Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Cutan Pathol ; 49(2): 116-122, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34322902

RESUMO

BACKGROUND: Expression of microRNA-21 (miR-21) is increased in psoriasis, leading to reduced levels of epidermal tissue inhibitor of matrix metalloproteinase 3 (TIMP-3), a highly potent inhibitor of the tumor necrosis factor alpha (TNFα) sheddase TACE (TNFα-converting enzyme)/ADAM17. We described the profile of miR-21 and TIMP-3 in paradoxical psoriasiform reactions induced by anti-TNFα drugs and in a control group to elucidate the pathogenesis of this reactions. METHODS: We performed an analytic, cross-sectional, prospective, experimental case-control study. We compared our findings with those of non-induced psoriasis. RESULTS: We included 15 patients with a change of morphology (plaque to guttate psoriasis) and 10 patients with induced psoriasis (six palmoplantar pustulosis and four plaque psoriasis). Consecutive patients with different subtypes of non-induced, non-systemically treated psoriasis were included as a control group. We found that most cases with guttate psoriasis and with induced plaque psoriasis cases showed high expression of TIMP-3 expression and decreased or poorly increased levels of miR-21. The expression pattern was not homogeneous in the cases of induced palmoplantar pustulosis. These profiles differ from those of non-induced psoriasis. CONCLUSION: We conclude that various pro-inflammatory cytokine profiles are involved in the pathogenesis of paradoxical psoriasiform reactions and non-induced psoriasis.


Assuntos
MicroRNAs/metabolismo , Psoríase/metabolismo , Inibidor Tecidual de Metaloproteinase-3/metabolismo , Inibidores do Fator de Necrose Tumoral/uso terapêutico , Adalimumab/uso terapêutico , Adulto , Biópsia , Estudos de Casos e Controles , Estudos Transversais , Feminino , Humanos , Infliximab/uso terapêutico , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Psoríase/tratamento farmacológico , Psoríase/patologia , Pele/metabolismo , Pele/patologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores
2.
Clin Exp Rheumatol ; 33(4 Suppl 92): S44-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26458100

RESUMO

Skin inflammation is a physiological reaction to tissue injury, pathogen invasion and irritants. During this process, innate and/or adaptive immune cells are activated and recruited to the site of inflammation to either promote or suppress inflammation. The sequential recruitment and activation of immune cells is modulated by a combination of cytokines and chemokines, which are regulated by transcription factors, such as AP-1 (Fos/Jun), NF-κB, NFATs, and STATs. Here we review the present evidence and the underlying mechanisms of how Jun/AP-1 proteins control skin inflammation. Genetically engineered mouse models (GEMMs) in which AP-1 proteins are deleted in the epidermis have revealed that these proteins control cytokine expression at multiple levels. Constitutive epidermal deletion of JunB in mice leads to a multi-organ disease characterised by increased levels of pro-inflammatory cytokines. These JunB-deficient mutant mice display several phenotypes from skin inflammation to a G-CSF-dependent myeloproliferative disease, as well as kidney atrophy and bone loss, reminiscent of psoriasis and systemic lupus erythematosus. Importantly, epidermal deletion of both JunB and c-Jun in an inducible manner in adult mice leads to a psoriasis-like disease, in which the epidermal proteome expression profile is comparable to the one from psoriasis patient samples. In this GEMM and in psoriasis patient-derived material, S100A8/A9-dependent C3/CFB complement activation, as well as a miR-21-dependent TIMP-3/TACE pathway leading to TNF-α shedding, plays causal roles in disease development. The newly identified therapeutic targets from GEMMs together with investigations in human patient samples open up new avenues for therapeutic interventions for psoriasis and related inflammatory skin diseases.


Assuntos
Dermatite/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Transdução de Sinais , Pele/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Dermatite/genética , Dermatite/imunologia , Dermatite/patologia , Regulação da Expressão Gênica , Genótipo , Humanos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-jun/genética , Pele/imunologia , Pele/patologia , Fator de Transcrição AP-1/genética
3.
Genes Dev ; 29(2): 144-56, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25547114

RESUMO

Altered epidermal differentiation characterizes numerous skin diseases affecting >25% of the human population. Here we identified Fra-2/AP-1 as a key regulator of terminal epidermal differentiation. Epithelial-restricted, ectopic expression of Fra-2 induced expression of epidermal differentiation genes located within the epidermal differentiation complex (EDC). Moreover, in a papilloma-prone background, a reduced tumor burden was observed due to precocious keratinocyte differentiation by Fra-2 expression. Importantly, loss of Fra-2 in suprabasal keratinocytes is sufficient to cause skin barrier defects due to reduced expression of differentiation genes. Mechanistically, Fra-2 binds and transcriptionally regulates EDC gene promoters, which are co-occupied by the transcriptional repressor Ezh2. Fra-2 remains transcriptionally inactive in nondifferentiated keratinocytes, where it was found monomethylated and dimethylated on Lys104 and interacted with Ezh2. Upon keratinocyte differentiation, Fra-2 is C-terminally phosphorylated on Ser320 and Thr322 by ERK1/2, leading to transcriptional activation. Thus, the induction of epidermal differentiation by Fra-2 is controlled by a dual mechanism involving Ezh2-dependent methylation and activation by ERK1/2-dependent phosphorylation.


Assuntos
Diferenciação Celular , Antígeno 2 Relacionado a Fos/metabolismo , Queratinócitos/citologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Células Cultivadas , Embrião de Mamíferos , Proteína Potenciadora do Homólogo 2 de Zeste , Regulação da Expressão Gênica no Desenvolvimento , Lisina/metabolismo , Metilação , Camundongos , Fosforilação
4.
Sci Transl Med ; 6(225): 225re1, 2014 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-24574341

RESUMO

Psoriasis is a common inflammatory skin disease with limited treatment options that is characterized by a complex interplay between keratinocytes, immune cells, and inflammatory mediators. MicroRNAs (miRNAs) are regulators of gene expression and play critical roles in many human diseases. A number of miRNAs have been described to be up-regulated in psoriasis, but their causal contribution to disease development has not been demonstrated. We confirm that miR-21 expression is increased in epidermal lesions of patients with psoriasis and that this leads to reduced epidermal TIMP-3 (tissue inhibitor of matrix metalloproteinase 3) expression and activation of TACE (tumor necrosis factor-α-converting enzyme)/ADAM17 (a disintegrin and metalloproteinase 17). Using patient-derived skin samples and mouse models of psoriasis, we demonstrate that increased miR-21 may be a consequence of impaired transcriptional activity of Jun/activating protein 1 (AP-1), leading to activation of the interleukin-6 (IL-6)/signal transducer and activator of transcription 3 (Stat3) pathway. Inhibition of miR-21 by locked nucleic acid (LNA)-modified anti-miR-21 compounds ameliorated disease pathology in patient-derived psoriatic skin xenotransplants in mice and in a psoriasis-like mouse model. Targeting miR-21 may represent a potential therapeutic option for the treatment of psoriasis.


Assuntos
Marcação de Genes , Terapia Genética/métodos , MicroRNAs/antagonistas & inibidores , Oligonucleotídeos/administração & dosagem , Psoríase/terapia , Pele/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Animais , Biópsia , Estudos de Casos e Controles , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , Psoríase/genética , Psoríase/metabolismo , Psoríase/patologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Pele/patologia , Transplante de Pele , Inibidor Tecidual de Metaloproteinase-3/genética , Inibidor Tecidual de Metaloproteinase-3/metabolismo , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica , Transfecção , Transplante Heterólogo , Regulação para Cima
5.
Immunity ; 39(6): 1171-81, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24332034

RESUMO

Psoriasis is a common heterogeneous inflammatory skin disease with a complex pathophysiology and limited treatment options. Here we performed proteomic analyses of human psoriatic epidermis and found S100A8-S100A9, also called calprotectin, as the most upregulated proteins, followed by the complement component C3. Both S100A8-S100A9 and C3 are specifically expressed in lesional psoriatic skin. S100A9 is shown here to function as a chromatin component modulating C3 expression in mouse and human cells by binding to a region upstream of the C3 start site. When S100A9 was genetically deleted in mouse models of skin inflammation, the psoriasis-like skin disease and inflammation were strongly attenuated, with a mild immune infiltrate and decreased amounts of C3. In addition, inhibition of C3 in the mouse model strongly reduced the inflammatory skin disease. Thus, S100A8-S100A9 can regulate C3 at the nuclear level and present potential new therapeutic targets for psoriasis.


Assuntos
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Complemento C3/genética , Regulação da Expressão Gênica , Psoríase/genética , Psoríase/fisiopatologia , Animais , Calgranulina A/genética , Calgranulina B/genética , Núcleo Celular/metabolismo , Células Cultivadas , Complemento C3/metabolismo , Modelos Animais de Doenças , Células Epidérmicas , Epiderme/imunologia , Humanos , Camundongos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteoma , Psoríase/imunologia , RNA Interferente Pequeno/metabolismo
6.
Genes Dev ; 27(18): 1959-73, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24029918

RESUMO

Skin squamous cell carcinomas (SCCs) are the second most prevalent skin cancers. Chronic skin inflammation has been associated with the development of SCCs, but the contribution of skin inflammation to SCC development remains largely unknown. In this study, we demonstrate that inducible expression of c-fos in the epidermis of adult mice is sufficient to promote inflammation-mediated epidermal hyperplasia, leading to the development of preneoplastic lesions. Interestingly, c-Fos transcriptionally controls mmp10 and s100a7a15 expression in keratinocytes, subsequently leading to CD4 T-cell recruitment to the skin, thereby promoting epidermal hyperplasia that is likely induced by CD4 T-cell-derived IL-22. Combining inducible c-fos expression in the epidermis with a single dose of the carcinogen 7,12-dimethylbenz(a)anthracene (DMBA) leads to the development of highly invasive SCCs, which are prevented by using the anti-inflammatory drug sulindac. Moreover, human SCCs display a correlation between c-FOS expression and elevated levels of MMP10 and S100A15 proteins as well as CD4 T-cell infiltration. Our studies demonstrate a bidirectional cross-talk between premalignant keratinocytes and infiltrating CD4 T cells in SCC development. Therefore, targeting inflammation along with the newly identified targets, such as MMP10 and S100A15, represents promising therapeutic strategies to treat SCCs.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Epiderme/metabolismo , Inflamação , Proteínas Proto-Oncogênicas c-fos/metabolismo , Neoplasias Cutâneas/fisiopatologia , Animais , Benzo(a)Antracenos , Linfócitos T CD4-Positivos/citologia , Carcinoma de Células Escamosas/fisiopatologia , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Hiperplasia/genética , Hiperplasia/metabolismo , Hiperplasia/fisiopatologia , Masculino , Metaloproteinase 10 da Matriz/genética , Metaloproteinase 10 da Matriz/metabolismo , Camundongos , Papiloma/induzido quimicamente , Papiloma/patologia , Proteínas Proto-Oncogênicas c-fos/genética , Transdução de Sinais
7.
EMBO J ; 32(16): 2248-63, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23860128

RESUMO

Squamous cell carcinomas (SCCs) are highly heterogeneous tumours, resulting from deranged expression of genes involved in squamous cell differentiation. Here we report that microRNA-34a (miR-34a) functions as a novel node in the squamous cell differentiation network, with SIRT6 as a critical target. miR-34a expression increases with keratinocyte differentiation, while it is suppressed in skin and oral SCCs, SCC cell lines, and aberrantly differentiating primary human keratinocytes (HKCs). Expression of this miRNA is restored in SCC cells, in parallel with differentiation, by reversion of genomic DNA methylation or wild-type p53 expression. In normal HKCs, the pro-differentiation effects of increased p53 activity or UVB exposure are miR-34a-dependent, and increased miR-34a levels are sufficient to induce differentiation of these cells both in vitro and in vivo. SIRT6, a sirtuin family member not previously connected with miR-34a function, is a direct target of this miRNA in HKCs, and SIRT6 down-modulation is sufficient to reproduce the miR-34a pro-differentiation effects. The findings are of likely biological significance, as SIRT6 is oppositely expressed to miR-34a in normal keratinocytes and keratinocyte-derived tumours.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Diferenciação Celular/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Queratinócitos/fisiologia , MicroRNAs/metabolismo , Sirtuínas/metabolismo , Diferenciação Celular/efeitos da radiação , Primers do DNA/genética , Humanos , Queratinócitos/metabolismo , Análise em Microsséries , Reação em Cadeia da Polimerase em Tempo Real , Receptor Notch1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta
8.
Mol Cell Biol ; 33(12): 2388-401, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23572561

RESUMO

Transforming growth factor ß1 (TGF-ß1) is a pleiotropic factor involved in the regulation of extracellular matrix (ECM) synthesis and remodeling. In search for novel genes mediating the action of TGF-ß1 on vascular ECM, we identified the member of the lysyl oxidase family of matrix-remodeling enzymes, lysyl oxidase-like 4 (LOXL4), as a direct target of TGF-ß1 in aortic endothelial cells, and we dissected the molecular mechanism of its induction. Deletion mapping and mutagenesis analysis of the LOXL4 promoter demonstrated the absolute requirement of a distal enhancer containing an activator protein 1 (AP-1) site and a Smad binding element for TGF-ß1 to induce LOXL4 expression. Functional cooperation between Smad proteins and the AP-1 complex composed of JunB/Fra2 accounted for the action of TGF-ß1, which involved the extracellular signal-regulated kinase (ERK)-dependent phosphorylation of Fra2. We furthermore provide evidence that LOXL4 was extracellularly secreted and significantly contributed to ECM deposition and assembly. These results suggest that TGF-ß1-dependent expression of LOXL4 plays a role in vascular ECM homeostasis, contributing to vascular processes associated with ECM remodeling and fibrosis.


Assuntos
Antígeno 2 Relacionado a Fos/metabolismo , Proteína-Lisina 6-Oxidase/metabolismo , Proteínas Smad/metabolismo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Aorta/metabolismo , Sequência de Bases , Sítios de Ligação/genética , Sítios de Ligação/fisiologia , Bovinos , Células Cultivadas , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Antígeno 2 Relacionado a Fos/genética , Células HEK293 , Humanos , Camundongos , Fosforilação , Regiões Promotoras Genéticas , Proteína-Lisina 6-Oxidase/biossíntese , Proteína-Lisina 6-Oxidase/genética , Interferência de RNA , RNA Interferente Pequeno , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/genética
9.
J Clin Invest ; 122(8): 2898-910, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22772468

RESUMO

Squamous cell carcinomas (SCCs) are heterogeneous and aggressive skin tumors for which innovative, targeted therapies are needed. Here, we identify a p53/TACE pathway that is negatively regulated by FOS and show that the FOS/p53/TACE axis suppresses SCC by inducing differentiation. We found that epidermal Fos deletion in mouse tumor models or pharmacological FOS/AP-1 inhibition in human SCC cell lines induced p53 expression. Epidermal cell differentiation and skin tumor suppression were caused by a p53-dependent transcriptional activation of the metalloprotease TACE/ADAM17 (TNF-α-converting enzyme), a previously unknown p53 target gene that was required for NOTCH1 activation. Although half of cutaneous human SCCs display p53-inactivating mutations, restoring p53/TACE activity in mouse and human skin SCCs induced tumor cell differentiation independently of the p53 status. We propose FOS/AP-1 inhibition or p53/TACE reactivating strategies as differentiation-inducing therapies for SCCs.


Assuntos
Proteínas ADAM/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas ADAM/genética , Proteína ADAM17 , Animais , Sequência de Bases , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/terapia , Diferenciação Celular , Linhagem Celular Tumoral , Primers do DNA/genética , Expressão Gênica , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-fos/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-fos/deficiência , Proteínas Proto-Oncogênicas c-fos/genética , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/terapia , Proteína Supressora de Tumor p53/genética
10.
Ann Rheum Dis ; 70 Suppl 1: i109-12, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21339212

RESUMO

Inflammation is a physiological response of the body to tissue injury, pathogen invasion and irritants. In the course of inflammation, immune cells of the innate and/or adaptive immune system are activated and recruited to the site of inflammation. Attraction and activation of immune cells is regulated by a variety of different cytokines and chemokines, which are predominantly regulated by transcription factors such as AP-1, NF-κB, NFATs and STATs. The evidence that Jun/AP-1 proteins control inflammation in the skin is summarised in this article. Genetic mouse models have demonstrated that a loss of Jun/AP-1 expression in epidermal cells controls cytokine expression through transcriptional and post-transcriptional pathways. The absence of JunB in epithelial K5-expressing tissues leads to a multiorgan disease, which is characterised by increased levels of granulocyte colony-stimulating factor and interleukin 6. Deletion of both JunB and c-Jun, in a constitutive or inducible manner, leads to perinatal death of newborn pups and to a psoriasis-like disease in adults, in which tumour necrosis factor α and the TIMP-3/TACE pathway have central roles. The loss or reduction of Jun expression in the epidermis relieves a block on cytokine expression. As a consequence, the increased levels of cytokines in mice lead to diseases reminiscent of psoriasis and systemic lupus erythematosus in human patients. New targets identified in mouse models, together with investigations on human samples, will provide important new avenues for therapeutic interventions in psoriasis and other inflammatory skin diseases.


Assuntos
Dermatite/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Camundongos , MicroRNAs/genética , Terapia de Alvo Molecular/métodos , Psoríase/metabolismo , Transdução de Sinais
11.
Nat Rev Rheumatol ; 6(12): 704-14, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20877306

RESUMO

Psoriasis is a common inflammatory skin disease of unknown etiology, for which there is no cure. This heterogeneous, cutaneous, inflammatory disorder is clinically characterized by prominent epidermal hyperplasia and a distinct inflammatory infiltrate. Crosstalk between immunocytes and keratinocytes, which results in the production of cytokines, chemokines and growth factors, is thought to mediate the disease. Given that psoriasis is only observed in humans, numerous genetic approaches to model the disease in mice have been undertaken. In this Review, we describe and critically assess the mouse models and transplantation experiments that have contributed to the discovery of novel disease-relevant pathways in psoriasis. Research performed using improved mouse models, combined with studies employing human cells, xenografts and patient material, will be key to our understanding of why such distinctive patterns of inflammation develop in patients with psoriasis. Indeed, a combination of genetic and immunological investigations will be necessary to develop both improved drugs for the treatment of psoriasis and novel curative strategies.


Assuntos
Modelos Animais de Doenças , Psoríase/genética , Psoríase/fisiopatologia , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular/fisiologia , Humanos , Queratinócitos/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Psoríase/tratamento farmacológico , Transplante Heterólogo
12.
Genes Dev ; 23(22): 2663-74, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19933155

RESUMO

Inducible epidermal deletion of JunB and c-Jun in adult mice causes a psoriasis-like inflammatory skin disease. Increased levels of the proinflammatory cytokine TNFalpha play a major role in this phenotype. Here we define the underlying molecular mechanism using genetic mouse models. We show that Jun proteins control TNFalpha shedding in the epidermis by direct transcriptional activation of tissue inhibitor of metalloproteinase-3 (TIMP-3), an inhibitor of the TNFalpha-converting enzyme (TACE). TIMP-3 is down-regulated and TACE activity is specifically increased, leading to massive, cell-autonomous TNFalpha shedding upon loss of both JunB and c-Jun. Consequently, a prominent TNFalpha-dependent cytokine cascade is initiated in the epidermis, inducing severe skin inflammation and perinatal death of newborns from exhaustion of energy reservoirs such as glycogen and lipids. Importantly, this metabolic "cachectic" phenotype can be genetically rescued in a TNFR1-deficient background or by epidermis-specific re-expression of TIMP-3. These findings reveal that Jun proteins are essential physiological regulators of TNFalpha shedding by controlling the TIMP-3/TACE pathway. This novel mechanism describing how Jun proteins control skin inflammation offers potential targets for the treatment of skin pathologies associated with increased TNFalpha levels.


Assuntos
Epiderme/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Caquexia/genética , Caquexia/mortalidade , Células Cultivadas , Citocinas/metabolismo , Epiderme/patologia , Regulação da Expressão Gênica , Inflamação/fisiopatologia , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-jun/deficiência , Proteínas Proto-Oncogênicas c-jun/genética , Dermatopatias/fisiopatologia , Inibidor Tecidual de Metaloproteinase-3/metabolismo
13.
Cancer Res ; 68(17): 6877-83, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18757399

RESUMO

Expression and function of the oncogenic transcription factor activator protein (AP-1; mainly composed of Jun and Fos proteins) is required for neoplastic transformation of keratinocytes in vitro and tumor promotion as well as malignant progression in vivo. Here, we describe the identification of 372 differentially expressed genes comparing skin tumor samples of K5-SOS-F transgenic mice (Fos(f/f) SOS(+)) with samples derived from animals with a specific deletion of c-Fos in keratinocytes (Fos(Deltaep) SOS(+)). Fos-dependent transcription of selected genes was confirmed by quantitative real-time PCR analysis using tumor samples and mouse back skin treated with the tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA). One of the most differentially expressed genes encodes the small mucin-like glycoprotein Podoplanin (Pdpn), whose expression correlates with malignant progression in mouse tumor model systems and human cancer. We found Pdpn and Fos expression in chemically induced mouse skin tumors, and detailed analysis of the Pdpn gene promoter revealed impaired activity in Fos-deficient mouse embryonic fibroblasts, which could be restored by ectopic Fos expression. Direct Fos protein binding to the Pdpn promoter was shown by chromatin immunoprecipitation and a TPA-induced complex at a TPA-responsive element-like motif in the proximal promoter was identified by electrophoretic mobility shift assays. In summary, we could define a Fos-dependent genetic program in a well-established model of skin tumors. Systematic analysis of these novel target genes will guide us in elucidating the molecular mechanisms of AP-1-regulated pathways that are critically implicated in neoplastic transformation and/or malignant progression.


Assuntos
Transformação Celular Neoplásica , Genes fos , Glicoproteínas de Membrana/genética , Neoplasias Cutâneas/genética , Animais , Progressão da Doença , Camundongos , Invasividade Neoplásica , Neoplasias Cutâneas/patologia
14.
Nat Cell Biol ; 10(8): 902-11, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18604200

RESUMO

The Notch1 gene has an important role in mammalian cell-fate decision and tumorigenesis. Upstream control mechanisms for transcription of this gene are still poorly understood. In a chemical genetics screen for small molecule activators of Notch signalling, we identified epidermal growth factor receptor (EGFR) as a key negative regulator of Notch1 gene expression in primary human keratinocytes, intact epidermis and skin squamous cell carcinomas (SCCs). The underlying mechanism for negative control of the Notch1 gene in human cells, as well as in a mouse model of EGFR-dependent skin carcinogenesis, involves transcriptional suppression of p53 by the EGFR effector c-Jun. Suppression of Notch signalling in cancer cells counteracts the differentiation-inducing effects of EGFR inhibitors while, at the same time, synergizing with these compounds in induction of apoptosis. Thus, our data reveal a key role of EGFR signalling in the negative regulation of Notch1 gene transcription, of potential relevance for combinatory approaches for cancer therapy.


Assuntos
Carcinoma de Células Escamosas/patologia , Receptores ErbB/fisiologia , Regulação da Expressão Gênica , Queratinócitos/citologia , Receptor Notch1/genética , Animais , Carcinoma de Células Escamosas/etiologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Humanos , Camundongos , Transdução de Sinais , Neoplasias Cutâneas , Proteína Supressora de Tumor p53/genética
15.
J Biol Chem ; 283(19): 13450-8, 2008 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-18316372

RESUMO

In response to oncogenic signals, cells have developed safe mechanisms to avoid transformation through activation of a senescence program. Upon v-H-Ras overexpression, normal cells undergo senescence through several cellular processes, including activation of the ERK1/2 pathway. Interestingly, the E1a gene from adenovirus 5 has been shown to rescue cells from senescence by a yet unknown mechanism. We investigated whether E1a was able to interfere with the ERK1/2 signaling pathway to rescue cells from v-H-Ras-mediated senescence. Our results show that, E1a overexpression blocks v-H-Ras-mediated ERK1/2 activation by two different and concomitant mechanisms. E1a through its ability to interfere with PKB/Akt activation induces the down-regulation of the PEA15 protein, an ERK1/2 nuclear export factor, leading to nuclear accumulation of ERK1/2. In addition to this, we show that E1a increases the expression of the inducible ERK1/2 nuclear phosphatases (MAPK phosphatases) MKP1/DUSP1 and DUSP5, which leads to ERK1/2 dephosphorylation. We confirmed our observations in the human normal diploid fibroblasts IMR90, in which we could also show that an E1a mutant, unable to bind retinoblastoma protein (pRb), cannot rescue cells from v-H-Ras-induced senescence. In conclusion, E1a is able to rescue from Ras-induced senescence by affecting ERK1/2 localization and phosphorylation.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Proteína Oncogênica p21(ras)/metabolismo , Regulação para Cima , Transporte Ativo do Núcleo Celular , Proteínas E1A de Adenovirus/genética , Animais , Linhagem Celular , Núcleo Celular/enzimologia , Senescência Celular/fisiologia , Chlorocebus aethiops , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação para Baixo , Ativação Enzimática , Regulação da Expressão Gênica , Humanos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Oncogênica p21(ras)/genética , Ligação Proteica , Proteína do Retinoblastoma/metabolismo , Soro
16.
Oncogene ; 22(26): 3998-4006, 2003 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-12821934

RESUMO

p38 MAPK has been implicated in the response to cancer therapy. To determine whether the activation of p38 MAPK could be specific to cancer therapy, we investigated the activation of p38 MAPK in response to several chemotherapeutic agents, such as cisplatin, doxorubicin and taxol in several human cell lines. Activation of p38 MAPK was measured after exposure to several chemotherapeutic agents, using specific phosphoantibodies. Only cisplatin was able to activate p38 MAPK in all the cell lines tested. Furthermore, other platinum compounds such as transplatin and platinum (IV) chloride can induce activation of p38 MAPK. The kinetics of this activation is a key event in the biological role of p38 MAPK in response to cisplatin, as we conclude from the differences observed after treatment with transplatin and cisplatin. The p38 MAPK activation is independent of the origin or genetic alterations of the cell lines and seems to be mediated through both upstream activators MKK6 and MKK3. Although the isoforms alpha/beta are mainly activated, we also demonstrated that other members of the p38 MAPK family were susceptible to activation by cisplatin when they were overexpressed in 293 T. Finally, pretreatment with specific inhibitors (SB 203580 and SKF 86002) induces a resistant phenotype in response to cisplatin. Furthermore, low activation of this SAPK pathway correlates with a resistant phenotype as demonstrated in our experimental model of head and neck cancer. Therefore, we conclude that the p38 MAPK pathway is a specific target for cisplatin-based therapy with clinical implications.


Assuntos
Cisplatino/farmacologia , Proteínas Quinases Ativadas por Mitógeno , Animais , Antineoplásicos/farmacologia , Células COS , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Linhagem Celular , Sobrevivência Celular , Reagentes de Ligações Cruzadas/farmacologia , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Resistencia a Medicamentos Antineoplásicos , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Imidazóis/farmacologia , Cinética , MAP Quinase Quinase 3 , MAP Quinase Quinase 6 , Proteína Quinase 12 Ativada por Mitógeno , Proteína Quinase 13 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fenótipo , Fosforilação , Plasmídeos/metabolismo , Compostos de Platina/farmacologia , Testes de Precipitina , Isoformas de Proteínas , Proteínas Tirosina Quinases/metabolismo , Piridinas/farmacologia , Transdução de Sinais , Tiazóis/farmacologia , Transfecção , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno
17.
Oncogene ; 21(46): 7131-6, 2002 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-12370836

RESUMO

In order to investigate the molecular mechanisms implicated in the induction of chemo sensitivity by adenovirus E1a gene expression, we decided to investigate which signal transduction pathways could be affected by the E1a gene in Human Normal Fibroblast (IMR90). No effect was observed in SAPK pathways (p38MAPK and JNK), but E1a was able to affect the Akt activation mediated by insulin. This result was confirmed by transient transfection experiments performed in Cos-7 cells and also observed in other transformed cell lines such as A431. Furthermore, E1a expression induces a decrease in the basal status of Akt activity. Finally we demonstrated that E1a is able to block the Akt activation mediated by cisplatin and correlates with a sensitive phenotype. In summary, our data demonstrate that specific inhibition of the PI3K/Akt pathway mediates some of the biological properties of E1a such as induction of chemosensitivity.


Assuntos
Proteínas E1A de Adenovirus/genética , Antineoplásicos/farmacologia , Cisplatino/farmacologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/fisiologia , Transdução de Sinais , Animais , Células COS , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Genes ras , Humanos , Insulina/farmacologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA