Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 110(31): 12643-8, 2013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-23858471

RESUMO

Organs are composites of tissue types with diverse developmental origins, and they rely on distinct stem and progenitor cells to meet physiological demands for cellular production and homeostasis. How diverse stem cell activity is coordinated within organs is not well understood. Here we describe a lineage-restricted, self-renewing common skeletal progenitor (bone, cartilage, stromal progenitor; BCSP) isolated from limb bones and bone marrow tissue of fetal, neonatal, and adult mice. The BCSP clonally produces chondrocytes (cartilage-forming) and osteogenic (bone-forming) cells and at least three subsets of stromal cells that exhibit differential expression of cell surface markers, including CD105 (or endoglin), Thy1 [or CD90 (cluster of differentiation 90)], and 6C3 [ENPEP glutamyl aminopeptidase (aminopeptidase A)]. These three stromal subsets exhibit differential capacities to support hematopoietic (blood-forming) stem and progenitor cells. Although the 6C3-expressing subset demonstrates functional stem cell niche activity by maintaining primitive hematopoietic stem cell (HSC) renewal in vitro, the other stromal populations promote HSC differentiation to more committed lines of hematopoiesis, such as the B-cell lineage. Gene expression analysis and microscopic studies further reveal a microenvironment in which CD105-, Thy1-, and 6C3-expressing marrow stroma collaborate to provide cytokine signaling to HSCs and more committed hematopoietic progenitors. As a result, within the context of bone as a blood-forming organ, the BCSP plays a critical role in supporting hematopoiesis through its generation of diverse osteogenic and hematopoietic-promoting stroma, including HSC supportive 6C3(+) niche cells.


Assuntos
Osso e Ossos/metabolismo , Cartilagem/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Transdução de Sinais/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Osso e Ossos/citologia , Cartilagem/citologia , Citocinas/genética , Citocinas/metabolismo , Regulação da Expressão Gênica/fisiologia , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Transgênicos , Células Estromais/citologia , Células Estromais/metabolismo
2.
Circulation ; 126(11 Suppl 1): S20-8, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22965984

RESUMO

BACKGROUND: Human cardiac progenitor cells (hCPCs) are a promising cell source for regenerative repair after myocardial infarction. Exploitation of their full therapeutic potential may require stable genetic modification of the cells ex vivo. Safe genetic engineering of stem cells, using facile methods for site-specific integration of transgenes into known genomic contexts, would significantly enhance the overall safety and efficacy of cellular therapy in a variety of clinical contexts. METHODS AND RESULTS: We used the phiC31 site-specific recombinase to achieve targeted integration of a triple fusion reporter gene into a known chromosomal context in hCPCs and human endothelial cells. Stable expression of the reporter gene from its unique chromosomal integration site resulted in no discernible genomic instability or adverse changes in cell phenotype. Namely, phiC31-modified hCPCs were unchanged in their differentiation propensity, cellular proliferative rate, and global gene expression profile when compared with unaltered control hCPCs. Expression of the triple fusion reporter gene enabled multimodal assessment of cell fate in vitro and in vivo using fluorescence microscopy, bioluminescence imaging, and positron emission tomography. Intramyocardial transplantation of genetically modified hCPCs resulted in significant improvement in myocardial function 2 weeks after cell delivery, as assessed by echocardiography (P=0.002) and MRI (P=0.001). We also demonstrated the feasibility and therapeutic efficacy of genetically modifying differentiated human endothelial cells, which enhanced hind limb perfusion (P<0.05 at day 7 and 14 after transplantation) on laser Doppler imaging. CONCLUSIONS: The phiC31 integrase genomic modification system is a safe, efficient tool to enable site-specific integration of reporter transgenes in progenitor and differentiated cell types.


Assuntos
Células-Tronco Fetais/transplante , Terapia Genética/métodos , Membro Posterior/irrigação sanguínea , Isquemia/cirurgia , Mutagênese Insercional/métodos , Infarto do Miocárdio/cirurgia , Animais , Diferenciação Celular , Divisão Celular , Cromossomos Humanos Par 19/genética , Células Endoteliais/citologia , Feminino , Coração Fetal/citologia , Células-Tronco Fetais/citologia , Células-Tronco Fetais/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Humanos , Integrases , Peptídeos e Proteínas de Sinalização Intracelular , Isquemia/fisiopatologia , Luciferases de Vaga-Lume/genética , Proteínas Luminescentes/genética , Imageamento por Ressonância Magnética , Camundongos , Camundongos SCID , Proteínas/genética , Distribuição Aleatória , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Timidina Quinase/genética , Transgenes , Proteínas de Transporte Vesicular , Proteínas Virais/genética , Integração Viral , Proteína Vermelha Fluorescente
3.
J Am Coll Cardiol ; 60(14): 1278-87, 2012 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-22841153

RESUMO

OBJECTIVES: The goal of this study was to demonstrate the enhancement of human cardiac progenitor cell (hCPC) reparative and regenerative potential by genetic modification for the treatment of myocardial infarction. BACKGROUND: Regenerative potential of stem cells to repair acute infarction is limited. Improved hCPC survival, proliferation, and differentiation into functional myocardium will increase efficacy and advance translational implementation of cardiac regeneration. METHODS: hCPCs isolated from the myocardium of heart failure patients undergoing left ventricular assist device implantation were engineered to express green fluorescent protein (hCPCe) or Pim-1-GFP (hCPCeP). Functional tests of hCPC regenerative potential were performed with immunocompromised mice by using intramyocardial adoptive transfer injection after infarction. Myocardial structure and function were monitored by echocardiographic and hemodynamic assessment for 20 weeks after delivery. hCPCe and hCPCeP expressing luciferase were observed by using bioluminescence imaging to noninvasively track persistence. RESULTS: hCPCeP exhibited augmentation of reparative potential relative to hCPCe control cells, as shown by significantly increased proliferation coupled with amelioration of infarction injury and increased hemodynamic performance at 20 weeks post-transplantation. Concurrent with enhanced cardiac structure and function, hCPCeP demonstrated increased cellular engraftment and differentiation with improved vasculature and reduced infarct size. Enhanced persistence of hCPCeP versus hCPCe was revealed by bioluminescence imaging at up to 8 weeks post-delivery. CONCLUSIONS: Genetic engineering of hCPCs with Pim-1 enhanced repair of damaged myocardium. Ex vivo gene delivery to modify stem cells has emerged as a viable option addressing current limitations in the field. This study demonstrates that efficacy of hCPCs from the failing myocardium can be safely and significantly enhanced through expression of Pim-1 kinase, setting the stage for use of engineered cells in pre-clinical settings.


Assuntos
Engenharia Genética , Infarto do Miocárdio/terapia , Miócitos Cardíacos/transplante , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Animais , Proliferação de Células , Ecocardiografia , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hemodinâmica , Humanos , Medições Luminescentes , Camundongos , Miócitos Cardíacos/enzimologia , Neovascularização Patológica , Proteínas Proto-Oncogênicas c-pim-1/genética , Transplante de Células-Tronco , Células-Tronco/enzimologia
4.
Circ Cardiovasc Imaging ; 5(4): 481-90, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22565608

RESUMO

BACKGROUND: Human cardiac progenitor cells have demonstrated great potential for myocardial repair in small and large animals, but robust methods for longitudinal assessment of their engraftment in humans is not yet readily available. In this study, we sought to optimize and evaluate the use of positron emission tomography (PET) reporter gene imaging for monitoring human cardiac progenitor cell (hCPC) transplantation in a mouse model of myocardial infarction. METHODS AND RESULTS: hCPCs were isolated and expanded from human myocardial samples and stably transduced with herpes simplex virus thymidine kinase (TK) PET reporter gene. Thymidine kinase-expressing hCPCs were characterized in vitro and transplanted into murine myocardial infarction models (n=57). Cardiac echocardiographic, magnetic resonance imaging and pressure-volume loop analyses revealed improvement in left ventricular contractile function 2 weeks after transplant (hCPC versus phosphate-buffered saline, P<0.03). Noninvasive PET imaging was used to track hCPC fate over a 4-week time period, demonstrating a substantial decline in surviving cells. Importantly, early cell engraftment as assessed by PET was found to predict subsequent functional improvement, implying a "dose-effect" relationship. We isolated the transplanted cells from recipient myocardium by laser capture microdissection for in vivo transcriptome analysis. Our results provide direct evidence that hCPCs augment cardiac function after their transplantation into ischemic myocardium through paracrine secretion of growth factors. CONCLUSIONS: PET reporter gene imaging can provide important diagnostic and prognostic information regarding the ultimate success of hCPC treatment for myocardial infarction.


Assuntos
Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/terapia , Miócitos Cardíacos/transplante , Tomografia por Emissão de Pósitrons/métodos , Transplante de Células-Tronco , Análise de Variância , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Ecocardiografia , Técnicas de Transferência de Genes , Genes Reporter/genética , Terapia Genética , Guanina/análogos & derivados , Humanos , Imuno-Histoquímica , Microdissecção e Captura a Laser , Modelos Lineares , Imageamento por Ressonância Magnética , Camundongos , Camundongos SCID , Contração Miocárdica/fisiologia , Infarto do Miocárdio/fisiopatologia , Comunicação Parácrina/fisiologia , Fenótipo , Recuperação de Função Fisiológica , Timidina Quinase/genética , Timidina Quinase/metabolismo , Proteínas Virais/metabolismo
5.
Circulation ; 124(11 Suppl): S27-34, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21911815

RESUMO

BACKGROUND: Although stem cell therapy has provided a promising treatment for myocardial infarction, the low survival of the transplanted cells in the infarcted myocardium is possibly a primary reason for failure of long-term improvement. Therefore, the development of novel prosurvival strategies to boost stem cell survival will be of significant benefit to this field. METHODS AND RESULTS: Cardiac progenitor cells (CPCs) were isolated from transgenic mice, which constitutively express firefly luciferase and green fluorescent protein. The CPCs were transduced with individual lentivirus carrying the precursor of miR-21, miR-24, and miR-221, a cocktail of these 3 microRNA precursors, or green fluorescent protein as a control. After challenge in serum free medium, CPCs treated with the 3 microRNA cocktail showed significantly higher viability compared with untreated CPCs. After intramuscular and intramyocardial injections, in vivo bioluminescence imaging showed that microRNA cocktail-treated CPCs survived significantly longer after transplantation. After left anterior descending artery ligation, microRNA cocktail-treated CPCs boost the therapeutic efficacy in terms of functional recovery. Histological analysis confirmed increased myocardial wall thickness and CPC engraftment in the myocardium with the microRNA cocktail. Finally, we used bioinformatics analysis and experimental validation assays to show that Bim, a critical apoptotic activator, is an important target gene of the microRNA cocktail, which collectively can bind to the 3'UTR region of Bim and suppress its expression. CONCLUSIONS: We have demonstrated that a microRNA prosurvival cocktail (miR-21, miR-24, and miR-221) can improve the engraftment of transplanted cardiac progenitor cells and therapeutic efficacy for treatment of ischemic heart disease.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Sobrevivência de Enxerto/fisiologia , MicroRNAs/genética , MicroRNAs/uso terapêutico , Infarto do Miocárdio/terapia , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Feminino , Injeções Intramusculares , Lentivirus , Camundongos , Camundongos SCID , Camundongos Transgênicos , Modelos Animais , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Células-Tronco/fisiologia , Transdução Genética , Resultado do Tratamento
6.
Circulation ; 124(11 Suppl): S46-54, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21911818

RESUMO

BACKGROUND: Under normoxic conditions, hypoxia-inducible factor (HIF)-1α is rapidly degraded by 2 hydroxylases: prolyl hydroxylase (PHD) and factor-inhibiting HIF-1 (FIH). Because HIF-1α mediates the cardioprotective response to ischemic injury, its upregulation may be an effective therapeutic option for ischemic heart failure. METHODS AND RESULTS: PHD and FIH were cloned from mouse embryonic stem cells. The best candidate short hairpin (sh) sequences for inhibiting PHD isoenzyme 2 and FIH were inserted into novel, nonviral, minicircle vectors. In vitro studies after cell transfection of mouse C2C12 myoblasts, HL-1 atrial myocytes, and c-kit(+) cardiac progenitor cells demonstrated higher expression of angiogenesis factors in the double-knockdown group compared with the single-knockdown and short hairpin scramble control groups. To confirm in vitro data, shRNA minicircle vectors were injected intramyocardially after left anterior descending coronary artery ligation in adult FVB mice (n=60). Functional studies using MRI, echocardiography, and pressure-volume loops showed greater improvement in cardiac function in the double-knockdown group. To assess mechanisms of this functional recovery, we performed a cell trafficking experiment, which demonstrated significantly greater recruitment of bone marrow cells to the ischemic myocardium in the double-knockdown group. Fluorescence-activated cell sorting showed significantly higher activation of endogenous c-kit(+) cardiac progenitor cells. Immunostaining showed increased neovascularization and decreased apoptosis in areas of injured myocardium. Finally, western blots and laser-capture microdissection analysis confirmed upregulation of HIF-1α protein and angiogenesis genes, respectively. CONCLUSIONS: We demonstrated that HIF-1α upregulation by double knockdown of PHD and FIH synergistically increases stem cell mobilization and myocardial angiogenesis, leading to improved cardiac function.


Assuntos
Células-Tronco Embrionárias/transplante , Técnicas de Silenciamento de Genes , Terapia Genética/métodos , Oxigenases de Função Mista/genética , Infarto do Miocárdio/terapia , Neovascularização Fisiológica/fisiologia , Pró-Colágeno-Prolina Dioxigenase/genética , Animais , Apoptose/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Endogâmicos , Oxigenases de Função Mista/metabolismo , Modelos Animais , Mioblastos Cardíacos/metabolismo , Mioblastos Cardíacos/patologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Transplante de Células-Tronco , Resultado do Tratamento
7.
Nat Biotechnol ; 29(9): 829-34, 2011 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-21841799

RESUMO

An important risk in the clinical application of human pluripotent stem cells (hPSCs), including human embryonic and induced pluripotent stem cells (hESCs and hiPSCs), is teratoma formation by residual undifferentiated cells. We raised a monoclonal antibody against hESCs, designated anti-stage-specific embryonic antigen (SSEA)-5, which binds a previously unidentified antigen highly and specifically expressed on hPSCs--the H type-1 glycan. Separation based on SSEA-5 expression through fluorescence-activated cell sorting (FACS) greatly reduced teratoma-formation potential of heterogeneously differentiated cultures. To ensure complete removal of teratoma-forming cells, we identified additional pluripotency surface markers (PSMs) exhibiting a large dynamic expression range during differentiation: CD9, CD30, CD50, CD90 and CD200. Immunohistochemistry studies of human fetal tissues and bioinformatics analysis of a microarray database revealed that concurrent expression of these markers is both common and specific to hPSCs. Immunodepletion with antibodies against SSEA-5 and two additional PSMs completely removed teratoma-formation potential from incompletely differentiated hESC cultures.


Assuntos
Anticorpos Monoclonais/metabolismo , Células-Tronco Pluripotentes/citologia , Polissacarídeos/metabolismo , Antígenos Embrionários Estágio-Específicos/metabolismo , Teratoma/patologia , Animais , Biomarcadores , Diferenciação Celular , Células Cultivadas , Biologia Computacional , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Análise em Microsséries , Células-Tronco Pluripotentes/química , Reação em Cadeia da Polimerase em Tempo Real , Teratoma/metabolismo
8.
J Biol Chem ; 286(37): 32697-704, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21719696

RESUMO

Derivation of patient-specific induced pluripotent stem cells (iPSCs) opens a new avenue for future applications of regenerative medicine. However, before iPSCs can be used in a clinical setting, it is critical to validate their in vivo fate following autologous transplantation. Thus far, preclinical studies have been limited to small animals and have yet to be conducted in large animals that are physiologically more similar to humans. In this study, we report the first autologous transplantation of iPSCs in a large animal model through the generation of canine iPSCs (ciPSCs) from the canine adipose stromal cells and canine fibroblasts of adult mongrel dogs. We confirmed pluripotency of ciPSCs using the following techniques: (i) immunostaining and quantitative PCR for the presence of pluripotent and germ layer-specific markers in differentiated ciPSCs; (ii) microarray analysis that demonstrates similar gene expression profiles between ciPSCs and canine embryonic stem cells; (iii) teratoma formation assays; and (iv) karyotyping for genomic stability. Fate of ciPSCs autologously transplanted to the canine heart was tracked in vivo using clinical positron emission tomography, computed tomography, and magnetic resonance imaging. To demonstrate clinical potential of ciPSCs to treat models of injury, we generated endothelial cells (ciPSC-ECs) and used these cells to treat immunodeficient murine models of myocardial infarction and hindlimb ischemia.


Assuntos
Tecido Adiposo/metabolismo , Regulação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Transplante de Células-Tronco , Tecido Adiposo/citologia , Animais , Modelos Animais de Doenças , Cães , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Masculino , Camundongos , Camundongos SCID , Isquemia Miocárdica/terapia , Análise de Sequência com Séries de Oligonucleotídeos , Células Estromais/citologia , Células Estromais/metabolismo , Transplante Autólogo , Transplante Heterólogo
9.
Biomaterials ; 32(20): 4647-58, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21477858

RESUMO

Polyethylenimine (PEI) based polymers are efficient agents for cell transfection. However, their use has been hampered due to high cell death associated with transfection thereby resulting in low efficiency of gene delivery within the cells. To circumvent the problem of cellular toxicity, metal binding peptides were linked to PEI. Eight peptide-PEI derivatives were synthesized to improve cell survival and transfection efficiency. TAT linked PEI was used as a control polymer. Peptides linked with PEI amines formed nanogels as shown by electron microscopy and atomic force microscopic measurements. Polymers were characterized by spectroscopic methods and their ability to form complexes with plasmids was tested using electrophoretic studies. These modifications improved polymer biocompatibility as well as cell survival markedly, when compared to PEI alone. A subset of the modified peptide-polymers also showed significantly higher transfection efficiency in primary human cells with respect to the widely used transfection agent, lipofectamine. Study of the underlying mechanism of the observed phenomena revealed lower levels of 'reactive oxygen species' (ROS) in the presence of the peptide-polymers when compared to PEI alone. This was further corroborated with global gene expression analysis which showed upregulation of multiple genes and pathways involved in regulating intracellular oxidative stress.


Assuntos
Técnicas de Transferência de Genes , Peptídeos/química , Peptídeos/metabolismo , Polietilenoimina , Polímeros , Sequência de Aminoácidos , Animais , Terapia Genética/métodos , Células HEK293 , Humanos , Teste de Materiais , Dados de Sequência Molecular , Estrutura Molecular , Estresse Oxidativo , Peptídeos/genética , Polietilenoimina/química , Polietilenoimina/metabolismo , Polímeros/química , Polímeros/metabolismo , Células Estromais/citologia , Células Estromais/fisiologia
10.
Adv Drug Deliv Rev ; 62(12): 1175-86, 2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20816906

RESUMO

Stem cell therapy has the potential to regenerate injured tissue. For stem cells to achieve their full therapeutic potential, stem cells must differentiate into the target cell, reach the site of injury, survive, and engraft. To fully characterize these cells, evaluation of cell morphology, lineage specific markers, cell specific function, and gene expression must be performed. To monitor survival and engraftment, cell fate imaging is vital. Only then can organ specific function be evaluated to determine the effectiveness of therapy. In this review, we will discuss methods for evaluating the function of transplanted cells for restoring the heart, nervous system, and pancreas. We will also highlight the specific challenges facing these potential therapeutic areas.


Assuntos
Linhagem da Célula , Células-Tronco Hematopoéticas/fisiologia , Transplante de Células-Tronco , Células-Tronco/fisiologia , Animais , Diferenciação Celular , Cardiopatias/metabolismo , Cardiopatias/terapia , Humanos , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/terapia , Pancreatopatias/metabolismo , Pancreatopatias/terapia , Regeneração
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA