Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 640
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Development ; 149(20)2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-35815787

RESUMO

Embryogenesis is guided by a limited set of signaling pathways dynamically expressed in different places. How a context-dependent signaling response is generated has been a central question of developmental biology, which can now be addressed with in vitro models of human embryos that are derived from embryonic stem cells (hESCs). Our previous work demonstrated that during early stages of hESC differentiation, cells chronicle signaling hierarchy. Only cells that have been exposed (primed) by WNT signaling can respond to subsequent activin exposure and differentiate to mesendodermal (ME) fates. Here, we show that WNT priming does not alter SMAD2 binding nor its chromatin opening but, instead, acts by inducing the expression of the SMAD2 co-factor EOMES. Expression of EOMES is sufficient to replace WNT upstream of activin-mediated ME differentiation, thus unveiling the mechanistic basis for priming and cellular memory in early development.


Assuntos
Células-Tronco Embrionárias Humanas , Ativinas/metabolismo , Ativinas/farmacologia , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias , Humanos , Via de Sinalização Wnt
2.
Nucleic Acids Res ; 50(18): 10526-10543, 2022 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-36134711

RESUMO

Transforming growth factor ß (TGF-ß) superfamily proteins are potent regulators of cellular development and differentiation. Nodal/Activin/TGF-ß and BMP ligands are both present in the intra- and extracellular milieu during early development, and cross-talk between these two branches of developmental signaling is currently the subject of intense research focus. Here, we show that the Nodal induced lncRNA-Smad7 regulates cell fate determination via repression of BMP signaling in mouse embryonic stem cells (mESCs). Depletion of lncRNA-Smad7 dramatically impairs cardiomyocyte differentiation in mESCs. Moreover, lncRNA-Smad7 represses Bmp2 expression through binding with the Bmp2 promoter region via (CA)12-repeats that forms an R-loop. Importantly, Bmp2 knockdown rescues defects in cardiomyocyte differentiation induced by lncRNA-Smad7 knockdown. Hence, lncRNA-Smad7 antagonizes BMP signaling in mESCs, and similarly regulates cell fate determination between osteocyte and myocyte formation in C2C12 mouse myoblasts. Moreover, lncRNA-Smad7 associates with hnRNPK in mESCs and hnRNPK binds at the Bmp2 promoter, potentially contributing to Bmp2 expression repression. The antagonistic effects between Nodal/TGF-ß and BMP signaling via lncRNA-Smad7 described in this work provides a framework for understanding cell fate determination in early development.


Assuntos
RNA Longo não Codificante , Proteína Smad7/metabolismo , Ativinas/metabolismo , Ativinas/farmacologia , Animais , Diferenciação Celular , Ligantes , Camundongos , RNA Longo não Codificante/metabolismo , Proteína Smad7/genética , Proteína Smad7/farmacologia , Fator de Crescimento Transformador beta/metabolismo
3.
Proc Natl Acad Sci U S A ; 117(22): 12269-12280, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32409602

RESUMO

In multiple sclerosis (MS), Th17 cells are critical drivers of autoimmune central nervous system (CNS) inflammation and demyelination. Th17 cells exhibit functional heterogeneity fostering both pathogenic and nonpathogenic, tissue-protective functions. Still, the factors that control Th17 pathogenicity remain incompletely defined. Here, using experimental autoimmune encephalomyelitis, an established mouse MS model, we report that therapeutic administration of activin-A ameliorates disease severity and alleviates CNS immunopathology and demyelination, associated with decreased activation of Th17 cells. In fact, activin-A signaling through activin-like kinase-4 receptor represses pathogenic transcriptional programs in Th17-polarized cells, while it enhances antiinflammatory gene modules. Whole-genome profiling and in vivo functional studies revealed that activation of the ATP-depleting CD39 and CD73 ectonucleotidases is essential for activin-A-induced suppression of the pathogenic signature and the encephalitogenic functions of Th17 cells. Mechanistically, the aryl hydrocarbon receptor, along with STAT3 and c-Maf, are recruited to promoter elements on Entpd1 and Nt5e (encoding CD39 and CD73, respectively) and other antiinflammatory genes, and control their expression in Th17 cells in response to activin-A. Notably, we show that activin-A negatively regulates the metabolic sensor, hypoxia-inducible factor-1α, and key inflammatory proteins linked to pathogenic Th17 cell states. Of translational relevance, we demonstrate that activin-A is induced in the CNS of individuals with MS and restrains human Th17 cell responses. These findings uncover activin-A as a critical controller of Th17 cell pathogenicity that can be targeted for the suppression of autoimmune CNS inflammation.


Assuntos
5'-Nucleotidase/metabolismo , Ativinas/farmacologia , Antígenos CD/metabolismo , Apirase/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/prevenção & controle , Esclerose Múltipla/imunologia , Células Th17/imunologia , Animais , Diferenciação Celular , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Proteínas Ligadas por GPI/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Células Th17/metabolismo
4.
Altern Lab Anim ; 51(6): 363-375, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37831588

RESUMO

Human umbilical cord-derived mesenchymal stem cells (hucMSCs) can differentiate into multiple cell lineages, but few methods have been developed to generate kidney lineage cells. Due to their human origin, pluripotent nature and immunomodulatory properties, these stem cells are attractive candidates for clinical applications such as the repair or regeneration of damaged organs. This study evaluated the renal differentiation potential of hucMSCs, when exposed for 10 days to optimised concentrations of retinoic acid, activin-A and bone morphogenetic protein-7 (BMP-7) in various combinations, with and without the priming of the cells with a Wnt signalling pathway activator (CHIR99021). The hucMSCs were isolated and characterised according to surface marker expression (CD73, CD90, CD44, CD146 and CD8) and tri-lineage differentiation potential. The expression of key marker genes (OSR1, TBXT, HOXA13, SIX2, PAX2, KRT18 and ZO1) was examined by qRT-PCR. Specific marker protein expression (E-cadherin, cytokeratin-8 and cytokeratin-19) was analysed by immunocytochemistry. CHIR99021-primed cells treated with the retinoic acid, activin-A and BMP-7 cocktail showed epithelial cell-like differentiation - i.e. distinct phenotypic changes, as well as upregulated gene and protein expression, were observed that were consistent with an epithelial cell phenotype. Thus, our results showed that hucMSCs can efficiently differentiate into renal epithelial-like cells. This work may help in the development of focused therapeutic strategies, in which lineage-defined human stem cells can be used for renal regeneration.


Assuntos
Proteína Morfogenética Óssea 7 , Células-Tronco Mesenquimais , Humanos , Proteína Morfogenética Óssea 7/metabolismo , Cordão Umbilical , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Células Epiteliais , Tretinoína/metabolismo , Ativinas/farmacologia , Ativinas/metabolismo , Células Cultivadas
5.
Genesis ; 60(1-2): e23466, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35104045

RESUMO

Allocation of cells to an endodermal fate in the gastrulating embryo is driven by Nodal signaling and consequent activation of TGFß pathway. In vitro methodologies striving to recapitulate the process of endoderm differentiation, however, use TGFß family member Activin in place of Nodal. This is despite Activin not known to have an in vivo role in endoderm differentiation. In this study, five epiblast stem cell lines were subjected to directed differentiation using both Activin A and Nodal to induce endodermal fate. A reporter line harboring endoderm markers FoxA2 and Sox17 was further analyzed for TGFß pathway activation and WNT response. We demonstrated that Activin A-treated cells remain more primitive streak-like when compared to Nodal-treated cells that have a molecular profile suggestive of more advanced differentiation. Activin A elicited a robust TGFß/SMAD activity, enhanced WNT signaling activity and promoted the generation of DE precursors. Nodal treatment resulted in lower TGFß/SMAD activity, and a weaker, sustained WNT response, and ultimately failed to upregulate endoderm markers. This is despite signaling response resembling more closely the activity seen in vivo. These findings emphasize the importance of understanding the downstream activities of Activin A and Nodal signaling in directing in vitro endoderm differentiation of primed-state epiblast stem cells.


Assuntos
Endoderma , Proteína Nodal , Ativinas/metabolismo , Ativinas/farmacologia , Diferenciação Celular/fisiologia , Endoderma/metabolismo , Camadas Germinativas , Proteína Nodal/genética , Proteína Nodal/metabolismo , Células-Tronco/metabolismo , Fator de Crescimento Transformador beta
6.
Hippocampus ; 32(5): 401-410, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35301773

RESUMO

Mossy cells (MCs) in the hilus of the dentate gyrus (DG) receive increasing attention as a major player controlling information processing in the DG network. Furthermore, disturbed MC activity has been implicated in widespread neuropsychiatric disorders such as epilepsy and major depression. Using whole-cell patch-clamp recordings from MCs in acute hippocampal slices from wild type and transgenic mice, we demonstrate that activin, a member of the transforming growth factor-ß (TGF-ß) family, has a strong neuromodulatory effect on MC activity. Disruption of activin receptor signaling reduced MC firing, dampened their excitatory input and augmented their inhibitory input. By contrast, acute application of recombinant activin A strongly increased MC activity and promoted excitatory synaptic drive. Notably, similar changes of MC activity have been observed in a rodent model of depression and after antidepressant drug therapy, respectively. Given that a rise in activin signaling particularly in the DG has been proposed as a mechanism of antidepressant action, our data suggest that the effect of activin on MC excitability might make a considerable contribution in this regard.


Assuntos
Hipocampo , Fibras Musgosas Hipocampais , Ativinas/farmacologia , Animais , Giro Denteado/fisiologia , Hipocampo/fisiologia , Camundongos , Camundongos Transgênicos , Fibras Musgosas Hipocampais/fisiologia
7.
FASEB J ; 35(2): e21220, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33230889

RESUMO

Activin A promotes human trophoblast invasion during the first trimester of pregnancy and is associated with preeclampsia and pregnancy-induced hypertension (PE/PIH) in naturally conceived pregnancies. However, whether integrin ß1 mediates activin A-increased trophoblast invasion remains unknown and the evidence is limited regarding the predictive value of activin A for PE/PIH in women receiving in vitro fertilization (IVF) treatment. Here, we studied the role and underlying molecular mechanisms of integrin ß1 in activin A-promoted invasion in immortalized (HTR8/SVneo) and primary human extravillous trophoblast (EVT) cells. A nest case-control study was designed to investigate the predictive/diagnostic value of activin A in IVF pregnancies. Results showed that integrin ß1 expression increased after activin A treatment and knockdown of integrin ß1 significantly decreased both basal and activin A-increased HTR8/SVneo cell invasion. SB431542 (TGF-ß type I receptors inhibitor) abolished activin A-induced SMAD2/SMAD3 phosphorylation and integrin ß1 overexpression. Activin A-upregulated integrin ß1 expression was attenuated after the depletion of ALK4 or SMAD4 in both HTR8/SVneo and primary EVT cells. Furthermore, we found similar first-trimester activin A levels in IVF patients with or without subsequent PE/PIH. These results reveal that integrin ß1 mediates activin A-promoted trophoblast invasion through ALK4-activated SMAD2/3-SMAD4 pathway, and the predictive/diagnostic value of first-trimester maternal serum activin A for hypertensive disorders of pregnancy might be different in IVF population.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Ativinas/farmacologia , Movimento Celular , Integrina beta1/metabolismo , Trofoblastos/metabolismo , Adulto , Benzamidas/farmacologia , Linhagem Celular , Células Cultivadas , Dioxóis/farmacologia , Feminino , Humanos , Integrina beta1/genética , Gravidez , Proteínas Smad/metabolismo , Trofoblastos/efeitos dos fármacos , Trofoblastos/fisiologia , Regulação para Cima
8.
Nature ; 529(7586): 403-407, 2016 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-26751055

RESUMO

Nanog, a core pluripotency factor in the inner cell mass of blastocysts, is also expressed in unipotent primordial germ cells (PGCs) in mice, where its precise role is yet unclear. We investigated this in an in vitro model, in which naive pluripotent embryonic stem (ES) cells cultured in basic fibroblast growth factor (bFGF) and activin A develop as epiblast-like cells (EpiLCs) and gain competence for a PGC-like fate. Consequently, bone morphogenetic protein 4 (BMP4), or ectopic expression of key germline transcription factors Prdm1, Prdm14 and Tfap2c, directly induce PGC-like cells (PGCLCs) in EpiLCs, but not in ES cells. Here we report an unexpected discovery that Nanog alone can induce PGCLCs in EpiLCs, independently of BMP4. We propose that after the dissolution of the naive ES-cell pluripotency network during establishment of EpiLCs, the epigenome is reset for cell fate determination. Indeed, we found genome-wide changes in NANOG-binding patterns between ES cells and EpiLCs, indicating epigenetic resetting of regulatory elements. Accordingly, we show that NANOG can bind and activate enhancers of Prdm1 and Prdm14 in EpiLCs in vitro; BLIMP1 (encoded by Prdm1) then directly induces Tfap2c. Furthermore, while SOX2 and NANOG promote the pluripotent state in ES cells, they show contrasting roles in EpiLCs, as Sox2 specifically represses PGCLC induction by Nanog. This study demonstrates a broadly applicable mechanistic principle for how cells acquire competence for cell fate determination, resulting in the context-dependent roles of key transcription factors during development.


Assuntos
Elementos Facilitadores Genéticos/genética , Células Germinativas/citologia , Células Germinativas/metabolismo , Camadas Germinativas/citologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Fatores de Transcrição/genética , Ativinas/farmacologia , Animais , Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular/genética , Cromatina/genética , Cromatina/metabolismo , Proteínas de Ligação a DNA , Epigênese Genética , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Genoma/genética , Camadas Germinativas/efeitos dos fármacos , Camadas Germinativas/metabolismo , Proteínas de Homeodomínio/antagonistas & inibidores , Masculino , Camundongos , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Proteína Homeobox Nanog , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Fator 1 de Ligação ao Domínio I Regulador Positivo , Ligação Proteica , Proteínas de Ligação a RNA , Fatores de Transcrição SOXB1/metabolismo , Fator de Transcrição AP-2/genética , Fator de Transcrição AP-2/metabolismo , Fatores de Transcrição/metabolismo
9.
Endocr J ; 69(10): 1193-1200, 2022 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-35584931

RESUMO

Recently, we reported that gonadotropin-releasing hormone (GnRH) stimulates annexin A1 (Anxa1) and A5 (Anxa5) mRNA expression through the GnRH-receptor-mitogen-activated protein kinase cascade in LßT2 cells. As LßT2 cells respond to activin, we examined the effect of activin A on Anxa1 and a5 expression in LßT2 cells. Activin A (0.4 and 4 ng/mL) treatment decreased Anxa5 mRNA levels in a dose-dependent manner, but did not affect Anxa1 mRNA levels at concentrations up to 40 ng/mL. After activin A treatment (4 ng/mL), Anxa5 mRNA levels significantly decreased at 6 h, gradually declined until 24 h, and remained low until 48 h, whereas Anxa1 mRNA levels did not significantly change following treatment. Pretreatment with activin A for 24 h increased GnRH agonist (GnRHa)-induced Anxa1 increase by approximately 7-fold compared with GnRHa stimulation alone, but Anxa5 was not affected. As previously reported, these activin A treatments increased gonadotropin ß subunit and GnRH receptor mRNA levels and slightly decreased common α-glycoprotein subunit mRNA levels. Furthermore, we examined the effect of activin A on Nr4a3, which is repressed by ANXA5 and which reduces Fshb expression, and found that activin A augmented Nr4a3 expression and slightly decreased the GnRHa-induced increase in Nr4a3. These results suggest that in gonadotrope cells, the mechanism regulating Anxa1 and a5 expression is differentially coupled with activin A signal transduction. Activin A suppresses Anxa5 expression under increased Nr4a3 expression, whereas activin A and GnRH synergistically stimulate Anxa1 expression. These GnRH-inducible annexins may have different specific functions in gonadotropes.


Assuntos
Ativinas , Hormônio Liberador de Gonadotropina , Hormônio Liberador de Gonadotropina/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Anexina A5/genética , Anexina A5/metabolismo , RNA Mensageiro/metabolismo , Ativinas/farmacologia , Ativinas/metabolismo
10.
Proc Natl Acad Sci U S A ; 116(11): 4989-4998, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30819898

RESUMO

WNT/ß-catenin signaling is crucial to all stages of life. It controls early morphogenetic events in embryos, maintains stem cell niches in adults, and is dysregulated in many types of cancer. Despite its ubiquity, little is known about the dynamics of signal transduction or whether it varies across contexts. Here we probe the dynamics of signaling by monitoring nuclear accumulation of ß-catenin, the primary transducer of canonical WNT signals, using quantitative live cell imaging. We show that ß-catenin signaling responds adaptively to constant WNT signaling in pluripotent stem cells, and that these dynamics become sustained on differentiation. Varying dynamics were also observed in the response to WNT in commonly used mammalian cell lines. Signal attenuation in pluripotent cells is observed even at saturating doses, where ligand stability does not affect the dynamics. TGFß superfamily ligands Activin and BMP, which coordinate with WNT signaling to pattern the gastrula, increase the ß-catenin response in a manner independent of their ability to induce new WNT ligand production. Our results reveal how variables external to the pathway, including differentiation status and cross-talk with other pathways, dramatically alter WNT/ß-catenin dynamics.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Via de Sinalização Wnt , Ativinas/farmacologia , Adaptação Biológica/efeitos dos fármacos , Proteína Morfogenética Óssea 4/farmacologia , Sistemas CRISPR-Cas/genética , Diferenciação Celular/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Ligantes , Células-Tronco Pluripotentes/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo
11.
BMC Immunol ; 22(1): 17, 2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33622252

RESUMO

BACKGROUND: Kawasaki disease is an autoimmune disease characterized by systemic vasculitis of unknown aetiology and most commonly occurs in children under 5 years old. Previous studies have found that the over-activation of lymphocytes is an important mechanism of Kawasaki disease. Activin A, also known as immunosuppressive factor P, is a multifunctional growth and transforming factor. However, whether activin A is involved in the regulation of peripheral lymphocytes activity in Kawasaki disease is unclear. Thus, we aimed to investigate the effect of activin A on the activity of peripheral lymphocytes in acute-phase Kawasaki disease. METHODS: Seven patients with Kawasaki disease and seven healthy controls were studied. Peripheral blood lymphocytes were isolated by Ficoll density gradient centrifugation. The activation of CD4+ and CD8+ T cells and CD19+ B cells was investigated by flow cytometry. The expression of activin type IIA receptors was investigated by flow cytometry. RESULTS: Immune imbalance in CD4 and CD8 lymphocytes were detected in acute-phase Kawasaki disease. The expression of activin type IIA receptors on CD8+ T cells and CD19+ B cells was increased in acute-phase Kawasaki disease and decreased following treatment with activin A. Activin A suppressed the expression of CD25 and CD69 on CD8+ T cells and the expression of CD69 on CD19+ B cells. CONCLUSIONS: The expression of activin type IIA receptor was increased on CD8+ T cells and CD19+ B cells in Kawasaki disease. Activin A suppressed the expression of CD25, CD69 and activin type IIA receptors on peripheral CD8+ T lymphocyte. Activin A plays different roles in different lymphocyte subsets and suppresses peripheral CD8+ T lymphocyte activity in acute-phase Kawasaki disease.


Assuntos
Ativinas/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Síndrome de Linfonodos Mucocutâneos/imunologia , Receptores de Activinas Tipo II/metabolismo , Antígenos CD/metabolismo , Antígenos CD19/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Linfócitos T CD8-Positivos/imunologia , Pré-Escolar , Feminino , Humanos , Lactente , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Lectinas Tipo C/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Contagem de Linfócitos , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/imunologia , Masculino
12.
J Cell Physiol ; 235(5): 4865-4877, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31667867

RESUMO

Activins regulate bone formation by controlling osteoclasts and osteoblasts. We investigated Activin-A mechanism of action on human osteoblast mineralization, RNA and microRNA (miRNA) expression profile. A single 2-day treatment of Activin-A at Day 5 of osteoblast differentiation significantly reduced matrix mineralization. Activin A-treated osteoblasts responded with transient change in gene expression, in a 2-wave-fashion. The 38 genes differentially regulated during the first wave (within 8 hr after Activin A start) were involved in transcription regulation. In the second wave (1-2 days after Activin A start), 65 genes were differentially regulated and related to extracellular matrix. Differentially expressed genes in both waves were associated to transforming growth factor beta signaling. We identified which microRNAs modulating osteoblast differentiation were regulated by Activin-A. In summary, 2-day treatment with Activin-A in premineralization period of osteoblast cultures influenced miRNAs, gene transcription, and reduced matrix mineralization. Modulation of Activin A signaling might be useful to control bone quality for therapeutic purposes.


Assuntos
Ativinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Transformada , Matriz Extracelular/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Osteoblastos/metabolismo , Osteogênese/genética , Fosforilação , Transdução de Sinais , Vírus 40 dos Símios , Proteína Smad3/metabolismo , Fatores de Tempo , Transcriptoma
13.
J Cell Physiol ; 235(11): 8640-8652, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32324269

RESUMO

We recently reported that epiblast stem cells (EpiSCs)-like cells could be derived from preimplantation embryos (named as AFSCs). Here, we established AFSCs from pre-implantation embryos of multiple mouse strains and showed that unlike EpiSCs, the derivation efficiency of AFSCs was affected by the genetic background. We then used AFSCs lines to dissect the roles of Activin A (Act A) and basic fibroblast growth factor and reported that Act A alone was capable of maintaining self-renewal but not developmental potential in vivo. Finally, we established a novel experimental system, in which AFSCs were efficiently converted to multipotent progenitor stem cells using Act A and bone morphogenetic protein 4 (named as ABSCs). Importantly, these ABSCs contributed to neural mesodermal progenitors and lateral plate mesoderm in postimplantation chimeras. Taken together, our study established a robust experimental system for the generation of specific multipotent progenitor stem cells that was self-renewable and capable of contributing to embryonic and extra-embryonic tissues.


Assuntos
Ativinas/farmacologia , Camadas Germinativas/efeitos dos fármacos , Células-Tronco Multipotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Ativinas/metabolismo , Animais , Blastocisto/efeitos dos fármacos , Blastocisto/metabolismo , Proteína Morfogenética Óssea 4/efeitos dos fármacos , Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Desenvolvimento Embrionário/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Camadas Germinativas/crescimento & desenvolvimento , Camundongos , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais/efeitos dos fármacos
14.
Biochem Biophys Res Commun ; 526(1): 41-47, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32192771

RESUMO

Human breast tumors are not fully autonomous. They are dependent on nutrients and growth-promoting signals provided by the supporting stromal cells. Within the tumor microenvironment, one of the secreted macromolecules by tumor cells is activin A, where we show to downregulate CD36 in fibroblasts. Downregulation of CD36 in fibroblasts also increases the secretion of activin A by fibroblasts. We hypothesize that overexpression of CD36 in fibroblasts inhibits the formation of solid tumors in subtypes of breast cancer models. For the first time, we show that co-culturing organoid models of breast cancer cell lines of MDA-MB-231 (e.g., a triple-negative line) or MCF7 (e.g., a luminal-A line) with CD36+ fibroblasts inhibit the growth and normalizes basal and lateral polarities, respectively. In the long-term anchorage-independent growth assay, the rate of colony formation is also reduced for MDA-MB-231. These observations are consistent with the mechanism of tumor suppression involving the downregulation of pSMAD2/3 and YY1 expression levels. Our integrated analytical methods leverage and extend quantitative assays at cell- and colony-scales in both short- and long-term cultures using brightfield or immunofluorescent microscopy and robust image analysis. Conditioned media are profiled with the ELISA assay.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Antígenos CD36/metabolismo , Fibroblastos/metabolismo , Glândulas Mamárias Humanas/patologia , Ativinas/farmacologia , Linhagem Celular Tumoral , Polaridade Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Humanos , Fosforilação/efeitos dos fármacos , Proteínas Smad/metabolismo , Ensaio Tumoral de Célula-Tronco , Fator de Transcrição YY1/metabolismo
15.
Reproduction ; 160(1): 141-154, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32484162

RESUMO

Disrupted fetal germline development underpins testicular germ cell neoplasia, which is increasing worldwide. The complex signaling milieu during normal testis development includes TGFß superfamily ligands; this study tests the hypothesis that, activin A, a TGFß superfamily member, can influence gonocyte development. The human seminoma-derived cell line, TCam-2, a model of fetal gonocytes, was cultured with activin A (1.25-25 ng/mL) for 48 h, or with 5 ng/mL activin A for short- (6, 24, and 48 h) and long-term (13 days) exposures, and downstream targets measured by qRT-PCR. Transcripts that exhibited significant dose-dependent responses to activin A included the early germ cell markers KIT, NODAL, and CRIPTO (NODALl co-receptor and activin inhibitor) which all increased and the differentiation marker DNMT3L which decreased. After 48 h, KIT, NODAL, and CRIPTO levels were significantly higher, while the differentiation marker NANOS2 was significantly lower. Interestingly, activin A exposure also significantly reduced both transcript and protein levels of the PIWI/piRNA pathway component DNMT3L. Because TCam-2 cells produce the activin inhibitor CRIPTO, CRIPTO was reduced using siRNA prior to activin A exposure. This selectively increased KIT in response to activin A. Other ligands present in the fetal testis (BMP4, FGF9, TGFß1, and TGFß2) induced distinct effects on germline marker expression. This study showed that activin A can directly modulate germline markers in this human gonocyte-like cell, promoting a less-differentiated phenotype. Additional findings indicate evidence of signaling crosstalk between activin A and NODAL, leading to target-specific effects on gonocyte differentiation.


Assuntos
Ativinas/farmacologia , Diferenciação Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Células Germinativas/patologia , Proteína Nodal/metabolismo , Seminoma/patologia , Fator de Crescimento Transformador beta/farmacologia , Perfilação da Expressão Gênica , Células Germinativas/metabolismo , Humanos , Masculino , Proteína Nodal/genética , Seminoma/tratamento farmacológico , Seminoma/genética , Neoplasias Testiculares/tratamento farmacológico , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia
16.
Stem Cells ; 37(1): 150-162, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30358011

RESUMO

In a previous study, we have shown that Activin B is a potent chemoattractant for bone marrow-derived mesenchymal stromal cells (BMSCs). As such, the combination of Activin B and BMSCs significantly accelerated rat skin wound healing. In another study, we showed that RhoA activation plays a key role in Activin B-induced BMSC migration. However, the role of the immediate downstream effectors of RhoA in this process is unclear. Here, we demonstrated that mammalian homolog of Drosophila diaphanous-1 (mDia1), a downstream effector of RhoA, exerts a crucial function in Activin B-induced BMSC migration by promoting membrane ruffling, microtubule morphology, and adhesion signaling dynamics. Furthermore, we showed that Activin B does not change Rac1 activity but increases Cdc42 activity in BMSCs. Inactivation of Cdc42 inhibited Activin B-stimulated Golgi reorientation and the cell migration of BMSCs. Furthermore, knockdown of mDia1 affected Activin B-induced BMSC-mediated wound healing in vivo. In conclusion, this study demonstrated that the RhoA-mDia1 and Cdc42 pathways regulate Activin B-induced BMSC migration. This study may help to optimize clinical MSC-based transplantation strategies to promote skin wound healing. Stem Cells 2019;37:150-162.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Forminas/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Ativinas/farmacologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Forminas/genética , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Cicatrização , Proteína cdc42 de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo
17.
Exp Cell Res ; 374(1): 114-121, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30458178

RESUMO

Activin A, a multifunctional cytokine of transforming growth factor-ß (TGF-ß) superfamily, can be produced by the diverse immune cells. NK cells in peripheral blood are one of the major immune cells applied to cancer therapy in recent years. However, whether activin A can be produced by natural killer (NK) cells and be involved in regulation of peripheral blood NK cells activities of mouse are not well characterized. Here, we found that activin type IIA and IIB receptors and signaling molecules Smad2, 3 were expressed in peripheral blood NK cells of mouse by flow cytometry and RT-PCR. The cultured blood NK cells of mouse not only produced activin ßA chain protein by intracellular cytokine staining, but also secreted mature activin A protein by enzyme-linked immunosorbent assay (ELISA), and the production was promoted by IL-2. In addition, IL-2 as a positive control obviously promoted IFNγ production of mouse blood NK cells in vitro. However, activin A suppressed IFNγ production, but enhanced IL-2 synthesis and did not alter IL-10 production. Moreover, we found that activin A significantly suppressed the ability of NK cells to lyse target cells. These data revealed that blood NK cells of mouse were not only the target cells in response to activin A, but also the source of activin A, suggesting that activin A may play an important role in regulation of NK cells activities of mouse in an autocrine / paracrine manner.


Assuntos
Ativinas/farmacologia , Comunicação Autócrina , Células Matadoras Naturais/metabolismo , Comunicação Parácrina , Receptores de Activinas Tipo II/sangue , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Animais , Comunicação Autócrina/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Folistatina/farmacologia , Subunidades beta de Inibinas/sangue , Subunidades beta de Inibinas/genética , Subunidades beta de Inibinas/metabolismo , Interferon gama/biossíntese , Interleucina-10/biossíntese , Interleucina-2/biossíntese , Células Matadoras Naturais/efeitos dos fármacos , Masculino , Camundongos Endogâmicos BALB C , Comunicação Parácrina/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Smad/sangue , Proteínas Smad/genética , Proteínas Smad/metabolismo
18.
Cell Mol Life Sci ; 76(1): 179-192, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30310934

RESUMO

Human pluripotent stem cells (hPSCs) provide a source for the generation of defined kidney cells and renal organoids applicable in regenerative medicine, disease modeling, and drug screening. These applications require the provision of hPSC-derived renal cells by reproducible, scalable, and efficient methods. We established a chemically defined protocol by application of Activin A, BMP4, and Retinoic acid followed by GDNF, which steered hPSCs to the renal lineage and resulted in populations of SIX2+/CITED1+ metanephric mesenchyme- (MM) and of HOXB7+/GRHL2+ ureteric bud (UB)-like cells already by 6 days. Transcriptome analysis corroborated that the PSC-derived cell types at day 8 resemble their renal vesicle and ureteric epithelial counterpart in vivo, forming tubular and glomerular renal cells 6 days later. We demonstrate that starting from hPSCs, our in vitro protocol generates a pool of nephrogenic progenitors at the renal vesicle stage, which can be further directed into specialized nephronal cell types including mesangial-, proximal tubular-, distal tubular, collecting duct epithelial cells, and podocyte precursors after 14 days. This simple and rapid method to produce renal cells from a common precursor pool in 2D culture provides the basis for scaled-up production of tailored renal cell types, which are applicable for drug testing or cell-based regenerative therapies.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Néfrons/citologia , Células-Tronco Pluripotentes/citologia , Ativinas/farmacologia , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Humanos , Néfrons/efeitos dos fármacos , Néfrons/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Transcriptoma/efeitos dos fármacos , Tretinoína/farmacologia
19.
Proc Natl Acad Sci U S A ; 114(9): 2271-2276, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-28167799

RESUMO

Steering the differentiation of induced pluripotent stem cells (iPSCs) toward specific cell types is crucial for patient-specific disease modeling and drug testing. This effort requires the capacity to predict and control when and how multipotent progenitor cells commit to the desired cell fate. Cell fate commitment represents a critical state transition or "tipping point" at which complex systems undergo a sudden qualitative shift. To characterize such transitions during iPSC to cardiomyocyte differentiation, we analyzed the gene expression patterns of 96 developmental genes at single-cell resolution. We identified a bifurcation event early in the trajectory when a primitive streak-like cell population segregated into the mesodermal and endodermal lineages. Before this branching point, we could detect the signature of an imminent critical transition: increase in cell heterogeneity and coordination of gene expression. Correlation analysis of gene expression profiles at the tipping point indicates transcription factors that drive the state transition toward each alternative cell fate and their relationships with specific phenotypic readouts. The latter helps us to facilitate small molecule screening for differentiation efficiency. To this end, we set up an analysis of cell population structure at the tipping point after systematic variation of the protocol to bias the differentiation toward mesodermal or endodermal cell lineage. We were able to predict the proportion of cardiomyocytes many days before cells manifest the differentiated phenotype. The analysis of cell populations undergoing a critical state transition thus affords a tool to forecast cell fate outcomes and can be used to optimize differentiation protocols to obtain desired cell populations.


Assuntos
Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/genética , Transcriptoma , Ativinas/farmacologia , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/genética , Endoderma/citologia , Endoderma/metabolismo , Perfilação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Mesoderma/citologia , Mesoderma/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Piridinas/farmacologia , Pirimidinas/farmacologia , Análise de Célula Única , Fatores de Transcrição/metabolismo
20.
Proc Natl Acad Sci U S A ; 114(14): E2891-E2900, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28320933

RESUMO

Type 1 regulatory T (Tr1) cells play a pivotal role in restraining human T-cell responses toward environmental allergens and protecting against allergic diseases. Still, the precise molecular cues that underlie their transcriptional and functional specification remain elusive. Here, we show that the cytokine activin-A instructs the generation of CD4+ T cells that express the Tr1-cell-associated molecules IL-10, inducible T-Cell costimulator (ICOS), lymphocyte activation gene 3 protein (LAG-3), and CD49b, and exert strongly suppressive functions toward allergic responses induced by naive and in vivo-primed human T helper 2 cells. Moreover, mechanistic studies reveal that activin-A signaling induces the activation of the transcription factor interferon regulatory factor (IRF4), which, along with the environmental sensor aryl hydrocarbon receptor, forms a multipartite transcriptional complex that binds in IL-10 and ICOS promoter elements and controls gene expression in human CD4+ T cells. In fact, IRF4 silencing abrogates activin-A-driven IL10 and ICOS up-regulation and impairs the suppressive functions of human activin-A-induced Tr1-like (act-A-iTr1) cells. Importantly, using a humanized mouse model of allergic asthma, we demonstrate that adoptive transfer of human act-A-iTr1 cells, both in preventive and therapeutic protocols, confers significant protection against cardinal asthma manifestations, including pulmonary inflammation. Overall, our findings uncover an activin-A-induced IRF4-aryl hydrocarbon receptor (AhR)-dependent transcriptional network, which generates suppressive human Tr1 cells that may be harnessed for the control of allergic diseases.


Assuntos
Ativinas/metabolismo , Asma/prevenção & controle , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores Reguladores de Interferon/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Linfócitos T Reguladores/imunologia , Ativinas/farmacologia , Animais , Asma/imunologia , Asma/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Hipersensibilidade/metabolismo , Hipersensibilidade/prevenção & controle , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Camundongos SCID , Regiões Promotoras Genéticas , Transdução de Sinais , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA