Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res ; 52: 102208, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33578365

RESUMO

Hypertrophic cardiomyopathy (HCM) is the most common form of genetic heart disease and is characterized by abnormal thickening of the left ventricular wall and interventricular septum. Here we describe the generation of two induced pluripotent stem cell (iPSC) clones from a HCM patient, heterozygous for the p.Arg723Gly (c.2169C > G) mutation in the MYH7 gene. The generated iPSC clones may provide a useful resource for disease modelling to study the mechanisms underlying HCM pathogenesis in iPSC derived progenies, in particular cardiomyocytes.


Assuntos
Cardiomiopatia Hipertrófica , Células-Tronco Pluripotentes Induzidas , Miosinas Cardíacas/genética , Cardiomiopatia Hipertrófica/genética , Células Clonais , Humanos , Mutação , Miócitos Cardíacos , Cadeias Pesadas de Miosina/genética
5.
Cytotherapy ; 10(4): 376-89, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18574770

RESUMO

BACKGROUND: Current efforts to direct differentiation of human embryonic stem cells (hESC) into a particular cell lineage usually lead to a heterogeneous cell population with only a fraction of the desired cell type present. We show the generation of an essentially pure population of human cardiomyocytes from hESC using lineage selection. METHODS: A construct comprising the murine alpha-myosin heavy chain (alpha-MHC) promoter driving the neomycin-resistance gene was introduced into hES3 cells to generate stable transgenic lines. Transgenic hESC lines were differentiated into cardiomyocytes and subjected to G418 selection. Both G418-selected and non-selected cardiomyocytes were characterized by immunocytochemistry and reverse transcriptase-polymerase chain reaction (RT-PCR) analysis. The teratoma-forming potential of differentiated cells was assessed by injection of about 2 million cells into the hind limb muscle of SCID mice. Results After cardiac differentiation and antibiotic selection in a suspension culture process, more than 99% of the transgenic cells showed immunoreactivity to alpha-MHC and alpha-actinin; this enrichment efficiency was observed for independent transgenic cell lines. Quantitative RT-PCR analysis revealed high levels of enrichment for cardiac-specific messages in the selected population. Importantly, injection of selected cells into six SCID mice resulted in no apparent teratoma formation, in contrast to differentiated but non-selected controls. DISCUSSION: Our results represent a significant step toward scalable production of pure human cardiomyocytes from stable, expandable hESC lines that will facilitate the development of cell therapies, safety pharmacology and drug discovery.


Assuntos
Linhagem da Célula , Células-Tronco Embrionárias , Miócitos Cardíacos , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem Celular , Eletrofisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Membro Posterior/patologia , Humanos , Camundongos , Camundongos SCID , Camundongos Transgênicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Transplante de Células-Tronco , Teratoma
6.
Gene Ther ; 11(23): 1685-93, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15470479

RESUMO

Local infusion of recombinant monocyte chemoattractant protein-1 (MCP-1) has been shown to enhance collateral artery formation in rabbit and pig hindlimb models. Owing to clinical disadvantages of protein infusion, a nonviral, liposome-based MCP-1 gene transfer was developed. Collateralization in a porcine hindlimb model served to provide a proof-of-principle for the functional benefit of MCP-1 overexpression. Development of arterial conductance as a measure of functionally relevant collateralization was evaluated in occluded as well as untreated hindlimbs in each animal. At the time of occlusion, MCP-1 and control DNA/DC-30 lipoplexes were transferred to femoral arteries of Goettingen minipigs (two therapeutic MCP-1 groups: 2 and 4 microg and one control group), using the Infiltrator local drug-delivery device. At 2 weeks following occlusion, collateralization was determined as changes in peripheral haemodynamic conductance, peripheral over aortic blood pressure ratio and angiographically visible morphology of the peripheral vessel tree. Nonviral MCP-1 gene transfer significantly improved peripheral conductance (control 11.69+/-2.78%, 2 microg 23.81+/-2.81%, P<0.05 and 4 microg 23.36+/-3.1%, P<0.05; n=12 per group) as well as the ratio of peripheral over aortic blood pressure (control 0.64+/-0.03%, 2 microg 0.75+/-0.02%, P<0.05 and 4 mug 0.75+/-0.02%, P<0.05; n=12 per group) when compared to the untreated controls 2 weeks after occlusion. Thus, it could be demonstrated for the first time that in situ overexpression of MCP-1 following local nonviral gene transfer is a potential approach to improve peripheral collateralization.


Assuntos
Arteriopatias Oclusivas/terapia , Quimiocina CCL2/genética , Circulação Colateral/genética , Artéria Femoral , Terapia Genética/métodos , Doenças Vasculares Periféricas/terapia , Animais , Arteriopatias Oclusivas/metabolismo , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Artéria Femoral/metabolismo , Expressão Gênica , Lipossomos , Doenças Vasculares Periféricas/metabolismo , Plasmídeos , Reação em Cadeia da Polimerase/métodos , Suínos , Porco Miniatura , Transfecção , Transgenes
7.
Cytotherapy ; 5(5): 399-413, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14578102

RESUMO

BACKGROUND: Cellular cardiomyoplasty is evolving as a new strategy to treat cardiac diseases. A prerequisite is a reliable source of pure cardiomyocytes, which could also help in the exploitation of recent advances in genomics and drug screening. Our goal was to establish a robust lab-scale process for the generation of embryonic stem (ES)-cell-derived cardiomyocytes in suspension. METHODS: A 71 ES cell clone carrying a construct consisting of the alpha-cardiac myosin heavy chain (alphaMHC) promoter driving the neomycin resistance gene was used for antibiotic-driven cardiomyocyte enrichment. Rotating suspension culture was established to initiate embryoid body (EB) formation. To track growth and differentiation kinetics, cell count and flow cytometry for SSEA-I, E-cadherin (stem-cell marker)and sarcomeric myosin (cardiomyocytes marker) was performed. Oct4 expression was measured via real time (RT)-PCR. RESULTS: Cultures comprising 2.5-8 x 10(6) differentiating FS cells/mL were obtained after 9 days in rotating suspension. Upon G418 addition,vigorous contracting spheres, termed cardiac bodies (CB), developed. These cultures consisted of about 2.1 x 10(5) enriched cardiomyocytes/mL after 6- 10 days of selection. Suspensions comprising 90- 95%viable single cells were generated using an improved dissociation method. Seeding of cardiomyocytes with 7 x 10(4) cell/cm(2) resulted in a homogeneous monolayer of synchronously contracting cells. Myocyte specific immunohistochemistry indicated purity of > 99%. DISCUSSION: We have established a reliable lab-scale protocol to generate cultures of highly enriched cardiomyocytes in suspension. This will facilitate development of larger-scale processes for stem-cell based cardiomyocyte supply. An improved method is provided to derive vital suspensions of cardiomyocytes, which could be utilized for transplantation as well as for drug screening purposes.


Assuntos
Técnicas de Cultura de Células/métodos , Separação Celular/métodos , Células-Tronco Multipotentes/citologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/transplante , Animais , Biomarcadores , Divisão Celular/fisiologia , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos/métodos , Cinética , Camundongos , Células-Tronco Multipotentes/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Reprodutibilidade dos Testes , Rotação , Transplante de Tecidos/métodos
8.
Tissue Eng ; 9(4): 767-78, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-13678453

RESUMO

Cardiomyocyte transplantation could offer a new approach to replace scarred, nonfunctional myocardium in a diseased heart. Clinical application of this approach would require the ability to generate large numbers of donor cells. The purpose of this study was to develop a scalable, robust, and reproducible process to derive purified cardiomyocytes from genetically engineered embryonic stem (ES) cells. ES cells transfected with a fusion gene consisting of the alpha-cardiac myosin heavy chain (MHC) promoter driving the aminoglycoside phosphotransferase (neomycin resistance) gene were used for cardiomyocyte enrichment. The transfected cells were aggregated into embyroid bodies (EBs), inoculated into stirred suspension cultures, and differentiated for 9 days before selection of cardiomyocytes by the addition of G418 with or without retinoic acid (RA). Throughout the culture period, EB and viable cell numbers were measured. In addition, flow cytometric analysis was performed to monitor sarcomeric myosin (a marker for cardiomyocytes) and Oct-4 (a marker for undifferentiated ES cells) expression. Enrichment of cardiomyocytes was achieved in cultures treated with either G418 and retinoic acid (RA) or with G418 alone. Eighteen days after differentiation, G418-selected flasks treated with RA contained approximately twice as many cells as the nontreated flasks, as well as undetectable levels of Oct-4 expression, suggesting that RA may promote cardiac differentiation and/or survival. Immunohistological and electron microscopic analysis showed that the harvested cardiomyocytes displayed many features characteristic of native cardiomyocytes. Our results demonstrate the feasibility of large-scale production of viable, ES cell-derived cardiomyocytes for tissue engineering and/or implantation, an approach that should be transferable to other ES cell derived lineages, as well as to adult stem cells with in vitro cardiomyogenic activity.


Assuntos
Diferenciação Celular/fisiologia , Miócitos Cardíacos/fisiologia , Células-Tronco/fisiologia , Engenharia Tecidual , Animais , Técnicas de Cultura de Células , Citometria de Fluxo , Complexo Principal de Histocompatibilidade/genética , Complexo Principal de Histocompatibilidade/fisiologia , Camundongos , Microscopia Eletrônica , Miócitos Cardíacos/ultraestrutura
9.
Mech Dev ; 95(1-2): 163-74, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10906459

RESUMO

To investigate functions of the homeodomain-containing transcription factor Nkx3.1 a null mutation was generated by targeted gene disruption introducing the bacterial LacZ gene as reporter into the locus. In addition to defects in duct morphogenesis of the prostate and bulbourethral gland displaying progressive epithelial hyperplasia and reduced ductal branching (Bhatia-Gaur, R., Donjacour, A.A., Sciavolino, P.J., Kim, M., Desai, N., Young, P., Norton, C.R., Gridley, T., Cardiff, R.D., Cunha, G.R., Abate-Shen, C., Shen, M.M., 1999. Genes Dev. 13, 966-977), we observed a novel phenotype in minor salivary glands of Nkx3.1 null mutants. Minor salivary glands in the oral cavity of mutant mice appeared reduced in size and exhibited severely altered duct morphology. Other Nkx3.1 expressing regions were unaffected by the mutation. The activity of the Nkx3. 1/LacZ allele faithfully reflected the known expression domains of Nkx3.1 in sclerotome, a subset of blood vessels, Rathke's pouch, and ductal epithelium in prostate and minor salivary glands during pre- and postnatal mouse development. However, it was additionally expressed in the heart, duodenum and lung. These ectopic expression domains resemble the pattern of the Nkx2.6 gene which is closely linked to Nkx3.1 in the mouse genome and its regulation may therefore be affected by the mutation. In Nkx3.1/Shh compound mutant mice we found that Nkx3.1 expression in sclerotome and prostate was strictly dependent on sonic hedgehog (Shh) signaling, while other expression domains including heart and gut were independent of Shh. Expression in lung appeared augmented in the absence of Shh. Our results suggest that Nkx3.1 plays a unique role in regulating proliferation of glandular epithelium and in the formation of ducts in prostate and minor salivary glands.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Próstata/embriologia , Glândulas Salivares/embriologia , Fatores de Transcrição/genética , Animais , Deleção de Genes , Masculino , Camundongos , Morfogênese/genética , Próstata/fisiologia , Glândulas Salivares/fisiologia
10.
Development ; 126(10): 2215-25, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10207146

RESUMO

The homeodomain transcription factor Nkx2-3 is expressed in gut mesenchyme and spleen of embryonic and adult mice. Targeted inactivation of the Nkx2-3 gene results in severe morphological alterations of both organs and early postnatal lethality in the majority of homozygous mutants. Villus formation in the small intestine appears considerably delayed in Nkx2-3(-)/- foetuses due to reduced proliferation of the epithelium, while massively increased growth of crypt cells ensues in surviving adult mutants. Interestingly, differentiated cell types of the intestinal epithelium are present in homozygous mutants, suggesting that Nkx2-3 is not required for their cell lineage allocation or migration-dependent differentiation. Hyperproliferation of the gut epithelium in adult mutants is associated with markedly reduced expression of BMP-2 and BMP-4, suggesting that these signalling molecules may be involved in mediating non-cell-autonomous control of intestinal cell growth. Spleens of Nkx2-3 mutants are generally smaller and contain drastically reduced numbers of lymphatic cells. The white pulp appears anatomically disorganized, possibly owing to a homing defect in the spleen parenchyme. Moreover, some of the Nkx2-3 mutants exhibit asplenia. Taken together these observations indicate that Nkx2-3 is essential for normal development and functions of the small intestine and spleen.


Assuntos
Proteínas Aviárias , Proteínas de Homeodomínio/fisiologia , Intestino Delgado/embriologia , Baço/embriologia , Fatores de Transcrição/fisiologia , Fator de Crescimento Transformador beta , Animais , Animais Recém-Nascidos , Proteína Morfogenética Óssea 2 , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/genética , Divisão Celular , Movimento Celular , Regulação da Expressão Gênica , Marcação de Genes , Proteínas de Homeodomínio/genética , Mucosa Intestinal , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênese , Fenótipo , Fatores de Transcrição/genética
11.
Dev Biol ; 192(1): 172-80, 1997 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9405106

RESUMO

Myf-5, a member of the family of four muscle-specific basic helix-loop-helix (bHLH) transcription factors is the first to be expressed in somites, branchial arches, and limb buds during prenatal mouse development. However, little is known about control mechanisms which actually regulate Myf-5 gene activity within these various muscle-forming domains. To identify control regions that contribute to the correct spatiotemporal activity pattern of the Myf-5 gene during mouse embryogenesis, here we report the characterization of yeast artificial chromosomes (YACs) which faithfully direct muscle-specific expression of the gene in chimeric mouse embryos. Forty-five kilobases of sequence 5' to the Myf-5 gene together with 500 kb of 3' flanking DNA drives the correct Myf-5 expression in the mesenchyme of the visceral arches and in somites but not in the hypaxial muscles of limb buds. An additional 50 kb of DNA at the 5' end is required to activate Myf-5 gene expression in developing limbs. These results demonstrate for the first time that unexpectedly distant regions of the Myf-5 gene are necessary to recapitulate its precise developmental expression pattern. We also show that Myf-5 expression in hypaxial muscles and in somites and visceral arches is regulated by separate and distinct far upstream regions. The identification of these remote regulatory elements on YACs carrying the mouse Myf-5 gene constitutes the first important step toward further dissection of the complex mechanisms by which cell-autonomous and external cues control Myf-5 expression during skeletal muscle formation in the mouse embryo.


Assuntos
Proteínas de Ligação a DNA , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Musculares/genética , Músculo Esquelético/embriologia , Músculo Esquelético/metabolismo , Transativadores , Animais , Linhagem Celular , Quimera , Cromossomos Artificiais de Levedura/genética , DNA/genética , Extremidades/embriologia , Genes Reguladores , Óperon Lac , Camundongos , Camundongos Transgênicos , Fator Regulador Miogênico 5 , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...