Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Clin Lab Med ; 31(2): 345-58, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21549247

RESUMO

Blue nevi and related lesions are characterized by the proliferation of dermal dendritic melanocytes. Although they share certain common clinical and histologic features, they encompass a spectrum of lesions ranging from benign melanocytic hamartomas and common blue nevi to borderline malignant pigmented epithelioid melanocytoma and aggressive malignant blue nevi. This article succinctly describes the common dermal dendritic proliferations and updates readers on newly classified entities and variants. The differential diagnosis of the main entities and strategies to distinguish them from their melanocytic and nonmelanocytic mimics is also presented.


Assuntos
Nevo Azul/patologia , Neoplasias Cutâneas/patologia , Diagnóstico Diferencial , Hamartoma/patologia , Histocitoquímica , Humanos , Melanócitos/patologia , Nevo Azul/diagnóstico , Nevo Pigmentado/patologia , Prognóstico , Neoplasias Cutâneas/diagnóstico
2.
Arch Pathol Lab Med ; 135(3): 327-36, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21366456

RESUMO

CONTEXT: Blue nevi are a subset of melanocytic proliferations containing cells reminiscent of the embryonal neural crest-derived dendritic melanocytic precursors. They are common specimens in a general pathology practice, but some of their rare variants may pose diagnostic difficulty. Recent molecular studies provide new insights into genetics of blue nevi. OBJECTIVE: To critically review clinical and histologic features of blue nevi with emphasis on diagnostic problems and rare variants, as well as to provide an update on the pathogenesis of blue nevi. DATA SOURCES: Published peer-reviewed literature and personal experience of the authors. CONCLUSIONS: Challenging areas in diagnosis of blue nevi include recognition of amelanotic, desmoplastic, atypical, and malignant variants of blue nevus. Recent data show that mutations in genes responsible for common nevi or melanomas such as BRAF , NRAS , or c- kit are rare in blue nevi. Benign and malignant blue nevi harbor frequent mutations in the Gαq class of G-protein α subunits, Gnaq and Gna11 proteins.


Assuntos
Melanócitos/patologia , Nevo Azul/diagnóstico , Neoplasias Cutâneas/diagnóstico , Biomarcadores Tumorais/metabolismo , Feminino , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Humanos , Masculino , Melanócitos/metabolismo , Mutação , Nevo Azul/genética , Nevo Azul/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo
3.
Am J Surg Pathol ; 35(5): 656-69, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21436676

RESUMO

The histopathologic interpretation of proliferative nodules (PNs) in congenital melanocytic nevi can present significant challenges as some PNs may exhibit atypical features that make the distinction from melanoma difficult. We compared histologic features, Ki-67%, PHH3, and CD117% expression levels by immunohistochemistry in 18 benign and 25 atypical PNs (from 41 patients) with that of background congenital nevi (of these 43 cases), 10 congenital nevi, and 3 dermal melanomas arising in congenital melanocytic lesions. In addition, we evaluated the presence of BRAF, GNAQ, HRAS, KRAS, and NRAS mutations in all groups using the SNaPshot Multiplex System. Follow-up was available on 19 patients (9 benign and 10 atypical PNs) (range, 2 to 20 y; median, 8 y) and all were alive with no evidence of disease. The specific histologic features of atypical PNs, such as sharp demarcation (P<0.001), expansile growth (P<0.001), epidermal effacement (P<0.001), nuclear pleomorphism (P<0.001), and increased mitoses (P<0.001), differed significantly from those of benign PNs. Immunohistochemical results showed that Ki-67% and PHH3 scores, but not CD117% expression, were significantly higher (P<0.05) in atypical PNs. Molecular analyses showed that the PNs and background congenital melanocytic nevi of the giant congenital nevi possess more frequent NRAS mutations and infrequent BRAF mutations when compared with those of the remaining cases. These findings suggest that histologic features and Ki-67 and PHH3 expression levels are the strongest parameters to distinguish between benign versus atypical PNs. The immunohistochemical results suggest that atypical PNs are distinct borderline lesions residing between benign PNs and dermal melanomas. Although numerous mutations are detected in the samples, the diagnostic use of molecular analysis in this regard is limited.


Assuntos
Biomarcadores Tumorais/análise , Melanoma/patologia , Nevo Pigmentado/patologia , Neoplasias Cutâneas/patologia , Adolescente , Adulto , Criança , Pré-Escolar , Análise Mutacional de DNA , Diagnóstico Diferencial , Feminino , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Genes ras , Humanos , Imuno-Histoquímica , Lactente , Antígeno Ki-67/metabolismo , Masculino , Melanoma/genética , Melanoma/metabolismo , Pessoa de Meia-Idade , Nevo Pigmentado/congênito , Nevo Pigmentado/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas Proto-Oncogênicas p21(ras) , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Adulto Jovem , Proteínas ras/genética
4.
Surg Pathol Clin ; 2(3): 483-96, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26838534

RESUMO

Blue nevi and related lesions are characterized by the proliferation of dermal dendritic melanocytes. Although they share certain common clinical and histologic features, they encompass a spectrum of lesions ranging from benign melanocytic hamartomas and common blue nevi to borderline malignant pigmented epithelioid melanocytoma and aggressive malignant blue nevi. This article succinctly describes the common dermal dendritic proliferations and updates readers on newly classified entities and variants. The differential diagnosis of the main entities and strategies to distinguish them from their melanocytic and nonmelanocytic mimics is also presented.

5.
J Biol Chem ; 283(42): 28354-60, 2008 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-18664570

RESUMO

That metastatic tumor cells grow in selective non-native environments suggests an ability to differentially respond to local microenvironments. BRMS1, like other metastasis suppressors, halts ectopic growth (metastasis) without blocking orthotopic tumor formation. BRMS1-expressing tumor cells reach secondary sites but do not colonize distant tissues, compelling the hypothesis that BRMS1 selectively restricts the ability of tumor cells to respond to exogenous regulators in different tissues. Here we report that BRMS1 expression in metastatic human breast cancer cells leads to a selective reduction in epidermal growth factor receptor expression and downstream (AKT) signaling. Signaling through another receptor tyrosine kinase, hepatocyte growth factor receptor (c-Met), remains unaltered despite reduced levels of the signaling intermediate phosphatidylinositol (4,5)-bisphosphate. Interestingly, reduced downstream calcium signaling is observed following treatment with platelet-derived growth factor, consistent with decreased phosphatidylinositol (4,5)-bisphosphate. However, platelet-derived growth factor receptor expression is unaltered. Thus, BRMS1 differentially attenuates cellular responses to mitogenic signals, not only dependent upon the specific signal received, but at varying steps within the same signaling cascade. Specific modulation of signaling responses received from the microenvironment may ultimately dictate which environments are permissive/restrictive for tumor cell growth and provide insights into the biology underlying metastasis.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Neoplasias/fisiologia , Cálcio/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Mitógenos , Modelos Biológicos , Metástase Neoplásica , Proteínas de Neoplasias/metabolismo , Fosfatidilinositol 4,5-Difosfato/química , Fosforilação , Receptores de Fatores de Crescimento/metabolismo , Proteínas Repressoras , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica
6.
Am J Pathol ; 172(3): 809-17, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18276787

RESUMO

Breast cancer metastasis suppressor 1 (BRMS1) inhibits formation of macroscopic lung metastases in breast, ovary, and melanoma xenograft models. Because it is unclear which step(s) of the metastatic cascade are affected by BRMS1, the major aim of this study was to determine when and how BRMS1 acts to suppress metastasis. We also examined whether BRMS1 expression globally blocks metastasis or selectively inhibits metastatic outgrowths in specific tissues. Metastatic human breast carcinoma cell lines MDA-MB-231 and -435 expressing enhanced green fluorescent protein (GFP; 231 GFP and 435 GFP) and cell lines transduced with the BRMS1 gene (231 GFP-BRMS1 and 435 GFP-BRMS1) were injected into the left cardiac ventricle to achieve the widest possible cellular distribution, by minimizing first-pass clearance in the lungs. Compared with parental cells, BRMS1-expressing clones formed significantly fewer metastases in all organs tested. When cells were injected directly into the vasculature, fewer of the BRMS1-expressing cells reached lungs or bone compared with parental cells, suggesting that restoration of BRMS1 expression increased cell death during transit. Susceptibility to anoikis was verified in vitro by demonstrating decreased survival on poly-hydroxyethyl methacrylate-coated dishes. Most of the BRMS1-expressing cells reaching secondary sites failed to proliferate, suggesting that BRMS1 also inhibits colonization. Coupled with previous reports showing modest effects of BRMS1 on adhesion and invasion, our results indicate that BRMS1 inhibits metastases in multiple organs by blocking several steps in the metastatic cascade.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma/genética , Carcinoma/patologia , Genes Supressores de Tumor , Metástase Neoplásica/prevenção & controle , Proteínas de Neoplasias/fisiologia , Animais , Anoikis/genética , Progressão da Doença , Feminino , Genes Supressores de Tumor/fisiologia , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Proteínas Recombinantes de Fusão/genética , Proteínas Repressoras , Transplante Heterólogo , Células Tumorais Cultivadas
7.
J Natl Cancer Inst ; 99(4): 309-21, 2007 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-17312308

RESUMO

BACKGROUND: The KISS1 protein suppresses metastasis of several tumor models without blocking orthotopic tumor growth, but the mechanism remains elusive. For its role in human sexual maturation, KISS1 protein is secreted and processed to kisspeptins, which bind to the G protein-coupled receptor GPR54. We tested the hypothesis that KISS1 secretion is required for metastasis suppression via GPR54. METHODS: KISS1 containing an internal FLAG epitope with (KFM) or without (KFMdeltaSS) a signal sequence was transfected into C8161.9 human melanoma cells, which do not express endogenous KISS1. Whole-cell lysates and conditioned medium from C8161.9(KFM) and C8161.9(KFMdeltaSS) cells were collected and analyzed for kisspeptins by immunoprecipitation and enzyme-linked immunosorbent assay. GPR54 levels were measured using real-time reverse transcription-polymerase chain reaction. The ability of conditioned medium from C8161.9(KFM) and C8161.9(KFMdeltaSS) cells to stimulate calcium mobilization in GPR54-expressing Chinese hamster ovary cells (CHO-G) and in C8161.9 cells was evaluated. Metastasis was monitored in athymic mice (groups of 10 per experiment) that were injected with C8161.9(KFM) or C8161.9(KFMdeltaSS) cells labeled with enhanced green fluorescent protein. Survival of mice injected with C8161.9 or C8161.9(KFM) cells was analyzed by Kaplan-Meier methods. RESULTS: Full-length KFM and KFMdeltaSS were detected in whole-cell lysates of C8161.9(KFM) and C8161.9(KFMdeltaSS) cells, respectively, but kisspeptins were detected only in conditioned medium of C8161.9(KFM) cells. In vivo, C8161.9(KFM), but not C8161.9(KFMdeltaSS), cells were suppressed for metastasis to lung, eye, kidney, and bone, with corresponding differences in mouse survival (median > 120 versus 42 days). C8161.9(KFM) cells seeded mouse lungs but did not form macroscopic metastases. Conditioned medium from C8161.9(KFM), but not C8161.9(KFMdeltaSS), cells stimulated calcium mobilization in CHO-G cells. GPR54 expression was low in C8161.9 cells, which were not stimulated by conditioned medium from C8161.9(KFM) cells. CONCLUSIONS: KISS1 secretion was required for multiple organ metastasis suppression and for maintenance of disseminated cells in a dormant state. The absence of GPR54 expression in C8161.9 cells (whose metastatic spread was suppressed by KFM) suggests that metastasis suppression is not mediated through this receptor. The results imply the existence of another KISS1 receptor and/or paracrine signaling. The findings raise the possibility that soluble KISS1, kisspeptins, or mimetics could be used to maintain tumor dormancy, rendering treatment of already disseminated tumor cells (i.e., micrometastases) a legitimate target.


Assuntos
Melanoma/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Proteínas de Fluorescência Verde , Humanos , Estimativa de Kaplan-Meier , Kisspeptinas , Camundongos , RNA Mensageiro/análise , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
8.
Biochem Biophys Res Commun ; 348(4): 1429-35, 2006 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-16919237

RESUMO

Breast cancer metastasis suppressor 1 (BRMS1) is a member of the mSin3-HDAC transcription co-repressor complex. However, the proteins associated with BRMS1 have not been fully identified. Yeast two-hybrid screen, immuno-affinity chromatography, and co-immunoprecipitation experiments were performed to identify BRMS1 interacting proteins (BIPs). In addition to known core mSin3 transcriptional complex components RBBP1 and mSDS3, BRMS1 interacted with other proteins including three chaperones: DNAJB6 (MRJ), Hsp90, and Hsp70. Hsp90 is a known target of HDAC6 and reversible acetylation is one of the mechanisms that is implicated in regulation of Hsp90 chaperone complex activity. BRMS1 interacted with class II HDACs, HDAC 4, 5, and 6. We further found that BRMS1 is stabilized by Hsp90, and its turnover is proteasome dependent. The stability of BRMS1 protein may be important in maintaining the functional role of BRMS1 in metastasis suppression.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Células COS , Chlorocebus aethiops , Histona Desacetilases/metabolismo , Humanos , Imunoprecipitação , Proteínas Repressoras , Técnicas do Sistema de Duplo-Híbrido
9.
Clin Cancer Res ; 12(5): 1431-40, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16533765

RESUMO

PURPOSE: In vivo studies have focused on the latter stages of the bone metastatic process (osteolysis), whereas little is known about earlier events, e.g., arrival, localization, and initial colonization. Defining these initial steps may potentially identify the critical points susceptible to therapeutic intervention. EXPERIMENTAL DESIGN: MDA-MB-435 human breast cancer cells engineered with green fluorescent protein were injected into the cardiac left ventricle of athymic mice. Femurs were analyzed by fluorescence microscopy, immunohistochemistry, real-time PCR, flow cytometry, and histomorphometry at times ranging from 1 hour to 6 weeks. RESULTS: Single cells were found in distal metaphyses at 1 hour postinjection and remained as single cells up to 72 hours. Diaphyseal arrest occurred rarely and few cells remained there after 24 hours. At 1 week, numerous foci (2-10 cells) were observed, mostly adjacent to osteoblast-like cells. By 2 weeks, fewer but larger foci (> or =50 cells) were seen. Most bones had a single large mass at 4 weeks (originating from a colony or coalescing foci) which extended into the diaphysis by 4 to 6 weeks. Little change (<20%) in osteoblast or osteoclast numbers was observed at 2 weeks, but at 4 to 6 weeks, osteoblasts were dramatically reduced (8% of control), whereas osteoclasts were reduced modestly (to approximately 60% of control). CONCLUSIONS: Early arrest in metaphysis and minimal retention in diaphysis highlight the importance of the local milieu in determining metastatic potential. These results extend the Seed and Soil hypothesis by demonstrating both intertissue and intratissue differences governing metastatic location. Ours is the first in vivo evidence that tumor cells influence not only osteoclasts, as widely believed, but also eliminate functional osteoblasts, thereby restructuring the bone microenvironment to favor osteolysis. The data may also explain why patients receiving bisphosphonates fail to heal bone despite inhibiting resorption, implying that concurrent strategies that restore osteoblast function are needed to effectively treat osteolytic bone metastases.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Animais , Linhagem Celular Tumoral/transplante , Feminino , Fêmur/patologia , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Humanos , Imuno-Histoquímica , Cinética , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Osteoblastos/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Breast Cancer Res ; 7(5): R819-27, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16168128

RESUMO

INTRODUCTION: Polyamines affect proliferation, differentiation, migration and apoptosis of cells, indicating their potential as a target for cancer chemotherapy. Ornithine decarboxylase converts ornithine to putrescine and is the rate-limiting step in polyamine synthesis.alpha-Difluoromethylornithine (DFMO) irreversibly inhibits ornithine decarboxylase and MDA-MB-435 human breast cancer metastasis to the lung without blocking orthotopic tumor growth. This study tested the effects of DFMO on orthotopic tumor growth and lung colonization of another breast cancer cell line (MDA-MB-231) and the effects on bone metastasis of MDA-MB-435 cells. METHODS: MDA-MB-231 cells were injected into the mammary fat pad of athymic mice. DFMO treatment (2% per orally) began at the day of tumor cell injection or 21 days post injection. Tumor growth was measured weekly. MDA-MB-231 cells were injected into the tail vein of athymic mice. DFMO treatment began 7 days prior to injection, or 7 or 14 days post injection. The number and incidence of lung metastases were determined. Green fluorescent protein-tagged MDA-MB-435 cells were injected into the left cardiac ventricle in order to assess the incidence and extent of metastasis to the femur. DFMO treatment began 7 days prior to injection. RESULTS: DFMO treatment delayed MDA-MB-231 orthotopic tumor growth to a greater extent than growth of MDA-MB-435 tumors. The most substantial effect on lung colonization by MDA-MB-231 cells occurred when DFMO treatment began 7 days before intravenous injection of tumor cells (incidence decreased 28% and number of metastases per lung decreased 35-40%). When DFMO treatment began 7 days post injection, the incidence and number of metastases decreased less than 10%. Surprisingly, treatment initiated 14 days after tumor cell inoculation resulted in a nearly 50% reduction in the number of lung metastases without diminishing the incidence. After intracardiac injection, DFMO treatment decreased the incidence of bone metastases (55% vs 87%) and the area occupied by the tumor (1.66 mm2 vs 4.51 mm2, P < 0.05). CONCLUSION: Taken together, these data demonstrate that DFMO exerts an anti-metastatic effect in more than one hormone-independent breast cancer, for which no standard form of biologically-based treatment exists. Importantly, the data show that DFMO is effective against metastasis to multiple sites and that treatment is generally more effective when administered early.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Ósseas/prevenção & controle , Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Eflornitina/uso terapêutico , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Metástase Neoplásica/prevenção & controle , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA