Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 185(2): 235-249, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-34995481

RESUMO

How cells become specialized, or "mature," is important for cell and developmental biology. While maturity is usually deemed a terminal fate, it may be more helpful to consider maturation not as a switch but as a dynamic continuum of adaptive phenotypic states set by genetic and environment programing. The hallmarks of maturity comprise changes in anatomy (form, gene circuitry, and interconnectivity) and physiology (function, rhythms, and proliferation) that confer adaptive behavior. We discuss efforts to harness their chemical (nutrients, oxygen, and growth factors) and physical (mechanical, spatial, and electrical) triggers in vitro and in vivo and how maturation strategies may support disease research and regenerative medicine.


Assuntos
Diferenciação Celular , Animais , Pesquisa Biomédica , Proliferação de Células , Humanos , Modelos Biológicos
2.
Cell ; 161(1): 12-17, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25815980

RESUMO

Nutrition presents unanswered scientific questions of high public health importance. We envision model systems composed of interacting gastrointestinal and metabolic tissues derived from human embryonic stem cells, populated by gut microbiota. The culture will be embedded in 3D scaffolds, creating a controlled experimental system that enables tissue sampling and imaging.


Assuntos
Células-Tronco Embrionárias/citologia , Trato Gastrointestinal/fisiologia , Modelos Biológicos , Animais , Dieta , Trato Gastrointestinal/microbiologia , Humanos , Modelos Animais , Fenômenos Fisiológicos da Nutrição
3.
Cell ; 159(2): 428-39, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25303535

RESUMO

The generation of insulin-producing pancreatic ß cells from stem cells in vitro would provide an unprecedented cell source for drug discovery and cell transplantation therapy in diabetes. However, insulin-producing cells previously generated from human pluripotent stem cells (hPSC) lack many functional characteristics of bona fide ß cells. Here, we report a scalable differentiation protocol that can generate hundreds of millions of glucose-responsive ß cells from hPSC in vitro. These stem-cell-derived ß cells (SC-ß) express markers found in mature ß cells, flux Ca(2+) in response to glucose, package insulin into secretory granules, and secrete quantities of insulin comparable to adult ß cells in response to multiple sequential glucose challenges in vitro. Furthermore, these cells secrete human insulin into the serum of mice shortly after transplantation in a glucose-regulated manner, and transplantation of these cells ameliorates hyperglycemia in diabetic mice.


Assuntos
Técnicas de Cultura de Células , Células Secretoras de Insulina/citologia , Animais , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Insulina/genética , Insulina/metabolismo , Ilhotas Pancreáticas , Camundongos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo
4.
Cell ; 153(4): 747-58, 2013 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-23623304

RESUMO

Replenishing insulin-producing pancreatic ß cell mass will benefit both type I and type II diabetics. In adults, pancreatic ß cells are generated primarily by self-duplication. We report on a mouse model of insulin resistance that induces dramatic pancreatic ß cell proliferation and ß cell mass expansion. Using this model, we identify a hormone, betatrophin, that is primarily expressed in liver and fat. Expression of betatrophin correlates with ß cell proliferation in other mouse models of insulin resistance and during gestation. Transient expression of betatrophin in mouse liver significantly and specifically promotes pancreatic ß cell proliferation, expands ß cell mass, and improves glucose tolerance. Thus, betatrophin treatment could augment or replace insulin injections by increasing the number of endogenous insulin-producing cells in diabetics.


Assuntos
Proliferação de Células , Células Secretoras de Insulina/metabolismo , Pâncreas/citologia , Hormônios Peptídicos/metabolismo , Tecido Adiposo Branco/metabolismo , Sequência de Aminoácidos , Proteína 8 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Animais , Feminino , Glucose/metabolismo , Humanos , Resistência à Insulina , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Pâncreas/metabolismo , Hormônios Peptídicos/química , Hormônios Peptídicos/genética , Peptídeos/administração & dosagem , Receptor de Insulina/antagonistas & inibidores , Alinhamento de Sequência
6.
Nature ; 569(7756): 368-373, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31068696

RESUMO

In vitro differentiation of human stem cells can produce pancreatic ß-cells; the loss of this insulin-secreting cell type underlies type 1 diabetes. Here, as a step towards understanding this differentiation process, we report the transcriptional profiling of more than 100,000 human cells undergoing in vitro ß-cell differentiation, and describe the cells that emerged. We resolve populations that correspond to ß-cells, α-like poly-hormonal cells, non-endocrine cells that resemble pancreatic exocrine cells and a previously unreported population that resembles enterochromaffin cells. We show that endocrine cells maintain their identity in culture in the absence of exogenous growth factors, and that changes in gene expression associated with in vivo ß-cell maturation are recapitulated in vitro. We implement a scalable re-aggregation technique to deplete non-endocrine cells and identify CD49a (also known as ITGA1) as a surface marker of the ß-cell population, which allows magnetic sorting to a purity of 80%. Finally, we use a high-resolution sequencing time course to characterize gene-expression dynamics during the induction of human pancreatic endocrine cells, from which we develop a lineage model of in vitro ß-cell differentiation. This study provides a perspective on human stem-cell differentiation, and will guide future endeavours that focus on the differentiation of pancreatic islet cells, and their applications in regenerative medicine.


Assuntos
Diferenciação Celular , Células Secretoras de Insulina/citologia , Células-Tronco/citologia , Animais , Biomarcadores/metabolismo , Linhagem da Célula , Separação Celular , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/classificação , Células Secretoras de Insulina/metabolismo , Integrina alfa1/metabolismo , Masculino , Camundongos , RNA-Seq , Análise de Célula Única , Células-Tronco/metabolismo
8.
Nature ; 560(7720): E34, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29925956

RESUMO

Change history: In this Insight Review, '1989' has been changed to '1998' in the sentence "This deep understanding of pancreatic development was put to the service of regenerative medicine in 1998, when human embryonic stem cells (hES cells) were successfully cultured and opened the door to developing methods of deriving pancreatic islets from hES cells66.". This error has been corrected online.

9.
Nature ; 557(7705): 351-358, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29769672

RESUMO

The pancreas is made from two distinct components: the exocrine pancreas, a reservoir of digestive enzymes, and the endocrine islets, the source of the vital metabolic hormone insulin. Human islets possess limited regenerative ability; loss of islet ß-cells in diseases such as type 1 diabetes requires therapeutic intervention. The leading strategy for restoration of ß-cell mass is through the generation and transplantation of new ß-cells derived from human pluripotent stem cells. Other approaches include stimulating endogenous ß-cell proliferation, reprogramming non-ß-cells to ß-like cells, and harvesting islets from genetically engineered animals. Together these approaches form a rich pipeline of therapeutic development for pancreatic regeneration.


Assuntos
Pâncreas/fisiologia , Regeneração/fisiologia , Medicina Regenerativa/métodos , Células-Tronco Adultas/citologia , Células-Tronco Adultas/transplante , Animais , Proliferação de Células , Reprogramação Celular , Humanos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiologia , Pâncreas/citologia , Pâncreas/crescimento & desenvolvimento , Pâncreas/patologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/transplante
10.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34504013

RESUMO

Islet transplantation for type 1 diabetes treatment has been limited by the need for lifelong immunosuppression regimens. This challenge has prompted the development of macroencapsulation devices (MEDs) to immunoprotect the transplanted islets. While promising, conventional MEDs are faced with insufficient transport of oxygen, glucose, and insulin because of the reliance on passive diffusion. Hence, these devices are constrained to two-dimensional, wafer-like geometries with limited loading capacity to maintain cells within a distance of passive diffusion. We hypothesized that convective nutrient transport could extend the loading capacity while also promoting cell viability, rapid glucose equilibration, and the physiological levels of insulin secretion. Here, we showed that convective transport improves nutrient delivery throughout the device and affords a three-dimensional capsule geometry that encapsulates 9.7-fold-more cells than conventional MEDs. Transplantation of a convection-enhanced MED (ceMED) containing insulin-secreting ß cells into immunocompetent, hyperglycemic rats demonstrated a rapid, vascular-independent, and glucose-stimulated insulin response, resulting in early amelioration of hyperglycemia, improved glucose tolerance, and reduced fibrosis. Finally, to address potential translational barriers, we outlined future steps necessary to optimize the ceMED design for long-term efficacy and clinical utility.


Assuntos
Encapsulamento de Células/métodos , Sistemas de Liberação de Medicamentos/métodos , Células Secretoras de Insulina/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Convecção , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Sistemas de Liberação de Medicamentos/instrumentação , Insulina/metabolismo , Secreção de Insulina/efeitos dos fármacos , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Masculino , Ratos
12.
Development ; 146(1)2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30602532

RESUMO

Blastema formation, a hallmark of limb regeneration, requires proliferation and migration of progenitors to the amputation plane. Although blastema formation has been well described, the transcriptional programs that drive blastemal progenitors remain unknown. We transcriptionally profiled dividing and non-dividing cells in regenerating stump tissues, as well as the wound epidermis, during early axolotl limb regeneration. Our analysis revealed unique transcriptional signatures of early dividing cells and, unexpectedly, repression of several core developmental signaling pathways in early regenerating stump tissues. We further identify an immunomodulatory role for blastemal progenitors through interleukin 8 (IL-8), a highly expressed cytokine in subpopulations of early blastemal progenitors. Ectopic il-8 expression in non-regenerating limbs induced myeloid cell recruitment, while IL-8 knockdown resulted in defective myeloid cell retention during late wound healing, delaying regeneration. Furthermore, the il-8 receptor cxcr-1/2 was expressed in myeloid cells, and inhibition of CXCR-1/2 signaling during early stages of limb regeneration prevented regeneration. Altogether, our findings suggest that blastemal progenitors are active early mediators of immune support, and identify CXCR-1/2 signaling as an important immunomodulatory pathway during the initiation of regeneration.


Assuntos
Blastoderma/imunologia , Diferenciação Celular/imunologia , Membro Posterior/fisiologia , Transdução de Sinais/imunologia , Células-Tronco/imunologia , Ambystoma mexicanum , Proteínas de Anfíbios/imunologia , Animais , Blastoderma/citologia , Interleucina-8/imunologia , Receptores de Interleucina-8A/imunologia , Receptores de Interleucina-8B/imunologia , Células-Tronco/citologia
13.
Proc Natl Acad Sci U S A ; 116(21): 10441-10446, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31040209

RESUMO

Polymorphic HLAs form the primary immune barrier to cell therapy. In addition, innate immune surveillance impacts cell engraftment, yet a strategy to control both, adaptive and innate immunity, is lacking. Here we employed multiplex genome editing to specifically ablate the expression of the highly polymorphic HLA-A/-B/-C and HLA class II in human pluripotent stem cells. Furthermore, to prevent innate immune rejection and further suppress adaptive immune responses, we expressed the immunomodulatory factors PD-L1, HLA-G, and the macrophage "don't-eat me" signal CD47 from the AAVS1 safe harbor locus. Utilizing in vitro and in vivo immunoassays, we found that T cell responses were blunted. Moreover, NK cell killing and macrophage engulfment of our engineered cells were minimal. Our results describe an approach that effectively targets adaptive as well as innate immune responses and may therefore enable cell therapy on a broader scale.


Assuntos
Engenharia Genética/métodos , Células-Tronco Pluripotentes/imunologia , Sistemas CRISPR-Cas , Linhagem Celular , Técnicas de Inativação de Genes , Genes MHC Classe I , Genes MHC da Classe II , Humanos
14.
Circulation ; 141(4): 285-300, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31707831

RESUMO

BACKGROUND: Current differentiation protocols to produce cardiomyocytes from human induced pluripotent stem cells (iPSCs) are capable of generating highly pure cardiomyocyte populations as determined by expression of cardiac troponin T. However, these cardiomyocytes remain immature, more closely resembling the fetal state, with a lower maximum contractile force, slower upstroke velocity, and immature mitochondrial function compared with adult cardiomyocytes. Immaturity of iPSC-derived cardiomyocytes may be a significant barrier to clinical translation of cardiomyocyte cell therapies for heart disease. During development, cardiomyocytes undergo a shift from a proliferative state in the fetus to a more mature but quiescent state after birth. The mechanistic target of rapamycin (mTOR)-signaling pathway plays a key role in nutrient sensing and growth. We hypothesized that transient inhibition of the mTOR-signaling pathway could lead cardiomyocytes to a quiescent state and enhance cardiomyocyte maturation. METHODS: Cardiomyocytes were differentiated from 3 human iPSC lines using small molecules to modulate the Wnt pathway. Torin1 (0 to 200 nmol/L) was used to inhibit the mTOR pathway at various time points. We quantified contractile, metabolic, and electrophysiological properties of matured iPSC-derived cardiomyocytes. We utilized the small molecule inhibitor, pifithrin-α, to inhibit p53 signaling, and nutlin-3a, a small molecule inhibitor of MDM2 (mouse double minute 2 homolog) to upregulate and increase activation of p53. RESULTS: Torin1 (200 nmol/L) increased the percentage of quiescent cells (G0 phase) from 24% to 48% compared with vehicle control (P<0.05). Torin1 significantly increased expression of selected sarcomere proteins (including TNNI3 [troponin I, cardiac muscle]) and ion channels (including Kir2.1) in a dose-dependent manner when Torin1 was initiated after onset of cardiomyocyte beating. Torin1-treated cells had an increased relative maximum force of contraction, increased maximum oxygen consumption rate, decreased peak rise time, and increased downstroke velocity. Torin1 treatment increased protein expression of p53, and these effects were inhibited by pifithrin-α. In contrast, nutlin-3a independently upregulated p53, led to an increase in TNNI3 expression and worked synergistically with Torin1 to further increase expression of both p53 and TNNI3. CONCLUSIONS: Transient treatment of human iPSC-derived cardiomyocytes with Torin1 shifts cells to a quiescent state and enhances cardiomyocyte maturity.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/metabolismo , Naftiridinas/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Benzotiazóis/farmacologia , Linhagem Celular , Humanos , Imidazóis/farmacologia , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/citologia , Piperazinas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Tolueno/análogos & derivados , Tolueno/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores
15.
Proc Natl Acad Sci U S A ; 115(15): 3924-3929, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29599125

RESUMO

Type 1 diabetes is characterized by autoimmune destruction of ß cells located in pancreatic islets. However, tractable in vivo models of human pancreatic ß cells have been limited. Here, we generated xenogeneic human pancreatic ß-like cells in the mouse pancreas by orthotopic transplantation of stem cell-derived ß (SC-ß) cells into the pancreas of neonatal mice. The engrafted ß-like cells expressed ß cell transcription factors and markers associated with functional maturity. Engrafted human cells recruited mouse endothelial cells, suggesting functional integration. Human insulin was detected in the blood circulation of transplanted mice for months after transplantation and increased upon glucose stimulation. In addition to ß-like cells, human cells expressing markers for other endocrine pancreas cell types, acinar cells, and pancreatic ductal cells were identified in the pancreata of transplanted mice, indicating that this approach allows studying other human pancreatic cell types in the mouse pancreas. Our results demonstrate that orthotopic transplantation of human SC-ß cells into neonatal mice is an experimental platform that allows the generation of mice with human pancreatic ß-like cells in the endogenous niche.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Células Secretoras de Insulina/citologia , Pâncreas/citologia , Células-Tronco Pluripotentes/transplante , Células Acinares/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatologia , Glucose/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/transplante , Camundongos , Pâncreas/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo
16.
Am J Transplant ; 19(7): 1930-1940, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30748094

RESUMO

Pancreatic ß-cell replacement by islet transplantation for the treatment of type 1 diabetes (T1D) is currently limited by donor tissue scarcity and the requirement for lifelong immunosuppression. The advent of in vitro differentiation protocols for generating functional ß-like cells from human pluripotent stem cells, also referred to as SC-ß cells, could eliminate these obstacles. To avoid the need for immunosuppression, alginate-microencapsulation is widely investigated as a safe path to ß-cell replacement. Nonetheless, inflammatory foreign body responses leading to pericapsular fibrotic overgrowth often causes microencapsulated islet-cell death and graft failure. Here we used a novel approach to evade the pericapsular fibrotic response to alginate-microencapsulated SC-ß cells; an immunomodulatory chemokine, CXCL12, was incorporated into clinical grade sodium alginate to microencapsulate SC-ß cells. CXCL12 enhanced glucose-stimulated insulin secretion activity of SC-ß cells and induced expression of genes associated with ß-cell function in vitro. SC-ß cells co-encapsulated with CXCL12 showed enhanced insulin secretion in diabetic mice and accelerated the normalization of hyperglycemia. Additionally, SC-ß cells co-encapsulated with CXCL12 evaded the pericapsular fibrotic response, resulting in long-term functional competence and glycemic correction (>150 days) without systemic immunosuppression in immunocompetent C57BL/6 mice. These findings lay the groundwork for further preclinical translation of this approach into large animal models of T1D.


Assuntos
Alginatos/química , Quimiocina CXCL12/metabolismo , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Sobrevivência de Enxerto , Células Secretoras de Insulina/citologia , Transplante das Ilhotas Pancreáticas/métodos , Células-Tronco/citologia , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Feminino , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/metabolismo
17.
Nature ; 491(7426): 765-8, 2012 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-23041930

RESUMO

One goal of regenerative medicine, to use stem cells to replace cells lost by injury or disease, depends on producing an excess of the relevant cell for study or transplantation. To this end, the stepwise differentiation of stem cells into specialized derivatives has been successful for some cell types, but a major problem remains the inefficient conversion of cells from one stage of differentiation to the next. If specialized cells are to be produced in large numbers it will be necessary to expand progenitor cells, without differentiation, at some steps of the process. Using the pancreatic lineage as a model for embryonic-stem-cell differentiation, we demonstrate that this is a solvable problem. Co-culture with organ-matched mesenchyme permits proliferation and self-renewal of progenitors, without differentiation, and enables an expansion of more than a million-fold for human endodermal cells with full retention of their developmental potential. This effect is specific both to the mesenchymal cell and to the progenitor being amplified. Progenitors that have been serially expanded on mesenchyme give rise to glucose-sensing, insulin-secreting cells when transplanted in vivo. Theoretically, the identification of stage-specific renewal signals can be incorporated into any scheme for the efficient production of large numbers of differentiated cells from stem cells and may therefore have wide application in regenerative biology.


Assuntos
Técnicas de Cocultura/métodos , Células-Tronco Embrionárias/citologia , Endoderma/citologia , Mesoderma/citologia , Pâncreas/citologia , Animais , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Glucose/farmacologia , Humanos , Insulina/metabolismo , Masculino , Camundongos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo
18.
Proc Natl Acad Sci U S A ; 112(50): 15498-503, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26621734

RESUMO

Type 2 diabetes is characterized by a reduction in insulin function and an increase in glucagon activity that together result in hyperglycemia. Glucagon receptor antagonists have been developed as drugs for diabetes; however, they often increase glucagon plasma levels and induce the proliferation of glucagon-secreting α-cells. We find that the secreted protein Angiopoietin-like 4 (Angptl4) is up-regulated via Pparγ activation in white adipose tissue and plasma following an acute treatment with a glucagon receptor antagonist. Induction of adipose angptl4 and Angptl4 supplementation promote α-cell proliferation specifically. Finally, glucagon receptor antagonist improves glycemia in diet-induced obese angptl4 knockout mice without increasing glucagon levels or α-cell proliferation, underscoring the importance of this protein. Overall, we demonstrate that triglyceride metabolism in adipose tissue regulates α-cells in the endocrine pancreas.


Assuntos
Tecido Adiposo/metabolismo , Angiopoietinas/metabolismo , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/metabolismo , Receptores de Glucagon/antagonistas & inibidores , Triglicerídeos/metabolismo , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/sangue , Animais , Restrição Calórica , Proliferação de Células , Regulação da Expressão Gênica , Glucagon/sangue , Camundongos Endogâmicos C57BL , Camundongos SCID , PPAR gama/agonistas , PPAR gama/metabolismo , Receptores de Glucagon/metabolismo
19.
Genes Dev ; 24(3): 312-26, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20123909

RESUMO

In embryonic stem (ES) cells, a well-characterized transcriptional network promotes pluripotency and represses gene expression required for differentiation. In comparison, the transcriptional networks that promote differentiation of ES cells and the blastocyst inner cell mass are poorly understood. Here, we show that Sox17 is a transcriptional regulator of differentiation in these pluripotent cells. ES cells deficient in Sox17 fail to differentiate into extraembryonic cell types and maintain expression of pluripotency-associated transcription factors, including Oct4, Nanog, and Sox2. In contrast, forced expression of Sox17 down-regulates ES cell-associated gene expression and directly activates genes functioning in differentiation toward an extraembryonic endoderm cell fate. We show these effects of Sox17 on ES cell gene expression are mediated at least in part through a competition between Sox17 and Nanog for common DNA-binding sites. By elaborating the function of Sox17, our results provide insight into how the transcriptional network promoting ES cell self-renewal is interrupted, allowing cellular differentiation.


Assuntos
Diferenciação Celular , Linhagem da Célula/genética , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas HMGB/metabolismo , Fatores de Transcrição SOXF/metabolismo , Animais , Células-Tronco Embrionárias/metabolismo , Redes Reguladoras de Genes , Proteínas HMGB/genética , Camundongos , Fatores de Transcrição SOXF/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
20.
Proc Natl Acad Sci U S A ; 111(8): 3038-43, 2014 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-24516164

RESUMO

Human pluripotent stem cells (hPSCs) have the potential to generate any human cell type, and one widely recognized goal is to make pancreatic ß cells. To this end, comparisons between differentiated cell types produced in vitro and their in vivo counterparts are essential to validate hPSC-derived cells. Genome-wide transcriptional analysis of sorted insulin-expressing (INS(+)) cells derived from three independent hPSC lines, human fetal pancreata, and adult human islets points to two major conclusions: (i) Different hPSC lines produce highly similar INS(+) cells and (ii) hPSC-derived INS(+) (hPSC-INS(+)) cells more closely resemble human fetal ß cells than adult ß cells. This study provides a direct comparison of transcriptional programs between pure hPSC-INS(+) cells and true ß cells and provides a catalog of genes whose manipulation may convert hPSC-INS(+) cells into functional ß cells.


Assuntos
Diferenciação Celular/fisiologia , Células Secretoras de Insulina/citologia , Pâncreas/citologia , Células-Tronco Pluripotentes/citologia , Adulto , Diferenciação Celular/genética , Feto/citologia , Feto/metabolismo , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Células Secretoras de Insulina/metabolismo , Análise em Microsséries , Células-Tronco Pluripotentes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA