Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Oncogene ; 27(40): 5373-84, 2008 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-18504437

RESUMO

Metastasis is the primary cause of death in patients with breast cancer. Overexpression of c-myc in humans correlates with metastases, but transgenic mice only show low rates of micrometastases. We have generated transgenic mice that overexpress both c-myc and vascular endothelial growth factor (VEGF) (Myc/VEGF) in the mammary gland, which develop high rates of pulmonary macrometastases. Gene expression profiling revealed a set of deregulated genes in Myc/VEGF tumors compared to Myc tumors associated with the increased metastatic phenotype. Cross-comparisons between this set of genes with a human breast cancer lung metastasis gene signature identified five common targets: tenascin-C(TNC), matrix metalloprotease-2, collagen-6-A1, mannosidase-alpha-1A and HLA-DPA1. Signaling blockade or knockdown of TNC in MDA-MB-435 cells resulted in a significant impairment of cell migration and anchorage-independent cell proliferation. Mice injected with clonal MDA-MB-435 cells with reduced expression of TNC demonstrated a significant decrease (P<0.05) in (1) primary tumor growth; (2) tumor relapse after surgical removal of the primary tumor and (3) incidence of lung metastasis. Our results demonstrate that VEGF induces complex alterations in tissue architecture and gene expression. The TNC signaling pathway plays an important role in mammary tumor growth and metastases, suggesting that TNC may be a relevant target for therapy against metastatic breast cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Tenascina/farmacologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Biomarcadores Tumorais/genética , Northern Blotting , Western Blotting , Adesão Celular , Movimento Celular , Proliferação de Células , Perfilação da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/prevenção & controle , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tenascina/antagonistas & inibidores , Tenascina/genética , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco
2.
Leukemia ; 19(5): 847-50, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15744335

RESUMO

GADD:45a-/- and p53-/- mice and cells derived from them share similar phenotypes, most notably genomic instability. However, p53-/- mice rapidly develop a variety of neoplasms, while Gadd45a-/- mice do not. The two proteins are involved in a regulatory feedback loop, whereby each can increase the expression or activity of the other, suggesting that common phenotypes might result from similar molecular mechanisms. Mice lacking both genes were generated to address this issue. Gadd45a-/-p53-/- mice developed tumors with a latency similar to that of tumor-prone p53-/- mice. However, while p53-/- mice developed a variety of tumor types, nearly all Gadd45a-/-p53-/- mice developed lymphoblastic lymphoma (LBL), often accompanied by mediastinal masses as is common in human patients with this tumor type. Deletion of Gadd45a in leukemia/lymphoma-prone AKR mice decreased the latency for LBL. These results indicate that Gadd45a may act as modifier locus for T-cell LBL, whereby deletion of Gadd45a enhances development of this tumor type in susceptible mice. Gadd45a is localized to 1p31.1, and 1p abnormalities have been described in T-cell lymphomas. Related human tumor samples did not show Gadd45a deletion or mutation, although changes in expression could not be ruled out.


Assuntos
Proteínas de Ciclo Celular/genética , Neoplasias Experimentais/genética , Proteínas Nucleares/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Fatores Etários , Alelos , Animais , Feminino , Humanos , Masculino , Camundongos , Neoplasias Experimentais/etiologia , Proteínas Nucleares/deficiência , Fenótipo , Análise de Sobrevida , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
3.
J Exp Med ; 194(12): 1731-41, 2001 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-11748275

RESUMO

We examined the effects of human immunodeficiency virus infection on the turnover of CD4 and CD8 T lymphocytes in 17 HIV-infected patients by 30 min in vivo pulse labeling with bromodeoxyuridine (BrdU). The percentage of labeled CD4 and CD8 T lymphocytes was initially higher in lymph nodes than in blood. Labeled cells equilibrated between the two compartments within 24 h. Based on mathematical modeling of the dynamics of BrdU-labeled cells in the blood, we identified rapidly and slowly proliferating subpopulations of CD4 and CD8 T lymphocytes. The percentage, but not the decay rate, of labeled CD4 or CD8 cells in the rapidly proliferating pool correlated significantly with plasma HIV RNA levels for both CD4 (r = 0.77, P < 0.001) and CD8 (r = 0.81, P < 0.001) T cells. In six patients there was a geometric mean decrease of greater than 2 logs in HIV levels within 2 to 6 mo after the initiation of highly active antiretroviral therapy; this was associated with a significant decrease in the percentage (but not the decay rate) of labeled cells in the rapidly proliferating pool for both CD4 (P = 0.03) and CD8 (P < 0.001) T lymphocytes. Neither plasma viral levels nor therapy had an effect on the decay rate constants or the percentage of labeled cells in the slowly proliferating pool. Monocyte production was inversely related to viral load (r = -0.56, P = 0.003) and increased with therapy (P = 0.01). These findings demonstrate that HIV does not impair CD4 T cell production but does increase CD4 and CD8 lymphocyte proliferation and death by inducing entry into a rapidly proliferating subpopulation of cells.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Infecções por HIV/imunologia , HIV-1/fisiologia , Adulto , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/patologia , Divisão Celular/imunologia , Feminino , Infecções por HIV/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Replicação Viral/imunologia
4.
Cancer Res ; 61(20): 7449-55, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11606379

RESUMO

Female transgenic mice that express SV40 T/t antigens under the regulatory control of the rat C3(1) gene spontaneously develop multifocal mammary lesions that predictably evolve into invasive, hormone-independent carcinomas, whereas male mice are prone to develop prostate cancer. Chemopreventive agents were administered to female C3(1)/SV40 large T-antigen mice from 7 to 19 weeks of age, during which time the mammary lesions developed and progressed to invasive carcinomas. No significant differences in the numbers of preinvasive mammary intraepithelial neoplasia lesions (histologically similar to human ductal carcinoma in situ) were observed after 2 or 8 weeks of treatment between mice receiving either vehicle alone, dehydroepiandrosterone (DHEA), or 2-difluoromethylornithine (DFMO). However, a dose-response reduction in invasive carcinoma growth was observed for both DFMO, an inhibitor of ornithine decarboxylase, and DHEA, the primary steroid precursor to both androgens and estrogens in primates. Despite unaltered expression of the transgene, tumor incidence was reduced approximately 20% by DFMO (8000 mg/kg) and 30% by DHEA (4000 mg/kg; P < 0.05). Tumor multiplicity was reduced by approximately 50% by both DFMO and DHEA (P < 0.05). DFMO had a dose-dependent effect on total tumor burden, which was reduced by 25% at low doses (4000 mg/kg) and 70% at high doses (8000 mg/kg). DHEA reduced tumor burden by 50% and 66% at low (2000 mg/kg) and high (4000 mg/kg) doses, respectively. Interestingly, despite its inhibitory effects on tumor development, DHEA caused a dose-dependent increase of serum estradiol levels that we have previously shown to increase mammary tumor formation in this model. No effect on the development of the prostate cancer precursor lesions (prostate intraepithelial neoplasia) was observed when mice were treated with DHEA, DFMO, tocopherol acetate, selenomethionine, or 9-cis-retinoic acid, although the effects on late-stage prostate cancer development were not determined. These results demonstrate that despite the expression of the highly transforming C3(1)/SV40 large T-antigen transgene, this transgenic model can be used to study the effects of chemopreventive agents on mammary cancer progression. The tumor-inhibitory effects of DHEA and DFMO on mammary cancer growth appear to occur after the development of preinvasive lesions, suggesting that these agents inhibit tumor progression but not initiation.


Assuntos
Anticarcinógenos/farmacologia , Desidroepiandrosterona/farmacologia , Eflornitina/farmacologia , Neoplasias Mamárias Experimentais/prevenção & controle , Neoplasias da Próstata/prevenção & controle , Animais , Anticarcinógenos/toxicidade , Antígenos Transformantes de Poliomavirus/biossíntese , Antígenos Transformantes de Poliomavirus/genética , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Desidroepiandrosterona/toxicidade , Modelos Animais de Doenças , Progressão da Doença , Eflornitina/toxicidade , Estradiol/sangue , Feminino , Expressão Gênica/efeitos dos fármacos , Masculino , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Camundongos , Lesões Pré-Cancerosas/tratamento farmacológico , Lesões Pré-Cancerosas/prevenção & controle , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Ratos , Transgenes/efeitos dos fármacos
5.
Nature ; 413(6853): 271-2, 2001 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-11565020

RESUMO

Retrospective epidemiological data have indicated that cutaneous malignant melanoma may arise as a consequence of intense, intermittent exposure of the skin to ultraviolet radiation, particularly in children, rather than from the cumulative lifetime exposure that is associated with other forms of skin cancer. Here we use a genetically engineered mouse model to show that a single dose of burning ultraviolet radiation to neonates, but not adults, is necessary and sufficient to induce tumours with high penetrance which are reminiscent of human melanoma. Our results provide experimental support for epidemiological evidence that childhood sunburn poses a significant risk of developing this potentially fatal disease.


Assuntos
Melanoma Experimental/etiologia , Neoplasias Induzidas por Radiação/etiologia , Neoplasias Cutâneas/etiologia , Queimadura Solar/complicações , Animais , Animais Recém-Nascidos , Criança , Modelos Animais de Doenças , Fator de Crescimento de Hepatócito/genética , Humanos , Camundongos , Camundongos Transgênicos , Raios Ultravioleta
6.
Anticancer Res ; 21(3C): 2141-7, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11501838

RESUMO

BACKGROUND: It is generally accepted that P-glycoprotein 170 (MDR1/Pgp170) expression in breast tumors results in poor response to chemotherapy due to its ability to export chemotherapeutic agents. Studies indicate that the use of non-steroidal anti-inflammatory drugs (NSAIDs) may enhance the anti-tumor activity of cancer chemotherapeutic agents and reduce the risk of many cancers. The best known function of NSAIDs is to block the enzyme cyclooxygenase (Cox), the rate limiting enzyme in the conversion of arachidonic acid to prostaglandins. In this study we investigated whether expression of the inducible isoform of Cox (Cox-2) is linked with the multidrug resistance phenotype in breast cancer. METHODS: Expression of Cox-2 and MDR1/Pgp170 was investigated in tumor specimens along with normal epithelium in breast cancer patients using immunohistochemisrty. Expression of Cox-2, MDR1/Pgp170, Protein Kinase C (PKC), and Activator Protein 1 (AP1) were investigated in a series of increasingly resistant human MCF-7 breast cancer cells compared to wild type using immunohistochemistry, Western blots, Northern blots, RT-PCR, and Southern blots. RESULTS: Immunohistochemical analyses of human breast tumor specimens revealed a strong correlation between expression of Cox-2 and MDR1/Pgp170. In drug resistant cell lines that over-express MDR1/Pgp170 there was also significant up-regulation of Cox-2 expression. In addition, PKC and AP1 subunits c-Jun and c-Fos were also upregulated. We hypothesized that increased prostaglandin production by Cox-2 induces PKC and the expression of transcriptional factor c-Jun, which in turn, induces the expression of MDR1/Pgp170. CONCLUSION: We propose that pretreatment with selective Cox-2 inhibitors may be useful in the prevention of multidrug resistance in response to cancer chemotherapy and should be further evaluated.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Resistência a Múltiplos Medicamentos/fisiologia , Isoenzimas/biossíntese , Prostaglandina-Endoperóxido Sintases/biossíntese , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Ciclo-Oxigenase 2 , Dinoprostona/biossíntese , Resistencia a Medicamentos Antineoplásicos/fisiologia , Indução Enzimática , Dosagem de Genes , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes MDR , Humanos , Isoenzimas/genética , Proteínas de Membrana , Inclusão em Parafina , Prostaglandina-Endoperóxido Sintases/genética , Proteína Quinase C/biossíntese , Fator de Transcrição AP-1/biossíntese , Células Tumorais Cultivadas
7.
Biol Blood Marrow Transplant ; 7(3): 136-43, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11302547

RESUMO

CD40 stimulation, by either antibody or ligand, has been shown to inhibit the growth of a variety of neoplastic cells, both in vivo and in vitro. In this study, we assessed the effects of CD40 stimulation using a murine agonistic CD40 monoclonal antibody (MoAb) (FGK115) or a soluble recombinant murine CD40 ligand (srmCD40L) in both lethally irradiated and nonirradiated BALB/c mice. Toxicity after CD40 stimulation was not observed in nonirradiated animals receiving up to 100 microg of the agonist anti-CD40 MoAb. However, as little as 10 microg of the agonistic anti-CD40 MoAb induced acute toxicity resulting in 100% morbidity of lethally irradiated animals by 4 days after irradiation. Histological evaluation of animals receiving anti-CD40 MoAb revealed severe intestinal lesions with disruption of the villi, goblet cell depletion, and crypt hyperplasia of the small intestine, colon, and cecum. Delaying the administration of anti-CD40 MoAb or reducing the amount of irradiation given resulted in increased survival and less severe lesions. Analysis of serum cytokine levels in lethally irradiated mice receiving agonistic anti-CD40 showed a marked increase of interferon (IFN)-gamma. Lethally irradiated IFN-gamma knockout mice given the agonistic anti-CD40 MoAb demonstrated significant increases in survival and minimal gut lesions compared with wild-type mice receiving the same regimen, suggesting that IFN-gamma plays a major role in this toxic reaction. These results indicate that CD40 stimulation using agonistic antibodies following lethal irradiation leads to a fatal, cytokine-induced disease affecting the intestine.


Assuntos
Anticorpos Monoclonais/imunologia , Transplante de Medula Óssea , Antígenos CD40/toxicidade , Interferon gama/fisiologia , Animais , Anticorpos Monoclonais/administração & dosagem , Antígenos CD40/imunologia , Colo/efeitos dos fármacos , Colo/patologia , Interferon gama/sangue , Interferon gama/genética , Enteropatias/etiologia , Enteropatias/patologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Taxa de Sobrevida , Fatores de Tempo , Irradiação Corporal Total
8.
Biochim Biophys Acta ; 1526(2): 211-20, 2001 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-11325543

RESUMO

Lsh is a member of the SNF2 family of chromatin remodelers, that regulate diverse biological processes such as replication, repair and transcription. Although expression of Lsh is highly tissue specific in adult animals, Lsh mRNA is detectable in multiple tissues during embryogenesis. In order to determine the physiologic role of Lsh during murine development and to assess its unique function in adult mice, we performed targeted deletion of the Lsh gene using homologous recombination in murine embryonic stem cells. Lsh-/- embryos occurred with the expected Mendelian frequency after implantation and during embryogenesis. However, Lsh-/- mice died within a few hours after birth. Furthermore, newborn mice were 22% lower in weight in comparison with their littermates and showed renal lesions. Thus Lsh is a non-redundant member of the SNF2 family and is essential for normal murine development and survival.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas de Transporte de Cátions , Regulação da Expressão Gênica no Desenvolvimento , Crescimento/fisiologia , Proteínas de Membrana/fisiologia , Proteínas Nucleares , Animais , Animais Recém-Nascidos , Peso ao Nascer , Proteínas de Transporte/genética , Clonagem Molecular , DNA Helicases , Proteínas de Ligação a DNA/fisiologia , Idade Gestacional , Heterozigoto , Rim/anormalidades , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Fatores de Transcrição/fisiologia
10.
J Immunol ; 166(5): 2924-8, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11207240

RESUMO

IL-7 is a critical cytokine in the development of T and B cells but little is known about its activity on nonhematopoietic cells. An unexpected finding was noted in allogeneic bone marrow transplant studies using IL-7 receptor null (IL-7R alpha(-/-)) mice as recipients. These mice exhibited a significantly greater weight loss after total body irradiation compared with wild type, IL-7R alpha(+/+), mice. Pathological assessment indicated greater intestinal crypt damage in IL-7R alpha(-/-) recipients, suggesting these mice may be predisposed to gut destruction. Therefore, we determined the effect of the conditioning itself on the intestinal tract of these mice. IL-7R alpha(-/-) mice and IL-7R alpha(+/+) mice were irradiated and examined for lesions and apoptosis within the small intestine. In moribund animals, IL-7R alpha(-/-) mice had extensive damage in the small intestine, including marked ablation of the crypts and extreme shortening of villi following 1500 cGy total body irradiation. In contrast, by 8 days after irradiation, the small intestines of IL-7R alpha(+/+) mice had regenerated as distinguished by normal villus length and hyperplastic crypts. Following 750 cGy irradiation, IL-7R alpha(-/-) mice had a higher proportion of apoptotic cells in the crypts and an accompanying increase in the pro-apoptotic protein Bak was expressed in intestinal epithelial cells. These results demonstrate the increased radiosensitivity of intestinal stem cells within the crypts in IL-7R alpha(-/-) mice and a role for IL-7 in the protection of radiation-induced apoptosis in these same cells. This study describes a novel role of IL-7 in nonhematopoietic tissues.


Assuntos
Raios gama , Mucosa Intestinal/imunologia , Mucosa Intestinal/efeitos da radiação , Intestino Delgado/imunologia , Intestino Delgado/efeitos da radiação , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/efeitos da radiação , Animais , Apoptose/genética , Apoptose/imunologia , Apoptose/efeitos da radiação , Transplante de Medula Óssea/imunologia , Transplante de Medula Óssea/patologia , Relação Dose-Resposta Imunológica , Relação Dose-Resposta à Radiação , Feminino , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Proteínas de Membrana/efeitos da radiação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/efeitos da radiação , Receptores de Interleucina-7/biossíntese , Receptores de Interleucina-7/deficiência , Transplante Homólogo , Redução de Peso/genética , Redução de Peso/imunologia , Redução de Peso/efeitos da radiação , Irradiação Corporal Total , Proteína Killer-Antagonista Homóloga a bcl-2 , Proteína bcl-X
11.
Am J Pathol ; 158(1): 323-32, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11141507

RESUMO

The C57BL/6, 129, and B6,129 mouse strains or stocks have been commonly used to generate targeted mutant mice. The pathology of these mice is not well characterized. In studies of these aging mice, we found high incidences of hyalinosis (eosinophilic cytoplasmic change) in the glandular stomach, respiratory tract, bile duct, and gall bladder of B6,129 CYP1A2-null and wild-type mice as well as in both sexes of the background 129S4/SvJae strain. The gastric lesions of the glandular stomach were found in 95.7% of female CYP1A2-null mice as well as in 45.7% of female 129S4/SvJae animals. The eosinophilic protein isolated from characteristic hyaline gastric lesions was identified as Ym2, a member of the chitinase family. Immunohistochemistry, using rabbit polyclonal antibodies to oligopeptides derived from the Ym1 sequence, detected focal to diffuse reactivity within both normal and abnormal nasal olfactory and respiratory epithelium, pulmonary alveolar macrophages, bone marrow myeloid cells, and the squamous epithelium of the forestomach and epithelium of the glandular stomach. Alveolar macrophages in acidophilic pneumonia, a major cause of death of aging 129 mice, and in mice with the me mutation also were highly immunoreactive. The possible cause of this protein excess in gastric and other lesions and its possible functions are discussed.


Assuntos
Fatores Quimiotáticos de Eosinófilos/genética , Citocromo P-450 CYP1A2/metabolismo , Mucosa Gástrica/metabolismo , Hialina/metabolismo , Lectinas/genética , Sistema Respiratório/metabolismo , beta-N-Acetil-Hexosaminidases/genética , Animais , Northern Blotting , Western Blotting , Fatores Quimiotáticos de Eosinófilos/metabolismo , Citocromo P-450 CYP1A2/genética , Eletroforese em Gel de Poliacrilamida , Feminino , Regulação da Expressão Gênica , Lectinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Mutantes , Microscopia Eletrônica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sistema Respiratório/patologia , Estômago/patologia , Estômago/ultraestrutura , Análise de Sobrevida , beta-N-Acetil-Hexosaminidases/metabolismo
12.
Biol Blood Marrow Transplant ; 6(6): 604-12, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11128810

RESUMO

Although alloreactive T cells are required for the induction of graft-versus-host disease (GVHD), other factors can influence outcome in murine models of the disease. Lethal total body irradiation (TBI) conditioning regimens followed by reconstitution with allogeneic lymphohematopoietic cells results in the generation of donor anti-host cytotoxic T lymphocyte (CTL)-mediated solid organ (gut, liver, skin) destruction. In contrast, donor anti-host CTL-mediated hematopoietic failure is the primary cause of morbidity following sublethal TBI. To determine the role of interferon (IFN)-gamma in graft-versus-host reactions against hematopoietic and solid organ targets, we used IFN-gamma knockout mice as donors in both lethal TBI and bone marrow transplantation (BMT) rescue and sublethal TBI models. In this report, we show that CD4+ T cells from IFN-gamma knockout (KO) mice resulted in accelerated GVHD after lethal TBI/BMT using a single major histocompatibility class II mismatch model. In marked contrast, the use of these same IFN-gamma KO CD4+ donor cells in combination with sublethal TBI significantly ameliorated GVHD-associated mortality. In these recipients, severe anemia, bone marrow aplasia, and intestinal lesions were observed in the presence but not the absence of donor-derived IFN-gamma. Administration of anti-IFN-gamma antibodies to sublethally irradiated recipients of wild-type donor cells confirmed the role of IFN-gamma depletion in CD4+ T cell-mediated GVHD. In conclusion, the extent of conditioning markedly affects the role of IFN-gamma in GVHD lesions mediated by CD4+ T cells. In models using sublethal TBI, the absence of IFN-gamma is protective from GVHD, whereas in lethal TBI situations, the loss is deleterious.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Doença Enxerto-Hospedeiro/imunologia , Transplante de Células-Tronco Hematopoéticas , Interferon gama/imunologia , Animais , Citotoxicidade Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Imunologia de Transplantes , Transplante Homólogo
13.
Int J Oncol ; 17(4): 811-8, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10995896

RESUMO

Helicobacter hepaticus causes chronic active hepatitis and liver tumors in mice, with associated increase in reactive oxygen species. Indigenous (I)-compounds are bulky DNA adducts present at low levels and detected by 32P-post-labeling. Some may be caused by reactive oxygen species; others occur normally and decrease during liver tumorigenesis. The identity of most is unknown. We investigated whether mouse liver infection by H. hepaticus and resulting progression of hepatic lesions would be associated with qualitative or quantitative changes in I-compounds. Mice were 3, 6, 9, and 12 months of age; liver disease ranged from minimal through marked. In control A/J mice, up to 20 I-compounds were detected, and the total level of these did not change with age, whereas 11 individual I-compounds showed marked age-related differences. These appeared to be coordinately regulated, as the total of these 11 adducts was constant at 6-12 months. In A/JNCr mice naturally infected with H. hepaticus, up to 12 hepatic I-compounds were found. Total levels varied markedly with age and were high at 6 and 12 months. Neither total adduct levels, nor the amount of any individual adduct, correlated positively with severity of hepatic lesions; in some cases, highest levels were found in livers with least disease. Thus, liver infection and tumorigenesis by H. hepaticus was not associated with an increase in any 32P-postlabeled DNA adduct. Marked, and distinct, age-related changes in total or individual adducts in control and infected mice suggest a role in the physiological alterations of aging and in host response to infection.


Assuntos
Envelhecimento , Adutos de DNA/análise , Infecções por Helicobacter/microbiologia , Helicobacter , Fígado/metabolismo , Animais , Cromatografia em Camada Fina , DNA/genética , DNA/metabolismo , Infecções por Helicobacter/complicações , Fígado/química , Fígado/patologia , Neoplasias Hepáticas Experimentais/etiologia , Neoplasias Hepáticas Experimentais/genética , Masculino , Camundongos , Camundongos Endogâmicos , Radioisótopos de Fósforo
14.
Cancer Res ; 60(14): 3738-43, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10919643

RESUMO

The dramatic rise in incidence of malignant melanoma experienced by populations both within the United States and throughout the world over the last several decades has been attributed to enhanced exposure to the UV spectrum of sunlight radiation. This hypothesis can now be tested using genetically engineered mouse models predisposed to malignant melanoma. Here we use melanoma-prone transgenic mice inappropriately expressing hepatocyte growth factor/scatter factor (HGF/SF) in the skin as an experimental model system to ascertain the consequences of a chronic regimen of suberythemal UV radiation on melanoma genesis. HGF/SF is a multifunctional regulator capable of stimulating growth, motility, invasiveness, and morphogenetic transformation in cells, including melanocytes, expressing its receptor tyrosine kinase Met. HGF/SF transgenic mice demonstrate ectopic interfollicular localization and accumulation of melanocytes within the truncal dermis, epidermis, and junction and if untreated develop primary cutaneous melanoma with a mean onset age of approximately 21 months. Transgenic mice and their wild-type littermates subjected to UV radiation three times weekly using FS40 sunlamps (60% UVB and 40% UVA), with daily UV doses graded from 2.25 to 6.0 kJ/m2, developed skin tumors with a mean onset age of 26 and 37 weeks, respectively (P < 0.001, Kaplan-Meier log rank test). However, the repeated doses of suberythemal UV radiation used in this study failed to accelerate melanoma genesis, instead inducing the development of nonmelanoma tumors that included squamous cell carcinomas, squamous papillomas, and sarcomas. The conspicuous absence of melanocytic tumors occurred despite the immunohistochemical detection of a significant stimulation (P < 0.001) in melanocyte-specific bromodeoxyuridine incorporation in response to only 2 weeks of UV irradiation (total UV dose of 13.5 kJ/m2), resulting in 2.6- and 4.6-fold increases in the number of melanocytes in the dermis and epidermis, respectively. These data indicate that chronic suberythemal UV radiation preferentially favors the development of nonmelanocytic over melanocytic neoplasms in this transgenic animal, consistent with the pathogenesis proposed for sun exposure-associated skin cancer based on retrospective studies in the human population. Our findings suggest that the HGF/SF transgenic mouse will be useful as an experimental model for determining the consequences of exposure to various regimens of UV radiation and for elucidating the mechanisms by which such consequences are realized.


Assuntos
Fator de Crescimento de Hepatócito/genética , Neoplasias Induzidas por Radiação/genética , Fatores Etários , Animais , DNA Complementar/metabolismo , Intervalo Livre de Doença , Relação Dose-Resposta à Radiação , Feminino , Masculino , Melanócitos/patologia , Melanócitos/efeitos da radiação , Melanoma Experimental/genética , Melanoma Experimental/patologia , Melanose/genética , Melanose/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Induzidas por Radiação/metabolismo , Neoplasias Induzidas por Radiação/patologia , Regiões Promotoras Genéticas , Pele/patologia , Pele/efeitos da radiação , Fatores de Tempo , Raios Ultravioleta
15.
Mol Cell Biol ; 20(11): 4075-83, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10805749

RESUMO

Ionizing radiation (IR) exposure causes mammalian cells to undergo p53-dependent cell cycle arrest and/or apoptosis. The in vivo role of DNA-dependent protein kinase (DNA-PK) in the transduction of the DNA damage signal to p53 remains unresolved. To determine the relationship between DNA-PK and p53, we studied the cell cycle and apoptotic responses to IR in mice deficient in DNA-PK. Using the slip mouse, which harbors an inactivating mutation of the DNA-PK catalytic subunit (DNA-PKcs), we demonstrated not only that these DNA-PKcs null mutants were highly radiosensitive but also that upon IR treatment, p53 accumulated in their cultured cells and tissue. Induced p53 was transcriptionally active and mediated the induction of p21 and Bax in slip cells. Examination of the thymic cell cycle response to IR treatment indicated that the slip G(1)/S-phase cell cycle checkpoint function was intact. We further show that slip mice exhibited a higher level of spontaneous thymic apoptosis as well as a more robust apoptotic response to IR than wild-type mice. Together, these data demonstrate that the p53-mediated response to DNA damage is intact in cells devoid of DNA-PK activity and suggest that other kinases, such as the product of the gene (ATM) mutated in ataxia telangiectasia, are better candidates for regulating IR-induced phosphorylation and accumulation of p53.


Assuntos
Dano ao DNA/efeitos da radiação , Proteínas de Ligação a DNA , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Ciclo Celular , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/biossíntese , Proteína Quinase Ativada por DNA , Feminino , Fase G1 , Raios gama , Masculino , Camundongos , Camundongos Mutantes , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Fase S , Proteína X Associada a bcl-2
16.
Oncogene ; 19(8): 1020-7, 2000 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-10713685

RESUMO

The 5' flanking region of the C3(1) component of the rat prostate steroid binding protein (PSBP) has been used to successfully target the expression of the SV40 large T-antigen (Tag) to the epithelium of both the mammary and prostate glands resulting in models of mammary and prostate cancers which histologically resemble the human diseases. Atypia of the mammary ductal epithelium develops at about 8 weeks of age, progressing to mammary intraepithelial neoplasia (resembling human ductal carcinoma in situ [DCIS]) at about 12 weeks of age with the development of invasive carcinomas at about 16 weeks of age in 100% of female mice. The carcinomas share features to what has been classified in human breast cancer as infiltrating ductal carcinomas. All FVB/N female mice carrying the transgene develop mammary cancer with about a 15% incidence of lung metastases. Approximately 10% of older male mice develop anaplastic mammary carcinomas. Unlike many other transgenic models in which hormones and pregnancy are used to induce a mammary phenotype, C3(1)/Tag mice develop mammary tumors in the mammary epithelium of virgin animals without hormone supplementation or pregnancy. Although mammary tumor development appears hormone-responsive at early stages, invasive carcinomas are hormone-independent, which corresponds to the loss of estrogen receptor-alpha expression during tumor progression. Molecular and biologic factors related to mammary tumor progression can be studied in this model since lesions evolve over a predictable time course. Genomic alterations have been identified during tumor progression, including an amplification of the distal portion of chromosome 6 containing ki-ras and loss of heterozygosity (LOH) in other chromosomal regions. We have demonstrated that stage specific alterations in the expression of genes which are critical regulators of the cell cycle and apoptosis are functionally important in vivo. C3(1)/Tag mice appear useful for testing particular therapies since growth of the mammary tumors can be reduced using chemopreventive agents, cytokines, and an anti-angiogenesis agent.


Assuntos
Proteína de Ligação a Androgênios/genética , Antígenos Transformantes de Poliomavirus/genética , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/patologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Proteínas Proto-Oncogênicas c-bcl-2 , Proteína de Ligação a Androgênios/metabolismo , Animais , Apoptose , Carcinoma Ductal de Mama/terapia , Ciclo Celular/genética , Modelos Animais de Doenças , Células Epiteliais/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Imunoterapia , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Transgênicos , Proteína de Ligação a Fosfatidiletanolamina , Gravidez , Prostateína , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Secretoglobinas , Uteroglobina , Proteína X Associada a bcl-2
17.
Oncogene ; 19(8): 968-88, 2000 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-10713680

RESUMO

NIH sponsored a meeting of medical and veterinary pathologists with mammary gland expertise in Annapolis in March 1999. Rapid development of mouse mammary models has accentuated the need for definitions of the mammary lesions in genetically engineered mice (GEM) and to assess their usefulness as models of human breast disease. The panel of nine pathologists independently reviewed material representing over 90% of the published systems. The GEM tumors were found to have: (1) phenotypes similar to those of non-GEM; (2) signature phenotypes specific to the transgene; and (3) some morphological similarities to the human disease. The current mouse mammary and human breast tumor classifications describe the majority of GEM lesions but unique morphologic lesions are found in many GEM. Since little information is available on the natural history of GEM lesions, a simple morphologic nomenclature is proposed that allows direct comparisons between models. Future progress requires rigorous application of guidelines covering pathologic examination of the mammary gland and the whole animal. Since the phenotype of the lesions is an essential component of their molecular pathology, funding agencies should adopt policies ensuring careful morphological evaluation of any funded research involving animal models. A pathologist should be part of each research team.


Assuntos
Neoplasias Mamárias Experimentais/classificação , Neoplasias Mamárias Experimentais/patologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Hiperplasia/genética , Hiperplasia/patologia , Hibridização In Situ , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Patologia/métodos , Lesões Pré-Cancerosas , Ratos , Terminologia como Assunto
18.
J Toxicol Environ Health A ; 58(4): 199-214, 1999 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-10591488

RESUMO

Based on the occurrence of pulmonary cancers in exposed populations, cadmium is classified as a human carcinogen. More controversial target sites for cadmium in humans include the prostate and kidney, where some studies have shown a link between cadmium and cancer. In Wistar rats cadmium induces tumors in the ventral prostate. The relevance of such lesions to humans is debated since the rat ventral lobe, unlike the dorsolateral lobe, has no embryological homolog in the human prostate. Cadmium has not been linked with renal tumors in rodents but is a potent nephrotoxin. In this work we studied the effects of oral cadmium in the Noble (NBL/Cr) rat with particular attention to proliferative lesions of the prostate and kidneys. Cadmium (as CdCl2) was given ad libitum throughout the study in the drinking water at doses of 0, 25, 50, 100, and 200 ppm Cd to groups (initial n = 30) of male rats, which were observed for up to 102 wk. At the lower doses of cadmium (< or =50 ppm) a clear dose-related increase in total proliferative lesions of the prostate (ventral and dorsolateral lesions combined) occurred (0 ppm = 21% incidence, 25 ppm = 46%, 50 ppm = 50%; trend p < .03). These lesions were described as intraepithelial hyperplasia with occasional areas of atypical epithelial cells without stromal invasion. The lesions occurred primarily in the dorsolateral prostate with cadmium exposure and most frequently showed three or more foci within each specimen. At higher doses, prostatic proliferative lesions declined to control levels. The loss of prostatic response at the higher doses was likely due to diminished testicular function secondary to cadmium treatment. This was reflected in lesions indicative of testicular hypofunction at the highest cadmium dose, namely, interstitial cell hyperplasia, and a strong correlation between cadmium dose and total proliferative lesions of the testes (hyperplasias and tumors combined). Renal tumors (mainly mesenchymal and pelvic transitional cell), although few in number, showed a positive correlation with cadmium dose, as did pelvic transitional epithelial hyperplasia. Renal lesions were not associated with any cadmium-induced changes in age-related chronic nephropathy. The incidence of pheochromocytomas of the adrenal was increased by cadmium but only at the 50 ppm dose. Inflammatory lesions of the liver and spleen were common at higher doses and showed strong trends based on dose. These results indicate that oral cadmium can induce proliferative lesions in the prostate and kidney of the Noble rat. The finding of proliferative lesions of dorsolateral prostate in rats has presumed relevance to human prostate cancers.


Assuntos
Neoplasias das Glândulas Suprarrenais/induzido quimicamente , Cádmio/toxicidade , Carcinógenos/toxicidade , Neoplasias Renais/induzido quimicamente , Hiperplasia Prostática/induzido quimicamente , Neoplasias da Próstata/induzido quimicamente , Neoplasias Testiculares/induzido quimicamente , Neoplasias das Glândulas Suprarrenais/patologia , Animais , Testes de Carcinogenicidade , Doença Hepática Induzida por Substâncias e Drogas/patologia , Hiperplasia/induzido quimicamente , Hiperplasia/patologia , Neoplasias Renais/patologia , Masculino , Feocromocitoma/induzido quimicamente , Feocromocitoma/patologia , Hiperplasia Prostática/patologia , Neoplasias da Próstata/patologia , Ratos , Ratos Endogâmicos , Esplenopatias/induzido quimicamente , Esplenopatias/patologia , Neoplasias Testiculares/patologia
19.
Am J Pathol ; 154(4): 1125-35, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10233851

RESUMO

The contribution of chemokines toward angiogenesis is currently a focus of intensive investigation. Certain members of the CXC chemokine family can induce bovine capillary endothelial cell migration in vitro and corneal angiogenesis in vivo, and apparently act via binding to their receptors CXCR1 and CXCR2. We used an RNAse protection assay that permitted the simultaneous detection of mRNA for various CXC chemokine receptors in resting human umbilical vein endothelial cells (HUVECs) and detected low levels of only CXCR4 mRNA. Stimulation of HUVECs with vascular endothelial growth factor (VEGF) or basic fibroblast growth factor (bFGF) up-regulated levels of only CXCR4 mRNA. CXCR4 specifically binds the chemokine stromal-derived factor-1alpha (SDF-1alpha). Competitive binding studies using 125I-labeled SDF-1alpha with Scatchard analysis indicated that VEGF or bFGF induced an average number of approximately 16,600 CXCR4 molecules per endothelial cell, with a Kd = 1.23 x 10(-9) mol/L. These receptors were functional as HUVECs and human aorta endothelial cells (HAECs) migrated toward SDF-1alpha. Although SDF-1alpha-induced chemotaxis was inhibited by the addition of a neutralizing monoclonal CXCR4 antibody, endothelial chemotaxis toward VEGF was not altered; therefore, the angiogenic effect of VEGF is independent of SDF-1alpha. Furthermore, subcutaneous SDF-1alpha injections into mice induced formation of local small blood vessels that was accompanied by leukocytic infiltrates. To test whether these effects were dependent on circulating leukocytes, we successfully obtained SDF-1alpha-induced neovascularization from cross sections of leukocyte-free rat aorta. Taken together, our data indicate that SDF-1alpha acts as a potent chemoattractant for endothelial cells of different origins bearing CXCR4 and is a participant in angiogenesis that is regulated at the receptor level by VEGF and bFGF.


Assuntos
Quimiocinas CXC/farmacologia , Fatores de Crescimento Endotelial/farmacologia , Endotélio Vascular/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Linfocinas/farmacologia , Receptores CXCR4/biossíntese , Animais , Aorta/efeitos dos fármacos , Aorta/fisiologia , Ligação Competitiva , Membrana Celular/metabolismo , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Quimiotaxia/efeitos dos fármacos , Fatores de Crescimento Endotelial/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Feminino , Humanos , Técnicas In Vitro , Linfocinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Fisiológica/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores CXCR4/metabolismo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
20.
Nat Med ; 4(7): 802-7, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9662371

RESUMO

Components of the transforming growth factor-beta (TGF-beta) signal pathway function as classic tumor suppressors, but the role of the TGF-betas themselves is less clear. Here we show that mice heterozygous for deletion of the TGF-beta1 gene express only 10-30% of wild-type TGF-beta1 protein levels. Although grossly normal, these mice have a subtly altered proliferative phenotype, with increased cell turnover in the liver and lung. Treatment of these mice with chemical carcinogens resulted in enhanced tumorigenesis when compared with wild-type littermates. However, tumors in the heterozygous mice did not lose the remaining wild-type TGF-beta1 allele, indicating that the TGF-beta1 ligand is a new form of tumor suppressor that shows true haploid insufficiency in its ability to protect against tumorigenesis.


Assuntos
Genes Supressores de Tumor , Fator de Crescimento Transformador beta/genética , Animais , Apoptose , Testes de Carcinogenicidade , Proteínas de Ciclo Celular/genética , Divisão Celular , Marcação de Genes , Fígado/citologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...