Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1861(6): 501-12, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27012521

RESUMO

High density lipoprotein (HDL) as well as annexin A1 have been reported to be associated with cardiovascular protection. However, the correlation between HDL and annexin A1 was still unknown. In this study, HDL increased endothelial annexin A1 and prevented the decrease of annexin A1 in TNF-α-activated endothelial cells in vitro and in vivo, and above effects were attenuated after knockdown of annexin A1. Annexin A1 modulation affected HDL-mediated inhibition of monocyte adhesion to TNF-α-activated endothelium (45.2±13.7% decrease for annexin A1 RNA interference; 78.7±16.3% decrease for anti-Annexin A1 antibody blocking; 11.2±6.9% increase for Ad-ANXA1 transfection). Additionally, HDL up-regulated annexin A1 through scavenger receptor class B type I, involving ERK, p38MAPK, Akt and PKC signaling pathways, and respective inhibitors of these pathways attenuated HDL-induced annexin A1 expression as well as impaired HDL-mediated inhibition of monocyte-endothelial cell adhesion. Apolipoprotein AI also increased annexin A1 and activated similar signaling pathways. Endothelial annexin A1 from apolipoprotein AI knockout mice was decreased in comparison to that from wild type mice. Finally, HDL-induced annexin A1 inhibited cell surface VCAM-1, ICAM-1 and E-selectin, and secretion of MCP-1, IL-8, VCAM-1 and E-selectin, thereby inhibiting monocyte adhesion.


Assuntos
Anexina A1/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Lipoproteínas HDL/farmacologia , Animais , Anexina A1/genética , Apolipoproteína A-I/farmacologia , Western Blotting , Antígenos CD36/genética , Antígenos CD36/metabolismo , Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Tempo , Fator de Necrose Tumoral alfa/farmacologia
2.
Nat Med ; 12(5): 557-67, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16648859

RESUMO

The mechanisms through which hematopoietic cytokines accelerate revascularization are unknown. Here, we show that the magnitude of cytokine-mediated release of SDF-1 from platelets and the recruitment of nonendothelial CXCR4+ VEGFR1+ hematopoietic progenitors, 'hemangiocytes,' constitute the major determinant of revascularization. Soluble Kit-ligand (sKitL), thrombopoietin (TPO, encoded by Thpo) and, to a lesser extent, erythropoietin (EPO) and granulocyte-macrophage colony-stimulating factor (GM-CSF) induced the release of SDF-1 from platelets, enhancing neovascularization through mobilization of CXCR4+ VEGFR1+ hemangiocytes. Although revascularization of ischemic hindlimbs was partially diminished in mice deficient in both GM-CSF and G-CSF (Csf2-/- Csf3-/-), profound impairment in neovascularization was detected in sKitL-deficient Mmp9-/- as well as thrombocytopenic Thpo-/- and TPO receptor-deficient (Mpl-/-) mice. SDF-1-mediated mobilization and incorporation of hemangiocytes into ischemic limbs were impaired in Thpo-/-, Mpl-/- and Mmp9-/- mice. Transplantation of CXCR4+ VEGFR1+ hemangiocytes into Mmp9-/- mice restored revascularization, whereas inhibition of CXCR4 abrogated cytokine- and VEGF-A-mediated mobilization of CXCR4+ VEGFR1+ cells and suppressed angiogenesis. In conclusion, hematopoietic cytokines, through graded deployment of SDF-1 from platelets, support mobilization and recruitment of CXCR4+ VEGFR1+ hemangiocytes, whereas VEGFR1 is essential for their angiogenic competency for augmenting revascularization. Delivery of SDF-1 may be effective in restoring angiogenesis in individuals with vasculopathies.


Assuntos
Quimiocinas CXC/metabolismo , Citocinas/sangue , Neovascularização Fisiológica , Receptores CXCR4/metabolismo , Regeneração , Células-Tronco/fisiologia , Animais , Plaquetas/metabolismo , Quimiocina CXCL12 , Quimiocinas CXC/genética , Humanos , Isquemia/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CXCR4/genética , Fator de Células-Tronco/metabolismo , Trombocitopenia/metabolismo , Trombopoetina/sangue , Trombopoetina/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
3.
Cancer Cell ; 4(5): 331-3, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14667498

RESUMO

Using in vivo phage display technology with murine tumorigenesis models, two reports have identified peptide motifs that selectively home to distinct molecular vascular targets in a tumor type- and stage-specific manner (Hoffman et al., 2003 and Joyce et al., 2003 [this issue of Cancer Cell]). By probing the surface-protein repertoire of these unique vascular beds, a pioneering draft of a molecular roadmap to decipher the heterogeneity of the vascular system in the context of carcinogenic progression has been plotted. Analysis of these phage peptides that differentially home to dysplastic or invasive tumor vasculature will lay the foundation for identifying unique functional tumor vascular-specific motifs that could potentially be applied to targeted therapeutic and imaging modalities.


Assuntos
Endotélio Vascular/metabolismo , Neovascularização Patológica , Biblioteca de Peptídeos , Peptídeos/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Carcinoma de Células Escamosas/metabolismo , Calicreínas/metabolismo , Camundongos , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica/genética , Pâncreas/metabolismo , Peptídeos/genética , Pele/metabolismo
4.
J Clin Invest ; 118(6): 2111-20, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18497886

RESUMO

Colon cancer stem cells are believed to originate from a rare population of putative CD133+ intestinal stem cells. Recent publications suggest that a small subset of colon cancer cells expresses CD133, and that only these CD133+ cancer cells are capable of tumor initiation. However, the precise contribution of CD133+ tumor-initiating cells in mediating colon cancer metastasis remains unknown. Therefore, to temporally and spatially track the expression of CD133 in adult mice and during tumorigenesis, we generated a knockin lacZ reporter mouse (CD133lacZ/+), in which the expression of lacZ is driven by the endogenous CD133 promoters. Using this model and immunostaining, we discovered that CD133 expression in colon is not restricted to stem cells; on the contrary, CD133 is ubiquitously expressed on differentiated colonic epithelium in both adult mice and humans. Using Il10-/-CD133lacZ mice, in which chronic inflammation in colon leads to adenocarcinomas, we demonstrated that CD133 is expressed on a full gamut of colonic tumor cells, which express epithelial cell adhesion molecule (EpCAM). Similarly, CD133 is widely expressed by human primary colon cancer epithelial cells, whereas the CD133- population is composed mostly of stromal and inflammatory cells. Conversely, CD133 expression does not identify the entire population of epithelial and tumor-initiating cells in human metastatic colon cancer. Indeed, both CD133+ and CD133- metastatic tumor subpopulations formed colonospheres in in vitro cultures and were capable of long-term tumorigenesis in a NOD/SCID serial xenotransplantation model. Moreover, metastatic CD133- cells form more aggressive tumors and express typical phenotypic markers of cancer-initiating cells, including CD44 (CD44+CD24-), whereas the CD133+ fraction is composed of CD44lowCD24+ cells. Collectively, our data suggest that CD133 expression is not restricted to intestinal stem or cancer-initiating cells, and during the metastatic transition, CD133+ tumor cells might give rise to the more aggressive CD133(- )subset, which is also capable of tumor initiation in NOD/SCID mice.


Assuntos
Antígenos CD/biossíntese , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/biossíntese , Células-Tronco/metabolismo , Antígeno AC133 , Animais , Células Epiteliais/metabolismo , Inflamação , Camundongos , Camundongos SCID , Camundongos Transgênicos , Modelos Biológicos , Modelos Genéticos , Metástase Neoplásica , Peptídeos , Fenótipo , Regiões Promotoras Genéticas , Células-Tronco/citologia
5.
Nat Med ; 10(1): 64-71, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14702636

RESUMO

The molecular pathways involved in the differentiation of hematopoietic progenitors are unknown. Here we report that chemokine-mediated interactions of megakaryocyte progenitors with sinusoidal bone marrow endothelial cells (BMECs) promote thrombopoietin (TPO)-independent platelet production. Megakaryocyte-active cytokines, including interleukin-6 (IL-6) and IL-11, did not induce platelet production in thrombocytopenic, TPO-deficient (Thpo(-/-)) or TPO receptor-deficient (Mpl(-/-)) mice. In contrast, megakaryocyte-active chemokines, including stromal-derived factor-1 (SDF-1) and fibroblast growth factor-4 (FGF-4), restored thrombopoiesis in Thpo(-/-) and Mpl(-/-) mice. FGF-4 and SDF-1 enhanced vascular cell adhesion molecule-1 (VCAM-1)- and very late antigen-4 (VLA-4)-mediated localization of CXCR4(+) megakaryocyte progenitors to the vascular niche, promoting survival, maturation and platelet release. Disruption of the vascular niche or interference with megakaryocyte motility inhibited thrombopoiesis under physiological conditions and after myelosuppression. SDF-1 and FGF-4 diminished thrombocytopenia after myelosuppression. These data suggest that TPO supports progenitor cell expansion, whereas chemokine-mediated interaction of progenitors with the bone marrow vascular niche allows the progenitors to relocate to a microenvironment that is permissive and instructive for megakaryocyte maturation and thrombopoiesis. Progenitor-active chemokines offer a new strategy to restore hematopoiesis in a clinical setting.


Assuntos
Medula Óssea/irrigação sanguínea , Quimiocinas/fisiologia , Células-Tronco Hematopoéticas/citologia , Trombopoese/fisiologia , Animais , Antígenos CD , Caderinas/fisiologia , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Megacariócitos/citologia , Camundongos , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/fisiologia , Receptores CXCR4/fisiologia , Receptores de Citocinas/genética , Receptores de Citocinas/fisiologia , Receptores de Trombopoetina , Trombopoetina/genética , Trombopoetina/fisiologia
6.
Nature ; 438(7069): 820-7, 2005 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-16341007

RESUMO

The cellular and molecular mechanisms by which a tumour cell undergoes metastasis to a predetermined location are largely unknown. Here we demonstrate that bone marrow-derived haematopoietic progenitor cells that express vascular endothelial growth factor receptor 1 (VEGFR1; also known as Flt1) home to tumour-specific pre-metastatic sites and form cellular clusters before the arrival of tumour cells. Preventing VEGFR1 function using antibodies or by the removal of VEGFR1(+) cells from the bone marrow of wild-type mice abrogates the formation of these pre-metastatic clusters and prevents tumour metastasis, whereas reconstitution with selected Id3 (inhibitor of differentiation 3)-competent VEGFR1+ cells establishes cluster formation and tumour metastasis in Id3 knockout mice. We also show that VEGFR1+ cells express VLA-4 (also known as integrin alpha4beta1), and that tumour-specific growth factors upregulate fibronectin--a VLA-4 ligand--in resident fibroblasts, providing a permissive niche for incoming tumour cells. Conditioned media obtained from distinct tumour types with unique patterns of metastatic spread redirected fibronectin expression and cluster formation, thereby transforming the metastatic profile. These findings demonstrate a requirement for VEGFR1+ haematopoietic progenitors in the regulation of metastasis, and suggest that expression patterns of fibronectin and VEGFR1+VLA-4+ clusters dictate organ-specific tumour spread.


Assuntos
Movimento Celular , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Metástase Neoplásica/patologia , Metástase Neoplásica/fisiopatologia , Neoplasias/metabolismo , Neoplasias/patologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Adesão Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Meios de Cultivo Condicionados/farmacologia , Fibronectinas/metabolismo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Proteínas Inibidoras de Diferenciação/metabolismo , Integrina alfa4beta1/metabolismo , Metaloproteinase 9 da Matriz , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos , Especificidade por Substrato , Regulação para Cima , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
7.
J Clin Invest ; 116(12): 3277-91, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17143334

RESUMO

Thrombopoietic cells may differentially promote or inhibit tissue vascularization by releasing both pro- and antiangiogenic factors. However, the molecular determinants controlling the angiogenic phenotype of thrombopoietic cells remain unknown. Here, we show that expression and release of thrombospondins (TSPs) by megakaryocytes and platelets function as a major antiangiogenic switch. TSPs inhibited thrombopoiesis, diminished bone marrow microvascular reconstruction following myelosuppression, and limited the extent of revascularization in a model of hind limb ischemia. We demonstrate that thrombopoietic recovery following myelosuppression was significantly enhanced in mice deficient in both TSP1 and TSP2 (TSP-DKO mice) in comparison with WT mice. Megakaryocyte and platelet levels in TSP-DKO mice were rapidly restored, thereby accelerating revascularization of myelosuppressed bone marrow and ischemic hind limbs. In addition, thrombopoietic cells derived from TSP-DKO mice were more effective in supporting neoangiogenesis in Matrigel plugs. The proangiogenic activity of TSP-DKO thrombopoietic cells was mediated through activation of MMP-9 and enhanced release of stromal cell-derived factor 1. Thus, TSP-deficient thrombopoietic cells function as proangiogenic agents, accelerating hemangiogenesis within the marrow and revascularization of ischemic hind limbs. As such, interference with the release of cellular stores of TSPs may be clinically effective in augmenting neoangiogenesis.


Assuntos
Medula Óssea/irrigação sanguínea , Células-Tronco Hematopoéticas/metabolismo , Neovascularização Patológica , Neovascularização Fisiológica , Trombospondinas/metabolismo , Animais , Plaquetas/citologia , Medula Óssea/metabolismo , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Células-Tronco Hematopoéticas/citologia , Membro Posterior/irrigação sanguínea , Membro Posterior/metabolismo , Membro Posterior/patologia , Isquemia/metabolismo , Isquemia/fisiopatologia , Megacariócitos/citologia , Megacariócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estilbenos/metabolismo , Trombopoese/fisiologia , Trombopoetina/genética , Trombopoetina/metabolismo , Trombospondina 1/genética , Trombospondina 1/metabolismo , Trombospondinas/genética
8.
Arterioscler Thromb Vasc Biol ; 28(2): 217-22, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18096826

RESUMO

Bone marrow-derived cells contribute to physiological and pathological vascular remodeling throughout ontogenesis and adult life. During tissue regeneration and tumor growth, the release of cytokines and chemokines mediates the recruitment of hematopoietic and endothelial progenitor cells that contribute to the assembly of neovessels. Current evidence implies that platelets contribute structurally and instructively to vascular remodeling. Platelets adhere almost immediately to exposed or activated endothelium, and they are major storage and delivery vehicles for pro- and antiangiogenic growth factors including VEGF-A and thrombospondin (TSP), and cytokines and chemokines, such as stromal-derived factor 1 (SDF-1). By site-specific deployment of these factors, platelets orchestrate the local angiogenic stimulus within a tissue and direct the recruitment and differentiation of circulating bone marrow-derived cells. These insights have profound clinical implications; inhibition of platelet-deployed growth factors or their receptors may be an effective strategy to block tumor growth, whereas activation of these pathways may be used to accelerate revascularization and tissue regeneration.


Assuntos
Plaquetas/fisiologia , Diferenciação Celular/fisiologia , Neovascularização Fisiológica/fisiologia , Células da Medula Óssea/fisiologia , Quimiocina CXCL12/fisiologia , Células Endoteliais/fisiologia , Humanos , Trombospondinas/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
9.
J Clin Invest ; 115(3): 653-63, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15765148

RESUMO

The neurotrophin brain-derived neurotrophic factor (BDNF) is required for the maintenance of cardiac vessel wall stability during embryonic development through direct angiogenic actions on endothelial cells expressing the tropomysin receptor kinase B (TrkB). However, the role of BDNF and a related neurotrophin ligand, neurotrophin-4 (NT-4), in the regulation of revascularization of the adult tissues is unknown. To study the potential angiogenic capacity of BDNF in mediating the neovascularization of ischemic and non-ischemic adult mouse tissues, we utilized a hindlimb ischemia and a subcutaneous Matrigel model. Recruitment of endothelial cells and promotion of channel formation within the Matrigel plug by BDNF and NT-4 was comparable to that induced by VEGF-A. The introduction of BDNF into non-ischemic ears or ischemic limbs induced neoangiogenesis, with a 2-fold increase in the capillary density. Remarkably, treatment with BDNF progressively increased blood flow in the ischemic limb over 21 days, similar to treatment with VEGF-A. The mechanism by which BDNF enhances capillary formation is mediated in part through local activation of the TrkB receptor and also by recruitment of Sca-1+CD11b+ pro-angiogenic hematopoietic cells. BDNF induces a potent direct chemokinetic action on subsets of marrow-derived Sca-1+ hematopoietic cells co-expressing TrkB. These studies suggest that local regional delivery of BDNF may provide a novel mechanism for inducing neoangiogenesis through both direct actions on local TrkB-expressing endothelial cells in skeletal muscle and recruitment of specific subsets of TrkB+ bone marrow-derived hematopoietic cells to provide peri-endothelial support for the newly formed vessels.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Endoteliais/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Neovascularização Fisiológica , Fatores de Crescimento Neural/metabolismo , Receptor trkB/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Biomarcadores/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Linhagem Celular , Quimiotaxia , Células Endoteliais/citologia , Feminino , Vetores Genéticos , Células-Tronco Hematopoéticas/citologia , Humanos , Isquemia/metabolismo , Isquemia/patologia , Masculino , Camundongos , Camundongos Endogâmicos , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
10.
Circ Res ; 96(9): 950-7, 2005 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-15831817

RESUMO

Connexins (Cxs) provide a means for intercellular communication and play important roles in the pathophysiology of vascular cardiac diseases. Infection of endothelial cells (ECs) with first-generation E1/E3-deleted E4+ adenovirus (AdE4+) selectively modulates the survival and angiogenic potential of ECs by as of yet unrecognized mechanisms. We show here that AdE4+ vectors potentiate Cx expression in ECs in vitro and in mouse heart tissue. Infection of ECs with AdE4+, but not AdE4-, resulted in a time- and dose-dependent induction of junctional Cx40 expression and suppression of Cx43 protein and mRNA expression. Treatment of ECs with PKA inhibitor H89 or PI3K inhibitor LY294002 prevented the AdE4+-mediated regulation of Cx40 and Cx43 that was associated with diminished AdE4+-mediated survival of ECs. Moreover, both PKA activity and cAMP-response element (CRE)-binding activity were enhanced by treatment of ECs with AdE4+. However, there is no causal evidence of a cross-talk between the 2 modulatory pathways, PKA and PI3K. Remarkably, Cx40 immunostaining was markedly increased and Cx43 was decreased in the heart tissue of mice treated with intra-tracheal AdE4+. Taken together, these results suggest that AdE4+ may play an important role in the regulation of Cx expression in ECs, and that these effects are mediated by both the PKA/CREB and PI3K signaling pathways.


Assuntos
Adenoviridae/genética , Proteínas E4 de Adenovirus/genética , Conexina 43/metabolismo , Conexinas/biossíntese , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Endotélio Vascular/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Conexina 43/análise , Conexina 43/genética , Conexinas/análise , Conexinas/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Células Endoteliais/química , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/enzimologia , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Vetores Genéticos , Humanos , Camundongos , Miocárdio/metabolismo , Toxina Pertussis/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , RNA Mensageiro/metabolismo , Transdução de Sinais , Proteína alfa-5 de Junções Comunicantes
11.
Cancer Res ; 65(8): 3185-92, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15833849

RESUMO

Induction of neoangiogenesis plays an important role in the pathogenesis of multiple myeloma. However, the mechanism by which expression of vascular endothelial growth factor (VEGF)-A and its receptors modulate the interaction of multiple myeloma cells with stromal cells is not known. Here, we describe a novel in vitro coculture system using fetal bone stromal cells as a feeder layer, which facilitates the survival and growth of human primary multiple myeloma cells. We show that stromal-dependent paracrine VEGF-A signaling promotes proliferation of human primary multiple myeloma cells. Primary multiple myeloma cells only expressed functional VEGF receptor (VEGFR)-1, but not VEGFR-2 or VEGFR-3. VEGFR-1 expression was detected in the cytoplasm and the nuclei of proliferating multiple myeloma cells. Inhibition of VEGFR-1 abrogated multiple myeloma cell proliferation and motility, suggesting that the functional interaction of VEGF-A with its cognate receptor is essential for the growth of primary multiple myeloma cells. Collectively, our results suggest that stromal-dependent paracrine and intracrine VEGF-A/VEGFR-1 signaling contributes to human primary multiple myeloma cell growth and therefore, VEGFR-1 blockade is a potential therapeutic strategy for the treatment of multiple myeloma.


Assuntos
Movimento Celular/fisiologia , Mieloma Múltiplo/patologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Processos de Crescimento Celular/fisiologia , Núcleo Celular/metabolismo , Sobrevivência Celular/fisiologia , Técnicas de Cocultura , Citoplasma/metabolismo , Humanos , Mieloma Múltiplo/metabolismo , Células Estromais/citologia , Células Estromais/metabolismo , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
12.
J Neurol Sci ; 383: 47-51, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29246620

RESUMO

The relationship between blood pressure(BP) and clinical outcome in patients with acute stroke is still controversial. The present study aimed to elucidate the impact of admission blood pressure on mortality in patients with acute stroke of different subtypes. Data were from ChinaQUEST (QUality Evaluation of Stroke Care and Treatment), a multicenter, prospective hospital registry study in 37 cities across China. A total of 6427 patients were admitted within 24h of onset and after following up for 12months, 5501 were included in the final analysis. Multivariate Cox regression model were used in data analysis. A "U-curve shaped" relationship was observed between admission systolic or diastolic BP and mortality at 12months in the overall study population. Compared to first quartile, the Hazard ratio (HR) for the systolic BP of top quartile was 1.444 (95%CI 1.854-1.636), while the HR was 0.692 (95%CI 0.802-0.930) for the second quartile. Similar associations were observed when we applied admission diastolic BP. In subgroup analysis, the U-shaped effect was remained only in patients with intracranial hemorrhage (ICH). The HR for the systolic BP of top quartile was 2.274 (95%CI 1.878-2.755), while the HR was 0.751 (95%CI 0.571-0.986) for the second quartile. Moreover, admission diastolic BP of top quartile was significantly associated with elevated risk of death for patients with ischemic stroke caused by small vessel diseases (LACI)(HR 1.470; CI 1.040-2.078). In addition, we found a heterogeneity of the admission BP distribution among different subtypes, which may explain the "U-curve" effect.


Assuntos
Pressão Sanguínea , Acidente Vascular Cerebral/mortalidade , Acidente Vascular Cerebral/fisiopatologia , Idoso , Determinação da Pressão Arterial , Isquemia Encefálica/diagnóstico , Isquemia Encefálica/mortalidade , Isquemia Encefálica/fisiopatologia , Isquemia Encefálica/terapia , China , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Admissão do Paciente , Prognóstico , Modelos de Riscos Proporcionais , Estudos Prospectivos , Sistema de Registros , Fatores de Risco , Acidente Vascular Cerebral/diagnóstico , Acidente Vascular Cerebral/terapia
13.
J Mol Diagn ; 8(2): 231-9, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16645210

RESUMO

Monitoring breakpoint cluster region-Abelson kinase (BCR-ABL) levels in patients treated for chronic myelogenous leukemia (CML) has become an integral part of patient management. Real-time reverse transcriptase-polymerase chain reaction is the method of choice for this purpose because of its high analytical sensitivity and reproducibility. Given the variation of RNA quality and quantity in clinical specimens, accurate quantitative assessment of BCR-ABL depends on normalization of the BCR-ABL signal to an appropriate internal reference. However, the controls used by different laboratories vary, and there is no clear consensus on an ideal reference due to limited investigations. In this study, we compared nine commonly used control genes for three criteria: mRNA abundance, levels in CML and non-CML cells, and their degradation kinetics in comparison with BCR-ABL. We found that beta-glucuronidase (GUSB) is the most suitable among the nine genes tested. Although ABL is most widely used, our data suggest that the amount of ABL is different in CML and non-CML cells. Moreover, ABL levels are regulated by cellular stress. These findings have a direct impact on current clinical laboratory practice and patient care because the use of a proper control gene affects the reported levels of BCR-ABL transcripts used for patient management decisions.


Assuntos
Proteínas de Fusão bcr-abl/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Adulto , Idoso , Sequência de Bases , Linhagem Celular Tumoral , Feminino , Humanos , Cinética , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Fatores de Tempo , Transcrição Gênica/genética
14.
Cancer Lett ; 220(2): 211-20, 2005 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-15766596

RESUMO

The level of alpha-fetoprotein (AFP) gene expression during liver regeneration in mice is regulated by the Afr2 gene. C3H/HeJ mice express 10-fold higher levels of AFP than C57BL/6J mice. We show that AFP gene expression is not Afr2 regulated after intoxication with the carcinogen diethylnitrosamine (DEN). Peak levels of AFP gene expression of the 2 strains were identical, although reached at different times following treatment. Analysis of the expression of AFP derived transgenes not subject to Afr2 regulation and genetic analyses showed that the difference in timing of peak AFP gene expression after DEN intoxication was independent of Afr2 regulation.


Assuntos
Alquilantes/farmacologia , Dietilnitrosamina/farmacologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , alfa-Fetoproteínas/biossíntese , alfa-Fetoproteínas/genética , Animais , Regeneração Hepática/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transgenes
15.
Neurosurgery ; 64(5): 819-26; discussion 826-7, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19404145

RESUMO

OBJECTIVE: The neovascularization of malignant brain tumors is a poorly understood phenomenon. Radiographic and histological evidence of increased vascularity correlate with clinical grade of gliomas. However, a quantitative noninvasive assay to assess glioma vascularity and associated clinical aggressiveness has not been developed. Circulating endothelial progenitor cells are unique vascular precursors recruited from the bone marrow through the circulation to form new tumor blood vessels. These cells were measured in patients undergoing surgery for glioblastoma multiforme (GBM). We hypothesized that this might reflect the extent of tumor vascularity, predict prognosis, or be useful as an assay to assess response to antiangiogenesis therapies. In addition, we report on a novel in vitro assay to assess the proangiogenic activity within the plasma samples obtained from glioma patients. METHODS: Fifty-six patients with various-grade gliomas had peripheral venous blood collected at the time of surgery and at subsequent visits during the follow-up period. The blood was separated into plasma and cellular fractions. The plasma was utilized in a human umbilical vein endothelial cell-based angiogenic assay. The cellular fraction containing endothelial progenitor cells was isolated, and specific cellular phenotypes were immunologically separated and counted using flow cytometry. Pathological samples were reviewed at the time of initial resection, and each patient's clinical course was monitored until the time of manuscript submission. RESULTS: Plasma derived from peripheral blood of patients with GBM scored significantly higher on the functional angiogenic scale compared with plasma derived from patients with low-grade gliomas and from controls. In addition, all patients with GBM had measurable numbers of bone marrow-derived endothelial precursor cells coexpressing CD133 and vascular endothelial growth factor receptor 2 in their peripheral circulation at the time of tumor resection. These cells range from less than 0.1% to 1.6% of the entire circulating mononuclear white blood cell population, or approximately 200,000 cells in some patients. A statistically significant relationship was observed between the percentage of endothelial progenitor cells in the peripheral blood at the time of initial GBM resection and survival. CONCLUSION: These studies suggest that plasma and circulating CD133+ vascular endothelial growth factor receptor 2+ proangiogenic cells are present in the peripheral blood of patients with glioma and can be used as a surrogate biomarker to measure tumor angiogenicity. These cells can be measured at the time of diagnosis and monitored in the postoperative period. These assays can be used to predict tumor aggressiveness. Also promising is their potential to identify patients with increased angiogenic activity who might respond maximally to antiangiogenesis therapies or to assess tumor response in patients using those therapies as the use of these adjuvant molecular modalities becomes more prevalent in neuro-oncology.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/sangue , Glioblastoma/sangue , Neovascularização Patológica/sangue , Antígeno AC133 , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos CD/metabolismo , Bioensaio/métodos , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Citometria de Fluxo , Seguimentos , Glioblastoma/mortalidade , Glioblastoma/patologia , Glicoproteínas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Peptídeos/metabolismo , Células-Tronco/metabolismo , Células-Tronco/patologia , Veias Umbilicais/patologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
16.
J Biol Chem ; 279(12): 11760-6, 2004 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-14660586

RESUMO

The early 4 region (E4) of the adenoviral vectors (AdE4(+)) prolongs human endothelial cell (EC) survival and alters the angiogenic response, although the mechanisms for the EC-specific, AdE4(+)-mediated effects remain unknown. We hypothesized that AdE4(+) modulates EC survival through activation of the vascular endothelial (VE)-cadherin/Akt pathway. Here, we showed that AdE4(+), but not AdE4(-) vectors, selectively stimulated phosphorylation of both Akt at Ser(473) and Src kinase in ECs. The phosphatidylinositol 3-kinase (PI3K) inhibitors LY294002 and wortmannin abrogated AdE4(+) induction of both phospho-Akt expression and prolonged EC survival. Regulation of phospho-Akt was found to be under the control of various factors, namely VE-cadherin activation, Src kinase, tyrosine kinase, extracellular signal-regulated kinase (ERK), and c-Jun N-terminal kinase (JNK). Downstream targets of Akt signaling resulted in glycogen synthase kinase-3alpha/beta phosphorylation, beta-catenin up-regulation, and caspase-3 suppression, all of which led to AdE4(+)-mediated EC survival. Furthermore, infection with AdE4(+) vectors increased the angiogenic potential of ECs by promoting EC migration and capillary tube formation in Matrigel plugs. This selective AdE4(+)-mediated enhanced motility of ECs was also blocked by PI3K inhibitors. Taken together, these results suggest that activation of the VE-cadherin/Akt pathway is critical for AdE4(+)-mediated survival of ECs and angiogenic potential.


Assuntos
Proteínas E4 de Adenovirus/genética , Caderinas/metabolismo , Sobrevivência Celular/genética , Endotélio Vascular/citologia , Neovascularização Fisiológica/genética , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Androstadienos/farmacologia , Apoptose/genética , Células Cultivadas , Cromonas/farmacologia , Endotélio Vascular/metabolismo , Inibidores Enzimáticos/farmacologia , Vetores Genéticos , Humanos , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt , Wortmanina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA