Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Biol Ther ; 15(9): 1208-18, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24921944

RESUMO

Stem cell factor receptor (c-Kit) exerts multiple biological effects on target cells upon binding its ligand stem cell factor (SCF). Aberrant activation of c-Kit results in dysregulated signaling and is implicated in the pathogenesis of numerous cancers. The development of more specific and effective c-Kit therapies is warranted given its essential role in tumorigenesis. In this study, we describe the biological properties of CK6, a fully human IgG1 monoclonal antibody against the extracellular region of human c-Kit. CK6 specifically binds c-Kit receptor with high affinity (EC 50 = 0.06 nM) and strongly blocks its interaction with SCF (IC 50 = 0.41 nM) in solid phase assays. Flow cytometry shows CK6 binding to c-Kit on the cell surface of human small cell lung carcinoma (SCLC), melanoma, and leukemia tumor cell lines. Furthermore, exposure to CK6 inhibits SCF stimulation of c-Kit tyrosine kinase activity and downstream signaling pathways such as mitogen-activated protein kinase (MAPK) and protein kinase B (AKT), in addition to reducing tumor cell line growth in vitro. CK6 treatment significantly decreases human xenograft tumor growth in NCI-H526 SCLC (T/C% = 57) and Malme-3M melanoma (T/C% = 58) models in vivo. The combination of CK6 with standard of care chemotherapy agents, cisplatin and etoposide for SCLC or dacarbazine for melanoma, more potently reduces tumor growth (SCLC T/C% = 24, melanoma T/C% = 38) compared with CK6 or chemotherapy alone. In summary, our results demonstrate that CK6 is a c-Kit antagonist antibody with tumor growth neutralizing properties and are highly suggestive of potential therapeutic application in treating human malignancies harboring c-Kit receptor.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Imunoglobulina G/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Melanoma Experimental/tratamento farmacológico , Proteínas Proto-Oncogênicas c-kit/metabolismo , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/administração & dosagem , Dacarbazina/administração & dosagem , Etoposídeo/administração & dosagem , Feminino , Xenoenxertos , Humanos , Imunoglobulina G/administração & dosagem , Imunoglobulina G/farmacologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos Nus , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/imunologia , Transdução de Sinais , Carcinoma de Pequenas Células do Pulmão/metabolismo , Carcinoma de Pequenas Células do Pulmão/patologia
2.
Mol Cancer Ther ; 11(11): 2473-82, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22933705

RESUMO

In lung cancer, platelet-derived growth factor receptor α (PDGFRα) is expressed frequently by tumor-associated stromal cells and by cancer cells in a subset of tumors. We sought to determine the effect of targeting stromal PDGFRα in preclinical lung tumor xenograft models (human tumor, mouse stroma). Effects of anti-human (IMC-3G3) and anti-mouse (1E10) PDGFRα monoclonal antibodies (mAb) on proliferation and PDGFRα signaling were evaluated in lung cancer cell lines and mouse fibroblasts. Therapy studies were conducted using established PDGFRα-positive H1703 cells and PDGFRα-negative Calu-6, H1993, and A549 subcutaneous tumors in immunocompromised mice treated with vehicle, anti-PDGFRα mAbs, chemotherapy, or combination therapy. Tumors were analyzed for growth and levels of growth factors. IMC-3G3 inhibited PDGFRα activation and the growth of H1703 cells in vitro and tumor growth in vivo, but had no effect on PDGFRα-negative cell lines or mouse fibroblasts. 1E10 inhibited growth and PDGFRα activation of mouse fibroblasts, but had no effect on human cancer cell lines in vitro. In vivo, 1E10-targeted inhibition of murine PDGFRα reduced tumor growth as single-agent therapy in Calu-6 cells and enhanced the effect of chemotherapy in xenografts derived from A549 cells. We also identified that low expression cancer cell expression of VEGF-A and elevated expression of PDGF-AA were associated with response to stromal PDGFRα targeting. We conclude that stromal PDGFRα inhibition represents a means for enhancing control of lung cancer growth in some cases, independent of tumor cell PDGFRα expression.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Terapia de Alvo Molecular , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Especificidade da Espécie , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Células Estromais/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Neoplasia ; 13(1): 49-59, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21245940

RESUMO

The benefits of inhibiting vascular endothelial growth factor (VEGF) signaling in cancer patients are predominantly attributed to effects on tumor endothelial cells. Targeting non-endothelial stromal cells to further impact tumor cell growth and survival is being pursued through the inhibition of additional growth factor pathways important for the survival and/or proliferation of these cells. However, recent data suggest that VEGF receptor (VEGFR)-specific inhibitors may target lymphatic vessels and pericytes in addition to blood vessels. Here, in fact, we demonstrate that DC101 (40 mg/kg, thrice a week), an antibody specific to murine VEGFR2, significantly reduces all three of these stromal components in subcutaneous (SKRC-29) and orthotopic (786-O-LP) models of renal cell carcinoma (RCC) established in nu/nu athymic mice. Sunitinib (40 mg/kg, once daily), a receptor tyrosine kinase inhibitor of VEGFR2 and other growth factor receptors, also caused significant loss of tumor blood vessels in RCC models but had weaker effects than DC101 on pericytes and lymphatic vessels. In combination, sunitinib did not significantly add to the effects of DC101 on tumor blood vessels, lymphatic vessels, or pericytes. Nevertheless, sunitinib increased the effect of DC101 on tumor burden in the SKRC-29 model, perhaps related to its broader specificity. Our data have important implications for combination therapy design, supporting the conclusion that targeting VEGFR2 alone in RCC has the potential to have pleiotropic effects on tumor stroma.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Indóis/farmacologia , Neoplasias Renais/tratamento farmacológico , Pirróis/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/irrigação sanguínea , Carcinoma de Células Renais/patologia , Modelos Animais de Doenças , Interações Medicamentosas , Feminino , Humanos , Fator 1 Induzível por Hipóxia/biossíntese , Indóis/uso terapêutico , Neoplasias Renais/irrigação sanguínea , Neoplasias Renais/patologia , Vasos Linfáticos/efeitos dos fármacos , Vasos Linfáticos/patologia , Camundongos , Camundongos Nus , Mutação , Transplante de Neoplasias , Neovascularização Patológica , Pericitos/efeitos dos fármacos , Pericitos/patologia , Pirróis/uso terapêutico , Células Estromais/efeitos dos fármacos , Células Estromais/patologia , Sunitinibe , Carga Tumoral , Células Tumorais Cultivadas , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia , Proteína Supressora de Tumor Von Hippel-Lindau/genética
4.
Circ Res ; 100(1): e1-11, 2007 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-17122441

RESUMO

The mechanisms that govern the capacity of the bone marrow stem cells to generate cardiac myocytes are still unknown. Herein we demonstrate that the cardiomyogenic potential of bone marrow-derived Oct3/4(+)/cKit(+/-)/CXCR4(+/-)/CD34(-)/Sca1(-) cells is governed by age-dependent paracrine/juxtacrine platelet-derived growth factor (PDGF) pathways. Specifically, bone marrow cell cultures from both 3- and 18-month-old mice formed aggregates of Oct3/4(+) cells circumscribed by PDGFRalpha(+)/Oct3/4(-)/Sca1(+) cells. In young (3-month) bone marrow cell cultures, induction of PDGF-AB preceded the induction of cardiac genes and was required for the generation of cardiomyogenesis. Indeed, in old (18-month) cultures, diminished PDGF-B induction was associated with impaired cardiomyogenic potential, despite having Oct3/4 levels similar to those in the young cells. Importantly, supplementation with PDGF-AB specifically restored the cardiac differentiation capacity of the old bone marrow cells. Together these results demonstrate that, regardless of age, the bone marrow niche contains Oct3/4 stem cells that are capable of differentiating into cardiac myocytes. Moreover, this differentiation is governed by age-dependent PDGF-AB-mediated paracrine/juxtacrine pathways that may be essential in the translation of bone marrow cell-mediated cardiomyogenesis.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular/fisiologia , Senescência Celular/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Comunicação Parácrina/fisiologia , Animais , Células da Medula Óssea/metabolismo , Agregação Celular , Células Cultivadas , Ligantes , Camundongos , Miócitos Cardíacos/fisiologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/fisiologia , Esferoides Celulares/citologia , Células-Tronco/citologia
5.
FASEB J ; 20(6): 717-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16461331

RESUMO

Microenvironmental cues mediate postnatal neovascularization via modulation of endothelial cell and bone marrow-derived endothelial progenitor cell (EPC) activity. Numerous signals regulate the activity of both of these cell types in response to vascular injury, which suggests that parallel mechanisms regulate angiogenesis in the vascular beds of both the heart and bone marrow. To identify mediators of such shared pathways, in vivo bone marrow/cardiac phage display biopanning was performed and led to the identification of tenascin-C as a candidate protein. Functionally, tenascin-C inhibits cardiac endothelial cell spreading and enhances migration in response to angiogenic growth factors. Analysis of human coronary thrombi revealed tenascin-C protein expression colocalized with the endothelial cell/EPC marker Tie-2 in intrathrombi vascular channels. Immunostains in the rodent heart demonstrated that tenascin-C also colocalizes with EPCs homing to sites of cardiac angiogenic induction. To determine the importance of tenascin-C in cardiac neovascularization, we used an established cardiac transplantation model and showed that unlike wild-type mice, tenascin-C-/- mice fail to vascularize cardiac allografts. This demonstrates for the first time that tenascin-C is essential for postnatal cardiac angiogenic function. Together, our data highlight the role of tenascin-C as a microenvironmental regulator of cardiac endothelial/EPC activity.


Assuntos
Vasos Coronários/crescimento & desenvolvimento , Endotélio Vascular/fisiologia , Neovascularização Fisiológica/fisiologia , Tenascina/metabolismo , Envelhecimento , Animais , Células da Medula Óssea/metabolismo , Adesão Celular , Movimento Celular , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Fenótipo , Ratos , Ratos Endogâmicos F344 , Tenascina/genética , Trombose/metabolismo
6.
Exp Gerontol ; 41(1): 63-8, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16337354

RESUMO

Recent studies have demonstrated that targeting of an age-associated impairment in platelet-derived growth factor (PDGF-AB) pathways could reduce histological measures of myocardial infarction in aging rat hearts. To facilitate preclinical developments of this approach, non-invasive measures of cardiac function were investigated in a 24-month-old rat myocardial infarction model employing intramyocardial PDGF-AB (100 ng) or vehicle control pretreatment. Electrocardiographic recordings post-coronary occlusion revealed ST segment elevation-myocardial injury patterns in both groups, which was confirmed histologically 2 weeks later by Masson's trichrome stains (PDGF-AB, 14.6+/-2.8% of left ventricular area (LVA) vs. control, 27.9+/-9.2%; P<0.05). Echocardiographic fractional shortening (FS) measurements revealed greater preservation of cardiac function in PDGF-AB-treated hearts compared with controls (PDGF-AB FS: 27.3+/-3.7% vs. control--16.7+/-4.1% (ANOVA P=0.005) vs. sham operation--34.5+/-6.7%), with a significant inverse relationship between FS and extent of myocardial injury (m=-0.68; r=-0.84). Notably, exercise testing did not correlate with myocardial injury. These findings provide an important functional foundation in preclinical translations of PDGF-AB-based cardioprotective treatment strategies. Moreover, demonstration of respective roles of electrocardiography and echocardiography in the confirmation and correlation of myocardial injury in the aging rat heart may serve to facilitate both PDGF-AB-based and other age-targeted approaches in large animal models of aging and cardiovascular disease.


Assuntos
Envelhecimento/fisiologia , Cardiotônicos/uso terapêutico , Infarto do Miocárdio/prevenção & controle , Fator de Crescimento Derivado de Plaquetas/uso terapêutico , Animais , Modelos Animais de Doenças , Ecocardiografia Transesofagiana , Eletrocardiografia , Teste de Esforço , Feminino , Coração/fisiopatologia , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes/uso terapêutico
7.
Am J Physiol Heart Circ Physiol ; 290(4): H1387-92, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16339836

RESUMO

Diabetes mellitus is associated with an increased risk of vascular disease, with significant alterations in systemic endothelial progenitor cells (EPCs) and peripheral vascular function. To identify the contribution of the different vascular compartments in the diabetic impairment of vascularization, we employed streptozotocin- and control-treated 3-mo-old C57Bl/6 mice in an isogeneic pinnal cardiac allograft model, revealing a significant delay in vascularization of wild-type cardiac tissue transplanted into diabetic mice. To investigate the basis of this impairment, the function of diabetic bone marrow cells was tested by transplantation of bone marrow cells isolated from diabetic and control mice into intact, unirradiated 18-mo-old C57Bl/6 mice, which have impaired function of both EPCs and peripheral endothelial cells. Importantly, cells derived from control, but not diabetic, bone marrow integrated into transplanted cardiac allografts. To assess the contribution of diabetic changes in the local vasculature, diabetic mice were treated with pinnal injections of platelet-derived growth factor (PDGF)-AB, which promotes cardiac angiogenesis in wild-type mice. However, whereas PDGF-AB enhanced allograft function in control mice, the activity of the cardiac transplants in the PDGF-AB-treated diabetic mice was significantly decreased. To decipher the potential interactions between systemic bone marrow-derived cells and local vascular pathways, diabetic mice were transplanted with wild-type bone marrow cells with or without PDGF-AB pinnal pretreatment, resulting in improved allograft function and donor cell recruitment only in the combination treatment arm. Overall, these studies show that the diabetic impairment in cardiac angiogenesis can be reversed by targeting the synergism between local trophic pathways and systemic cell function.


Assuntos
Transplante de Medula Óssea/métodos , Angiopatias Diabéticas/fisiopatologia , Angiopatias Diabéticas/terapia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/cirurgia , Fator de Crescimento Derivado de Plaquetas/administração & dosagem , Animais , Angiopatias Diabéticas/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estreptozocina , Resultado do Tratamento
8.
Physiol Genomics ; 24(3): 191-7, 2006 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-16352696

RESUMO

Aging is associated with shifts in autocrine and paracrine pathways in the cardiac vasculature that may contribute to the risk of cardiovascular disease in older persons. To elucidate the molecular basis of these changes in vivo, phage-display biopanning of 3- and 18-mo-old mouse hearts was performed that identified peptide epitopes with homology to brain-derived neurotrophic factor (BDNF) in old but not young phage pools. Quantification of cardiac phage binding by titration and immunostaining after injection with BDNF-like phage identified a twofold increased density of the BDNF receptor, truncated Trk B, in the aging hearts. Studies focused on the receptor ligand using a rat model of transient myocardial ischemia revealed increases in cardiac BDNF associated with local mononuclear infiltrates in 24- but not 4-mo-old rats. To investigate these changes, both 4- and 24-mo-old rat hearts were treated with intramyocardial injections of BDNF (or PBS control), demonstrating significant inflammatory increases with activated macrophage (ED1+) in BDNF-treated aging hearts compared with aging controls and similarly treated young hearts. Additional studies with permanent coronary occlusion following intramyocardial growth factor pretreatment revealed that BDNF significantly increased the extent of myocardial injury in older rat hearts (BDNF 35 +/- 10% vs. PBS 16.2 +/- 7.9% left ventricular injury; P < 0.05) without affecting younger hearts (BDNF 15 +/- 5.1% vs. PBS 14.5 +/- 6.0% left ventricular injury). Overall, these studies suggest that age-associated changes in BDNF-Trk B pathways may predispose the aging heart to increased injury after acute myocardial infarction and potentially contribute to the enhanced severity of cardiovascular disease in older individuals.


Assuntos
Envelhecimento/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Inflamação/metabolismo , Miocárdio/metabolismo , Animais , Feminino , Camundongos , Infarto do Miocárdio/metabolismo , Miocárdio/enzimologia , Biblioteca de Peptídeos , Ratos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor trkB/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo
9.
Am J Physiol Heart Circ Physiol ; 290(2): H525-30, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16183723

RESUMO

Based on the role of tumor necrosis factor-alpha (TNF-alpha) in ischemic preconditioning (IPC) and the age-associated loss of both TNF-alpha-induced platelet-derived growth factor-AB (PDGF-AB)-mediated cardioprotection and IPC-mediated cardioprotection, we hypothesized that targeting of PDGF-AB-based pathways would restore cardioprotection by IPC in the aging heart. To study this, IPC was induced in 4- and 24-mo-old F344 rats. Sections of young hearts isolated 1 day post-IPC revealed increased TNF-alpha compared with controls. In old rats, TNF-alpha was higher at baseline than IPC young rats and was not significantly altered after IPC. Treatment of old rats with PDGF-AB with vascular endothelial growth factor and angiopoietin-2 (a combination termed PVA), but not PDGF-AB alone, at the time of IPC decreased TNF-alpha. In addition, when compared with young hearts, IPC induced greater apoptosis in the old hearts, which was decreased with PVA treatment but was markedly increased with PDGF-AB. To test the significance of these findings, additional rats underwent permanent coronary ligation 1 day post-IPC. IPC was cardioprotective in young rats [14 days postmyocardial infarction (MI), fractional shortening 29 +/- 6% vs. control MI 17 +/- 4%, P < 0.05; Masson's trichrome stain MI size: 13 +/- 2% vs. control MI 17 +/- 4% left ventricular area (LVA); P < 0.05]. In old rats, however, IPC reduced the post-MI 14-day survival (33% vs. controls 67%; P < 0.05). Treatment of IPC-aging rats with PVA, but not PDGF-AB-alone, reversed IPC-induced mortality (PVA-IPC-MI survival, 88%; PDGF-AB-IPC-MI, 14%) and reduced myocardial injury (fractional shortening: PVA-IPC, 31 +/- 1% vs. control MI, 21 +/- 6%, P < 0.05; MI size: PVA-IPC, 12 +/- 2% vs. control MI, 18 +/- 3% LVA, P < 0.05) and thus demonstrated that PDGF-AB-based pathways can reverse the senescent impairment in IPC-mediated cardioprotection.


Assuntos
Envelhecimento , Cardiotônicos/farmacologia , Coração/efeitos dos fármacos , Coração/fisiopatologia , Precondicionamento Isquêmico Miocárdico , Fator de Crescimento Derivado de Plaquetas/farmacologia , Angiopoietina-2/farmacologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Combinação de Medicamentos , Ecocardiografia Transesofagiana , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Ratos , Ratos Endogâmicos F344 , Análise de Sobrevida , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
10.
Coron Artery Dis ; 15(1): 59-64, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15201622

RESUMO

OBJECTIVES: The translation of cardioprotective therapies for myocardial infarction requires a preclinical demonstration of improved cardiovascular function following acute coronary occlusion. We previously showed that pretreatment of rodent hearts with platelet-derived growth factor (PDGF) promotes angiogenesis and decreases the extent of myocardial injury measured by histology. The present study aimed to determine the correlation of these histological findings with noninvasive measures of improvement in cardiac function. METHODS: Rats were treated with intramyocardial injections of PDGF (100 ng) or phosphate buffer solution (PBS) (n = 6 per group) 24 h prior to acute, permanent ligation of the left anterior descending artery and the extent of myocardial injury was assessed by Masson's trichrome staining 14 days later. To assess the physiological effects of PDGF pretreatment after coronary occlusion, cardiac function was assessed noninvasively by electrocardiography, exercise testing and echocardiography and correlated with direct histological measures. RESULTS: Physiological studies demonstrated that PDGF resulted in lower ST-segment elevation at the time of coronary occlusion (0.12 +/- 0.02 mV above baseline) than in PBS control rats (0.35 +/- 0.05 mV; P < 0.05). Exercise testing 14 days after coronary occlusion revealed that PDGF pretreatment resulted in faster maximal exercise speeds (28.54 +/- 3.98 m/min) than in control rats (24.98 +/- 3.13 m/min; P < 0.05). Echocardiography also revealed that the left ventricular factional shortening in the PDGF-pretreated rats was significantly greater (18.47 +/- 12.21%) than in control animals (4.91 +/- 7.21%; P<0.05). CONCLUSIONS: These studies demonstrate that PDGF pretreatment improves cardiac function following acute coronary occlusion. Strategies based on the cardioprotective actions of PDGF may provide a significant advance in the treatment of myocardial infarction.


Assuntos
Coração/fisiologia , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/fisiopatologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Animais , Cardiotônicos/farmacologia , Doença das Coronárias/diagnóstico por imagem , Doença das Coronárias/tratamento farmacológico , Doença das Coronárias/fisiopatologia , Modelos Animais de Doenças , Ecocardiografia , Eletrocardiografia , Técnicas Eletrofisiológicas Cardíacas , Teste de Esforço , Coração/efeitos dos fármacos , Modelos Cardiovasculares , Infarto do Miocárdio/diagnóstico por imagem , Ratos , Estatística como Assunto
11.
Physiol Genomics ; 18(3): 255-60, 2004 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-15187201

RESUMO

Age-associated alterations in the actions of tumor necrosis factor-alpha (TNFalpha) in the heart with impaired cardioprotective pathways and enhanced apoptotic induction may contribute to the increased severity of cardiovascular pathology in older persons. To identify the molecular events mediating these changes in the microvasculature of the aging rodent heart, the biochemical properties of in vivo phage-display cyclic peptide cardiac biopanning were studied. Analysis of individual amino acid positions revealed that the center of the peptide motif (amino acid position 4) had a significantly higher frequency of aromatic amino acid side chains in phage homing to the old hearts compared with young controls (18 mo old, 11% vs. 3 mo old, 3%, P < 0.05). This subset of phage motifs revealed an age-associated homology with oxidoreductase enzymes (homology: 18 mo, 7/7; 3 mo, 0/2), suggesting the substrates and/or binding sites of these enzymes are increased in the aging hearts. Immunostaining for the oxidoreductase substrate 4-hydroxy-2-nonenal (HNE), a cardiotoxic lipid peroxidation product, demonstrated a twofold higher density of HNE(+) cells in PBS-treated hearts of old mice (18 mo) compared with young controls (3 mo) (18 mo, 3.2 +/- 2.8 vs. 3 mo, 1.0 +/- 0.9 cells/HPF, P < 0.05). Moreover, intracardiac injection of TNFalpha resulted in a significantly greater increase in HNE staining in the old hearts (18 mo, 16.9 +/- 13.8 vs. 3 mo, 9.1 +/- 6.0 cells/HPF, P < 0.05). Overall, these studies demonstrate that aging-associated alterations in TNFalpha-mediated pathways with induction of reactive oxidative species and changes in vascular surface binding sites may contribute mechanistically to the increased cardiovascular pathology of the aging heart.


Assuntos
Envelhecimento/fisiologia , Estresse Oxidativo/fisiologia , Biblioteca de Peptídeos , Fator de Necrose Tumoral alfa/fisiologia , Motivos de Aminoácidos , Animais , Imuno-Histoquímica/métodos , Ponto Isoelétrico , Peroxidação de Lipídeos , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/química , Miocárdio/patologia , Peptídeos/química , Peptídeos/imunologia , Espécies Reativas de Oxigênio/química , Espécies Reativas de Oxigênio/metabolismo , Homologia de Sequência de Aminoácidos
12.
J Exp Med ; 199(6): 797-804, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15007092

RESUMO

Pretreatment of rodent hearts with platelet-derived growth factor (PDGF)-AB decreases myocardial injury after coronary occlusion. However, PDGF-AB cardioprotection is diminished in older animals, suggesting that downstream elements mediating and/or synergizing the actions of PDGF-AB may be limited in aging cardiac vasculature. In vitro PDGF-AB induced vascular endothelial growth factor (VEGF) and angiopoietin (Ang)-2 expression in 4-mo-old rat cardiac endothelial cells, but not in 24-mo-old heart cells. In vivo injection of young hearts with PDGF-AB increased densities of microvessels staining for VEGF and its receptor, Flk-1, and Ang-2 and its receptor, Tie-2, as well as PDGF receptor (PDGFR)-alpha. In older hearts, PDGF-AB-mediated induction was primarily limited to PDGFR-alpha. Studies in a murine cardiac transplantation model demonstrated that synergist interactions of PDGF-AB plus VEGF plus Ang-2 (PVA) provided an immediate restoration of senescent cardiac vascular function. Moreover, PVA injection in young rat hearts, but not PDGF-AB alone or other cytokine combinations, at the time of coronary occlusion suppressed acute myocardial cell death by >50%. However, PVA also reduced the extent of myocardial infarction with an age-associated cardioprotective benefit (4-mo-old with 45% reduction vs. 24-mo-old with 24%; P < 0.05). These studies showed that synergistic cytokine pathways augmenting the actions of PDGF-AB are limited in older hearts, suggesting that strategies based on these interactions may provide age-dependent clinical cardiovascular benefit.


Assuntos
Envelhecimento/metabolismo , Angiopoietina-2/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/metabolismo , Fator de Crescimento Derivado de Plaquetas/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Animais , Cardiotônicos/uso terapêutico , Células Cultivadas , Doença das Coronárias/complicações , Primers do DNA , Sinergismo Farmacológico , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/complicações , Ratos , Ratos Endogâmicos F344 , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor TIE-2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
13.
Circ Res ; 94(5): E39-45, 2004 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-14963008

RESUMO

The directed generation of cardiac myocytes from endogenous stem cells offers the potential for novel therapies for cardiovascular disease. To facilitate the development of such approaches, we sought to identify and exploit the pathways directing the generation of cardiac myocytes from adult rodent bone marrow cells (BMCs). In vitro cultures supporting the spontaneous generation of functional cardiac myocytes from murine BMCs demonstrated induced expression of platelet-derived growth factor (PDGF)-A and -B isoforms with alpha- and beta-myosin heavy chains as well as connexin43. Supplementation of PDGF-AB speeded the kinetics of myocyte development in culture by 2-fold. In a rat heart, myocardial infarction pretreatment model PDGF-AB also promoted the derivation of cardiac myocytes from BMCs, resulting in a significantly greater number of islands of cardiac myocyte bundles within the myocardial infarction scar compared with other treatment groups. However, gap junctions were detected only between the cardiac myocytes receiving BMCs alone, but not BMCs injected with PDGF-AB. Echocardiography and exercise testing revealed that the functional improvement of hearts treated with the combination of BMCs and PDGF-AB was no greater than with injections of BMCs or PDGF-AB alone. These studies demonstrated that PDGF-AB enhances the generation of BMC-derived cardiac myocytes in rodent hearts, but suggest that alterations in cellular patterning may limit the functional benefit from the combined injection of PDGF-AB and BMCs. Strategies based on the synergistic interactions of PDGF-AB and endogenous stem cells will need to maintain cellular patterning in order to promote the restoration of cardiac function after acute coronary occlusion.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Miócitos Cardíacos/citologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Animais , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Diferenciação Celular/efeitos dos fármacos , Sistemas Computacionais , Teste de Esforço , Fator 2 de Crescimento de Fibroblastos/farmacologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia de Vídeo , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/farmacologia , Fator de Crescimento Derivado de Plaquetas/uso terapêutico , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos F344 , Ultrassonografia , Fator A de Crescimento do Endotélio Vascular/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...