Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Am J Pathol ; 173(3): 689-99, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18688025

RESUMO

Primary abnormalities in permeability barrier function appear to underlie atopic dermatitis and epidermal trauma; a concomitant barrier dysfunction could also drive other inflammatory dermatoses, including psoriasis. Central to this outside-inside view of disease pathogenesis is the epidermal generation of cytokines/growth factors, which in turn signal downstream epidermal repair mechanisms. Yet, this cascade, if sustained, signals downstream epidermal hyperplasia and inflammation. We found here that acute barrier disruption rapidly stimulates mRNA and protein expression of epidermal vascular endothelial growth factor-A (VEGF-A) in normal hairless mice, a specific response to permeability barrier requirements because up-regulation is blocked by application of a vapor-impermeable membrane. Moreover, epidermal vegf(-/-) mice display abnormal permeability barrier homeostasis, attributable to decreased VEGF signaling of epidermal lamellar body production; a paucity of dermal capillaries with reduced vascular permeability; and neither angiogenesis nor epidermal hyperplasia in response to repeated tape stripping (a model of psoriasiform hyperplasia). These results support a central role for epidermal VEGF in the maintenance of epidermal permeability barrier homeostasis and a link between epidermal VEGF production and both dermal angiogenesis and the development of epidermal hyperplasia. Because psoriasis is commonly induced by external trauma [isomorphic (Koebner) phenomenon] and is associated with a prominent permeability barrier abnormality, excess VEGF production, prominent angiogenesis, and epidermal hyperplasia, these results could provide a potential outside-inside mechanistic basis for the development of psoriasis.


Assuntos
Epiderme/metabolismo , Epiderme/patologia , Homeostase/fisiologia , Neovascularização Patológica/metabolismo , Psoríase/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Northern Blotting , Derme/irrigação sanguínea , Derme/metabolismo , Hiperplasia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Pelados , Camundongos Knockout , Permeabilidade
2.
J Invest Dermatol ; 126(10): 2316-22, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16741507

RESUMO

Hemangioma of infancy is the most common neoplasm of childhood. While hemangiomas are classic examples of angiogenesis, the angiogenic factors responsible for hemangiomas are not fully understood. Previously, we demonstrated that malignant endothelial tumors arise in the setting of autocrine loops involving vascular endothelial growth factor (VEGF) and its major mitogenic receptor vascular endothelial growth factor receptor 2. Hemangiomas of infancy differ from malignant endothelial tumors in that they usually regress, or can be induced to regress by pharmacologic means, suggesting that angiogenesis in hemangiomas differs fundamentally from that of malignant endothelial tumors. Here, we demonstrate constitutive activation of the endothelial tie-2 receptor in human hemangioma of infancy and, using a murine model of hemangioma, bEnd.3 cells; we show that bEnd.3 hemangiomas produce both angiopoietin-2 (ang-2) and its receptor, tie-2, in vivo. We also demonstrate that inhibition of tie-2 signaling with a soluble tie-2 receptor decreases bEnd.3 hemangioma growth in vivo. The efficacy of tie-2 blockade suggests that either tie-2 activation or ang-2 may be required for in vivo growth. To address this issue, we used tie-2-deficient bEnd.3 hemangioma cells, which, surprisingly, were fully proficient in in vivo growth. Previous studies from our laboratory and others have implicated reactive oxygen-generating nox enzymes in the angiogenic switch, so we examined the effect of nox inhibitors on ang-2 production in vitro and on bEnd.3 tumor growth in vivo. We then inhibited ang-2 production pharmacologically using novel inhibitors of nox enzymes and found that this treatment nearly abolished bEnd.3 hemangioma growth in vivo. Signal-transduction blockade targeting ang-2 production may be useful in the treatment of human hemangiomas in vivo.


Assuntos
Angiopoietina-2/antagonistas & inibidores , Hemangioma/tratamento farmacológico , Angiopoietina-2/biossíntese , Angiopoietina-2/genética , Animais , Violeta Genciana/farmacologia , Hemangioma/etiologia , Hemangioma/patologia , Humanos , Camundongos , NADPH Oxidases/antagonistas & inibidores , Compostos de Amônio Quaternário/farmacologia , RNA Mensageiro/análise , Receptor TIE-2/antagonistas & inibidores , Receptor TIE-2/fisiologia , Transdução de Sinais
3.
Arch Dermatol ; 141(10): 1297-300, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16230568

RESUMO

BACKGROUND: Vascular malformations and hemangiomas, which are endothelial lesions of childhood, may result in considerable morbidity because they can cause discomfort and functional impairment and have a negative affect on the patient's appearance. Although vascular malformations may initially appear very similar to hemangiomas, they have distinct clinical courses. Infantile hemangiomas progress through 3 stages: proliferative, involuting, and involuted. The proliferative phase is characterized by clinical growth. Once hemangiomas reach their maximum size, they begin to regress or involute. Histologically, this stage is characterized by endothelial apoptosis. Finally, the involuted stage of the hemangioma occurs when the original lesion is replaced by a connective tissue remnant. In contrast to hemangiomas, vascular malformations do not involute but continue to enlarge as the patient grows. OBSERVATIONS: The biochemical differences between hemangiomas, which involute, and vascular malformations, which do not involute, are not well understood. We found that the transcription factor encoded by the Wilms tumor 1 (WT1) gene is expressed in the endothelium of hemangiomas but not in vascular malformations. CONCLUSIONS: Defects in WT1 signaling may underlie the inability of malformation endothelial cells to undergo physiologic apoptosis and remodeling. The availability of WT1 staining in hospital laboratories may allow the clinician to distinguish hemangiomas from vascular malformations and thus to give appropriate therapy to the patient.


Assuntos
Malformações Arteriovenosas/diagnóstico , Malformações Arteriovenosas/genética , Expressão Gênica , Genes do Tumor de Wilms , Hemangioma/diagnóstico , Hemangioma/genética , Apoptose , Malformações Arteriovenosas/patologia , Diagnóstico Diferencial , Progressão da Doença , Hemangioma/patologia , Hemangioma/fisiopatologia , Humanos , Imuno-Histoquímica
4.
Clin Cancer Res ; 8(12): 3728-33, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12473582

RESUMO

PURPOSE: Melanoma is the most common cause of death from cutaneous malignancy, and is the cancer that is most rapidly rising in incidence. Because current therapeutic methods for metastatic melanoma are poorly efficacious, enhanced understanding of signal transduction in melanoma progression is warranted. Prior experimental studies in murine models and human tissues have shown a correlation among activation of mitogen activated protein kinase (MAPK) signaling, angiogenesis, and tumorigenesis. Because of these findings, we wanted to assess the role of MAPK signaling in melanoma progression and angiogenesis. EXPERIMENTAL DESIGN: We studied expression of phosphorylated (active) MAPK and two target genes known to be induced by MAPK signaling, tissue factor and vascular endothelial growth factor, in 131 melanocytic lesions, ranging from atypical nevi to metastatic melanoma. RESULTS: We observed little staining for activated (phosphorylated) MAPK and low amounts of angiogenesis in atypical nevi, but angiogenesis and MAPK activation were activated in radial growth melanoma and in later stage lesions. CONCLUSIONS: Our findings implicate MAPK activation as an early event in melanoma progression, and MAPK may be a potential target for pharmacologic intervention.


Assuntos
Melanoma/enzimologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Nevo Pigmentado/enzimologia , Neoplasias Cutâneas/enzimologia , Fator A de Crescimento do Endotélio Vascular , Adulto , Idoso , Idoso de 80 Anos ou mais , Indutores da Angiogênese/metabolismo , Progressão da Doença , Ativação Enzimática , Feminino , Humanos , Técnicas Imunoenzimáticas , Masculino , Melanoma/patologia , Pessoa de Meia-Idade , Invasividade Neoplásica , Nevo Pigmentado/patologia , Fosforilação , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/secundário , Tromboplastina/metabolismo , Células Tumorais Cultivadas
5.
J Invest Dermatol ; 123(4): 788-90, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15373786

RESUMO

Recessive dystrophic epidermolysis bullosa (RDEB) is an autosomal recessive disorder characterized by the loss of collagen type VII, an intrinsic component of the anchoring fibrils, which attach the epidermis to the dermis. Of the genetic blistering disorders, RDEB has the highest rate of morbidity and mortality, with morbidity arising from fusion of digits in a mitten-glove deformity and growth retardation associated with anemia. The leading cause of death in RDEB is cutaneous squamous cell carcinoma, which causes death through invasion and metastasis. In order to better understand the pathogenesis of these rare but aggressive squamous cell carcinoma (SCC), we analyzed them for mutations in p53 and loss of p16ink4a. Three tumors demonstrated mutations in the p53 tumor suppressor gene. We also analyzed SCC from patients with RDEB for the presence of p16ink4a hypermethylation, and found two tumors that have loss of p16ink4a through hypermethylation. This is the first description of specific abnormalities in tumor suppressor genes in RDEB associated SCC, and demonstrates that alterations in both p53 and p16ink4a can contribute to RDEB associated SCC.


Assuntos
Carcinoma de Células Escamosas/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Epidermólise Bolhosa Distrófica/genética , Neoplasias Cutâneas/genética , Proteína Supressora de Tumor p53/genética , Metilação de DNA , Éxons , Genes Recessivos , Humanos , Mutação
6.
Proc Natl Acad Sci U S A ; 102(1): 175-9, 2005 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-15611471

RESUMO

Burkitt's lymphoma (BL) is an aggressive B cell neoplasm, which is one of the most common neoplasms of childhood. It is highly widespread in East Africa, where it appears in endemic form associated with Epstein-Barr virus (EBV) infection, and around the world in a sporadic form in which EBV infection is much less common. In addition to being the first human neoplasm to be associated with EBV, BL is associated with a characteristic translocation, in which the Ig promoter is translocated to constitutively activate the c-myc oncogene. Although many BLs respond well to chemotherapy, a significant fraction fails to respond to therapy, leading to death. In this article, we demonstrate that EBV-positive BL expresses high levels of activated mitogen-activated protein kinase and reactive oxygen species (ROS), and that ROS directly regulate NF-kappaB activation. EBV-negative BLs exhibit activation of phosphoinositol 3-kinase, but do not have elevated levels of ROS. Elevated reactive oxygen may play a role in diverse forms of viral carcinogenesis in humans, including cancers caused by EBV, hepatitis B, C, and human T cell lymphotropic virus. Our findings imply that inhibition of ROS may be useful in the treatment of EBV-induced neoplasia.


Assuntos
Linfoma de Burkitt/virologia , Herpesvirus Humano 4/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Linfoma de Burkitt/enzimologia , Linfoma de Burkitt/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Humanos , NF-kappa B/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas Virais
7.
J Biol Chem ; 280(7): 5870-4, 2005 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-15576369

RESUMO

Tuberous sclerosis (TS) is a common autosomal dominant disorder caused by loss or malfunction of hamartin (tsc1) or tuberin (tsc2). Many lesions in TS do not demonstrate loss of heterozygosity for these genes, implying that dominant negative forms of these genes may account for some hamartomas and neoplasms in TS. To test this hypothesis, we expressed a dominant negative allele of tuberin (DeltaRG) behind the cytomegalovirus promoter in NIH3T3 cells and transgenic mice. This allele binds hamartin but has a deletion in the C terminus of tuberin, leading to constitutive activation of rap1 and rab5/rabaptin. Expression of DeltaRG in NIH3T3 cells led to a strong induction of reactive oxygen species, induction of vascular endothelial growth factor, and malignant transformation in vivo. Expression of DeltaRG driven by the constitutive cytomegalovirus promoter led to high level expression in all murine tissues examined, including skin, kidney, liver, and brain. Surprisingly, mice expressing the DeltaRG transgene developed a fibrovascular collagenoma in the dermis, which closely resembles the Shagreen patch observed in human patients with TS. In addition, numerous small subpial collections of external granule cells in the cerebellum were observed, which may be the murine equivalent of subependymal giant cell astrocytomas or tubers commonly seen in TS patients. Thus, expression of a dominant negative tuberin in multiple tissues can lead to a tissue-specific phenotype resembling some of the findings in human TS. Our data are the first to demonstrate that specific signaling abnormalities underlie specific hamartomas in a model of a human genetic disorder.


Assuntos
Neoplasias Encefálicas/patologia , Genes Dominantes/genética , Regiões Promotoras Genéticas/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Neoplasias Cutâneas/patologia , Esclerose Tuberosa/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Alelos , Animais , Apoptose , Neoplasias Encefálicas/genética , Quimiocina CCL2/metabolismo , Camundongos , Camundongos Transgênicos , Fibras Musculares Esqueléticas , Mutação/genética , Células NIH 3T3 , Especificidade de Órgãos , Fenótipo , Fosforilação , Fator de Crescimento Derivado de Plaquetas/farmacologia , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores CCR2 , Receptores de Quimiocinas/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Transgenes/genética , Esclerose Tuberosa/patologia , Proteína 2 do Complexo Esclerose Tuberosa , Fator A de Crescimento do Endotélio Vascular/genética
8.
Am J Pathol ; 163(4): 1321-7, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14507641

RESUMO

Pathological angiogenesis, the development of a microvasculature by neoplastic processes, is a critical component of the development of tumors. The role of oncogenes in the induction of angiogenesis has been extensively studied in benign and malignant tumors. However, the role of infection in inducing angiogenesis is not well understood. Verruga peruana is a clinical syndrome caused by the bacterium Bartonella bacilliformis, and is characterized by the development of hemangioma-like lesions, in which bacteria colonize endothelial cells. To gain insight into how this bacteria induces angiogenesis in vivo, we performed in situ hybridization of clinical specimens of verruga peruana for the angiogenesis factors vascular endothelial growth factor (VEGF), its receptors VEGFR1 and VEGFR2, and angiopoietin-2. High-level expression of angiopoietin-2 and VEGF receptors was observed in the endothelium of verruga peruana. Surprisingly, the major source of VEGF production in verruga peruana is the overlying epidermis. Infection of cultured endothelium with B. bacilliformis also resulted in induction of angiopoetin-2 in vitro. These findings imply a collaboration between infected endothelium and overlying epidermis to induce angiogenesis.


Assuntos
Indutores da Angiogênese/metabolismo , Infecções por Bartonella/complicações , Fatores de Crescimento Endotelial/biossíntese , Endotélio Vascular/metabolismo , Epiderme/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Linfocinas/biossíntese , Neovascularização Patológica/metabolismo , Neovascularização Patológica/microbiologia , Angiopoietina-2 , Células Cultivadas , Humanos , Imuno-Histoquímica , Hibridização In Situ , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
9.
J Virol ; 76(10): 5208-19, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11967335

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) encodes a cellular dihydrofolate reductase (DHFR) homologue. Methotrexate (MTX), a potent anti-inflammatory agent, inhibits cellular DHFR activity. We investigated the effect of noncytotoxic doses of MTX on latency and lytic KSHV replication in two KSHV-infected primary effusion lymphoma cell lines (BC-3 and BC-1) and in MTX-resistant BC-3 cells (MTX-R-BC-3 cells). Treatment with MTX completely prevented tetradecanoyl phorbol acetate-induced viral DNA replication and strongly decreased viral lytic transcript levels, even in MTX-resistant cells. However, the same treatment had no effect on transcription of cellular genes and KSHV latent genes. One of the lytic transcripts inhibited by MTX, ORF50/Rta (open reading frame), is an immediate-early gene encoding a replication-transcription activator required for expression of other viral lytic genes. Therefore, transcription of genes downstream of ORF50/Rta was inhibited, including those encoding the viral G-protein-coupled receptor (GPCR), viral interleukin-6, and K12/kaposin, which have been shown to be transforming in vitro and oncogenic in mice. Resistance to MTX has been documented in cultured cells and also in patients treated with this drug. However, MTX showed an inhibitory activity even in MTX-R-BC-3 cells. Two currently available antiherpesvirus drugs, cidofovir and foscarnet, had no effect on the transcription of these viral oncogenes and ORF50/Rta. MTX is the first example of a compound shown to downregulate the expression of ORF50/Rta and therefore prevent viral transforming gene transcription. Given that the expression of these genes may be important for tumor development, MTX could play a role in the future management of KSHV-associated malignancies.


Assuntos
Herpesvirus Humano 8/fisiologia , Proteínas Imediatamente Precoces/biossíntese , Metotrexato/farmacologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , Organofosfonatos , Transativadores/biossíntese , Replicação Viral , Anti-Inflamatórios não Esteroides/farmacologia , Antivirais/farmacologia , Cidofovir , Citosina/análogos & derivados , Citosina/farmacologia , Regulação para Baixo , Resistência a Medicamentos , Foscarnet/farmacologia , Humanos , Proteínas Imediatamente Precoces/genética , Interleucina-6/biossíntese , Neuropeptídeos/biossíntese , Fases de Leitura Aberta , Compostos Organofosforados/farmacologia , Transativadores/genética , Transcrição Gênica/efeitos dos fármacos , Células Tumorais Cultivadas , Proteínas Virais/biossíntese , Ativação Viral , Latência Viral/efeitos dos fármacos
10.
J Biol Chem ; 278(37): 35501-7, 2003 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-12816951

RESUMO

Natural products comprise a major source of small molecular weight angiogenesis inhibitors. We have used the transformed endothelial cell line SVR as an effective screen of natural product extracts to isolate anti-angiogenesis and anti-tumor compounds. Aqueous extracts of Magnolia grandiflora exhibit potent activity in our SVR proliferation assays. We found that the small molecular weight compound honokiol is the active principle of magnolia extract. Honokiol exhibited potent anti-proliferative activity against SVR cells in vitro. In addition, honokiol demonstrated preferential inhibition of primary human endothelial cells compared with fibroblasts and this inhibition was antagonized by antibodies against TNF alpha-related apoptosis-inducing ligand. In vivo, honokiol was highly effective against angiosarcoma in nude mice. Our preclinical data suggests that honokiol is a systemically available and non-toxic inhibitor of angiogenesis and should be further evaluated as a potential chemotherapeutic agent.


Assuntos
Inibidores da Angiogênese/farmacologia , Compostos de Bifenilo/farmacologia , Endotélio Vascular/efeitos dos fármacos , Hemangiossarcoma/patologia , Lignanas , Fitoterapia , Inibidores da Angiogênese/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Compostos de Bifenilo/uso terapêutico , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Transformada , Endotélio Vascular/transplante , Hemangiossarcoma/tratamento farmacológico , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Magnolia , Camundongos , Camundongos Nus , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA