Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
2.
J Am Soc Nephrol ; 34(6): 969-987, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36787763

RESUMO

SIGNIFICANCE STATEMENT: Ischemia-reperfusion AKI (IR-AKI) is common and causes significant morbidity. Effective treatments are lacking. However, preclinical studies suggest that inhibition of angiopoietin-Tie2 vascular signaling promotes injury, whereas activation of Tie2 is protective. We show that kidney ischemia leads to increased levels of the endothelial-specific phosphatase vascular endothelial protein tyrosine phosphatase (VE-PTP; PTPRB), which inactivates Tie2. Activation of Tie2 through VE-PTP deletion, or delivery of a novel angiopoietin mimetic (Hepta-ANG1), abrogated IR-AKI in mice. Single-cell RNAseq analysis showed Tie2 activation promotes increased Entpd1 expression, downregulation of FOXO1 target genes in the kidney vasculature, and emergence of a new subpopulation of glomerular endothelial cells. Our data provide a molecular basis and identify a candidate therapeutic to improve endothelial integrity and kidney function after IR-AKI. BACKGROUND: Ischemia-reperfusion AKI (IR-AKI) is estimated to affect 2%-7% of all hospitalized patients. The significant morbidity and mortality associated with AKI indicates urgent need for effective treatments. Previous studies have shown activation of the vascular angiopoietin-Tie2 tyrosine kinase signaling pathway abrogates ischemia-reperfusion injury (IRI). We extended previous studies to (1) determine the molecular mechanism(s) underlying kidney injury and protection related to decreased or increased activation of Tie2, respectively, and (2) to test the hypothesis that deletion of the Tie2 inhibitory phosphatase vascular endothelial protein tyrosine phosphatase (VE-PTP) or injection of a new angiopoietin mimetic protects the kidney from IRI by common molecular mechanism(s). METHODS: Bilateral IR-AKI was performed in VE-PTP wild-type or knockout mice and in C57BL/6J mice treated with Hepta-ANG1 or vehicle. Histologic, immunostaining, and single-cell RNA sequencing analyses were performed. RESULTS: The phosphatase VE-PTP, which negatively regulates the angiopoietin-Tie2 pathway, was upregulated in kidney endothelial cells after IRI, and genetic deletion of VE-PTP in mice protected the kidney from IR-AKI. Injection of Hepta-ANG1 potently activated Tie2 and protected the mouse kidney from IRI. Single-cell RNAseq analysis of kidneys from Hepta-ANG1-treated and vehicle-treated mice identified endothelial-specific gene signatures and emergence of a new glomerular endothelial subpopulation associated with improved kidney function. Overlap was found between endothelial-specific genes upregulated by Hepta-ANG1 treatment and those downregulated in HUVECs with constitutive FOXO1 activation, including Entpd1 / ENTPD1 that modulates purinergic receptor signaling. CONCLUSIONS: Our data support a key role of the endothelium in the development of IR-AKI, introduce Hepta-ANG1 as a putative new therapeutic biologic, and report a model to explain how IRI reduces Tie2 signaling and how Tie2 activation protects the kidney. PODCAST: This article contains a podcast at https://dts.podtrac.com/redirect.mp3/www.asn-online.org/media/podcast/JASN/2023_05_23_JSN_Ang_EP23_052323.mp3.


Assuntos
Injúria Renal Aguda , Células Endoteliais , Camundongos , Animais , Células Endoteliais/metabolismo , Angiopoietinas/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Camundongos Endogâmicos C57BL , Endotélio/metabolismo , Rim/metabolismo , Transdução de Sinais , Receptor TIE-2/genética , Angiopoietina-1/uso terapêutico , Camundongos Knockout , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/metabolismo , Isquemia/complicações , Isquemia/metabolismo
3.
Int J Hyperthermia ; 39(1): 888-896, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35848416

RESUMO

OBJECTIVES: Angiogenesis occurs during tumor progression of hepatocellular carcinoma (HCC) after insufficient radiofrequency ablation (RFA). Arsenic trioxide (ATO) shows promising therapeutic potential in advanced HCC. Whether ATO regulates angiogenesis and can be used to prevent tumor progression in HCC after insufficient RFA is still unknown. METHODS: Insufficient RFA was simulated using a water bath. MTT assay and tube formation assay were used to evaluate the effects of ATO on viability and proangiogenic abilities of SMMC7721 and HepG2 cells after insufficient RFA in vitro. The molecular changes with the treatment of ATO were evaluated through Western blot. An ectopic nude mice model was used to evaluate the effect of ATO on the tumor of SMMC7721 cells in vivo after insufficient RFA. RESULTS: In this study, HepG2 and SMMC7721 cells after insufficient RFA (named HepG2-H and SMMC7721-H, respectively) showed higher proliferation than the untreated cells and promoted tube formation of endothelial cells in a paracrine manner. ATO eliminated the difference in proliferation between untreated and RFA-treated cells and suppressed angiogenesis induced by HCC cells after insufficient RFA through the Ang-1 (angiopoietin-1)/Ang-2 (angiopoietin-2)/Tie2 pathway. Hif-1α overexpression abolished the inhibitory effect of ATO on angiogenesis in HCC after insufficient RFA. ATO inhibited tumor growth and angiogenesis in HCC after insufficient RFA. CONCLUSIONS: Our results demonstrate that ATO blocks the paracrine signaling of Ang-1 and Ang-2 by inhibiting p-Akt/Hif-1α and further suppresses the angiogenesis of HCC after insufficient RFA.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Ablação por Radiofrequência , Angiopoietina-1/uso terapêutico , Angiopoietina-2/uso terapêutico , Animais , Trióxido de Arsênio/farmacologia , Trióxido de Arsênio/uso terapêutico , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Ablação por Radiofrequência/métodos
4.
Nephrol Dial Transplant ; 37(10): 1844-1856, 2022 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-35451482

RESUMO

BACKGROUND: Renal artery stenosis (RAS) is an important cause of chronic kidney disease and secondary hypertension. In animal models, renal ischemia leads to downregulation of growth factor expression and loss of intrarenal microcirculation. However, little is known about the sequelae of large-vessel occlusive disease on the microcirculation within human kidneys. METHOD: This study included five patients who underwent nephrectomy due to renovascular occlusion and seven nonstenotic discarded donor kidneys (four deceased donors). Micro-computed tomography was performed to assess microvascular spatial densities and tortuosity, an index of microvascular immaturity. Renal protein expression, gene expression and histology were studied in vitro using immunoblotting, polymerase chain reaction and staining. RESULTS: RAS demonstrated a loss of medium-sized vessels (0.2-0.3 mm) compared with donor kidneys (P = 0.037) and increased microvascular tortuosity. RAS kidneys had greater protein expression of angiopoietin-1, hypoxia-inducible factor-1α and thrombospondin-1 but lower protein expression of vascular endothelial growth factor (VEGF) than donor kidneys. Renal fibrosis, loss of peritubular capillaries (PTCs) and pericyte detachment were greater in RAS, yet they had more newly formed PTCs than donor kidneys. Therefore, our study quantified significant microvascular remodeling in the poststenotic human kidney. RAS induced renal microvascular loss, vascular remodeling and fibrosis. Despite downregulated VEGF, stenotic kidneys upregulated compensatory angiogenic pathways related to angiopoietin-1. CONCLUSIONS: These observations underscore the nature of human RAS as a microvascular disease distal to main vessel stenosis and support therapeutic strategies directly targeting the poststenotic kidney microcirculation in patients with RAS.


Assuntos
Obstrução da Artéria Renal , Angiopoietina-1/metabolismo , Angiopoietina-1/uso terapêutico , Animais , Fibrose , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Rim/patologia , Obstrução da Artéria Renal/complicações , Circulação Renal/fisiologia , Trombospondinas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Microtomografia por Raio-X
5.
Vascul Pharmacol ; 141: 106919, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34583025

RESUMO

The Angiopoietin-1/2 system is an opportune target for therapeutic intervention in a wide range of vascular pathologies, particularly through its association with endothelium. The complex multi-domain structure of native human Angiopoietin-1 has hindered its widespread applicability as a therapeutic agent, prompting the search for alternative approaches to mimicking the Ang1:Tie2 signalling axis; a system with highly complex patterns of regulation involving multiple structurally similar molecules. An engineered variant, Cartilage Oligomeric Matrix Protein - Angiopoietin-1 (COMP-Ang1), has been demonstrated to overcome the limitations of the native molecule and activate the Tie2 pathway with several fold greater potency than Ang1, both in vitro and in vivo. The therapeutic efficacy of COMP-Ang1, at both the vascular and systemic levels, is evident from multiple studies. Beneficial impacts on skeletal muscle regeneration, wound healing and angiogenesis have been reported alongside renoprotective, anti-hypertensive and anti-inflammatory effects. COMP-Ang1 has also demonstrated synergy with other compounds to heighten bone repair, has been leveraged for potential use as a co-therapeutic for enhanced targeted cancer treatment, and has received considerable attention as an anti-leakage agent for microvascular diseases like diabetic retinopathy. This review examines the vascular Angiopoietin:Tie2 signalling mechanism, evaluates the potential therapeutic merits of engineered COMP-Ang1 in both vascular and systemic contexts, and addresses the inherent translational challenges in moving this potential therapeutic from bench-to-bedside.


Assuntos
Angiopoietina-1 , Proteína de Matriz Oligomérica de Cartilagem , Transdução de Sinais , Angiopoietina-1/genética , Angiopoietina-1/uso terapêutico , Proteína de Matriz Oligomérica de Cartilagem/genética , Humanos , Engenharia de Proteínas , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Cicatrização
6.
Aging (Albany NY) ; 13(14): 19048-19063, 2021 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-34326273

RESUMO

Dystonia is a disorder associated with abnormalities in many brain regions including the basal ganglia and cerebellum. The toxin 3-Nitropropionic acid (3-NP) can induce neuropathologies in the mice striatum and nigra substance, including excitotoxicity, neuroinflammation, and extensive neuronal atrophy, characterized by progressive motor dysfunction, dystonia, and memory loss, mimicking those observed in humans. We established a mouse model of dystonia by administering 3-NP. Given the reported neuroprotective effects of the endothelial growth factor angiopoietin-1 (Ang-1) and the anti-inflammatory integrin αvß3 binding peptide C16, we performed this study to evaluate their combined effects on 3-NP striatal toxicity and their therapeutic potential with multiple methods using an in vivo mouse model. Sixty mice were equally and randomly divided into three groups: control, 3-NP treatment, and 3-NP+C16+Ang-1 treatment. Behavioral and electrophysiological tests were conducted and the effect of the combined C16+Ang-1 treatment on neural function recovery was determined. We found that C16+Ang-1 treatment alleviated 3-NP-induced behavioral, biochemical, and cellular alterations in the central nervous system and promoted function recovery by restoring vascular permeability and reducing inflammation in the micro-environment. In conclusion, our results confirmed the neuroprotective effect of combined C16+Ang-1 treatment and suggest their potential as a complementary therapeutic against 3-NP-induced dystonia.


Assuntos
Angiopoietina-1/uso terapêutico , Encéfalo/efeitos dos fármacos , Distonia/tratamento farmacológico , Inflamação/tratamento farmacológico , Neuroproteção , Peptídeos/uso terapêutico , Angiopoietina-1/farmacologia , Animais , Anti-Inflamatórios , Encéfalo/patologia , Encéfalo/fisiopatologia , Permeabilidade Capilar , Sistema Nervoso Central , Corpo Estriado , Modelos Animais de Doenças , Quimioterapia Combinada , Distonia/induzido quimicamente , Distonia/patologia , Distonia/fisiopatologia , Fatores de Crescimento Endotelial , Masculino , Camundongos Endogâmicos C57BL , Neurônios , Nitrocompostos , Peptídeos/farmacologia , Propionatos , Distribuição Aleatória
7.
Exp Biol Med (Maywood) ; 245(18): 1683-1696, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32915636

RESUMO

C16 peptide and angiopoietin-1 (Ang-1) have been found to have anti-inflammatory activity in various inflammation-related diseases. However, their combined role in acute respiratory distress syndrome (ARDS) has not been investigated yet. The objective of this study was to investigate the effects of C16 peptide and Ang-1 in combination with lipopolysaccharide (LPS)-induced inflammatory insult in vitro and in vivo. Human pulmonary microvascular endothelial cells and human pulmonary alveolar epithelial cells were used as cell culture systems, and an ARDS rodent model was used for in vivo studies. Our results demonstrated that C16 and Ang-1 in combination significantly suppressed inflammatory cell transmigration by 33% in comparison with the vehicle alone, and decreased the lung tissue wet-to-dry lung weight ratio to a maximum of 1.53, compared to 3.55 in the vehicle group in ARDS rats. Moreover, C + A treatment reduced the histology injury score to 60% of the vehicle control, enhanced arterial oxygen saturation (SO2), decreased arterial carbon dioxide partial pressure (PCO2), and increased oxygen partial pressure (PO2) in ARDS rats, while also improving the survival rate from 47% (7/15) to 80% (12/15) and diminishing fibrosis, necrosis, and apoptosis in lung tissue. Furthermore, when C + A therapy was administered 4 h following LPS injection, the treatment showed significant alleviating effects on pulmonary inflammatory cell infiltration 24 h postinsult. In conclusion, our in vitro and in vivo studies show that C16 and Ang-1 exert protective effects against LPS-induced inflammatory insult. C16 and Ang-1 hold promise as a novel agent against LPS-induced ARDS. Further studies are needed to determine the potential for C16 and Ang-1 in combination in treating inflammatory lung diseases.


Assuntos
Angiopoietina-1/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Peptídeos/uso terapêutico , Substâncias Protetoras/uso terapêutico , Síndrome do Desconforto Respiratório/tratamento farmacológico , Sequência de Aminoácidos , Angiopoietina-1/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Inflamação/patologia , Lipopolissacarídeos , Lesão Pulmonar/complicações , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/patologia , Masculino , Monócitos/efeitos dos fármacos , Monócitos/patologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Peptídeos/química , Peptídeos/farmacologia , Substâncias Protetoras/farmacologia , Fibrose Pulmonar/complicações , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/patologia , Troca Gasosa Pulmonar , Ratos Wistar , Síndrome do Desconforto Respiratório/induzido quimicamente , Síndrome do Desconforto Respiratório/complicações , Síndrome do Desconforto Respiratório/patologia
8.
J Clin Invest ; 130(10): 5562-5575, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32673288

RESUMO

Pathologic lymphatic remodeling in lymphedema evolves during periods of tissue inflammation and hypoxia through poorly defined processes. In human and mouse lymphedema, there is a significant increase of hypoxia inducible factor 1 α (HIF-1α), but a reduction of HIF-2α protein expression in lymphatic endothelial cells (LECs). We questioned whether dysregulated expression of these transcription factors contributes to disease pathogenesis and found that LEC-specific deletion of Hif2α exacerbated lymphedema pathology. Even without lymphatic vascular injury, the loss of LEC-specific Hif2α caused anatomic pathology and a functional decline in fetal and adult mice. These findings suggest that HIF-2α is an important mediator of lymphatic health. HIF-2α promoted protective phosphorylated TIE2 (p-TIE2) signaling in LECs, a process also replicated by upregulating TIE2 signaling through adenovirus-mediated angiopoietin-1 (Angpt1) gene therapy. Our study suggests that HIF-2α normally promotes healthy lymphatic homeostasis and raises the exciting possibility that restoring HIF-2α pathways in lymphedema could mitigate long-term pathology and disability.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Sistema Linfático/metabolismo , Sistema Linfático/patologia , Linfedema/metabolismo , Linfedema/patologia , Angiopoietina-1/genética , Angiopoietina-1/uso terapêutico , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Sistema Linfático/embriologia , Linfedema/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Gravidez , Receptor TIE-2/metabolismo , Transdução de Sinais
9.
Br J Anaesth ; 121(5): 1041-1051, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30336848

RESUMO

BACKGROUND: Cardiopulmonary bypass (CPB) during cardiac surgery impairs microcirculatory perfusion and is paralleled by vascular leakage. The endothelial angiopoietin/Tie2 system controls microvascular leakage. This study investigated whether targeting Tie2 with the angiopoietin-1 mimetic vasculotide reduces vascular leakage and preserves microcirculatory perfusion in a rat CPB model. METHODS: Rats were subjected to 75 min of CPB after treatment with vasculotide or phosphate buffered solution as control or underwent a sham procedure. Microcirculatory perfusion and leakage were assessed with intravital microscopy (n=10 per group) and Evans blue dye extravasation (n=13 per group), respectively. Angiopoietin-1, -2, and Tie2 protein and gene expression were determined in plasma, kidney, and lung. RESULTS: CPB immediately impaired microcirculatory perfusion [5 (4-8) vs 10 (7-12) vessels per recording, P=0.002] in untreated CPB rats compared with sham, which persisted after weaning from CPB. CPB increased circulating angiopoeietin-1, -2, and soluble Tie2 concentrations and reduced Tie2 messenger ribonucleic acid (mRNA) expression in kidney and lung. Moreover, CPB increased Evans blue dye leakage in kidney [12 (8-25) vs 7 (1-12) µg g-1, P=0.04] and lung [and 23 (13-60) vs 6 (4-16) µg g-1, P=0.001] compared with sham. Vasculotide treatment preserved microcirculatory perfusion during and after CPB. Moreover, vasculotide treatment reduced Evans blue dye extravasation in lung compared with CPB control [18 (6-28) µg g-1vs 23 (13-60) µg g-1, P=0.04], but not in kidney [10 (3-23) vs 12 (8-25) µg g-1, P=0.38]. Vasculotide did not affect circulating or mRNA expression of angiopoietin-1, -2, and Tie2 concentrations compared with untreated CPB controls. CONCLUSIONS: Treatment with the angiopoietin-1 mimetic vasculotide reduced pulmonary vascular leakage and preserved microcirculatory perfusion during CPB in a rat model.


Assuntos
Angiopoietina-1/uso terapêutico , Ponte Cardiopulmonar/efeitos adversos , Fragmentos de Peptídeos/uso terapêutico , Circulação Pulmonar/efeitos dos fármacos , Angiopoietina-1/biossíntese , Angiopoietina-1/genética , Angiopoietina-2/biossíntese , Angiopoietina-2/genética , Animais , Capilares/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Masculino , Microcirculação/efeitos dos fármacos , Ratos , Ratos Wistar , Receptor TIE-2/biossíntese , Receptor TIE-2/genética , Receptor TIE-2/metabolismo
10.
Acta Cir Bras ; 33(4): 386-395, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29768541

RESUMO

PURPOSE: To investigate the safety and clinical, hemodynamic and tissue improvement ability in mini pigs undergoing cell and gene therapy for the treatment of acute myocardial infarction. METHODS: Thirty-two mini pigs Br1 lineage, 12 months old, undergoing induction of acute myocardial infarction by occlusion of the diagonal branch of the paraconal coronary. They were divided into 4 groups: one control group and 3 treatment groups (cell therapy and gene cell therapy). Echocardiography reviews were performed on three occasions and histopathological analysis was performed after 4 weeks. Analysis of variance (ANOVA), Tukey and Wilcoxon tests, were performed. RESULTS: Association of vascular endothelial growth factor (VEGF) with angiopoietin1 (Ang1) presented satisfactory results in the improvement of ventricular function following ischemic cardiomyopathy in mini pigs when compared to the results of the other treated groups. CONCLUSION: The therapy with VEGF and the combination of VEGF with Ang1, promoted recovered function of the myocardium, characterized by reduced akinetic area and induction of neovascularization.


Assuntos
Angiopoietina-1/uso terapêutico , Terapia Baseada em Transplante de Células e Tecidos/métodos , Terapia Genética/métodos , Infarto do Miocárdio/terapia , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Função Ventricular/fisiologia , Animais , Modelos Animais de Doenças , Ecocardiografia , Hemodinâmica/fisiologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Necrose , Neovascularização Fisiológica , Reprodutibilidade dos Testes , Suínos , Porco Miniatura , Resultado do Tratamento , Cicatrização
11.
Mediators Inflamm ; 2018: 4187347, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29670463

RESUMO

Neuromyelitis optica (NMO) is an autoimmune inflammatory demyelinating disease that mainly affects the spinal cord and optic nerve, causing blindness and paralysis in some individuals. Moreover, NMO may cause secondary complement-dependent cytotoxicity (CDC), leading to oligodendrocyte and neuronal damage. In this study, a rodent NMO model, showing typical NMO pathogenesis, was induced with NMO-IgG from patient serum and human complement. We then tested whether the combination of C16, an αvß3 integrin-binding peptide, and angiopoietin-1 (Ang1), a member of the endothelial growth factor family, could alleviate NMO in the model. Our results demonstrated that this combination therapy significantly decreased disease severity, inflammatory cell infiltration, secondary demyelination, and axonal loss, thus reducing neural death. In conclusion, our study suggests a possible treatment that can relieve progressive blindness and paralysis in an animal model of NMO through improvement of the inflammatory milieu.


Assuntos
Angiopoietina-1/uso terapêutico , Neuromielite Óptica/tratamento farmacológico , Peptídeos/uso terapêutico , Animais , Axônios/metabolismo , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/fisiopatologia , Modelos Animais de Doenças , Feminino , Imunoglobulina G/metabolismo , Neurofisiologia/métodos , Oligodendroglia/metabolismo , Ratos
12.
Acta cir. bras ; 33(4): 386-395, Apr. 2018. tab, graf
Artigo em Inglês | LILACS | ID: biblio-886279

RESUMO

Abstract Purpose: To investigate the safety and clinical, hemodynamic and tissue improvement ability in mini pigs undergoing cell and gene therapy for the treatment of acute myocardial infarction. Methods: Thirty-two mini pigs Br1 lineage, 12 months old, undergoing induction of acute myocardial infarction by occlusion of the diagonal branch of the paraconal coronary. They were divided into 4 groups: one control group and 3 treatment groups (cell therapy and gene cell therapy). Echocardiography reviews were performed on three occasions and histopathological analysis was performed after 4 weeks. Analysis of variance (ANOVA), Tukey and Wilcoxon tests, were performed. Results: Association of vascular endothelial growth factor (VEGF) with angiopoietin1 (Ang1) presented satisfactory results in the improvement of ventricular function following ischemic cardiomyopathy in mini pigs when compared to the results of the other treated groups. Conclusion: The therapy with VEGF and the combination of VEGF with Ang1, promoted recovered function of the myocardium, characterized by reduced akinetic area and induction of neovascularization.


Assuntos
Animais , Terapia Genética/métodos , Função Ventricular/fisiologia , Angiopoietina-1/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Terapia Baseada em Transplante de Células e Tecidos/métodos , Infarto do Miocárdio/terapia , Suínos , Porco Miniatura , Cicatrização , Ecocardiografia , Reprodutibilidade dos Testes , Resultado do Tratamento , Neovascularização Fisiológica , Modelos Animais de Doenças , Hemodinâmica/fisiologia , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/patologia , Necrose
13.
J Cell Physiol ; 233(11): 8567-8577, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29377123

RESUMO

Optic nerve injury triggered retinal ganglion cell (RGC) death and optic nerve atrophy lead to visual loss. Bone marrow mesenchymal stem cells (BMSCs) are stromal cells, capable of proliferating and differentiating into different types of tissues. This aims of this study is to investigate the role of BMSCs transfected with angiopoietin-1 (Ang-1) in optic nerve injury induced by hyperoxia in a neonatal mice model. Ang-1 overexpression vector was constructed and used to transfect BMSCs. Reverse transcription-quantitative polymerase chain reaction was performed to detect Ang-1 expression in BMSCs. The hyperoxia-induced optic nerve injury model was established. The optic nerves at 6-7 mm posterior to the eyeball were extracted, and were treated with luxol fast blue staining, immunohistochemistry, immunofluorescence, and transmission electron microscopy to examine the effects of Ang-1-modified BMSCs on optic nerve injury induced by hyperoxia. The mice in the Ang-1 + BMSCs and BMSCs groups showed remarkably improved myelin sheaths of nerve fibers compared to the hyperoxia saline group. The positive expression and integrated optic density of Ang-1 in the Ang-1 + BMSCs group were significantly higher compared to the air control, hyperoxia saline and BMSCs groups. The number and diameter of myelinated nerve fibers, the diameter of axons and the thickness of myelin sheath in the air control and Ang-1 + BMSCs groups were higher compared to the hyperoxia saline group. Our study provides evidence supporting that Ang-1-modified BMSCs may have preventive and therapeutic effects on hyperoxia-induced optic nerve injury in neonatal mice.


Assuntos
Angiopoietina-1/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Traumatismos do Nervo Óptico/terapia , Angiopoietina-1/uso terapêutico , Animais , Animais Recém-Nascidos , Axônios/efeitos dos fármacos , Axônios/metabolismo , Modelos Animais de Doenças , Humanos , Hiperóxia/complicações , Camundongos , Traumatismos do Nervo Óptico/etiologia , Traumatismos do Nervo Óptico/genética , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Transfecção
14.
PLoS One ; 12(3): e0174315, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28334037

RESUMO

Vascular endothelial growth factor (VEGF) and other pro-angiogenic growth factors have been investigated to enhance muscle tissue perfusion and repair in Duchenne muscular dystrophy (DMD). Current understanding is limited by a lack of functional data following in vivo delivery of these growth factors. We previously used dynamic contrast-enhanced computed tomography to monitor disease progression in murine models of DMD, but no study to date has utilized this imaging technique to assess vascular therapy in a preclinical model of DMD. In the current study, we locally delivered VEGF and ANG1 alone or in combination to dystrophic hind limb skeletal muscle. Using functional imaging, we found the combination treatment as well as ANG1 alone prevented decline in muscle perfusion whereas VEGF alone had no effect compared to controls. These findings were validated histologically as demonstrated by increased alpha-smooth muscle actin-positive vessels in muscles that received either VEGF+ANG1 or ANG1 alone compared to the sham group. We further show that ANG1 alone slows progression of fibrosis compared to either sham or VEGF treatment. The findings from this study shed new light on the functional effects of vascular therapy and suggest that ANG1 alone may be a candidate therapy in the treatment of DMD.


Assuntos
Angiopoietina-1/uso terapêutico , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Animais , Modelos Animais de Doenças , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Camundongos , Microscopia , Músculo Esquelético/diagnóstico por imagem , Músculo Esquelético/efeitos dos fármacos , Distrofia Muscular de Duchenne/diagnóstico por imagem , Distrofia Muscular de Duchenne/patologia , Proteínas Recombinantes , Tomografia Computadorizada por Raios X/métodos , Fator A de Crescimento do Endotélio Vascular/uso terapêutico
15.
Acta Biomater ; 48: 58-67, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27756647

RESUMO

Acute myocardial infarction (MI) caused by ischemia is the most common cause of cardiac dysfunction. While growth factor therapy is promising, the retention in the highly vascularized myocardium is limited and prevents sustained activation needed for adequate cellular responses. Here, we demonstrated the use of polyethylene glycol-fibrinogen (PF) hydrogels for sustained dual delivery of vascular endothelial growth factor (VEGF) and angiopoietin-1 (ANG-1) to enhance myocardial repair and function. VEGF and ANG-1 were incorporated in PF hydrogels and their in vitro characteristics were studied. Acute MI was generated in a rodent model with rats randomly assigned to 4 groups; sham, saline, PF and PF-VEGF-ANG1 (n=10 each group). Saline or hydrogel was injected in infarct and peri-infarct areas of the myocardium. After 4weeks, myocardial function was assessed using echocardiography. Tissue samples were harvested for Hematoxylin and Eosin, Masson Trichrome and capillary staining to assess the extent of fibrotic scar and arteriogenesis. Both VEGF and ANG-1 were released in a sustained and controlled manner over 30days. PF-VEGF-ANG1 treated animals showed the best improvement in cardiac function, highest degree of cardiac muscle preservation, and arteriogenesis. This study demonstrates that PF hydrogels can simultaneously provide mechanical support to attenuate adverse myocardial remodelling, and a pro-angiogenic benefit from the sustained VEGF and ANG1 delivery that culminates in a restorative effect following MI. The utility of this synergistic, biomaterial-based growth factor delivery may have clinical implications in the prevention of post-MI cardiac dysfunction. STATEMENT OF SIGNIFICANCE: Acute myocardial infarction (MI) caused by ischemia is the most common cause of cardiac dysfunction. Here, we demonstrated the use of polyethylene glycol-fibrinogen (PF) hydrogels for sustained dual delivery of vascular endothelial growth factor (VEGF) and angiopoietin-1 (ANG-1) to enhance myocardial repair and function. Treated animals showed the best improvement in cardiac function, highest degree of cardiac muscle preservation, and arteriogenesis. This study demonstrates that PF hydrogels can simultaneously provide mechanical support to attenuate adverse myocardial remodelling, and a pro-angiogenic benefit from the sustained VEGF and ANG1 delivery that culminates in a restorative effect following MI.


Assuntos
Angiopoietina-1/administração & dosagem , Angiopoietina-1/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Isquemia Miocárdica/tratamento farmacológico , Miocárdio/patologia , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Angiopoietina-1/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Fibrinogênio/química , Imunofluorescência , Testes de Função Cardíaca , Hemodinâmica/efeitos dos fármacos , Humanos , Cinética , Masculino , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Polietilenoglicóis/química , Ratos Wistar , Coloração e Rotulagem , Fator A de Crescimento do Endotélio Vascular/farmacologia
16.
CNS Neurol Disord Drug Targets ; 15(4): 496-513, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26553158

RESUMO

Breakdown of normal blood-brain barrier function and accompanying vascular leakage are fundamental stages in the onset of multiple sclerosis and its animal counterpart, experimental allergic encephalomyelitis. In the present study, angiopoietin-1, an endothelial growth factor well known for its role in establishing and maintaining vascular integrity, and C16, a peptide that competitively binds to integrin αvß3 expressed on endothelial cells, were used to treat acute experimental allergic encephalomyelitis in Lewis rats. Angiopoietin-1 was more effective than C16 for reducing inflammation-induced vascular leakage. Moreover, treatment with a combination of angiopoietin-1 and C16 resulted in greater effects, not only in alleviating inflammation and reducing axonal loss/demyelination but also in down-regulating pro-inflammatory cytokine expression and improving electrophysiological dysfunction, than treatment with either angiopoietin-1 or C16 alone. Different protective effects were observed with angiopoietin-1 and C16 treatment suggesting that these proteins target specific receptors to act through different pathways. Furthermore, angiopoietin-1 and C16 may form the basis of a promising therapeutic strategy for experimental allergic encephalomyelitis and multiple sclerosis.


Assuntos
Angiopoietina-1/uso terapêutico , Traumatismo Cerebrovascular/tratamento farmacológico , Encefalomielite Autoimune Experimental/complicações , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/tratamento farmacológico , Oligopeptídeos/uso terapêutico , Animais , Vasos Sanguíneos/patologia , Vasos Sanguíneos/ultraestrutura , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/ultraestrutura , Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Traumatismo Cerebrovascular/etiologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Quimioterapia Combinada , Encefalomielite Autoimune Experimental/etiologia , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/prevenção & controle , Regulação da Expressão Gênica/imunologia , Inflamação/etiologia , Injeções Subcutâneas , Masculino , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/etiologia , Ratos , Ratos Endogâmicos Lew , Tempo de Reação/efeitos dos fármacos , Fatores de Tempo , Fator de Transcrição RelA/metabolismo
17.
Diabetes ; 64(12): 4247-59, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26340930

RESUMO

Diabetic retinopathy (DR) is the leading cause of blindness in the working-age population in the U.S. The vision-threatening processes of neuroglial and vascular dysfunction in DR occur in concert, driven by hyperglycemia and propelled by a pathway of inflammation, ischemia, vasodegeneration, and breakdown of the blood retinal barrier. Currently, no therapies exist for normalizing the vasculature in DR. Here, we show that a single intravitreal dose of adeno-associated virus serotype 2 encoding a more stable, soluble, and potent form of angiopoietin 1 (AAV2.COMP-Ang1) can ameliorate the structural and functional hallmarks of DR in Ins2Akita mice, with sustained effects observed through six months. In early DR, AAV2.COMP-Ang1 restored leukocyte-endothelial interaction, retinal oxygenation, vascular density, vascular marker expression, vessel permeability, retinal thickness, inner retinal cellularity, and retinal neurophysiological response to levels comparable with nondiabetic controls. In late DR, AAV2.COMP-Ang1 enhanced the therapeutic benefit of intravitreally delivered endothelial colony-forming cells by promoting their integration into the vasculature and thereby stemming further visual decline. AAV2.COMP-Ang1 single-dose gene therapy can prevent neurovascular pathology, support vascular regeneration, and stabilize vision in DR.


Assuntos
Angiopoietina-1/uso terapêutico , Proteína de Matriz Oligomérica de Cartilagem/uso terapêutico , Diabetes Mellitus Tipo 1/complicações , Retinopatia Diabética/terapia , Modelos Animais de Doenças , Terapia Genética , Retina/patologia , Angiopoietina-1/química , Angiopoietina-1/genética , Angiopoietina-1/metabolismo , Animais , Proteína de Matriz Oligomérica de Cartilagem/química , Proteína de Matriz Oligomérica de Cartilagem/genética , Proteína de Matriz Oligomérica de Cartilagem/metabolismo , Células Cultivadas , Terapia Combinada/efeitos adversos , Cruzamentos Genéticos , Retinopatia Diabética/imunologia , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/transplante , Terapia Genética/efeitos adversos , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/imunologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Injeções Intravítreas , Leucócitos/citologia , Leucócitos/imunologia , Leucócitos/metabolismo , Leucócitos/patologia , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Estabilidade Proteica , Distribuição Aleatória , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Retina/imunologia , Retina/metabolismo , Solubilidade
18.
Exp Neurol ; 261: 245-57, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24852101

RESUMO

Multiple sclerosis (MS) is characterized by perivascular inflammatory infiltration, secondary demyelination, and axonal loss in the central nervous system. Angiopoietin-1 (Ang-1) constitutes a family of endothelial growth factors that can inhibit MS-associated, inflammation-induced blood vascular leakage and lessen increased blood vessel permeability. This study was designed to investigate the effects of Ang-1 on a model of acute experimental autoimmune encephalomyelitis (EAE). Evans blue and the luciferase assay were employed to test blood vessel permeability, while immunohistochemistry, ELISA, and Western blotting were used to assess the degree of inflammation. Electron microscopy and cortical somatosensory evoked potentials were also used to observe axonal loss, white matter demyelination, and functional impairment in EAE groups. Our results showed that Ang-1 treatment could ameliorate inflammation-induced leakage, inhibit inflammatory cell infiltration into the brain and spinal cord, and improve functional impairment associated with EAE in a dose-dependent manner.


Assuntos
Angiopoietina-1/uso terapêutico , Doenças Desmielinizantes/tratamento farmacológico , Encefalomielite Autoimune Experimental/complicações , Inflamação/tratamento farmacológico , Inflamação/etiologia , Animais , Vasos Sanguíneos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/ultraestrutura , Permeabilidade Capilar/efeitos dos fármacos , Citocinas/metabolismo , Doenças Desmielinizantes/etiologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Encefalomielite Autoimune Experimental/imunologia , Fatores de Crescimento Endotelial/metabolismo , Potenciais Somatossensoriais Evocados/efeitos dos fármacos , Masculino , Microscopia Eletrônica de Transmissão , Infiltração de Neutrófilos/efeitos dos fármacos , Ratos , Ratos Endogâmicos Lew , Tempo de Reação/efeitos dos fármacos , Coloração pela Prata
19.
J Sex Med ; 10(12): 2912-27, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23937122

RESUMO

INTRODUCTION: Erectile dysfunction (ED) is a highly prevalent complication of diabetes, and the severity of endothelial dysfunction is one of the most important factors in reduced responsiveness to oral phosphodiesterase type 5 inhibitors. AIM: To study the effects of human angiopoietin-4 (Ang-4) protein on erectile function in diabetic mice. METHODS: Diabetes was induced by intraperitoneal injection of streptozotocin into 8-week-old C57BL/6J male mice. At 8 weeks after the induction of diabetes, the animals were divided into four groups: control nondiabetic mice and diabetic mice receiving two successive intracavernous injections of phosphate buffered saline (days -3 and 0), a single intracavernous injection of Ang-4 protein (day 0), or two successive intracavernous injections of Ang-4 protein (days -3 and 0). MAIN OUTCOME MEASURES: One week after treatment, we measured erectile function by electrical stimulation of the cavernous nerve. The penis was harvested and stained with hydroethidine or antibodies to Ang-4, platelet/endothelial cell adhesion molecule-1, and phosphorylated endothelial nitric oxide synthase (eNOS). We also determined the differential expression of Ang-4 in cavernous tissue in the control and diabetic mice. The effect of Ang-4 protein on the phosphorylation of Tie-2, Akt, and eNOS was determined in human umbilical vein endothelial cells (HUVECs) by Western blot. RESULTS: The cavernous expression of Ang-4 was downregulated in diabetic mice; Ang-4 was mainly expressed in endothelial cells. Local delivery of Ang-4 protein significantly increased cavernous endothelial content, induced eNOS phosphorylation, and decreased the generation of superoxide anion and apoptosis in diabetic mice. Ang-4 protein strongly increased the phosphorylation of Tie-2, Akt, and eNOS in HUVECs. Repeated intracavernous injections of Ang-4 induced significant restoration of erectile function in diabetic mice (87% of control values), whereas a single intracavernous injection of Ang-4 protein elicited modest improvement. CONCLUSIONS: Cavernous endothelial regeneration by use of Ang-4 protein may have potential for the treatment of vascular disease-induced ED, such as diabetic ED.


Assuntos
Angiopoietinas/administração & dosagem , Diabetes Mellitus Experimental/complicações , Disfunção Erétil/tratamento farmacológico , Ereção Peniana/efeitos dos fármacos , Pênis/efeitos dos fármacos , Angiopoietina-1/metabolismo , Angiopoietina-1/farmacologia , Angiopoietina-1/uso terapêutico , Angiopoietinas/metabolismo , Animais , Diabetes Mellitus Experimental/fisiopatologia , Disfunção Erétil/etiologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico Sintase Tipo III/fisiologia , Ereção Peniana/fisiologia , Pênis/irrigação sanguínea , Fosforilação , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regeneração/efeitos dos fármacos
20.
Cytokine Growth Factor Rev ; 24(6): 579-92, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23838360

RESUMO

Angiopoietin-1 and -2 are endogenous ligands for the vascular endothelial receptor tyrosine kinase Tie2. Signalling by angiopoietin-1 promotes vascular endothelial cell survival and the sprouting and reorganisation of blood vessels, as well as inhibiting activation of the vascular endothelial barrier to reduce leakage and leucocyte migration into tissues. Angiopoietin-2 generally has an opposing action, and is released naturally at times of vascular growth and inflammation. There is a significant body of emerging evidence that promoting the actions of angiopoietin-1 through Tie2 is of benefit in pathologies of vascular activation, such as sepsis, stroke, diabetic retinopathy and asthma. Similarly, methods to inhibit the actions of angiopoietin-2 are emerging and have been demonstrated to be of preclinical and clinical benefit in reducing tumour angiogenesis. Here the author reviews the evidence for potential benefits of modulation of the interaction of angiopoietins with Tie2, and the potential applications. Additionally, methods for delivery of the complex protein angiopoietin-1 are discussed, as well as potentially deleterious consequences of administering angiopoietin-1.


Assuntos
Angiopoietina-1/metabolismo , Angiopoietina-2/metabolismo , Receptor TIE-2/metabolismo , Doenças Vasculares/metabolismo , Angiopoietina-1/farmacologia , Angiopoietina-1/uso terapêutico , Animais , Humanos , Doenças Vasculares/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA