Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Proc Natl Acad Sci U S A ; 109(32): 13016-21, 2012 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-22826236

RESUMO

Unique insights for the reprograming of cell lineages have come from embryonic development in the ascidian Ciona, which is dependent upon the transcription factors Ci-ets1/2 and Ci-mesp to generate cardiac progenitors. We tested the idea that mammalian v-ets erythroblastosis virus E26 oncogene homolog 2 (ETS2) and mesoderm posterior (MESP) homolog may be used to convert human dermal fibroblasts into cardiac progenitors. Here we show that murine ETS2 has a critical role in directing cardiac progenitors during cardiopoiesis in embryonic stem cells. We then use lentivirus-mediated forced expression of human ETS2 to convert normal human dermal fibroblasts into replicative cells expressing the cardiac mesoderm marker KDR(+). However, although neither ETS2 nor the purported cardiac master regulator MESP1 can by themselves generate cardiac progenitors de novo from fibroblasts, forced coexpression of ETS2 and MESP1 or cell treatment with purified proteins reprograms fibroblasts into cardiac progenitors, as shown by the de novo appearance of core cardiac transcription factors, Ca(2+) transients, and sarcomeres. Our data indicate that ETS2 and MESP1 play important roles in a genetic network that governs cardiopoiesis.


Assuntos
Transdiferenciação Celular/fisiologia , Fibroblastos/citologia , Mioblastos Cardíacos/citologia , Proteína Proto-Oncogênica c-ets-2/metabolismo , Pele/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Western Blotting , Transdiferenciação Celular/genética , Citometria de Fluxo , Imunofluorescência , Técnicas de Inativação de Genes , Humanos , Camundongos , Mioblastos Cardíacos/fisiologia , Reação em Cadeia da Polimerase , Proteína Proto-Oncogênica c-ets-2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
Stem Cells ; 31(1): 12-22, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22961723

RESUMO

The differentiation of stem-like tumor cells may contribute to the cellular heterogeneity of breast cancers. We report the propagation of highly enriched mouse mammary cancer stem cells that retain the potential to differentiate both in vivo and in culture and their use to identify chemical compounds that influence both self-renewal and differentiation. We identify epithelial tumor-initiating cells (ETICs) that express lineage markers of both basal and luminal mammary cell lineages and retain the potential, from even single cells, to generate heterogeneous tumors similar to the tumor of origin. ETICs can progress through a Rho-associated coiled-coil containing protein kinase 1 dependent, epithelial to mesenchymal transition to generate mesenchymal tumor-initiating cells capable of initiating tumors of limited heterogeneity. The propagation of ETICs may allow for the identification of new therapeutic compounds that may inhibit or prevent progression of some types of breast cancer.


Assuntos
Neoplasias Mamárias Animais/metabolismo , Células-Tronco Neoplásicas/metabolismo , Quinases Associadas a rho/antagonistas & inibidores , Animais , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Transição Epitelial-Mesenquimal , Feminino , Perfilação da Expressão Gênica , Glândulas Mamárias Animais/patologia , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Camundongos Transgênicos , Interferência de RNA , RNA Interferente Pequeno , Células Tumorais Cultivadas , Proteína Wnt1/metabolismo , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
3.
ACS Pharmacol Transl Sci ; 6(7): 1028-1042, 2023 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-37470014

RESUMO

Triple-negative breast cancer (TNBC) remains a disease with a paucity of targeted treatment opportunities. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that is involved in a wide range of physiological processes, including the sensing of xenobiotics, immune function, development, and differentiation. Different small-molecule AhR ligands drive strikingly varied cellular and organismal responses. In certain cancers, AhR activation by select small molecules induces cell cycle arrest or apoptosis via activation of tumor-suppressive transcriptional programs. AhR is expressed in triple-negative breast cancers, presenting a tractable therapeutic opportunity. Here, we identify a novel ligand of the aryl hydrocarbon receptor that potently and selectively induces cell death in triple-negative breast cancer cells and TNBC stem cells via the AhR. Importantly, we found that this compound, Analog 523, exhibits minimal cytotoxicity against multiple normal human primary cells. Analog 523 represents a high-affinity AhR ligand with potential for future clinical translation as an anticancer agent.

4.
Stem Cells ; 29(3): 430-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21425406

RESUMO

Ets2 has both tumor repressive and supportive functions for different types of cancer. We have investigated the role of Ets2 within intestinal epithelial cells in postnatal mouse colon development and tumorigenesis. Conditional inactivation of Ets2 within intestinal epithelial cells results in over representation of Ets2-deficient colon crypts within young and adult animals. This preferential representation is associated with an increased number of proliferative cells within the stem cell region and an increased rate of crypt fission in young mice that result in larger patches of Ets2-deficient crypts. These effects are consistent with a selective advantage of Ets2-deficient intestinal stem cells in colonizing colonic crypts and driving crypt fission. Ets2-deficient colon crypts have an increased mucosal thickness, an increased number of goblet cells, and an increased density. Mice with Ets2-deficient intestinal cells develop more colon tumors in response to treatment with azoxymethane and dextran sulfate sodium. The selective population of colon crypts, the altered differentiation state and increased sensitivity to carcinogen-induced tumors all indicate that Ets2 deficiency alters colon stem cell number or behavior. Ets2-dependent, epithelial cell-autonomous repression of intestinal tumors may contribute to protection from colon cancer of persons with increased dosage of chromosome 21.


Assuntos
Adenoma/genética , Células-Tronco Adultas/patologia , Transformação Celular Neoplásica/genética , Colo/citologia , Neoplasias do Colo/genética , Proteína Proto-Oncogênica c-ets-2/fisiologia , Adenoma/patologia , Células-Tronco Adultas/metabolismo , Animais , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Colo/metabolismo , Colo/patologia , Neoplasias do Colo/patologia , Regulação para Baixo/genética , Regulação para Baixo/fisiologia , Feminino , Predisposição Genética para Doença , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos/genética , Proteína Proto-Oncogênica c-ets-2/genética , Proteína Proto-Oncogênica c-ets-2/metabolismo
5.
J Biol Chem ; 285(8): 5836-47, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-19955175

RESUMO

The transcription factor FoxN1 is essential for differentiation of thymic epithelial cell (TEC) progenitors during thymic organogenesis. However, limited information is available on the postnatal contribution of FoxN1 to thymic maintenance. To address this question, we generated a loxP-floxed FoxN1 (fx) mouse with three different promoter-driven inducible CreER(T) transgenes. Postnatal ubiquitous deletion of FoxN1 caused dramatic thymic atrophy in 5 days and more severe deterioration in medullary TECs (mTECs) than in cortical TECs (cTECs). Induction of FoxN1 deletion selectively in K5 promoter-driven somatic epithelial cells (mostly mTECs and possibly some adult epithelial stem cells) was sufficient to cause significant thymic atrophy, whereas FoxN1 deletion in K18 promoter-driven somatic epithelial cells (mostly cTECs) was not. Thymic atrophy resulted from increased apoptosis and was associated with activation of the p53 gene in mature mTECs. Although FoxN1 is required for the development of both mTECs and cTECs in thymic organogenesis, it is most important for the maintenance of mTECs in the postnatal thymus, which are in turn necessary to prevent thymic atrophy.


Assuntos
Células Epiteliais/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regiões Promotoras Genéticas , Células-Tronco/metabolismo , Timo/crescimento & desenvolvimento , Proteína Supressora de Tumor p53/biossíntese , Animais , Apoptose , Atrofia , Células Epiteliais/patologia , Fatores de Transcrição Forkhead/genética , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Transgênicos , Células-Tronco/patologia , Timo/patologia , Fatores de Tempo , Transgenes , Proteína Supressora de Tumor p53/genética
6.
Blood ; 114(5): 1123-30, 2009 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-19411629

RESUMO

The ras/Raf/Mek/Erk pathway plays a central role in coordinating endothelial cell activities during angiogenesis. Transcription factors Ets1 and Ets2 are targets of ras/Erk signaling pathways that have been implicated in endothelial cell function in vitro, but their precise role in vascular formation and function in vivo remains ill-defined. In this work, mutation of both Ets1 and Ets2 resulted in embryonic lethality at midgestation, with striking defects in vascular branching having been observed. The action of these factors was endothelial cell autonomous as demonstrated using Cre/loxP technology. Analysis of Ets1/Ets2 target genes in isolated embryonic endothelial cells demonstrated down-regulation of Mmp9, Bcl-X(L), and cIAP2 in double mutants versus controls, and chromatin immunoprecipitation revealed that both Ets1 and Ets2 were loaded at target promoters. Consistent with these observations, endothelial cell apoptosis was significantly increased both in vivo and in vitro when both Ets1 and Ets2 were mutated. These results establish essential and overlapping functions for Ets1 and Ets2 in coordinating endothelial cell functions with survival during embryonic angiogenesis.


Assuntos
Apoptose/genética , Desenvolvimento Embrionário/genética , Células Endoteliais/citologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neovascularização Fisiológica/genética , Proteína Proto-Oncogênica c-ets-1/fisiologia , Proteína Proto-Oncogênica c-ets-2/fisiologia , Animais , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/ultraestrutura , Sobrevivência Celular/genética , Quimera , Edema/embriologia , Edema/genética , Transferência Embrionária , Morte Fetal/genética , Morte Fetal/patologia , Doenças Fetais/genética , Doenças Fetais/patologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Hemorragia/embriologia , Hemorragia/genética , Homozigoto , Camundongos , Camundongos Knockout , Fenótipo , Proteína Proto-Oncogênica c-ets-1/deficiência , Proteína Proto-Oncogênica c-ets-1/genética , Proteína Proto-Oncogênica c-ets-2/deficiência , Proteína Proto-Oncogênica c-ets-2/genética
7.
J Cell Biol ; 171(6): 931-7, 2005 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-16365160

RESUMO

Keratins 8 and 18 (K8/18) are major constituents of Mallory bodies (MBs), which are hepatocyte cytoplasmic inclusions seen in several liver diseases. K18-null but not K8-null or heterozygous mice form MBs, which indicates that K8 is important for MB formation. Early stages in MB genesis include K8/18 hyperphosphorylation and overexpression. We used transgenic mice that overexpress K8, K18, or K8/18 to test the importance of K8 and/or K18 in MB formation. MBs were induced by feeding 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC). Livers of young K8 or K8/K18 overexpressors had no histological abnormalities despite increased keratin protein and phosphorylation. In aging mice, only K8-overexpressing livers spontaneously developed small "pre-MB" aggregates. Only K8-overexpressing young mice are highly susceptible to MB formation after short-term DDC feeding. Thus, the K8 to K18 ratio, rather than K8/18 overexpression by itself, plays an essential role in MB formation. K8 overexpression is sufficient to form pre-MB and primes animals to accumulate MBs upon DDC challenge, which may help explain MB formation in human liver diseases.


Assuntos
Regulação da Expressão Gênica/fisiologia , Hepatócitos/ultraestrutura , Corpos de Inclusão/ultraestrutura , Queratinas/metabolismo , Proteínas/metabolismo , Animais , Humanos , Queratina-18 , Queratina-8 , Fígado/enzimologia , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Modelos Biológicos , RNA Mensageiro/metabolismo
8.
Mol Cell Biol ; 27(14): 5201-13, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17502352

RESUMO

Extraembryonic ectoderm differentiation and chorioallantoic attachment are fibroblast growth factor (FGF)- and transforming growth factor beta-regulated processes that are the first steps in the development of the placenta labyrinth and the establishment of the fetal-maternal circulation in the developing embryo. Only a small number of genes have been demonstrated to be important in trophoblast stem cell differentiation. Erf is a ubiquitously expressed Erk-regulated, ets domain transcriptional repressor expressed throughout embryonic development and adulthood. However, in the developing placenta, after 7.5 days postcoitum (dpc) its expression is restricted to the extraembryonic ectoderm, and its expression is restricted after 9.5 dpc in a subpopulation of labyrinth cells. Homozygous deletion of Erf in mice leads to a block of chorionic cell differentiation before chorioallantoic attachment, resulting in a persisting chorion layer, a persisting ectoplacental cone cavity, failure of chorioallantoic attachment, and absence of labyrinth. These defects result in embryo death by 10.5 dpc. Trophoblast stem cell lines derived from Erf(dl1/dl1) knockout blastocysts exhibit delayed differentiation and decreased expression of spongiotrophoblast markers, consistent with the persisting chorion layer, the expanded giant cell layer, and the diminished spongiotrophoblast layer observed in vivo. Our data suggest that attenuation of FGF/Erk signaling and consecutive Erf nuclear localization and function is required for extraembryonic ectoderm differentiation, ectoplacental cone cavity closure, and chorioallantoic attachment.


Assuntos
Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Ectoderma/citologia , Proteínas Repressoras/metabolismo , Animais , Membrana Corioalantoide/citologia , Cruzamentos Genéticos , Proteínas de Ligação a DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Genótipo , Masculino , Camundongos , Camundongos Mutantes , Modelos Biológicos , Neuropeptídeos/metabolismo , Fenótipo , Placenta/anormalidades , Placenta/embriologia , Placenta/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Células-Tronco/citologia , Fatores de Transcrição
9.
Nat Biotechnol ; 25(2): 233-7, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17220877

RESUMO

Placental dysfunction underlies many complications during pregnancy, and better understanding of gene function during placentation could have considerable clinical relevance. However, the lack of a facile method for placenta-specific gene manipulation has hampered investigation of placental organogenesis and the treatment of placental dysfunction. We showed previously that transduction of fertilized mouse eggs with lentiviral vectors leads to transgene expression in both the fetus and the placenta. Here we report placenta-specific gene incorporation by lentiviral transduction of mouse blastocysts after removal of the zona pellucida. All of the placentas analyzed, but none of the fetuses, were transgenic. Application of this method substantially rescued mice deficient in Ets2, Mapk14 (also known as p38alpha) and Mapk1 (also known as Erk2) from embryonic lethality caused by placental defects. Ectopic expression of Mapk11 also complemented Mapk14 deficiency during placentation.


Assuntos
Embrião de Mamíferos/fisiologia , Lentivirus/genética , Camundongos Transgênicos/genética , Placenta/fisiologia , Placenta/virologia , Transdução Genética/métodos , Trofoblastos/virologia , Animais , Feminino , Vetores Genéticos/genética , Camundongos , Doenças Placentárias/genética , Gravidez , Análise de Sobrevida
10.
Genesis ; 47(12): 793-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19830817

RESUMO

Transgenic and knockout studies have advanced our understanding of the genetic control of embryonic development over the past decades. However, interpretation of the phenotype of mutant mice is potentially complicated, since the commonly used knockout approach modifies both the fetal and placental genome. To circumvent this problem, we previously developed a placenta-specific gene manipulation system by lentiviral vector transduction of embryos at the blastocyst stage. In the present study, by combination with the Cre/LoxP system, we successfully demonstrate placenta-specific gene activation and inactivation in EGFP reporter mice and Ets2 floxed mice, respectively. Transient expression using integrase-defective lentiviral (IDLV) vectors diminished the toxic effect of Cre expression and solved the dilemma of mosaic recombination with lower concentrations and toxic effects with higher concentrations of Cre recombinase. We also show that placenta-specific Ets2 disruption causes embryonic lethality and reconfirmed the critical role of Ets2 during placentation. This technology facilitates both gain and loss of gene function analyses in placental development during pregnancy. Since IDLV vectors can efficiently transduce a variety of cell types similarly to wild-type vectors, our IDLV-Cre strategy is potentially useful for a wide range of applications.


Assuntos
Integrases/genética , Lentivirus/genética , Placenta/metabolismo , Animais , Sítios de Ligação/genética , Blastocisto/citologia , Blastocisto/metabolismo , Vírus Defeituosos/genética , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Feminino , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Integrases/metabolismo , Lentivirus/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Placenta/citologia , Gravidez , Recombinação Genética , Transfecção/métodos
11.
J Cell Biol ; 161(4): 749-56, 2003 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-12771125

RESUMO

The intermediate filament protein keratin 8 (K8) is critical for the development of most mouse embryos beyond midgestation. We find that 68% of K8-/- embryos, in a sensitive genetic background, are rescued from placental bleeding and subsequent death by cellular complementation with wild-type tetraploid extraembryonic cells. This indicates that the primary defect responsible for K8-/- lethality is trophoblast giant cell layer failure. Furthermore, the genetic absence of maternal but not paternal TNF doubles the number of viable K8-/- embryos. Finally, we show that K8-/- concepti are more sensitive to a TNF-dependent epithelial apoptosis induced by the administration of concanavalin A (ConA) to pregnant mothers. The ConA-induced failure of the trophoblast giant cell barrier results in hematoma formation between the trophoblast giant cell layer and the embryonic yolk sac in a phenocopy of dying K8-deficient concepti in a sensitive genetic background. We conclude the lethality of K8-/- embryos is due to a TNF-sensitive failure of trophoblast giant cell barrier function. The keratin-dependent protection of trophoblast giant cells from a maternal TNF-dependent apoptotic challenge may be a key function of simple epithelial keratins.


Assuntos
Queratinas/metabolismo , Placenta/fisiologia , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Apoptose/efeitos dos fármacos , Concanavalina A/farmacologia , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Feminino , Deleção de Genes , Células Gigantes/efeitos dos fármacos , Células Gigantes/metabolismo , Células Gigantes/patologia , Hematoma/metabolismo , Hematoma/patologia , Queratina-8 , Queratinas/genética , Masculino , Camundongos , Camundongos Knockout , Placenta/efeitos dos fármacos , Gravidez , Receptores do Fator de Necrose Tumoral/deficiência , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Trofoblastos/patologia , Fator de Necrose Tumoral alfa/metabolismo
12.
J Cell Biol ; 158(6): 1051-66, 2002 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-12235123

RESUMO

Apoptosis depends critically on regulated cytoskeletal reorganization events in a cell. We demonstrate that death effector domain containing DNA binding protein (DEDD), a highly conserved and ubiquitous death effector domain containing protein, exists predominantly as mono- or diubiquitinated, and that diubiquitinated DEDD interacts with both the K8/18 intermediate filament network and pro-caspase-3. Early in apoptosis, both cytosolic DEDD and its close homologue DEDD2 formed filaments that colocalized with and depended on K8/18 and active caspase-3. Subsequently, these filamentous structures collapsed into intracellular inclusions that migrated into cytoplasmic blebs and contained DEDD, DEDD2, active caspase-3, and caspase-3-cleaved K18 late in apoptosis. Biochemical studies further confirmed that DEDD coimmunoprecipitated with both K18 and pro-caspase-3, and kinetic analyses placed apoptotic DEDD staining prior to caspase-3 activation and K18 cleavage. In addition, both caspase-3 activation and K18 cleavage was inhibited by expression of DEDDDeltaNLS1-3, a cytosolic form of DEDD that cannot be ubiquitinated. Finally, siRNA mediated DEDD knockdown cells exhibited inhibition of staurosporine-induced DNA degradation. Our data suggest that DEDD represents a novel scaffold protein that directs the effector caspase-3 to certain substrates facilitating their ordered degradation during apoptosis.


Assuntos
Apoptose , Proteínas de Ligação a DNA/fisiologia , Filamentos Intermediários/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/metabolismo , Caspase 3 , Caspases/metabolismo , DNA/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte , Inibidores Enzimáticos/farmacologia , Precursores Enzimáticos/metabolismo , Feminino , Células HeLa , Humanos , Corpos de Inclusão/química , Corpos de Inclusão/fisiologia , Corpos de Inclusão/ultraestrutura , Filamentos Intermediários/ultraestrutura , Células Jurkat , Queratinas/metabolismo , Cinética , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Proteínas Nucleares/metabolismo , RNA Interferente Pequeno , RNA não Traduzido/metabolismo , Estaurosporina/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/metabolismo
13.
Dev Biol ; 312(1): 284-99, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17977525

RESUMO

The Ets2 transcription factor is essential for the development of the mouse placenta and for generating signals for embryonic mesoderm and axis formation. Using a conditional targeted Ets2 allele, we show that Ets2 is essential for trophoblast stem (TS) cells self-renewal. Inactivation of Ets2 results in TS cell slower growth, increased expression of a subset of differentiation-associated genes and decreased expression of several genes implicated in TS self-renewal. Among the direct TS targets of Ets2 is Cdx2, a key master regulator of TS cell state. Thus Ets2 contributes to the regulation of multiple genes important for maintaining the undifferentiated state of TS cells and as candidate signals for embryonic development.


Assuntos
Proteína Proto-Oncogênica c-ets-2/metabolismo , Células-Tronco/citologia , Trofoblastos/citologia , Alelos , Animais , Fator de Transcrição CDX2 , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Colo/metabolismo , Perda do Embrião , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Proteínas de Homeodomínio/genética , Humanos , Integrases/metabolismo , Camundongos , Camundongos Mutantes , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica , Trofoblastos/metabolismo
14.
Oncotarget ; 9(38): 25057-25074, 2018 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-29861853

RESUMO

Di(1H-indol-3-yl)(4-trifluoromethylphenyl)methane (DIM-Ph-4-CF3) is an analog of orphan nuclear receptor 4A1 (NR4A1) ligand cytosporone B. We have synthesized several oxidation products of DIM-Ph-4-CF3, focusing on analogs with electron-withdrawing or donating groups at their phenyl ring 4-positions, and examined their anti-cancer activity and mechanism-of-action. Mesylates (DIM-Ph-4-X+ OMs-s) having CF3, CO2Me and Cl groups were more effective inhibitors of cancer cell viability than their precursors. 19F NMR spectroscopy and differential scanning calorimetry strongly indicated interactions of DIM-Ph-4-CF3+ OMs- with the NR4A1 ligand binding domain, and compound-induced apoptosis of prostate cancer cells was dependent on NR4A1. DIM-Ph-4-CF3+ OMs- showed robust inhibition of LNCaP prostate cancer xenografts with no apparent toxicity. In vitro and in vivo, DIM-Ph-4-CF3+ OMs- activated proapoptotic unfolded protein response (UPR) signaling in prostate cancer cells. Independently of DIM-Ph-4-CF3+ OMs-, the bulk of NR4A1 localized to the cytoplasm in various cancer cell lines, suggesting a cytoplasmic mechanism-of-action of DIM-Ph-4-CF3+ OMs- in UPR induction and cell death. In summary, the data suggest that oxidized analogs of DIM-Ph-4-CF3 possess potent and safe anti-cancer activity which is mediated through UPR signaling downstream of NR4A1 binding.

15.
Gene ; 391(1-2): 161-70, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17270363

RESUMO

SHEP1, BCAR3 and NSP1 are the three members of a family of cytoplasmic proteins involved in cell adhesion/migration and antiestrogen resistance. All three proteins contain an SH2 domain and an exchange factor-like domain that binds both Ras GTPases and the scaffolding protein Cas. SHEP1, BCAR3 and NSP1 mRNAs are widely expressed in tissues, and SHEP1 and BCAR3 have multiple splice variants that differ in their 5' untranslated regions and in some cases the beginning of their coding regions. Interestingly, our data suggest that SHEP1 is highly expressed in blood vessels in mouse breast cancer models. In contrast, BCAR3 and NSP1 are more highly expressed than SHEP1 in breast cancer cells. These expression patterns suggest differential roles for the three genes during breast cancer progression in either the vasculature or the tumor cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Processamento Alternativo , Perfilação da Expressão Gênica , Integrinas/fisiologia , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular , Linhagem Celular Tumoral , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Fatores de Troca do Nucleotídeo Guanina , Humanos , Immunoblotting , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Mol Cell Biol ; 23(23): 8614-25, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14612405

RESUMO

The Ets2 transcription factor is regulated by mitogen-activated protein (MAP) kinase phosphorylation of a single threonine residue. We generated by gene targeting a single codon mutation in Ets2 substituting Ala for the critical Thr-72 phosphorylation site (Ets2A72), to investigate the importance of MAP kinase activation of Ets2 in embryo and tumor development. Ets2(A72/A72) mice are viable and develop normally. However, combining the Ets2A72 allele with a deletion mutant of Ets2 results in lethality at E11.5 and shows that Ets2A72 is a hypomorphic allele. Mammary tumors caused by transgenic polyomavirus middle T antigen, activated Neu(Erbb2), or the combination of Neu and transgenic VEGF (Neu; VEGF-25) were all restricted in Ets2(A72/A72) females. The Ets2(A72/A72) restriction on Neu; VEGF-25 tumor growth was associated with increased p21Cip1 expression. The size of tumors transplanted into fat pads of mice with Ets2 targeted alleles was correlated directly with Ets2 activity and fewer stromal cells expressing matrix metalloproteinase 9 (MMP-9). Decreased MMP-3 and MMP-9 mRNAs were confirmed in Ets2(A72/A72) macrophages. Activation of Ets2 at Thr-72 acts in the stroma, downstream of vascular endothelial growth factor production, in part through the regulation of macrophage proteases to support the progression of Neu- and polyomavirus middle-T-initiated mammary tumors.


Assuntos
Proteínas de Ligação a DNA , Neoplasias Mamárias Experimentais/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras , Transativadores/metabolismo , Fatores de Transcrição , Alelos , Substituição de Aminoácidos , Animais , Sequência de Bases , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , DNA Complementar/genética , Desenvolvimento Embrionário e Fetal , Feminino , Expressão Gênica , Marcação de Genes , Heterozigoto , Macrófagos/metabolismo , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fenótipo , Fosforilação , Gravidez , Proteína Proto-Oncogênica c-ets-2 , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Fator A de Crescimento do Endotélio Vascular/genética
17.
Cancer Res ; 65(12): 5325-36, 2005 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15958580

RESUMO

The ErbB2 (Neu) receptor tyrosine kinase is frequently overexpressed in human breast cancers, and this phenotype correlates with a poor clinical prognosis. We examined the effects of the mammalian target of rapamycin inhibitor, rapamycin, on mammary tumorigenesis in transgenic mice bearing an activated ErbB2 (NeuYD) transgene in the absence or presence of a second transgene encoding vascular endothelial growth factor (VEGF). Treatment of NeuYD or NeuYD x VEGF mice with rapamycin dramatically inhibited tumor growth accompanied by a marked decrease in tumor vascularization. Two key events that may underlie the antitumor activity of rapamycin were decreased expression of ErbB3 and inhibition of hypoxia-inducible factor-1-dependent responses to hypoxic stress. Rapamycin exposure caused only a modest inhibition of the proliferation of tumor-derived cell lines in standard monolayer cultures, but dramatically inhibited the growth of the same cells in three-dimensional cultures, due in part to the induction of apoptotic cell death. These studies underscore the therapeutic potential of mammalian target of rapamycin inhibitors in ErbB2-positive breast cancers and indicate that, relative to monolayer cultures, three-dimensional cell cultures are more predictive in vitro models for studies of the antitumor mechanisms of rapamycin and related compounds.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Receptor ErbB-2/biossíntese , Sirolimo/farmacologia , Animais , Apoptose/efeitos dos fármacos , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/genética , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/biossíntese , Feminino , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Masculino , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Transgênicos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Proteínas Nucleares/biossíntese , Fosforilação , Proteínas Quinases/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-3/biossíntese , Esferoides Celulares , Serina-Treonina Quinases TOR , Fatores de Transcrição/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
18.
Oncogene ; 24(46): 6870-6, 2005 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-16007139

RESUMO

Decreasing the amount of active mouse Ets2 transcription factor by half in mice or use of a MAP kinase insensitive hypomorphic targeted Ets2 allele restricts the appearance of transgenic mammary tumors caused by either Polyoma middle T antigen (PyMT) or activated Neu/ErbB2. In addition, the early growth of transplanted mammary tumors is limited by restricted Ets2 activity of the host. Here we have tested genetically, with the use of a conditional Ets2flox allele and tissue specific Cre recombinase expression, whether Ets2 also functions within tumor cells by inactivating Ets2 within mammary luminal epithelial cells from which transgenic PyMTY315/322F tumors arise. We find that inactivation of Ets2 within tumor cells has no effect on tumor appearance or growth. By contrast, complete inactivation of Ets2 in both epithelial and stromal cells moderates the early hyperplastic phase of tumor development and the time of tumor appearance but does not prevent tumor occurrence and has no detectable effect on tumor growth. Thus, Ets2 supports mammary tumors exclusively through their microenvironment.


Assuntos
Neoplasias Mamárias Experimentais/patologia , Proteína Proto-Oncogênica c-ets-2/fisiologia , Animais , Sequência de Bases , Primers do DNA , Feminino , Genótipo , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteína Proto-Oncogênica c-ets-2/genética , Proteína Proto-Oncogênica c-ets-2/metabolismo
19.
Cancer Res ; 64(1): 169-79, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14729621

RESUMO

The Neu (ErbB2, HER2) member of the epidermal growth factor receptor family is implicated in many human breast cancers. We have tested the importance of increased angiogenic signaling in the NeuYD [mouse mammary tumor virus (MMTV)-Neu(ndl)-YD5] mammary tumor model. Transgenic mice expressing vascular endothelial growth factor (VEGF)(164) from the MMTV promoter were generated. These mice expressed VEGF(164) RNA and protein at 20- to 40-fold higher levels throughout mammary gland development but exhibited normal mammary gland development and function. However, in combination with the NeuYD oncogene, VEGF(164) expression resulted in increased vascularization of hyperplastic mammary epithelium and dramatic acceleration of tumor appearance from 111 to 51 days. Gene expression profiling also indicated that the VEGF-accelerated tumors were substantially more vascularized and less hypoxic. The preferential vascularization of early hyperplastic portions of mammary epithelia in NeuYD;MMTV-VEGF animals was associated with NeuYD RNA expression, disorganization of the tight junctions, and overlapping transgenic VEGF expression. NeuYD;MMTV-VEGF(164) bigenic, tumor-bearing animals resulted in an average of 10 tumor cell colonies/lung lodged within vascular spaces. No similar lung colonies were found in control NeuYD mice with similar tumor burdens. Overall, these results demonstrate the angiogenic restriction of early hyperplastic mammary lesions. They also reinforce in vivo the importance of activated Neu in causing disorganization of mammary luminal epithelial cell junctions and provide support for an invasion-independent mechanism of metastasis.


Assuntos
Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/patologia , Neovascularização Patológica/patologia , Receptor ErbB-2/fisiologia , Animais , Sequência de Bases , Primers do DNA , Progressão da Doença , Vírus do Tumor Mamário do Camundongo , Camundongos , Camundongos Transgênicos , Metástase Neoplásica , Reação em Cadeia da Polimerase , Fator A de Crescimento do Endotélio Vascular/genética
20.
Oncotarget ; 7(50): 83278-83293, 2016 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-27829216

RESUMO

Cancers are heterogeneous by nature. While traditional oncology screens commonly use a single endpoint of cell viability, altering the phenotype of tumor-initiating cells may reveal alternative targets that regulate cellular growth by processes other than apoptosis or cell division. We evaluated the impact of knocking down expression of 420 kinases in bi-lineage triple-negative breast cancer (TNBC) cells that express characteristics of both myoepithelial and luminal cells. Knockdown of ERN1 or ALPK1 induces bi-lineage MDA-MB-468 cells to lose the myoepithelial marker keratin 5 but not the luminal markers keratin 8 and GATA3. In addition, these cells exhibit increased ß-casein production. These changes are associated with decreased proliferation and clonogenicity in spheroid cultures and anchorage-independent growth assays. Confirmation of these assays was completed in vivo, where ERN1- or ALPK1-deficient TNBC cells are less tumorigenic. Finally, treatment with K252a, a kinase inhibitor active on ERN1, similarly impairs anchorage-independent growth of multiple breast cancer cell lines. This study supports the strategy to identify new molecular targets for types of cancer driven by cells that retain some capacity for normal differentiation to a non-tumorigenic phenotype. ERN1 and ALPK1 are potential targets for therapeutic development.


Assuntos
Diferenciação Celular , Endorribonucleases/metabolismo , Células-Tronco Neoplásicas/enzimologia , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Neoplasias de Mama Triplo Negativas/enzimologia , Animais , Antineoplásicos/farmacologia , Carbazóis/farmacologia , Caseínas/genética , Caseínas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Relação Dose-Resposta a Droga , Endorribonucleases/antagonistas & inibidores , Endorribonucleases/genética , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Alcaloides Indólicos/farmacologia , Queratina-5/genética , Queratina-5/metabolismo , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Fenótipo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Transfecção , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Carga Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA