Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 10(68): 7251-7275, 2019 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-31921386

RESUMO

Chloride intracellular channel 4 (CLIC4) is a tumor suppressor implicated in processes including growth arrest, differentiation, and apoptosis. CLIC4 protein expression is diminished in the tumor parenchyma during progression in squamous cell carcinoma (SCC) and other neoplasms, but the underlying mechanisms have not been identified. Data from The Cancer Genome Atlas suggest this is not driven by genomic alterations. However, screening and functional assays identified miR-142-3p as a regulator of CLIC4. CLIC4 and miR-142-3p expression are inversely correlated in head and neck (HN) SCC and cervical SCC, particularly in advanced stage cancers. In situ localization revealed that stromal immune cells, not tumor cells, are the predominant source of miR-142-3p in HNSCC. Furthermore, HNSCC single-cell expression data demonstrated that CLIC4 is lower in tumor epithelial cells than in stromal fibroblasts and endothelial cells. Tumor-specific downregulation of CLIC4 was confirmed in an SCC xenograft model concurrent with immune cell infiltration and miR-142-3p upregulation. These findings provide the first evidence of CLIC4 regulation by miRNA. Furthermore, the distinct localization of CLIC4 and miR-142-3p within the HNSCC tumor milieu highlight the limitations of bulk tumor analysis and provide critical considerations for both future mechanistic studies and use of miR-142-3p as a HNSCC biomarker.

2.
Cancer J ; 24(3): 115-120, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29794535

RESUMO

Inspired by the Cancer Moonshot, a dedicated team of professionals worked with leaders across the cancer ecosystem to look for an opportunity to radically reduce cancer mortality globally by focusing on early cancer detection. After an initial survey of cancer innovation, progress, and pitfalls, the team believed that if new rapid, affordable, and accurate early detection solutions were appropriately brought to market, it would be possible to intervene earlier when cancer is most treatable.An extensive process began, informed by dozens of experts in the cancer ecosystem. The Cancer XPRIZE team designed a prize competition where "the winning team will develop a means to rapidly, accurately, and affordably screen for early cancers where intervention can reduce human suffering."The following outlines the Cancer XPRIZE's experience using a powerful approach-the radical prize design-to catch more cancers in time to make a difference saving lives, dollars, and suffering.


Assuntos
Detecção Precoce de Câncer/métodos , Neoplasias/diagnóstico , Custos e Análise de Custo/métodos , Atenção à Saúde/métodos , Humanos
3.
Sci Signal ; 9(433): ra62, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27330189

RESUMO

The receptor tyrosine kinase MET is abundant in many human squamous cell carcinomas (SCCs), but its functional significance in tumorigenesis is not clear. We found that the incidence of carcinogen-induced skin squamous tumors was substantially increased in transgenic MT-HGF (mouse metallothionein-hepatocyte growth factor) mice, which have increased abundance of the MET ligand HGF. Squamous tumors also erupted spontaneously on the skin of MT-HGF mice that were promoted by wounding or the application of 12-O-tetradecanoylphorbol 13-acetate, an activator of protein kinase C. Carcinogen-initiated tumors had Ras mutations, but spontaneous tumors did not. Cultured keratinocytes from MT-HGF mice and oncogenic RAS-transduced keratinocytes shared phenotypic and biochemical features of initiation that were dependent on autocrine activation of epidermal growth factor receptor (EGFR) through increased synthesis and release of EGFR ligands, which was mediated by the kinase SRC, the pseudoproteases iRhom1 and iRhom2, and the metallopeptidase ADAM17. Pharmacological inhibition of EGFR caused the regression of MT-HGF squamous tumors that developed spontaneously in orthografts of MT-HGF keratinocytes combined with dermal fibroblasts and implanted onto syngeneic mice. The global gene expression profile in MET-transformed keratinocytes was highly concordant with that in RAS-transformed keratinocytes, and a core RAS/MET coexpression network was activated in precancerous and cancerous human skin lesions. Tissue arrays revealed that many human skin SCCs have abundant HGF at both the transcript and protein levels. Thus, through the activation of EGFR, MET activation parallels a RAS pathway to contribute to human and mouse cutaneous cancers.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Receptores ErbB/metabolismo , Queratinócitos/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Animais , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Queratinócitos/patologia , Camundongos , Proteínas Proto-Oncogênicas c-met/genética , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Acetato de Tetradecanoilforbol/toxicidade
4.
J Invest Dermatol ; 133(7): 1731-41, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23364477

RESUMO

Selenoproteins are essential molecules for the mammalian antioxidant network. We previously demonstrated that targeted loss of all selenoproteins in mouse epidermis disrupted skin and hair development, and caused premature death. In the current study, we targeted specific selenoproteins for epidermal deletion to determine whether similar phenotypes developed. Keratinocyte-specific knockout mice lacking either the glutathione peroxidase 4 (GPx4) or thioredoxin reductase 1 (TR1) gene were generated by cre-lox technology using K14-cre. TR1 knockout mice had a normal phenotype in resting skin, whereas GPx4 loss in the epidermis caused epidermal hyperplasia, dermal inflammatory infiltrate, dysmorphic hair follicles, and alopecia in perinatal mice. Unlike epidermal ablation of all selenoproteins, mice ablated for GPx4 recovered after 5 weeks and had a normal life span. GPx1 and TR1 were upregulated in the skin and keratinocytes of GPx4-knockout mice. GPx4 deletion reduces keratinocyte adhesion in culture and increases lipid peroxidation and cyclooxygenase-2 (COX-2) levels in cultured keratinocytes and whole skin. Feeding a COX-2 inhibitor to nursing mothers partially prevents development of the abnormal skin phenotype in knockout pups. These data link the activity of cutaneous GPx4 to the regulation of COX-2 and hair follicle morphogenesis, and provide insight into the function of individual selenoprotein activity in maintaining cutaneous homeostasis.


Assuntos
Inibidores de Ciclo-Oxigenase 2/farmacologia , Ciclo-Oxigenase 2/efeitos dos fármacos , Células Epiteliais/metabolismo , Glutationa Peroxidase/deficiência , Folículo Piloso/crescimento & desenvolvimento , Morfogênese/fisiologia , Pele/metabolismo , Animais , Animais Recém-Nascidos , Adesão Celular/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Células Epiteliais/citologia , Feminino , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Peroxidação de Lipídeos/fisiologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fenótipo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Pele/citologia , Tiorredoxina Redutase 1/deficiência , Tiorredoxina Redutase 1/genética , Tiorredoxina Redutase 1/metabolismo
5.
Am J Pathol ; 181(1): 74-84, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22613027

RESUMO

Cutaneous wound healing is a complex process involving blood clotting, inflammation, migration of keratinocytes, angiogenesis, and, ultimately, tissue remodeling and wound closure. Many of these processes involve transforming growth factor-ß (TGF-ß) signaling, and mice lacking components of the TGF-ß signaling pathway are defective in wound healing. We show herein that CLIC4, an integral component of the TGF-ß pathway, is highly up-regulated in skin wounds. We genetically deleted murine CLIC4 and generated a colony on a C57Bl/6 background. CLIC4(NULL) mice were viable and fertile but had smaller litters than did wild-type mice. After 6 months of age, up to 40% of null mice developed spontaneous skin erosions. Reepithelialization of induced full-thickness skin wounds and superficial corneal wounds was delayed in CLIC4(NULL) mice, resolution of inflammation was delayed, and expression of ß4 integrin and p21 was reduced in lysates of constitutive and wounded CLIC4(NULL) skin. The induced level of phosphorylated Smad2 in response to TGF-ß was reduced in cultured CLIC4(NULL) keratinocytes relative to in wild-type cells, and CLIC4(NULL) keratinocytes migrated slower than did wild-type keratinocytes and did not increase migration in response to TGF-ß. CLIC4(NULL) keratinocytes were also less adherent on plates coated with matrix secreted by wild-type keratinocytes. These results indicate that CLIC4 participates in skin healing and corneal wound reepithelialization through enhancement of epithelial migration by a mechanism that may involve a compromised TGF-ß pathway.


Assuntos
Canais de Cloreto/fisiologia , Lesões da Córnea , Proteínas Mitocondriais/fisiologia , Úlcera Cutânea/fisiopatologia , Pele/lesões , Cicatrização/fisiologia , Animais , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Canais de Cloreto/deficiência , Córnea/patologia , Córnea/fisiologia , Relação Dose-Resposta a Droga , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/fisiologia , Camundongos , Camundongos Knockout , Microscopia Confocal , Proteínas Mitocondriais/deficiência , Proteínas/metabolismo , Transdução de Sinais/fisiologia , Pele/metabolismo , Pele/patologia , Úlcera Cutânea/patologia , Fatores de Tempo , Fator de Crescimento Transformador beta/administração & dosagem , Fator de Crescimento Transformador beta/farmacologia
6.
Carcinogenesis ; 33(5): 986-95, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22387366

RESUMO

Chloride intracellular channel (CLIC) 4 is a member of a redox-regulated, metamorphic multifunctional protein family, first characterized as intracellular chloride channels. Current knowledge indicates that CLICs participate in signaling, cytoskeleton integrity and differentiation functions of multiple tissues. In metabolically stressed skin keratinocytes, cytoplasmic CLIC4 is S-nitrosylated and translocates to the nucleus where it enhances transforming growth factor-ß (TGF-ß) signaling by protecting phospho-Smad 2 and 3 from dephosphorylation. CLIC4 expression is diminished in multiple human epithelial cancers, and the protein is excluded from the nucleus. We now show that CLIC4 expression is reduced in chemically induced mouse skin papillomas, mouse and human squamous carcinomas and squamous cancer cell lines, and the protein is excluded from the nucleus. The extent of reduction in CLIC4 coincides with progression of squamous tumors from benign to malignant. Inhibiting antioxidant defense in tumor cells increases S-nitrosylation and nuclear translocation of CLIC4. Adenoviral-mediated reconstitution of nuclear CLIC4 in squamous cancer cells enhances TGF-ß-dependent transcriptional activity and inhibits growth. Adenoviral targeting of CLIC4 to the nucleus of tumor cells in orthografts inhibits tumor growth, whereas elevation of CLIC4 in transgenic epidermis reduces de novo chemically induced skin tumor formation. In parallel, overexpression of exogenous CLIC4 in squamous tumor orthografts suppresses tumor growth and enhances TGF-ß signaling. These results indicate that CLIC4 suppresses the growth of squamous cancers, that reduced CLIC4 expression and nuclear residence detected in cancer cells is associated with the altered redox state of tumor cells and the absence of detectable nuclear CLIC4 in cancers contributes to TGF-ß resistance and enhances tumor development.


Assuntos
Canais de Cloreto/biossíntese , Proteínas Mitocondriais/biossíntese , Neoplasias de Células Escamosas/metabolismo , Neoplasias Cutâneas/metabolismo , Proteínas Supressoras de Tumor/biossíntese , Animais , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Fibroblastos/metabolismo , Humanos , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos SENCAR , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Neoplasias de Células Escamosas/genética , Oxirredução , Papiloma/genética , Papiloma/metabolismo , Transporte Proteico , Transdução de Sinais , Neoplasias Cutâneas/genética , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
7.
Clin Cancer Res ; 18(1): 170-83, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22068661

RESUMO

PURPOSE: Epidermal growth factor receptor (EGFR)-targeted therapy is in clinical use to treat squamous cell carcinoma of the head and neck and other cancers of lining epithelium. RAS mutations in these tumors are a negative prognostic factor for response, and skin inflammation is an adverse reaction to therapy. We investigated transcriptional and biochemical changes that could account for the confounding effects of RAS activation and inflammation in a squamous tissue. EXPERIMENTAL DESIGN: We carried out gene expression profiling on oncogenic Ras-transformed and wild-type mouse and human keratinocytes with EGFR ablated chronically by genetic deletion or acutely by drug treatment and followed leads provided by pathway analysis with biochemical studies. RESULTS: We identified a 25-gene signature specific to the Ras-EGFR ablation interaction and a distinct 19-gene EGFR ablation signature on normal keratinocytes. EGFR ablation in the context of wild-type Ras reduces ontologies favoring cell-cycle control and transcription, whereas oncogenic Ras enriches ontologies for ion channels and membrane transporters, particularly focused on calcium homeostasis. Ontologies between chronic EGFR ablation and acute pharmacologic ablation were unique, both with and without Ras activation. p38α is activated in response to abrogation of EGFR signaling under conditions of Ras activation in both mouse and human keratinocytes and in RAS-transformed tumor orthografts of EGFR-ablated mouse keratinocytes. EGFR ablation in the absence of oncogenic Ras revealed Erk and interleukin-1ß-related pathways. CONCLUSION: These findings reveal unrecognized interactions between Ras and EGFR signaling in squamous tumor cells that could influence the therapeutic response to EGFR ablation therapy.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Transformação Celular Neoplásica/patologia , Receptores ErbB/fisiologia , Perfilação da Expressão Gênica , Queratinócitos/patologia , Proteínas ras/metabolismo , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Carcinoma de Células Escamosas/etiologia , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Humanos , Técnicas Imunoenzimáticas , Queratinócitos/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 14 Ativada por Mitógeno/genética , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais
8.
PLoS One ; 5(8): e12249, 2010 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-20805887

RESUMO

Dietary selenium is known to protect skin against UV-induced damage and cancer and its topical application improves skin surface parameters in humans, while selenium deficiency compromises protective antioxidant enzymes in skin. Furthermore, skin and hair abnormalities in humans and rodents may be caused by selenium deficiency, which are overcome by dietary selenium supplementation. Most important biological functions of selenium are attributed to selenoproteins, proteins containing selenium in the form of the amino acid, selenocysteine (Sec). Sec insertion into proteins depends on Sec tRNA; thus, knocking out the Sec tRNA gene (Trsp) ablates selenoprotein expression. We generated mice with targeted removal of selenoproteins in keratin 14 (K14) expressing cells and their differentiated descendents. The knockout progeny had a runt phenotype, developed skin abnormalities and experienced premature death. Lack of selenoproteins in epidermal cells led to the development of hyperplastic epidermis and aberrant hair follicle morphogenesis, accompanied by progressive alopecia after birth. Further analyses revealed that selenoproteins are essential antioxidants in skin and unveiled their role in keratinocyte growth and viability. This study links severe selenoprotein deficiency to abnormalities in skin and hair and provides genetic evidence for the role of these proteins in keratinocyte function and cutaneous development.


Assuntos
Queratinócitos/citologia , Queratinócitos/metabolismo , Selenoproteínas/metabolismo , Pele/crescimento & desenvolvimento , Pele/metabolismo , Animais , Animais Recém-Nascidos , Antioxidantes/metabolismo , Biomarcadores/metabolismo , Adesão Celular , Diferenciação Celular , Proliferação de Células , Células Epidérmicas , Epiderme/metabolismo , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Folículo Piloso/citologia , Folículo Piloso/metabolismo , Queratina-14/genética , Queratina-14/metabolismo , Peroxidação de Lipídeos , Macrófagos/citologia , Macrófagos/imunologia , Masculino , Camundongos , Especificidade de Órgãos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA de Transferência Aminoácido-Específico/genética , RNA de Transferência Aminoácido-Específico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Recombinação Genética , Selênio/deficiência , Selenoproteínas/deficiência , Selenoproteínas/genética , Pele/citologia
9.
J Invest Dermatol ; 130(9): 2295-303, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20410912

RESUMO

Overexpression of transforming growth factor-beta1 (TGFbeta1) in the normal epidermis can provoke an inflammatory response, but whether this occurs within a developing tumor is not clear. To test this, we used an inducible transgenic mouse to overexpress TGFbeta1 in premalignant squamous lesions. Within 48 hours of TGFbeta1 induction, there was an increase in IL-17 production by both CD4(+) and gammadelta(+) T cells, together with increased expression of T-helper-17 (Th17)-polarizing cytokines. Induction of TGFbeta1 in premalignant primary keratinocytes elevated the expression of proinflammatory and Th17-polarizing cytokines, and the keratinocyte-conditioned media caused IL-17 production by naive T cells that was dependent on T-cell TGFbeta1 signaling. Microarray analysis showed significant upregulation of proinflammatory genes 2 days after TGFbeta1 induction, and this was followed by increased MPO(+), F4/80(+), and CD8(+) cells in tumors, increased CD8(+) effectors and IFNgamma(+) cells in skin-draining LNs, and tumor regression. In parallel, the percentage of tumor CD11b(+)Ly6G(+) neutrophils was reduced. Neutralization of IL-17 blocked TGFbeta1-induced CD11b(+) Ly6G(-) tumor infiltration but did not alter the reduction of neutrophils or tumor regression. Thus, TGFbeta1 overexpression causes IL-17-dependent and IL-17-independent changes in the premalignant tumor inflammatory microenvironment.


Assuntos
Carcinoma de Células Escamosas/imunologia , Dermatite/imunologia , Interleucina-17 , Lesões Pré-Cancerosas/imunologia , Neoplasias Cutâneas/imunologia , Fator de Crescimento Transformador beta1 , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Carcinoma de Células Escamosas/patologia , Células Cultivadas , Dermatite/patologia , Progressão da Doença , Epiderme/imunologia , Epiderme/metabolismo , Epiderme/patologia , Interleucina-17/genética , Interleucina-17/imunologia , Interleucina-17/metabolismo , Queratinócitos/imunologia , Queratinócitos/metabolismo , Queratinócitos/patologia , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Papiloma/imunologia , Papiloma/patologia , Lesões Pré-Cancerosas/patologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Transdução de Sinais/imunologia , Neoplasias Cutâneas/patologia , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/imunologia , Fator de Crescimento Transformador beta1/metabolismo
10.
Mol Carcinog ; 49(4): 363-73, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20082322

RESUMO

Syndecan-1 (sdc-1) is a cell surface proteoglycan that mediates the interaction of cells with their matrix, influencing attachment, migration, and response to growth factors. In keratinocytes, loss of sdc-1 delays wound healing, reduces migration, and increases Transforming growth factor beta (TGFbeta) 1 expression. In this study we show that sdc-1 expression is significantly reduced in basal cell, squamous cell, and metastatic human skin cancers compared to normal human skin. In experimental mouse skin tumor induction, compared to wildtype (wt) BALB/c mice, papilloma formation in sdc-1 null mice was reduced by 50% and the percent of papillomas converting to squamous cell carcinoma (SCC) was enhanced. sdc-1 expression on wt mouse papillomas decreased as they converted to SCC. Furthermore, papillomas forming on sdc-1 null mice expressed suprabasal alpha3 and beta4 integrins; suprabasal beta4 integrin is a marker of a high risk for progression. While the proliferative response to phorbol-12-myristate-13-acetate (TPA) did not differ among the genotypes, sdc-1 null mice had an enhanced inflammatory response and retained higher levels of total TGFbeta1 within their skin after TPA treatment. sdc-1 null keratinocytes, transduced in vitro by oncogenic ras(Ha), expressed higher levels of beta4 integrin and had enhanced pSmad2 signaling and reduced senescence when compared to wt ras(Ha)-transduced keratinocytes. When ras(Ha)-transduced cells of both genotypes were grafted onto nude mice, null tumors converted to SCC with higher frequency confirming the skin painting experiments. These data indicate that sdc-1 is important both early in the development of skin tumors and in progression of skin cancers suggesting that reduced expression of sdc-1 could be a useful marker for progression in neoplastic skin lesions.


Assuntos
Transformação Celular Neoplásica , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Sindecana-1/genética , Animais , Carcinógenos/toxicidade , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Papiloma/patologia , Ésteres de Forbol/toxicidade , Neoplasias Cutâneas/metabolismo , Sindecana-1/metabolismo , Testes de Toxicidade Aguda , Testes de Toxicidade Crônica
11.
Mol Cancer Res ; 6(3): 509-16, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18337457

RESUMO

Cripto-1 is an epidermal growth factor-Cripto/FRL1/Cryptic family member that plays a role in early embryogenesis as a coreceptor for Nodal and is overexpressed in human tumors. Here we report that in the two-stage mouse skin carcinogenesis model, Cripto-1 is highly up-regulated in tumor promoter-treated normal skin and in benign papillomas. Treatment of primary mouse keratinocytes with Cripto-1 stimulated proliferation and induced expression of keratin 8 but blocked induction of the normal epidermal differentiation marker keratin 1, changes that are hallmarks of tumor progression in squamous cancer. Chemical or genetic blockade of the transforming growth factor (TGF)-beta1 signaling pathway using the ALK5 kinase inhibitor SB431542 and dominant negative TGF-beta type II receptor, respectively, had similar effects on keratinocyte differentiation. Our results show that Cripto-1 could block TGF-beta1 receptor binding, phosphorylation of Smad2 and Smad3, TGF-beta-responsive luciferase reporter activity, and TGF-beta1-mediated senescence of keratinocytes. We suggest that inhibition of TGF-beta1 by Cripto-1 may play an important role in altering the differentiation state of keratinocytes and promoting outgrowth of squamous tumors in the mouse epidermis.


Assuntos
Diferenciação Celular/fisiologia , Fator de Crescimento Epidérmico/fisiologia , Glicoproteínas de Membrana/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias Cutâneas/fisiopatologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores , 9,10-Dimetil-1,2-benzantraceno , Animais , Carcinoma de Células Escamosas , Diferenciação Celular/genética , Replicação do DNA , Fator de Crescimento Epidérmico/genética , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Queratinócitos/fisiologia , Glicoproteínas de Membrana/genética , Camundongos , Proteínas de Neoplasias/genética , Proteínas Serina-Treonina Quinases/fisiologia , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Transdução de Sinais , Neoplasias Cutâneas/genética , Fator de Crescimento Transformador beta1/fisiologia
12.
J Cell Sci ; 120(Pt 15): 2631-40, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17636002

RESUMO

Keratinocyte differentiation requires integrating signaling among intracellular ionic changes, kinase cascades, sequential gene expression, cell cycle arrest, and programmed cell death. We now show that Cl(-) intracellular channel 4 (CLIC4) expression is increased in both mouse and human keratinocytes undergoing differentiation induced by Ca(2+), serum and the protein kinase C (PKC)-activator, 12-O-tetradecanoyl-phorbol-13-acetate (TPA). Elevation of CLIC4 is associated with signaling by PKCdelta, and knockdown of CLIC4 protein by antisense or shRNA prevents Ca(2+)-induced keratin 1, keratin 10 and filaggrin expression and cell cycle arrest in differentiating keratinocytes. CLIC4 is cytoplasmic in actively proliferating keratinocytes in vitro, but the cytoplasmic CLIC4 translocates to the nucleus in keratinocytes undergoing growth arrest by differentiation, senescence or transforming growth factor beta (TGFbeta) treatment. Targeting CLIC4 to the nucleus of keratinocytes via adenoviral transduction increases nuclear Cl(-) content and enhances expression of differentiation markers in the absence of elevated Ca(2+). In vivo, CLIC4 is localized to the epidermis in mouse and human skin, where it is predominantly nuclear in quiescent cells. These results suggest that CLIC4 participates in epidermal homeostasis through both alterations in the level of expression and subcellular localization. Nuclear CLIC4, possibly by altering the Cl(-) and pH of the nucleus, contributes to cell cycle arrest and the specific gene expression program associated with keratinocyte terminal differentiation.


Assuntos
Cálcio/metabolismo , Diferenciação Celular , Canais de Cloreto/metabolismo , Queratinócitos/citologia , Proteína Quinase C/metabolismo , Animais , Núcleo Celular/metabolismo , Células Cultivadas , Canais de Cloreto/isolamento & purificação , Proteínas Filagrinas , Expressão Gênica , Humanos , Proteínas de Filamentos Intermediários/metabolismo , Queratinócitos/metabolismo , Queratinas/metabolismo , Camundongos , Isoformas de Proteínas/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Fator de Transcrição AP-1/metabolismo
13.
Mol Carcinog ; 46(8): 605-10, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17538943

RESUMO

Benign tumors that form following chemical initiation and promotion in the mouse skin can be grouped into two classes. The majority of papillomas do not progress to squamous cell carcinoma (SCC), and these are designated as low-risk or terminally benign papillomas. In contrast, a much smaller group forms the true precursor to the SCC, and these have a significantly higher frequency and rate of malignant conversion than the bulk of low-risk papillomas. In standard two-stage carcinogenesis studies both tumor types are present, but grossly indistinguishable. Here we describe properties and potential origins of high-risk papillomas and discuss the relevance of this model for certain human cancers with defined premalignant states.


Assuntos
Carcinoma de Células Escamosas/etiologia , Modelos Animais de Doenças , Papiloma/etiologia , Neoplasias Cutâneas/etiologia , Animais , Carcinoma de Células Escamosas/patologia , Humanos , Papiloma/patologia , Fatores de Risco , Neoplasias Cutâneas/patologia
14.
Clin Cancer Res ; 13(1): 121-31, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17200346

RESUMO

PURPOSE: CLIC4, a member of a family of intracellular chloride channels, is regulated by p53, c-Myc, and tumor necrosis factor-alpha. Regulation by factors involved in cancer pathogenesis, together with the previously shown proapoptotic activity of CLIC4, suggests that the protein may have a tumor suppressor function. To address this possibility, we characterized the expression profile, subcellular localization, and gene integrity of CLIC4 in human cancers and determined the functional consequences of CLIC4 expression in tumor epithelium and stromal cells. EXPERIMENTAL DESIGN: CLIC4 expression profiles were analyzed by genomics, proteomics, bioinformatics, and tissue microarrays. CLIC4 expression, as a consequence of crosstalk between stroma and epithelium, was tested in vitro by coculture of breast epithelial tumor cells and normal fibroblasts, and the functional consequences of CLIC4 expression was tested in vivo in xenografts of human breast tumor cell lines reconstituted with CLIC4 or mixed with fibroblasts that overexpress CLIC4 transgenically. RESULTS: In cDNA arrays of matched human normal and tumor tissues, CLIC4 expression was reduced in renal, ovarian, and breast cancers. However, CLIC4 protein levels were variable in tumor lysate arrays. Transcript sequences of CLIC4 from the human expressed sequence tag database and manual sequencing of cDNA from 60 human cancer cell lines (NCI60) failed to reveal deletion or mutations in the CLIC4 gene. On matched tissue arrays, CLIC4 was predominantly nuclear in normal human epithelial tissues but not cancers. With advancing malignant progression, CLIC4 staining became undetectable in tumor cells, but expression increased in stromal cells coincident with up-regulation of alpha-smooth muscle actin, suggesting that CLIC4 is up-regulated in myofibroblasts. Coculture of cancer cells and fibroblasts induced the expression of both CLIC4 and alpha-smooth muscle actin in fibroblasts adjacent to tumor nests. Introduction of CLIC4 or nuclear targeted CLIC4 via adenovirus into human breast cancer xenografts inhibited tumor growth, whereas overexpression of CLIC4 in stromal cells of xenografts enhanced tumor growth. CONCLUSION: Loss of CLIC4 in tumor cells and gain in tumor stroma is common to many human cancers and marks malignant progression. Up-regulation of CLIC4 in tumor stroma is coincident with myofibroblast conversion, generally a poor prognostic indicator. Reactivation and restoration of CLIC4 in tumor cells or the converse in tumor stromal cells could provide a novel approach to inhibit tumor growth.


Assuntos
Canais de Cloreto/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias/patologia , Regulação para Cima , Actinas/metabolismo , Animais , Linhagem Celular Tumoral , Canais de Cloreto/genética , Análise Mutacional de DNA , Progressão da Doença , Epitélio/metabolismo , Fibroblastos/metabolismo , Genes Supressores de Tumor , Humanos , Camundongos , Transplante de Neoplasias , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA