Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Cell Regen ; 13(1): 11, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38780677

RESUMO

The family of secreted dimeric proteins known as the Transforming Growth Factor-ß (TGF-ß) family plays a critical role in facilitating intercellular communication within multicellular animals. A recent symposium on TGF-ß Biology - Signaling, Development, and Diseases, held on December 19-21, 2023, in Hangzhou, China, showcased some latest advances in our understanding TGF-ß biology and also served as an important forum for scientific collaboration and exchange of ideas. More than twenty presentations and discussions at the symposium delved into the intricate mechanisms of TGF-ß superfamily signaling pathways, their roles in normal development and immunity, and the pathological conditions associated with pathway dysregulation.

3.
Cell Syst ; 13(9): 724-736.e9, 2022 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-36057257

RESUMO

Identifying the chemical regulators of biological pathways is a time-consuming bottleneck in developing therapeutics and research compounds. Typically, thousands to millions of candidate small molecules are tested in target-based biochemical screens or phenotypic cell-based screens, both expensive experiments customized to each disease. Here, our uncustomized, virtual, profile-based screening approach instead identifies compounds that match to pathways based on the phenotypic information in public cell image data, created using the Cell Painting assay. Our straightforward correlation-based computational strategy retrospectively uncovered the expected, known small-molecule regulators for 32% of positive-control gene queries. In prospective, discovery mode, we efficiently identified new compounds related to three query genes and validated them in subsequent gene-relevant assays, including compounds that phenocopy or pheno-oppose YAP1 overexpression and kill a Yap1-dependent sarcoma cell line. This image-profile-based approach could replace many customized labor- and resource-intensive screens and accelerate the discovery of biologically and therapeutically useful compounds.


Assuntos
Estudos Prospectivos , Linhagem Celular , Estudos Retrospectivos
5.
J Pathol ; 254(4): 358-373, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33834494

RESUMO

Many chronic diseases are marked by fibrosis, which is defined by an abundance of activated fibroblasts and excessive deposition of extracellular matrix, resulting in loss of normal function of the affected organs. The initiation and progression of fibrosis are elaborated by pro-fibrotic cytokines, the most critical of which is transforming growth factor-ß1 (TGF-ß1). This review focuses on the fibrogenic roles of increased TGF-ß activities and underlying signaling mechanisms in the activated fibroblast population and other cell types that contribute to progression of fibrosis. Insight into these roles and mechanisms of TGF-ß as a universal driver of fibrosis has stimulated the development of therapeutic interventions to attenuate fibrosis progression, based on interference with TGF-ß signaling. Their promise in preclinical and clinical settings will be discussed. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Fibrose , Fator de Crescimento Transformador beta , Animais , Humanos
6.
Dev Cell ; 56(6): 726-746, 2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33756119

RESUMO

Epithelial cells repress epithelial characteristics and elaborate mesenchymal characteristics to migrate to other locations and acquire new properties. Epithelial plasticity responses are directed through cooperation of signaling pathways, with TGF-ß and TGF-ß-related proteins playing prominent instructive roles. Epithelial-mesenchymal transitions (EMTs) directed by activin-like molecules, bone morphogenetic proteins, or TGF-ß regulate metazoan development and wound healing and drive fibrosis and cancer progression. In carcinomas, diverse EMTs enable stem cell generation, anti-cancer drug resistance, genomic instability, and localized immunosuppression. This review discusses roles of TGF-ß and TGF-ß-related proteins, and underlying molecular mechanisms, in epithelial plasticity in development and wound healing, fibrosis, and cancer.


Assuntos
Células Epiteliais/patologia , Transição Epitelial-Mesenquimal , Neoplasias/patologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Humanos , Neoplasias/metabolismo
7.
Nat Rev Clin Oncol ; 18(1): 9-34, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32710082

RESUMO

TGFß signalling has key roles in cancer progression: most carcinoma cells have inactivated their epithelial antiproliferative response and benefit from increased TGFß expression and autocrine TGFß signalling through effects on gene expression, release of immunosuppressive cytokines and epithelial plasticity. As a result, TGFß enables cancer cell invasion and dissemination, stem cell properties and therapeutic resistance. TGFß released by cancer cells, stromal fibroblasts and other cells in the tumour microenvironment further promotes cancer progression by shaping the architecture of the tumour and by suppressing the antitumour activities of immune cells, thus generating an immunosuppressive environment that prevents or attenuates the efficacy of anticancer immunotherapies. The repression of TGFß signalling is therefore considered a prerequisite and major avenue to enhance the efficacy of current and forthcoming immunotherapies, including in tumours comprising cancer cells that are not TGFß responsive. Herein, we introduce the mechanisms underlying TGFß signalling in tumours and their microenvironment and discuss approaches to inhibit these signalling mechanisms as well as the use of these approaches in cancer immunotherapies and their potential adverse effects.


Assuntos
Imunoterapia/métodos , Neoplasias/terapia , Fator de Crescimento Transformador beta/metabolismo , Animais , Progressão da Doença , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Transdução de Sinais
8.
Nat Rev Mol Cell Biol ; 21(6): 341-352, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32300252

RESUMO

Epithelial-mesenchymal transition (EMT) encompasses dynamic changes in cellular organization from epithelial to mesenchymal phenotypes, which leads to functional changes in cell migration and invasion. EMT occurs in a diverse range of physiological and pathological conditions and is driven by a conserved set of inducing signals, transcriptional regulators and downstream effectors. With over 5,700 publications indexed by Web of Science in 2019 alone, research on EMT is expanding rapidly. This growing interest warrants the need for a consensus among researchers when referring to and undertaking research on EMT. This Consensus Statement, mediated by 'the EMT International Association' (TEMTIA), is the outcome of a 2-year-long discussion among EMT researchers and aims to both clarify the nomenclature and provide definitions and guidelines for EMT research in future publications. We trust that these guidelines will help to reduce misunderstanding and misinterpretation of research data generated in various experimental models and to promote cross-disciplinary collaboration to identify and address key open questions in this research field. While recognizing the importance of maintaining diversity in experimental approaches and conceptual frameworks, we emphasize that lasting contributions of EMT research to increasing our understanding of developmental processes and combatting cancer and other diseases depend on the adoption of a unified terminology to describe EMT.


Assuntos
Pesquisa Biomédica/normas , Transição Epitelial-Mesenquimal , Animais , Movimento Celular , Plasticidade Celular , Consenso , Biologia do Desenvolvimento/normas , Humanos , Neoplasias/patologia , Terminologia como Assunto
9.
Sci Rep ; 9(1): 16992, 2019 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-31740700

RESUMO

Insulin signaling governs many processes including glucose homeostasis and metabolism, and is therapeutically used to treat hyperglycemia in diabetes. We demonstrated that insulin-induced Akt activation enhances the sensitivity to TGF-ß by directing an increase in cell surface TGF-ß receptors from a pool of intracellular TGF-ß receptors. Consequently, increased autocrine TGF-ß signaling in response to insulin participates in insulin-induced angiogenic responses of endothelial cells. With TGF-ß signaling controlling many cell responses, including differentiation and extracellular matrix deposition, and pathologically promoting fibrosis and cancer cell dissemination, we addressed to which extent autocrine TGF-ß signaling participates in insulin-induced gene responses of human endothelial cells. Transcriptome analyses of the insulin response, in the absence or presence of a TGF-ß receptor kinase inhibitor, revealed substantial positive and negative contributions of autocrine TGF-ß signaling in insulin-responsive gene responses. Furthermore, insulin-induced responses of many genes depended on or resulted from autocrine TGF-ß signaling. Our analyses also highlight extensive contributions of autocrine TGF-ß signaling to basal gene expression in the absence of insulin, and identified many novel TGF-ß-responsive genes. This data resource may aid in the appreciation of the roles of autocrine TGF-ß signaling in normal physiological responses to insulin, and implications of therapeutic insulin usage.


Assuntos
Perfilação da Expressão Gênica/métodos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Insulina/farmacologia , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteínas Smad/genética , Fator de Crescimento Transformador beta/farmacologia , Benzamidas/farmacologia , Células Cultivadas , Dioxóis/farmacologia , Ontologia Genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hipoglicemiantes/farmacologia , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteínas Smad/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo
10.
Dev Cell ; 49(3): 313-316, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-31063750

RESUMO

Epithelial cells acquire mesenchymal characteristics during development, wound healing and inflammation, and in cancer and fibrosis. With increasing appreciation of different roles of epithelial-mesenchymal transition (EMT), we address the question of how to define and recognize EMT processes and discuss their properties in cancer progression.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Neoplasias/metabolismo , Animais , Diferenciação Celular/fisiologia , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Humanos , Transdução de Sinais , Cicatrização/fisiologia
11.
J Biol Chem ; 294(21): 8490-8504, 2019 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-30948511

RESUMO

Functional activation of the transforming growth factor-ß (TGF-ß) receptors (TGFBRs) is carefully regulated through integration of post-translational modifications, spatial regulation at the cellular level, and TGFBR availability at the cell surface. Although the bulk of TGFBRs resides inside the cells, AKT Ser/Thr kinase (AKT) activation in response to insulin or other growth factors rapidly induces transport of TGFBRs to the cell surface, thereby increasing the cell's responsiveness to TGF-ß. We now demonstrate that TGF-ß itself induces a rapid translocation of its own receptors to the cell surface and thus amplifies its own response. This mechanism of response amplification, which hitherto has not been reported for other cell-surface receptors, depended on AKT activation and TGF-ß type I receptor kinase. In addition to an increase in cell-surface TGFBR levels, TGF-ß treatment promoted TGFBR internalization, suggesting an overall amplification of TGFBR cycling. The TGF-ß-induced increase in receptor presentation at the cell surface amplified TGF-ß-induced SMAD family member (SMAD) activation and gene expression. Furthermore, bone morphogenetic protein 4 (BMP-4), which also induces AKT activation, increased TGFBR levels at the cell surface, leading to enhanced autocrine activation of TGF-ß-responsive SMADs and gene expression, providing context for the activation of TGF-ß signaling in response to BMP during development. In summary, our results indicate that TGF-ß- and BMP-induced activation of low levels of cell surface-associated TGFBRs rapidly mobilizes additional TGFBRs from intracellular stores to the cell surface, increasing the abundance of cell-surface TGFBRs and cells' responsiveness to TGF-ß signaling.


Assuntos
Receptor do Fator de Crescimento Transformador beta Tipo I/biossíntese , Transdução de Sinais , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima , Células A549 , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Humanos , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Proteínas Smad/genética , Fator de Crescimento Transformador beta/genética
12.
Sci Signal ; 12(570)2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30808818

RESUMO

Encoded in mammalian cells by 33 genes, the transforming growth factor-ß (TGF-ß) family of secreted, homodimeric and heterodimeric proteins controls the differentiation of most, if not all, cell lineages and many aspects of cell and tissue physiology in multicellular eukaryotes. Deregulation of TGF-ß family signaling leads to developmental anomalies and disease, whereas enhanced TGF-ß signaling contributes to cancer and fibrosis. Here, we review the fundamentals of the signaling mechanisms that are initiated upon TGF-ß ligand binding to its cell surface receptors and the dependence of the signaling responses on input from and cooperation with other signaling pathways. We discuss how cells exquisitely control the functional presentation and activation of heteromeric receptor complexes of transmembrane, dual-specificity kinases and, thus, define their context-dependent responsiveness to ligands. We also introduce the mechanisms through which proteins called Smads act as intracellular effectors of ligand-induced gene expression responses and show that the specificity and impressive versatility of Smad signaling depend on cross-talk from other pathways. Last, we discuss how non-Smad signaling mechanisms, initiated by distinct ligand-activated receptor complexes, complement Smad signaling and thus contribute to cellular responses.


Assuntos
Regulação da Expressão Gênica/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Ligantes , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Ligação Proteica , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia
13.
Sci Signal ; 12(570)2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30808819

RESUMO

Tumors comprise cancer stem cells (CSCs) and their heterogeneous progeny within a stromal microenvironment. In response to transforming growth factor-ß (TGF-ß), epithelial and carcinoma cells undergo a partial or complete epithelial-mesenchymal transition (EMT), which contributes to cancer progression. This process is seen as reversible because cells revert to an epithelial phenotype upon TGF-ß removal. However, we found that prolonged TGF-ß exposure, mimicking the state of in vivo carcinomas, promotes stable EMT in mammary epithelial and carcinoma cells, in contrast to the reversible EMT induced by a shorter exposure. The stabilized EMT was accompanied by stably enhanced stem cell generation and anticancer drug resistance. Furthermore, prolonged TGF-ß exposure enhanced mammalian target of rapamycin (mTOR) signaling. A bitopic mTOR inhibitor repressed CSC generation, anchorage independence, cell survival, and chemoresistance and efficiently inhibited tumorigenesis in mice. These results reveal a role for mTOR in the stabilization of stemness and drug resistance of breast cancer cells and position mTOR inhibition as a treatment strategy to target CSCs.


Assuntos
Antineoplásicos/farmacologia , Transformação Celular Neoplásica/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Animais , Benzamidas/farmacologia , Linhagem Celular Transformada , Transformação Celular Neoplásica/genética , Células Cultivadas , Dioxóis/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Humanos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Células-Tronco Neoplásicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
14.
iScience ; 11: 474-491, 2019 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-30684493

RESUMO

Angiogenesis, the development of new blood vessels, is a key process in disease. We reported that insulin promotes translocation of transforming growth factor ß (TGF-ß) receptors to the plasma membrane of epithelial and fibroblast cells, thus enhancing TGF-ß responsiveness. Since insulin promotes angiogenesis, we addressed whether increased autocrine TGF-ß signaling participates in endothelial cell responses to insulin. We show that insulin enhances TGF-ß responsiveness and autocrine TGF-ß signaling in primary human endothelial cells, by inducing a rapid increase in cell surface TGF-ß receptor levels. Autocrine TGF-ß/Smad signaling contributed substantially to insulin-induced gene expression associated with angiogenesis, including TGF-ß target genes encoding angiogenic mediators; was essential for endothelial cell migration; and participated in endothelial cell invasion and network formation. Blocking TGF-ß signaling impaired insulin-induced microvessel outgrowth from neonatal aortic rings and modified insulin-stimulated blood vessel formation in zebrafish. We conclude that enhanced autocrine TGF-ß signaling is integral to endothelial cell and angiogenic responses to insulin.

15.
J Biol Chem ; 293(34): 13059-13072, 2018 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-29907569

RESUMO

The epithelial-to-mesenchymal transdifferentiation (EMT) is crucial for tissue differentiation in development and drives essential steps in cancer and fibrosis. EMT is accompanied by reprogramming of gene expression and has been associated with the epithelial stem-cell state in normal and carcinoma cells. The cytokine transforming growth factor ß (TGF-ß) drives this program in cooperation with other signaling pathways and through TGF-ß-activated SMAD3 as the major effector. TGF-ß-induced SMAD3 activation is inhibited by SMAD7 and to a lesser extent by SMAD6, and SMAD6 and SMAD7 both inhibit SMAD1 and SMAD5 activation in response to the TGF-ß-related bone morphogenetic proteins (BMPs). We previously reported that, in response to BMP, protein arginine methyltransferase 1 (PRMT1) methylates SMAD6 at the BMP receptor complex, thereby promoting its dissociation from the receptors and enabling BMP-induced SMAD1 and SMAD5 activation. We now provide evidence that PRMT1 also facilitates TGF-ß signaling by methylating SMAD7, which complements SMAD6 methylation. We found that PRMT1 is required for TGF-ß-induced SMAD3 activation, through a mechanism similar to that of BMP-induced SMAD6 methylation, and thus promotes the TGF-ß-induced EMT and epithelial stem-cell generation. This critical mechanism positions PRMT1 as an essential mediator of TGF-ß signaling that controls the EMT and epithelial cell stemness through SMAD7 methylation.


Assuntos
Arginina/química , Células Epiteliais/citologia , Transição Epitelial-Mesenquimal , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Proteína Smad7/metabolismo , Células-Tronco/citologia , Fator de Crescimento Transformador beta1/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Células Cultivadas , Células Epiteliais/fisiologia , Humanos , Metilação , Proteína-Arginina N-Metiltransferases/genética , Proteínas Repressoras/genética , Pele/citologia , Pele/metabolismo , Proteína Smad7/genética , Células-Tronco/fisiologia , Fator de Crescimento Transformador beta1/genética
16.
EMBO Rep ; 19(1): 135-155, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29233829

RESUMO

During epithelial-mesenchymal transition (EMT), reprogramming of gene expression is accompanied by histone modifications. Whether EMT-promoting signaling directs functional changes in histone methylation has not been established. We show here that the histone lysine methyltransferase SETDB1 represses EMT and that, during TGF-ß-induced EMT, cells attenuate SETDB1 expression to relieve this inhibition. SETDB1 also controls stem cell generation, cancer cell motility, invasion, metastatic dissemination, as well as sensitivity to certain cancer drugs. These functions may explain the correlation of breast cancer patient survival with SETDB1 expression. At the molecular level, TGF-ß induces SETDB1 recruitment by Smad3, to repress Smad3/4-activated transcription of SNAI1, encoding the EMT "master" transcription factor SNAIL1. Suppression of SNAIL1-mediated gene reprogramming by SETDB1 occurs through H3K9 methylation at the SNAI1 gene that represses its H3K9 acetylation imposed by activated Smad3/4 complexes. SETDB1 therefore defines a TGF-ß-regulated balance between histone methylation and acetylation that controls EMT.


Assuntos
Neoplasias da Mama/genética , Carcinoma Ductal/genética , Transição Epitelial-Mesenquimal/genética , Histonas/genética , Proteínas Metiltransferases/genética , Proteína Smad3/genética , Fatores de Transcrição da Família Snail/genética , Acetilação , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Ductal/metabolismo , Carcinoma Ductal/patologia , Linhagem Celular Tumoral , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Humanos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/patologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Metilação , Camundongos , Proteínas Metiltransferases/metabolismo , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Proteína Smad3/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Fator de Crescimento Transformador beta/farmacologia
17.
J Clin Invest ; 127(10): 3675-3688, 2017 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-28872461

RESUMO

TGF-ß1 signaling is a critical driver of collagen accumulation and fibrotic disease but also a vital suppressor of inflammation and epithelial cell proliferation. The nature of this multifunctional cytokine has limited the development of global TGF-ß1 signaling inhibitors as therapeutic agents. We conducted phenotypic screens for small molecules that inhibit TGF-ß1-induced epithelial-mesenchymal transition without immediate TGF-ß1 receptor (TßR) kinase inhibition. We identified trihydroxyphenolic compounds as potent blockers of TGF-ß1 responses (IC50 ~50 nM), Snail1 expression, and collagen deposition in vivo in models of pulmonary fibrosis and collagen-dependent lung cancer metastasis. Remarkably, the functional effects of trihydroxyphenolics required the presence of active lysyl oxidase-like 2 (LOXL2), thereby limiting effects to fibroblasts or cancer cells, the major LOXL2 producers. Mechanistic studies revealed that trihydroxyphenolics induce auto-oxidation of a LOXL2/3-specific lysine (K731) in a time-dependent reaction that irreversibly inhibits LOXL2 and converts the trihydrophenolic to a previously undescribed metabolite that directly inhibits TßRI kinase. Combined inhibition of LOXL2 and TßRI activities by trihydrophenolics resulted in potent blockade of pathological collagen accumulation in vivo without the toxicities associated with global inhibitors. These findings elucidate a therapeutic approach to attenuate fibrosis and the disease-promoting effects of tissue stiffness by specifically targeting TßRI kinase in LOXL2-expressing cells.


Assuntos
Inibidores Enzimáticos , Transição Epitelial-Mesenquimal , Fibroblastos/metabolismo , Neoplasias Pulmonares , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Fibrose Pulmonar , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo , Células A549 , Aminoácido Oxirredutases/genética , Aminoácido Oxirredutases/metabolismo , Animais , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Fibroblastos/patologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Metástase Neoplásica , Proteínas de Neoplasias/genética , Fenóis/química , Fenóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/genética
18.
Trends Cell Biol ; 27(9): 658-672, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28552280

RESUMO

Transforming growth factor (TGF)-ß family proteins control cell physiology, proliferation, and growth, and direct cell differentiation, thus playing key roles in normal development and disease. The mechanisms of how TGF-ß family ligands interact with heteromeric complexes of cell surface receptors to then activate Smad signaling that directs changes in gene expression are often seen as established. Even though TGF-ß-induced Smad signaling may be seen as a linear signaling pathway with predictable outcomes, this pathway provides cells with a versatile means to induce different cellular responses. Fundamental questions remain as to how, at the molecular level, TGF-ß and TGF-ß family proteins activate the receptor complexes and induce a context-dependent diversity of cell responses. Among the areas of progress, we summarize new insights into how cells control TGF-ß responsiveness by controlling the TGF-ß receptors, and into the key roles and versatility of Smads in directing cell differentiation and cell fate selection.


Assuntos
Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Smad/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Humanos , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo
19.
Proc Natl Acad Sci U S A ; 113(44): 12478-12483, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27738243

RESUMO

Developmental signaling molecules are used for cell fate determination, and understanding how their combinatorial effects produce the variety of cell types in multicellular organisms is a key problem in biology. Here, we demonstrate that the combination of leukemia inhibitory factor (LIF), bone morphogenetic protein 4 (BMP4), lysophosphatidic acid (LPA), and ascorbic acid (AA) efficiently converts mouse primed pluripotent stem cells (PSCs) into naive PSCs. Signaling by the lipid LPA through its receptor LPAR1 and downstream effector Rho-associated protein kinase (ROCK) cooperated with LIF signaling to promote this conversion. BMP4, which also stimulates conversion to naive pluripotency, bypassed the need for exogenous LPA by increasing the activity of the extracellular LPA-producing enzyme autotaxin (ATX). We found that LIF and LPA-LPAR1 signaling affect the abundance of signal transducer and activator of transcription 3 (STAT3), which induces a previously unappreciated Kruppel-like factor (KLF)2-KLF4-PR domain 14 (PRDM14) transcription factor circuit key to establish naive pluripotency. AA also affects this transcription factor circuit by controlling PRDM14 expression. Thus, our study reveals that ATX-mediated autocrine lipid signaling promotes naive pluripotency by intersecting with LIF and BMP4 signaling.


Assuntos
Proteína Morfogenética Óssea 4/farmacologia , Fator Inibidor de Leucemia/farmacologia , Lisofosfolipídeos/farmacologia , Diester Fosfórico Hidrolases/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Animais , Ácido Ascórbico/farmacologia , Linhagem Celular , Reprogramação Celular/efeitos dos fármacos , Reprogramação Celular/genética , Sinergismo Farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , Fator 4 Semelhante a Kruppel , Camundongos Endogâmicos C57BL , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição/genética , Vitaminas/farmacologia
20.
Artigo em Inglês | MEDLINE | ID: mdl-27328871

RESUMO

Transforming growth factors (TGFs) were discovered as activities that were secreted by cancer cells, and later by normal cells, and had the ability to phenotypically and reversibly transform immortalized fibroblasts. TGF-ß distinguished itself from TGF-α because it did not bind to the same epidermal growth factor (EGF) receptor as TGF-α and, therefore, acted through different cell-surface receptors and signaling mediators. This review summarizes the discovery of TGF-ß, the early developments in its molecular and biological characterization with its many biological activities in different cell and tissue contexts and its roles in disease, the realization that there is a family of secreted TGF-ß-related proteins with many differentiation functions in development and activities in normal cell and tissue physiology, and the subsequent identification and characterization of the receptors and effectors that mediate TGF-ß family signaling responses.


Assuntos
Fator de Crescimento Transformador beta/fisiologia , Animais , Clonagem Molecular , DNA Complementar/genética , Humanos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...