Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Ann Rheum Dis ; 82(12): 1547-1557, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37679035

RESUMO

OBJECTIVES: Progressive pseudorheumatoid arthropathy of childhood (PPAC), caused by deficiency of WNT1 inducible signalling pathway protein 3 (WISP3), has been challenging to study because no animal model of the disease exists and cartilage recovered from affected patients is indistinguishable from common end-stage osteoarthritis. Therefore, to gain insights into why precocious articular cartilage failure occurs in this disease, we made in vitro derived articular cartilage using isogenic WISP3-deficient and WISP3-sufficient human pluripotent stem cells (hPSCs). METHODS: We generated articular cartilage-like tissues from induced-(i) PSCs from two patients with PPAC and one wild-type human embryonic stem cell line in which we knocked out WISP3. We compared these tissues to in vitro-derived articular cartilage tissues from two isogenic WISP3-sufficient control lines using histology, bulk RNA sequencing, single cell RNA sequencing and in situ hybridisation. RESULTS: WISP3-deficient and WISP3-sufficient hPSCs both differentiated into articular cartilage-like tissues that appeared histologically similar. However, the transcriptomes of WISP3-deficient tissues differed significantly from WISP3-sufficient tissues and pointed to increased TGFß, TNFα/NFκB, and IL-2/STAT5 signalling and decreased oxidative phosphorylation. Single cell sequencing and in situ hybridisation revealed that WISP3-deficient cartilage contained a significantly higher fraction (~4 fold increase, p<0.001) of superficial zone chondrocytes compared with deeper zone chondrocytes than did WISP3-sufficient cartilage. CONCLUSIONS: WISP3-deficient and WISP3-sufficient hPSCs can be differentiated into articular cartilage-like tissues, but these tissues differ in their transcriptomes and in the relative abundances of chondrocyte subtypes they contain. These findings provide important starting points for in vivo studies when an animal model of PPAC or presymptomatic patient-derived articular cartilage becomes available.


Assuntos
Cartilagem Articular , Células-Tronco Pluripotentes , Animais , Humanos , Cartilagem Articular/metabolismo , Mutação , Condrócitos/metabolismo , Diferenciação Celular/genética
2.
Development ; 144(1): 12-16, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28049688

RESUMO

Over 100 scientists with common interests in human development, disease and regeneration gathered in late September 2016 for The Company of Biologists' second 'From Stem Cells to Human Development' meeting held in historic Southbridge. In this Meeting Review, we highlight some of the exciting new findings that were presented, and discuss emerging themes and convergences in human development and disease that arose during these discussions.


Assuntos
Diferenciação Celular , Pesquisas com Embriões , Embriologia , Desenvolvimento Embrionário/fisiologia , Células-Tronco Embrionárias/fisiologia , Pesquisa Translacional Biomédica , Animais , Diferenciação Celular/genética , Embriologia/métodos , Embriologia/tendências , Humanos , Pesquisa Translacional Biomédica/métodos , Pesquisa Translacional Biomédica/tendências
3.
Development ; 140(12): 2597-610, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23715552

RESUMO

Osteoarthritis primarily affects the articular cartilage of synovial joints. Cell and/or cartilage replacement is a promising therapy, provided there is access to appropriate tissue and sufficient numbers of articular chondrocytes. Embryonic stem cells (ESCs) represent a potentially unlimited source of chondrocytes and tissues as they can generate a broad spectrum of cell types under appropriate conditions in vitro. Here, we demonstrate that mouse ESC-derived chondrogenic mesoderm arises from a Flk-1(-)/Pdgfrα(+) (F(-)P(+)) population that emerges in a defined temporal pattern following the development of an early cardiogenic F(-)P(+) population. Specification of the late-arising F(-)P(+) population with BMP4 generated a highly enriched population of chondrocytes expressing genes associated with growth plate hypertrophic chondrocytes. By contrast, specification with Gdf5, together with inhibition of hedgehog and BMP signaling pathways, generated a population of non-hypertrophic chondrocytes that displayed properties of articular chondrocytes. The two chondrocyte populations retained their hypertrophic and non-hypertrophic properties when induced to generate spatially organized proteoglycan-rich cartilage-like tissue in vitro. Transplantation of either type of chondrocyte, or tissue generated from them, into immunodeficient recipients resulted in the development of cartilage tissue and bone within an 8-week period. Significant ossification was not observed when the tissue was transplanted into osteoblast-depleted mice or into diffusion chambers that prevent vascularization. Thus, through stage-specific manipulation of appropriate signaling pathways it is possible to efficiently and reproducibly derive hypertrophic and non-hypertrophic chondrocyte populations from mouse ESCs that are able to generate distinct cartilage-like tissue in vitro and maintain a cartilage tissue phenotype within an avascular and/or osteoblast-free niche in vivo.


Assuntos
Cartilagem Articular/citologia , Condrócitos/citologia , Condrogênese , Células-Tronco Embrionárias/citologia , Animais , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Cartilagem Articular/metabolismo , Diferenciação Celular , Linhagem da Célula , Condrócitos/metabolismo , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/transplante , Feminino , Fator 5 de Diferenciação de Crescimento/genética , Fator 5 de Diferenciação de Crescimento/metabolismo , Hipertrofia/metabolismo , Imuno-Histoquímica , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Osteoblastos/metabolismo , Osteogênese , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Fatores de Tempo
4.
J Orthop Res ; 42(8): 1841-1851, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38433390

RESUMO

The ideal cell source for articular cartilage repair remains elusive. Using developmentally inspired differentiation protocols, we induced human pluripotent stem cells (hPSCs) toward articular chondrocytes capable of joint cartilage repair in rodent models, which were distinct from growth plate chondrocytes, fated to be replaced by bone in vivo. Working toward clinical translation, we demonstrated controlled differentiation into chondrocytes by comprehensive gene expression analysis at each step of the differentiation. Articular chondrocytes derived from hPSCs could be expanded several passages in vitro without losing chondrogenic potential. Furthermore, chondrocytes isolated from these articular cartilage tissues had the potential to serially regenerate new articular and growth plate cartilage tissues. Finally, the ability to cryopreserve articular chondrocytes with the desired phenotype is critical for clinical translation and here we report no loss in cell viability or regenerative potential following cryopreservation. These results support the immense potential of hPSC-derived articular chondrocytes as a cell-based therapy for cartilage repair.


Assuntos
Cartilagem Articular , Diferenciação Celular , Condrócitos , Células-Tronco Pluripotentes , Condrócitos/fisiologia , Condrócitos/citologia , Humanos , Cartilagem Articular/citologia , Cartilagem Articular/fisiologia , Células-Tronco Pluripotentes/fisiologia , Células-Tronco Pluripotentes/citologia , Animais , Regeneração/fisiologia , Criopreservação
5.
bioRxiv ; 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38915712

RESUMO

Cartilage plays a crucial role in skeletal development and function, and abnormal development contributes to genetic and age-related skeletal disease. To better understand how human cartilage develops in vivo , we jointly profiled the transcriptome and open chromatin regions in individual nuclei recovered from distal femurs at 2 fetal timepoints. We used these multiomic data to identify transcription factors expressed in distinct chondrocyte subtypes, link accessible regulatory elements with gene expression, and predict transcription factor-based regulatory networks that are important for growth plate or epiphyseal chondrocyte differentiation. We developed a human pluripotent stem cell platform for interrogating the function of predicted transcription factors during chondrocyte differentiation and used it to test NFATC2 . We expect new regulatory networks we uncovered using multiomic data to be important for promoting cartilage health and treating disease, and our platform to be a useful tool for studying cartilage development in vitro . Statement of Significance: The identity and integrity of the articular cartilage lining our joints are crucial to pain-free activities of daily living. Here we identified a gene regulatory landscape of human chondrogenesis at single cell resolution, which is expected to open new avenues of research aimed at mitigating cartilage diseases that affect hundreds of millions of individuals world-wide.

6.
Dev Cell ; 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39216481

RESUMO

Tendons, which transmit force from muscles to bones, are highly prone to injury. Understanding the mechanisms driving tendon fate would impact efforts to improve tendon healing, yet this knowledge is limited. To find direct regulators of tendon progenitor emergence, we performed a zebrafish high-throughput chemical screen. We established forskolin as a tenogenic inducer across vertebrates, functioning through Creb1a, which is required and sufficient for tendon fate. Putative enhancers containing cyclic AMP (cAMP) response elements (CREs) in humans, mice, and fish drove specific expression in zebrafish cranial and fin tendons. Analysis of these genomic regions identified motifs for early B cell factor (Ebf/EBF) transcription factors. Mutation of CRE or Ebf/EBF motifs significantly disrupted enhancer activity and specificity in tendons. Zebrafish ebf1a/ebf3a mutants displayed defects in tendon formation. Notably, Creb1a/CREB1 and Ebf1a/Ebf3a/EBF1 overexpression facilitated tenogenic induction in zebrafish and human pluripotent stem cells. Together, our work identifies the functional conservation of two transcription factors in promoting tendon fate.

7.
bioRxiv ; 2023 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-37066225

RESUMO

Objectives: Progressive Pseudorheumatoid Arthropathy of Childhood (PPAC), caused by deficiency of WNT1 inducible signaling pathway protein 3 ( WISP3 ), has been challenging to study because no animal model of the disease exists and cartilage recovered from affected patients is indistinguishable from common end-stage osteoarthritis. Therefore, to gain insights into why precocious articular cartilage failure occurs in this disease, we made in vitro derived articular cartilage using isogenic WISP3 -deficient and WISP3 -sufficient human pluripotent stem cells (hPSCs). Methods: We generated articular cartilage-like tissues from induced-(i)PSCs from 2 patients with PPAC and 1 wild-type human embryonic stem cell line in which we knocked out WISP3. We compared these tissues to in vitro -derived articular cartilage tissues from 2 isogenic WISP3 -sufficient control lines using histology, bulk RNA sequencing, single cell RNA sequencing, and in situ hybridization. Results: WISP3 -deficient and WISP3 -sufficient hPSCs both differentiated into articular cartilage-like tissues that appeared histologically similar. However, the transcriptomes of WISP3 -deficient tissues differed significantly from WISP3 -sufficient tissues and pointed to increased TGFß, TNFα/NFkB, and IL-2/STAT5 signaling and decreased oxidative phosphorylation. Single cell sequencing and in situ hybridization revealed that WISP3 -deficient cartilage contained a significantly higher fraction (∼ 4-fold increase, p < 0.001) of superficial zone chondrocytes compared to deeper zone chondrocytes than did WISP3 -sufficient cartilage. Conclusions WISP3 -deficient and WISP3 -sufficient hPSCs can be differentiated into articular cartilage-like tissues, but these tissues differ in their transcriptomes and in the relative abundances of chondrocyte sub-types they contain. These findings provide important starting points for in vivo studies when an animal model of PPAC or presymptomtic patient-derived articular cartilage becomes available. KEY MESSAGES: What is already known on this topic: Loss-of-function mutations in WISP3 cause Progressive Pseudorheumatoid Arthropathy of Childhood (PPAC), yet the precise function of WISP3 in cartilage is unknown due to the absence of cartilage disease Wisp3 knockout mice and the lack of available PPAC patient cartilage that is not end-stage. Thus, most functional studies of WISP3 have been performed in vitro using WISP3 over-expressing cell lines (i.e., not wild-type) and WISP3 -deficient chondrocytes. What this study adds: We describe 3 new WISP3 -deficient human pluripotent stem cell (hPSC) lines and show they can be differentiated into articular cartilage-like tissue. We compare in vitro -derived articular cartilage made from WISP3 -deficient and isogenic WISP3 - sufficient hPSCs using bulk RNA sequencing, single cell RNA sequencing, and in situ hybridization. We observe significant differences in the expression of genes previously associated with cartilage formation and homeostasis in the TGFß, TNFα/NFkB, and IL-2/STAT5 signaling pathways. We also observe that WISP3-deficient cartilage-like tissues contain significantly higher fractions of chondrocytes that express superficial zone transcripts. These data suggest precocious cartilage failure in PPAC is the result of abnormal articular cartilage formation, dysregulated homeostatic signaling, or both.How this study might affect research, practice or policy: This study uses in vitro -derived articular cartilage to generate hypotheses for why cartilage fails in children with PPAC. This work prioritizes downstream studies to be performed when pre-symptomatic patient-derived cartilage samples or animal model of PPAC becomes available. It is essential to know how WISP3 functions in cartilage to develop therapies that benefit patients with PPAC and other degenerative joint diseases.

8.
Elife ; 122023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36920035

RESUMO

To address large gaps in our understanding of the molecular regulation of articular and growth plate cartilage development in humans, we used our directed differentiation approach to generate these distinct cartilage tissues from human embryonic stem cells. The resulting transcriptomic profiles of hESC-derived articular and growth plate chondrocytes were similar to fetal epiphyseal and growth plate chondrocytes, with respect to genes both known and previously unknown to cartilage biology. With the goal to characterize the regulatory landscapes accompanying these respective transcriptomes, we mapped chromatin accessibility in hESC-derived chondrocyte lineages, and mouse embryonic chondrocytes, using ATAC-sequencing. Integration of the expression dataset with the differentially accessible genomic regions revealed lineage-specific gene regulatory networks. We validated functional interactions of two transcription factors (TFs) (RUNX2 in growth plate chondrocytes and RELA in articular chondrocytes) with their predicted genomic targets. The maps we provide thus represent a framework for probing regulatory interactions governing chondrocyte differentiation. This work constitutes a substantial step towards comprehensive and comparative molecular characterizations of distinct chondrogenic lineages and sheds new light on human cartilage development and biology.


Assuntos
Cartilagem , Condrócitos , Humanos , Animais , Camundongos , Condrócitos/metabolismo , Diferenciação Celular/genética , Lâmina de Crescimento , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Condrogênese/genética
9.
Cell Stem Cell ; 29(12): 1615-1616, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36459962

RESUMO

Fang and colleagues provide a comprehensive transcriptomic analysis of the cell types occupying the interface between tendon and bone, the enthesis. They establish a framework for understanding enthesis maturation and identify a potent Gli1-lineage progenitor with clonogenicity and multipotency that improves enthesis healing in an adult injury model.


Assuntos
Perfilação da Expressão Gênica , Cicatrização , Adulto , Humanos
10.
J Virol ; 84(14): 7360-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20463073

RESUMO

Viral vector-based gene expression libraries from normal or diseased tissues offer opportunities to interrogate cellular functions that influence or participate directly in specific biological processes. Here we report the creation and characterization of a herpes simplex virus (HSV)-based expression library consisting of cDNAs derived from PC12 pheochromocytoma cells. A replication-defective HSV vector backbone was engineered to contain both a bacterial artificial chromosome (BAC) and the Invitrogen in vitro Gateway recombination system, creating DBAC-GW. A cDNA library was produced and transferred into the DBAC-GW genome by in vitro recombination and selection in bacteria to produce DBAC-L. DBAC-L contained at least 15,000 unique cDNAs, as shown by DNA array analysis of PCR-amplified cDNA inserts, representing a wide range of cancer- and neuron-related cellular functions. Transfection of the recombinant DBAC-L DNA into complementing animal cells produced more than 1 million DBAC-L virus particles representing the library genes. By microarray analysis of vector-infected cells, we observed that individual members of this vector population expressed unique PC12 cDNA-derived mRNA, demonstrating the power of this system to transfer and express a variety of gene activities. We discuss the potential utility of this and similarly derived expression libraries for genome-wide approaches to identify cellular functions that participate in complex host-pathogen interactions or processes related to disease and to cell growth and development.


Assuntos
Expressão Gênica , Biblioteca Gênica , Vetores Genéticos , Simplexvirus/genética , Animais , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Células PC12 , Ratos
11.
Stem Cells ; 26(12): 3119-29, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18787207

RESUMO

The ability of embryonic stem cells to develop into multiple cell lineages provides a powerful resource for tissue repair and regeneration. Gene transfer offers a means to dissect the complex events in lineage determination but is limited by current delivery systems. We designed a high-efficiency replication-defective herpes simplex virus gene transfer vector (JDbetabeta) for robust and transient expression of the transcription factors Pax3 and MyoD, which are known to be involved in skeletal muscle differentiation. JDbetabeta-mediated expression of each gene in day 4 embryoid bodies (early-stage mesoderm) resulted in the induction of unique alterations in gene expression profiles, including the upregulation of known target genes relevant to muscle and neural crest development, whereas a control enhanced green fluorescent protein expression vector was relatively inert. This vector delivery system holds great promise for the use of gene transfer to analyze the impact of specific genes on both regulatory genetic events and commitment of stem cells to particular lineages.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/virologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Proteína MyoD/biossíntese , Fatores de Transcrição Box Pareados/biossíntese , Simplexvirus/metabolismo , Animais , Linhagem Celular Tumoral , Linhagem da Célula , Chlorocebus aethiops , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/metabolismo , Humanos , Músculo Esquelético/metabolismo , Fator de Transcrição PAX3 , Células Vero
12.
Stem Cell Res ; 31: 201-215, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30118958

RESUMO

Directed differentiation of pluripotent stem cells provides an accessible system to model development. However, the distinct cell types that emerge, their dynamics, and their relationship to progenitors in the early embryo has been difficult to decipher because of the cellular heterogeneity inherent to differentiation. Here, we used a combination of bulk RNA-Seq, single cell RNA-Seq, and bioinformatics analyses to dissect the cell types that emerge during directed differentiation of mouse embryonic stem cells as embryoid bodies and we compared them to spatially and temporally resolved transcriptional profiles of early embryos. Our single cell analyses of the day 4 embryoid bodies revealed three populations which had retained related yet distinct pluripotent signatures that resemble the pre- or post-implantation epiblast, one population of presumptive neuroectoderm, one population of mesendoderm, and four populations of neural progenitors. By day 6, the neural progenitors predominated the embryoid bodies, but both a small population of pluripotent-like cells and an anterior mesoderm-like Brachyury-expressing population were present. By comparing the day 4 and day 6 populations, we identified candidate differentiation paths, transcription factors, and signaling pathways that mark the in vitro correlate of the transition from the mid-to-late primitive streak stage.


Assuntos
Corpos Embrioides/fisiologia , Desenvolvimento Embrionário/fisiologia , Mesoderma/metabolismo , Diferenciação Celular , Humanos
13.
J Clin Invest ; 126(5): 1649-63, 2016 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-27018594

RESUMO

Both the WNT/ß-catenin and hedgehog signaling pathways are important in the regulation of limb development, chondrocyte differentiation, and degeneration of articular cartilage in osteoarthritis (OA). It is not clear how these signaling pathways interact in interzone cell differentiation and synovial joint morphogenesis. Here, we determined that constitutive activation of hedgehog signaling specifically within interzone cells induces joint morphological changes by selectively inhibiting ß-catenin-induced Fgf18 expression. Stabilization of ß-catenin or treatment with FGF18 rescued hedgehog-induced phenotypes. Hedgehog signaling induced expression of a dominant negative isoform of TCF7L2 (dnTCF7L2) in interzone progeny, which may account for the selective regulation of ß-catenin target genes observed. Knockdown of TCF7L2 isoforms in mouse chondrocytes rescued hedgehog signaling-induced Fgf18 downregulation, while overexpression of the human dnTCF7L2 orthologue (dnTCF4) in human chondrocytes promoted the expression of catabolic enzymes associated with OA. Similarly, expression of dnTCF4 in human chondrocytes positively correlated with the aggrecanase ADAMTS4. Consistent with our developmental findings, activation of ß-catenin also attenuated hedgehog-induced or surgically induced articular cartilage degeneration in mouse models of OA. Thus, our results demonstrate that hedgehog inhibits selective ß-catenin target gene expression to direct interzone progeny fates and articular cartilage development and disease. Moreover, agents that increase ß-catenin activity have the potential to therapeutically attenuate articular cartilage degeneration as part of OA.


Assuntos
Condrócitos/imunologia , Proteínas Hedgehog/imunologia , Osteoartrite/imunologia , Membrana Sinovial/imunologia , Via de Sinalização Wnt/imunologia , beta Catenina/imunologia , Proteína ADAMTS4/genética , Proteína ADAMTS4/imunologia , Animais , Condrócitos/patologia , Modelos Animais de Doenças , Regulação para Baixo/imunologia , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/imunologia , Proteínas Hedgehog/genética , Humanos , Camundongos , Camundongos Knockout , Osteoartrite/genética , Osteoartrite/patologia , Membrana Sinovial/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/imunologia , Via de Sinalização Wnt/genética , beta Catenina/genética
14.
Nat Biotechnol ; 33(6): 638-45, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25961409

RESUMO

The replacement of articular cartilage through transplantation of chondrogenic cells or preformed cartilage tissue represents a potential new avenue for the treatment of degenerative joint diseases. Although many studies have described differentiation of human pluripotent stem cells (hPSCs) to the chondrogenic lineage, the generation of chondrocytes able to produce stable articular cartilage in vivo has not been demonstrated. Here we show that activation of the TGFß pathway in hPSC-derived chondrogenic progenitors promotes the efficient development of articular chondrocytes that can form stable cartilage tissue in vitro and in vivo. In contrast, chondrocytes specified by BMP4 signaling display characteristics of hypertrophy and give rise to cartilage tissues that initiate the endochondral ossification process in vivo. These findings provide a simple serum-free and efficient approach for the routine generation of hPSC-derived articular chondrocytes for modeling diseases of the joint and developing cell therapy approaches to treat them.


Assuntos
Cartilagem Articular/citologia , Diferenciação Celular/genética , Artropatias/terapia , Células-Tronco Pluripotentes/transplante , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Cartilagem Articular/crescimento & desenvolvimento , Condrócitos/citologia , Humanos , Artropatias/patologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Pluripotentes/citologia , Transdução de Sinais/genética
16.
Nat Biotechnol ; 29(11): 1011-8, 2011 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-22020386

RESUMO

To identify cell-surface markers specific to human cardiomyocytes, we screened cardiovascular cell populations derived from human embryonic stem cells (hESCs) against a panel of 370 known CD antibodies. This screen identified the signal-regulatory protein alpha (SIRPA) as a marker expressed specifically on cardiomyocytes derived from hESCs and human induced pluripotent stem cells (hiPSCs), and PECAM, THY1, PDGFRB and ITGA1 as markers of the nonmyocyte population. Cell sorting with an antibody against SIRPA allowed for the enrichment of cardiac precursors and cardiomyocytes from hESC/hiPSC differentiation cultures, yielding populations of up to 98% cardiac troponin T-positive cells. When plated in culture, SIRPA-positive cells were contracting and could be maintained over extended periods of time. These findings provide a simple method for isolating populations of cardiomyocytes from human pluripotent stem cell cultures, and thereby establish a readily adaptable technology for generating large numbers of enriched cardiomyocytes for therapeutic applications.


Assuntos
Antígenos de Diferenciação/metabolismo , Separação Celular/métodos , Citometria de Fluxo/métodos , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/citologia , Receptores Imunológicos/metabolismo , Antígenos de Superfície/análise , Biomarcadores/metabolismo , Diferenciação Celular , Regulação da Expressão Gênica , Humanos , Miócitos Cardíacos/metabolismo , Células-Tronco Pluripotentes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA