Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Angiogenesis ; 26(1): 63-76, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35947328

RESUMO

Progression of atherosclerosis is associated with a maladaptive form of angiogenesis which contributes to intraplaque hemorrhage and plaque disruption. Hypoxia has been implicated in mechanisms of angiogenic neovessel fragility and atherosclerotic plaque destabilization. We used ex vivo and in vivo models to characterize the effect of oxygen (O2) on the formation, stability and tendency to bleed of human plaque-induced neovessels. Plaque explants potently stimulated the ex vivo angiogenic response of rat aortic rings at atmospheric O2 levels. Severe hypoxia (1% O2) inhibited plaque-induced angiogenesis and pericyte recruitment causing neovessel breakdown, whereas increasing O2 levels dose dependently enhanced pericyte numbers and neovessel stability. Plaque fragments implanted subcutaneously with or without aortic rings in SCID mice stimulated the host angiogenic response with plaques causing minimal or no hemorrhages and plaques co-implanted with aortic rings causing marked hemorrhages. Plaque/aortic ring-induced hemorrhages were reduced in mice exposed to moderate hyperoxia (50% O2). Hyperoxia downregulated expression of the hypoxia-sensitive genes Ca9, Ca12 and VegfA and increased influx into implants of mesenchymal cells reactive for the pericyte marker NG2. In both ex vivo and in vivo models, O2 promoted expression of vasostabilizing genes required for pericyte recruitment (Angpt1, Pdgfb), basement membrane assembly (Col4A1), and tight junction formation (Cldn5 and/or Ocln). Our results suggest that formation of neovessels that are stable, pericyte-coated, and resistant to bleeding requires adequate tissue oxygenation. Understanding the mechanisms by which O2 stabilizes neovessels and mitigates neovessel bleeding may lead to new therapies for the prevention of atherosclerosis complications.


Assuntos
Aterosclerose , Hiperóxia , Placa Aterosclerótica , Humanos , Ratos , Camundongos , Animais , Neovascularização Patológica , Camundongos SCID , Hemorragia , Hipóxia
2.
Diabetologia ; 65(10): 1687-1700, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35871651

RESUMO

AIMS/HYPOTHESIS: The islet vasculature, including its constituent islet endothelial cells, is a key contributor to the microenvironment necessary for normal beta cell health and function. In type 2 diabetes, islet amyloid polypeptide (IAPP) aggregates, forming amyloid deposits that accumulate between beta cells and islet capillaries. This process is known to be toxic to beta cells but its impact on the islet vasculature has not previously been studied. Here, we report the first characterisation of the effects of IAPP aggregation on islet endothelial cells/capillaries using cell-based and animal models. METHODS: Primary and immortalised islet endothelial cells were treated with amyloidogenic human IAPP (hIAPP) alone or in the presence of the amyloid blocker Congo Red or the Toll-like receptor (TLR) 2/4 antagonist OxPAPc. Cell viability was determined0 along with mRNA and protein levels of inflammatory markers. Islet capillary abundance, morphology and pericyte coverage were determined in pancreases from transgenic mice with beta cell expression of hIAPP using conventional and confocal microscopy. RESULTS: Aggregated hIAPP decreased endothelial cell viability in immortalised and primary islet endothelial cells (by 78% and 60%, respectively) and significantly increased expression of inflammatory markers Il6, Vcam1 and Edn1 mRNA relative to vehicle treatment in both cell types (p<0.05; n=4). Both cytotoxicity and the proinflammatory response were ameliorated by Congo Red (p<0.05; n=4); whereas TLR2/4-inhibition blocked inflammatory gene expression (p<0.05; n=6) without improving viability. Islets from high-fat-diet-fed amyloid-laden hIAPP transgenic mice also exhibited significantly increased expression of most markers of endothelial inflammation (p<0.05; n=5) along with decreased capillary density compared with non-transgenic littermates fed the same diet (p<0.01). Moreover, a 16% increase in capillary diameter was observed in amyloid-adjacent capillaries (p<0.01), accompanied by a doubling in pericyte structures positive for neuron-glial antigen 2 (p<0.001). CONCLUSIONS/INTERPRETATION: Islet endothelial cells are susceptible to hIAPP-induced cytotoxicity and exhibit a TLR2/4-dependent proinflammatory response to aggregated hIAPP. Additionally, we observed amyloid-selective effects that decreased islet capillary density, accompanied by increased capillary diameter and increased pericyte number. Together, these data demonstrate that the islet vasculature is a target of the cytotoxic and proinflammatory effects of aggregated hIAPP that likely contribute to the detrimental effects of hIAPP aggregation on beta cell function and survival in type 2 diabetes.


Assuntos
Amiloidose , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Ilhotas Pancreáticas , Amiloide/metabolismo , Amiloidose/metabolismo , Animais , Vermelho Congo/metabolismo , Vermelho Congo/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Células Endoteliais/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Interleucina-6/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Transgênicos , RNA Mensageiro/metabolismo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo
3.
Angiogenesis ; 22(3): 421-431, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30968256

RESUMO

Progression of atherosclerotic plaques into life-threatening lesions is associated with angiogenesis which contributes to intraplaque hemorrhages and plaque instability. The lack of adequate models for the study of human plaque-induced angiogenesis has limited progress in this field. We describe here a novel ex vivo model which fills this gap. Plaques obtained from 15 patients who underwent endarterectomy procedures were co-cultured in collagen gels with rat aorta rings which served as read-out of human plaque angiogenic activity. The majority of plaque fragments markedly stimulated angiogenic sprouting from the aortic rings while concurrently promoting the outgrowth of resident macrophages from the aortic adventitia. This stimulatory activity correlated with the presence of intraplaque macrophages. Proteomic analysis of plaque secretomes revealed heterogeneity of macrophage-stimulatory cytokine and angiogenic factor production by different plaques. VEGF was identified in some of the plaque secretomes. Antibody-mediated blockade of VEGF had significant but transient inhibitory effect on angiogenesis, which suggested redundancy of plaque-derived angiogenic stimuli. Pharmacologic ablation of adventitial macrophages permanently impaired the angiogenic response of aortic rings to plaque stimuli. Our results show that human plaque-induced angiogenesis can be reproduced ex vivo using rat aortic rings as read-out of plaque angiogenic activity. This model can be used to identify key cellular and molecular mechanisms responsible for the neovascularization of human plaques.


Assuntos
Aterosclerose/patologia , Bioensaio/métodos , Neovascularização Patológica/patologia , Placa Aterosclerótica/patologia , Túnica Adventícia/patologia , Idoso , Indutores da Angiogênese/metabolismo , Animais , Aorta , Polaridade Celular , Quimiocinas/metabolismo , Humanos , Macrófagos/patologia , Masculino , Pessoa de Meia-Idade , Ratos Endogâmicos F344 , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Angiogenesis ; 21(3): 425-532, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29766399

RESUMO

The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.


Assuntos
Bioensaio/métodos , Neoplasias , Neovascularização Patológica , Animais , Bioensaio/instrumentação , Guias como Assunto , Humanos , Camundongos , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia
5.
Angiogenesis ; 19(2): 133-46, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26748649

RESUMO

This study was designed to investigate how changes in O2 levels affected angiogenesis in vascular organ culture. Although hypoxia is a potent inducer of angiogenesis, aortic rings cultured in collagen paradoxically failed to produce an angiogenic response in 1-4 % O2. Additionally, aortic neovessels preformed in atmospheric O2 lost pericytes and regressed at a faster rate than control when exposed to hypoxia. Aortic explants remained viable in hypoxia and produced an angiogenic response when returned to atmospheric O2. Hypoxic aortic rings were unresponsive to VEGF, while increased oxygenation of the system dose-dependently enhanced VEGF-induced angiogenesis. Hypoxia-induced refractoriness to angiogenic stimulation was not restricted to the aorta because similar results were obtained with vena cava explants or isolated endothelial cells. Unlike endothelial cells, aorta-derived mural cells were unaffected by hypoxia. Hypoxia downregulated expression in aortic explants of key signaling molecules including VEGFR2, NRP1 and Prkc-beta while upregulating expression of VEGFR1. Medium conditioned by hypoxic cultures exhibited angiostatic and anti-VEGF activities likely mediated by sVEGFr1. Hypoxia reduced expression of VEGFR1 and VEGFR2 in endothelial cells while upregulating VEGFR1 in macrophages and VEGF in both macrophages and mural cells. Thus, changes in O2 levels profoundly affect the endothelial response to angiogenic stimuli. These results suggest that hypoxia-induced angiogenesis is fine-tuned by complex regulatory mechanisms involving not only production of angiogenic factors including VEGF but also differential regulation of VEGFR expression in different cell types and production of inhibitors of VEGF function such as sVEGFR1.


Assuntos
Vasos Sanguíneos/fisiologia , Neovascularização Fisiológica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Antioxidantes/farmacologia , Aorta/efeitos dos fármacos , Becaplermina , Vasos Sanguíneos/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Separação Celular , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Oxigênio/farmacologia , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Proteínas Proto-Oncogênicas c-sis/farmacologia , Ratos Endogâmicos F344 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Técnicas de Cultura de Tecidos , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
Blood ; 121(13): 2574-8, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23303818

RESUMO

The treatment of festering wounds is one of the most important aspects of medical care. Macrophages are important components of wound repair, both in fending off infection and in coordinating tissue repair. Here we show that macrophages use a Wnt-Calcineurin-Flt1 signaling pathway to suppress wound vasculature and delay repair. Conditional mutants deficient in both Wntless/GPR177, the secretory transporter of Wnt ligands, and CNB1, the essential component of the nuclear factor of activated T cells dephosporylation complex, displayed enhanced angiogenesis and accelerated repair. Furthermore, in myeloid-like cells, we show that noncanonical Wnt activates Flt1, a naturally occurring inhibitor of vascular endothelial growth factor-A-mediated angiogenesis, but only when calcineurin function is intact. Then, as expected, conditional deletion of Flt1 in macrophages resulted in enhanced wound angiogenesis and repair. These results are consistent with the published link between enhanced angiogenesis and enhanced repair, and establish novel therapeutic approaches for treatment of wounds.


Assuntos
Calcineurina/metabolismo , Macrófagos/metabolismo , Neovascularização Fisiológica , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Via de Sinalização Wnt/fisiologia , Cicatrização , Animais , Calcineurina/genética , Células Cultivadas , Derme/irrigação sanguínea , Derme/lesões , Derme/metabolismo , Macrófagos/fisiologia , Camundongos , Camundongos Transgênicos , Neovascularização Fisiológica/genética , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteínas Wnt/fisiologia , Via de Sinalização Wnt/genética , Proteína Wnt-5a , Cicatrização/genética , Cicatrização/fisiologia
7.
Angiogenesis ; 17(1): 147-61, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24091496

RESUMO

The angiogenic response to injury can be studied by culturing rat or mouse aortic explants in collagen gels. Gene expression studies show that aortic angiogenesis is preceded by an immune reaction with overexpression of Toll-like receptors (TLRs) and TLR-inducible genes. TLR1, 3, and 6 are transiently upregulated at 24 h whereas TLR2, 4, and 8 expression peaks at 24 h but remains elevated during angiogenesis and vascular regression. Expression of TLR5, 7 and 9 steadily increases over time and is highest during vascular regression. Studies with isolated cells show that TLRs are expressed at higher levels in aortic macrophages compared to endothelial or mural cells with the exception of TLR2 and TLR9 which are more abundant in the aortic endothelium. LPS and other TLR ligands dose dependently stimulate angiogenesis and vascular endothelial growth factor production. TLR9 ligands also influence the behavior of nonendothelial cell types by blocking mural cell recruitment and inducing formation of multinucleated giant cells by macrophages. TLR9-induced mural cell depletion is associated with reduced expression of the mural cell recruiting factor PDGFB. The spontaneous angiogenic response of the aortic rings to injury is reduced in cultures from mice deficient in myeloid differentiation primary response 88 (MyD88), a key adapter molecule of TLRs, and following treatment with an inhibitor of the NFκB pathway. These results suggest that the TLR system participates in the angiogenic response of the vessel wall to injury and may play an important role in the regulation of inflammatory angiogenesis in reactive and pathologic processes.


Assuntos
Túnica Adventícia/metabolismo , Aorta/metabolismo , Diferenciação Celular/fisiologia , Macrófagos/metabolismo , Neovascularização Fisiológica/fisiologia , Receptores Toll-Like/biossíntese , Túnica Adventícia/citologia , Animais , Aorta/citologia , Diferenciação Celular/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Macrófagos/citologia , Masculino , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis/genética , Proteínas Proto-Oncogênicas c-sis/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptores Toll-Like/genética , Túnica Íntima/citologia , Túnica Íntima/metabolismo
8.
Arterioscler Thromb Vasc Biol ; 31(5): 1151-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21372301

RESUMO

OBJECTIVE: The goal of this study was to define the role of tumor necrosis factor-α (TNFα) in the cascade of gene activation that regulates aortic angiogenesis in response to injury. METHODS AND RESULTS: Angiogenesis was studied by culturing rat or mouse aortic rings in collagen gels. Gene expression was evaluated by quantitative reverse transcription-polymerase chain reaction, microarray analysis, immunocytochemistry, and ELISA. TNFα gene disruption and recombinant TNFα or blocking antibodies against vascular endothelial growth factor (VEGF) or TNF receptors were used to investigate TNFα-mediated angiogenic mechanisms. Resident aortic macrophages were depleted with liposomal clodronate. Angiogenesis was preceded by overexpression of TNFα and TNFα-inducible genes. Studies with isolated cells showed that macrophages were the main source of TNFα. Angiogenesis, VEGF production, and macrophage outgrowth were impaired by TNFα gene disruption and promoted by exogenous TNFα. Antibody-mediated inhibition of TNF receptor 1 significantly inhibited angiogenesis. The proangiogenic effect of TNFα was suppressed by blocking VEGF or by ablating aortic macrophages. Exogenous TNFα, however, maintained a limited proangiogenic capacity in the absence of macrophages and macrophage-mediated VEGF production. CONCLUSIONS: Overexpression of TNFα is required for optimal VEGF production and angiogenesis in response to injury. This TNFα/VEGF-mediated angiogenic pathway requires macrophages. The residual capacity of TNFα to stimulate angiogenesis in macrophage-depleted aortic cultures implies the existence of a VEGF-independent alternate pathway of TNFα-induced angiogenesis.


Assuntos
Aorta Torácica/imunologia , Macrófagos/imunologia , Neovascularização Fisiológica , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Lesões do Sistema Vascular/imunologia , Animais , Anticorpos/farmacologia , Aorta Torácica/lesões , Aorta Torácica/fisiopatologia , Western Blotting , Células Cultivadas , Ácido Clodrônico/farmacologia , Ensaio de Imunoadsorção Enzimática , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Imuno-Histoquímica , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Tempo , Técnicas de Cultura de Tecidos , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/fisiopatologia
9.
J Endocrinol ; 248(2): 95-106, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33337344

RESUMO

Islet endothelial cells produce paracrine factors important for islet beta-cell function and survival. Under conditions of type 2 diabetes, islet endothelial cells exhibit a dysfunctional phenotype including increased expression of genes involved in cellular adhesion and inflammation. We sought to determine whether treatment of hyperglycemia with the sodium glucose co-transporter 2 inhibitor empagliflozin, either alone or in combination with metformin, would improve markers of endothelial cell function in islets, assessed ex vivo, and if such an improvement is associated with improved insulin secretion in a mouse model of diabetes in vivo. For these studies, db/db diabetic mice and non-diabetic littermate controls were treated for 6 weeks with empagliflozin or metformin, either alone or in combination. For each treatment group, expression of genes indicative of islet endothelial dysfunction was quantified. Islet endothelial and beta-cell area was assessed by morphometry of immunochemically stained pancreas sections. Measurements of plasma glucose and insulin secretion during an intravenous glucose tolerance test were performed on vehicle and drug treated diabetic animals. We found that expression of endothelial dysfunction marker genes is markedly increased in diabetic mice. Treatment with either empagliflozin or metformin lowered expression of the dysfunction marker genes ex vivo, which correlated with improved glycemic control, and increased insulin release in vivo. Empagliflozin treatment was more effective than metformin alone, with a combination of the two drugs demonstrating the greatest effects. Improving islet endothelial function through strategies such as empagliflozin/metformin treatment may provide an effective approach for improving insulin release in human type 2 diabetes.


Assuntos
Compostos Benzidrílicos/uso terapêutico , Diabetes Mellitus Experimental/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Glucosídeos/uso terapêutico , Secreção de Insulina/efeitos dos fármacos , Inibidores do Transportador 2 de Sódio-Glicose/uso terapêutico , Animais , Compostos Benzidrílicos/farmacologia , Glicemia/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Quimioterapia Combinada , Glucosídeos/farmacologia , Hipoglicemiantes/uso terapêutico , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Metformina/uso terapêutico , Camundongos , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia
10.
Angiogenesis ; 13(3): 219-26, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20571857

RESUMO

Rings of rat or mouse aorta embedded in collagen gels produce angiogenic outgrowths in response to the injury of the dissection procedure. Aortic outgrowths are composed of branching endothelial tubes and surrounding mural cells. Mural cells emerge following endothelial sprouting and gradually increase during the maturation of the neovessels. Treatment of aortic cultures with angiopoietin-1 (Ang-1), an angiogenic factor implicated in vascular maturation and remodeling, stimulates the mural cell recruitment process. Ang-1 induces expression of many cytokines and chemokines including monocyte chemotactic protein-1 (MCP-1). Inhibition of p38 MAP kinase, a signaling molecule required for mural cell recruitment, blocks Ang1-induced MCP-1 expression. Recombinant MCP-1 dose-dependently increases mural cell number while an anti-MCP-1 blocking antibody reduces it. In addition, antibody mediated neutralization of MCP-1 abrogates the stimulatory effect of Ang-1 on mural cell recruitment. Aortic rings from genetically modified mice deficient in MCP-1 or its receptor CCR2 have fewer mural cells than controls. MCP-1 deficiency also impairs the mural cell recruitment activity of Ang-1. Our studies indicate that spontaneous and Ang1-induced mural cell recruitment in the aortic ring of model of angiogenesis are in part mediated by MCP-1. These results implicate MCP-1 as one of the mediators of mural cell recruitment in the aortic ring model, and suggest that chemokine pathways may contribute to the assembly of the vessel wall during the angiogenesis response to injury.


Assuntos
Aorta/citologia , Movimento Celular , Quimiocina CCL2/metabolismo , Modelos Biológicos , Neovascularização Fisiológica , Angiopoietina-1/farmacologia , Animais , Contagem de Células , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/antagonistas & inibidores , Quimiocina CCL2/deficiência , Humanos , Imidazóis/farmacologia , Técnicas In Vitro , Camundongos , Mutação/genética , Neovascularização Fisiológica/efeitos dos fármacos , Piridinas/farmacologia , Ratos , Receptores CCR2/genética , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
J Immunol ; 181(8): 5711-9, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18832730

RESUMO

The purpose of this study was to define early events during the angiogenic response of the aortic wall to injury. Rat aortic rings produced neovessels in collagen culture but lost this capacity over time. These quiescent rings responded to vascular endothelial growth factor but not to a mixture of macrophage-stimulatory cytokines and chemokines that was angiogenically active on fresh rings. Analysis of cytokine receptor expression revealed selective loss in quiescent rings of the proangiogenic chemokine receptor CXCR2, which was expressed predominantly in aortic macrophages. Pharmacologic inhibition of CXCR2 impaired angiogenesis from fresh rings but had no effect on vascular endothelial growth factor-induced angiogenesis from quiescent explants. Angiogenesis was also impaired in cultures of aortic rings from CXCR2-deficient mice. Reduced CXCR2 expression in quiescent rat aortic rings correlated with marked macrophage depletion. Pharmacologic ablation of macrophages from aortic explants blocked formation of neovessels in vitro and reduced aortic ring-induced angiogenesis in vivo. The angiogenic response of macrophage-depleted rings was completely restored by adding exogenous macrophages. Moreover, angiogenesis from fresh rings was promoted by macrophage CSF (CSF-1) and inhibited with anti-CSF-1 Ab. Thus, aortic angiogenic sprouting following injury is strongly influenced by conditions that modulate resident macrophage numbers and function.


Assuntos
Aorta/imunologia , Aorta/lesões , Regulação da Expressão Gênica/imunologia , Macrófagos/imunologia , Neovascularização Fisiológica/imunologia , Receptores de Interleucina-8B/imunologia , Animais , Anticorpos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Fator Estimulador de Colônias de Macrófagos/imunologia , Masculino , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Ratos , Ratos Endogâmicos F344 , Fator A de Crescimento do Endotélio Vascular/imunologia
12.
J Pharmacol Exp Ther ; 328(2): 378-89, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18988770

RESUMO

The proteolytic activation by thrombin of the proteinase-activated receptor 1 unveils the tethered peptide ligand and cleaves a 41-amino acid peptide. In this report, we show that this peptide, which we have designated as "parstatin," is a potent inhibitor of angiogenesis. Synthesized parstatin suppressed both the basic angiogenesis and that stimulated by basic fibroblast growth factor and vascular endothelial growth factor in the chick embryo model in vivo and in the rat aortic ring assay. Parstatin also abrogated endothelial cell migration and capillary-like network formation on the Matrigel and fibrin angiogenesis models in vitro. Treatment of endothelial cells with parstatin resulted in inhibition of cell growth by inhibiting the phosphorylation of extracellular signal-regulated kinases in a specific and reversible fashion and by promoting cell cycle arrest and apoptosis through a mechanism involving activation of caspases. We have shown that parstatin acts as a cell-penetrating peptide, exerting its biological effects intracellularly. The uptake into cells and the inhibitory activity were dependent on parstatin hydrophobic region. These results support the notion that parstatin may represent an important negative regulator of angiogenesis with possible therapeutic applications.


Assuntos
Inibidores da Angiogênese/farmacologia , Movimento Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptor PAR-1/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Capilares/efeitos dos fármacos , Capilares/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Humanos , Peptídeos/farmacologia , Ratos , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Biomaterials ; 141: 314-329, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28711779

RESUMO

Loss of the microvascular (MV) network results in tissue ischemia, loss of tissue function, and is a hallmark of chronic diseases. The incorporation of a functional vascular network with that of the host remains a challenge to utilizing engineered tissues in clinically relevant therapies. We showed that vascular-bed-specific endothelial cells (ECs) exhibit differing angiogenic capacities, with kidney microvascular endothelial cells (MVECs) being the most deficient, and sought to explore the underlying mechanism. Constitutive activation of the phosphatase PTEN in kidney MVECs resulted in impaired PI3K/AKT activity in response to vascular endothelial growth factor (VEGF). Suppression of PTEN in vivo resulted in microvascular regeneration, but was insufficient to improve tissue function. Promoter analysis of the differentially regulated genes in KMVECs suggests that the transcription factor FOXO1 is highly active and RNAseq analysis revealed that hyperactive FOXO1 inhibits VEGF-Notch-dependent tip-cell formation by direct and indirect inhibition of DLL4 expression in response to VEGF. Inhibition of FOXO1 enhanced angiogenesis in human bio-engineered capillaries, and resulted in microvascular regeneration and improved function in mouse models of injury-repair.


Assuntos
Proteína Forkhead Box O1/metabolismo , Rim/irrigação sanguínea , Rim/fisiopatologia , Microvasos/fisiopatologia , Neovascularização Fisiológica , Adulto , Animais , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Rim/lesões , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/metabolismo , Microvasos/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
14.
Physiol Genomics ; 27(1): 20-8, 2006 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-17018690

RESUMO

The purpose of this study was to identify novel transcriptional events occurring in the aortic wall before angiogenesis. We used a defined tissue culture system that takes advantage of the capacity of rat aortic rings to generate neovessels ex vivo in response to angiogenic factor stimulation. Total RNA isolated from aortic rings 18 h posttreatment with angiopoietin (Ang)-1 or vascular endothelial growth factor (VEGF) was used to probe oligonucleotide microarrays. Many genes were up- or downregulated by either Ang-1 or VEGF, with a subset being affected by treatment with both growth factors. Grouping of genes by biological function revealed that Ang-1 and VEGF both upregulated a host of immune-related genes including many inflammatory cytokines. A mixture of the Ang-1- and VEGF-induced cytokines stimulated the spontaneous angiogenic response of aortic rings and was synergistic with a low dose of recombinant VEGF. This effect was associated with enhanced recruitment of adventitial macrophages and dendritic cells in the angiogenic outgrowths. Thus Ang-1 and VEGF activate the innate immune system of the vessel wall, stimulating the production of proangiogenic inflammatory cytokines before the emergence of neovessels. This hitherto unreported feature of the angiogenic response might represent an important early component of the cellular and molecular cascade responsible for the angiogenic response of the aortic wall.


Assuntos
Angiopoietina-1/farmacologia , Citocinas/biossíntese , Neovascularização Fisiológica/imunologia , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Aorta/crescimento & desenvolvimento , Aorta/imunologia , Citocinas/genética , Células Dendríticas/imunologia , Endotélio Vascular/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Perfilação da Expressão Gênica , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Neovascularização Fisiológica/genética , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Técnicas de Cultura de Tecidos
15.
Methods Mol Biol ; 1464: 63-72, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27858356

RESUMO

This chapter describes protocols developed in our laboratory to prepare and analyze angiogenic cultures of rat aorta. Rings of rat aorta cultured in collagen gels produce neovessel outgrowths which reproduce ex vivo key steps of the angiogenic process including endothelial migration, proliferation, proteolytic digestion of the extracellular matrix, capillary tube formation, pericyte recruitment, and vascular regression. The angiogenic response of aortic explants can be stimulated with growth factors or inhibited with anti-angiogenic molecules. Aortic ring cultures can also be used to study tumor angiogenesis. Protocols outlined in this chapter describe how this assay can be modified to investigate the angiogenic activity of cancer cells.


Assuntos
Aorta/citologia , Técnicas de Cocultura/métodos , Colágeno/metabolismo , Neovascularização Patológica/patologia , Inibidores da Angiogênese/farmacologia , Animais , Aorta/efeitos dos fármacos , Diferenciação Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Modelos Biológicos , Neovascularização Fisiológica , Ratos , Inclusão do Tecido
16.
Methods Mol Biol ; 1214: 255-64, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25468610

RESUMO

Protocols outlined in this chapter illustrate how to prepare and analyze angiogenic cultures of rat aorta. Aortic rings embedded in gels of extracellular matrix generate vascular outgrowths that can easily be monitored over time with inverted microscopy. The angiogenic response can be measured by counting vessels or with image analysis. Aortic ring cultures can be used to investigate molecular mechanisms of angiogenesis and test the efficacy of stimulators and inhibitors of the angiogenic process. As such this assay is an invaluable tool for both basic and applied angiogenesis research.


Assuntos
Aorta/fisiologia , Modelos Biológicos , Neovascularização Fisiológica , Técnicas de Cultura de Tecidos , Animais , Aorta/citologia , Colágeno/química , Matriz Extracelular/metabolismo , Imagem Molecular , Ratos
17.
Open J Clin Diagn ; 3(2): 37-51, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23956953

RESUMO

The gut hormone apelin is a major therapeutic focus for several diseases involving inflammation and aberrant cell growth. We investigated whether apelin-36 contained alternative bioactive peptides associated with normal physiology or disease. Amino acid sequence analysis of apelin-36 identified an amidation motif consistent with the formation of a secondary bioactive peptide (SCNH2). SCNH2 is proven to be mitogenic and chemotactic in normal/malignant cells and augments angiogenesis via a PTX-resistant/CT-X-sensitive G protein-coupled receptor (GPCR). Notably, SCNH2 is substantially more potent and sensitive than apelin-13 and vascular endothelial growth factor-A. Endogenous SCNH2 is highly expressed in human tumors and placenta and in mouse embryonic tissues. Our findings demonstrate that SCNH2 is a new apelinergic member with critical pluripotent roles in angiogenesis related diseases and embryogenesis via a non-APJ GPCR.

18.
PLoS One ; 7(8): e41387, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22916107

RESUMO

BACKGROUND: Tissues respond to injury by releasing acute phase reaction (APR) proteins which regulate inflammation and angiogenesis. Among the genes upregulated in wounded tissues are tumor necrosis factor-alpha (TNFα) and the acute phase reactant orosomucoid-1 (ORM1). ORM1 has been shown to modulate the response of immune cells to TNFα, but its role on injury- and TNFα-induced angiogenesis has not been investigated. This study was designed to characterize the role of ORM1 in the angiogenic response to injury and TNFα. METHODS AND RESULTS: Angiogenesis was studied with in vitro, ex vivo, and in vivo angiogenesis assays. Injured rat aortic rings cultured in collagen gels produced an angiogenic response driven by macrophage-derived TNFα. Microarray analysis and qRT-PCR showed that TNFα and ORM1 were upregulated prior to angiogenic sprouting. Exogenous ORM1 delayed the angiogenic response to injury and inhibited the proangiogenic effect of TNFα in cultures of aortic rings or isolated endothelial cells, but stimulated aortic angiogenesis over time while promoting VEGF production and activity. ORM1 inhibited injury- and TNFα-induced phosphorylation of MEK1/2 and p38 MAPK in aortic rings, but not of NFκB. This effect was injury/TNFα-specific since ORM1 did not inhibit VEGF-induced signaling, and cell-specific since ORM1 inhibited TNFα-induced phosphorylation of MEK1/2 and p38 MAPK in macrophages and endothelial cells, but not mural cells. Experiments with specific inhibitors demonstrated that the MEK/ERK pathway was required for angiogenesis. ORM1 inhibited angiogenesis in a subcutaneous in vivo assay of aortic ring-induced angiogenesis, but stimulated developmental angiogenesis in the chorioallantoic membrane (CAM) assay. CONCLUSION: ORM1 regulates injury-induced angiogenesis in a time- and context-dependent manner by sequentially dampening the initial TNFα-induced angiogenic response and promoting the downstream stimulation of the angiogenic process by VEGF. The context-dependent nature of ORM1 angioregulatory function is further demonstrated in the CAM assay where ORM1 stimulates developmental angiogenesis without exerting any inhibitory activity.


Assuntos
Reação de Fase Aguda , Neovascularização Patológica/fisiopatologia , Orosomucoide/fisiologia , Animais , Aorta/enzimologia , Aorta/fisiopatologia , Western Blotting , Colágeno/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Técnicas In Vitro , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Proteínas Quinases/metabolismo , Ratos , Fator de Necrose Tumoral alfa/fisiologia , Regulação para Cima/fisiologia
19.
Int J Dev Biol ; 55(4-5): 447-53, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21858770

RESUMO

The development of blood vessels during angiogenesis is the result of paracrine interactions between tube-forming endothelial cells and angiogenic factor-producing nonendothelial cells. This process can be reproduced and studied under chemically defined culture conditions by culturing vascular explants in three-dimensional gels of extracellular matrix. Rings of rat or mouse aorta cultured in collagen, fibrin or basement membrane gels produce angiogenic outgrowths composed of a mixed population of endothelial cells and nonendothelial cells. Aortic angiogenesis is regulated by endogenous angiogenic factors, inflammatory cytokines, chemokines, extracellular matrix molecules, and proteolytic enzymes produced by cells of the vessel wall in response to the injury of the dissection procedure. In this paper, we review how macrophages, mural cells and fibroblasts regulate different stages of the angiogenic process, from the formation of immature endothelial sprouts to the reabsorption of the neovessels. We also describe how aortic cultures can be used to study interactions between angiogenic outgrowths and nonvascular cell types such as bone marrow macrophages, platelets or cancer cells. Morphologic, genetic and functional studies of this model have provided invaluable information on how vessels form, mature, interact with nonvascular cell types, and are eventually reabsorbed. Further analysis of the paracrine cross-talk between aortic endothelial and nonendothelial cells is likely to provide new insights into the angiogenic process and its key mechanisms.


Assuntos
Aorta/citologia , Aorta/crescimento & desenvolvimento , Neovascularização Fisiológica/fisiologia , Comunicação Parácrina/fisiologia , Animais , Aorta/lesões , Plaquetas/fisiologia , Células Endoteliais/fisiologia , Fibroblastos/fisiologia , Macrófagos/fisiologia , Camundongos , Microvasos/citologia , Microvasos/crescimento & desenvolvimento , Neoplasias Experimentais/irrigação sanguínea , Neovascularização Patológica/fisiopatologia , Ratos , Técnicas de Cultura de Tecidos
20.
J Leukoc Biol ; 88(5): 1051-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20628067

RESUMO

Angiogenesis in the aortic ring model is preceded by activation of the immune system and impaired by ablation of adventitial macrophages. Treatment of aortic cultures with M-CSF induced extensive periaortic outgrowth of CD45(+) CD68(+) mononuclear cells with ultrastructural features of macrophages and DCs. Periaortic lysis of collagen caused many CD45(+) CD68(+) cells to attach to the bottom of the culture dish. Lifting the collagen gels left behind patches of CD45(+) CD68(+) cells, which focally organized into branching cords. These cells also expressed CD14, CD169, F4/80, and α-SMA but not CD31, vWF, desmin, or CD163. DNA synthesis studies showed that M-CSF-stimulated cells were actively proliferating. Aortic patch cells showed phagocytic properties and responded to IL-4 and GM-CSF by expressing MHC II, differentiating into DCs, and forming multinucleated giant cells. They also stimulated angiogenesis and VEGF production in aortic ring cultures. This study demonstrates that the rat aorta contains a distinct subset of immature immunocytes capable of proliferating, differentiating into macrophages and DCs, and stimulating angiogenesis. Isolation of these cells in patches from M-CSF-stimulated aortic rings provides a reproducible system to study the biology and angiogenic role of the resident immune system of the aortic wall.


Assuntos
Aorta/citologia , Fagócitos/citologia , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Aorta/efeitos dos fármacos , Aorta/imunologia , Aorta/ultraestrutura , Divisão Celular , Primers do DNA , Técnicas Imunoenzimáticas , Antígenos Comuns de Leucócito/imunologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Microscopia Confocal/métodos , Neovascularização Fisiológica , Fagócitos/efeitos dos fármacos , Fagocitose , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA