Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 156
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(19): 4007-4037, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-37714133

RESUMEN

The TGF-ß regulatory system plays crucial roles in the preservation of organismal integrity. TGF-ß signaling controls metazoan embryo development, tissue homeostasis, and injury repair through coordinated effects on cell proliferation, phenotypic plasticity, migration, metabolic adaptation, and immune surveillance of multiple cell types in shared ecosystems. Defects of TGF-ß signaling, particularly in epithelial cells, tissue fibroblasts, and immune cells, disrupt immune tolerance, promote inflammation, underlie the pathogenesis of fibrosis and cancer, and contribute to the resistance of these diseases to treatment. Here, we review how TGF-ß coordinates multicellular response programs in health and disease and how this knowledge can be leveraged to develop treatments for diseases of the TGF-ß system.


Asunto(s)
Transducción de Señal , Factor de Crecimiento Transformador beta , Animales , Adaptación Fisiológica , Proliferación Celular , Desarrollo Embrionario
2.
Nat Rev Mol Cell Biol ; 21(6): 341-352, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32300252

RESUMEN

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.


Asunto(s)
Investigación Biomédica/normas , Transición Epitelial-Mesenquimal , Animales , Movimiento Celular , Plasticidad de la Célula , Consenso , Biología Evolutiva/normas , Humanos , Neoplasias/patología , Terminología como Asunto
3.
Cell ; 168(6): 1101-1113.e13, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28283064

RESUMEN

We molecularly dissected leptomeningeal metastasis, or spread of cancer to the cerebrospinal fluid (CSF), which is a frequent and fatal condition mediated by unknown mechanisms. We selected lung and breast cancer cell lines for the ability to infiltrate and grow in CSF, a remarkably acellular, mitogen-poor metastasis microenvironment. Complement component 3 (C3) was upregulated in four leptomeningeal metastatic models and proved necessary for cancer growth within the leptomeningeal space. In human disease, cancer cells within the CSF produced C3 in correlation with clinical course. C3 expression in primary tumors was predictive of leptomeningeal relapse. Mechanistically, we found that cancer-cell-derived C3 activates the C3a receptor in the choroid plexus epithelium to disrupt the blood-CSF barrier. This effect allows plasma components, including amphiregulin, and other mitogens to enter the CSF and promote cancer cell growth. Pharmacologic interference with C3 signaling proved therapeutically beneficial in suppressing leptomeningeal metastasis in these preclinical models.


Asunto(s)
Complemento C3/metabolismo , Neoplasias Meníngeas/secundario , Metástasis de la Neoplasia/patología , Animales , Neoplasias de la Mama/patología , Líquido Cefalorraquídeo , Plexo Coroideo/irrigación sanguínea , Complemento C3/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias Pulmonares/patología , Antígeno de Macrófago-1/metabolismo , Ratones , Transducción de Señal , Microambiente Tumoral , Regulación hacia Arriba
4.
Cell ; 165(1): 45-60, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-27015306

RESUMEN

Metastasis frequently develops years after the removal of a primary tumor, from a minority of disseminated cancer cells that survived as latent entities through unknown mechanisms. We isolated latency competent cancer (LCC) cells from early stage human lung and breast carcinoma cell lines and defined the mechanisms that suppress outgrowth, support long-term survival, and maintain tumor-initiating potential in these cells during the latent metastasis stage. LCC cells show stem-cell-like characteristics and express SOX2 and SOX9 transcription factors, which are essential for their survival in host organs under immune surveillance and for metastatic outgrowth under permissive conditions. Through expression of the WNT inhibitor DKK1, LCC cells self-impose a slow-cycling state with broad downregulation of ULBP ligands for NK cells and evasion of NK-cell-mediated clearance. By expressing a Sox-dependent stem-like state and actively silencing WNT signaling, LCC cells can enter quiescence and evade innate immunity to remain latent for extended periods.


Asunto(s)
Comunicación Autocrina , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Metástasis de la Neoplasia/inmunología , Metástasis de la Neoplasia/patología , Escape del Tumor , Vía de Señalización Wnt , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Vigilancia Inmunológica , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción SOXB1/metabolismo
5.
Cell ; 164(5): 1015-30, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26898331

RESUMEN

TGF-ß signaling can be pro-tumorigenic or tumor suppressive. We investigated this duality in pancreatic ductal adenocarcinoma (PDA), which, with other gastrointestinal cancers, exhibits frequent inactivation of the TGF-ß mediator Smad4. We show that TGF-ß induces an epithelial-mesenchymal transition (EMT), generally considered a pro-tumorigenic event. However, in TGF-ß-sensitive PDA cells, EMT becomes lethal by converting TGF-ß-induced Sox4 from an enforcer of tumorigenesis into a promoter of apoptosis. This is the result of an EMT-linked remodeling of the cellular transcription factor landscape, including the repression of the gastrointestinal lineage-master regulator Klf5. Klf5 cooperates with Sox4 in oncogenesis and prevents Sox4-induced apoptosis. Smad4 is required for EMT but dispensable for Sox4 induction by TGF-ß. TGF-ß-induced Sox4 is thus geared to bolster progenitor identity, whereas simultaneous Smad4-dependent EMT strips Sox4 of an essential partner in oncogenesis. Our work demonstrates that TGF-ß tumor suppression functions through an EMT-mediated disruption of a lineage-specific transcriptional network.


Asunto(s)
Carcinoma Ductal/genética , Transición Epitelial-Mesenquimal , Redes Reguladoras de Genes , Neoplasias Pancreáticas/genética , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Apoptosis , Carcinoma Ductal/patología , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Organoides/metabolismo , Organoides/patología , Neoplasias Pancreáticas/patología , Factores de Transcripción SOXC/metabolismo , Proteína Smad4/metabolismo
6.
Immunity ; 54(5): 1037-1054.e7, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33756102

RESUMEN

Immune cells identify and destroy tumors by recognizing cellular traits indicative of oncogenic transformation. In this study, we found that myocardin-related transcription factors (MRTFs), which promote migration and metastatic invasion, also sensitize cancer cells to the immune system. Melanoma and breast cancer cells with high MRTF expression were selectively eliminated by cytotoxic lymphocytes in mouse models of metastasis. This immunosurveillance phenotype was further enhanced by treatment with immune checkpoint blockade (ICB) antibodies. We also observed that high MRTF signaling in human melanoma is associated with ICB efficacy in patients. Using biophysical and functional assays, we showed that MRTF overexpression rigidified the filamentous actin cytoskeleton and that this mechanical change rendered mouse and human cancer cells more vulnerable to cytotoxic T lymphocytes and natural killer cells. Collectively, these results suggest that immunosurveillance has a mechanical dimension, which we call mechanosurveillance, that is particularly relevant for the targeting of metastatic disease.


Asunto(s)
Linfocitos/inmunología , Neoplasias/inmunología , Citoesqueleto de Actina/inmunología , Actinas/inmunología , Animales , Comunicación Celular/inmunología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/inmunología , Femenino , Células HEK293 , Humanos , Células Asesinas Naturales/inmunología , Células MCF-7 , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología , Factores de Transcripción/inmunología
7.
Nat Rev Mol Cell Biol ; 19(7): 419-435, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29643418

RESUMEN

Few cell signals match the impact of the transforming growth factor-ß (TGFß) family in metazoan biology. TGFß cytokines regulate cell fate decisions during development, tissue homeostasis and regeneration, and are major players in tumorigenesis, fibrotic disorders, immune malfunctions and various congenital diseases. The effects of the TGFß family are mediated by a combinatorial set of ligands and receptors and by a common set of receptor-activated mothers against decapentaplegic homologue (SMAD) transcription factors, yet the effects can differ dramatically depending on the cell type and the conditions. Recent progress has illuminated a model of TGFß action in which SMADs bind genome-wide in partnership with lineage-determining transcription factors and additionally integrate inputs from other pathways and the chromatin to trigger specific cellular responses. These new insights clarify the operating logic of the TGFß pathway in physiology and disease.

8.
Nat Rev Mol Cell Biol ; 19(7): 479, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29740128

RESUMEN

In the section 'Combinatorial ligand perception' of the original article, DMP1 was incorrectly used in place of BMP. This has now been corrected in the HTML and PDF versions of the article.

9.
Nature ; 626(8000): 864-873, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38326607

RESUMEN

Macrophage activation is controlled by a balance between activating and inhibitory receptors1-7, which protect normal tissues from excessive damage during infection8,9 but promote tumour growth and metastasis in cancer7,10. Here we report that the Kupffer cell lineage-determining factor ID3 controls this balance and selectively endows Kupffer cells with the ability to phagocytose live tumour cells and orchestrate the recruitment, proliferation and activation of natural killer and CD8 T lymphoid effector cells in the liver to restrict the growth of a variety of tumours. ID3 shifts the macrophage inhibitory/activating receptor balance to promote the phagocytic and lymphoid response, at least in part by buffering the binding of the transcription factors ELK1 and E2A at the SIRPA locus. Furthermore, loss- and gain-of-function experiments demonstrate that ID3 is sufficient to confer this potent anti-tumour activity to mouse bone-marrow-derived macrophages and human induced pluripotent stem-cell-derived macrophages. Expression of ID3 is therefore necessary and sufficient to endow macrophages with the ability to form an efficient anti-tumour niche, which could be harnessed for cell therapy in cancer.


Asunto(s)
Proteínas Inhibidoras de la Diferenciación , Macrófagos del Hígado , Neoplasias , Animales , Humanos , Ratones , Células de la Médula Ósea/citología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linaje de la Célula , Células Madre Pluripotentes Inducidas/citología , Proteínas Inhibidoras de la Diferenciación/deficiencia , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Macrófagos del Hígado/citología , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/metabolismo , Hígado/inmunología , Hígado/patología , Activación de Macrófagos , Proteínas de Neoplasias , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Fagocitosis
10.
Cell ; 156(5): 1002-16, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24581498

RESUMEN

Brain metastasis is an ominous complication of cancer, yet most cancer cells that infiltrate the brain die of unknown causes. Here, we identify plasmin from the reactive brain stroma as a defense against metastatic invasion, and plasminogen activator (PA) inhibitory serpins in cancer cells as a shield against this defense. Plasmin suppresses brain metastasis in two ways: by converting membrane-bound astrocytic FasL into a paracrine death signal for cancer cells, and by inactivating the axon pathfinding molecule L1CAM, which metastatic cells express for spreading along brain capillaries and for metastatic outgrowth. Brain metastatic cells from lung cancer and breast cancer express high levels of anti-PA serpins, including neuroserpin and serpin B2, to prevent plasmin generation and its metastasis-suppressive effects. By protecting cancer cells from death signals and fostering vascular co-option, anti-PA serpins provide a unifying mechanism for the initiation of brain metastasis in lung and breast cancers.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Encéfalo/metabolismo , Fibrinolisina/metabolismo , Neuropéptidos/metabolismo , Inhibidor 2 de Activador Plasminogénico/metabolismo , Serpinas/metabolismo , Adenocarcinoma/secundario , Animales , Astrocitos/metabolismo , Encéfalo/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma/secundario , Línea Celular Tumoral , Supervivencia Celular , Modelos Animales de Enfermedad , Proteína Ligando Fas/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neuropéptidos/genética , Inhibidor 2 de Activador Plasminogénico/genética , Activadores Plasminogénicos/metabolismo , Serpinas/genética , Neuroserpina
11.
Nature ; 616(7958): 806-813, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36991128

RESUMEN

Metastasis frequently develops from disseminated cancer cells that remain dormant after the apparently successful treatment of a primary tumour. These cells fluctuate between an immune-evasive quiescent state and a proliferative state liable to immune-mediated elimination1-6. Little is known about the clearing of reawakened metastatic cells and how this process could be therapeutically activated to eliminate residual disease in patients. Here we use models of indolent lung adenocarcinoma metastasis to identify cancer cell-intrinsic determinants of immune reactivity during exit from dormancy. Genetic screens of tumour-intrinsic immune regulators identified the stimulator of interferon genes (STING) pathway as a suppressor of metastatic outbreak. STING activity increases in metastatic progenitors that re-enter the cell cycle and is dampened by hypermethylation of the STING promoter and enhancer in breakthrough metastases or by chromatin repression in cells re-entering dormancy in response to TGFß. STING expression in cancer cells derived from spontaneous metastases suppresses their outgrowth. Systemic treatment of mice with STING agonists eliminates dormant metastasis and prevents spontaneous outbreaks in a T cell- and natural killer cell-dependent manner-these effects require cancer cell STING function. Thus, STING provides a checkpoint against the progression of dormant metastasis and a therapeutically actionable strategy for the prevention of disease relapse.


Asunto(s)
Neoplasias Pulmonares , Metástasis de la Neoplasia , Animales , Ratones , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/inmunología , Adenocarcinoma del Pulmón/patología , Ciclo Celular , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/inmunología , Metástasis de la Neoplasia/patología , Recurrencia Local de Neoplasia/tratamiento farmacológico , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta , Células Asesinas Naturales/inmunología
12.
Immunity ; 50(4): 924-940, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30995507

RESUMEN

Transforming growth factor (TGF)-ß is a crucial enforcer of immune homeostasis and tolerance, inhibiting the expansion and function of many components of the immune system. Perturbations in TGF-ß signaling underlie inflammatory diseases and promote tumor emergence. TGF-ß is also central to immune suppression within the tumor microenvironment, and recent studies have revealed roles in tumor immune evasion and poor responses to cancer immunotherapy. Here, we present an overview of the complex biology of the TGF-ß family and its context-dependent nature. Then, focusing on cancer, we discuss the roles of TGF-ß signaling in distinct immune cell types and how this knowledge is being leveraged to unleash the immune system against the tumor.


Asunto(s)
Neoplasias/inmunología , Factor de Crecimiento Transformador beta/fisiología , Inmunidad Adaptativa , Animales , Células Dendríticas/inmunología , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Fibroblastos/inmunología , Humanos , Inmunidad Innata , Inflamación , Macrófagos/inmunología , Ratones Noqueados , Neutrófilos/inmunología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/inmunología , Escape del Tumor , Microambiente Tumoral
14.
Cell ; 154(5): 1060-1073, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23993096

RESUMEN

How organ-specific metastatic traits arise in primary tumors remains unknown. Here, we show a role of the breast tumor stroma in selecting cancer cells that are primed for metastasis in bone. Cancer-associated fibroblasts (CAFs) in triple-negative (TN) breast tumors skew heterogeneous cancer cell populations toward a predominance of clones that thrive on the CAF-derived factors CXCL12 and IGF1. Limiting concentrations of these factors select for cancer cells with high Src activity, a known clinical predictor of bone relapse and an enhancer of PI3K-Akt pathway activation by CXCL12 and IGF1. Carcinoma clones selected in this manner are primed for metastasis in the CXCL12-rich microenvironment of the bone marrow. The evidence suggests that stromal signals resembling those of a distant organ select for cancer cells that are primed for metastasis in that organ, thus illuminating the evolution of metastatic traits in a primary tumor and its distant metastases.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Metástasis de la Neoplasia , Transducción de Señal , Animales , Médula Ósea/metabolismo , Neoplasias Óseas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Quimiocina CXCL12/metabolismo , Fibroblastos/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Trasplante de Neoplasias , Transcripción Genética , Trasplante Heterólogo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
15.
Immunity ; 48(4): 626-628, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29669246

RESUMEN

Immune checkpoint therapy can induce durable remissions, but many tumors demonstrate resistance. In a recent issue of Nature, Mariathasan et al. (2018) and Tauriello et al. (2018) identify stromal TGF-ß signaling as a determinant of immune exclusion. Combination TGF-ß inhibition and immunotherapy induces complete responses in mouse models.


Asunto(s)
Linfocitos T , Factor de Crecimiento Transformador beta , Animales , Modelos Animales de Enfermedad , Inmunoterapia , Ratones , Neoplasias
16.
Cell ; 149(6): 1179-81, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22682238

RESUMEN

Mutations in keratinocyte and melanocyte precursors that are caused by extensive sun exposure are well-established contributors to skin cancer. Now Hu et al. provide evidence that the sun's harmful rays may also cause tumor-promoting epigenetic modifications in dermal fibroblasts, highlighting further the importance of tumor-stroma interactions in cancer.

17.
Cell ; 150(1): 165-78, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22770218

RESUMEN

Metastasis and chemoresistance in cancer are linked phenomena, but the molecular basis for this link is unknown. We uncovered a network of paracrine signals between carcinoma, myeloid, and endothelial cells that drives both processes in breast cancer. Cancer cells that overexpress CXCL1 and 2 by transcriptional hyperactivation or 4q21 amplification are primed for survival in metastatic sites. CXCL1/2 attract CD11b(+)Gr1(+) myeloid cells into the tumor, which produce chemokines including S100A8/9 that enhance cancer cell survival. Although chemotherapeutic agents kill cancer cells, these treatments trigger a parallel stromal reaction leading to TNF-α production by endothelial and other stromal cells. TNF-α via NF-kB heightens the CXCL1/2 expression in cancer cells, thus amplifying the CXCL1/2-S100A8/9 loop and causing chemoresistance. CXCR2 blockers break this cycle, augmenting the efficacy of chemotherapy against breast tumors and particularly against metastasis. This network of endothelial-carcinoma-myeloid signaling interactions provides a mechanism linking chemoresistance and metastasis, with opportunities for intervention.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma/patología , Quimiocina CXCL1/metabolismo , Resistencia a Antineoplásicos , Metástasis de la Neoplasia , Comunicación Paracrina , Animales , Neoplasias de la Mama/metabolismo , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Carcinoma/metabolismo , Quimiocina CXCL1/genética , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/secundario , Ganglios Linfáticos/patología , Metástasis Linfática , Ratones , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Trasplante de Neoplasias , Trasplante Heterólogo
18.
Genes Dev ; 33(21-22): 1506-1524, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31582430

RESUMEN

TGF-ß receptors phosphorylate SMAD2 and SMAD3 transcription factors, which then form heterotrimeric complexes with SMAD4 and cooperate with context-specific transcription factors to activate target genes. Here we provide biochemical and structural evidence showing that binding of SMAD2 to DNA depends on the conformation of the E3 insert, a structural element unique to SMAD2 and previously thought to render SMAD2 unable to bind DNA. Based on this finding, we further delineate TGF-ß signal transduction by defining distinct roles for SMAD2 and SMAD3 with the forkhead pioneer factor FOXH1 as a partner in the regulation of differentiation genes in mouse mesendoderm precursors. FOXH1 is prebound to target sites in these loci and recruits SMAD3 independently of TGF-ß signals, whereas SMAD2 remains predominantly cytoplasmic in the basal state and set to bind SMAD4 and join SMAD3:FOXH1 at target promoters in response to Nodal TGF-ß signals. The results support a model in which signal-independent binding of SMAD3 and FOXH1 prime mesendoderm differentiation gene promoters for activation, and signal-driven SMAD2:SMAD4 binds to promoters that are preloaded with SMAD3:FOXH1 to activate transcription.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Modelos Moleculares , Transducción de Señal , Proteína Smad2 , Proteína smad3 , Factor de Crecimiento Transformador beta/metabolismo , Animales , Embrión de Mamíferos , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Estructura Terciaria de Proteína , Proteína Smad2/química , Proteína Smad2/metabolismo , Proteína smad3/química , Proteína smad3/metabolismo
19.
Cell ; 147(7): 1511-24, 2011 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-22196728

RESUMEN

Specific chromatin marks keep master regulators of differentiation silent yet poised for activation by extracellular signals. We report that nodal TGF-ß signals use the poised histone mark H3K9me3 to trigger differentiation of mammalian embryonic stem cells. Nodal receptors induce the formation of companion Smad4-Smad2/3 and TRIM33-Smad2/3 complexes. The PHD-Bromo cassette of TRIM33 facilitates binding of TRIM33-Smad2/3 to H3K9me3 and H3K18ac on the promoters of mesendoderm regulators Gsc and Mixl1. The crystal structure of this cassette, bound to histone H3 peptides, illustrates that PHD recognizes K9me3, and Bromo binds an adjacent K18ac. The interaction between TRIM33-Smad2/3 and H3K9me3 displaces the chromatin-compacting factor HP1γ, making nodal response elements accessible to Smad4-Smad2/3 for Pol II recruitment. In turn, Smad4 increases K18 acetylation to augment TRIM33-Smad2/3 binding. Thus, nodal effectors use the H3K9me3 mark as a platform to switch master regulators of stem cell differentiation from the poised to the active state.


Asunto(s)
Ensamble y Desensamble de Cromatina , Células Madre Embrionarias/metabolismo , Histonas/metabolismo , Proteínas Smad/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Proteína Goosecoide/genética , Proteínas de Homeodominio/genética , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Alineación de Secuencia
20.
Nature ; 578(7793): E11, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31937917

RESUMEN

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA