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.
Stem Cell Res Ther ; 6: 83, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25900017

RESUMO

INTRODUCTION: Induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) may be suitable for myocardial repair. While their functional and structural properties have been extensively investigated, their response to ischemia-like conditions has not yet been clearly defined. METHODS: iPS-CMs were differentiated and enriched from murine induced pluripotent stem cells expressing enhanced green fluorescent protein (eGFP) and puromycin resistance genes under the control of an α-myosin heavy chain (α-MHC) promoter. iPS-CMs maturity and function were characterized by microscopy, real-time PCR, calcium transient recordings, electrophysiology, and mitochondrial function assays, and compared to those from neonatal murine cardiomyocytes. iPS-CMs as well as neonatal murine cardiomyocytes were exposed for 3 hours to hypoxia (1% O2) and glucose/serum deprivation, and viability, apoptosis markers, reactive oxygen species, mitochondrial membrane potential and intracellular stress signaling cascades were investigated. Then, the iPS-CMs response to mesenchymal stromal cell-conditioned medium was determined. RESULTS: iPS-CMs displayed key morphological and functional properties that were comparable to those of neonatal cardiomyocytes, but several parameters indicated an earlier iPS-CMs maturation stage. During hypoxia and glucose/serum deprivation, iPS-CMs exhibited a significantly higher proportion of poly-caspase-active, 7-aminoactinomycin D-positive and TUNEL-positive cells than neonatal cardiomyocytes. The average mitochondrial membrane potential was reduced in "ischemic" iPS-CMs but remained unchanged in neonatal cardiomyocytes; reactive oxygen species production was only increased in "ischemic" iPS-CMs, and oxidoreductase activity in iPS-CMs dropped more rapidly than in neonatal cardiomyocytes. In iPS-CMs, hypoxia and glucose/serum deprivation led to upregulation of Hsp70 transcripts and decreased STAT3 phosphorylation and total PKCε protein expression. Treatment with mesenchymal stromal cell-conditioned medium preserved oxidoreductase activity and restored pSTAT3 and PKCε levels. CONCLUSION: iPS-CMs appear to be particularly sensitive to hypoxia and nutrient deprivation. Counteracting the ischemic susceptibility of iPS-CMs with mesenchymal stromal cell-conditioned medium may help enhance their survival and efficacy in cell-based approaches for myocardial repair.


Assuntos
Hipóxia Celular , Miócitos Cardíacos/metabolismo , Animais , Cálcio/metabolismo , Diferenciação Celular , Meios de Cultivo Condicionados/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Imuno-Histoquímica , Células-Tronco Pluripotentes Induzidas/citologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteína Quinase C-épsilon/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT3/metabolismo
2.
Cell Transplant ; 24(6): 985-1002, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24256742

RESUMO

The amniotic epithelium consists of cells exhibiting mature epithelial cell characteristics, but also varying degrees of stemness. We tested the hypothesis that induction of epithelial-to-mesenchymal transition (EMT) in amniotic epithelial cells (AECs) derived from human placenta enhances their capacity to support the ischemic myocardium. In response to incubation with transforming growth factor-ß1 (TGF-ß1) protein, AECs lost their cobblestone morphology and acquired a fibroblastoid shape, associated with downregulation of E-cadherin, upregulation of N-cadherin, Akt phosphorylation, and intracellular periostin translocation. EMT-AECs displayed greatly enhanced mobility and secreted gelatinase activity compared with naive AECs. The surface presentation of CD105 and CD73 decreased, and RNA microarray analysis mirrored the loss of epithelial characteristics and transcriptional profile. Unmodified AECs and EMT-AECs were then injected intramyocardially in fully immunocompetent mice after permanent LAD ligation, and heart function was followed by MRI as well as 2D speckle tracking echocardiography after 4 weeks. EMT-AEC-treated infarct hearts displayed better global systolic function and improved longitudinal strain rate in the area of interest. Although no signals of human cells were detectable by histology, infarct size was smaller in EMT-AEC-treated hearts, associated with fewer TUNEL-positive cells and upregulation of periostin, while blood vessel density was increased in both ACE- and EMT-AEC-treated hearts. We conclude that EMT enhances the cardioprotective effects of human AECs.


Assuntos
Âmnio/citologia , Células Epiteliais/citologia , Transição Epitelial-Mesenquimal , Coração/fisiopatologia , Substâncias Protetoras/metabolismo , Animais , Antígenos de Superfície/metabolismo , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Forma Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Feminino , Citometria de Fluxo , Gelatinases/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos Endogâmicos BALB C , Microvasos/efeitos dos fármacos , Microvasos/patologia , Microvasos/fisiopatologia , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Análise de Sequência com Séries de Oligonucleotídeos , Fator de Crescimento Transformador beta1/farmacologia , Ultrassonografia
3.
Eur J Cardiothorac Surg ; 47(3): 416-25; discussion 425, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24778452

RESUMO

OBJECTIVES: Cross-talk between organ-specific extracellular matrix (ECM) and stem cells is often assumed but has not been directly demonstrated. We developed a protocol for the preparation of human cardiac ECM (cECM) and studied whether cECM has effects on pluripotent stem cell differentiation that may be useful for future cardiac regeneration strategies in patients with end-stage heart failure. METHODS: Of note, 0.3 mm-thick cECM slices were prepared from samples of myocardium from patients with end-stage non-ischaemic dilated cardiomyopathy, using a three-step protocol involving hypotonic lysis buffer, sodium dodecyl sulphate (SDS) and foetal bovine serum (FBS). Murine embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and mesenchymal stromal cells (MSCs) were seeded and grown in standard culture, on cECM or on non-specific ECM preparations (Matrigel® or Geltrex®). Cell attachment, apoptosis induction (Caspase 3/7 activity) and metabolic activity (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium conversion) were followed. Transcriptional activation of genes involved in pluripotency; early and late myocardial development; and endothelial, ectodermal or endodermal commitment were monitored by quantitative real-time polymerase chain reaction (rtPCR). Protein expression of selected markers was confirmed by immunohistology. RESULTS: cECM supported the proliferation of ESCs and iPSCs, and Caspase 3/7 activity was significantly lower compared with standard culture. Cardiac lineage commitment was favoured when ESCs or iPSCs were grown on cECM, as evidenced by the significantly increased mRNA expression of cardiac alpha myosin heavy polypeptide 6 (Myh6), cardiac troponin T2 (Tnnt2) and NK2 homeobox 5 (Nkx2.5) as well as positive immunohistology for cardiac troponin T and heavy-chain cardiac myosin protein. In contrast, Matrigel or Geltrex did not induce cardiac-specific markers. MSCs showed no evidence of cardiomyocyte differentiation. CONCLUSIONS: Human cardiac ECM seems to direct differentiation of pluripotent stem cells towards a cardiomyocyte phenotype. This phenomenon supports the use of cardiac ECM preparations for guided stem cell differentiation and myocardial repair, and may ultimately increase the therapeutic efficacy of cell therapy in heart failure patients.


Assuntos
Matriz Extracelular/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Animais , Apoptose , Biomarcadores/análise , Biomarcadores/metabolismo , Miosinas Cardíacas/análise , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , Matriz Extracelular/metabolismo , Humanos , Camundongos , Miócitos Cardíacos/química , Miócitos Cardíacos/fisiologia , Células-Tronco Pluripotentes/química , Células-Tronco Pluripotentes/fisiologia , Troponina T/análise , Troponina T/química , Troponina T/metabolismo
4.
Cell Physiol Biochem ; 34(3): 646-57, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25170622

RESUMO

BACKGROUND/AIMS: Cell-based therapies may be useful for treating ischemic diseases, but the underlying mechanisms are incompletely understood. We investigated the impact of cord blood mesenchymal stromal cell (CBMSC)- or fibroblast (FB)-secreted factors on starved endothelial cells and determined the relevant intracellular signaling pathways. METHODS: HUVECs were subjected to glucose/serum deprivation (GSD) in hypoxia or normoxia, in presence of CBMSC- or FB-conditioned medium (CM). Viability and proliferation were determined via WST-8 conversion and BrdU incorporation. Apoptosis was quantified by annexin V/ethidium homodimer-III staining, nuclear fragmentation and cell morphology. mRNA expression and protein phosphorylation were determined by real-time qPCR and western blot. Experiments were repeated in presence of small-molecule inhibitors. RESULTS: The negative impact of GSD was most pronounced at 21% O2. Here, medium of CBMSCs and FBs increased viability and proliferation and reduced apoptosis of HUVECs. This was associated with increased STAT3 and ERK1/2 phosphorylation and BCL-2 expression. Under STAT3 inhibition, the beneficial effect of CBMSC-CM on viability and BCL-2 expression was abolished. CONCLUSION: Factors released by CBMSCs protect endothelial cells from the deleterious impact of GSD by activation of the STAT3 survival pathway. However, this phenomenon is not CBMSC-specific and can be reproduced using juvenile fibroblasts.


Assuntos
Meios de Cultivo Condicionados , Sangue Fetal/citologia , Células-Tronco Mesenquimais/citologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Sequência de Bases , Primers do DNA , Células Endoteliais da Veia Umbilical Humana , Humanos , Reação em Cadeia da Polimerase em Tempo Real
5.
Eur J Cardiothorac Surg ; 45(6): 983-92, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24562010

RESUMO

OBJECTIVES: Among the mechanisms by which somatic stem cells may improve left ventricular function in ischaemic heart disease are pro-survival stimuli mediated by secreted factors. This phenomenon is frequently referred to, but remains poorly understood. We therefore investigated the non-regenerative cardioprotective effects of cord blood mesenchymal stromal cells (CBMSCs) in vitro and sought to identify relevant intracellular signalling pathways. METHODS: Conditioned medium from CBMSCs and fibroblasts was prepared, and secreted factors were analysed by Luminex(®) immunobead assay. Murine cardiomyocyte-derived HL-1 cells were subjected to simulated ischaemia by glucose and serum deprivation and hypoxia in CBMSC-conditioned or cell-free control medium or in medium conditioned by foreskin fibroblasts. The proportions of vital, apoptotic and necrotic cells (poly-caspase activity, annexin V and ethidium homodimer-III staining) were quantified using a high-content imaging system. Metabolic activity and proliferation rate were determined via 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium and 5-bromo-2-deoxyuridine assays. Phosphorylation of Akt, extracellular-signal-regulated kinase (ERK)1/2, signal transducer and activator of transcription 3 (STAT3) and glycogen synthase kinase 3ß was determined by western blot, and experiments were repeated in the presence of specific small-molecule inhibitors (Wortmannin, UO126 and Stattic). RESULTS: CBMSC medium reduced the proportion of dead HL-1 cardiomyocytes from 39 ± 3 to 28 ± 1% (P < 0.05) and the rate of late apoptotic cells to 68 ± 2% of that in control medium (P < 0.001). Metabolic activity was increased by 12 ± 1% compared with control (P < 0.05), while in fibroblast medium it was not (5 ± 2%, P = 1). This was associated with increased phosphorylation of Akt (2-fold, P < 0.05), ERK1/2 (3-fold, P < 0.01) and STAT3 (12-fold, P < 0.001). Combined blocking of the phosphatidylinositol-4,5-bisphosphate 3-kinase/Akt and mitogen-activated protein kinase/ERK signalling abolished the protective CBMSC effect, while blocking the pathways individually had no effect. Inhibition of STAT3 phosphorylation drastically lowered HL-1 cell viability in control medium, but not in medium conditioned by CBMSCs. CONCLUSIONS: The factors released by CBMSCs protect cardiomyocyte-like HL-1 cells from simulated ischaemia more than those released from fibroblasts. While CBMSC-triggered Akt and ERK1/2 activation provides protection in a compensatory manner, STAT3 is crucial for cardiomyocyte survival in ischaemia, but is not a key mediator of cytoprotective stem cell actions.


Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Modelos Cardiovasculares , Isquemia Miocárdica , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Animais , Sobrevivência Celular , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Sangue Fetal/citologia , Sistema de Sinalização das MAP Quinases , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia
6.
J Biomed Mater Res A ; 102(9): 3263-72, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24142588

RESUMO

Extracellular matrix (ECM) derived by tissue decellularization has applications as a tissue engineering scaffold and for support of cellular regeneration. Myocardial ECM from animals has been produced by whole-organ perfusion or immersion processes, but methods for preparation of human myocardial ECM for therapy and research have not been compared in detail, yet. We analyzed the impact of decellularization processes on human myocardial ECM, and tested its ability to serve as a scaffold for cell seeding. Sodium dodecyl sulfate (SDS)-based decellularization, but not treatments based on Triton X-100, deoxycholate or hypo/hypertonic incubations, removed cells satisfactorily, and incubation with fetal bovine serum (FBS) eliminated residual DNA. ECM architecture was best preserved by a protocol consisting of 2 h lysis, 6 h SDS, and 3 h FBS, but age and pathology of the donor tissue are highly important for producing reproducible, high-quality scaffolds. We also studied ECM repopulation with mesenchymal stem cells (CB-MSC), cardiomyocytes derived from induced pluripotent stem cells (iPS-CM), and na€ive neonatal mouse cardiomyocytes. Cells attached to the matrix and proliferated and displayed higher viability than in standard culture. We conclude that human cardiac ECM sheets may be suitable scaffold for cell-matrix interaction studies and as a biomaterial for tissue regeneration and engineering.


Assuntos
Matriz Extracelular/química , Células-Tronco Mesenquimais/citologia , Miocárdio/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Células Cultivadas , Coração/fisiologia , Humanos , Camundongos , Regeneração
7.
J Transl Med ; 11: 236, 2013 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-24074138

RESUMO

BACKGROUND: Clinical cardiac cell therapy using autologous somatic stem cells is restricted by age and disease-associated impairment of stem cell function. Juvenile cells possibly represent a more potent alternative, but the impact of patient-related variables on such cell products is unknown. We therefore evaluated the behavior of neonatal cord blood mesenchymal stem cells (CB-MSC) in the presence of serum from patients with advanced heart failure (HF). METHODS: Human serum was obtained from patients with severe HF (n = 21) and from healthy volunteers (n = 12). To confirm the systemic quality of HF in the sera, TNF-α and IL-6 were quantified. CB-MSC from healthy neonates were cultivated for up to 14 days in medium supplemented with 10% protein-normalized human HF or control serum or fetal calf serum (FCS). RESULTS: All HF sera contained increased cytokine concentrations (IL-6, TNF-α). When exposed to HF serum, CB-MSC maintained basic MSC properties as confirmed by immunophenotyping and differentiation assays, but clonogenic cells were reduced in number and gave rise to substantially smaller colonies in the CFU-F assay. Cell cycle analysis pointed towards G1 arrest. CB-MSC metabolic activity and proliferation were significantly impaired for up to 3 days as measured by MTS turnover, BrdU incorporation and DAPI + nuclei counting. On day 5, however, CB-MSC growth kinetics approached control serum levels, though protein expression of cell cycle inhibitors (p21, p27), and apoptosis marker Caspase 3 remained elevated. Signal transduction included the stress and cytokine-induced JNK and ERK1/2 MAP kinase pathways. CONCLUSIONS: Heart failure temporarily inhibits clonality and proliferation of "healthy" juvenile MSC in vitro. Further studies should address the in vivo and clinical relevance of this finding.


Assuntos
Insuficiência Cardíaca/patologia , Células-Tronco Mesenquimais/citologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Biomarcadores/sangue , Estudos de Casos e Controles , Ciclo Celular , Proliferação de Células , Células Clonais , Ensaio de Unidades Formadoras de Colônias , Citocinas/sangue , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sangue Fetal/citologia , Insuficiência Cardíaca/sangue , Humanos , Recém-Nascido , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Soro/metabolismo , Transdução de Sinais , Adulto Jovem
8.
J Surg Res ; 185(1): 70-83, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23830369

RESUMO

BACKGROUND: Autologous cells for cell therapy of ischemic cardiomyopathy often display age- and disease-related functional impairment, whereas an allogenic immunotolerant cell product would allow off-the-shelf application of uncompromised donor cells. We investigated the cardiac regeneration potential of a novel, clinical-grade placenta-derived human stromal cell product (PLX-PAD). METHODS: PLX-PAD cells derived from human donor placentas and expanded in a three-dimensional bioreactor system were tested for surface marker expression, proangiogenic, anti-inflammatory, and immunomodulatory properties in vitro. In BALB/C mice, the left anterior descending artery was ligated and PLX-PAD cells (n = 10) or vehicle (n = 10) were injected in the infarct border zone. Four weeks later, heart function was analyzed by two-dimensional and M-mode echocardiography. Scar size, microvessel density, extracellular matrix composition, myocyte apoptosis, and PLX-PAD cell retention were studied by histology. RESULTS: In vitro, PLX-PAD cells displayed both proangiogenesis and anti-inflammatory properties, represented by the secretion of both vascular endothelial growth factor and angiopoietin-1 that was upregulated by hypoxia, as well as by the capacity to suppress T-cell proliferation and augment IL-10 secretion when co-cultured with peripheral blood mononuclear cells. Compared with control mice, PLX-PAD-treated hearts had better contractile function, smaller infarct size, greater regional left ventricular wall thickness, and less apoptosis after 4 wk. PLX-PAD stimulated both angiogenesis and arteriogenesis in the infarct border zone, and periostin expression was upregulated in PLX-PAD-treated hearts. CONCLUSIONS: Clinical-grade PLX-PAD cells exert beneficial effects on ischemic myocardium that are associated with improved contractile function, and may be suitable for further evaluation aiming at clinical pilot trials of cardiac cell therapy.


Assuntos
Circulação Coronária/fisiologia , Infarto do Miocárdio/terapia , Neovascularização Fisiológica/fisiologia , Placenta/citologia , Células Estromais/transplante , Animais , Células Cultivadas , Modelos Animais de Doenças , Ecocardiografia , Feminino , Sobrevivência de Enxerto , Humanos , Interleucina-10/sangue , Masculino , Camundongos Endogâmicos BALB C , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/fisiopatologia , Comunicação Parácrina/fisiologia , Gravidez , Células Estromais/citologia , Transplante Heterólogo
9.
Glia ; 44(1): 67-75, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12951658

RESUMO

During development and in the adult, erbB2, erbB3, and erbB4 are expressed in many tissues and as heterodimers (B2/B3, B2/B4) serve as receptors for neuregulins. The general importance of neuregulin receptors for development is underlined by the observed embryonic (erbB2, erbB4) or perinatal (erbB3) lethality in mouse mutants. These mutants further revealed the fundamental role of the erbB2/erbB3 heterodimer for proper Schwann cell development, the ensheathing glia of the peripheral nervous system. However, only little is known about the functions of neuregulins and their receptors during postnatal development and in the adult. erbB2 and erbB3 during late embryogenesis and postnatally are expressed in different areas and cell types of the central nervous system, including oligodendrocytes, the ensheathing glia of the central nervous system. As terminal differentiation of oligodendrocytes peaks during postnatal development, it is not possible to use the neuregulin receptor mouse mutants to study terminal differentiation of oligodendrocytes in their absence in vivo. In order to investigate possible functions of the erbB3 gene in oligodendrocytes, we employed two different techniques. First, we directed the differentiation of erbB3-deficient embryonic stem cells into neural cell types to analyze the development of oligodendrocytes in the absence of erbB3 in vitro. Second, we grafted neural stem cells from spinal cords of erbB3 mutants into the retina of young mice to monitor oligodendrocyte differentiation and myelination in vivo. Results of both experimental approaches clearly show that erbB3 is not required for normal oligodendrocyte development and myelination.


Assuntos
Diferenciação Celular/genética , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/crescimento & desenvolvimento , Neurregulinas/metabolismo , Oligodendroglia/metabolismo , Receptor ErbB-3/deficiência , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Sistema Nervoso Central/ultraestrutura , Feminino , Feto , Masculino , Camundongos , Camundongos Mutantes , Microscopia Eletrônica , Bainha de Mielina/metabolismo , Bainha de Mielina/ultraestrutura , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/ultraestrutura , Oligodendroglia/ultraestrutura , Receptor ErbB-3/genética , Retina/citologia , Retina/crescimento & desenvolvimento , Retina/cirurgia , Medula Espinal/citologia , Medula Espinal/embriologia , Medula Espinal/transplante , Células-Tronco/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...