H2 O2 (100 µmol/L) decreased iPSC viability and inhibited the capacity of iPSC migration and transendothelial migration. iPSC were sensitive to H2 O2 -induced G2/M arrest, senescence and apoptosis when exposed to H2 O2 at concentrations above 25 µmol/L. H2 O2 increased the expression of stress defence genes, including catalase, cytochrome B alpha, lactoperoxidase and thioredoxin domain containing 2. H2 O2 upregulated the expression of osteoblast- and adipocyte-associated genes in iPSC during their differentiation; however, short-term H2 O2 -induced oxidative stress did not affect the protein expression of the pluripotency markers, octamer-binding transcription factor 4 and sex-determining region Y-box 2. The present results suggest that iPSC are sensitive to H2 O2 toxicity, and inhibition of oxidative stress might be a strategy for improving their functions.


Assuntos
Apoptose/fisiologia , Adesão Celular/fisiologia , Senescência Celular/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Estresse Oxidativo/fisiologia , Migração Transendotelial e Transepitelial/fisiologia , Animais , Apoptose/genética , Adesão Celular/genética , Moléculas de Adesão Celular/genética , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Movimento Celular/genética , Movimento Celular/fisiologia , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Senescência Celular/genética , Regulação para Baixo/genética , Regulação para Baixo/fisiologia , Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Expressão Gênica/genética , Expressão Gênica/fisiologia , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo/genética , Migração Transendotelial e Transepitelial/genética , Regulação para Cima/genética , Regulação para Cima/fisiologia
15.
Front Immunol ; 14: 1136290, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37275899

RESUMO

The mortality rate associated with acute lung injury (ALI) and its severe form, acute respiratory distress syndrome, is high. Induced pluripotent stem cell (iPSC) therapy is a potential treatment method for ALI, but its therapeutic efficacy is limited in injured lungs. Nitric oxide (NO) has various physiological actions. The current study investigated the effect of iPSCs pretreated with NO donors in paraquat (PQ)-induced ALI mouse model. Male C57BL/6 mice were intraperitoneally injected with PQ, followed by infusion of phosphate-buffered saline, iPSCs, L-arginine pretreated iPSCs, or Nitro-L-arginine methylester (L-NAME) pretreated iPSCs through the tail veins. Histopathological changes, pulmonary microvascular permeability, and inflammatory cytokine levels were analyzed after 3 or 28 d. The effects on iPSC proliferation, migration, and adhesion were evaluated in vitro. More L-arginine-pretreated iPSCs were selectively trafficked into the injured pulmonary tissue of mice with LPS-induced ALI, drastically diminishing the histopathologic changes and inflammatory cytokine levels (IL-1ß and IL-6). There was also markedly improved pulmonary microvascular permeability and pulmonary function. The NO inhibitor abolished the protective effects of iPSCs. In addition, the ability of L-arginine to promote the proliferation and migration of iPSCs was decreased by L-NAME pretreatment, suggesting that NO might mediate the therapeutic benefits of iPSC. The improvement of the iPSC physiological changes by the endogenous gaseous molecule NO reduces lung injury severity. L-Arginine represents a pharmacologically important strategy for enhancing the therapeutic potential of iPSCs.


Assuntos
Lesão Pulmonar Aguda , Células-Tronco Pluripotentes Induzidas , Óxido Nítrico/metabolismo , Paraquat/toxicidade , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Camundongos Endogâmicos C57BL , Masculino , Animais , Camundongos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Arginina/uso terapêutico
16.
Aging (Albany NY) ; 15(12): 5854-5872, 2023 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-37367945

RESUMO

Heterochronic parabiosis has shown that aging individuals can be rejuvenated by a youthful circulatory system; however, the underlying mechanisms remain unclear. Here, we evaluated the effect of exosomes isolated from mouse induced pluripotent stem cells (iPSCs) on angiogenesis in naturally aged mice. To achieve this, the angiogenic capacity of aortic ring, the total antioxidant capacity (TAOC), p53 and p16 expression levels of major organs, the proliferation of adherent bone marrow cells, and the function and content of serum exosomes in aged mice administered iPSC-derived exosomes were examined. Additionally, the effect of iPSC-derived exosomes on injured human umbilical vein endothelial cells (HUVECs) was assessed. The angiogenic capacity of aortic rings and clonality of bone marrow cells from young mice were significantly higher than those from aged mice; moreover, the organs of aged mice had a higher expression of aging genes and lower total TAOC. However, in vitro and in vivo experiments showed that the administration of iPSC-derived exosomes significantly improved these parameters in aged mice. The synergistic effect of both in vivo and in vitro treatments of aortic rings with iPSC-derived exosomes improved the angiogenic capacity of aortic rings from aged mice to levels similar to that of young mice. Compared with untreated aged mice, serum exosomal protein content and their promoted effect on endothelial cell proliferation and angiogenesis were significantly higher in untreated young mice and aged mice treated with iPSC-derived exosomes. Overall, these results showed that iPSC-derived exosomes may rejuvenate the body by anti-aging the vascular system.


Assuntos
Exossomos , Células-Tronco Pluripotentes Induzidas , MicroRNAs , Humanos , Camundongos , Animais , Células-Tronco Pluripotentes Induzidas/metabolismo , Exossomos/metabolismo , Neovascularização Fisiológica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Fenômenos Fisiológicos Cardiovasculares , Proliferação de Células , MicroRNAs/metabolismo
17.
Cancer Sci ; 103(6): 1000-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22380844

RESUMO

Estrogens are involved in the complex regulation of cell proliferation and apoptosis of hormone sensitive tumors including breast and endometrial cancers. Sulfation is the main pathway for estrogen metabolism, which is believed to be involved in the inactivation of estrogens in target tissues. SULT1E1 and PAPSS (PAPSS1 and PAPSS2) are responsible for the estrogen sulfation by providing catalyzing enzyme and universal sulfate donor. The present study showed the expression patterns of SULT1E1 and PAPSS in the breast and endometrial tissues by tissue array analysis and the assessment of clinical samples. The estrogen sulfation enzymes were comparatively higher in the tumorous tissues than their adjacent normal tissues. SULT1E1 overexpression inhibited the tumorigenesis in subcutaneous xenograft model. By CCK-8 assay and flow cytometry assay, overexpression of SULT1E1 and PAPSS1 by adenovirus blocked the estrogen pro-proliferating effect and promoted cell apoptosis induced by H(2)O(2) in MCF-7 cells. By real-time reverse transcription-polymerase chain reaction and western-blot assays, overexpression of SULT1E1 and PAPSS1 suppressed cell growth and triggered apoptosis by downregulating the levels of c-myc, cyclin D1 and bcl-2, meanwhile, upregulating bax expression. In conclusion, the discrepancies in expressions of SULT1E1 and PAPSS between breast and endometrial tumorous tissues and their adjacent normal tissues were prominent. Overexpression of SULT1E1 and PAPSS1 retarded MCF-7 cells growth in vivo and in vitro by arresting cell cycles and inducing apoptosis. Thus, targeting SULT1E1 and PAPSS expressions might be an important approach for estrogen-dependent cancers.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias do Endométrio/metabolismo , Estrogênios/metabolismo , Complexos Multienzimáticos/metabolismo , Neoplasias Hormônio-Dependentes/metabolismo , Sulfato Adenililtransferase/metabolismo , Sulfotransferases/metabolismo , Animais , Apoptose , Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Ciclina D1/biossíntese , Neoplasias do Endométrio/patologia , Endométrio/metabolismo , Feminino , Humanos , Peróxido de Hidrogênio/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Complexos Multienzimáticos/genética , Transplante de Neoplasias , Neoplasias Hormônio-Dependentes/patologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-myc/biossíntese , Sulfato Adenililtransferase/genética , Sulfotransferases/genética , Análise Serial de Tecidos , Transplante Heterólogo , Proteína X Associada a bcl-2/biossíntese
18.
Lipids Health Dis ; 11: 23, 2012 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-22316301

RESUMO

BACKGROUND: Dysregulation of circadian rhythms can contribute to diseases of lipid metabolism. NAD-dependent deacetylase sirtuin-1(SIRT1) is an important hub which links lipid metabolism with circadian clock by its deacetylation activity depends on intracellular NAD+/NADH content ratio. Hydrogen sulfide (H2S) is an endogenous reductant which can affect the intracellular redox state. Therefore, we hypothesized that exogenous H2S can affect the expression of circadian clock genes mediated by sirt1 thereby affecting body's lipid metabolism. And also because the liver is a typical peripheral circadian clock oscillator that is intimately linked to lipid metabolism. Thus the effect of H2S were observed on 24-hour dynamic expression of 4 central circadian clock genes and sirt1gene in primary cultured hepatocytes. RESULTS: We established a hepatocyte model that showed a circadian rhythm by serum shock method. And detected that the expression level and the peak of circadian clock genes decreased gradually and H2S could maintain the expression and amplitude of circadian clock genes such as Clock, Per2, Bmal1 and Rev-erbαwithin a certain period time. Accordingly the expression level of sirt1 in H2S group was significantly higher than that in the control group. CONCLUSION: Exogenous reductant H2S maintain the circadian rhythm of clock gene in isolated liver cells. We speculated that H2S has changed NAD+/NADH content ratio in hepatocytes and enhanced the activity of SIRT1 protein directly or indirectly, so as to maintain the rhythm of expression of circadian clock genes, they play a role in the prevention and treatment of lipid metabolism-related disease caused by the biological clock disorders.


Assuntos
Ritmo Circadiano/efeitos dos fármacos , Hepatócitos/fisiologia , Sulfeto de Hidrogênio/farmacologia , Substâncias Redutoras/farmacologia , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Animais , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Células Cultivadas , Ritmo Circadiano/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Metabolismo dos Lipídeos/genética , Camundongos , Camundongos Endogâmicos C57BL , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Cultura Primária de Células , Sirtuína 1/genética , Sirtuína 1/metabolismo
19.
Stem Cell Res Ther ; 13(1): 389, 2022 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-35908001

RESUMO

BACKGROUND: Induced pluripotent stem cells (iPSCs) have the potential to promote wound healing; however, their adhesion to the extracellular matrix (ECM) might decrease iPSC migration, thereby limiting their therapeutic potential. Integrin ß1 (Itgb1) is the major integrin subunit that mediates iPSC-ECM adhesion, suggesting that knocking out Itgb1 might be an effective method for enhancing the therapeutic efficacy of iPSCs. METHODS: We knocked out Itgb1 in mouse iPSCs and evaluated its effects on the therapeutic potential of topically applied iPSCs, as well as their underlying in vivo and in vitro mechanisms. RESULTS: The Itgb1-knockout (Itgb1-KO) did not change iPSC pluripotency, function, or survival in the absence of embedding in an ECM gel but did accelerate wound healing, angiogenesis, blood perfusion, and survival in skin-wound lesions. However, embedding in an ECM gel inhibited the in vivo effects of wild-type iPSCs but not those of Itgb1-knockout iPSCs. Additionally, in vitro results showed that Itgb1-knockout decreased iPSC-ECM adhesion while increasing ECM-crossing migration. Moreover, ECM coating on the culture surface did not change cell survival, regardless of Itgb1 status; however, the in vivo and in vitro functions of both Itgb1-knockout and wild-type iPSCs were not affected by the presence of agarose gel, which does not contain integrin-binding sites. Knockout of Integrin α4 (Itga4) did not change the above-mentioned cellular and therapeutic functions of iPSCs. CONCLUSIONS: Itgb1-knockout increased iPSCs migration and the wound-healing-promoting effect of topically applied iPSCs. These findings suggest the inhibition of Itgb1 expression is a possible strategy for increasing the efficacy of iPSC therapies.


Assuntos
Células-Tronco Pluripotentes Induzidas , Integrina beta1/metabolismo , Animais , Movimento Celular , Matriz Extracelular/fisiologia , Integrina beta1/genética , Integrinas , Camundongos , Camundongos Knockout , Cicatrização/genética
20.
EBioMedicine ; 71: 103575, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34488017

RESUMO

BACKGROUND: Human sinoatrial cardiomyocytes are essential building blocks for cell therapies of conduction system disorders. However, current differentiation protocols for deriving nodal cardiomyocytes from human pluripotent stem cells (hPSCs) are very inefficient. METHODS: By employing the hPSCs to cardiomyocyte (CM) in vitro differentiation system and generating E2A-knockout hESCs using CRISPR/Cas9 gene editing technology, we analyze the functions of E2A in CM differentiation. FINDINGS: We found that knockout of the transcription factor E2A substantially increased the proportion of nodal-like cells in hESC-derived CMs. The E2A ablated CMs displayed smaller cell size, increased beating rates, weaker contractile force, and other functional characteristics similar to sinoatrial node (SAN) cells. Transcriptomic analyses indicated that ion channel-encoding genes were up-regulated in E2A ablated CMs. E2A directly bounded to the promoters of genes key to SAN development via conserved E-box motif, and promoted their expression. Unexpect enhanced activity of NOTCH pathway after E2A ablation could also facilate to induct ventricle workingtype CMs reprogramming into SAN-like cells. INTERPRETATION: Our study revealed a new role for E2A during directed cardiac differentiation of hESCs and may provide new clues for enhancing induction efficiency of SAN-like cardiomyocytes from hPSCs in the future. FUNDING: This work was supported by the NSFC (No.82070391, N.S.; No.81870175 and 81922006, P.L.), the National Key R&D Program of China (2018YFC2000202, N.S.; 2017YFA0103700, P.L.), the Haiju program of National Children's Medical Center EK1125180102, and Innovative research team of high-level local universities in Shanghai and a key laboratory program of the Education Commission of Shanghai Municipality (ZDSYS14005).


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Células-Tronco Embrionárias Humanas/citologia , Miócitos Cardíacos/citologia , Potenciais de Ação , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Células Cultivadas , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos , Camundongos SCID , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA