Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Br J Cancer ; 113(11): 1541-7, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26554652

RESUMO

BACKGROUND: PI3K-AKT-mTOR inhibitors (PAMi) are promising anticancer treatments. Hyperglycaemia is a mechanism-based toxicity of these agents and is becoming increasingly important with their use in larger numbers of patients. METHODS: Retrospective case-control study comparing incidence and severity of hyperglycaemia (all grades) between a case group of 387 patients treated on 18 phase I clinical trials with PAMi (78 patients with PI3Ki, 138 with mTORi, 144 with AKTi and 27 with PI3K/mTORi) and a control group of 109 patients treated on 10 phase I clinical trials with agents not directly targeting the PAM pathway. Diabetic patients were excluded in both groups. RESULTS: The incidence of hyperglycaemia was not significantly different between cases and controls (86.6% vs 80.7%, respectively, P=0.129). However, high grade (grade 3-4) hyperglycaemia was more frequent in the PAMi group than in controls (6.7% vs 0%, respectively, P=0.005). The incidence of grade 3-4 hyperglycaemia was greater with AKT and multikinase inhibitors compared with other PAMi (P<0.001). All patients with high-grade hyperglycaemia received antihyperglycemic treatment and none developed severe metabolic complications (diabetic ketoacidosis or hyperosmolar hyperglycemic nonketotic state). High-grade hyperglycaemia was the cause of permanent PAMi discontinuation in nine patients. CONCLUSIONS: PI3K-AKT-mTOR inhibitors are associated with small (6.7%) but statistically significant increased risk of high-grade hyperglycaemia compared with non-PAM targeting agents. However, PAMi-induced hyperglycaemia was not found to be associated with severe metabolic complications in this non-diabetic population of patients with advanced cancers.


Assuntos
Antineoplásicos/efeitos adversos , Hiperglicemia/epidemiologia , Hiperglicemia/etiologia , Neoplasias/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/efeitos adversos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Glicemia/efeitos dos fármacos , Estudos de Casos e Controles , Ensaios Clínicos Fase I como Assunto , Feminino , Humanos , Hiperglicemia/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Masculino , Pessoa de Meia-Idade , Neoplasias/patologia , Estudos Retrospectivos , Fatores de Risco , Índice de Gravidade de Doença , Transdução de Sinais/efeitos dos fármacos , Adulto Jovem
2.
Andrology ; 2(4): 607-14, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24902969

RESUMO

The optimal markers for human spermatogonial stem cells (SSCs) are not known. Among the genes recently linked to SSCs in mice and other animals are the basic helix-loop-helix transcription factor ID4 and the orphan G-protein-coupled receptor GPR125. While ID4 and GPR125 are considered putative markers for SSCs, they have not been evaluated for coexpression in human tissue. Furthermore, neither the size nor the character of the human spermatogonial populations that express ID4 and GPR125, respectively, are known. A major barrier to addressing these questions is the availability of healthy adult testis tissue from donors with no known reproductive health problems. To overcome this obstacle, we have employed healthy testicular tissue from a novel set of organ donors (n = 16; aged 17-68 years) who were undergoing post-mortem clinical organ procurement. Using immunolabelling, we found that ID4 and GPR125 are expressed on partially overlapping spermatogonial populations and are more broadly expressed in the normal adult human testis. In addition, we found that expression of ID4 remained stable during ageing. These findings suggest that ID4 and GPR125 could be efficacious for identifying previously unrecognized human spermatogonial subpopulations in conjunction with other putative human stem cell markers, both in younger and older donors.


Assuntos
Biomarcadores/metabolismo , Sequências Hélice-Alça-Hélice/fisiologia , Proteínas Inibidoras de Diferenciação/biossíntese , Receptores Acoplados a Proteínas G/biossíntese , Espermatogônias/metabolismo , Células-Tronco/metabolismo , Adolescente , Adulto , Idoso , Cadáver , Humanos , Masculino , Pessoa de Meia-Idade , Doadores de Tecidos
3.
Leukemia ; 28(10): 1969-77, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24721792

RESUMO

Recent technological advances led to an appreciation of the genetic complexity of human acute myeloid leukemia (AML), but underlying progenitor cells remain poorly understood because their rarity precludes direct study. We developed a co-culture method integrating hypoxia, aryl hydrocarbon receptor inhibition and micro-environmental support via human endothelial cells to isolate these cells. X-chromosome inactivation studies of the least mature precursors derived following prolonged culture of CD34(+)/CD33(-) cells revealed polyclonal growth in highly curable AMLs, suggesting that mutations necessary for clonal expansion were acquired in more mature progenitors. Consistently, in core-binding factor (CBF) leukemias with known complementing mutations, immature precursors derived following prolonged culture of CD34(+)/CD33(-) cells harbored neither mutation or the CBF mutation alone, whereas more mature precursors often carried both mutations. These results were in contrast to those with leukemias with poor prognosis that showed clonal dominance in the least mature precursors. These data indicate heterogeneity among progenitors in human AML that may have prognostic and therapeutic implications.


Assuntos
Células-Tronco Hematopoéticas/citologia , Leucemia Mieloide Aguda/genética , Mutação , Antígenos CD34/metabolismo , Hipóxia Celular , Separação Celular , Técnicas de Cocultura , Fatores de Ligação ao Core/metabolismo , Citometria de Fluxo , Sistema Hematopoético , Humanos , Leucemia Mieloide Aguda/metabolismo , Prognóstico , Receptores de Hidrocarboneto Arílico/metabolismo , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo
4.
Ann Oncol ; 24(6): 1491-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23406736

RESUMO

BACKGROUND: Bone marrow-derived endothelial progenitor cells (EPCs) are critical for metastatic progression. This study explores the effect of tetrathiomolybdate (TM), an anti-angiogenic copper chelator, on EPCs in patients at high risk for breast cancer recurrence. PATIENTS AND METHODS: This phase 2 study enrolled breast cancer patients with stage 3 and stage 4 without evidence of disease (NED), and stage 2 if triple-negative. TM 100 mg orally was administered to maintain ceruloplasmin <17 mg/dl for 2 years or until relapse. The primary end point was change in EPCs. RESULTS: Forty patients (28 stage 2/3, 12 stage 4 NED) were enrolled. Seventy-five percent patients achieved the copper depletion target by 1 month. Ninety-one percent of triple-negative patients copper-depleted compared with 41% luminal subtypes. In copper-depleted patients only, there was a significant reduction in EPCs/ml by 27 (P = 0.04). Six patients relapsed while on study, of which only one patient had EPCs maintained below baseline. The 10-month relapse-free survival was 85.0% (95% CI 74.6%-96.8%). Only grade 3/4 toxicity was hematologic: neutropenia (3.1% of cycles), febrile neutropenia (0.2%), and anemia (0.2%). CONCLUSIONS: TM is safe and appears to maintain EPCs below baseline in copper-depleted patients. TM may promote tumor dormancy and ultimately prevent relapse.


Assuntos
Neoplasias da Mama/sangue , Cobre/sangue , Células Endoteliais/metabolismo , Molibdênio/uso terapêutico , Recidiva Local de Neoplasia/prevenção & controle , Células-Tronco/metabolismo , Adulto , Idoso , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/tratamento farmacológico , Quelantes/uso terapêutico , Células Endoteliais/efeitos dos fármacos , Feminino , Humanos , Pessoa de Meia-Idade , Molibdênio/farmacologia , Recidiva Local de Neoplasia/sangue , Fatores de Risco , Células-Tronco/efeitos dos fármacos
5.
Iran Red Crescent Med J ; 14(4): 235-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22754687

RESUMO

BACKGROUND: Public hospitals can privatize management activities by contracting with a private organization or person to perform the work. Management contract is a method which uses private sector for major government projects like hospitals. This study evaluates contract management in health care institutes of developing countries. METHODS: Information has been collected by reviewing the management contract condition of selected countries. Different forms of public private partnership for private participation in hospitals were surveyed. RESULTS: The effects of management contract is expanding market opportunities to include public sector clients, capturing a market to be protected from competitors and providing a reliable and timely source of revenue. CONCLUSION: Contracting with non-governmental entities will provide better results than government provision of the same services. Contracting initiatives must be regulated and monitored at the highest level of government by experienced and astute policy makers, economists and operational personnel.

6.
Leukemia ; 26(8): 1771-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22343591

RESUMO

Advances in the treatment of acute leukemia have resulted in significantly improved remission rates, although disease relapse poses a significant risk. By utilizing sensitive, non-invasive imaging guidance, detection of early leukemic infiltration and the extent of residual tumor burden after targeted therapy can be expedited, leading to more efficient treatment planning. We demonstrated marked survival benefit and therapeutic efficacy of a new-generation vascular disrupting agent, combretastatin-A1-diphosphate (OXi4503), using reporter gene-imaging technologies and mice systemically administered luc+ and GFP+ human leukemic cells (LCs). Before treatment, homing of double-transduced cells was serially monitored and whole-body cellular distributions were mapped using bioluminescence imaging (BLI). Imaging findings strongly correlated with quantitative GFP expression levels in solid organs/tissues, suggesting that the measured BLI signal provides a highly sensitive and reliable biomarker of tumor tissue burden in systemic leukemic models. Such optical technologies can thereby serve as robust non-invasive imaging tools for preclinical drug discovery and for rapidly screening promising therapeutic agents to establish potency, treatment efficacy and survival advantage. We further show that GFP+ HL-60 cells reside in close proximity to VE-cadherin- and CD31-expressing endothelial cells, suggesting that the perivascular niche may have a critical role in the maintenance and survival of LCs.


Assuntos
Antineoplásicos/administração & dosagem , Rastreamento de Células/métodos , Difosfatos/administração & dosagem , Leucemia/tratamento farmacológico , Estilbenos/administração & dosagem , Animais , Antineoplásicos/farmacologia , Movimento Celular , Difosfatos/farmacologia , Genes Reporter , Células HL-60 , Humanos , Leucemia/mortalidade , Leucemia/patologia , Medições Luminescentes , Camundongos , Estilbenos/farmacologia , Transdução Genética , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Ann Oncol ; 20(3): 413-24, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19088170

RESUMO

Angiogenesis, the growth of new blood vessels, requires dynamic expansion, assembly and stabilization of vascular endothelial cells in response to proangiogenic stimuli. Antiangiogenic strategies have become an important therapeutic modality for solid tumors. While many aspects of postnatal pathological angiogenesis have been extensively studied in the context of nonhematopoietic neoplasms, the precise role of these processes in lymphoma pathogenesis is under active investigation. Lymphoma growth and progression is potentiated by at least two distinct angiogenic mechanisms: autocrine stimulation of tumor cells via expression of vascular endothelial growth factor (VEGF) and VEGF receptors by lymphoma cells, as well as paracrine influences of proangiogenic tumor microenvironment on both local neovascular transformation and recruitment of circulating bone marrow-derived progenitors. Lymphoma-associated infiltrating host cells including hematopoietic monocytes, T cells and mesenchymal pericytes have increasingly been associated with the pathogenesis and prognosis of lymphoma, in part providing perivascular guidance and support to neoangiogenesis. Collectively, these distinct angiogenic mechanisms appear to be important therapeutic targets in selected non-Hodgkin's lymphoma (NHL) subtypes. Understanding these pathways has led to the introduction of antiangiogenic treatment strategies into the clinic where they are currently under assessment in several ongoing studies of NHL patients.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Linfoma não Hodgkin/tratamento farmacológico , Neovascularização Patológica , Humanos , Linfoma não Hodgkin/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Histol Histopathol ; 22(9): 971-6, 2007 09.
Artigo em Inglês | MEDLINE | ID: mdl-17523074

RESUMO

The enzyme beta-galactosidase, encoded by the bacterial gene lac-Z, is commonly used as a histochemical reporter to track transplanted cells in vivo or to analyze temporospatial gene expression patterns by coupling expression of specific target genes to beta-galactosidase activity. Previously, endogenous beta-galactosidase activity has been recognized as a confounding factor in the study of different soft tissues, but there is no description of the typical background on bone marrow sections when using the chromogenic substrate 5-Bromo-4-chloro-3-indolyl beta-D-Galactoside (X-Gal). In this report, we show that osteoclasts in bone marrow sections specifically and robustly stain blue with X-Gal. This leads to a typical background when bone marrow is examined that is present from the first day post partum throughout the adult life of experimental mice and can be confused with transgenic, bacterial beta-galactosidase expressing hematopoietic or stromal cells. Experimental variations in the X-Gal staining procedure, such as pH and time of exposure to substrate, were not sufficient to avoid this background. Therefore, these data demonstrate the need for strenuous controls when evaluating beta-galactosidase positive bone marrow cells. Verifiable bacterial beta-galactosidase positive bone marrow cells should be further identified using immunohistological or other approaches. Specifically, beta-galactosidase positive hematopoietic or stromal cells should be proven specifically not to be osteoclasts by co-staining or staining adjacent sections for specific markers of hematopoietic and stromal cells.


Assuntos
Células da Medula Óssea/citologia , Histocitoquímica/métodos , Osteoclastos/metabolismo , beta-Galactosidase/análise , beta-Galactosidase/metabolismo , Fosfatase Ácida/metabolismo , Animais , Biomarcadores/metabolismo , Osso e Ossos/metabolismo , Fêmur/metabolismo , Genes Reporter , Sistema Hematopoético/metabolismo , Concentração de Íons de Hidrogênio , Imuno-Histoquímica , Isoenzimas/metabolismo , Óperon Lac , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Radiografia Dentária Digital , Coloração e Rotulagem , Células Estromais/metabolismo , Especificidade por Substrato , Fosfatase Ácida Resistente a Tartarato , Fatores de Tempo
9.
Ann N Y Acad Sci ; 1106: 175-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17395736

RESUMO

Megakaryocytes and platelets have been known to secrete angiogenic growth factors for a long time. However, there is little in vivo data on the regulation of angiogenesis by thrombopoietic cells. Both megakaryocytes and platelets are known to carry and release a multitude of both pro- and antiangiogenic mediators. Thus, it remained unknown how the "angiogenic phenotype" of thrombopoietic cells would be determined. Our group established that platelets contribute to angiogenesis as carriers of SDF-1, which is released by platelets in response to stimulation with hematopoietic cytokines. Indeed, even the action of VEGF-A seems to be mediated in part by the release of SDF-1 from stimulated platelets, thereby attracting proangiogenic hematopoietic cells. Moreover, the analysis of murine plasma and serum showed that similar to VEGF-A, SDF-1 is almost exclusively derived from platelets, and only trace amounts are detectable in platelet poor plasma. Because tumor patients' platelets have been shown to contain lower amounts of thrombospondin (Tsp), we generated Tsp-1 and Tsp-2 double knockout mice by crossing the single knockout lines. Interestingly, megakaryocytes and platelets derived from these mice confer a proangiogenic phenotype both in the bone marrow and in reperfusion of ischemic hindlimbs, thereby verifying the hypothesis of pro- and antiangiogenic platelet constituents "in balance."


Assuntos
Plaquetas/citologia , Células da Medula Óssea/citologia , Megacariócitos/citologia , Animais , Plaquetas/metabolismo , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Endotélio Vascular/citologia , Humanos , Megacariócitos/metabolismo , Camundongos , Camundongos Knockout , Modelos Biológicos , Neovascularização Patológica , Fenótipo
11.
Leukemia ; 20(6): 979-86, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16598308

RESUMO

Fibroblast growth factors (FGFs) are important regulators of hematopoiesis and have been implicated in the tumorigenesis of solid tumors. Recent evidence suggests that FGF signaling through FGF receptors (FGFRs) may play a role in the proliferation of subsets of acute myeloid leukemias (AMLs). However, the precise mechanism and specific FGF receptors that support leukemic cell growth are not known. We show that FGF-2, through activation of FGFR1beta signaling, promotes survival, proliferation and migration of AML cells. Stimulation of FGFR1beta results in phosphoinositide 3-kinase (PI3-K)/Akt activation and inhibits chemotherapy-induced apoptosis of leukemic cells. Neutralizing FGFR1-specific antibody abrogates the physiologic and chemoprotective effects of FGF-2/FGFR1beta signaling and inhibits tumor growth in mice xenotransplanted with human AML. These data suggest that activation of FGF-2/FGFR1beta supports progression and chemoresistance in subsets of AML. Therefore, FGFR1 targeting may be of therapeutic benefit in subsets of AML.


Assuntos
Movimento Celular , Resistencia a Medicamentos Antineoplásicos , Leucemia Mieloide/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Doença Aguda , Idoso de 80 Anos ou mais , Proteína Relacionada com Agouti , Animais , Anticorpos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/genética , Masculino , Camundongos , Camundongos SCID , Fosforilação , Subunidades Proteicas/efeitos dos fármacos , Subunidades Proteicas/metabolismo , RNA Mensageiro/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/efeitos dos fármacos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Leuk Res ; 28(9): 979-85, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15234576

RESUMO

This meeting was convened by Richard T. Silver, M.D. and co-chaired by Jerry L. Spivak, M.D. It was held from 16 to 18 October 2003 in New York City, New York, USA. Thirty-nine invited speakers from nine different countries participated in the conference. There were more than 350 attendees. There were formal presentations and discussions on biology, clinical aspects, and management of patients with these diverse bone marrow stem cell disorders linked by a variable progression to acute myeloid leukemia. Of considerable interest, a clinical symposium exclusively for patients was held the day preceding the meeting at which John Bennett, Tiziano Barbui, Richard Silver, Jerry Spivak, and Ayalew Tefferi spoke on various topics pertaining to these diseases. This proved to be highly informative to the patients who reported that they enjoyed the program immensely. This was sponsored by the Cancer Research & Treatment Fund, Inc. Representatives of the Myelodysplasia Foundation were also present. This meeting report provides a summary of five different sections prepared by one or more of the session chairs. The keynote address was given by Shahin Rafii (Cornell Medical Center). Most appropriately, this talk focused on the expression and activation of angiogenic factors which play a crucial role in the progression of both myeloproliferative disorders and myelodysplastic syndromes (MDS). Among the known factors, vascular endothelial growth tyrosine kinase receptors (VEGF-R1, R2, and R3) support proliferation, survival, and mobility. Rafii's team has demonstrated that these receptors are expressed on subsets of primary hematopoietic cells as well as leukemic cells. Some leukemic cells express both VEGF-A and VEGF-R2, resulting in the generation of an autocrine loop that supports survival and within the osteoblastic zone translocating these cells to the vascular enriched niche for receipt of molecular instructions required for proliferation and differentiation. A pathologic correlation can be seen in some patients with the identification of abnormal localization of immature precursors (ALIP) in the central portions of the medullary cavity. Misplaced megakaryocytes can release pro-fibrotic factors, including platelet derived growth factors and transforming growth factor-beta. Collectively, these data suggest that chronic disregulation of angiogenic factors alter the microenvironment dislocating marrow stem cells that force both proliferation and differentiation in varying degrees, contributing to these hematological disorders.


Assuntos
Síndromes Mielodisplásicas , Transtornos Mieloproliferativos , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva , Síndromes Mielodisplásicas/diagnóstico , Síndromes Mielodisplásicas/etiologia , Síndromes Mielodisplásicas/genética , Transtornos Mieloproliferativos/diagnóstico , Transtornos Mieloproliferativos/etiologia , Transtornos Mieloproliferativos/genética , Policitemia , Mielofibrose Primária , Trombocitose
13.
Leukemia ; 17(3): 604-11, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12646950

RESUMO

Vascular endothelial growth factor (VEGF) and its receptors (VEGFR) have been implicated in promoting solid tumor growth and metastasis via stimulating tumor-associated angiogenesis. We recently showed that certain 'liquid' tumors such as leukemia not only produce VEGF, but also express functional VEGFR, resulting in an autocrine loop for tumor growth and propagation. A chimeric anti-VEGFR2 (or kinase insert domain-containing receptor, KDR) antibody, IMC-1C11, was shown to be able to inhibit VEGF-induced proliferation of human leukemia cells in vitro, and to prolong survival of nonobese diabetic-severe combined immune deficient (NOD-SCID) mice inoculated with human leukemia cells. Here we produced two fully human anti-KDR antibodies (IgG1), IMC-2C6 and IMC-1121, from Fab fragments originally isolated from a large antibody phage display library. These antibodies bind specifically to KDR with high affinities: 50 and 200 pM for IMC-1121 and IMC-2C6, respectively, as compared to 270 pM for IMC-1C11. Like IMC-1C11, both human antibodies block VEGF/KDR interaction with an IC(50) of approximately 1 nM, but IMC-1121 is a more potent inhibitor to VEGF-stimulated proliferation of human endothelial cells. These anti-KDR antibodies strongly inhibited VEGF-induced migration of human leukemia cells in vitro, and when administered in vivo, significantly prolonged survival of NOD-SCID mice inoculated with human leukemia cells. It is noteworthy that the mice treated with antibody of the highest affinity, IMC-1121, survived the longest period of time, followed by mice treated with IMC-2C6 and IMC-1C11. Taken together, our data suggest that anti-KDR antibodies may have broad applications in the treatment of both solid tumors and leukemia. It further underscores the efforts to identify antibodies of high affinity for enhanced antiangiogenic and antitumor activities.


Assuntos
Anticorpos Monoclonais/farmacologia , Leucemia Experimental/tratamento farmacológico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Afinidade de Anticorpos , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Divisão Celular/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Fatores de Crescimento Endotelial/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Feminino , Células HL-60 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Leucemia Experimental/mortalidade , Linfocinas/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Recombinantes de Fusão , Taxa de Sobrevida , Veias Umbilicais/citologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
14.
Gene Ther ; 9(10): 631-41, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12032709

RESUMO

Adult bone marrow (BM) is a rich reservoir for endothelial and hematopoietic stem and progenitor cells that contribute to revascularization of injured and tumor tissue. Physiological stress results in the release of specific chemo-cytokines that promote mobilization of stem cells to the circulation and direct their incorporation into the target tissues. In order to dissect the mechanism and identify the cellular mediators that regulate stem cell recruitment, we have developed an in vivo murine model, in which the plasma levels of chemokines are elevated by introducing adenoviral vectors (Advectors) expressing such chemokines. Among the known stem cell-active chemokines, the angiogenic factor VEGF through interaction with its receptors, VEGFR2 and VEGFR1 expressed on endothelial and hematopoietic stem cells, promotes mobilization and recruitment of these cells into the neo-angiogenic sites, thereby accelerating the revascularization process. Based on these studies, it has become apparent that mobilization of stem cells is a dynamic process and requires sequential release of chemocytokines, expression of adhesion molecules and activation of proteases that facilitate egress of cells from the BM to the circulation. Chemokine-activation of metalloproteinases is essential for the release of bio-active cytokines, thereby enhancing stem cell mobilization potential. Advectors are ideal for delivery of chemocytokines since they allow for long-term robust expression facilitating in vivo proliferation and mobilization of large numbers of an otherwise rare population of stem cells. VEGF-mobilized endothelial and hematopoietic stem cells provide for an enriched source of adult pluripotent cells that can be used for revascularization, tissue regeneration or gene therapy.


Assuntos
Endotélio Vascular/citologia , Terapia Genética/métodos , Substâncias de Crescimento/genética , Mobilização de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas , Adenoviridae/genética , Adulto , Animais , Células da Medula Óssea , Moléculas de Adesão Celular/genética , Divisão Celular , Movimento Celular , Quimiocinas/genética , Fatores de Crescimento Endotelial/genética , Endotélio Vascular/metabolismo , Vetores Genéticos , Humanos , Linfocinas/genética , Metaloendopeptidases/metabolismo , Camundongos , Modelos Animais , Fator de Células-Tronco/genética , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
16.
Exp Hematol ; 29(12): 1417-24, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11750100

RESUMO

OBJECTIVE: The mechanism for anagrelide's potent platelet lowering activity in human subjects is not well defined. Studies related to anagrelide function have been hampered by its lack of activity in nonhuman primates and water insolubility. In an effort to define the mechanism whereby anagrelide exerts its therapeutic effect, we identified a water-soluble metabolite (anagrelide.met). The availability of anagrelide.met allowed, for the first time, parallel in vitro and in vivo animal studies centered on the mechanisms by which anagrelide lowers platelet levels. MATERIALS AND METHODS: The effects of anagrelide.met on proliferation and maturation of mega-karyocytes (MKs) as well as platelet production were studied both in vitro and in vivo. RESULTS: Anagrelide.met is capable of blocking in vitro MK migration by 20% to 40%. At 100 ng/mL, anagrelide.met selectively blocked in vitro MK maturation, resulting in a 50% decrease in the total number of CD41a(+) MKs, corresponding with a 30% decrease in MK ploidy by day 10 and a 60% decrease by day 20. Daily intraperitoneal injections of anagrelide.met 100 microg into BALB/c mice was sufficient to significantly decrease platelet counts within 24 to 48 hours, stabilizing to 40 to 50% of normal levels by day 5. This was associated with a 45% decrease in the number of developing MKs and an increase in thrombopoietin levels. Anagrelide.met did not alter WBC counts, hematocrit, or bleeding time, or lead to any apparent signs of toxicity. Furthermore, unlike the parent anagrelide compound, anagrelide.met did not inhibit ADP-induced platelet aggregation even at high concentrations (10 microg/mL). CONCLUSIONS: We describe a cross-species reactive anagrelide metabolite that selectively inhibits MK maturation and migration, lowering platelet levels without influencing platelet aggregation.


Assuntos
Megacariócitos/patologia , Trombocitopenia/fisiopatologia , Acetilcolinesterase/análise , Animais , Antígenos CD34/análise , Biomarcadores/análise , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/farmacologia , Sangue Fetal/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Agregação Plaquetária , Inibidores da Agregação Plaquetária/farmacologia , Contagem de Plaquetas , Ploidias , Quinazolinas/farmacologia
17.
Nat Med ; 7(11): 1194-201, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11689883

RESUMO

The role of bone marrow (BM)-derived precursor cells in tumor angiogenesis is not known. We demonstrate here that tumor angiogenesis is associated with recruitment of hematopoietic and circulating endothelial precursor cells (CEPs). We used the angiogenic defective, tumor resistant Id-mutant mice to show that transplantation of wild-type BM or vascular endothelial growth factor (VEGF)-mobilized stem cells restore tumor angiogenesis and growth. We detected donor-derived CEPs throughout the neovessels of tumors and Matrigel-plugs in an Id1+/-Id3-/- host, which were associated with VEGF-receptor-1-positive (VEGFR1+) myeloid cells. The angiogenic defect in Id-mutant mice was due to impaired VEGF-driven mobilization of VEGFR2+ CEPs and impaired proliferation and incorporation of VEGFR1+ cells. Although targeting of either VEGFR1 or VEGFR2 alone partially blocks the growth of tumors, inhibition of both VEGFR1 and VEGFR2 was necessary to completely ablate tumor growth. These data demonstrate that recruitment of VEGF-responsive BM-derived precursors is necessary and sufficient for tumor angiogenesis and suggest new clinical strategies to block tumor growth.


Assuntos
Células-Tronco Hematopoéticas/patologia , Proteínas de Neoplasias , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/patologia , Neovascularização Patológica , Proteínas Repressoras , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Endotélio Vascular/patologia , Transplante de Células-Tronco Hematopoéticas , Proteína 1 Inibidora de Diferenciação , Proteínas Inibidoras de Diferenciação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Mutação , Neovascularização Patológica/genética , Testes de Neutralização , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Receptores de Fatores de Crescimento do Endotélio Vascular , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular
19.
Proc Natl Acad Sci U S A ; 98(19): 10857-62, 2001 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-11553814

RESUMO

Antiangiogenic agents block the effects of tumor-derived angiogenic factors (paracrine factors), such as vascular endothelial growth factor (VEGF), on endothelial cells (EC), inhibiting the growth of solid tumors. However, whether inhibition of angiogenesis also may play a role in liquid tumors is not well established. We recently have shown that certain leukemias not only produce VEGF but also selectively express functional VEGF receptors (VEGFRs), such as VEGFR-2 (Flk-1, KDR) and VEGFR1 (Flt1), resulting in the generation of an autocrine loop. Here, we examined the relative contribution of paracrine (EC-dependent) and autocrine (EC-independent) VEGF/VEGFR signaling pathways, by using a human leukemia model, where autocrine and paracrine VEGF/VEGFR loops could be selectively inhibited by neutralizing mAbs specific for murine EC (paracrine pathway) or human tumor (autocrine) VEGFRs. Blocking either the paracrine or the autocrine VEGF/VEGFR-2 pathway delayed leukemic growth and engraftment in vivo, but failed to cure inoculated mice. Long-term remission with no evidence of disease was achieved only if mice were treated with mAbs against both murine and human VEGFR-2, whereas mAbs against human or murine VEGFR-1 had no effect on mice survival. Therefore, effective antiangiogenic therapies to treat VEGF-producing, VEGFR-expressing leukemias may require blocking both paracrine and autocrine VEGF/VEGFR-2 angiogenic loops to achieve remission and long-term cure.


Assuntos
Comunicação Autócrina , Fatores de Crescimento Endotelial/metabolismo , Leucemia Promielocítica Aguda/fisiopatologia , Linfocinas/metabolismo , Comunicação Parácrina , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Transdução de Sinais , Animais , Divisão Celular , Células Cultivadas , Técnicas de Cocultura , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/metabolismo , Camundongos , Invasividade Neoplásica , Transplante de Neoplasias , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/fisiopatologia , Neovascularização Patológica/fisiopatologia , Receptores de Fatores de Crescimento do Endotélio Vascular , Fatores de Tempo , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
20.
Microvasc Res ; 62(2): 83-93, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11516238

RESUMO

Idiopathic thrombotic thrombocytopenic purpura (TTP) is a disease characterized by the apoptotic injury of all microvascular endothelial cells (MVEC) except those of pulmonary origin. It notably also spares EC of large vessel origin. It is fatal unless treated with plasma exchange. The EC lineage restriction of the apoptotic lesions in vivo is reproduced in vitro following exposure of primary human MVEC derived from various tissues to TTP plasma. Oligonucleotide genechip technology was used to identify genes that may contribute to the resistance of lung MVEC to apoptosis induced by TTP plasma and to explore the intrinsic genotypic heterogeneity between MVEC of TTP-sensitive (skin) versus resistant (lung) lineage. Exposure of cells to TTP or normal plasma yielded 157 genes that were differentially expressed in primary human lung MVEC. A global change in expression of pro- and anti-apoptotic genes was seen, including increases in caspase 1, Fas, and Bcl-xl, already shown by experimental means to be involved in TTP pathogenesis. Additional differences suggest the importance of pathways related to the death receptor ligand TRAIL, as well as a role for disruption of EC-extracellular matrix interactions in the initiation of apoptosis. Maintenance of specific prosurvival signals at baseline may be a feature of lung MVEC resistance in TTP as suggested by higher expression than skin EC of the TRAIL antagonist, osteoprotegerin, and the vascular endothelial growth factors, VEGF/VPF and VEGF-C, and their receptors, VEGFR-2 (KDR) and VEGFR-3 (Flt4).


Assuntos
Apoptose/genética , Endotélio Vascular/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Púrpura Trombocitopênica Trombótica/fisiopatologia , Biomarcadores , Endotélio Vascular/citologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Síndrome Hemolítico-Urêmica/fisiopatologia , Humanos , Pulmão/irrigação sanguínea , Microcirculação , Pessoa de Meia-Idade , Púrpura Trombocitopênica Trombótica/sangue , Púrpura Trombocitopênica Trombótica/genética , Pele/irrigação sanguínea
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...