Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(5): 1033-1038, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28096337

RESUMO

Breast tumors contain tumorigenic cancer cells, termed "tumor-initiating cells" (TICs), which are capable of both replenishing themselves and giving rise to populations of nontumorigenic breast cancer cells (non-TICs). However, the molecular mechanisms responsible for breast tumor initiation remain poorly understood. Here we describe a chemical screening strategy to identify small molecules that enhance the effect of chemotherapeutic agents on TIC-enriched breast cancer cells. We identified proteins that interact with the lead compound C108, including the stress granule-associated protein, GTPase-activating protein (SH3 domain)-binding protein 2, G3BP2. G3BP2 regulates breast tumor initiation through the stabilization of Squamous cell carcinoma antigen recognized by T cells 3 (SART3) mRNA, which leads to increased expression of the pluripotency transcription factors Octamer-binding protein 4 (Oct-4) and Nanog Homeobox (Nanog). Our findings suggest that G3BP2 is important for the process of breast cancer initiation. Furthermore, these data suggest a possible connection between stress granule formation and tumor initiation in breast cancer cells.


Assuntos
Neoplasias da Mama/etiologia , Carcinogênese , Proteínas de Transporte/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Antineoplásicos/isolamento & purificação , Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Grânulos Citoplasmáticos/fisiologia , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteína Homeobox Nanog/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Paclitaxel/farmacologia , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Neoplásico/metabolismo , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/genética , Bibliotecas de Moléculas Pequenas
2.
Proc Natl Acad Sci U S A ; 108(28): 11596-601, 2011 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-21709229

RESUMO

Antivascular agents have become a standard of treatment for many malignancies. However, most of them target the VEGF pathway and lead to refractoriness. To improve the diversity of options for antivascular therapy, we applied a high-throughput screen for small molecules targeting cell adhesion. We then assayed the resulting antiadhesion hits in a transgenic zebrafish line with endothelial expression of EGFP (Tg(fli1:EGFP)(y1)) to identify nontoxic molecules with antivascular activity selective to neovasculature. This screen identified dehydro-α-lapachone (DAL), a natural plant product. We found that DAL inhibits vessel regeneration, interferes with vessel anastomosis, and limits plexus formation in zebrafish. Furthermore, DAL induces vascular pruning and growth delay in orthotopic mammary tumors in mice. We show that DAL targets cell adhesion by promoting ubiquitination of the Rho-GTPase Rac1, which is frequently up-regulated in many different cancers.


Assuntos
Inibidores da Angiogênese/farmacologia , Naftoquinonas/farmacologia , Inibidores da Angiogênese/isolamento & purificação , Animais , Animais Geneticamente Modificados , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Feminino , Proteínas de Fluorescência Verde/genética , Ensaios de Triagem em Larga Escala , Humanos , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos SCID , Naftoquinonas/isolamento & purificação , Plantas Medicinais/química , Tabebuia/química , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
3.
Cancer Cell ; 6(6): 553-63, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15607960

RESUMO

The recent landmark Phase III clinical trial with a VEGF-specific antibody suggests that antiangiogenic therapy must be combined with cytotoxic therapy for the treatment of solid tumors. However, there are no guidelines for optimal scheduling of these therapies. Here we show that VEGFR2 blockade creates a "normalization window"--a period during which combined radiation therapy gives the best outcome. This window is characterized by an increase in tumor oxygenation, which is known to enhance radiation response. During the normalization window, but not before or after it, VEGFR2 blockade increases pericyte coverage of brain tumor vessels via upregulation of Ang1 and degrades their pathologically thick basement membrane via MMP activation.


Assuntos
Vasos Sanguíneos/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Inibidores da Angiogênese/uso terapêutico , Angiopoietina-1/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antígenos/análise , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Membrana Basal/efeitos dos fármacos , Membrana Basal/metabolismo , Membrana Basal/patologia , Vasos Sanguíneos/química , Vasos Sanguíneos/efeitos da radiação , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Colágeno Tipo IV/análise , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Terapia Combinada/métodos , Dipeptídeos/farmacologia , Efrina-B2/genética , Angiofluoresceinografia , Raios gama/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/metabolismo , Glioma/radioterapia , Humanos , Imuno-Histoquímica , Masculino , Inibidores de Metaloproteinases de Matriz , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Nus , Modelos Biológicos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/radioterapia , Análise de Sequência com Séries de Oligonucleotídeos , Oxigênio/metabolismo , Pericitos/química , Pericitos/citologia , Pericitos/fisiologia , Proteoglicanas/análise , Receptor TIE-2/antagonistas & inibidores , Receptor TIE-2/imunologia , Fatores de Tempo , Transfecção , Regulação para Cima/genética
4.
Mol Oncol ; 2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-33525064

RESUMO

Mounting evidence suggests that cancer stemness and immunosuppression are related, but the underlying mechanisms behind these are not clear. We previously reported that the stress granule-associated protein G3BP2 is involved in the regulation of tumor-initiating (stem) cells. In this study, we show that this protein also upregulates the immune checkpoint molecule PD-L1 under conditions of stress in breast and glioblastoma cancer cells, revealing a previously unknown connection between stemness programs, stress responses, and immune checkpoint control. We also identified a significant correlation between G3BP2 and PD-L1 co-expression in tumor tissues from cancer patients. To assess the targetability of G3BP2, we employed a small molecule (C108) that binds G3BP2 and interferes with the stress response. Tumors treated with C108 had increased CD8 T-cell proliferation and infiltration. Moreover, treatment of breast tumor-bearing mice with C108 resulted in a significant survival benefit and long-term cures. Cancer cells treated with C108 or cancer cells with genetically repressed G3BP2 had decreased PD-L1 expression due to enhanced mRNA degradation. Our study provides a compelling mechanism linking stress granule formation and immune checkpoint program of cancer, suggesting this link may provide new opportunities for improving anticancer immunotherapy.

5.
Nature ; 428(6980): 328-32, 2004 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-15029197

RESUMO

Gliomas are the most common primary tumours of the central nervous system, with nearly 15,000 diagnosed annually in the United States and a lethality approaching 80% within the first year of glioblastoma diagnosis. The marked induction of angiogenesis in glioblastomas suggests that it is a necessary part of malignant progression; however, the precise molecular mechanisms underlying the regulation of brain tumour growth and angiogenesis remain unresolved. Here we report that a candidate tumour suppressor gene, ING4, is involved in regulating brain tumour growth and angiogenesis. Expression of ING4 is significantly reduced in gliomas as compared with normal human brain tissue, and the extent of reduction correlates with the progression from lower to higher grades of tumours. In mice, xenografts of human glioblastoma U87MG, which has decreased expression of ING4, grow significantly faster and have higher vascular volume fractions than control tumours. We show that ING4 physically interacts with p65 (RelA) subunit of nuclear factor NF-kappaB, and that ING4 regulates brain tumour angiogenesis through transcriptional repression of NF-kappaB-responsive genes. These results indicate that ING4 has an important role in brain tumour pathogenesis.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Neovascularização Patológica , Proteínas Supressoras de Tumor/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proteínas de Ciclo Celular , Divisão Celular , Linhagem Celular Tumoral , Ciclo-Oxigenase 2 , Progressão da Doença , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/irrigação sanguínea , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Proteínas de Homeodomínio , Humanos , Interleucina-6/genética , Interleucina-8/genética , Isoenzimas/genética , Proteínas de Membrana , Camundongos , Camundongos SCID , NF-kappa B/metabolismo , Transplante de Neoplasias , Prostaglandina-Endoperóxido Sintases/genética , Ligação Proteica , Fator de Transcrição RelA , Proteínas Supressoras de Tumor/genética
6.
Cancer Res ; 67(11): 5076-82, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17545583

RESUMO

The effects of antiangiogenic therapy on tumors relapsing after irradiation are not known. To this end, we irradiated human tumors growing s.c. in nude mice with a single dose of 20 or 30 Gy. Compared with primary (treatment-naive) xenografts, the growth rate of recurrent tumors was 1.6-fold slower, which is consistent with the known "tumor bed effect." For similar size tumors, recurrences had fewer functional vessels, a reduced vessel coverage by perivascular cells, and were more necrotic. Placenta growth factor concentration was significantly lower in relapses, whereas vascular endothelial growth factor (VEGF) levels were similar between primary and recurrent tumors. On the other hand, fibrillar collagen deposition was significantly increased in recurrent tumors. This radiation-induced fibrosis was partially responsible for the slower growth of recurrences; the i.t. injection of collagenase increased the growth rate of tumor relapses without affecting primary tumor growth. The mouse-specific VEGF receptor 2-blocking antibody DC101 induced a 2.2-fold longer growth delay in recurrent tumors compared with treatment-naive tumors. DC101 significantly decreased the interstitial fluid pressure and did not change the functional vessel density and perivascular cell coverage in both tumor variants. Interestingly, DC101 induced a rapid (2 days after treatment initiation) and significant decrease in tumor cell proliferation in recurrent but not in primary tumors. Thus, our results show that the stromal compartment and the response to antiangionenic therapy of primary and in-field recurrent tumors are significantly different. Our findings suggest that antiangiogenic agents could be effective in the treatment of patients with relapses after radiotherapy.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/radioterapia , Recidiva Local de Neoplasia/irrigação sanguínea , Recidiva Local de Neoplasia/tratamento farmacológico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Inibidores da Angiogênese/farmacologia , Animais , Processos de Crescimento Celular/efeitos dos fármacos , Colágeno Tipo I/metabolismo , Colagenases/farmacologia , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Camundongos , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Fator de Crescimento Placentário , Proteínas da Gravidez/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Oncotarget ; 8(65): 108292-108302, 2017 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-29312532

RESUMO

SLPI has been implicated in the progression and metastasis of certain cancers. However, the effects of SLPI seem to be tumor-specific and the mechanisms remain poorly defined. Here, we demonstrate that highly metastatic, triple-negative breast cancer (TNBC) 4T1 cells secreted more SLPI compared to their non-metastatic counterparts. Furthermore, SLPI secretion directly correlated with spontaneous lung metastasis from 4T1 tumors orthotopically implanted in mice. Consistent with our experimental results, we also found that higher SLPI expression levels correlate with worse clinical outcome in basal/TNBC patients. Using high-throughput screening we identified a novel compound, C74, which significantly inhibits SLPI secretion. C74 administration in our mouse model slows the growth of primary 4T1 tumors and inhibits their dissemination to the lung. We also discovered that SLPI physically interacts with the retinoblastoma tumor suppressor protein (Rb) and releases FoxM1 from the Rb-FoxM1 complex, which may activate FoxM1 target genes involved in breast cancer metastasis.

8.
Circ Res ; 93(9): e88-97, 2003 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-14525808

RESUMO

With an increasing incidence of obesity worldwide, rational strategies are needed to control adipogenesis. Growth of any tissue requires the formation of a functional and mature vasculature. To gain mechanistic insight into the link between active adipogenesis and angiogenesis, we developed a model to visualize noninvasively and in real time both angiogenesis and adipogenesis using intravital microscopy. Implanted murine preadipocytes induced vigorous angiogenesis and formed fat pads in a mouse dorsal skin-fold chamber. The newly formed vessels subsequently remodeled into a mature network consisting of arterioles, capillaries, and venules, whereas the preadipocytes differentiated into adipocytes as confirmed by increased aP2 expression. Inhibition of adipocyte differentiation by transfection of preadipocytes with a peroxisome proliferator-activated receptor gamma dominant-negative construct not only abrogated fat tissue formation but also reduced angiogenesis. Surprisingly, inhibition of angiogenesis by vascular endothelial growth factor receptor-2 (VEGFR2) blocking antibody not only reduced angiogenesis and tissue growth but also inhibited preadipocyte differentiation. We found that part of this inhibition stems from the paracrine interaction between endothelial cells and preadipocytes and that VEGF-VEGFR2 signaling in endothelial cells, but not preadipocytes, mediates this process. These findings reveal a reciprocal regulation of adipogenesis and angiogenesis, and suggest that blockade of VEGF signaling can inhibit in vivo adipose tissue formation. The full text of this article is available online at http://www.circresaha.org.


Assuntos
Adipócitos/fisiologia , Diferenciação Celular/fisiologia , Neovascularização Fisiológica , Comunicação Parácrina/fisiologia , Células 3T3 , Adipócitos/citologia , Adipócitos/transplante , Animais , Anticorpos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Cultura em Câmaras de Difusão , Genes Dominantes , Masculino , Camundongos , Camundongos SCID , Neovascularização Fisiológica/efeitos dos fármacos , Comunicação Parácrina/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/genética , Transplante de Células-Tronco , Fatores de Transcrição/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
9.
Oncotarget ; 7(28): 44406-44416, 2016 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-27259271

RESUMO

Gastric cancer is a prevalent tumor that is usually detected at an advanced metastatic stage. Currently, standard therapies are mostly ineffective. Here, we report that Glypican-3 (GPC3) is absent in invasive tumors and metastatic lymph nodes, in particular in aggressive and highly disseminated signet ring cell carcinomas. We demonstrate that loss of GPC3 correlates with poor overall survival in patients. Moreover, we show that absence of GPC3 causes up-regulation of MAPK/FoxM1 signaling and that blockade of this pathway alters cellular invasion. An inverse correlation between GPC3 and FoxM1 is also shown in patient samples. These data identify GPC3 as a potential metastasis suppressor gene and suggest its value as a prognostic marker in gastric cancer. Development of therapies targeting signaling downstream of GPC3 are warranted.


Assuntos
Glipicanas/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Feminino , Genes Supressores de Tumor , Glipicanas/deficiência , Glipicanas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Neoplasias Gástricas/metabolismo
10.
FEBS Lett ; 524(1-3): 163-71, 2002 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-12135761

RESUMO

The p53 tumor suppressor protein induces cell cycle arrest or apoptosis in response to cellular stresses. We have identified PRG3 (p53-responsive gene 3), which is induced specifically under p53-dependent apoptotic conditions in human colon cancer cells, and encodes a novel polypeptide of 373 amino acids with a predicted molecular mass of 40.5 kDa. PRG3 has significant homology to bacterial oxidoreductases and the apoptosis-inducing factor, AIF, and the gene was assigned to chromosome 10q21.3-q22.1. Expression of PRG3 was induced by the activation of endogenous p53 and it contains a p53-responsive element. Unlike AIF, PRG3 localizes in the cytoplasm and its ectopic expression induces apoptosis. An amino-terminal deletion mutant of PRG3 that lacks a putative oxidoreductase activity retains its apoptotic activity, suggesting that the oxidoreductase activity is dispensable for the apoptotic function of PRG3. The PRG3 gene is thus a novel p53 target gene in a p53-dependent apoptosis pathway.


Assuntos
Flavoproteínas/genética , Regulação da Expressão Gênica/fisiologia , Proteínas de Membrana/genética , Proteínas Mitocondriais , Proteínas/genética , Proteína Supressora de Tumor p53/fisiologia , Difosfato de Adenosina/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Fator de Indução de Apoptose , Proteínas Reguladoras de Apoptose , Sequência de Bases , Sítios de Ligação , Mapeamento Cromossômico , Cromossomos Humanos Par 10 , Clonagem Molecular , DNA , Genes Reporter , Humanos , Hibridização in Situ Fluorescente , Dados de Sequência Molecular , Proteínas/química , RNA Mensageiro/genética , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas
12.
Neoplasia ; 14(2): 84-94, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22431917

RESUMO

Neurofibromatosis type 2 (NF2) is an autosomal-dominant multiple neoplasia syndrome that results from mutations in the NF2 tumor suppressor gene. Patients with NF2 develop hallmark schwannomas that require surgery or radiation, both of which have significant adverse effects. Recent studies have indicated that the tumor microenvironment-in particular, tumor blood vessels-of schwannomas may be an important therapeutic target. Furthermore, although much has been done to understand how merlin, the NF2 gene product, functions as a tumor suppressor gene in schwannoma cells, the functional role of merlin in the tumor microenvironment and the mechanism(s) by which merlin regulates angiogenesis to support schwannoma growth is largely unexplored. Here we report that the expression of semaphorin 3F (SEMA3F) was specifically downregulated in schwannoma cells lacking merlin/NF2. When we reintroduced SEMA3F in schwannoma cells, we observed normalized tumor blood vessels, reduced tumor burden, and extended survival in nude mice bearing merlin-deficient brain tumors. Next, using chemical inhibitors and gene knockdown with RNA interference, we found that merlin regulated expression of SEMA3F through Rho GTPase family member Rac1. This study shows that, in addition to the tumor-suppressing activity of merlin, it also functions to maintain physiological angiogenesis in the nervous system by regulating antiangiogenic factors such as SEMA3F. Restoring the relative balance of proangiogenic and antiangiogenic factors, such as increases in SEMA3F, in schwannoma microenvironment may represent a novel strategy to alleviate the clinical symptoms of NF2-related schwannomas.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Proteínas de Membrana/metabolismo , Neovascularização Patológica , Proteínas do Tecido Nervoso/metabolismo , Neurilemoma/irrigação sanguínea , Neurofibromina 2/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Vasos Sanguíneos/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Regulação para Baixo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Estimativa de Kaplan-Meier , Proteínas de Membrana/genética , Camundongos , Camundongos Nus , Transplante de Neoplasias , Proteínas do Tecido Nervoso/genética , Neurilemoma/metabolismo , Neurilemoma/patologia , Neurofibromatose 2/metabolismo , Neurofibromatose 2/patologia , Neurofibromina 2/genética , Permeabilidade , Transdução de Sinais , Trombospondinas/genética , Trombospondinas/metabolismo
13.
Cancer Res ; 71(13): 4527-38, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21571860

RESUMO

Functional roles for the cancer cell-associated membrane type I matrix metalloproteinase (MT1-MMP) during early steps of the metastatic cascade in primary tumors remain unresolved. In an effort to determine its significance, we determined the in vivo effects of RNAi-mediated downregulation in mammary cancer cells on the migration, blood and lymphatic vessel invasion (LVI), and lymph node and lung metastasis. We also correlated the expression of cancer cell MT1-MMP with blood vessel invasion (BVI) in 102 breast cancer biopsies. MT1-MMP downregulation in cancer cells decreased lung metastasis without affecting primary tumor growth. The inhibition of lung metastasis correlated with reduced cancer cell migration and BVI. Furthermore, cancer cell-expressed MT1-MMP upregulated the expression of MT1-MMP in vascular endothelial cells, but did not affect MT1-MMP expression in lymphatic endothelial cells, LVI, or lymph node metastasis. Of clinical importance, we observed that elevated MT1-MMP expression correlated with BVI in biopsies from triple-negative breast cancers (TNBC), which have a poor prognosis and high incidence of distant metastasis, relative to other breast cancer subtypes. Together, our findings established that MT1-MMP activity in breast tumors is essential for BVI, but not LVI, and that MT1-MMP should be further explored as a predictor and therapeutic target of hematogenous metastasis in TNBC patients.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Metaloproteinase 14 da Matriz/biossíntese , Animais , Neoplasias da Mama/enzimologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Regulação para Baixo , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Metástase Linfática , Metaloproteinase 14 da Matriz/metabolismo , Camundongos , Camundongos SCID , Metástase Neoplásica , Neovascularização Patológica/enzimologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Ratos , Receptor ErbB-2/biossíntese , Receptor ErbB-2/deficiência , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/deficiência , Receptores de Progesterona/biossíntese , Receptores de Progesterona/deficiência
15.
Neoplasia ; 9(12): 1038-45, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18084611

RESUMO

Blood vascular endothelial cells (BECs) and the developmentally related lymphatic endothelial cells (LECs) create complementary, yet distinct vascular networks. Each endothelial cell type interacts with flowing fluid and circulating cells, yet each vascular system has evolved specialized gene expression programs and thus both cell types display different phenotypes. BECs and LECs express distinct genes that are unique to their specific vascular microenvironment. Tumors also take advantage of the molecules that are expressed in these vascular systems to enhance their metastatic potential. We completed transcriptome analyses on primary cultured LECs and BECs, where each comparative set was isolated from the same individual. Differences were resolved in the expression of several major categories, such as cell adhesion molecules (CAMs), cytokines, and cytokine receptors. We have identified new molecules that are associated with BECs (e.g., claudin-9, CXCL11, neurexin-1, neurexin-2, and the neuronal growth factor regulator-1) and LECs (e.g., claudin-7, CD58, hyaluronan and proteoglycan link protein 1 (HAPLN1), and the poliovirus receptor-related 3 molecule) that may lead to novel therapeutic treatments for diseases of lymphatic or blood vessels, including metastasis of cancer to lymph nodes or distant organs.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Regulação da Expressão Gênica , Vasos Linfáticos/citologia , Células Cultivadas/metabolismo , Perfilação da Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa