Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Transl Med ; 15(1): 175, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28810879

RESUMO

BACKGROUND: Speckle-type POZ protein (SPOP) is an E3 ubiquitin ligase adaptor protein that functions as a potential tumor suppressor, and SPOP mutations have been identified in ~10% of human prostate cancers. However, it remains unclear if mutant SPOP proteins can be utilized as biomarkers for early detection, diagnosis, prognosis or targeted therapy of prostate cancer. Moreover, the SPOP mutation sites are distributed in a relatively short region with multiple lysine residues, posing significant challenges for bottom-up proteomics analysis of the SPOP mutations. METHODS: To address this issue, PRISM (high-pressure, high-resolution separations coupled with intelligent selection and multiplexing)-SRM (selected reaction monitoring) mass spectrometry assays have been developed for quantifying wild-type SPOP protein and 11 prostate cancer-derived SPOP mutations. RESULTS: Despite inherent limitations due to amino acid sequence constraints, all the PRISM-SRM assays developed using Arg-C digestion showed a linear dynamic range of at least two orders of magnitude, with limits of quantification ranged from 0.1 to 1 fmol/µg of total protein in the cell lysate. Applying these SRM assays to analyze HEK293T cells with and without expression of the three most frequent SPOP mutations in prostate cancer (Y87N, F102C or F133V) led to confident detection of all three SPOP mutations in corresponding positive cell lines but not in the negative cell lines. Expression of the F133V mutation and wild-type SPOP was at much lower levels compared to that of F102C and Y87N mutations; however, at present, it is unknown if this also affects the biological activity of the SPOP protein. CONCLUSIONS: In summary, PRISM-SRM enables multiplexed, isoform-specific detection of mutant SPOP proteins in cell lysates, providing significant potential in biomarker development for prostate cancer.


Assuntos
Espectrometria de Massas/métodos , Mutação/genética , Proteínas Nucleares/genética , Neoplasias da Próstata/genética , Proteômica/métodos , Proteínas Repressoras/genética , Sequência de Aminoácidos , Células HEK293 , Humanos , Limite de Detecção , Masculino , Peptídeos/química , Peptídeos/metabolismo
2.
bioRxiv ; 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38895216

RESUMO

Osteosarcoma (OS) is the most common primary pediatric bone malignancy. One promising new therapeutic target is SKP2, encoding a substrate recognition factor of the SCF E3 ubiquitin ligase responsible for ubiquitination and proteasome degradation of substrate p27, thus driving cellular proliferation. We have shown previously that knockout of Skp2 in an immunocompetent transgenic mouse model of OS improved survival, drove apoptosis, and induced tumor inflammation. Here, we applied single-cell RNA-sequencing (scRNA-seq) to study primary OS tumors derived from Osx-Cre driven conditional knockout of Rb1 and Trp53. We showed that murine OS models recapitulate the tumor heterogeneity and microenvironment complexity observed in patient tumors. We further compared this model with OS models with functional disruption of Skp2: one with Skp2 knockout and the other with the Skp2-p27 interaction disrupted (resulting in p27 overexpression). We found reduction of T cell exhaustion and upregulation of interferon activation, along with evidence of replicative and endoplasmic reticulum-related stress in the Skp2 disruption models, and showed that interferon induction was correlated with improved survival in OS patients. Additionally, our scRNA-seq analysis uncovered decreased activities of metastasis-related gene signatures in the Skp2-disrupted OS, which we validated by observation of a strong reduction in lung metastasis in the Skp2 knockout mice. Finally, we report several potential mechanisms of escape from targeting Skp2 in OS, including upregulation of Myc targets, DNA copy number amplification and overexpression of alternative E3 ligase genes, and potential alternative lineage activation. These mechanistic insights into OS tumor biology and Skp2 function suggest novel targets for new, synergistic therapies, while the data and our comprehensive analysis may serve as a public resource for further big data-driven OS research.

3.
Nat Methods ; 7(8): 655-60, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20581828

RESUMO

Not all tumor vessels are equal. Tumor-associated vasculature includes immature vessels, regressing vessels, transport vessels undergoing arteriogenesis and peritumor vessels influenced by tumor growth factors. Current techniques for analyzing tumor blood flow do not discriminate between vessel subtypes and only measure average changes from a population of dissimilar vessels. We developed methodologies for simultaneously quantifying blood flow (velocity, flux, hematocrit and shear rate) in extended networks at single-capillary resolution in vivo. Our approach relies on deconvolution of signals produced by labeled red blood cells as they move relative to the scanning laser of a confocal or multiphoton microscope and provides fully resolved three-dimensional flow profiles within vessel networks. Using this methodology, we show that blood velocity profiles are asymmetric near intussusceptive tissue structures in tumors in mice. Furthermore, we show that subpopulations of vessels, classified by functional parameters, exist in and around a tumor and in normal brain tissue.


Assuntos
Eritrócitos/citologia , Microcirculação , Neoplasias/irrigação sanguínea , Animais , Velocidade do Fluxo Sanguíneo , Hematócrito , Hemorreologia , Camundongos
4.
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
5.
Nat Commun ; 11(1): 5549, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33144576

RESUMO

Advanced prostate cancer initially responds to hormonal treatment, but ultimately becomes resistant and requires more potent therapies. One mechanism of resistance observed in around 10-20% of these patients is lineage plasticity, which manifests in a partial or complete small cell or neuroendocrine prostate cancer (NEPC) phenotype. Here, we investigate the role of the mammalian SWI/SNF (mSWI/SNF) chromatin remodeling complex in NEPC. Using large patient datasets, patient-derived organoids and cancer cell lines, we identify mSWI/SNF subunits that are deregulated in NEPC and demonstrate that SMARCA4 (BRG1) overexpression is associated with aggressive disease. We also show that SWI/SNF complexes interact with different lineage-specific factors in NEPC compared to prostate adenocarcinoma. These data point to a role for mSWI/SNF complexes in therapy-related lineage plasticity, which may also be relevant for other solid tumors.


Assuntos
Linhagem da Célula , Plasticidade Celular , Proteínas Cromossômicas não Histona/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Fatores de Transcrição/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Linhagem Celular Tumoral , Estudos de Coortes , DNA Helicases/genética , DNA Helicases/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Masculino , Modelos Biológicos , Invasividade Neoplásica , Proteínas de Neoplasias/metabolismo , Tumores Neuroendócrinos/metabolismo , Tumores Neuroendócrinos/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias da Próstata/genética , Subunidades Proteicas/metabolismo , Fatores de Transcrição/genética , Transcriptoma/genética
6.
J Clin Invest ; 114(8): 1082-9, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15489955

RESUMO

Angiogenesis, or new blood vessel formation, is critical for the growth and spread of tumors. Multiple phases of this process, namely, migration, proliferation, morphogenesis, and vascular stabilization, are needed for optimal tumor growth beyond a diffusion-limited size. The sphingosine 1-phosphate (S1P) receptor-1 (S1P(1)) is required for stabilization of nascent blood vessels during embryonic development. Here we show that S1P(1) expression is strongly induced in tumor vessels. We developed a multiplex RNA interference technique to downregulate S1P(1) in mice. The small interfering RNA (siRNA) for S1P(1) specifically silenced the cognate transcript in endothelial cells and inhibited endothelial cell migration in vitro and the growth of neovessels into subcutaneous implants of Matrigel in vivo. Local injection of S1P(1) siRNA, but not a negative control siRNA, into established tumors inhibited the expression of S1P(1) polypeptide on neovessels while concomitantly suppressing vascular stabilization and angiogenesis, which resulted in dramatic suppression of tumor growth in vivo. These data suggest that S1P(1) is a critical component of the tumor angiogenic response and argue for the utility of siRNA technology in antiangiogenic therapeutics.


Assuntos
Neoplasias/irrigação sanguínea , Neovascularização Patológica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Células Cultivadas , Colágeno/metabolismo , Combinação de Medicamentos , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Regulação Neoplásica da Expressão Gênica , Laminina/metabolismo , Camundongos , Transplante de Neoplasias , Neoplasias/metabolismo , Proteoglicanas/metabolismo , RNA Interferente Pequeno/genética , Receptores de Lisoesfingolipídeo/genética , Transplante Heterólogo
7.
Elife ; 42015 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-26374986

RESUMO

Genomic instability is a fundamental feature of human cancer often resulting from impaired genome maintenance. In prostate cancer, structural genomic rearrangements are a common mechanism driving tumorigenesis. However, somatic alterations predisposing to chromosomal rearrangements in prostate cancer remain largely undefined. Here, we show that SPOP, the most commonly mutated gene in primary prostate cancer modulates DNA double strand break (DSB) repair, and that SPOP mutation is associated with genomic instability. In vivo, SPOP mutation results in a transcriptional response consistent with BRCA1 inactivation resulting in impaired homology-directed repair (HDR) of DSB. Furthermore, we found that SPOP mutation sensitizes to DNA damaging therapeutic agents such as PARP inhibitors. These results implicate SPOP as a novel participant in DSB repair, suggest that SPOP mutation drives prostate tumorigenesis in part through genomic instability, and indicate that mutant SPOP may increase response to DNA-damaging therapeutics.


Assuntos
Instabilidade Genômica , Proteínas Nucleares/deficiência , Neoplasias da Próstata/patologia , Proteínas Repressoras/deficiência , Animais , Células Cultivadas , Quebras de DNA de Cadeia Dupla , Dano ao DNA/efeitos dos fármacos , Reparo do DNA , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Mutagênicos/metabolismo , Complexos Ubiquitina-Proteína Ligase , Peixe-Zebra
8.
Prostaglandins Other Lipid Mediat ; 73(1-2): 141-50, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15165038

RESUMO

S1P1 (also known as EDG-1) is a G-protein coupled receptor for the bioactive lipid, sphingosine-1-phosphate (S1P). Activation of S1P1 receptor in endothelial cells induces diverse cellular effects, including cell proliferation, survival, migration and morphogenesis. Recent in vivo studies showed that the S1P1 receptor is required in vascular maturation during development. While a number of studies reported a functional role of S1P1 in vascular system and the presence of S1P1 transcripts in various mouse organs, tissue distribution of S1P1 has not been fully defined. In this study, we determined the expression pattern of S1P1 by beta-galactosidase reporter gene expression, which is knocked into the S1P1 locus. We show that S1P1 is widely expressed in various cell types of adult mouse tissues, suggesting a regulatory role of this receptor in numerous physiological processes in both vascular and non-vascular tissues.


Assuntos
Estruturas Animais/metabolismo , Lisofosfolipídeos/metabolismo , Receptores de Lisoesfingolipídeo/biossíntese , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Animais , Animais Geneticamente Modificados , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Endoteliais/metabolismo , Galactosidases , Histocitoquímica , Camundongos , Especificidade de Órgãos , Receptores de Lisoesfingolipídeo/genética
9.
Mol Oncol ; 8(7): 1169-80, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25266362

RESUMO

Fusions between the transmembrane protease serine 2 (TMPRSS2) and ETS related gene (ERG) represent one of the most specific biomarkers that define a distinct molecular subtype of prostate cancer. Studies of TMPRSS2-ERG gene fusions have seldom been performed at the protein level, primarily due to the lack of high-quality antibodies suitable for quantitative studies. Herein, we applied a recently developed PRISM (high-pressure high-resolution separations with intelligent selection and multiplexing)-SRM (selected reaction monitoring) strategy for quantifying ERG protein in prostate cancer cell lines and tumors. The highly sensitive PRISM-SRM assays provided confident detection of 6 unique ERG peptides in both TMPRSS2-ERG positive cell lines and tissues, but not in cell lines or tissues lacking the TMPRSS2-ERG rearrangement, clearly indicating that ERG protein expression is significantly increased in the presence of the TMPRSS2-ERG gene fusion. Significantly, our results provide evidence that two distinct ERG protein isoforms are simultaneously expressed in TMPRSS2-ERG positive samples as evidenced by the concomitant detection of two mutually exclusive peptides in two patient tumors and in the VCaP prostate cancer cell line. Three peptides, shared across almost all fusion protein products, were determined to be the most abundant peptides, providing "signature" peptides for detection of ERG over-expression resulting from TMPRSS2-ERG gene fusion. The PRISM-SRM assays provide valuable tools for studying TMPRSS2-ERG gene fusion protein products in prostate cancer.


Assuntos
Proteínas de Fusão Oncogênica/análise , Próstata/patologia , Neoplasias da Próstata/patologia , Sequência de Aminoácidos , Linhagem Celular Tumoral , Rearranjo Gênico , Humanos , Masculino , Dados de Sequência Molecular , Proteínas de Fusão Oncogênica/genética , Peptídeos/análise , Peptídeos/genética , Próstata/metabolismo , Neoplasias da Próstata/genética
10.
Nat Genet ; 44(6): 685-9, 2012 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-22610119

RESUMO

Prostate cancer is the second most common cancer in men worldwide and causes over 250,000 deaths each year. Overtreatment of indolent disease also results in significant morbidity. Common genetic alterations in prostate cancer include losses of NKX3.1 (8p21) and PTEN (10q23), gains of AR (the androgen receptor gene) and fusion of ETS family transcription factor genes with androgen-responsive promoters. Recurrent somatic base-pair substitutions are believed to be less contributory in prostate tumorigenesis but have not been systematically analyzed in large cohorts. Here, we sequenced the exomes of 112 prostate tumor and normal tissue pairs. New recurrent mutations were identified in multiple genes, including MED12 and FOXA1. SPOP was the most frequently mutated gene, with mutations involving the SPOP substrate-binding cleft in 6-15% of tumors across multiple independent cohorts. Prostate cancers with mutant SPOP lacked ETS family gene rearrangements and showed a distinct pattern of genomic alterations. Thus, SPOP mutations may define a new molecular subtype of prostate cancer.


Assuntos
Fator 3-alfa Nuclear de Hepatócito/genética , Complexo Mediador/genética , Proteínas Associadas aos Microtúbulos/genética , Mutação , Neoplasias da Próstata/genética , Exoma , Humanos , Masculino , Análise de Sequência de DNA
11.
Am J Cancer Res ; 1(2): 144-54, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21822499

RESUMO

We sought to characterize the function of bone marrow stromal cell (BMSC) populations in tumor progression. Because this function may depend on the cell-lineage and mouse strain heterogeneity, we first characterized ex vivo the BMSCs harvested from C57BL/6 versus FVB mice and established their in vivo function in tumor growth and metastasis experiments. All plastic-adherent BMSCs expressed platelet-derived growth factor receptor beta (PDGFRß) and stem cell antigen 1 (Sca1), consistent with a mesenchymal precursor phenotype, as well as CD80. Moreover, these BMSCs were capable of differentiation along mesenchymal lineage into adipocytes, osteoblasts, chondrocytes or myofibroblasts. However, further phenotypic analysis detected a distinct populations of myeloid (CD11b(+)) precursor cells amongst the ex vivo expanded BMSCs -with specific surface marker phenotypes and gene expression pattern. When co-implanted with metastatic cancer cells, all the BMSCs persisted and integrated into tumor stroma, but only myeloid BMSCs significantly promoted tumor growth and metastasis. These data demonstrate the differential effect of BMSCs sub-populations on tumor progression. These results may have important implications for anti-tumor therapy and for the use of mesenchymal BMSCs as cell-based therapies.

12.
Cancer Discov ; 1(6): 487-95, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22389870

RESUMO

UNLABELLED: Neuroendocrine prostate cancer (NEPC) is an aggressive subtype of prostate cancer that most commonly evolves from preexisting prostate adenocarcinoma (PCA). Using Next Generation RNA-sequencing and oligonucleotide arrays, we profiled 7 NEPC, 30 PCA, and 5 benign prostate tissue (BEN), and validated findings on tumors from a large cohort of patients (37 NEPC, 169 PCA, 22 BEN) using IHC and FISH. We discovered significant overexpression and gene amplification of AURKA and MYCN in 40% of NEPC and 5% of PCA, respectively, and evidence that that they cooperate to induce a neuroendocrine phenotype in prostate cells. There was dramatic and enhanced sensitivity of NEPC (and MYCN overexpressing PCA) to Aurora kinase inhibitor therapy both in vitro and in vivo, with complete suppression of neuroendocrine marker expression following treatment. We propose that alterations in Aurora kinase A and N-myc are involved in the development of NEPC, and future clinical trials will help determine from the efficacy of Aurora kinase inhibitor therapy. SIGNIFICANCE: We report on the largest in-depth molecular analysis of NEPC and provide new insight into molecular events involved in the progression of prostate cancer.


Assuntos
Antineoplásicos/uso terapêutico , Tumores Neuroendócrinos/tratamento farmacológico , Tumores Neuroendócrinos/genética , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Inibidores de Proteínas Quinases/uso terapêutico , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Aurora Quinase A , Aurora Quinases , Carcinoma de Células Pequenas/tratamento farmacológico , Carcinoma de Células Pequenas/genética , Carcinoma de Células Pequenas/metabolismo , Linhagem Celular Tumoral , Estudos de Coortes , Progressão da Doença , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Humanos , Masculino , Terapia de Alvo Molecular , Proteína Proto-Oncogênica N-Myc , Tumores Neuroendócrinos/metabolismo , Tumores Neuroendócrinos/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Próstata/efeitos dos fármacos , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo
14.
Clin Cancer Res ; 16(14): 3618-27, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20501615

RESUMO

PURPOSE: In brain tumors, cerebral edema is a significant source of morbidity and mortality. Recent studies have shown that inhibition of vascular endothelial growth factor (VEGF) signaling induces transient vascular normalization and reduces cerebral edema, resulting in a modest survival benefit in glioblastoma patients. During anti-VEGF treatment, circulating levels of angiopoietin (Ang)-2 remained high after an initial minor reduction. It is not known, however, whether Ang-2 can modulate anti-VEGF treatment of glioblastoma. Here, we used an orthotopic glioma model to test the hypothesis that Ang-2 is an additional target for improving the efficacy of current anti-VEGF therapies in glioma patients. EXPERIMENTAL DESIGN: To recapitulate high levels of Ang-2 in glioblastoma patients during anti-VEGF treatment, Ang-2 was ectopically expressed in U87 glioma cells. Animal survival and tumor growth were assessed to determine the effects of Ang-2 and anti-VEGF receptor 2 (VEGFR2) treatment. We also monitored morphologic and functional vascular changes using multiphoton laser scanning microscopy and immunohistochemistry. RESULTS: Ectopic expression of Ang-2 had no effect on vascular permeability, tumor growth, or survival, although it resulted in higher vascular density, with dilated vessels and reduced mural cell coverage. On the other hand, when combined with anti-VEGFR2 treatment, Ang-2 destabilized vessels without affecting vessel regression and compromised the survival benefit of VEGFR2 inhibition by increasing vascular permeability. VEGFR2 inhibition normalized tumor vasculature whereas ectopic expression of Ang-2 diminished the beneficial effects of VEGFR2 blockade by inhibiting vessel normalization. CONCLUSION: Cancer treatment regimens combining anti-VEGF and anti-Ang-2 agents may be an effective strategy to improve the efficacy of current anti-VEGF therapies.


Assuntos
Angiopoietina-2/metabolismo , Glioma/terapia , Neovascularização Patológica/terapia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Angiopoietina-2/genética , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Glioma/irrigação sanguínea , Humanos , Imuno-Histoquímica , Camundongos , Transplante de Neoplasias , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
15.
PLoS One ; 4(9): e6525, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19763275

RESUMO

The role of vascular endothelial growth factor receptor 1 (VEGFR1/Flt1) in tumor metastasis remains incompletely characterized. Recent reports suggested that blocking VEGFR1 activity or the interaction with its ligands (VEGF and PlGF) has anti-tumor effects. Moreover, several studies showed that VEGFR1 mediates tumor progression to distant metastasis. All these effects may be exerted indirectly by recruitment of bone marrow-derived cells (BMDCs), such as myeloid cells. We investigated the role of VEGFR1 activity in BMDCs during the pre-metastatic phase, i.e., prior to metastatic nodule formation in mice after surgical removal of the primary tumor. Using pharmacologic blockade or genetic deletion of the tyrosine kinase domain of VEGFR1, we demonstrate that VEGFR1 activity is not required for the infiltration of de novo myeloid BMDCs in the pre-metastatic lungs in two tumor models and in two mouse models. Moreover, in line with emerging clinical observations, we show that blockade of VEGFR1 activity neither prevents nor changes the rate of spontaneous metastasis formation after primary tumor removal. Prevention of metastasis will require further identification and exploration of cellular and molecular pathways that mediate the priming of the metastatic soil.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Animais , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Linhagem Celular Tumoral , Ligantes , Neoplasias Pulmonares/patologia , Masculino , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/citologia , Metástase Neoplásica , Estrutura Terciária de Proteína , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
16.
Cell Stem Cell ; 5(5): 540-53, 2009 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-19896444

RESUMO

The PI3K-AKT-FoxO pathway is integral to lifespan regulation in lower organisms and essential for the stability of long-lived cells in mammals. Here, we report the impact of combined FoxO1, 3, and 4 deficiencies on mammalian brain physiology with a particular emphasis on the study of the neural stem/progenitor cell (NSC) pool. We show that the FoxO family plays a prominent role in NSC proliferation and renewal. FoxO-deficient mice show initial increased brain size and proliferation of neural progenitor cells during early postnatal life, followed by precocious significant decline in the NSC pool and accompanying neurogenesis in adult brains. Mechanistically, integrated transcriptomic, promoter, and functional analyses of FoxO-deficient NSC cultures identified direct gene targets with known links to the regulation of human brain size and the control of cellular proliferation, differentiation, and oxidative defense. Thus, the FoxO family coordinately regulates diverse genes and pathways to govern key aspects of NSC homeostasis in the mammalian brain.


Assuntos
Encéfalo/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Multipotentes/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Encéfalo/patologia , Proteínas de Ligação a Calmodulina , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Fatores de Transcrição Forkhead/genética , Camundongos , Camundongos Knockout , Células-Tronco Multipotentes/citologia , Proteínas do Tecido Nervoso/genética , Neurogênese/genética , Tamanho do Órgão/genética , Oxigênio/metabolismo , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/genética , Proteínas Wnt/genética
17.
Dev Biol ; 268(2): 441-7, 2004 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15063179

RESUMO

Angiogenesis, also known as new blood vessel formation, is regulated coordinately with other tissue differentiation events during limb development. Although vascular endothelial cell growth factor (VEGF) is important in the regulation of angiogenesis, chondrogenesis and osteogenesis during limb development, the role of other angiogenic factors is not well understood. Sphingosine 1-phosphate, a platelet-derived lipid mediator, regulates angiogenesis and vascular maturation via its action on the G-protein-coupled receptor S1P(1) (also known as EDG-1). In addition to vascular defects, abnormal limb development was also observed in S1p(1)(-/-) mice. Here we show that strong induction of S1P(1) expression is observed in the blood vessels and the interdigital mesenchymal cells during limb development. Deletion of S1P(1) results in aberrant chondrocyte condensation and defective digit morphogenesis. Interestingly, the vasculature in the S1p(1)(-/-) limbs was hyperplastic and morphologically altered. In addition, the hypoxia inducible factor (HIF)-1 alpha and its response gene VEGF were induced in S1p(1)(-/-) limbs. However, aberrant regulation of HIF-1 alpha and VEGF were not observed in embryonic fibroblasts derived from S1p(1)(-/-) mice, suggesting a non-cell autonomous effect of S1P(1) on VEGF expression. Indeed, similar limb defects were observed in endothelium-specific S1P(1) null mice in vivo. These data suggest that the function of S1P(1) in the developing vasculature is essential for proper limb development.


Assuntos
Extremidades/embriologia , Hipóxia/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camundongos , Receptores de Lisofosfolipídeos
18.
Genes Dev ; 18(19): 2392-403, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15371328

RESUMO

Vascular stabilization, a process by which nascent vessels are invested with mural cells, is important in angiogenesis. Here we describe the molecular basis of vascular stabilization regulated by sphingosine 1-phosphate (S1P), a platelet-derived lipid mediator. S1P1 receptor-dependent cell-surface trafficking and activation of the cell-cell adhesion molecule N-cadherin is essential for interactions between endothelial and mural cells. Endothelial cell S1P1/Gi/Rac pathway induces microtubule polymerization, resulting in trafficking of N-cadherin to polarized plasma membrane domains. S1P treatment modulated the phosphorylation of N-cadherin as well as p120-catenin and induced the formation of cadherin/catenin/actin complexes containing novel regulatory and trafficking factors. The net result of endothelial cell S1P1 receptor activation is the proper trafficking and strengthening of N-cadherin-dependent cell-cell adhesion with mural cells. Perturbation of N-cadherin expression with small interfering RNA profoundly attenuated vascular stabilization in vitro and in vivo. S1P-induced trafficking and activation of N-cadherin provides a novel mechanism for the stabilization of nascent blood vessels by mural cells and may be exploited to control angiogenesis and vascular diseases.


Assuntos
Vasos Sanguíneos/fisiologia , Caderinas/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Camundongos , Fosforilação , Transporte Proteico , Receptores de Lisofosfolipídeos , Transdução de Sinais
19.
J Biol Chem ; 277(8): 6667-75, 2002 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-11741921

RESUMO

The enzyme sphingosine kinase (SK) catalyzes the formation of sphingosine 1-phosphate (S1P), a bioactive lipid that acts extracellularly on G protein-coupled receptors of the S1P(1)/EDG-1 subfamily. Although S1P is formed in the cytosol of various cells, S1P release is not understood and is controversial because this lipid mediator is also regarded as a second messenger. In this report, we describe the existence of an extracellular S1P-generating system in vascular endothelial cells. Endothelial cells release SK constitutively and form S1P in the range of receptor stimulation. Levels of sphingosine but not ATP in the extracellular environment are rate-limiting. Treatment of endothelial cells with small interfering RNA for SK-1 transcript specifically inhibited SK export, and SK-1-transfected human embryonic kidney 293 cells exhibited enhanced release of SK-1. The export of SK-1 is constitutive and is inhibited by cytochalasin D and treatment at 4 degrees C but not by brefeldin A or nocodazole, suggesting that a nonclassical secretory pathway that requires the actin cytoskeleton dynamics is involved. Because S1P regulates angiogenesis and vascular maturation, we overexpressed SK-1 using an adenoviral vector in vivo in the Matrigel system of angiogenesis. Overexpression of SK-1 resulted in enhanced release of SK activity and induced angiogenesis and vascular maturation. These findings suggest that S1P is made in the extracellular milieu and that extracellular export of SK contributes to the action of S1P in the vascular system.


Assuntos
Endotélio Vascular/metabolismo , Regulação Enzimológica da Expressão Gênica , Lisofosfolipídeos , Neovascularização Fisiológica , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Proteínas de Xenopus , Animais , Cálcio/metabolismo , Linhagem Celular , Meios de Cultivo Condicionados , Citosol/metabolismo , Feminino , Inativação Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Proteínas Imediatamente Precoces/metabolismo , Rim , Cinética , Oócitos/fisiologia , Transporte Proteico , RNA Interferente Pequeno , RNA não Traduzido/genética , Receptores de Lisofosfolipídeos , Proteínas Repressoras/metabolismo , Transfecção , Xenopus , Homeobox 2 de Ligação a E-box com Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA