Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Semin Cancer Biol ; 96: 100-114, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37852342

RESUMO

Epithelial-to-mesenchymal transition or transformation (EMT) is a cell shape-changing process that is utilized repeatedly throughout embryogenesis and is critical to the attainment of a precise body plan. In the adult, EMT is observed under both normal and pathological conditions, such as during normal wounding healing, during development of certain fibrotic states and vascular anomalies, as well as in some cancers when malignant cells progress to become more aggressive, invasive, and metastatic. Epithelia derived from any of the three embryonic germ layers can undergo EMT, including those derived from mesoderm, such as endothelial cells (sometimes termed Endo-MT) and those derived from endoderm such as fetal liver stroma. At the cellular level, EMT is defined as the transformation of epithelial cells towards a mesenchymal phenotype and is marked by attenuation of expression of epithelial markers and de novo expression of mesenchymal markers. This process is induced by extracellular factors and can be reversible, resulting in mesenchymal-to-epithelial transformation (MET). It is now clear that a cell can simultaneously express properties of both epithelia and mesenchyme, and that such transitional cell-types drive tumor cell heterogeneity, an important aspect of cancer progression, development of a stem-like cell state, and drug resistance. Here we review some of the earliest studies demonstrating the existence of EMT during embryogenesis and discuss the discovery of the extracellular factors and intracellular signaling pathways that contribute to this process, with components of the TGFß signaling superfamily playing a prominent role. We mention early controversies surrounding in vivo EMT during embryonic development and in adult diseased states, and the maturation of the field to a stage wherein targeting EMT to control disease states is an aspirational goal.


Assuntos
Transição Epitelial-Mesenquimal , Neoplasias , Gravidez , Feminino , Adulto , Humanos , Transição Epitelial-Mesenquimal/genética , Células Endoteliais , Células Epiteliais/patologia , Neoplasias/patologia , Desenvolvimento Embrionário/genética
2.
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
3.
PLoS Biol ; 13(12): e1002325, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26680585

RESUMO

Epithelial-mesenchymal transition (EMT) is a normal cell differentiation event during development and contributes pathologically to carcinoma and fibrosis progression. EMT often associates with increased transforming growth factor-ß (TGF-ß) signaling, and TGF-ß drives EMT, in part through Smad-mediated reprogramming of gene expression. TGF-ß also activates the Erk MAPK pathway through recruitment and Tyr phosphorylation of the adaptor protein ShcA by the activated TGF-ß type I receptor. We found that ShcA protects the epithelial integrity of nontransformed cells against EMT by repressing TGF-ß-induced, Smad-mediated gene expression. p52ShcA competed with Smad3 for TGF-ß receptor binding, and down-regulation of ShcA expression enhanced autocrine TGF-ß/Smad signaling and target gene expression, whereas increased p52ShcA expression resulted in decreased Smad3 binding to the TGF-ß receptor, decreased Smad3 activation, and increased Erk MAPK and Akt signaling. Furthermore, p52ShcA sequestered TGF-ß receptor complexes to caveolin-associated membrane compartments, and reducing ShcA expression enhanced the receptor localization in clathrin-associated membrane compartments that enable Smad activation. Consequently, silencing ShcA expression induced EMT, with increased cell migration, invasion, and dissemination, and increased stem cell generation and mammosphere formation, dependent upon autocrine TGF-ß signaling. These findings position ShcA as a determinant of the epithelial phenotype by repressing TGF-ß-induced Smad activation through differential partitioning of receptor complexes at the cell surface.


Assuntos
Transição Epitelial-Mesenquimal , Queratinócitos/metabolismo , Glândulas Mamárias Animais/metabolismo , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Proteína Smad3/agonistas , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular , Membrana Celular/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Queratinócitos/citologia , Queratinócitos/patologia , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/patologia , Camundongos , Fosforilação , Processamento de Proteína Pós-Traducional , Transporte Proteico , Interferência de RNA , Proteínas Adaptadoras da Sinalização Shc/antagonistas & inibidores , Proteínas Adaptadoras da Sinalização Shc/genética , Proteína Smad2/agonistas , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
4.
Proc Natl Acad Sci U S A ; 111(21): 7723-8, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24812125

RESUMO

Outcome of TGFß1 signaling is context dependent and differs between individuals due to germ-line genetic variation. To explore innate genetic variants that determine differential outcome of reduced TGFß1 signaling, we dissected the modifier locus Tgfbm3, on mouse chromosome 12. On a NIH/OlaHsd genetic background, the Tgfbm3b(C57) haplotype suppresses prenatal lethality of Tgfb1(-/-) embryos and enhances nuclear accumulation of mothers against decapentaplegic homolog 2 (Smad2) in embryonic cells. Amino acid polymorphisms within a disintegrin and metalloprotease 17 (Adam17) can account, at least in part, for this Tgfbm3b effect. ADAM17 is known to down-regulate Smad2 signaling by shedding the extracellular domain of TGFßRI, and we show that the C57 variant is hypomorphic for down-regulation of Smad2/3-driven transcription. Genetic variation at Tgfbm3 or pharmacological inhibition of ADAM17, modulates postnatal circulating endothelial progenitor cell (CEPC) numbers via effects on TGFßRI activity. Because CEPC numbers correlate with angiogenic potential, this suggests that variant Adam17 is an innate modifier of adult angiogenesis, acting through TGFßR1. To determine whether human ADAM17 is also polymorphic and interacts with TGFß signaling in human vascular disease, we investigated hereditary hemorrhagic telangiectasia (HHT), which is caused by mutations in TGFß/bone morphogenetic protein receptor genes, ENG, encoding endoglin (HHT1), or ACVRL1 encoding ALK1 (HHT2), and considered a disease of excessive abnormal angiogenesis. HHT manifests highly variable incidence and severity of clinical features, ranging from small mucocutaneous telangiectases to life-threatening visceral and cerebral arteriovenous malformations (AVMs). We show that ADAM17 SNPs associate with the presence of pulmonary AVM in HHT1 but not HHT2, indicating genetic variation in ADAM17 can potentiate a TGFß-regulated vascular disease.


Assuntos
Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Vasos Sanguíneos/patologia , Regulação da Expressão Gênica/fisiologia , Variação Genética , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Proteína ADAM17 , Animais , Regulação da Expressão Gênica/genética , Humanos , Imuno-Histoquímica , Luciferases , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Transdução de Sinais/genética , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta1/genética
5.
Proc Natl Acad Sci U S A ; 109(44): 18042-7, 2012 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-23064636

RESUMO

TGFß activation and signaling have been extensively studied in experimental models of allergen-induced asthma as potential therapeutic targets during chronic or acute phases of the disease. Outcomes of experimental manipulation of TGFß activity have been variable, in part due to use of different model systems. Using an ovalbumin (OVA)-induced mouse model of asthma, we here show that innate variation within TGFß1 genetic modifier loci, Tgfbm2 and Tgfbm3, alters disease susceptibility. Specifically, Tgfbm2(129) and Tgfbm3(C57) synergize to reverse accentuated airway hyperresponsiveness (AHR) caused by low TGFß1 levels in Tgfb1(+/-) mice of the NIH/OlaHsd strain. Moreover, epistatic interaction between Tgfbm2(129) and Tgfbm3(C57) uncouples the inflammatory response to ovalbumin from those of airway remodeling and airway hyperresponsiveness, illustrating independent genetic control of these responses. We conclude that differential inheritance of genetic variants of Tgfbm genes alters biological responses to reduced TGFß1 signaling in an experimental asthma model. TGFß antagonists for treatment of lung diseases might therefore give diverse outcomes, dependent on genetic variation.


Assuntos
Asma/genética , Epistasia Genética , Fator de Crescimento Transformador beta1/genética , Animais , Predisposição Genética para Doença , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
6.
J Cell Sci ; 125(Pt 5): 1259-73, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22399812

RESUMO

In cancer progression, carcinoma cells gain invasive behavior through a loss of epithelial characteristics and acquisition of mesenchymal properties, a process that can lead to epithelial-mesenchymal transition (EMT). TGF-ß is a potent inducer of EMT, and increased TGF-ß signaling in cancer cells is thought to drive cancer-associated EMT. Here, we examine the physiological requirement for mTOR complex 2 (mTORC2) in cells undergoing EMT. TGF-ß rapidly induces mTORC2 kinase activity in cells undergoing EMT, and controls epithelial cell progression through EMT. By regulating EMT-associated cytoskeletal changes and gene expression, mTORC2 is required for cell migration and invasion. Furthermore, inactivation of mTORC2 prevents cancer cell dissemination in vivo. Our results suggest that the mTORC2 pathway is an essential downstream branch of TGF-ß signaling, and represents a responsive target to inhibit EMT and prevent cancer cell invasion and metastasis.


Assuntos
Proteínas de Transporte/metabolismo , Transição Epitelial-Mesenquimal , Serina-Treonina Quinases TOR/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Movimento Celular , Progressão da Doença , Células Epiteliais/metabolismo , Metaloproteinase 9 da Matriz/biossíntese , Camundongos , Invasividade Neoplásica , Neoplasias/metabolismo , Neoplasias/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Interferente Pequeno , Proteína Companheira de mTOR Insensível à Rapamicina , Transdução de Sinais/fisiologia , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Proteína rhoA de Ligação ao GTP/metabolismo
7.
Nat Cell Biol ; 9(9): 1000-4, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17762890

RESUMO

During development, stem and progenitor cells gradually commit to differentiation pathways. Cell fate decisions are regulated by differentiation factors, which activate transcription programmes that specify lineage and differentiation status. Among these factors, the transforming growth factor (TGF)-beta family is important in both lineage selection and progression of differentiation of most, if not all, cell and tissue types. There is now increasing evidence that TGF-beta family proteins have the ability to redirect the differentiation of cells that either have fully differentiated or have engaged in differentiation along a particular lineage, and can thereby elicit 'transdifferentiation'. This capacity for cellular plasticity is critical for normal embryonic development, but when recapitulated in the adult it can give rise to, or contribute to, a variety of diseases. This is illustrated by the ability of TGF-beta family members to redirect epithelial cells into mesenchymal differentiation and to cause switching of mesenchymal cells from one lineage to another. Hence, various pathologies in adults may be considered diseases of abnormal development and differentiation.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem da Célula , Epitélio/crescimento & desenvolvimento , Fibrose/metabolismo , Humanos , Mesoderma/citologia , Mesoderma/crescimento & desenvolvimento , Morfogênese , Metástase Neoplásica , Fator de Crescimento Transformador beta/genética
8.
Science ; 384(6699): eadi7453, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38815020

RESUMO

Stem cells play a critical role in cancer development by contributing to cell heterogeneity, lineage plasticity, and drug resistance. We created gene expression networks from hundreds of mouse tissue samples (both normal and tumor) and integrated these with lineage tracing and single-cell RNA-seq, to identify convergence of cell states in premalignant tumor cells expressing markers of lineage plasticity and drug resistance. Two of these cell states representing multilineage plasticity or proliferation were inversely correlated, suggesting a mutually exclusive relationship. Treatment of carcinomas in vivo with chemotherapy repressed the proliferative state and activated multilineage plasticity whereas inhibition of differentiation repressed plasticity and potentiated responses to cell cycle inhibitors. Manipulation of this cell state transition point may provide a source of potential combinatorial targets for cancer therapy.


Assuntos
Carcinoma de Células Escamosas , Linhagem da Célula , Células-Tronco Neoplásicas , Neoplasias Cutâneas , Animais , Camundongos , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Células-Tronco Neoplásicas/patologia , Análise de Célula Única , Diferenciação Celular , Resistencia a Medicamentos Antineoplásicos/genética , Plasticidade Celular , Proliferação de Células , Redes Reguladoras de Genes , RNA-Seq , Regulação Neoplásica da Expressão Gênica
9.
J Neurosci ; 32(4): 1197-206, 2012 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-22279205

RESUMO

Deletions of the genes encoding the integrin αVß8 (Itgav, Itgb8) have been shown to result in abnormal vascular development in the CNS, including prenatal and perinatal hemorrhage. Other work has indicated that a major function of this integrin in vivo is to promote TGFß activation. In this paper, we show that Itgb8 mRNA is strongly expressed in murine Müller glia and retinal ganglion cells, but not astrocytes. We further show that Itgb8 deletion in the entire retina severely perturbs development of the murine retinal vasculature, elevating vascular branch point density and vascular coverage in the superficial vascular plexus, while severely impairing formation of the deep vascular plexus. The stability of the mutant vasculature is also impaired as assessed by the presence of hemorrhage and vascular basal lamina sleeves lacking endothelial cells. Specific deletion of Itgb8 in Müller glia and neurons, but not deletion in astrocytes, recapitulates the phenotype observed following Itgb8 in the entire retina. Consistent with αVß8's role in TGFß1 activation, we show that retinal deletion of Tgfb1 results in very similar retinal vascular abnormalities. The vascular deficits appear to reflect impaired TGFß signaling in vascular endothelial cells because retinal deletion of Itgb8 reduces phospho-SMAD3 in endothelial cells and endothelial cell-specific deletion of the TGFßRII gene recapitulates the major deficits observed in the Itgb8 and TGFß1 mutants. Of special interest, the retinal vascular phenotypes observed in each mutant are remarkably similar to those of others following inhibition of neuropilin-1, a receptor previously implicated in TGFß activation and signaling.


Assuntos
Diferenciação Celular , Células Endoteliais/patologia , Integrinas/fisiologia , Vasos Retinianos/crescimento & desenvolvimento , Vasos Retinianos/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Diferenciação Celular/fisiologia , Células Endoteliais/fisiologia , Feminino , Integrinas/antagonistas & inibidores , Integrinas/deficiência , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Camundongos Transgênicos , Vasos Retinianos/patologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores
10.
bioRxiv ; 2023 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-37215032

RESUMO

Adult mammalian stem cells play critical roles in normal tissue homeostasis, as well as in tumor development, by contributing to cell heterogeneity, plasticity, and development of drug resistance. The relationship between different types of normal and cancer stem cells is highly controversial and poorly understood. Here, we carried out gene expression network analysis of normal and tumor samples from genetically heterogeneous mice to create network metagenes for visualization of stem-cell networks, rather than individual stem-cell markers, at the single-cell level during multistage carcinogenesis. We combined this approach with lineage tracing and single-cell RNASeq of stem cells and their progeny, identifying a previously unrecognized hierarchy in which Lgr6+ stem cells from tumors generate progeny that express a range of other stem-cell markers including Sox2, Pitx1, Foxa1, Klf5, and Cd44. Our data identify a convergence of multiple stem-cell and tumor-suppressor pathways in benign tumor cells expressing markers of lineage plasticity and oxidative stress. This same single-cell population expresses network metagenes corresponding to markers of cancer drug resistance in human tumors of the skin, lung and prostate. Treatment of mouse squamous carcinomas in vivo with the chemotherapeutic cis-platin resulted in elevated expression of the genes that mark this cell population. Our data have allowed us to create a simplified model of multistage carcinogenesis that identifies distinct stem-cell states at different stages of tumor progression, thereby identifying networks involved in lineage plasticity, drug resistance, and immune surveillance, providing a rich source of potential targets for cancer therapy.

11.
Annu Rev Cancer Biol ; 6(1): 123-146, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36382146

RESUMO

Discovered over four decades ago, transforming growth factor ß (TGFß) is a potent pleiotropic cytokine that has context-dependent effects on most cell types. It acts as a tumor suppressor in some cancers and/or supports tumor progression and metastasis through its effects on the tumor stroma and immune microenvironment. In TGFß-responsive tumors it can promote invasion and metastasis through epithelial-mesenchymal transformation, the appearance of cancer stem cell features, and resistance to many drug classes, including checkpoint blockade immunotherapies. Here we consider the biological activities of TGFß action on different cells of relevance toward improving immunotherapy outcomes for patients, with a focus on the adaptive immune system. We discuss recent advances in the development of drugs that target the TGFß signaling pathway in a tumor-specific or cell type-specific manner to improve the therapeutic window between response rates and adverse effects.

12.
Nat Cell Biol ; 4(7): 487-94, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12105419

RESUMO

Metastasis is a multistep process that involves local tumour invasion followed by dissemination to, and re-establishment at, distant sites. Here we show that during multistage tumorigenesis, discrete expression thresholds of activated Smad2 and H-ras are sequentially surpassed, driving tumour progression through distinct phases from a differentiated squamous carcinoma to a motile invasive stage, followed by an overt change from epithelial to mesenchymal cell type, finally culminating in metastatic tumour spread. Smad2 activation alone induces migration of tumour cells. Elevated H-ras levels, however, are required for nuclear accumulation of Smad2, both of which are essential for the epithelial mesenchymal transition (EMT). Having undergone EMT, fibroblastoid carcinoma cells with elevated levels of activated Smad2, gain the capability to spread to a wide variety of tissues by a further increase in Smad2 expression. These findings have far-reaching implications for the prevention of tumour growth, invasion and metastasis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Genes ras , Invasividade Neoplásica/fisiopatologia , Metástase Neoplásica/fisiopatologia , Transativadores/metabolismo , Animais , Carcinoma de Células Escamosas/fisiopatologia , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteína Smad2
14.
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
15.
Cell Rep ; 36(1): 109309, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34233193

RESUMO

αvß8 integrin, a key activator of transforming growth factor ß (TGF-ß), inhibits anti-tumor immunity. We show that a potent blocking monoclonal antibody against αvß8 (ADWA-11) causes growth suppression or complete regression in syngeneic models of squamous cell carcinoma, mammary cancer, colon cancer, and prostate cancer, especially when combined with other immunomodulators or radiotherapy. αvß8 is expressed at the highest levels in CD4+CD25+ T cells in tumors, and specific deletion of ß8 from T cells is as effective as ADWA-11 in suppressing tumor growth. ADWA-11 increases expression of a suite of genes in tumor-infiltrating CD8+ T cells normally inhibited by TGF-ß and involved in tumor cell killing, including granzyme B and interferon-γ. The in vitro cytotoxic effect of tumor CD8 T cells is inhibited by CD4+CD25+ cells, and this suppressive effect is blocked by ADWA-11. These findings solidify αvß8 integrin as a promising target for cancer immunotherapy.


Assuntos
Imunidade , Imunoterapia , Integrinas/metabolismo , Modelos Biológicos , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T/imunologia , Animais , Anticorpos Antineoplásicos/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígeno CTLA-4/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Granzimas/metabolismo , Interferon gama/metabolismo , Depleção Linfocítica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Neoplasias/genética , Neoplasias/patologia , Transdução de Sinais , Proteína Smad3/metabolismo , Análise de Sobrevida , Linfócitos T Citotóxicos/imunologia , Fator de Crescimento Transformador beta/metabolismo , Microambiente Tumoral/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
17.
Breast Cancer Res ; 11(1): 202, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19291273

RESUMO

The contribution of transforming growth factor (TGF)beta to breast cancer has been studied from a myriad perspectives since seminal studies more than two decades ago. Although the action of TGFbeta as a canonical tumor suppressor in breast is without a doubt, there is compelling evidence that TGFbeta is frequently subverted in a malignant plexus that drives breast cancer. New knowledge that TGFbeta regulates the DNA damage response, which underlies cancer therapy, reveals another facet of TGFbeta biology that impedes cancer control. Too much TGFbeta, too late in cancer progression is the fundamental motivation for pharmaceutical inhibition.


Assuntos
Neoplasias/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Humanos , Transdução de Sinais
18.
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.

19.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA