Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
1.
Oncogene ; 36(35): 4997-5005, 2017 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-28459465

RESUMO

Inturned (INTU), a cilia and planar polarity effector, performs prominent ciliogenic functions during morphogenesis, such as in the skin. INTU is expressed in adult tissues but its role in tissue maintenance is unknown. Here, we report that the expression of the INTU gene is aberrantly elevated in human basal cell carcinoma (BCC), coinciding with increased primary cilia formation and activated hedgehog (Hh) signaling. Disrupting Intu in an oncogenic mutant Smo (SmoM2)-driven BCC mouse model prevented the formation of BCC through suppressing primary cilia formation and Hh signaling, suggesting that Intu performs a permissive role during BCC formation. INTU is essential for intraflagellar transport A complex assembly during ciliogenesis. To further determine whether Intu is directly involved in the activation of Hh signaling downstream of ciliogenesis, we examined the Hh signaling pathway in mouse embryonic fibroblasts, which readily responds to the Hh pathway activation. Depleting Intu blocked Smo agonist-induced Hh pathway activation, whereas the expression of Gli2ΔN, a constitutively active Gli2, restored Hh pathway activation in Intu-deficient cells, suggesting that INTU functions upstream of Gli2 activation. In contrast, overexpressing Intu did not promote ciliogenesis or Hh signaling. Taken together, data obtained from this study suggest that INTU is indispensable during BCC tumorigenesis and that its aberrant upregulation is likely a prerequisite for primary cilia formation during Hh-dependent tumorigenesis.


Assuntos
Carcinoma Basocelular/metabolismo , Cílios/metabolismo , Cílios/patologia , Proteínas Hedgehog/metabolismo , Proteínas de Membrana/genética , Neoplasias Cutâneas/metabolismo , Animais , Carcinoma Basocelular/genética , Carcinoma Basocelular/patologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Proteínas Hedgehog/genética , Humanos , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Transfecção
2.
Oncogene ; 35(3): 290-300, 2016 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-25893296

RESUMO

The limitations of cancer cell lines have led to the development of direct patient-derived xenograft models. However, the interplay between the implanted human cancer cells and recruited mouse stromal and immune cells alters the tumor microenvironment and limits the value of these models. To overcome these constraints, we have developed a technique to expand human hematopoietic stem and progenitor cells (HSPCs) and use them to reconstitute the radiation-depleted bone marrow of a NOD/SCID/IL2rg(-/-) (NSG) mouse on which a patient's tumor is then transplanted (XactMice). The human HSPCs produce immune cells that home into the tumor and help replicate its natural microenvironment. Despite previous passage on nude mice, the expression of epithelial, stromal and immune genes in XactMice tumors aligns more closely to that of the patient tumor than to those grown in non-humanized mice-an effect partially facilitated by human cytokines expressed by both the HSPC progeny and the tumor cells. The human immune and stromal cells produced in the XactMice can help recapitulate the microenvironment of an implanted xenograft, reverse the initial genetic drift seen after passage on non-humanized mice and provide a more accurate tumor model to guide patient treatment.


Assuntos
Neoplasias de Cabeça e Pescoço/genética , Células-Tronco Hematopoéticas/patologia , Microambiente Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Medula Óssea/patologia , Linhagem Celular Tumoral , Citocinas/biossíntese , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Camundongos
3.
Oncogene ; 31(21): 2680-90, 2012 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-21963848

RESUMO

Clinical observations, as well as data obtained from the analysis of genetically engineered mouse models, firmly established the gain-of-function (GOF) properties of certain p53 mutations. However, little is known about the underlying mechanisms. We have used two independent microarray platforms to perform a comprehensive and global analysis of tumors arising in a model of metastatic skin cancer progression, which compares the consequences of a GOF p53(R172H) mutant vs p53 deficiency. DNA profiling revealed a higher level of genomic instability in GOF vs loss-of-function (LOF) p53 squamous cell carcinomas (SCCs). Moreover, GOF p53 SCCs showed preferential amplification of Myc with a corresponding increase in its expression and deregulation of Aurora Kinase A. Fluorescent in situ hybridization confirmed amplification of Myc in primary GOF p53 SCCs and its retention in metastatic tumors. We also identified by RNA profiling distinct gene expression profiles in GOF p53 tumors, which included enriched integrin and Rho signaling, independent of tumor stage. Thus, the progression of GOF p53 papillomas to carcinoma was marked by the acquisition of epithelial-to-mesenchymal transition and metastatic signatures. In contrast, LOF p53 tumors showed enrichment of genes associated with cancer proliferation and chromosomal instability. Collectively, these observations suggest that genomic instability has a prominent role in the early stages of GOF p53 tumor progression (that is, papillomas), whereas it is implicated at a later stage in LOF p53 tumors (that is, SCCs). This model will allow us to identify specific targets in mutant p53 SCCs, which may lead to the development of new therapeutic agents for the treatment of metastatic SCCs.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/secundário , Genes myc , Mutação , Papiloma/genética , Proteínas Serina-Treonina Quinases/metabolismo , Neoplasias Cutâneas/genética , Proteína Supressora de Tumor p53 , Animais , Aurora Quinase A , Aurora Quinases , Modelos Animais de Doenças , Progressão da Doença , Transição Epitelial-Mesenquimal , Amplificação de Genes , Instabilidade Genômica , Integrinas/genética , Camundongos , Neoplasias Cutâneas/patologia , Regulação para Cima
4.
Oncogene ; 30(43): 4399-409, 2011 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-21532625

RESUMO

Progenitor cells are considered an important cell of origin of human malignancies. However, there has not been any single gene that can define mammary bipotential progenitor cells, and as such it has not been possible to use genetic methods to introduce oncogenic alterations into these cells in vivo to study tumorigenesis from them. Keratin 6a is expressed in a subset of mammary luminal epithelial cells and body cells of terminal end buds. By generating transgenic mice using the Keratin 6a (K6a) gene promoter to express tumor virus A (tva), which encodes the receptor for avian leukosis virus subgroup A (ALV/A), we provide direct evidence that K6a(+) cells are bipotential progenitor cells, and the first demonstration of a non-basal location for some biopotential progenitor cells. These K6a(+) cells were readily induced to form mammary tumors by intraductal injection of RCAS (an ALV/A-derived vector) carrying the gene encoding the polyoma middle T antigen. Tumors in this K6a-tva line were papillary and resembled the normal breast-like subtype of human breast cancer. This is the first model of this subtype of human tumors and thus may be useful for preclinical testing of targeted therapy for patients with normal-like breast cancer. These observations also provide direct in vivo evidence for the hypothesis that the cell of origin affects mammary tumor phenotypes.


Assuntos
Neoplasias da Mama/metabolismo , Modelos Animais de Doenças , Queratina-6/metabolismo , Neoplasias Experimentais/metabolismo , Células-Tronco/metabolismo , Animais , Vírus da Leucose Aviária/genética , Transição Epitelial-Mesenquimal , Feminino , Humanos , Queratina-6/genética , Camundongos , Camundongos Transgênicos
6.
Oncogene ; 25(29): 4027-32, 2006 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-16518415

RESUMO

Minichromosomal maintenance protein 7 (MCM7) is an essential component of the replication helicase complex (MCM2-7) required for DNA replication. Although this function is highly conserved among eukaryotes, additional functions for the MCM molecules continue to be described. Minichromosomal maintenance protein 7 is a marker for proliferation and is upregulated in a variety of tumors including neuroblastoma, prostate, cervical and hypopharyngeal carcinomas. To further investigate the general role of MCM7 in tumorigenesis, we generated a mouse model with deregulated MCM7 expression targeted to the basal layer of the epidermis using the keratin 14 (K14) promoter (K14.MCM7). When subjected to a two-stage chemical carcinogenesis protocol (dimethylbenz[alpha]anthracene (DMBA) initiation with 12-ortho-tetradecanoylphorbol-13-acetate promotion), K14.MCM7 mice showed significantly increased incidence and prevalence of tumor development relative to controls. Furthermore, within 40 weeks of treatment over 45% K14.MCM7 mice exhibited tumors that had converted to squamous cell carcinomas versus none in the control group. As predicted from previous skin carcinogenesis studies using DMBA as the initiating agent, Ras mutations where found in more than 90% of tumors isolated from K14.MCM7 mice. Whereas previous studies have shown that MCM7 is useful as a proliferation marker, our data suggest that deregulated MCM7 expression actively contributes to tumor formation, progression and malignant conversion.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/genética , Proteínas de Ciclo Celular/genética , Transformação Celular Neoplásica/genética , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Neoplasias Cutâneas/genética , 9,10-Dimetil-1,2-benzantraceno/farmacologia , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Biomarcadores Tumorais/biossíntese , Carcinógenos/farmacologia , Carcinógenos/toxicidade , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Proteínas de Ciclo Celular/biossíntese , Proliferação de Células , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/metabolismo , Proteínas de Ligação a DNA/biossíntese , Genes ras/genética , Humanos , Camundongos , Camundongos Transgênicos , Componente 7 do Complexo de Manutenção de Minicromossomo , Mutação , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Proteínas Nucleares/biossíntese , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia , Acetato de Tetradecanoilforbol/toxicidade
7.
Trends Mol Med ; 7(9): 422-4, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11530338

RESUMO

Hereditary skin blistering disorders comprise a group of genodermatoses whose common primary feature is the formation of blisters following minor trauma. Examples of such conditions include epidermolysis bullosa and several bullous forms of ichthyosis. Distinct mutations in various genes encoding intra- and extra-cellular structural components of the skin reflect the clinical heterogeneity of these disorders. Several animal models are currently used to study the role of these molecules in the disease process. Some of these models will find their place in evaluating new therapeutic strategies for this devastating group of diseases.


Assuntos
Vesícula/genética , Modelos Animais de Doenças , Animais , Vesícula/metabolismo , Vesícula/patologia , Humanos , Hiperceratose Epidermolítica/genética , Hiperceratose Epidermolítica/metabolismo , Hiperceratose Epidermolítica/patologia , Camundongos , Camundongos Knockout , Camundongos Mutantes , Mutação/genética , Fenótipo
8.
J Natl Cancer Inst ; 93(15): 1166-70, 2001 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-11481389

RESUMO

BACKGROUND: The phase II enzyme NAD(P)H :quinone oxidoreductase 1 (NQO1) catalyzes quinone detoxification, protecting cells from redox cycling, oxidative stress, mutagenicity, and cytotoxicity induced by quinones and its precursors. We have used NQO1(-/-) C57BL/6 mice to show that NQO1 protects them from skin cancer induced by the polycyclic aromatic hydrocarbon benzo[a]pyrene. Herein, we used NQO1(-/-) mice to investigate whether NQO1 also protects them against 7,12-dimethylbenz[a]anthracene (DMBA), where methyl substituents diminish primary quinone formation. METHODS: Dorsal skin of NQO1(-/-) or wild-type C57BL/6 mice was shaved. When tested as a complete carcinogen, DMBA (500 or 750 microg in 100 microL of acetone) alone was applied to the shaved area. When tested as a tumor initiator, DMBA (200 or 400 nmol in 100 microL of acetone) was applied to the shaved area; 1 week later, twice-weekly applications of phorbol 12-myristate 13-acetate (PMA)-10 microg dissolved in 200 microL of acetone-to the same area began and were continued for 20 weeks. Tumor development was monitored in all mice (12-15 per group). All statistical tests were two-sided. RESULTS: When DMBA (750 microg) was tested as a complete carcinogen, about 50% of the DMBA-treated NQO1(-/-) mice but no DMBA-treated wild-type mouse developed skin tumors. When DMBA (both concentrations) was used as a tumor initiator, NQO1(-/-) mice developed larger tumors at a greater frequency than their wild-type littermates. Twenty-three weeks after the first PMA treatment in the tumor initiator test, all 30 NQO1(-/-) mice given 400 nmol of DMBA had developed skin tumors, compared with 33% (10 of 30) of treated wild-type mice (P<.001). CONCLUSIONS: NQO1(-/-) mice are more susceptible to DMBA-induced skin cancer than are their wild-type littermates, suggesting that NQO1 may protect cells from DMBA carcinogenesis.


Assuntos
Quinona Redutases/deficiência , Neoplasias Cutâneas/enzimologia , 9,10-Dimetil-1,2-benzantraceno , Animais , Carcinógenos , Suscetibilidade a Doenças/enzimologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Cutâneas/induzido quimicamente
9.
J Cell Biol ; 154(3): 619-30, 2001 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-11489919

RESUMO

The murine genome is known to have two keratin 6 (K6) genes, mouse K6 (MK6)a and MK6b. These genes display a complex expression pattern with constitutive expression in the epithelia of oral mucosa, hair follicles, and nail beds. We generated mice deficient for both genes through embryonic stem cell technology. The majority of MK6a/b-/- mice die of starvation within the first two weeks of life. This is due to a localized disintegration of the dorsal tongue epithelium, which results in the build up of a plaque of cell debris that severely impairs feeding. However, approximately 25% of MK6a/b-/- mice survive to adulthood. Remarkably, the surviving MK6a/b-/- mice have normal hair and nails. To our surprise, we discovered MK6 staining both in the hair follicle and the nail bed of MK6a/b-/- mice, indicating the presence of a third MK6 gene. We cloned this previously unknown murine keratin gene and found it to be highly homologous to human K6hf, which is expressed in hair follicles. We therefore termed this gene MK6 hair follicle (MK6hf). The presence of MK6hf in the MK6a/b-/- follicles and nails offers an explanation for the absence of hair and nail defects in MK6a/b-/- animals.


Assuntos
Doenças do Cabelo/genética , Doenças do Cabelo/patologia , Queratinas/genética , Doenças da Unha/genética , Doenças da Unha/patologia , Animais , Células Epiteliais/patologia , Deleção de Genes , Doenças do Cabelo/mortalidade , Hiperplasia , Isomerismo , Queratinas/química , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Dados de Sequência Molecular , Doenças da Boca/genética , Doenças da Boca/mortalidade , Doenças da Boca/patologia , Doenças da Unha/mortalidade , Fenótipo , Homologia de Sequência de Aminoácidos , Pele/patologia , Inanição/genética , Inanição/mortalidade , Inanição/patologia , Língua/patologia , Língua/ultraestrutura , Cicatrização/genética
10.
Nat Genet ; 28(2): 165-8, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11381265

RESUMO

The beta-catenin/TCF signaling pathway is essential for the maintenance of epithelial stem cells in the small intestine. c-Myc a downstream target of beta-catenin/TCF (ref. 2), can induce differentiation of epidermal stem cells in vitro. To determine the role of c-Myc in epidermal stem cells in vivo, we have targeted expression of human MYC2 to the hair follicles and the basal layer of mouse epidermis using a keratin 14 vector (K14.MYC2). Adult K14.MYC2 mice gradually lose their hair and develop spontaneous ulcerated lesions due to a severe impairment in wound healing; their keratinocytes show impaired migration in response to wounding. The expression of beta1 integrin, which is preferentially expressed in epidermal stem cells is unusually low in the epidermis of K14.MYC2 mice. Label-retaining analysis to identify epidermal stem cells reveals a 75% reduction in the number of stem cells in 3-month-old K14.MYC2 mice, compared with wildtype mice. We conclude that deregulated expression of c-Myc in stem cells reduces beta1 integrin expression, which is essential to both keratinocyte migration and stem cell maintenance.


Assuntos
Proteínas Proto-Oncogênicas c-myc/genética , Pele/patologia , Células-Tronco/fisiologia , Animais , Bromodesoxiuridina/metabolismo , Divisão Celular/genética , Movimento Celular/genética , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Hiperplasia/genética , Integrina beta1/genética , Integrina beta1/metabolismo , Queratina-14 , Queratinócitos/patologia , Queratinas/genética , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Pele/embriologia , Células-Tronco/patologia , Cicatrização/genética
11.
J Cell Biol ; 152(3): 645-9, 2001 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-11157989

RESUMO

Stem cells are crucial for the formation and maintenance of tissues and organs. The role of stem cells in the pathogenesis of mosaic skin disorders remains unclear. To study the molecular and cellular basis of mosaicism, we established a mouse model for the autosomal-dominant skin blistering disorder, epidermolytic hyperkeratosis (MIM 113800), which is caused by mutations in either keratin K1 or K10. This genetic model allows activation of a somatic K10 mutation in epidermal stem cells in a spatially and temporally controlled manner using an inducible Cre recombinase. Our results indicate that lack of selective pressure against certain mutations in epidermal stem cells leads to mosaic phenotypes. This finding has important implications for the development of new strategies for somatic gene therapy of dominant genodermatoses.


Assuntos
Hiperceratose Epidermolítica/genética , Queratinas/genética , Mosaicismo/genética , Mutação Puntual/genética , Pele/patologia , Células-Tronco/fisiologia , Proteínas Virais , Animais , Modelos Animais de Doenças , Feminino , Marcação de Genes , Humanos , Hiperceratose Epidermolítica/patologia , Hiperceratose Epidermolítica/fisiopatologia , Integrases/genética , Integrases/metabolismo , Queratina-10 , Queratinas/metabolismo , Camundongos , Camundongos Transgênicos , Mifepristona/farmacologia , Pele/efeitos dos fármacos , Pele/fisiopatologia
12.
J Cell Biol ; 152(3): 651-6, 2001 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-11157990

RESUMO

The Dowling-Meara variant of epidermolysis bullosa simplex (EBS-DM) is a severe blistering disease inherited in an autosomal-dominant fashion. Here we report the generation of a mouse model that allows focal activation of a mutant keratin 14 allele in epidermal stem cells upon topical administration of an inducer, resulting in EBS phenotypes in treated areas. Using laser capture microdissection, we show that induced blisters healed by migration of surrounding nonphenotypic stem cells into the wound bed. This observation provides an explanation for the lack of mosaic forms of EBS-DM. In addition, we show that decreased mutant keratin 14 expression resulted in normal morphology and functions of the skin. Our results have important implications for gene therapy of EBS and other dominantly inherited diseases.


Assuntos
Modelos Animais de Doenças , Epidermólise Bolhosa Simples/genética , Regulação da Expressão Gênica , Queratinas/genética , Pele/fisiopatologia , Proteínas Virais , Animais , Southern Blotting , Epidermólise Bolhosa Simples/patologia , Epidermólise Bolhosa Simples/fisiopatologia , Epidermólise Bolhosa Simples/terapia , Terapia Genética , Humanos , Integrases/genética , Integrases/metabolismo , Queratina-14 , Queratinas/metabolismo , Luteolíticos/farmacologia , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Mifepristona/administração & dosagem , Mifepristona/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/efeitos dos fármacos , Pele/ultraestrutura
13.
Hum Gene Ther ; 11(16): 2297-300, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11084689

RESUMO

Epidermal keratinocytes can secrete polypeptides into the bloodstream, and they can be easily expanded in culture and genetically modified. It is thus possible to use epidermal keratinocytes for the systemic delivery of transgene products. Here we review the development of epidermal secretory systems, from cultured keratinocytes to skin grafts and transgenic mouse models. We also discuss a gene-switch approach for regulated cutaneous gene delivery.


Assuntos
Epiderme/metabolismo , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Queratinócitos/metabolismo , Neoplasias/terapia , Dermatopatias/terapia , Pele/metabolismo , Transgenes , Animais , Humanos , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Dermatopatias/genética
14.
Mol Carcinog ; 29(2): 67-75, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11074603

RESUMO

To investigate the role of loss of the p53 tumor suppressor gene in skin carcinogenesis, p53 knockout (p53(-/-)) mice were mated with transgenic mice coexpressing v-Ha-ras, v-fos, or human transforming growth factor alpha (TGFalpha) exclusively in the epidermis by using human keratin 1 (HK1)-based vectors (HK1.ras/fos, HK1.ras/alpha, and HK1.fos/alpha). HK1.ras/fos and HK1.ras/alpha mice displayed epidermal hyperplasia and autonomous benign papillomas to an identical degree between p53(+/+) and p53(+/-) genotypes. However, HK1.ras/fos mice with the p53(-/-) genotype were born with papillomatous skin and died soon after birth. HK1.ras/alpha-p53(-/-) mice also exhibited an increased epidermal hyperplasia, and, similar to HK1.ras/alpha mice with p53(+/+) and p53(+/-) genotypes, these mice rapidly developed spontaneous and 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced papillomas. These results are in contrast to our previous observation that, HK1.ras, HK1.fos, and HK1.TGFalpha transgenic mice with the p53(-/-) genotype display an unexpected delay in both spontaneous and TPA-promoted papilloma formation compared with mice with p53(+/+) and p53(+/-) genotypes. Taken collectively, our mating experiments between HK1 oncogenic transgenic mice and p53 knockout mice may identify a backup system that effectively compensates for p53 loss. Activation of multiple oncogenes not only partly overcomes such compensation but also synergizes with p53 loss. However, HK1.fos/alpha-p53(-/-) mice failed to exhibit either an increased newborn epidermal hyperplasia or an accelerated spontaneous or TPA-induced papillomas, suggesting that certain combinations of oncogenes, such as with activated Ha-ras, are required for this process. Because neither spontaneous nor TPA-elicited papillomas in p53(-/-) mice progressed to malignancy, additional genetic insults appear to be required for malignant progression.


Assuntos
Genes fos/fisiologia , Genes p53/fisiologia , Genes ras/fisiologia , Papiloma/genética , Neoplasias Cutâneas/genética , Fator de Crescimento Transformador alfa/genética , Animais , Carcinógenos , Epiderme/metabolismo , Epiderme/fisiologia , Feminino , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Queratinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Camundongos Transgênicos , Papiloma/induzido quimicamente , Neoplasias Cutâneas/induzido quimicamente , Acetato de Tetradecanoilforbol
15.
Cancer Res ; 60(21): 5913-5, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11085502

RESUMO

NAD(P)H:quinone oxidoreductase 1 (NQO1) is a flavoprotein that catalyzes the metabolic detoxification of quinones and their derivatives. This protects cells against quinone-induced oxidative stress, cytotoxicity, and mutagenicity. C57BL6 NQO1-/- mice, deficient in NQO1 RNA and protein, were generated in our laboratory. To investigate the role of NQO1 in chemical carcinogenesis, the dorsal skin of NQO1-deficient (NQO1-/-) and wild-type (NQO1+/+) mice were treated with a single dose of benzo(a)pyrene, followed by twice weekly applications of phorbol-12-myristate-13-acetate. The NQO1-/- mice showed a much higher frequency of skin tumor development when compared with their wild-type littermates. Interestingly, the male NQO1-/- mice were slower to develop skin tumors than their NQO1-/- female littermates. Histological analysis of the NQO1-/- tumors showed proliferative activity. These results demonstrate that NQO1 acts as an endogenous factor in protection against benzo(a)pyrene carcinogenicity.


Assuntos
Benzo(a)pireno/toxicidade , Carcinógenos/toxicidade , Cocarcinogênese , NAD(P)H Desidrogenase (Quinona)/deficiência , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/enzimologia , Animais , Benzo(a)pireno/farmacocinética , Biotransformação , Carcinógenos/farmacocinética , Suscetibilidade a Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos A , Modelos Animais , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Pele/efeitos dos fármacos , Pele/enzimologia , Acetato de Tetradecanoilforbol/toxicidade
16.
J Cell Biol ; 151(2): 401-12, 2000 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-11038186

RESUMO

Mutations in the cornified cell envelope protein loricrin have been reported recently in some patients with Vohwinkel syndrome (VS) and progressive symmetric erythrokeratoderma (PSEK). To establish a causative relationship between loricrin mutations and these diseases, we have generated transgenic mice expressing a COOH-terminal truncated form of loricrin that is similar to the protein expressed in VS and PSEK patients. At birth, transgenic mice (ML.VS) exhibited erythrokeratoderma with an epidermal barrier dysfunction. 4 d after birth, high-expressing transgenic animals showed a generalized scaling of the skin, as well as a constricting band encircling the tail and, by day 7, a thickening of the footpads. Histologically, ML. VS transgenic mice also showed retention of nuclei in the stratum corneum, a characteristic feature of VS and PSEK. Immunofluorescence and immunoelectron microscopy showed the mutant loricrin protein in the nucleus and cytoplasm of epidermal keratinocytes, but did not detect the protein in the cornified cell envelope. Transfection experiments indicated that the COOH-terminal domain of the mutant loricrin contains a nuclear localization signal. To determine whether the ML.VS phenotype resulted from dominant-negative interference of the transgene with endogenous loricrin, we mated the ML.VS transgenics with loricrin knockout mice. A severe phenotype was observed in mice that lacked expression of wild-type loricrin. Since loricrin knockout mice are largely asymptomatic (Koch, P.K., P. A. de Viragh, E. Scharer, D. Bundman, M.A. Longley, J. Bickenbach, Y. Kawachi, Y. Suga, Z. Zhou, M. Huber, et al., J. Cell Biol. 151:389-400, this issue), this phenotype may be attributed to expression of the mutant form of loricrin. Thus, deposition of the mutant protein in the nucleus appears to interfere with late stages of epidermal differentiation, resulting in a VS-like phenotype.


Assuntos
Surdez/etiologia , Ceratose/etiologia , Proteínas de Membrana/genética , Sequência de Aminoácidos , Animais , Compartimento Celular , Membrana Celular/química , Surdez/genética , Mutação da Fase de Leitura , Ceratose/genética , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Dados de Sequência Molecular , Sinais de Localização Nuclear , Fenótipo , Transporte Proteico , Pele/patologia , Fenômenos Fisiológicos da Pele/genética , Síndrome
17.
J Cell Biol ; 151(2): 389-400, 2000 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-11038185

RESUMO

The epidermal cornified cell envelope (CE) is a complex protein-lipid composite that replaces the plasma membrane of terminally differentiated keratinocytes. This lamellar structure is essential for the barrier function of the skin and has the ability to prevent the loss of water and ions and to protect from environmental hazards. The major protein of the epidermal CE is loricrin, contributing approximately 70% by mass. We have generated mice that are deficient for this protein. These mice showed a delay in the formation of the skin barrier in embryonic development. At birth, homozygous mutant mice weighed less than control littermates and showed skin abnormalities, such as congenital erythroderma with a shiny, translucent skin. Tape stripping experiments suggested that the stratum corneum stability was reduced in newborn Lor(-/-) mice compared with wild-type controls. Isolated mutant CEs were more easily fragmented by sonication in vitro, indicating a greater susceptibility to mechanical stress. Nevertheless, we did not detect impaired epidermal barrier function in these mice. Surprisingly, the skin phenotype disappeared 4-5 d after birth. At least one of the compensatory mechanisms preventing a more severe skin phenotype in newborn Lor(-/-) mice is an increase in the expression of other CE components, such as SPRRP2D and SPRRP2H, members of the family of "small proline rich proteins", and repetin, a member of the "fused gene" subgroup of the S100 gene family.


Assuntos
Epiderme/fisiologia , Proteínas de Membrana/genética , Fenômenos Fisiológicos da Pele/genética , Adaptação Biológica , Sequência de Aminoácidos , Animais , Fenômenos Biomecânicos , Membrana Celular , Clonagem Molecular , Proteínas Ricas em Prolina do Estrato Córneo , Proteínas de Filamentos Intermediários/biossíntese , Proteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Permeabilidade , Proteínas S100/biossíntese , Regulação para Cima
18.
Cancer Res ; 60(15): 4016-20, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10945600

RESUMO

In the epidermis, p53 plays an important role in UV-B protection that led us to examine the role, if any, that p63, a p53 homologue highly expressed in the basal layer of the epidermis, might play in the epidermal UV-B response. One p63 isoform, deltaNp63alpha, decreased dramatically in normal keratinocytes or newborn epidermis at both the protein and RNA levels after UV-B irradiation. In an attempt to further investigate the significance of the UV-B-induced decrease of this p63 isoform as well as further delineate the function of p63 in the epidermis, we generated transgenic mice that constitutively express deltaNp63alpha in the mouse epidermis using the loricrin promoter (ML.deltaNp63alpha). The ML.deltaNp63alpha mouse epidermis developed normally, with no overt phenotype and an unaltered proliferation rate. When challenged by UV-B exposure, the ML.deltaNp63alpha mice exhibited a 40-45% decrease in the number of apoptotic cells in the epidermis as compared with nontransgenic littermates. These results suggest that aberrant expression of deltaNp63alpha altered the UV-B-induced apoptotic pathway in the transgenic epidermis, proving that down-regulation of deltaNp63alpha in response to UV-B is important to epidermal apoptosis. The forced overexpression of deltaNp63alpha may act via a dominant negative effect on the endogenous p53 transcriptional activity required for UV-B-induced apoptosis.


Assuntos
Apoptose/efeitos da radiação , Epiderme/efeitos da radiação , Proteínas de Membrana , Fosfoproteínas/fisiologia , Transativadores , Raios Ultravioleta/efeitos adversos , Animais , Animais Recém-Nascidos , Apoptose/genética , Apoptose/fisiologia , Regulação para Baixo/efeitos da radiação , Células Epidérmicas , Epiderme/metabolismo , Expressão Gênica , Queratinócitos/metabolismo , Queratinócitos/efeitos da radiação , Camundongos , Camundongos Endogâmicos ICR , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Fenômenos Fisiológicos da Pele/genética , Fenômenos Fisiológicos da Pele/efeitos da radiação , Transgenes
19.
Mol Cell Biol ; 20(14): 5248-55, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10866680

RESUMO

Keratin 6 (K6) expression in the epidermis has two components: constitutive expression in the innermost layer of the outer root sheath (ORS) of hair follicles and inducible expression in the interfollicular epidermis in response to stressful stimuli such as wounding. Mice express two K6 isoforms, MK6a and MK6b. To gain insight into the functional significance of these isoforms, we generated MK6a-deficient mice through mouse embryonic stem cell technology. Upon wounding, MK6a was induced in the outer ORS and the interfollicular epidermis including the basal cell layer of MK6a(+/+) mice, whereas MK6b induction in MK6a(-/-) mice was restricted to the suprabasal layers of the epidermis. After superficial wounding of the epidermis by tape stripping, MK6a(-/-) mice showed a delay in reepithelialization from the hair follicle. However, the healing of full-thickness skin wounds was not impaired in MK6a(-/-) animals. Migration and proliferation of MK6a(-/-) keratinocytes were not impaired in vitro. Furthermore, the migrating and the proliferating keratinocytes of full-thickness wounds in MK6a(-/-) animals expressed neither MK6a nor MK6b. These data indicate that MK6a does not play a major role in keratinocyte proliferation or migration but point to a role in the activation of follicular keratinocytes after wounding. This study represents the first report of a keratin null mutation that results in a wound healing defect.


Assuntos
Queratinas/genética , Cicatrização/fisiologia , Animais , Divisão Celular , Movimento Celular/genética , Células Cultivadas , Epiderme/metabolismo , Células Epiteliais/patologia , Deleção de Genes , Engenharia Genética , Folículo Piloso/metabolismo , Folículo Piloso/patologia , Queratinócitos/citologia , Queratinócitos/patologia , Queratinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Recombinação Genética , Células-Tronco , Cicatrização/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...