Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Oncologist ; 28(12): e1248-e1258, 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-37260332

RESUMO

INTRODUCTION: Targeted therapy in non-small cell lung cancer (NSCLC) patients with mesenchymal epithelial transition (MET) exon 14 skipping mutations (METex14) and MET amplifications has improved patients' outcomes. The development of more potent MET kinase inhibitors could further benefit these patients. The aim of this trial is to determine the safety and recommended phase 2 dose (RP2D) of OMO-1 (an oral dual MET kinase/OCT-2 inhibitor) and to assess preliminary clinical efficacy in METex14-positive NSCLC and other MET-positive solid tumors. MATERIALS AND METHODS: This was a first-in-patient, open-label, multicenter study of OMO-1 in patients with locally advanced or metastatic solid malignancies. A standard 3 + 3 dose escalation design was utilized starting at a dose level of 100 mg BID continuously. Preliminary efficacy was investigated in patients with METex14-positive NSCLC, and MET amplified NSCLC and other solid tumors (MET basket). RESULTS: In the dose-escalation part, 24 patients were included in 5 dose levels ranging from 100 mg twice daily (BID) to 400 mg BID. Most common adverse events (≥ 20%) were nausea, fatigue, vomiting, increased blood creatinine, and headache. The RP2D was determined at 250 mg BID. In the expansion cohorts, 15 patients were included (10 in METex14-positive NSCLC cohort and 5 in MET basket cohort) and received either 200 or 250 mg BID. Eight out of the 10 patients with METex14 positive NSCLC had stable disease as the best response. CONCLUSION: OMO-1 was tolerated at the dose of 250 mg BID and shows initial signs of MET inhibition and anti-tumor activity in METex14 mutated NSCLC patients.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Segunda Neoplasia Primária , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Proteínas Proto-Oncogênicas c-met/genética , Inibidores de Proteínas Quinases/efeitos adversos , Segunda Neoplasia Primária/genética , Éxons , Mutação
2.
Eur Heart J ; 40(4): 372-382, 2019 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-30452556

RESUMO

Aims: The E3-ligase CBL-B (Casitas B-cell lymphoma-B) is an important negative regulator of T cell activation that is also expressed in macrophages. T cells and macrophages mediate atherosclerosis, but their regulation in this disease remains largely unknown; thus, we studied the function of CBL-B in atherogenesis. Methods and results: The expression of CBL-B in human atherosclerotic plaques was lower in advanced lesions compared with initial lesions and correlated inversely with necrotic core area. Twenty weeks old Cblb-/-Apoe-/- mice showed a significant increase in plaque area in the aortic arch, where initial plaques were present. In the aortic root, a site containing advanced plaques, lesion area rose by 40%, accompanied by a dramatic change in plaque phenotype. Plaques contained fewer macrophages due to increased apoptosis, larger necrotic cores, and more CD8+ T cells. Cblb-/-Apoe-/- macrophages exhibited enhanced migration and increased cytokine production and lipid uptake. Casitas B-cell lymphoma-B deficiency increased CD8+ T cell numbers, which were protected against apoptosis and regulatory T cell-mediated suppression. IFNγ and granzyme B production was enhanced in Cblb-/-Apoe-/- CD8+ T cells, which provoked macrophage killing. Depletion of CD8+ T cells in Cblb-/-Apoe-/- bone marrow chimeras rescued the phenotype, indicating that CBL-B controls atherosclerosis mainly through its function in CD8+ T cells. Conclusion: Casitas B-cell lymphoma-B expression in human plaques decreases during the progression of atherosclerosis. As an important regulator of immune responses in experimental atherosclerosis, CBL-B hampers macrophage recruitment and activation during initial atherosclerosis and limits CD8+ T cell activation and CD8+ T cell-mediated macrophage death in advanced atherosclerosis, thereby preventing the progression towards high-risk plaques.


Assuntos
Aterosclerose/etiologia , Linfócitos T CD8-Positivos/imunologia , Linfoma de Células B/complicações , Macrófagos/patologia , Proteína Oncogênica v-cbl/metabolismo , Placa Aterosclerótica/etiologia , Animais , Apoptose , Aterosclerose/metabolismo , Aterosclerose/patologia , Modelos Animais de Doenças , Humanos , Linfoma de Células B/metabolismo , Linfoma de Células B/patologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia
3.
EMBO J ; 30(20): 4142-56, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21847094

RESUMO

Histone deacetylases (HDACs) deacetylate histones and non-histone proteins, thereby affecting protein activity and gene expression. The regulation and function of the cytoplasmic class IIb HDAC6 in endothelial cells (ECs) is largely unexplored. Here, we demonstrate that HDAC6 is upregulated by hypoxia and is essential for angiogenesis. Silencing of HDAC6 in ECs decreases sprouting and migration in vitro and formation of functional vascular networks in matrigel plugs in vivo. HDAC6 regulates zebrafish vessel formation, and HDAC6-deficient mice showed a reduced formation of perfused vessels in matrigel plugs. Consistently, overexpression of wild-type HDAC6 increases sprouting from spheroids. HDAC6 function requires the catalytic activity but is independent of ubiquitin binding and deacetylation of α-tubulin. Instead, we found that HDAC6 interacts with and deacetylates the actin-remodelling protein cortactin in ECs, which is essential for zebrafish vessel formation and which mediates the angiogenic effect of HDAC6. In summary, we show that HDAC6 is necessary for angiogenesis in vivo and in vitro, involving the interaction and deacetylation of cortactin that regulates EC migration and sprouting.


Assuntos
Movimento Celular , Cortactina/metabolismo , Histona Desacetilases/metabolismo , Neovascularização Fisiológica , Tubulina (Proteína)/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Acetilação , Animais , Carcinoma Pulmonar de Lewis/enzimologia , Células Cultivadas , Células Endoteliais/metabolismo , Feminino , Desacetilase 6 de Histona , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Knockout
4.
FASEB J ; 28(5): 2202-13, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24481967

RESUMO

Modulation of hematopoietic stem and progenitor cells (HSPCs) determines immune cell function. In this study, we investigated how hypercholesterolemia affects HSPC biology and atherosclerosis. Hypercholesterolemia induced loss of HSPC quiescence, characterized by increased proliferation and expression of cyclin B1, C1, and D1, and a decreased expression of Rb, resulting in a 3.6- fold increase in the number of HSPCs in hypercholesterolemic Ldlr(-/-) mice. Competitive bone marrow (BM) transplantations showed that a hypercholesterolemic BM microenvironment activates HSPCs and skews their development toward myeloid lineages. Conversely, hypercholesterolemia-primed HSPCs acquired an enhanced propensity to generate myeloid cells, especially granulocytes and Ly6C(high) monocytes, even in a normocholesterolemic BM microenvironment. In conformity, macrophages differentiated from hypercholesterolemia-primed HSPCs produced 17.0% more TNF-α, 21.3% more IL-6, and 10.5% more MCP1 than did their normocholesterolemic counterparts. Hypercholesterolemia-induced priming of HSPCs generated leukocytes that more readily migrated into the artery, which resulted in a 2.1-fold increase in atherosclerotic plaque size. In addition, these plaques had a more advanced phenotype and exhibited a 1.2-fold increase in macrophages and 1.8-fold increase in granulocytes. These results identify hypercholesterolemia-induced activation and priming of HSPCs as a novel pathway in the development of atherosclerosis. Inhibition of this proinflammatory differentiation pathway on the HSPC level has the potential to reduce atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Células-Tronco Hematopoéticas/citologia , Hipercolesterolemia/metabolismo , Animais , Aterosclerose/patologia , Transplante de Medula Óssea , Diferenciação Celular , Células Cultivadas , Granulócitos/metabolismo , Inflamação , Interleucina-6/metabolismo , Lipoproteínas/sangue , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fenótipo , Receptores de LDL/genética , Fator de Necrose Tumoral alfa/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 34(6): 1187-92, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24675658

RESUMO

OBJECTIVE: Platelets abundantly express the membrane receptor CD36 and store its ligand thrombospondin-1 (TSP1) in the α-granules. We investigated whether released TSP1 can support platelet adhesion and thrombus formation via interaction with CD36. APPROACH AND RESULTS: Mouse platelets deficient in CD36 showed reduced adhesion to TSP1 and subsequent phosphatidylserine expression. Deficiency in either CD36 or TSP1 resulted in markedly increased dissolution of thrombi formed on collagen, although thrombus buildup was unchanged. In mesenteric vessels in vivo, deficiency in CD36 prolonged the time to occlusion and enhanced embolization, which was in agreement with earlier observations in TSP1-deficient mice. Thrombi formed using wild-type blood stained positively for secreted TSP1. Releasate from wild-type but not from TSP1-deficient platelets enhanced platelet activation, phosphatidylserine expression, and thrombus formation on collagen. The enhancement was dependent on CD36 because it was without effect on thrombus formation by CD36-deficient platelets. CONCLUSIONS: These results demonstrate an anchoring role of platelet-released TSP1 via CD36 in platelet adhesion and collagen-dependent thrombus stabilization. Thus, the TSP1-CD36 tandem is another platelet ligand-receptor axis contributing to the maintenance of a stable thrombus.


Assuntos
Antígenos CD36/fisiologia , Colágeno/metabolismo , Trombose/etiologia , Trombospondina 1/fisiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Ativação Plaquetária , Adesividade Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia
6.
EMBO J ; 29(2): 424-41, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-20010698

RESUMO

Vascular endothelial growth factor (VEGF) and beta-catenin both act broadly in embryogenesis and adulthood, including in the skeletal and vascular systems. Increased or deregulated activity of these molecules has been linked to cancer and bone-related pathologies. By using novel mouse models to locally increase VEGF levels in the skeleton, we found that embryonic VEGF over-expression in osteo-chondroprogenitors and their progeny largely pheno-copied constitutive beta-catenin activation. Adult induction of VEGF in these cell populations dramatically increased bone mass, associated with aberrant vascularization, bone marrow fibrosis and haematological anomalies. Genetic and pharmacological interventions showed that VEGF increased bone mass through a VEGF receptor 2- and phosphatidyl inositol 3-kinase-mediated pathway inducing beta-catenin transcriptional activity in endothelial and osteoblastic cells, likely through modulation of glycogen synthase kinase 3-beta phosphorylation. These insights into the actions of VEGF in the bone and marrow environment underscore its power as pleiotropic bone anabolic agent but also warn for caution in its therapeutic use. Moreover, the finding that VEGF can modulate beta-catenin activity may have widespread physiological and clinical ramifications.


Assuntos
Osso e Ossos/metabolismo , Osso e Ossos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta Catenina/metabolismo , Animais , Osso e Ossos/embriologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Células Endoteliais/citologia , Humanos , Mesoderma/citologia , Camundongos , Camundongos Transgênicos , Morfogênese , Osteoblastos/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Células-Tronco/citologia , Células Estromais/citologia , Fator A de Crescimento do Endotélio Vascular/genética , beta Catenina/genética
7.
Cancer Cell ; 7(1): 3-4, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15652744

RESUMO

The first angiogenesis inhibitor has recently been approved for cancer treatment. Nonetheless, optimizing the dose of novel angiogenesis inhibitors remains a formidable challenge--primarily because we lack reliable surrogate markers of tumor angiogenesis. In this issue, Shaked et al. provide evidence that the levels of circulating endothelial precursor cells (CEPs), which contribute to the formation of tumor vessels, are genetically predetermined and regulated by angiogenic factors, and reflect the antitumor efficacy of angiogenesis inhibitors. These findings highlight the potential usefulness of CEPs as a surrogate marker to monitor and adjust antiangiogenic therapy.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Neoplasias/tratamento farmacológico , Neovascularização Patológica , Inibidores da Angiogênese/metabolismo , Animais , Biomarcadores , Ensaios Clínicos como Assunto , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Humanos , Neoplasias/patologia
8.
Cancer Cell ; 8(2): 131-41, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16098466

RESUMO

The hypoxia-inducible transcription factors HIF-1alpha and HIF-2alpha are activated in hypoxic tumor regions. However, their role in tumorigenesis remains controversial, as tumor growth promoter and suppressor activities have been ascribed to HIF-1alpha, while the role of HIF-2alpha remains largely unknown. Here, we show that overexpression of HIF-2alpha in rat glioma tumors enhances angiogenesis but reduces growth of these tumors, in part by increasing tumor cell apoptosis. Moreover, siRNA knockdown of HIF-2alpha reduced apoptosis in hypoxic human malignant glioblastoma cells. Furthermore, inhibition of HIF by overexpression of a dominant-negative HIF transgene in glioma cells or HIF-2alpha deficiency in teratomas reduced vascularization but accelerated growth of these tumor types. These findings urge careful consideration of using HIF inhibitors as cancer therapeutic strategies.


Assuntos
Glioma/irrigação sanguínea , Neovascularização Patológica , Fatores de Transcrição/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Glioma/genética , Glioma/metabolismo , Humanos , Camundongos , Neoplasias Neuroepiteliomatosas/irrigação sanguínea , Neoplasias Neuroepiteliomatosas/genética , Neoplasias Neuroepiteliomatosas/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , RNA Interferente Pequeno/genética , Ratos , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Ativação Transcricional , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/genética
9.
Blood ; 115(11): 2264-73, 2010 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-19965679

RESUMO

The transforming and tumor growth-promoting properties of Axl, a member of the Tyro3, Axl, and Mer (TAM) family of receptor tyrosine kinases (TAMRs), are well recognized. In contrast, little is known about the role of the TAMR ligand growth arrest-specific gene 6 (Gas6) in tumor biology. By using Gas6-deficient (Gas6(-/-)) mice, we show that bone marrow-derived Gas6 promotes growth and metastasis in different experimental cancer models, including one resistant to vascular endothelial growth factor inhibitors. Mechanistic studies reveal that circulating leukocytes produce minimal Gas6. However, once infiltrated in the tumor, leukocytes up-regulate Gas6, which is mitogenic for tumor cells. Consistent herewith, impaired tumor growth in Gas6(-/-) mice is rescued by transplantation of wild-type bone marrow and, conversely, mimicked by transplantation of Gas6(-/-) bone marrow into wild-type hosts. These findings highlight a novel role for Gas6 in a positive amplification loop, whereby tumors promote their growth by educating infiltrating leukocytes to up-regulate the production of the mitogen Gas6. Hence, inhibition of Gas6 might offer novel opportunities for the treatment of cancer.


Assuntos
Movimento Celular , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Leucócitos/metabolismo , Leucócitos/patologia , Mitógenos/biossíntese , Neoplasias/patologia , Animais , Transplante de Medula Óssea , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Inflamação/complicações , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/genética , Interleucina-10/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Metástase Neoplásica , Neoplasias/irrigação sanguínea , Neoplasias/enzimologia , Neoplasias/genética , Neovascularização Patológica/complicações , Neovascularização Patológica/genética , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia , Regulação para Cima/genética
10.
J Cell Biol ; 178(6): 1039-51, 2007 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-17785520

RESUMO

Duchenne muscular dystrophy (DMD) is a fatal and incurable muscle degenerative disorder. We identify a function of the protease urokinase plasminogen activator (uPA) in mdx mice, a mouse model of DMD. The expression of uPA is induced in mdx dystrophic muscle, and the genetic loss of uPA in mdx mice exacerbated muscle dystrophy and reduced muscular function. Bone marrow (BM) transplantation experiments revealed a critical function for BM-derived uPA in mdx muscle repair via three mechanisms: (1) by promoting the infiltration of BM-derived inflammatory cells; (2) by preventing the excessive deposition of fibrin; and (3) by promoting myoblast migration. Interestingly, genetic loss of the uPA receptor in mdx mice did not exacerbate muscular dystrophy in mdx mice, suggesting that uPA exerts its effects independently of its receptor. These findings underscore the importance of uPA in muscular dystrophy.


Assuntos
Distrofia Muscular de Duchenne/metabolismo , Mioblastos/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/deficiência , Animais , Transplante de Medula Óssea , Movimento Celular , Células Cultivadas , Fibrina/metabolismo , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Mioblastos/patologia , Receptores de Superfície Celular/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase
11.
Eur Heart J ; 32(5): 627-36, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21193434

RESUMO

AIMS: The identification of factors that mobilize subsets of endogenous progenitor cells may provide new therapeutic tools to enhance the repair of ischaemic tissue. We previously identified circulating mesenchymal cells that co-express endothelial markers (so-called circulating mesoangioblasts, cMABs) in children undergoing heart surgery with cardiopulmonary bypass (CPB). However, the mechanisms by which these cells are mobilized and their origin is unclear. METHODS AND RESULTS: Circulating CD73(+)CD45(-)KDR(+) cMABs were analysed in adults undergoing heart surgery with (n = 21) or without CPB (n = 8). During surgery with CPB, cMABs are mobilized with a maximal response at the end of the operation. In contrast, off-pump heart surgery does not stimulate cMAB mobilization, indicating that the stress mediated by CPB induces the mobilization of cMAB. Circulating mesoangioblasts were enriched in blood obtained from the coronary sinus. Histologically, CD73(+) cells were detected around vessels in the heart, indicating that the heart is one of the niches of cMABs. Consistently, studies in gender mismatched bone marrow transplanted patients demonstrated that cMABs did not originate from the bone marrow. Cytokine profiling of serum samples revealed that hepatocyte growth factor (HGF) was profoundly increased at the time point of maximal mobilization of cMABs. Hepatocyte growth factor stimulated the migration of cMABs. Importantly, injection of recombinant HGF increased cMABs in rats. CONCLUSIONS: Hepatocyte growth factor induces mobilization of non-haematopoietic progenitor cells with a cardiac repair capacity. This newly identified function together with the known pleiotrophic effects of HGF makes HGF an attractive therapeutic option for the treatment of ischaemic heart disease.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Leucócitos Mononucleares/citologia , Células-Tronco Mesenquimais/citologia , Idoso , Animais , Ponte Cardiopulmonar , Criança , Feminino , Humanos , Hibridização in Situ Fluorescente , Ligadura , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Infarto do Miocárdio/patologia , Ratos , Ratos Endogâmicos Lew , Proteínas Recombinantes/farmacologia
12.
J Clin Invest ; 118(2): 583-96, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18188450

RESUMO

Many patients with anemia fail to respond to treatment with erythropoietin (Epo), a commonly used hormone that stimulates erythroid progenitor production and maturation by human BM or by murine spleen. The protein product of growth arrest-specific gene 6 (Gas6) is important for cell survival across several cell types, but its precise physiological role remains largely enigmatic. Here, we report that murine erythroblasts released Gas6 in response to Epo and that Gas6 enhanced Epo receptor signaling by activating the serine-threonine kinase Akt in these cells. In the absence of Gas6, erythroid progenitors and erythroblasts were hyporesponsive to the survival activity of Epo and failed to restore hematocrit levels in response to anemia. In addition, Gas6 may influence erythropoiesis via paracrine erythroblast-independent mechanisms involving macrophages. When mice with acute anemia were treated with Gas6, the protein normalized hematocrit levels without causing undesired erythrocytosis. In a transgenic mouse model of chronic anemia caused by insufficient Epo production, Gas6 synergized with Epo in restoring hematocrit levels. These findings may have implications for the treatment of patients with anemia who fail to adequately respond to Epo.


Assuntos
Anemia/tratamento farmacológico , Anemia/genética , Eritropoese/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/uso terapêutico , Animais , Adesão Celular/genética , Sobrevivência Celular , Modelos Animais de Doenças , Resistência a Medicamentos , Eritroblastos/efeitos dos fármacos , Eritroblastos/metabolismo , Eritropoetina/genética , Eritropoetina/farmacologia , Eritropoetina/uso terapêutico , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Mutantes , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptores da Eritropoetina/agonistas , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , c-Mer Tirosina Quinase , Receptor Tirosina Quinase Axl
13.
Blood ; 113(3): 755-64, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18824596

RESUMO

Allograft vasculopathy is the leading cause of death in patients with heart transplantation. Accelerated endothelial regeneration mediated by enhanced endothelial progenitor cell (EPC) incorporation may attenuate the development of allograft vasculopathy. We investigated the hypothesis that modulation of EPC biology and attenuation of allograft vasculopathy by increased high-density lipoprotein cholesterol after human apo A-I (AdA-I) transfer requires scavenger receptor (SR)-BI expression in bone marrow-derived EPCs. After AdA-I transfer, the number of circulating EPCs increased 2.0-fold (P < .001) at different time points in C57BL/6 mice transplanted with SR-BI(+/+) bone marrow but remained unaltered in mice with SR-BI(-/-) bone marrow. The effect of high-density lipoprotein on EPC migration in vitro requires signaling via SR-BI and extracellular signal-regulated kinases and is dependent on increased nitric oxide (NO) production in EPCs. Human apo A-I transfer 2 weeks before paratopic artery transplantation reduced intimal area at day 21 3.7-fold (P < .001) in mice with SR-BI(+/+) bone marrow but had no effect in mice with SR-BI(-/-) bone marrow. AdA-I transfer potently stimulated EPC incorporation and accelerated endothelial regeneration in chimeric SR-BI(+/+) mice but not in chimeric SR-BI(-/-) mice. In conclusion, human apo A-I transfer accelerates endothelial regeneration mediated via SR-BI expressing bone marrow-derived EPCs, thereby preventing allograft vasculopathy.


Assuntos
Apolipoproteína A-I/metabolismo , Vasos Sanguíneos/patologia , Células Endoteliais/metabolismo , Receptores Depuradores Classe B/metabolismo , Células-Tronco/metabolismo , Animais , Apolipoproteína A-I/genética , Western Blotting , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Artérias Carótidas/transplante , Movimento Celular , HDL-Colesterol/sangue , Ensaio de Imunoadsorção Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Óxido Nítrico/metabolismo , Fosforilação , Regeneração , Receptores Depuradores Classe B/genética , Transdução de Sinais/fisiologia , Transplante Homólogo
14.
Nat Med ; 8(7): 702-10, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12053176

RESUMO

Respiratory distress syndrome (RDS) due to insufficient production of surfactant is a common and severe complication of preterm delivery. Here, we report that loss of the hypoxia-inducible transcription factor-2alpha (HIF-2alpha) caused fatal RDS in neonatal mice due to insufficient surfactant production by alveolar type 2 cells. VEGF, a target of HIF-2alpha, regulates fetal lung maturation: because VEGF levels in alveolar cells were reduced in HIF-2alpha-deficient fetuses; mice with a deficiency of the VEGF(164) and VEGF(188) isoforms or of the HIF-binding site in the VEGF promotor died of RDS; intrauterine delivery of anti-VEGF-receptor-2 antibodies caused RDS and VEGF stimulated production of surfactant proteins by cultured type 2 pneumocytes. Intrauterine delivery or postnatal intratracheal instillation of VEGF stimulated conversion of glycogen to surfactant and protected preterm mice against RDS. The pneumotrophic effect of VEGF may have therapeutic potential for lung maturation in preterm infants.


Assuntos
Fatores de Crescimento Endotelial/fisiologia , Pneumopatias/prevenção & controle , Linfocinas/fisiologia , Transativadores/fisiologia , Envelhecimento , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Modelos Animais de Doenças , Fatores de Crescimento Endotelial/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/crescimento & desenvolvimento , Pulmão/patologia , Pulmão/fisiopatologia , Pneumopatias/patologia , Pneumopatias/fisiopatologia , Linfocinas/farmacologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Alvéolos Pulmonares/crescimento & desenvolvimento , Alvéolos Pulmonares/patologia , Síndrome do Desconforto Respiratório/patologia , Mucosa Respiratória/patologia , Mucosa Respiratória/fisiopatologia , Transativadores/deficiência , Transativadores/genética , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
15.
Nat Med ; 8(8): 831-40, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12091877

RESUMO

The therapeutic potential of placental growth factor (PlGF) and its receptor Flt1 in angiogenesis is poorly understood. Here, we report that PlGF stimulated angiogenesis and collateral growth in ischemic heart and limb with at least a comparable efficiency to vascular endothelial growth factor (VEGF). An antibody against Flt1 suppressed neovascularization in tumors and ischemic retina, and angiogenesis and inflammatory joint destruction in autoimmune arthritis. Anti-Flt1 also reduced atherosclerotic plaque growth and vulnerability, but the atheroprotective effect was not attributable to reduced plaque neovascularization. Inhibition of VEGF receptor Flk1 did not affect arthritis or atherosclerosis, indicating that inhibition of Flk1-driven angiogenesis alone was not sufficient to halt disease progression. The anti-inflammatory effects of anti-Flt1 were attributable to reduced mobilization of bone marrow-derived myeloid progenitors into the peripheral blood; impaired infiltration of Flt1-expressing leukocytes in inflamed tissues; and defective activation of myeloid cells. Thus, PlGF and Flt1 constitute potential candidates for therapeutic modulation of angiogenesis and inflammation.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Arteriosclerose/tratamento farmacológico , Artrite Experimental/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Neovascularização Fisiológica , Proteínas da Gravidez/farmacologia , Proteínas Proto-Oncogênicas/imunologia , Receptores Proteína Tirosina Quinases/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Arteriosclerose/imunologia , Arteriosclerose/patologia , Artrite Experimental/patologia , Vasos Sanguíneos/citologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/patologia , Modelos Animais de Doenças , Fatores de Crescimento Endotelial/farmacologia , Feminino , Células-Tronco Hematopoéticas/efeitos dos fármacos , Membro Posterior/irrigação sanguínea , Humanos , Isquemia/tratamento farmacológico , Isquemia/patologia , Articulações/patologia , Linfocinas/farmacologia , Camundongos , Camundongos Nus , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Neovascularização Fisiológica/efeitos dos fármacos , Fator de Crescimento Placentário , Proteínas da Gravidez/genética , Proteínas da Gravidez/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento/imunologia , Receptores de Fatores de Crescimento/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fator A de Crescimento do Endotélio Vascular , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
16.
J Clin Invest ; 117(11): 3188-97, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17975666

RESUMO

Although studies have suggested a role for angiogenesis in determining heart size during conditions demanding enhanced cardiac performance, the role of EC mass in determining the normal organ size is poorly understood. To explore the relationship between cardiac vasculature and normal heart size, we generated a transgenic mouse with a regulatable expression of the secreted angiogenic growth factor PR39 in cardiomyocytes. A significant change in adult mouse EC mass was apparent by 3 weeks following PR39 induction. Heart weight; cardiomyocyte size; vascular density normalization; upregulation of hypertrophy markers including atrial natriuretic factor, beta-MHC, and GATA4; and activation of the Akt and MAP kinase pathways were observed at 6 weeks post-induction. Treatment of PR39-induced mice with the eNOS inhibitor L-NAME in the last 3 weeks of a 6-week stimulation period resulted in a significant suppression of heart growth and a reduction in hypertrophic marker expression. Injection of PR39 or another angiogenic growth factor, VEGF-B, into murine hearts during myocardial infarction led to induction of myocardial hypertrophy and restoration of myocardial function. Thus stimulation of vascular growth in normal adult mouse hearts leads to an increase in cardiac mass.


Assuntos
Cardiomegalia , Coração , Miocárdio , Neovascularização Fisiológica , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Animais , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Células Cultivadas , Ecocardiografia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Inibidores Enzimáticos/metabolismo , Coração/anatomia & histologia , Hemodinâmica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , NG-Nitroarginina Metil Éster/metabolismo , Tamanho do Órgão , Ratos , Ratos Sprague-Dawley , Transgenes
17.
Basic Res Cardiol ; 105(6): 703-12, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20652278

RESUMO

Diabetes is characterized by a chronic stage of hyperglycemia associated with endothelial progenitor cell dysfunction and reduced neovascularization in response to tissue ischemia. The underlying mechanisms are not entirely clear. The bone marrow niches provide the essential microenvironment for maintenance of stem cell function in the bone marrow. A disturbed stem cell niche might lead to stem cell dysfunction, thereby, impairing progenitor cell-dependent vascular repair. Therefore, we investigated the effects of streptozotocin-induced diabetes on the bone marrow stem cell niches and stem cell function in mice. Here, we show that long-term diabetes induced a reduction in Lin⁻Sca-1(+)c-kit(+) hematopoietic progenitor cells and reduced the repopulation capacity in a competitive engraftment experiment. Consistently, the expression of Bmi1, which prevents hematopoietic progenitor cell senescence, was significantly reduced in diabetic bone marrow cells. To address the mechanism underlying the progenitor cell dysfunction, we analyzed the composition of the stem cell niche and the cytokine environment. Although the morphology of the vascular and endosteal niche was not affected by diabetes, diabetic mice showed a significant deterioration of cytokine expression patterns in the bone marrow. In summary, these data indicate that diabetes imposes a long-term effect on the stem cell niche and affects important hematopoietic progenitor cell functions in mice.


Assuntos
Células da Medula Óssea/patologia , Proliferação de Células , Citocinas/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/patologia , Células-Tronco Hematopoéticas/patologia , Nicho de Células-Tronco , Animais , Antígenos Ly/metabolismo , Células da Medula Óssea/imunologia , Transplante de Medula Óssea , Diferenciação Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/fisiopatologia , Células Endoteliais/imunologia , Células Endoteliais/patologia , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/imunologia , Membro Posterior , Isquemia/imunologia , Isquemia/patologia , Isquemia/fisiopatologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Proteínas Nucleares/metabolismo , Osteoblastos/imunologia , Osteoblastos/patologia , Osteoclastos/imunologia , Osteoclastos/patologia , Complexo Repressor Polycomb 1 , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Tempo
18.
Blood ; 112(10): 4048-50, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18723692

RESUMO

The role of proteinases in the mobilization of hematopoietic progenitor cells (HPCs) after granulocyte colony-stimulating factor (G-CSF) remains unclear. Here we report that genetic loss of the plasminogen activator inhibitor Pai-1 or of the plasmin inhibitor alpha2-antiplasmin increases HPC mobilization in response to G-CSF. Moreover, thrombolytic agents, such as tenecteplase and microplasmin, enhance HPC mobilization in mice and humans. Taken together, these findings identify a novel role for plasmin in augmenting HPC mobilization in response to G-CSF.


Assuntos
Fibrinolisina/administração & dosagem , Fibrinolíticos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Mobilização de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/citologia , Animais , Feminino , Humanos , Masculino , Camundongos , Fragmentos de Peptídeos/administração & dosagem , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Serpina E2 , Serpinas/genética , Serpinas/metabolismo , Tenecteplase , Ativador de Plasminogênio Tecidual/administração & dosagem , alfa 2-Antiplasmina/genética , alfa 2-Antiplasmina/metabolismo
19.
Circ Res ; 103(10): 1181-9, 2008 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-18818405

RESUMO

Thrombospondin (TSP)1 is implicated in various inflammatory processes, but its role in atherosclerotic plaque formation and progression is unclear. Therefore, the development of atherosclerosis was compared in ApoE(-/-) and Tsp1(-/-)ApoE(-/-) mice kept on a normocholesterolemic diet. At 6 months, morphometric analysis of the aortic root of both mouse genotypes showed comparable lesion areas. Even when plaque burden increased approximately 5-fold in ApoE(-/-) and 10-fold in Tsp1(-/-)ApoE(-/-) mice, during the subsequent 3 months, total plaque areas were comparable at 9 months. In contrast, plaque composition differed substantially between genotypes: smooth muscle cell areas, mostly located in the fibrous cap of ApoE(-/-) plaques, both at 6 and 9 months, were 3-fold smaller in Tsp1(-/-)ApoE(-/-) plaques, which, in addition, were also more fibrotic. Moreover, inflammation by macrophages was twice as high in Tsp1(-/-)ApoE(-/-) plaques. This correlated with a 30-fold elevated incidence of elastic lamina degradation, with matrix metalloproteinase-9 accumulation, underneath plaques and manifestation of ectasia, exclusively in Tsp1(-/-)ApoE(-/-) mice. At 9 months, the necrotic core was 1.4-fold larger and 4-fold higher numbers of undigested disintegrated apoptotic cells were found in Tsp1(-/-)ApoE(-/-) plaques. Phagocytosis of platelets by cultured Tsp1(-/-) macrophages revealed the instrumental role of TSP1 in phagocytosis, corroborating the defective intraplaque phagocytosis of apoptotic cells. Hence, the altered smooth muscle cell phenotype in Tsp1(-/-)ApoE(-/-) mice has limited quantitative impact on atherosclerosis, but defective TSP1-mediated phagocytosis enhanced plaque necrotic core formation, accelerating inflammation and macrophage-induced elastin degradation by metalloproteinases, speeding up plaque maturation and vessel wall degeneration.


Assuntos
Apolipoproteínas E , Aterosclerose/metabolismo , Macrófagos Peritoneais/metabolismo , Miócitos de Músculo Liso/metabolismo , Fagocitose , Trombospondina 1 , Animais , Apolipoproteínas E/genética , Apoptose/genética , Aterosclerose/genética , Aterosclerose/patologia , Plaquetas/metabolismo , Plaquetas/patologia , Elastina/genética , Elastina/metabolismo , Genótipo , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/patologia , Necrose/metabolismo , Necrose/patologia , Fagocitose/genética , Trombospondina 1/genética
20.
Curr Opin Lipidol ; 20(5): 386-92, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19644365

RESUMO

PURPOSE OF REVIEW: Growth arrest-specific gene 6 (Gas6) belongs to the family of vitamin K-dependent coagulation proteins, but in contrast to its other members, has only a limited role in hemostasis. Instead, Gas6 plays a prominent role in conditions of injury, inflammation and repair. Gas6 amplifies the activation of various cell types including endothelial cells and platelets in different models of thrombosis and inflammation, processes also important in atherosclerosis. RECENT FINDINGS: Recently, we showed that in human and murine atherosclerotic plaques, Gas6 is expressed by endothelial cells, smooth muscle cells and most abundantly by macrophages, and that its expression increases with atherosclerosis severity. Moreover, genetic loss of Gas6 in ApoE mice reduced the influx of inflammatory cells in the plaque and induced plaque fibrosis, hence creating a stable plaque phenotype. Consistent herewith, Gas6 plasma levels are increased in patients with unstable angina pectoris, which is a common consequence of atherosclerotic plaque rupture. SUMMARY: Inhibition of Gas6 would be an attractive therapeutic target for stabilizing atherosclerotic plaques and for the prevention of vascular thrombotic occlusion after plaque rupture. Here we will critically review the existing literature on the potential roles of Gas6 and its receptors in the different stages of atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Animais , Aterosclerose/imunologia , Aterosclerose/patologia , Células Endoteliais/metabolismo , Humanos , Leucócitos/imunologia , Leucócitos/metabolismo , Necrose/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA