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.
Microorganisms ; 8(5)2020 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-32370298

RESUMO

Cigarette smoke (CS) exacerbates symptoms in Crohn's disease (CD) patients while protecting those with ulcerative colitis (UC). CD has been associated with immuno-dysregulation, mucosal dysfunction, and infection. Among the CD-debated pathogens are Mycobacterium avium subsp. paratuberculosis (MAP), adherent invasive Escherichia coli (AIEC), and Klebsiella pneumoniae. The mechanism of how CS modulates nicotinic acetylcholine receptor-α7 (α7nAChR) and elicits inflammatory response in CD-like macrophages is unknown. Here, we investigated the effect of CS/nicotine on macrophages infected with CD-associated pathogens. We measured apoptosis, bacterial viability, macrophage polarization, and gene expression/cytokine levels involved in macrophage response to nicotine/CS extracts from Havana-Leave extract (HLE-nicotine rich) and germplasm line of Maryland tobacco (LAMD-nicotine less). Nicotine (4 µg/mL) and HLE extracts (0.18%) significantly favored anti-inflammatory response in macrophages (increased CD-206 (M2) and IL-10, and decreased M1/M2 ratio; p < 0.05). While macrophages infected with MAP or treated with LPS promoted pro-inflammatory response. Further treatment of these macrophages with nicotine or HLE extracts caused higher inflammatory response (increased iNOS (M1), TNF-α, IL-6, and M1/M2 ratio, p < 0.05), increased MAP burden, and decreased apoptosis. Pre-conditioning macrophages with nicotine ahead of infection resulted in lower pro-inflammatory response. Blocking α7nAChR with an antagonist voided the effect of nicotine on macrophages. Overall, the study provides an insight toward understanding the contradictory effect of nicotine on Inflammatory Bowel Disease patients and about the mechanistic role of α7nAChR in modulation of macrophages in tobacco smokers.

2.
Sci Rep ; 9(1): 8824, 2019 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-31217543

RESUMO

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by glia over-proliferation, neuro-inflammation, perturbed neural circuitry, and gastrointestinal symptoms. The role of gut dys-biosis in ASD is intriguing and should be elucidated. We investigated the effect of Propionic acid (PPA), a short-chain fatty acid (SCFA) and a product of dys-biotic ASD gut, on human neural stem cells (hNSCs) proliferation, differentiation and inflammation. hNSCs proliferated to 66 neuropsheres when exposed to PPA versus 45 in control. The neurosphere diameter also increased at day 10 post PPA treatment to (Mean: 193.47 um ± SEM: 6.673 um) versus (154.16 um ± 9.95 um) in control, p < 0.001. Pre-treatment with ß-HB, SCFA receptor inhibitor, hindered neurosphere expansion (p < 0.001). While hNSCs spontaneously differentiated to (48.38% ± 6.08%) neurons (Tubulin-IIIß positive) and (46.63% ± 2.5%) glia (GFAP positive), PPA treatment drastically shifted differentiation to 80% GFAP cells (p < 0.05). Following 2 mM PPA exposure, TNF-α transcription increased 4.98 fold and the cytokine increased 3.29 fold compared to control (P < 0.001). Likewise, GPR41 (PPA receptor) and pro-survival p-Akt protein were elevated (p < 0.001). PTEN (Akt inhibitor) level decreased to (0.42 ug/ul ± 0.04 ug/ul) at 2 mM PPA compared to (0.83 ug/ul ± 0.09 ug/ul) in control (p < 0.001). PPA at 2 mM decreased neurite outgrowth to (80.70 um ± 5.5 um) compared to (194.93 um ± 19.7 um) in control. Clearly, the data supports a significant role for PPA in modulating hNSC patterning leading to gliosis, disturbed neuro-circuitry, and inflammatory response as seen in ASD.


Assuntos
Transtorno do Espectro Autista/enzimologia , Transtorno do Espectro Autista/patologia , Gliose/patologia , Inflamação/patologia , Neurônios/patologia , PTEN Fosfo-Hidrolase/metabolismo , Propionatos/efeitos adversos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Biomarcadores Tumorais/metabolismo , Ácido Butírico/farmacologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tubulina (Proteína)/metabolismo
3.
World J Gastroenterol ; 24(25): 2764-2775, 2018 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-29991880

RESUMO

AIM: To perform a meta-analysis on the risk of developing Mycobacterium tuberculosis (TB) infection in Crohn's disease (CD) patients treated with tumor necrosis factor-alpha (TNFα) inhibitors. METHODS: A meta-analysis of randomized, double-blind, placebo-controlled trials of TNFα inhibitors for treatment of CD in adults was conducted. Arcsine transformation of TB incidence was performed to estimate risk difference. A novel epidemiologically-based correction (EBC) enabling inclusions of studies reporting no TB infection cases in placebo and treatment groups was developed to estimate relative odds. RESULTS: Twenty-three clinical trial studies were identified, including 5669 patients. Six TB infection cases were reported across 5 studies, all from patients receiving TNFα inhibitors. Eighteen studies reported no TB infection cases in placebo and TNFα inhibitor treatment arms. TB infection risk was significantly increased among patients receiving TNFα inhibitors, with a risk difference of 0.028 (95%CI: 0.0011-0.055). The odds ratio was 4.85 (95%CI: 1.02-22.99) with EBC and 5.85 (95%CI: 1.13-30.38) without EBC. CONCLUSION: The risk of TB infection is higher among CD patients receiving TNFα inhibitors. Understanding the immunopathogenesis of CD is crucial, since using TNFα inhibitors in these patients could favor mycobacterial infections, particularly Mycobacterium avium subspecies paratuberculosis, which ultimately could worsen their clinical condition.


Assuntos
Doença de Crohn/tratamento farmacológico , Fármacos Gastrointestinais/efeitos adversos , Paratuberculose/epidemiologia , Tuberculose/epidemiologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Doença de Crohn/imunologia , Humanos , Sistema Imunitário/efeitos dos fármacos , Incidência , Mycobacterium avium subsp. paratuberculosis/imunologia , Mycobacterium avium subsp. paratuberculosis/isolamento & purificação , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/isolamento & purificação , Paratuberculose/imunologia , Paratuberculose/microbiologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Tuberculose/imunologia , Tuberculose/microbiologia , Fator de Necrose Tumoral alfa/imunologia
4.
Mol Cell Biochem ; 406(1-2): 101-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25920449

RESUMO

Cardiac cell regeneration from endogenous cardiac stem cells (CSCs) following MI is rather low. Therefore, identifying mechanisms to boost endogenous CSC activation and participation in cardiac repair appears to be the most promising strategy for MI patients. We previously engineered tissue inhibitor of metalloproteinases-1 (TIMP-1) overexpressing embryonic stem (ES-TIMP-1) cells and transplanted them into the infarcted murine heart. Collected data demonstrated that TIMP-1 enhanced transplanted ES cell engraftment, survival and differentiation into cardiac myocytes post-transplantation. Therefore, we postulated that there may be a new stem cell population present in the heart that is regulated by extracellular protein TIMP-1. Furthermore, we hypothesized that this cell population has a potential for cell proliferation and differentiation into cardiac cell types. Therefore, we isolated CSCs from 4 weeks old C57BL/6 mice and cultured them in vitro in presence of ESCM, ES-TIMP-1-CM or TIMP-1. Our immunostaining data demonstrated the existence of a novel CSC subpopulation, CD63(+ve)/c-kit(+ve). When treated with TIMP-1, these cells showed significantly (p < 0.05) increased proliferation rates compared to control cells, enhanced TIMP-1 receptor (CD63), along with improved expression of phospho and total ß-catenin proteins as demonstrated by Western blot analysis. Next, we demonstrate significantly (p < 0.05) improved cardiac myocyte, vascular smooth muscle cell, and endothelial cell differentiation. Furthermore, our RT-PCR data shows increase in cardiac gene (GATA-4, Mef2C, and Nkx-2.5) expression when compared to ESCM and control cells. Collectively, these data, for the first time, establish the existence of a new CD63(+ve)/c-kit(+ve) CSC subpopulation that has a significant potential for proliferation and differentiation into cardiac cell types once stimulated with TIMP-1.


Assuntos
Células-Tronco Adultas/fisiologia , Miocárdio/citologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Tetraspanina 30/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Separação Celular , Camundongos Endogâmicos C57BL , Inibidor Tecidual de Metaloproteinase-1/fisiologia , Ativação Transcricional
5.
PLoS One ; 10(3): e0120739, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25768089

RESUMO

Inflammation has been implicated as a perpetrator of diabetes and its associated complications. Monocytes, key mediators of inflammation, differentiate into pro-inflammatory M1 macrophages and anti-inflammatory M2 macrophages upon infiltration of damaged tissue. However, the inflammatory cell types, which propagate diabetes progression and consequential adverse disorders, remain unclear. The current study was undertaken to assess monocyte infiltration and the role of fibroblast growth factor-9 (FGF-9) on monocyte to macrophage differentiation and cardioprotection in the diabetic infarcted heart. Db/db diabetic mice were assigned to sham, myocardial infarction (MI), and MI+FGF-9 groups. MI was induced by permanent coronary artery ligation and animals were subjected to 2D transthoracic echocardiography two weeks post-surgery. Immunohistochemical and immunoassay results from heart samples collected suggest significantly increased infiltration of monocytes (Mean ± SEM; MI: 2.02% ± 0.23% vs. Sham 0.75% ± 0.07%; p<0.05) and associated pro-inflammatory cytokines (TNF-α, MCP-1, and IL-6), adverse cardiac remodeling (Mean ± SEM; MI: 33% ± 3.04% vs. Sham 2.2% ± 0.33%; p<0.05), and left ventricular dysfunction (Mean ± SEM; MI: 35.4% ± 1.25% vs. Sham 49.19% ± 1.07%; p<0.05) in the MI group. Importantly, treatment of diabetic infarcted myocardium with FGF-9 resulted in significantly decreased monocyte infiltration (Mean ± SEM; MI+FGF-9: 1.39% ± 0.1% vs. MI: 2.02% ± 0.23%; p<0.05), increased M2 macrophage differentiation (Mean ± SEM; MI+FGF-9: 4.82% ± 0.86% vs. MI: 0.85% ± 0.3%; p<0.05) and associated anti-inflammatory cytokines (IL-10 and IL-1RA), reduced adverse remodeling (Mean ± SEM; MI+FGF-9: 11.59% ± 1.2% vs. MI: 33% ± 3.04%; p<0.05), and improved cardiac function (Fractional shortening, Mean ± SEM; MI+FGF-9: 41.51% ± 1.68% vs. MI: 35.4% ± 1.25%; p<0.05). In conclusion, our data suggest FGF-9 possesses novel therapeutic potential in its ability to mediate monocyte to M2 differentiation and confer cardiac protection in the post-MI diabetic heart.


Assuntos
Diferenciação Celular/fisiologia , Complicações do Diabetes/fisiopatologia , Fator 9 de Crescimento de Fibroblastos/metabolismo , Monócitos/fisiologia , Infarto do Miocárdio/fisiopatologia , Análise de Variância , Animais , Remodelamento Atrial/fisiologia , Citocinas , Primers do DNA/genética , Ecocardiografia , Ensaio de Imunoadsorção Enzimática , Testes de Função Cardíaca , Imuno-Histoquímica , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos NOD , Infarto do Miocárdio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Cell Transplant ; 24(6): 1043-52, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24593859

RESUMO

Vascular apoptosis plays a pivotal role in the development and progression of a myriad of cardiac dysfunctions, but has yet to be investigated in doxorubicin-induced cardiomyopathy (DIC). Additionally, the neovascularization potential and resulting functional consequences of embryonic stem (ES) cells and factors released from these cells in the chronic DIC myocardium remain largely unknown. To this end, we transplanted conditioned media (CM) and ES cells in the DIC-injured heart and evaluated their potential to inhibit vascular cell death, activate endogenous c-kit(+) and FLK-1(+) cells, enhance neovascularization, and augment left ventricular dysfunction. Data presented suggest transplanted CM and ES cells significantly blunt vascular cell apoptosis consequent to DIC. Quantitative immunohistochemistry data demonstrate significantly increased c-kit(+) and FLK-1(+) cells, as well as enhanced differentiated CD31(+) cells in the CM and ES cell groups relative to DIC controls. Heart function, including fractional shortening and ejection fraction, assessed by transthoracic echocardiography, was significantly improved following CM and ES cell transplantation. In conclusion, our data suggest that transplantation of CM and ES cells inhibit vascular apoptosis, activate endogenous c-kit(+) and FLK-1(+) cells and differentiate them into endothelial cells, enhance neovascularization, and improve cardiac function in the DIC-injured myocardium.


Assuntos
Cardiomiopatias/induzido quimicamente , Doxorrubicina/efeitos adversos , Células-Tronco Embrionárias Murinas/citologia , Neovascularização Fisiológica , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Apoptose/efeitos dos fármacos , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/crescimento & desenvolvimento , Cardiomiopatias/patologia , Cardiomiopatias/fisiopatologia , Doença Crônica , Meios de Cultivo Condicionados/farmacologia , Feminino , Testes de Função Cardíaca , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Transplante de Células-Tronco
7.
PLoS One ; 8(9): e75580, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086577

RESUMO

Thymosin ß4 (Tß4), a small G-actin sequestering peptide, mediates cell proliferation, migration, and angiogenesis. Whether embryonic stem (ES) cells, overexpressing Tß4, readily differentiate into cardiac myocytes in vitro and in vivo and enhance cardioprotection following transplantation post myocardial infarction (MI) remains unknown. Accordingly, we established stable mouse ES cell lines, RFP-ESCs and Tß4-ESCs, expressing RFP and an RFP-Tß4 fusion protein, respectively. In vitro, the number of spontaneously beating embryoid bodies (EBs) was significantly increased in Tß4-ESCs at day 9, 12 and 15, compared with RFP-ESCs. Enhanced expression of cardiac transcriptional factors GATA-4, Mef2c and Txb6 in Tß4-EBs, as confirmed with real time-PCR analysis, was accompanied by the increased number of EB areas stained positive for sarcomeric α-actin in Tß4-EBs, compared with the RFP control, suggesting a significant increase in functional cardiac myocytes. Furthermore, we transplanted Tß4-ESCs into the infarcted mouse heart and performed morphological and functional analysis 2 weeks after MI. There was a significant increase in newly formed cardiac myocytes associated with the Notch pathway, a decrease in apoptotic nuclei mediated by an increase in Akt and a decrease in levels of PTEN. Cardiac fibrosis was significantly reduced, and left ventricular function was significantly augmented in the Tß4-ESC transplanted group, compared with controls. It is concluded that genetically modified Tß4-ESCs, potentiates their ability to turn into cardiac myocytes in vitro as well as in vivo. Moreover, we also demonstrate that there was a significant decrease in both cardiac apoptosis and fibrosis, thus improving cardiac function in the infarcted heart.


Assuntos
Células-Tronco Embrionárias/fisiologia , Desenvolvimento Muscular/fisiologia , Infarto do Miocárdio/fisiopatologia , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Timosina/metabolismo , Remodelação Ventricular/fisiologia , Animais , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular , Células-Tronco Embrionárias/metabolismo , Fibrose/metabolismo , Fibrose/fisiopatologia , Coração/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Transplante de Células-Tronco , Fatores de Transcrição/metabolismo , Função Ventricular Esquerda/fisiologia
8.
Can J Physiol Pharmacol ; 90(3): 307-15, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22372593

RESUMO

Cell therapy is emerging as a new strategy to circumvent the adverse effects of heart disease. Many experimental and clinical studies investigating the transplantation of cells into the injured myocardium have yielded promising results. Moreover, data from these reports show that transplanted stem cells can engraft within the myocardium, differentiate into major cardiac cell types, and improve cardiac function. However, results from clinical trials show conflicting results. These trials demonstrate significant improvements in cardiac function for up to 6 months. However, these improved functions were diminished when examined at 18 months. In this review, we will discuss the current literature available on cell transplantation, covering studies ranging from animal models to clinical trials.


Assuntos
Cardiomiopatias/terapia , Transplante de Células-Tronco , Células-Tronco Adultas/transplante , Animais , Células-Tronco Embrionárias/transplante , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Mioblastos Esqueléticos , Miócitos Cardíacos/transplante
9.
Mol Pharm ; 8(5): 1602-10, 2011 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-21851072

RESUMO

Recently, we proclaimed that induced pluripotent stem (iPS) cells generated from H9c2 cells, following transplantation into infarcted nondiabetic mice, can inhibit apoptosis and differentiate into cardiac myocytes. iPS cells can be an ideal candidate to expand regenerative medicine to the clinic. Therefore, examining the wide range of their potential to differentiate into neovascular cell types remains a major interest. We hypothesized that transplanted iPS cells in the infarcted diabetic db/db and nondiabetic mice can differentiate into vascular smooth muscle (VSM) and endothelial cells (ECs) as well as activate endogenous c-kit progenitor cells to enhance neovascularization along with improved cardiac function. We transplanted intramyocardially 50,000 iPS cells in the peri-infarct zone of infarcted db/db and C57BL/6 mice and hearts were examined at D14 post-MI. Cardiac function was examined using echocardiography. Our data implies that there was a significant (p < 0.001) increase in VSM and ECs in the infarcted heart following iPS cell transplantation compared with MI and sham groups in both db/db and C57BL/6 animals. Furthermore, the MI+iPS cell transplanted group also displayed a significant (p < 0.001) increase in c-kit(+ve) activated VSM and ECs confirmed with combined stainings of c-kit and cell specific markers, compared with respective controls. Next, our histology data in the MI+iPS cell group also establishes a significant (p < 0.05) increase in coronary artery vessels compared with MI, suggesting neovascularization. Furthermore, our data demonstrates significant improved cardiac function following iPS cell transplantation compared with MI. Overall increased neovascularization in the infarcted db/db and C57BL/6 mice is associated with improved cardiac function following iPS cell transplantation.


Assuntos
Vasos Coronários/fisiopatologia , Angiopatias Diabéticas/terapia , Coração/fisiopatologia , Células-Tronco Pluripotentes Induzidas/transplante , Infarto do Miocárdio/terapia , Neovascularização Fisiológica , Células-Tronco Adultas/metabolismo , Células-Tronco Adultas/patologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem Celular , Vasos Coronários/diagnóstico por imagem , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Angiopatias Diabéticas/diagnóstico por imagem , Angiopatias Diabéticas/fisiopatologia , Endotélio Vascular/citologia , Endotélio Vascular/diagnóstico por imagem , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Genes Reporter , Imuno-Histoquímica , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Músculo Liso Vascular/citologia , Músculo Liso Vascular/diagnóstico por imagem , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Ultrassonografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA