Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
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
2.
Nat Methods ; 9(9): 897-900, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22863881

RESUMO

We here establish a mouse cancer model called Multi-Hit that allows for the evaluation of oncogene cooperativities in tumor development. The model is based on the stochastic expression of oncogene combinations ('hits') that are mediated by Cre in a given tissue. Cells with cooperating hits are positively selected and give rise to tumors. We used this approach to evaluate the requirement of Ras downstream effector pathways in tumorigenesis.


Assuntos
Modelos Animais de Doenças , Neoplasias Pulmonares/metabolismo , Neoplasias Experimentais/metabolismo , Proteína Oncogênica p21(ras)/metabolismo , Transdução de Sinais , Animais , Feminino , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Proteína Oncogênica p21(ras)/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Transdução de Sinais/genética
3.
Proc Natl Acad Sci U S A ; 106(48): 20423-8, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19918056

RESUMO

Systemic lupus erythematosus (SLE) is a complex autoimmune disease affecting various tissues. Involvement of B and T cells as well as increased cytokine levels have been associated with disease manifestation. Recently, we demonstrated that mice with epidermal loss of JunB (JunB(Deltaep)) develop a myeloproliferative syndrome (MPS) due to high levels of G-CSF which are secreted by JunB-deficient keratinocytes. In addition, we show that JunB(Deltaep) mice develop a SLE phenotype linked to increased epidermal interleukin 6 (IL-6) secretion. Intercrosses with IL-6-deficient mice could rescue the SLE phenotype. Furthermore, we show that JunB binds to the IL-6 promoter and transcriptionally suppresses IL-6. Facial skin biopsies of human SLE patients similarly revealed low JunB protein expression and high IL-6, activated Stat3, Socs-1, and Socs-3 levels within lupus lesions. Thus, keratinocyte-induced IL-6 secretion can cause SLE and systemic autoimmunity. Our results support trials to use alpha-IL-6 receptor antibody therapy for treatment of SLE.


Assuntos
Epiderme/metabolismo , Regulação da Expressão Gênica/fisiologia , Queratinócitos/metabolismo , Lúpus Eritematoso Sistêmico/patologia , Proteínas Proto-Oncogênicas c-jun/deficiência , Animais , Imunoprecipitação da Cromatina , Cruzamentos Genéticos , Ensaio de Imunoadsorção Enzimática , Técnica Direta de Fluorescência para Anticorpo , Fator Estimulador de Colônias de Granulócitos/metabolismo , Humanos , Interleucina-6/metabolismo , Luciferases , Lúpus Eritematoso Sistêmico/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Proteínas Proto-Oncogênicas c-jun/metabolismo
4.
Nature ; 437(7057): 369-75, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16163348

RESUMO

Psoriasis is a frequent, inflammatory disease of skin and joints with considerable morbidity. Here we report that in psoriatic lesions, epidermal keratinocytes have decreased expression of JunB, a gene localized in the psoriasis susceptibility region PSORS6. Likewise, inducible epidermal deletion of JunB and its functional companion c-Jun in adult mice leads (within two weeks) to a phenotype resembling the histological and molecular hallmarks of psoriasis, including arthritic lesions. In contrast to the skin phenotype, the development of arthritic lesions requires T and B cells and signalling through tumour necrosis factor receptor 1 (TNFR1). Prior to the disease onset, two chemotactic proteins (S100A8 and S100A9) previously mapped to the psoriasis susceptibility region PSORS4, are strongly induced in mutant keratinocytes in vivo and in vitro. We propose that the abrogation of JunB/activator protein 1 (AP-1) in keratinocytes triggers chemokine/cytokine expression, which recruits neutrophils and macrophages to the epidermis thereby contributing to the phenotypic changes observed in psoriasis. Thus, these data support the hypothesis that epidermal alterations are sufficient to initiate both skin lesions and arthritis in psoriasis.


Assuntos
Artrite Psoriásica/genética , Epiderme/metabolismo , Deleção de Genes , Genes jun/genética , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Envelhecimento/fisiologia , Animais , Artrite Psoriásica/metabolismo , Artrite Psoriásica/patologia , Quimiotaxia/efeitos dos fármacos , Citocinas/metabolismo , Regulação para Baixo , Humanos , Camundongos , Fenótipo , Proteínas Proto-Oncogênicas c-jun/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Proteínas S100/metabolismo , Transdução de Sinais , Linfócitos T/metabolismo , Linfócitos T/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
5.
Proc Natl Acad Sci U S A ; 105(30): 10525-30, 2008 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-18641127

RESUMO

Studies using genetically modified mice have revealed fundamental functions of the transcription factor Fos/AP-1 in bone biology, inflammation, and cancer. However, the biological role of the Fos-related protein Fra-2 is not well defined in vivo. Here we report an unexpected profibrogenic function of Fra-2 in transgenic mice, in which ectopic expression of Fra-2 in various organs resulted in generalized fibrosis with predominant manifestation in the lung. The pulmonary phenotype was characterized by vascular remodeling and obliteration of pulmonary arteries, which coincided with expression of osteopontin, an AP-1 target gene involved in vascular remodeling and fibrogenesis. These alterations were followed by inflammation; release of profibrogenic factors, such as IL-4, insulin-like growth factor 1, and CXCL5; progressive fibrosis; and premature mortality. Genetic experiments and bone marrow reconstitutions suggested that fibrosis developed independently of B and T cells and was not mediated by autoimmunity despite the marked inflammation observed in transgenic lungs. Importantly, strong expression of Fra-2 was also observed in human samples of idiopathic and autoimmune-mediated pulmonary fibrosis. These findings indicate that Fra-2 expression is sufficient to cause pulmonary fibrosis in mice, possibly by linking vascular remodeling and fibrogenesis, and suggest that Fra-2 has to be considered a contributing pathogenic factor of pulmonary fibrosis in humans.


Assuntos
Antígeno 2 Relacionado a Fos/biossíntese , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia , Fator de Transcrição AP-1/metabolismo , Animais , Quimiocina CXCL5/metabolismo , Feminino , Fibrose , Humanos , Inflamação , Fator de Crescimento Insulin-Like I/metabolismo , Interleucina-4/metabolismo , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Neoplasias/imunologia , Neoplasias/patologia
6.
Oncogene ; 40(6): 1091-1105, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33323974

RESUMO

Metastatic melanoma is hallmarked by its ability of phenotype switching to more slowly proliferating, but highly invasive cells. Here, we tested the impact of signal transducer and activator of transcription 3 (STAT3) on melanoma progression in association with melanocyte inducing transcription factor (MITF) expression levels. We established a mouse melanoma model for deleting Stat3 in melanocytes with specific expression of human hyperactive NRASQ61K in an Ink4a-deficient background, two frequent driver mutations in human melanoma. Mice devoid of Stat3 showed early disease onset with higher proliferation in primary tumors, but displayed significantly diminished lung, brain, and liver metastases. Whole-genome expression profiling of tumor-derived cells also showed a reduced invasion phenotype, which was further corroborated by 3D melanoma model analysis. Notably, loss or knockdown of STAT3 in mouse or human cells resulted in the upregulation of MITF and induction of cell proliferation. Mechanistically we show that STAT3-induced CAAT Box Enhancer Binding Protein (CEBP) expression was sufficient to suppress MITF transcription. Epigenetic analysis by ATAC-seq confirmed that CEBPa/b binding to the MITF enhancer region silenced the MITF locus. Finally, by classification of patient-derived melanoma samples, we show that STAT3 and MITF act antagonistically and hence contribute differentially to melanoma progression. We conclude that STAT3 is a driver of the metastatic process in melanoma and able to antagonize MITF via direct induction of CEBP family member transcription.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/genética , Melanoma/genética , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição STAT3/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Melanócitos/efeitos dos fármacos , Melanoma/patologia , Camundongos , Metástase Neoplásica , Transdução de Sinais/efeitos dos fármacos
7.
Dev Cell ; 4(6): 879-89, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12791272

RESUMO

To investigate the function of c-Jun during skin development and skin tumor formation, we conditionally inactivated c-jun in the epidermis. Mice lacking c-jun in keratinocytes (c-jun(Deltaep)) develop normal skin but express reduced levels of EGFR in the eyelids, leading to open eyes at birth, as observed in EGFR null mice. Primary keratinocytes from c-jun(Deltaep) mice proliferate poorly, show increased differentiation, and form prominent cortical actin bundles, most likely because of decreased expression of EGFR and its ligand HB-EGF. In the absence of c-Jun, tumor-prone K5-SOS-F transgenic mice develop smaller papillomas, with reduced expression of EGFR in basal keratinocytes. Thus, using three experimental systems, we show that EGFR and HB-EGF are regulated by c-Jun, which controls eyelid development, keratinocyte proliferation, and skin tumor formation.


Assuntos
Receptores ErbB/metabolismo , Pálpebras/embriologia , Genes jun , Papiloma/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Animais , Apoptose/genética , Carcinógenos/farmacologia , Divisão Celular , Células Epidérmicas , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Epiderme/lesões , Receptores ErbB/genética , Pálpebras/anormalidades , Pálpebras/metabolismo , Pálpebras/ultraestrutura , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Papiloma/etiologia , Papiloma/patologia , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/patologia , Acetato de Tetradecanoilforbol/farmacologia , Transgenes
8.
Biotechniques ; 43(5): 659-60, 662, 664, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18072595

RESUMO

The use of bacterial artificial chromosomes (BACs) modified via homologous recombination in Escherichia coli has become a powerful tool in the transgenic field. Homologous recombination allows the manipulation of BACs in very different ways. However this process can be cumbersome and problematic when using large targeting constructs containing several repeated elements. In order to address this problem, we have established a phiC31 integrase-mediated cassette exchange into a BAC. As an example of this technique, we have exchanged a cassette previously recombined into a BAC containing the Rosa 26 locus, by a 16.5-kb incoming construct containing several repeated elements. The combination of homologous recombination in E. coli and cassette exchange should expand the tools for manipulating BACs, thus facilitating the generation of constructs with higher complexity.


Assuntos
Bacteriófagos/enzimologia , Cromossomos Artificiais Bacterianos/genética , Integrases/metabolismo , Mutagênese Insercional/métodos
9.
Int J Biochem Cell Biol ; 38(7): 1043-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16423552

RESUMO

The Jun proteins Jun, JunB and JunD are core members of activator protein-1 (AP-1), a dimeric transcription factor complex consisting of homo- and heterodimers of the Jun, Fos, activating transcription factor (ATF) and musculoaponeurotic fibrosarcoma (Maf) families. Growth factors, hormones and a variety of environmental stresses activate mitogen activated protein kinase (MAPK) cascades that enhance Jun/AP-1 activity, e.g. through phosphorylation thereby regulating cell proliferation, differentiation, transformation and/or apoptosis. Embryonic lethality of various AP-1 knock-outs, e.g. for Jun, JunB, Fra-1 and Fra-2 largely prevented functional studies in vivo. Therefore, conditional knock-out strategies, in particular for the epidermis, have become an important model to study the regulation and function of AP-1 subunits in physiological and pathological processes in vivo. Jun is regarded as a positive regulator of keratinocyte proliferation/differentiation during development and in skin cancer through its direct transcriptional effect on epidermal growth factor receptor (EGFR) expression. In contrast, JunB can antagonize proliferation of keratinocytes and hematopoietic stem cells. Furthermore, it has been demonstrated in patient's samples and an inducible mouse model that down-regulation of JunB/AP-1 in keratinocytes is one initiating event in the aetiology of psoriasis which is characterized by increased cell proliferation and deregulated cytokine expression.


Assuntos
Epiderme/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Psoríase/fisiopatologia , Neoplasias Cutâneas/fisiopatologia , Fatores Ativadores da Transcrição/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Biologia do Desenvolvimento , Humanos , Queratinócitos/metabolismo , MAP Quinase Quinase 1/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Transdução de Sinais/fisiologia , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/metabolismo
10.
Nat Cell Biol ; 16(10): 972-7, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25173978

RESUMO

Hepatocellular carcinoma (HCC) is a frequent cancer with limited treatment options and poor prognosis. Tumorigenesis has been linked with macrophage-mediated chronic inflammation and diverse signalling pathways, including the epidermal growth factor receptor (EGFR) pathway. The precise role of EGFR in HCC is unknown, and EGFR inhibitors have shown disappointing clinical results. Here we discover that EGFR is expressed in liver macrophages in both human HCC and in a mouse HCC model. Mice lacking EGFR in macrophages show impaired hepatocarcinogenesis, whereas mice lacking EGFR in hepatocytes unexpectedly develop more HCC owing to increased hepatocyte damage and compensatory proliferation. Mechanistically, following interleukin-1 stimulation, EGFR is required in liver macrophages to transcriptionally induce interleukin-6, which triggers hepatocyte proliferation and HCC. Importantly, the presence of EGFR-positive liver macrophages in HCC patients is associated with poor survival. This study demonstrates a tumour-promoting mechanism for EGFR in non-tumour cells, which could lead to more effective precision medicine strategies.


Assuntos
Carcinoma Hepatocelular/metabolismo , Receptores ErbB/metabolismo , Neoplasias Hepáticas/metabolismo , Fígado/metabolismo , Macrófagos/metabolismo , Animais , Western Blotting , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/genética , Células Cultivadas , Dietilnitrosamina , Receptores ErbB/genética , Hepatócitos/metabolismo , Humanos , Imuno-Histoquímica , Interleucina-1beta/farmacologia , Células de Kupffer/efeitos dos fármacos , Células de Kupffer/metabolismo , Fígado/patologia , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/genética , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos
11.
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
12.
Nat Cell Biol ; 10(8): 1003-11, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18641637

RESUMO

Mice that lack JunB in epidermal cells are born with normal skin; however, keratinocytes hyperproliferate in vitro and on TPA treatment in vivo. Loss of JunB expression in the epidermis of adult mice affects the skin, the proliferation of haematopoietic cells and bone formation. G-CSF is a direct transcriptional target of JunB and mutant epidermis releases large amounts of G-CSF that reach high systemic levels and cause skin ulcerations, myeloproliferative disease and low bone mass. The absence of G-CSF significantly improves hyperkeratosis and prevents the development of myeloproliferative disease, but does not affect bone loss. This study describes a mechanism by which the absence of JunB in epithelial cells causes multi-organ disease, suggesting that the epidermis can act as an endocrine-like organ.


Assuntos
Epiderme , Fator Estimulador de Colônias de Granulócitos/genética , Hematopoese , Osteogênese , Proteínas Proto-Oncogênicas c-jun/fisiologia , Transcrição Gênica , Animais , Camundongos , Camundongos Mutantes , Proteínas Repressoras
13.
Arthritis Res Ther ; 10(1): 201, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18226189

RESUMO

Activator protein 1 (AP-1) (Fos/Jun) is a transcriptional regulator composed of members of the Fos and Jun families of DNA binding proteins. The functions of AP-1 were initially studied in mouse development as well as in the whole organism through conventional transgenic approaches, but also by gene targeting using knockout strategies. The importance of AP-1 proteins in disease pathways including the inflammatory response became fully apparent through conditional mutagenesis in mice, in particular when employing gene inactivation in a tissue-specific and inducible fashion. Besides the well-documented roles of Fos and Jun proteins in oncogenesis, where these genes can function both as tumor promoters or tumor suppressors, AP-1 proteins are being recognized as regulators of bone and immune cells, a research area termed osteoimmunology. In the present article, we review recent data regarding the functions of AP-1 as a regulator of cytokine expression and an important modulator in inflammatory diseases such as rheumatoid arthritis, psoriasis and psoriatic arthritis. These new data provide a better molecular understanding of disease pathways and should pave the road for the discovery of new targets for therapeutic applications.


Assuntos
Doenças Ósseas/metabolismo , Inflamação/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Dermatopatias/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Artrite Psoriásica/metabolismo , Artrite Reumatoide/metabolismo , Citocinas/metabolismo , Epiderme , Humanos , Psoríase/metabolismo
14.
Genesis ; 45(7): 447-51, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17607691

RESUMO

Loss of function mouse models comprise knock-out mice, where a gene is deleted in the germline, and conditional knock-out mice with somatic deletion of a floxed allele in defined tissues. Both types of mice are used for comprehensive studies of gene functions in vivo. Here, we describe a simple method for simultaneous generation of mice with conditional or knock-out alleles for the transcription factor fra-2 (Fos-related antigen 2) using a single embryonic stem (ES) cell clone. ES cells with a floxed fra-2 allele were transiently transfected with a Cre-recombinase expression plasmid and plated at low density. Most of the resulting ES cell colonies consisted of a mixture of cells that have either retained or lost the conditional allele. We demonstrate that these mixed ES cell clones can be directly used for generation of chimeras that give rise to offspring with conditional or knock-out alleles simultaneously. This strategy shortens the time and reduces the number of germline transmission events to generate genetically modified mice.


Assuntos
Antígeno 2 Relacionado a Fos/deficiência , Antígeno 2 Relacionado a Fos/genética , Genes Letais , Animais , Camundongos , Camundongos Knockout
15.
Cell ; 112(2): 181-92, 2003 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-12553907

RESUMO

The transcription factor c-Jun mediates several cellular processes, including proliferation and survival, and is upregulated in many carcinomas. Liver-specific inactivation of c-Jun at different stages of tumor development was used to study its role in chemically induced hepatocellular carcinomas (HCCs) in mice. The requirement for c-jun was restricted to early stages of tumor development, and the number and size of hepatic tumors was dramatically reduced when c-jun was inactivated after the tumor had initiated. The impaired tumor development correlated with increased levels of p53 and its target gene noxa, resulting in the induction of apoptosis without affecting cell proliferation. Primary hepatocytes lacking c-Jun showed increased sensitivity to TNF-alpha-induced apoptosis, which was abrogated in the absence of p53. These data indicate that c-Jun prevents apoptosis by antagonizing p53 activity, illustrating a mechanism that might contribute to the early stages of human HCC development.


Assuntos
Apoptose , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Fatores Etários , Animais , Divisão Celular , Sobrevivência Celular , Transformação Celular Neoplásica , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Mutação , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa