Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Differentiation ; 99: 62-69, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29239730

RESUMO

Corneal tissue is the most transplanted of all body tissues. Currently, cadaveric donor tissues are used for transplantation. However, a global shortage of transplant grade material has prompted development of alternative, cell-based therapies for corneal diseases. Pluripotent stem cells are attractive sources of cells for regenerative medicine, because large numbers of therapeutically useful cells can be generated. However, a detailed understanding of how to differentiate clinically relevant cell types from stem cells is fundamentally required. Periocular mesenchyme (POM), a subtype of cranial neural crest, is vital for development of multiple cell types in the cornea, including clinically relevant cells such as corneal endothelium and stromal keratocytes. Herein, we describe protocols for differentiation of POM from pluripotent stem cells. Using defined media containing inhibitors of TGFß and WNT signalling, we generated neural crest cells that express high levels of the POM transcription factors PITX2 and FOXC1. Furthermore, we identified cells resembling POM in the adult cornea, located in a niche between the trabecular meshwork and peripheral endothelium. The generation and expansion of POM is an important step in the generation of a number of cells types that could prove to be clinically useful for a number of diseases of the cornea.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias Humanas/citologia , Crista Neural/citologia , Células-Tronco Pluripotentes/citologia , Células Cultivadas , Córnea/citologia , Humanos , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo
2.
Stem Cells Transl Med ; 5(9): 1171-81, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27388238

RESUMO

UNLABELLED: : Familial osteochondritis dissecans (FOCD) is an inherited skeletal defect characterized by the development of large cartilage lesions in multiple joints, short stature, and early onset of severe osteoarthritis. It is associated with a heterozygous mutation in the ACAN gene, resulting in a Val-Met replacement in the C-type lectin domain of aggrecan. To understand the cellular pathogenesis of this condition, we studied the chondrogenic differentiation of patient bone marrow mesenchymal stromal cells (BM-MSCs). We also looked at cartilage derived from induced pluripotent stem cells (iPSCs) generated from patient fibroblasts. Our results revealed several characteristics of the differentiated chondrocytes that help to explain the disease phenotype and susceptibility to cartilage injury. First, patient chondrogenic pellets had poor structural integrity but were rich in glycosaminoglycan. Second, it was evident that large amounts of aggrecan accumulated within the endoplasmic reticulum of chondrocytes differentiated from both BM-MSCs and iPSCs. In turn, there was a marked absence of aggrecan in the extracellular matrix. Third, it was evident that matrix synthesis and assembly were globally dysregulated. These results highlight some of the abnormal aspects of chondrogenesis in these patient cells and help to explain the underlying cellular pathology. The results suggest that FOCD is a chondrocyte aggrecanosis with associated matrix dysregulation. The work provides a new in vitro model of osteoarthritis and cartilage degeneration based on the use of iPSCs and highlights how insights into disease phenotype and pathogenesis can be uncovered by studying differentiation of patient stem cells. SIGNIFICANCE: The isolation and study of patient stem cells and the development of methods for the generation of iPSCs have opened up exciting opportunities in understanding causes and exploring new treatments for major diseases. This technology was used to unravel the cellular phenotype in a severe form of inherited osteoarthritis, termed familial osteochondritis dissecans. The phenotypic abnormalities that give rise to cartilage lesions in these patients were able to be described via the generation of chondrocytes from bone marrow-derived mesenchymal stromal cells and iPSCs, illustrating the extraordinary value of these approaches in disease modeling.


Assuntos
Condrócitos/patologia , Estresse do Retículo Endoplasmático/fisiologia , Matriz Extracelular/patologia , Osteocondrite Dissecante/congênito , Adulto , Agrecanas/genética , Animais , Cartilagem/metabolismo , Técnicas de Cultura de Células/métodos , Condrócitos/metabolismo , Condrogênese/fisiologia , Humanos , Imuno-Histoquímica , Células-Tronco Pluripotentes Induzidas/citologia , Masculino , Espectrometria de Massas , Células-Tronco Mesenquimais/citologia , Camundongos , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Osteocondrite Dissecante/genética , Osteocondrite Dissecante/metabolismo , Osteocondrite Dissecante/patologia , Fenótipo
3.
Sci Rep ; 6: 28112, 2016 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-27346849

RESUMO

The high proliferation rate of embryonic stem cells (ESCs) is thought to arise partly from very low expression of p21. However, how p21 is suppressed in ESCs has been unclear. We found that p53 binds to the p21 promoter in human ESCs (hESCs) as efficiently as in differentiated human mesenchymal stem cells, however it does not promote p21 transcription in hESCs. We observed an enrichment for both the repressive histone H3K27me3 and activating histone H3K4me3 chromatin marks at the p21 locus in hESCs, suggesting it is a suppressed, bivalent domain which overrides activation by p53. Reducing H3K27me3 methylation in hESCs rescued p21 expression, and ectopic expression of p21 in hESCs triggered their differentiation. Further, we uncovered a subset of bivalent promoters bound by p53 in hESCs that are similarly induced upon differentiation in a p53-dependent manner, whereas p53 promotes the transcription of other target genes which do not show an enrichment of H3K27me3 in ESCs. Our studies reveal a unique epigenetic strategy used by ESCs to poise undesired p53 target genes, thus balancing the maintenance of pluripotency in the undifferentiated state with a robust response to differentiation signals, while utilizing p53 activity to maintain genomic stability and homeostasis in ESCs.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/genética , Histonas/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Sequência de Bases , Diferenciação Celular , Linhagem Celular , Epigênese Genética , Técnica Indireta de Fluorescência para Anticorpo , Células-Tronco Embrionárias Humanas/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Metilação , Regiões Promotoras Genéticas/genética , Ligação Proteica , Estabilidade Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ativação Transcricional , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética
4.
Elife ; 42015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-26312502

RESUMO

Hutchinson-Gilford progeria (HGPS) is a premature ageing syndrome caused by a mutation in LMNA, resulting in a truncated form of lamin A called progerin. Progerin triggers loss of the heterochromatic marker H3K27me3, and premature senescence, which is prevented by telomerase. However, the mechanism how progerin causes disease remains unclear. Here, we describe an inducible cellular system to model HGPS and find that LAP2α (lamina-associated polypeptide-α) interacts with lamin A, while its interaction with progerin is significantly reduced. Super-resolution microscopy revealed that over 50% of telomeres localize to the lamina and that LAP2α association with telomeres is impaired in HGPS. This impaired interaction is central to HGPS since increasing LAP2α levels rescues progerin-induced proliferation defects and loss of H3K27me3, whereas lowering LAP2 levels exacerbates progerin-induced defects. These findings provide novel insights into the pathophysiology underlying HGPS, and how the nuclear lamina regulates proliferation and chromatin organization.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Lamina Tipo A/metabolismo , Proteínas de Membrana/metabolismo , Progéria/patologia , Telômero/metabolismo , Humanos , Microscopia , Ligação Proteica
5.
Cell Transplant ; 24(2): 287-304, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24268186

RESUMO

Corneal endothelium-associated corneal blindness is the most common indication for corneal transplantation. Restorative corneal transplant surgery is the only option to reverse the blindness, but a global shortage of donor material remains an issue. There are immense clinical interests in the development of alternative treatment strategies to alleviate current reliance on donor materials. For such endeavors, ex vivo propagation of human corneal endothelial cells (hCECs) is required, but current methodology lacks consistency, with expanded hCECs losing cellular morphology to a mesenchymal-like transformation. In this study, we describe a novel dual media culture approach for the in vitro expansion of primary hCECs. Initial characterization included analysis of growth dynamics of hCECs grown in either proliferative (M4) or maintenance (M5) medium. Subsequent comparisons were performed on isolated hCECs cultured in M4 alone against cells expanded using the dual media approach. Further characterizations were performed using immunocytochemistry, quantitative real-time PCR, and gene expression microarray. At the third passage, results showed that hCECs propagated using the dual media approach were homogeneous in appearance, retained their unique polygonal cellular morphology, and expressed higher levels of corneal endothelium-associated markers in comparison to hCECs cultured in M4 alone, which were heterogeneous and fibroblastic in appearance. Finally, for hCECs cultured using the dual media approach, global gene expression and pathway analysis between confluent hCECs before and after 7-day exposure to M5 exhibited differential gene expression associated predominately with cell proliferation and wound healing. These findings showed that the propagation of primary hCECs using the novel dual media approach presented in this study is a consistent method to obtain bona fide hCECs. This, in turn, will elicit greater confidence in facilitating downstream development of alternative corneal endothelium replacement using tissue-engineered graft materials or cell injection therapy.


Assuntos
Proliferação de Células/efeitos dos fármacos , Meios de Cultura/farmacologia , Endotélio Corneano/metabolismo , Adolescente , Adulto , Células Cultivadas , Pré-Escolar , Regulação para Baixo , Endotélio Corneano/citologia , Endotélio Corneano/efeitos dos fármacos , Feminino , Humanos , Imuno-Histoquímica , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Regulação para Cima , Adulto Jovem
6.
Cell Stem Cell ; 15(5): 531-2, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25517458

RESUMO

Somatic cell nuclear transfer offers an alternative approach to the use of exogenous transcription factors for the reprogramming of somatic cells. But is it a better way? Two groups have performed detailed molecular comparisons between human cell lines made by the two methods and report different conclusions.


Assuntos
Impressão Genômica/genética , Taxa de Mutação , Técnicas de Transferência Nuclear , Fases de Leitura Aberta/genética , Células-Tronco Pluripotentes/citologia , Humanos , Masculino
7.
Stem Cell Reports ; 1(5): 379-86, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24286026

RESUMO

Human embryonic stem cells (hESCs) regularly acquire nonrandom genomic aberrations during culture, raising concerns about their safe therapeutic application. The International Stem Cell Initiative identified a copy number variant (CNV) amplification of chromosome 20q11.21 in 25% of hESC lines displaying a normal karyotype. By comparing four cell lines paired for the presence or absence of this CNV, we show that those containing this amplicon have higher population doubling rates, attributable to enhanced cell survival through resistance to apoptosis. Of the three genes encoded within the minimal amplicon and expressed in hESCs, only overexpression of BCL2L1 (BCL-XL isoform) provides control cells with growth characteristics similar to those of CNV-containing cells, whereas inhibition of BCL-XL suppresses the growth advantage of CNV cells, establishing BCL2L1 as a driver mutation. Amplification of the 20q11.21 region is also detectable in human embryonal carcinoma cell lines and some teratocarcinomas, linking this mutation with malignant transformation.


Assuntos
Cromossomos Humanos Par 20/genética , Variações do Número de Cópias de DNA , Células-Tronco Embrionárias/metabolismo , Seleção Genética , Proteína bcl-X/metabolismo , Linhagem Celular , Amplificação de Genes , Loci Gênicos , Humanos , Mutação , Proteína bcl-X/genética
8.
PLoS One ; 8(7): e67546, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23844023

RESUMO

Considerable interest has been generated for the development of suitable corneal endothelial graft alternatives through cell-tissue engineering, which can potentially alleviate the shortage of corneal transplant material. The advent of less invasive suture-less key-hole surgery options such as Descemet's Stripping Endothelial Keratoplasty (DSEK) and Descemet's Membrane Endothelial Keratoplasty (DMEK), which involve transplantation of solely the endothelial layer instead of full thickness cornea, provide further impetus for the development of alternative endothelial grafts for clinical applications. A major challenge for this endeavor is the lack of specific markers for this cell type. To identify genes that reliably mark corneal endothelial cells (CECs) in vivo and in vitro, we performed RNA-sequencing on freshly isolated human CECs (from both young and old donors), CEC cultures, and corneal stroma. Gene expression of these corneal cell types was also compared to that of other human tissue types. Based on high throughput comparative gene expression analysis, we identified a panel of markers that are: i) highly expressed in CECs from both young donors and old donors; ii) expressed in CECs in vivo and in vitro; and iii) not expressed in corneal stroma keratocytes and the activated corneal stroma fibroblasts. These were SLC4A11, COL8A2 and CYYR1. The use of this panel of genes in combination reliably ascertains the identity of the CEC cell type.


Assuntos
Proteínas de Transporte de Ânions/genética , Antiporters/genética , Colágeno Tipo VIII/genética , Células Endoteliais/metabolismo , Endotélio Corneano/metabolismo , Expressão Gênica , Proteínas de Membrana/genética , Adulto , Idoso , Proteínas de Transporte de Ânions/metabolismo , Antiporters/metabolismo , Autopsia , Biomarcadores/metabolismo , Colágeno Tipo VIII/metabolismo , Ceratócitos da Córnea/citologia , Ceratócitos da Córnea/metabolismo , Substância Própria/citologia , Substância Própria/metabolismo , Células Endoteliais/citologia , Endotélio Corneano/citologia , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Especificidade de Órgãos , Cultura Primária de Células
9.
J Cell Biol ; 200(5): 605-17, 2013 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-23439683

RESUMO

The nuclear lamina consists of A- and B-type lamins. Mutations in LMNA cause many human diseases, including progeria, a premature aging syndrome, whereas LMNB1 duplication causes adult-onset autosomal dominant leukodystrophy (ADLD). LMNB1 is reduced in cells from progeria patients, but the significance of this reduction is unclear. In this paper, we show that LMNB1 protein levels decline in senescent human dermal fibroblasts and keratinocytes, mediated by reduced transcription and inhibition of LMNB1 messenger ribonucleic acid (RNA) translation by miRNA-23a. This reduction is also observed in chronologically aged human skin tissue. To determine whether altered LMNB1 levels cause senescence, we either increased or reduced LMNB1. Both LMNB1 depletion and overexpression inhibited proliferation, but only LMNB1 overexpression induced senescence, which was prevented by telomerase expression or inactivation of p53. This phenotype was exacerbated by a simultaneous reduction of LMNA/C. Our results demonstrate that altering LMNB1 levels inhibits proliferation and are relevant to understanding the molecular pathology of ADLD.


Assuntos
Proliferação de Células , Senescência Celular , Fibroblastos/metabolismo , Queratinócitos/metabolismo , Lamina Tipo B/metabolismo , Diferenciação Celular , Células Cultivadas , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Fibroblastos/patologia , Genótipo , Humanos , Queratinócitos/patologia , Lamina Tipo A/metabolismo , Lamina Tipo B/genética , Proteínas de Membrana/metabolismo , MicroRNAs/metabolismo , Lâmina Nuclear/metabolismo , Doença de Pelizaeus-Merzbacher/genética , Doença de Pelizaeus-Merzbacher/metabolismo , Doença de Pelizaeus-Merzbacher/patologia , Fenótipo , Interferência de RNA , RNA Mensageiro/metabolismo , Envelhecimento da Pele , Telomerase/metabolismo , Fatores de Tempo , Transcrição Gênica , Transfecção , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
10.
Neurobiol Aging ; 34(2): 602-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22595370

RESUMO

The mitochondrial theory of aging argues that oxidative stress, caused by mitochondrial DNA mutations, is associated with decreased adenosine triphosphate (ATP) production leading to cellular degeneration. The rate of this degradation is linked to metabolic demand, with the outer retina having the greatest in the body, showing progressive inflammation, macrophage invasion, and cell loss, resulting in visual decline. Mitochondrial function shifts in vitro after 670-nm light exposure, reducing oxidative stress and increasing ATP production. In vivo, it ameliorates induced pathology. Here, we ask whether 670 nm light shifts mitochondrial function and reduces age-related retinal inflammation. Aged mice were exposed to only five 90-second exposures over 35 hours. This significantly increased mitochondrial membrane polarization and significantly reduced macrophage numbers and tumor necrosis factor (TNF)-alpha levels, a key proinflammatory cytokine. Three additional inflammatory markers were assessed; complement component 3d (C3d), a marker of chronic inflammation and calcitonin, and a systemic inflammatory biomarker were significantly reduced. Complement component 3b (C3b), a marker of acute inflammation, was not significantly altered. These results provide a simple route to combating inflammation in an aging population with declining visual function and may be applicable to clinical conditions where retinal inflammation is a key feature.


Assuntos
Inflamação/terapia , Potencial da Membrana Mitocondrial/fisiologia , Fototerapia , Retina/patologia , Animais , Calcitonina/metabolismo , Complemento C3d/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Inflamação/fisiopatologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Estresse Oxidativo/fisiologia , Retina/metabolismo , Retina/fisiopatologia , Fator de Necrose Tumoral alfa/metabolismo
11.
Nat Biotechnol ; 29(12): 1132-44, 2011 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-22119741

RESUMO

The International Stem Cell Initiative analyzed 125 human embryonic stem (ES) cell lines and 11 induced pluripotent stem (iPS) cell lines, from 38 laboratories worldwide, for genetic changes occurring during culture. Most lines were analyzed at an early and late passage. Single-nucleotide polymorphism (SNP) analysis revealed that they included representatives of most major ethnic groups. Most lines remained karyotypically normal, but there was a progressive tendency to acquire changes on prolonged culture, commonly affecting chromosomes 1, 12, 17 and 20. DNA methylation patterns changed haphazardly with no link to time in culture. Structural variants, determined from the SNP arrays, also appeared sporadically. No common variants related to culture were observed on chromosomes 1, 12 and 17, but a minimal amplicon in chromosome 20q11.21, including three genes expressed in human ES cells, ID1, BCL2L1 and HM13, occurred in >20% of the lines. Of these genes, BCL2L1 is a strong candidate for driving culture adaptation of ES cells.


Assuntos
Células-Tronco Embrionárias/citologia , Crescimento/genética , Células-Tronco Pluripotentes Induzidas/citologia , Proteínas de Ligação a RNA/metabolismo , Proteína bcl-X/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Cromossomos Humanos Par 20/genética , Evolução Clonal/genética , Metilação de DNA , Etnicidade/genética , Regulação da Expressão Gênica no Desenvolvimento , Variação Genética , Genótipo , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Proteína 1 Inibidora de Diferenciação/metabolismo , Polimorfismo de Nucleotídeo Único , Proteínas de Ligação a RNA/genética , Seleção Genética/genética , Proteína bcl-X/genética
12.
Cell Stem Cell ; 9(2): 156-65, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21816366

RESUMO

Somatic tissues in female eutherian mammals are mosaic due to random X inactivation. In contrast to mice, X chromosome reactivation does not occur during the reprogramming of human female somatic cells to induced pluripotent stem cells (iPSCs), although this view is contested. Using balanced populations of female Rett patient and control fibroblasts, we confirm that all cells in iPSC colonies contain an inactive X, and additionally find that all colonies made from the same donor fibroblasts contain the same inactive X chromosome. Notably, this extreme "skewing" toward a particular dominant, active X is also a general feature of primary female fibroblasts during proliferation, and the skewing seen in reprogramming and fibroblast culture can be alleviated by overexpression of telomerase. These results have important implications for in vitro modeling of X-linked diseases and the interpretation of long-term culture studies in cancer and senescence using primary female fibroblast cell lines.


Assuntos
Reprogramação Celular/genética , Cromossomos Humanos X/metabolismo , Telomerase/metabolismo , Animais , Sequência de Bases , Proliferação de Células , Células Cultivadas , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Inativação do Cromossomo X
13.
Cell Stem Cell ; 8(1): 31-45, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21185252

RESUMO

The segmental premature aging disease Hutchinson-Gilford Progeria syndrome (HGPS) is caused by a truncated and farnesylated form of Lamin A called progerin. HGPS affects mesenchymal lineages, including the skeletal system, dermis, and vascular smooth muscle (VSMC). To understand the underlying molecular pathology of HGPS, we derived induced pluripotent stem cells (iPSCs) from HGPS dermal fibroblasts. The iPSCs were differentiated into neural progenitors, endothelial cells, fibroblasts, VSMCs, and mesenchymal stem cells (MSCs). Progerin levels were highest in MSCs, VSMCs, and fibroblasts, in that order, with these lineages displaying increased DNA damage, nuclear abnormalities, and HGPS-VSMC accumulating numerous calponin-staining inclusion bodies. Both HGPS-MSC and -VSMC viability was compromised by stress and hypoxia in vitro and in vivo (MSC). Because MSCs reside in low oxygen niches in vivo, we propose that, in HGPS, this causes additional depletion of the MSC pool responsible for replacing differentiated cells lost to progerin toxicity.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Mesenquimais/patologia , Músculo Liso Vascular/patologia , Senilidade Prematura/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Fibroblastos/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Músculo Liso Vascular/citologia , Oxigênio/metabolismo , Progéria/patologia , Progéria/terapia
14.
EMBO Rep ; 10(7): 714-21, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19543232

RESUMO

For much of the last century, the differentiated state that characterizes the many cell types of an adult organism was thought to be stable and abrogated only in rare instances by transdifferentiation, metaplasia or cancer. This stability was thought to reside in the autoregulatory molecular circuitry that exists between the cytoplasm and the nucleus, a status quo that could be disrupted during somatic cell nuclear transfer, to reprogramme cells to a pluripotent state. Pioneering work in the 1980s showed that transdifferentiation of cell lineages could be induced by the addition of transcription factors. However, these conversions were usually confined to cell types from the same germ layer, and proof of conversion was difficult to obtain. This deficiency has now been overturned by demonstrations that exogenously added transcription factors can convert differentiated cell types into embryonic-like induced pluripotent stem cells. Here, we highlight the recent progress, and the implications of this work for our understanding of the relationship between the pluripotent and more differentiated cell states.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes/citologia , Animais , Reprogramação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Humanos , Cinética , Células-Tronco Pluripotentes/metabolismo
16.
Differentiation ; 76(9): 958-70, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18557764

RESUMO

Many applications of human embryonic stem cells (hESCs) will require fully defined growth and differentiation conditions including media devoid of fetal calf serum. To identify factors that control lineage differentiation we have analyzed a serum-free (SF) medium conditioned by the cell line END2, which efficiently induces hESCs to form cardiomyocytes. Firstly, we noted that insulin, a commonly used medium supplement, acted as a potent inhibitor of cardiomyogenesis in multiple hESC lines and was rapidly cleared by medium conditioning. In the presence of insulin or IGF-1, which also suppressed cardiomyocyte differentiation, the PI3/Akt pathway was activated in undifferentiated hESC, suggesting that insulin/IGF-1 effects were mediated by this signaling cascade. Time course analysis and quantitative RT-PCR revealed impaired expression of endoderm and mesoderm markers in the presence of insulin, particularly if added during early stages of hESC differentiation. Relatively high levels of the neural ectoderm marker Sox1 were expressed under these conditions. Secondly, comparative gene expression showed that two key enzymes in the prostaglandin I2 (PGI2) synthesis pathway were highly up-regulated in END2 cells compared with a related, but non-cardiogenic, cell line. Biochemical analysis confirmed 6-10-fold higher PGI2 levels in END2 cell-conditioned medium (END2-CM) vs. controls. Optimized concentrations of PGI2 in a fully synthetic, insulin-free medium resulted in a cardiogenic activity equivalent to END2-CM. Addition of the p38 mitogen-activated protein kinase-inhibitor SB203580, which we have shown previously to enhance hESC cardiomyogenesis, to these insulin-free and serum-free conditions resulted in a cardiomyocyte content of >10% in differentiated cultures without any preselection. This study represents a significant step toward developing scalable production for cardiomyocytes from hESC using clinically compliant reagents compatible with Good Manufacturing Practice.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Miócitos Cardíacos/citologia , Células Cultivadas , Meios de Cultivo Condicionados , Meios de Cultura Livres de Soro , Células-Tronco Embrionárias/metabolismo , Epoprostenol/metabolismo , Humanos , Imidazóis/farmacologia , Insulina/metabolismo , Insulina/farmacologia , Miócitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/farmacologia , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
J Mol Cell Cardiol ; 43(4): 504-16, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17707399

RESUMO

Human embryonic stem cell (hESC)-derived cardiomyocytes are a promising cell source for cardiac repair. Whether these cells can be transported long distance, survive, and mature in hearts subjected to ischemia/reperfusion with minimal infarction is unknown. Taking advantage of a constitutively GFP-expressing hESC line we investigated whether hESC-derived cardiomyocytes could be shipped and subsequently form grafts when transplanted into the left ventricular wall of athymic nude rats subjected to ischemia/reperfusion with minimal infarction. Co-localization of GFP-epifluorescence and cardiomyocyte-specific marker staining was utilized to analyze hESC-derived cardiomyocyte fate in a rat ischemia/reperfused myocardium. Differentiated, constitutively green fluorescent protein (GFP)-expressing hESCs (hES3-GFP; Envy) containing about 13% cardiomyocytes were differentiated in Singapore, and shipped in culture medium at 4 degrees C to Los Angeles (shipping time approximately 3 days). The cells were dissociated and a cell suspension (2 x 10(6) cells for each rat, n=10) or medium (n=10) was injected directly into the myocardium within the ischemic risk area 5 min after left coronary artery occlusion in athymic nude rats. After 15 min of ischemia, the coronary artery was reperfused. The hearts were harvested at various time points later and processed for histology, immunohistochemical staining, and fluorescence microscopy. In order to assess whether the hESC-derived cardiomyocytes might evade immune surveillance, 2 x 10(6) cells were injected into immune competent Sprague-Dawley rat hearts (n=2), and the hearts were harvested at 4 weeks after cell injection and examined as in the previous procedures. Even following 3 days of shipping, the hESC-derived cardiomyocytes within embryoid bodies (EBs) showed active and rhythmic contraction after incubation in the presence of 5% CO(2) at 37 degrees C. In the nude rats, following cell implantation, H&E, immunohistochemical staining and GFP epifluorescence demonstrated grafts in 9 out of 10 hearts. Cells that demonstrated GFP epifluorescence also stained positive (co-localized) for the muscle marker alpha-actinin and exhibited cross striations (sarcomeres). Furthermore, cells that stained positive for the antibody to GFP (immunohistochemistry) also stained positive for the muscle marker sarcomeric actin and demonstrated cross striations. At 4 weeks engrafted hESCs expressed connexin 43, suggesting the presence of nascent gap junctions between donor and host cells. No evidence of rejection was observed in nude rats as determined by inspection for lymphocytic infiltrate and/or giant cells. In contrast, hESC-derived cardiomyocytes injected into immune competent Sprague-Dawley rats resulted in an overt lymphocytic infiltrate. hESCs-derived cardiomyocytes can survive several days of shipping. Grafted cells survived up to 4 weeks after transplantation in hearts of nude rats subjected to ischemia/reperfusion with minimal infarction. They continued to express cardiac muscle markers and exhibit sarcomeric structure and they were well interspersed with the endogenous myocardium. However, hESC-derived cells did not escape immune surveillance in the xenograft setting in that they elicited a rejection phenomenon in immune competent rats.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/transplante , Isquemia Miocárdica/terapia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Animais , Sobrevivência Celular , Células Cultivadas , Conexina 43/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Isquemia Miocárdica/patologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Ratos , Ratos Nus , Transplante de Células-Tronco , Transplante Heterólogo
19.
Stem Cells Dev ; 16(1): 75-89, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17348806

RESUMO

The absence of efficient and directed methods for the differentiation of adult pancreatic progenitor cell populations to pancreatic islet cells has raised doubts concerning the regeneration potential inherent in the adult pancreas. Relatively low levels of islet cell differentiation have been reported using adult pancreatic cells in vivo and in vitro. In the present study, we initially enriched for a nonendocrine epithelial component of the adult human pancreas and defined conditions that are permissive to islet cell differentiation in vitro. Sequential progression of cell differentiation in the permissive conditions allowed for incremental evaluation of changes occurring in the cell population. Optimization of the differentiation process, for the efficient production of islet endocrine cells, was accomplished by identifying specific factors and culture conditions that increased islet progenitor production 250-fold. Ultimately, 85% percent of the nonendocrine epithelial cells isolated from human pancreatic tissue and cultured in the optimized conditions for 8 days, readily re-expressed pancreatic duodenal homeobox-1 (Pdx1). Sixty-five percent of these Pdx1-expressing cells were capable of additional islet endocrine cell differentiation. This represents a significant advancement in the differentiation of an adult pancreatic progenitor cell population in vitro and suggests that the nonendocrine compartment of the human pancreas remains an important cell resource for the generation of transplantable islets to treat diabetes.


Assuntos
Células-Tronco Adultas/citologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Ilhotas Pancreáticas/citologia , Ductos Pancreáticos/citologia , Adolescente , Adulto , Criança , Células Epiteliais/citologia , Proteínas de Homeodomínio/genética , Humanos , Transativadores/genética
20.
Trends Biotechnol ; 25(1): 24-32, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17084475

RESUMO

Recent developments in the identification, in vitro culture and differentiation of stem cells point to the unprecedented potential of these cells, or their derivatives, to cure degenerative disorders. Human embryonic stem cells (hESC) offer the particular advantage of prolonged proliferative capacity and great versatility in the lineages that can be formed in culture. Translating these advantages into clinical benefits faces many challenges, including efficient differentiation into the desired cell type(s), maintaining genetic stability during long-term culture and, finally, ensuring the absence of potentially tumorigenic hESC from the final product. It is this final safety issue that will form the focus of this review.


Assuntos
Transformação Celular Neoplásica , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células-Tronco Embrionárias/transplante , Segurança , Transplante de Células-Tronco/efeitos adversos , Técnicas de Cultura de Células , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/imunologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA