Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
Biomed Pharmacother ; 143: 112186, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34649339

RESUMO

INTRODUCTION: Patients with peripheral artery disease have poor prognosis despite advances in vascular surgery. Therefore, attempts have been made at using gene and cell therapy to stimulate angiogenesis in the lower limbs in patients with critical lower limb ischemia (CLI). METHODS: The study included 30 rats divided into 3 groups. An intramuscular injection of a therapeutic gene or cells in the right hind limb was administered in each group: angiopoietin-1 (ANG1) plasmid in group 1, ANG1/vascular endothelial growth factor (ANG1/VEGF) bicistronic construct in group 2, and naked plasmid in group 3 (control). After 3 months of follow-up, tissue samples were harvested, and vessels that stained positively for CD34 cells were quantified. RESULTS: The highest CD34+ cell count was noted in the ANG1/VEGF group (98.26 cells), followed by the ANG1 group (80.31) and control group (47.93). The CD34+ cell count was significantly higher in the ANG1/VEGF and ANG1 groups than in the control group. There was no significant difference in the CD34+ cell count between the ANG1/VEGF and ANG1 groups. CONCLUSION: Our study confirmed that therapy with ANG1 plasmid alone or ANG1/VEGF bicistronic construct is safe and effective in a rat model. The therapy resulted in the recruitment of more CD34+ vascular endothelial cells than in the control group receiving naked plasmid.


Assuntos
Angiopoietina-1/biossíntese , Antígenos CD34/metabolismo , Movimento Celular , Células Progenitoras Endoteliais/metabolismo , Terapia Genética , Isquemia/terapia , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/biossíntese , Angiopoietina-1/genética , Animais , Modelos Animais de Doenças , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Membro Posterior , Injeções Intramusculares , Isquemia/genética , Isquemia/metabolismo , Isquemia/fisiopatologia , Masculino , Ratos Endogâmicos BUF , Fator A de Crescimento do Endotélio Vascular/genética
2.
Biotechnol Bioeng ; 118(1): 423-432, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32970320

RESUMO

Vascular leak is a key driver of organ injury in diseases, and strategies that reduce enhanced permeability and vascular inflammation are promising therapeutic targets. Activation of the angiopoietin-1 (ANG1)-Tie2 tyrosine kinase signaling pathway is an important regulator of vascular quiescence. Here we describe the design and construction of a new soluble ANG1 mimetic that is a potent activator of endothelial Tie2 in vitro and in vivo. Using a chimeric fusion strategy, we replaced the extracellular matrix (ECM) binding and oligomerization domain of ANG1 with a heptameric scaffold derived from the C-terminus of serum complement protein C4-binding protein α. We refer to this new fusion protein biologic as Hepta-ANG1, which forms a stable heptamer and induces Tie2 phosphorylation in cultured cells, and in the lung following intravenous injection of mice. Injection of Hepta-ANG1 ameliorates vascular endothelial growth factor- and lipopolysaccharide-induced vascular leakage, in keeping with the known functions of Angpt1-Tie2 in maintaining quiescent vascular stability. The new Hepta-ANG1 fusion is easy to produce and displays remarkable stability with high multimericity that can potently activate Tie2. It could be a new candidate ANG1 mimetic therapy for treatments of inflammatory vascular leak, such as acute respiratory distress syndrome and sepsis.


Assuntos
Angiopoietina-1 , Proteína de Ligação ao Complemento C4b , Células Endoteliais da Veia Umbilical Humana/metabolismo , Proteínas Recombinantes de Fusão , Doenças Vasculares/tratamento farmacológico , Angiopoietina-1/biossíntese , Angiopoietina-1/genética , Angiopoietina-1/farmacologia , Animais , Proteína de Ligação ao Complemento C4b/biossíntese , Proteína de Ligação ao Complemento C4b/genética , Proteína de Ligação ao Complemento C4b/farmacologia , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Domínios Proteicos , Coelhos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Síndrome do Desconforto Respiratório/tratamento farmacológico , Síndrome do Desconforto Respiratório/metabolismo , Síndrome do Desconforto Respiratório/patologia , Sepse/tratamento farmacológico , Sepse/metabolismo , Sepse/patologia , Doenças Vasculares/metabolismo , Doenças Vasculares/patologia
3.
J Biol Chem ; 295(34): 12045-12057, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32616651

RESUMO

Ambra1 is considered an autophagy and trafficking protein with roles in neurogenesis and cancer cell invasion. Here, we report that Ambra1 also localizes to the nucleus of cancer cells, where it has a novel nuclear scaffolding function that controls gene expression. Using biochemical fractionation and proteomics, we found that Ambra1 binds to multiple classes of proteins in the nucleus, including nuclear pore proteins, adaptor proteins such as FAK and Akap8, chromatin-modifying proteins, and transcriptional regulators like Brg1 and Atf2. We identified biologically important genes, such as Angpt1, Tgfb2, Tgfb3, Itga8, and Itgb7, whose transcription is regulated by Ambra1-scaffolded complexes, likely by altering histone modifications and Atf2 activity. Therefore, in addition to its recognized roles in autophagy and trafficking, Ambra1 scaffolds protein complexes at chromatin, regulating transcriptional signaling in the nucleus. This novel function for Ambra1, and the specific genes impacted, may help to explain the wider role of Ambra1 in cancer cell biology.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cromatina/metabolismo , Regulação da Expressão Gênica , Complexos Multiproteicos/metabolismo , Transdução de Sinais , Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ancoragem à Quinase A/metabolismo , Fator 2 Ativador da Transcrição/genética , Fator 2 Ativador da Transcrição/metabolismo , Transporte Ativo do Núcleo Celular/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Angiopoietina-1/biossíntese , Angiopoietina-1/genética , Linhagem Celular , Cromatina/genética , DNA Helicases/genética , DNA Helicases/metabolismo , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Humanos , Cadeias alfa de Integrinas/biossíntese , Cadeias alfa de Integrinas/genética , Cadeias beta de Integrinas/biossíntese , Cadeias beta de Integrinas/genética , Complexos Multiproteicos/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta2/biossíntese , Fator de Crescimento Transformador beta2/genética , Fator de Crescimento Transformador beta3/biossíntese , Fator de Crescimento Transformador beta3/genética
4.
J Cereb Blood Flow Metab ; 39(12): 2343-2354, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31581897

RESUMO

Clinical studies report that low circulating angiopoietin-1 concentration at presentation predicts worse outcomes after ischaemic stroke. Upregulating angiopoietin-1 may therefore have therapeutic benefit for ischaemic stroke. This systematic review assessed whether upregulating angiopoietin-1 improved outcomes in rodent models of ischaemic stroke. Random-effects models quantified the effect of angiopoietin-1 upregulation on stroke severity in terms of the size of cerebral infarction and the extent of blood-brain barrier permeability. Eleven studies utilising rat and mouse models of ischaemic stroke fulfilled the inclusion criteria. Meta-analyses demonstrated that angiopoietin-1 upregulation significantly reduced cerebral infarction size (standardised mean difference: -3.02; 95% confidence intervals: -4.41, -1.63; p < 0.001; n = 171 animals) and improved blood-brain barrier integrity (standardized mean difference: -2.02; 95% confidence intervals: -3.27, -0.77; p = 0.002; n = 129 animals). Subgroup analyses demonstrated that angiopoietin-1 upregulation improved outcomes in models of transient, not permanent cerebral ischaemia. Six studies assessed the effect of angiopoietin-1 upregulation on neurological function; however, inter-study heterogeneity prevented meta-analysis. In conclusion, published rodent data suggest that angiopoietin-1 upregulation improves outcome following temporary cerebral ischaemia by reducing cerebral infarction size and improving blood-brain barrier integrity. Additional research is required to examine the effect of angiopoietin-1 upregulation on neurological function during stroke recovery and investigate the benefit and risks in patients.


Assuntos
Angiopoietina-1/biossíntese , Barreira Hematoencefálica , Infarto Cerebral , Acidente Vascular Cerebral , Regulação para Cima , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Infarto Cerebral/metabolismo , Infarto Cerebral/patologia , Infarto Cerebral/fisiopatologia , Modelos Animais de Doenças , Humanos , Camundongos , Ratos , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia
5.
Acta Biomater ; 97: 578-586, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31349056

RESUMO

Cellular attachment and response to biomaterials are mediated by integrin receptor binding to extracellular matrix proteins adsorbed onto the material surface. Osteoblasts interact with their substrates via several integrin complexes including fibronectin-binding α5ß1 and collagen-binding α1ß1 and α2ß1. Knockdown of α2 or ß1 integrin subunits inhibits the production of factors that promote an osteogenic microenvironment, including osteocalcin, osteoprotegerin, and TGFß1. Osteoblasts also secrete several angiogenic growth factors such as VEGF-A (VEGF165), FGF-2, and angiopoietin 1, which are regulated by titanium surface topography and surface energy. Here, we examined whether signaling through integrin receptor complexes regulates production and secretion of angiogenic factors during osteoblast differentiation on microtextured Ti surfaces. To do this, integrin subunits α1, α2, α5, and ß1 were stably silenced in MG63 osteoblast-like cells cultured on grit-blasted/acid-etched hydrophobic Ti (SLA) or on hydrophilic SLA (modSLA). VEGF-A production increased in response to Ti surface topography and energy in integrin α2, α5, and ß1 silenced cells but decreased in α1-silenced cells. FGF-2 decreased on modSLA substrates in both α1 and α2-silenced cells but was unchanged in response to silencing of either α5 or ß1. In integrin α1, α2, and ß1-silenced cells, Ang-1 increased on modSLA but α5-silencing did not affect Ang-1 production during surface mediated differentiation. These results suggest that signaling through specific integrin receptor complexes during osteoblast differentiation on microstructured Ti substrates, regulates the production of angiogenic factors by those cells, and this is differentially regulated by surface hydrophilicity. STATEMENT OF SIGNIFICANCE: Successful implantation of synthetic biomaterials into bone depends on the biological process known as osseointegration. Osseointegration is a highly regulated communication of cells that orchestrates the migration of progenitor cells towards the implant site and promotes the deposition and mineralization of extracellular matrix proteins within the implant microenvironment, to tightly join the implant to native bone. In this process, angiogenesis functions as the initiation site of progenitor cell migration and is necessary for matrix deposition by providing the necessary nutrients for bone formation. In the present study, we show a novel regulation of specific angiogenic growth factors by integrin receptor complexes. This research is important to develop biomaterials that promote and maintain osseointegration through proper vascularization and prevent implant failure in patients lacking sufficient angiogenesis.


Assuntos
Angiopoietina-1/biossíntese , Fator 2 de Crescimento de Fibroblastos/biossíntese , Integrinas/metabolismo , Osteoblastos/metabolismo , Titânio/farmacologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Humanos , Osteoblastos/citologia , Propriedades de Superfície , Titânio/química
6.
Int J Cancer ; 144(9): 2279-2289, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30548585

RESUMO

Hematopoietic progenitors, residing in the bone marrow (BM) niche, are supported by mesenchymal stromal cells (MSCs). Cytogenetic and molecular aberrations in these progenitors lead to acute myeloid leukemia (AML). The BM-MSC role in leukemogenesis is not fully elucidated. In the current study, an ex-vivo system of patient's own stroma (POS), best mimicking the in-vivo BM niche, has been developed aiming to unravel interactions and crosstalk between MSCs and AML cells. POS derived from AML patients at diagnosis (Dx), relapse (Rx) and remission (Rm) was compared to healthy donor MSCs in terms of their morphology, growth pattern, support of leukemia cell viability and cytokine profile. Compared to control MSCs, POS (Dx/Rx, Rm) demonstrated a reduced proliferation rate (35%), significantly slower expansion, enlarged cell area (3-4-fold) and provided preferential support to leukemic cells of the same individual. Cytokine profiling showed significantly higher secreted phosphoprotein-1 (SPP1) expression in Dx/Rx and Rm POS compared to healthy MSCs. Additionally, the angiopoietin-1 expression was elevated in Dx/Rx POS with a further increase in the AML cell presence. In conclusion, the fact that POS derived in active disease and remission exhibited similar morphological and functional characteristics, might imply the involvement of the BM niche in leukemogenesis.


Assuntos
Células da Medula Óssea/patologia , Medula Óssea/patologia , Carcinogênese/patologia , Células-Tronco Hematopoéticas/patologia , Leucemia Mieloide Aguda/patologia , Células-Tronco Mesenquimais/patologia , Adulto , Angiopoietina-1/biossíntese , Animais , Linhagem Celular , Proliferação de Células , Humanos , Camundongos , Osteopontina/metabolismo
7.
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
8.
Inhal Toxicol ; 30(4-5): 187-194, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30257148

RESUMO

OBJECTIVE: Angiopoietin-1 (Ang1) is reported to have the ability to attenuate endothelial permeability and inflammation during the stress condition and is considered to play a critical role in vascular stabilization. The aim of this study was to investigate the mechanisms involved in the protective effects of adenovirus-delivered Ang1 in phosgene-induced acute lung injury (ALI). METHODS: ALI was induced in rats by phosgene exposure at 8.33 g/m3 for 5 min, followed by an intravenous injection of adenovirus-Ang1 (Ad/Ang1). The histologic changes of the lung were evaluated with H&E staining. The levels of cytokines in the serum and bronchoalveolar lavage fluid (BALF) were determined by ELISA. NLRP3 inflammasome activation was assessed with immunohistochemistry, RT-PCR, Western blotting and TUNEL staining. RESULTS: Histologic analyses suggested that reduced severity in phosgene-induced ALI with Ad/Ang1 treatment. Reduced levels of IL-1ß, IL-18 and IL-33 were found in both serum and BALF samples from Ad/Ang1-treated ALI rats induced by phosgene. Moreover, immunohistochemistry analysis revealed that Ad/Ang1 treatment inhibited the NLRP3 inflammasome activation. Decreased mRNA and protein levels of NLRP3 and caspase-1 were found in phosgene-exposed rats treated with Ad/Ang1. In addition, TUNEL staining indicated a decrease in pyroptosis in phosgene-exposed rats treated with Ad/Ang1. CONCLUSIONS: Ang1 exerts beneficial effects on phosgene-induced lung injury via inhibition of NLRP3 inflammasome activation. Disruption of NLRP3 inflammasome activation might be served as therapeutic modality for the treatment of phosgene-induced ALI.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Adenoviridae/genética , Angiopoietina-1/biossíntese , Terapia Genética/métodos , Vetores Genéticos , Inflamassomos/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fosgênio/toxicidade , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/metabolismo , Angiopoietina-1/genética , Animais , Líquido da Lavagem Broncoalveolar/química , Citocinas/sangue , Inflamassomos/genética , Inflamassomos/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Piroptose/efeitos dos fármacos , Ratos Sprague-Dawley
9.
Connect Tissue Res ; 59(1): 12-20, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28165799

RESUMO

Bridging large bone defects with mesenchymal stromal cells-seeded scaffolds remains a big challenge in orthopedic surgery, due to the lack of vascularization. Within such a cell-scaffold construct, cells are exposed to ischemic conditions. When human mesenchymal stem cells (hMSCs) encounter hypoxic conditions, they show higher cell proliferation than at ambient oxygen levels. However, when hMSCs are exposed to prolonged ischemia, cell proliferation ceases completely. Exposure of hMSCs to hypoxic conditions is known to result in the transcription of angiogenic factors (AGF), which can promote the development of new blood vessels. In this study, we investigated at which oxygen level hMSC proliferation and the transcription of AGF were optimal. Human bone marrow-derived hMSCs were cultured at 0.1, 1, 2, 3, 4, 5, and 21% oxygen. Cell proliferation over 14 days was assayed using a DNA quantification method. hMSC metabolic activity over 14 days was measured using a MTT test. Quantitative RT-PCR was used to assess mRNA levels of angiogenic factors at the tested oxygen percentages. hMSCs showed the highest cell proliferation rate at 1% oxygen. The highest corrected cell metabolic rate was found at 21% oxygen, followed by 2% oxygen. HIF1α transcription did not increase under hypoxic conditions compared to 21% oxygen conditions. However, transcription of VEGF and ANG-1 was significantly higher at 2% oxygen than at 21% O2. The optimum oxygen range at which hMSCs proliferated rapidly and angiogenic factors ANG-1 and VEGF simultaneously came to expression was from 1 to 2% oxygen.


Assuntos
Angiopoietina-1/biossíntese , Células da Medula Óssea/metabolismo , Proliferação de Células , Células-Tronco Mesenquimais/metabolismo , Transcrição Gênica , Fator A de Crescimento do Endotélio Vascular/biossíntese , Idoso , Idoso de 80 Anos ou mais , Células da Medula Óssea/citologia , Hipóxia Celular , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Mesenquimais/citologia , Pessoa de Meia-Idade
10.
Ann Vasc Surg ; 48: 214-221, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29197611

RESUMO

BACKGROUND: To investigate whether angiopoietin-1 (Ang-1) could regulate the endothelial progenitor cells (EPCs) survival and the effect of accelerating intra-aneurysmal organization and occlusion of the aneurysm neck. METHODS: EPCs were isolated from Wistar rats. EPCs were cultured and transfected with lentivirus-Ang-1-endothelial progenitor cells (Ang-1-EPCs) and lentivirus-NC-endothelial progenitor cells (NC-EPCs). The effects of Ang-1 on viability and functioning of EPCs were explored via tube formation, migration, and MTT (3-[4,5-dimethylthiazolyl-2]-2,5-diphenyltetrazolium bromide) assays. Eighteen Wistar rats were randomly allocated into 3 groups. Eighteen bare coils were inserted into the ligated external carotid artery (ECA) sacs of rats. The ECA sacs were removed 2 weeks after the coils were implanted and examined by histology assay. RESULTS: Ang-1 significantly promoted EPCs tube formation, migration, and proliferation ability in vitro. Histology analyses revealed that the organized areas in the ECA sacs in the Ang-1-EPCs group are higher than NC-EPCs group and control group at 2 weeks. Immunofluorescence revealed that organized tissues were characterized by an accumulation of cells positive for α-smooth muscle actin-positive cells in aneurysm sacs. CONCLUSIONS: Overexpression of Ang-1 enhanced the tube formation, migration, and proliferation ability of EPCs. Ang-1 gene-modified EPCs accelerated organization within the aneurysms and occlusion of aneurysm neck. Transplantation of Ang-1-transfected EPCs may be a new method for the treatment of aneurysm.


Assuntos
Aneurisma/cirurgia , Angiopoietina-1/biossíntese , Doenças das Artérias Carótidas/cirurgia , Artéria Carótida Externa/metabolismo , Células Progenitoras Endoteliais/transplante , Neovascularização Fisiológica , Actinas/metabolismo , Aneurisma/metabolismo , Aneurisma/patologia , Angiopoietina-1/genética , Animais , Doenças das Artérias Carótidas/metabolismo , Doenças das Artérias Carótidas/patologia , Artéria Carótida Externa/patologia , Artéria Carótida Externa/cirurgia , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Células Progenitoras Endoteliais/metabolismo , Ligadura , Ratos Wistar , Transdução de Sinais , Fatores de Tempo , Regulação para Cima
11.
J Cell Biochem ; 119(1): 524-535, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28608561

RESUMO

This experiment investigated the impact of serum from patients with type 2 diabetes mellitus on the angiogenic behavior of human mesenchymal stem cells in vitro. Changes in the level of Ang-1, Ang-2, cell migration, and trans-differentiation into pericytes and endothelial lineage were monitored after 7 days. The interaction of mesenchymal stem cells with endothelial cells were evaluated using surface plasmon resonance technique. Paracrine restorative effect of diabetic stem cells was tested on pancreatic ß cells. Compared to data from FBS and normal serum, diabetic serum reduced the stem cell survival and chemotaxis toward VEGF and SDF-1α (P < 0.05). Diabetic condition were found to decline cell migration rate and the activity of MMP-2 and -9 (P < 0.05). The down-regulation of VEGFR-2 and CXCR-4 was observed with an increase in the level of miR-1-3p and miR-15b-5p at the same time. The paracrine angiogenic potential of diabetic stem cells was disturbed via the changes in the dynamic of Ang-1, Ang-2, and VEGF. Surface plasmon resonance analysis showed that diabetes could induce an aberrant increase in the interaction of stem cells with endothelial cells. After treatment with diabetic serum, the expression of VE-cadherin and NG2 and ability for uptake of Dil-Ac-LDL were reduced (P < 0.01). Conditioned media prepared from diabetic stem cells were unable to decrease fatty acid accumulation in ß-cells (P < 0.05). The level of insulin secreted by ß-cells was not affected after exposure to supernatant from diabetic or non-diabetic mesenchymal stem cells. Data suggest diabetes could decrease angiogenic and restorative effect of stem cells in vitro. J. Cell. Biochem. 119: 524-535, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Diabetes Mellitus Tipo 2/sangue , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica , Soro , Idoso , Angiopoietina-1/biossíntese , Angiopoietina-2/biossíntese , Humanos , Masculino , Metaloproteinase 2 da Matriz/biossíntese , Pessoa de Meia-Idade , Receptores CXCR4/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese
12.
J Thromb Haemost ; 15(12): 2408-2418, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28981191

RESUMO

Essentials Platelets express retinoic acid receptor (RAR)α protein, specifically binding target mRNAs. mRNAs under RARα control include MAP1LC3B2, SLAIN2, and ANGPT1. All-trans retinoic acid (atRA) releases RARα from its target mRNA. RARα expressed in human platelets exerts translational control via direct mRNA binding. SUMMARY: Background Translational control mechanisms in platelets are incompletely defined. Here, we determined whether the nuclear transcription factor RARα controls protein translational events in human platelets. Methods Isolated human platelets were treated with the pan-RAR agonist all-trans-retinoic acid (atRA). Global and targeted translational events were examined. Results Stimulation of platelets with atRA significantly increased global protein expression. RARα protein bound to a subset of platelet mRNAs, as measured by next-generation RNA-sequencing. In-depth analyses of 5' and 3'-untranslated regions of the RARα-bound mRNAs revealed consensus RARα binding sites in microtubule-associated protein 1 light chain 3 beta 2 (MAP1LC3B2), SLAIN motif-containing protein 2 (SLAIN2) and angiopoietin-1 (ANGPT1) transcripts. When platelets were treated with atRA, binding interactions between RARα protein and mRNA for MAP1LC3B2, SLAIN2 and ANGPT1 were significantly decreased. Consistent with the release of bound RARα protein from MAP1LCB2mRNA, we observed an increase in the synthesis of MAP1LC3B2 protein. Conclusions These findings provide the first evidence that RARα, a nuclear transcriptional factor, regulates synthetic events in anucleate human platelets. They also reveal an additional non-genomic role for RARα in platelets that may have implications for the vitamin A-dependent signaling in humans.


Assuntos
Plaquetas/metabolismo , Proteínas Sanguíneas/biossíntese , Proteínas Sanguíneas/genética , Receptor alfa de Ácido Retinoico/sangue , Angiopoietina-1/biossíntese , Angiopoietina-1/sangue , Angiopoietina-1/genética , Sequência de Bases , Sítios de Ligação/genética , Plaquetas/efeitos dos fármacos , Humanos , Técnicas In Vitro , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/sangue , Proteínas Associadas aos Microtúbulos/genética , Biossíntese de Proteínas , RNA Mensageiro/sangue , RNA Mensageiro/genética , Receptor alfa de Ácido Retinoico/genética , Tretinoína/farmacologia
13.
Ann Hematol ; 96(12): 2103-2112, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28956132

RESUMO

As a site of complicated interactions among cytokines, bone marrow niche has been the subject of many scientific studies, mainly in the context of the proteins influencing damage or recovery of endothelium after allogeneic hematopoietic stem cell transplantation (HSCT). In this study, we aimed at exploring mutual correlations of bone marrow niche cytokines involved in the homing and mobilization of hematopoietic stem cells, as well as in angiogenesis. The aim of our study was to evaluate levels of cytokines: VEGF, angiopoietin-1 (ANGPT1), angiopoietin-2 (ANGPT2), and matrix metalloproteinase 9 (MMP-9) during autologous HSCT and to examine their influence on hematological recovery. Forty-three patients with hematological malignancies (33 multiple myeloma, 10 lymphoma) were enrolled in the study. Plasma samples were taken at five time points: before conditioning treatment (BC), on transplantation day (0) and 7 (+7), 14 (+14), and 21 (+21) days after HSCT. The cytokine levels were evaluated by ELISA method. Our study revealed decreased levels of VEGF, ANGPT1, and MMP-9 in the early post-transplant period as compared to the baseline (BC). ANGPT2 was decreased after conditioning treatment, but tended to increase from day +7. On day +7, positive correlations between ANGPT1 level as well as MMP-9 and the time to engraftment were observed. As opposite to ANGPT1, negative correlation between ANGPT2 level on day +7 after HSCT and the time to hematological recovery was noticed. Our study suggests that investigated cytokines are an important part of bone marrow environment and significantly influence the time to engraftment after HSCT.


Assuntos
Angiopoietina-1/biossíntese , Angiopoietina-2/biossíntese , Regulação Neoplásica da Expressão Gênica , Neoplasias Hematológicas , Transplante de Células-Tronco Hematopoéticas , Linfoma , Metaloproteinase 9 da Matriz/biossíntese , Mieloma Múltiplo , Proteínas de Neoplasias/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Adulto , Idoso , Feminino , Neoplasias Hematológicas/sangue , Neoplasias Hematológicas/terapia , Humanos , Linfoma/sangue , Linfoma/terapia , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/sangue , Mieloma Múltiplo/terapia
14.
ACS Synth Biol ; 6(11): 2003-2013, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-28793186

RESUMO

The precise spatial and temporal control of gene expression, cell differentiation, and tissue morphogenesis has widespread application in regenerative medicine and the study of tissue development. In this work, we applied optogenetics to control cell differentiation and new tissue formation. Specifically, we engineered an optogenetic "on" switch that provides permanent transgene expression following a transient dose of blue light illumination. To demonstrate its utility in controlling cell differentiation and reprogramming, we incorporated an engineered form of the master myogenic factor MyoD into this system in multipotent cells. Illumination of cells with blue light activated myogenic differentiation, including upregulation of myogenic markers and fusion into multinucleated myotubes. Cell differentiation was spatially patterned by illumination of cell cultures through a photomask. To demonstrate the application of the system to controlling in vivo tissue development, the light inducible switch was used to control the expression of VEGF and angiopoietin-1, which induced angiogenic sprouting in a mouse dorsal window chamber model. Live intravital microscopy showed illumination-dependent increases in blood-perfused microvasculature. This optogenetic switch is broadly useful for applications in which sustained and patterned gene expression is desired following transient induction, including tissue engineering, gene therapy, synthetic biology, and fundamental studies of morphogenesis.


Assuntos
Angiopoietina-1 , Diferenciação Celular , Regulação da Expressão Gênica , Proteína MyoD , Optogenética/métodos , Fator A de Crescimento do Endotélio Vascular , Angiopoietina-1/biossíntese , Angiopoietina-1/genética , Animais , Linhagem Celular , Camundongos , Proteína MyoD/genética , Proteína MyoD/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
15.
Tumour Biol ; 39(7): 1010428317711658, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28720059

RESUMO

Metastatic or recurrent cervical cancer has limited treatment options and a high rate of mortality. Although anti-vascular endothelial growth factor drugs have shown great promise as a therapeutic target for treatment of advanced cervical cancer, drug resistance and class-specific side effects negate long-term benefits. The identification of alternative anti-angiogenic factors will be critical for future drug development for advanced or recurrent cervical cancer. In this study, we found that angiopoietins and Tie receptors were highly expressed in cervical cancer cells. Tie-2 expression in tumor cells predicted poorer prognosis. Wound closure assay and Transwell assay showed that upregulated or downregulated Ang-1 and Ang-2 expression promoted or reduced cervical cancer cell lines migration and invasion, respectively. In subcutaneous xenograft models of cervical cancer, downregulation of Ang-1 and Ang-2 attenuated tumor growth. The expression of vimentin and endomucin and microvessel density were all significantly decreased in the siAng-1 group and siAng-2 group relative to the infection control group. Our data support that dual inhibition of Ang-1 and Ang-2 may be an alternative target for anti-angiogenic adjuvant therapy in advanced or recurrent cervical squamous cell cancer.


Assuntos
Angiopoietina-1/genética , Receptor TIE-2/genética , Neoplasias do Colo do Útero/genética , Proteínas de Transporte Vesicular/genética , Adulto , Idoso , Angiopoietina-1/biossíntese , Animais , Carcinogênese/genética , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Células HeLa , Humanos , Camundongos , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Prognóstico , Receptor TIE-2/biossíntese , Neoplasias do Colo do Útero/patologia , Proteínas de Transporte Vesicular/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
16.
PLoS One ; 12(4): e0174555, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28369068

RESUMO

Esophageal squamous cell carcinoma (ESCC) is the second common cancer in Henan province and is well-known for aggressiveness and dismal prognosis. Adjuvant therapies, chemotherapy, radiotherapy and endoscopic treatment have not improved survival rates in patients with late stage esophageal carcinoma. All-trans retinoic acid (ATRA) is the active ingredient of Vitamin A and affects a wide spectrum of biological processes including development, growth, neural function, immune function, reproduction, and vision. It is one of the most potent therapeutic agents used for treating cancers, especially lung adenocarcinomas. ATRA inhibits metastatic potential and angiogenesis in several tumor models. We investigated the effects of ATRA on the expression of angiopoietin 1 (Ang-1), angiopoietin 2 (Ang-2) and receptor Tie-2 in EC1 cells in vitro. We also assessed the growth and migration of EC1 cells in vitro. ATRA treatment caused 29.5% and 40.3% reduction of the growth of EC1 cells after 24 hours and 48 hours, relative to the control. ATRA plus fluorouracil treatment reduced the viability more strongly than either drug alone, indicating an additive effect. Moreover, ATRA decreased EC1 migration by 87%. Furthermore, ATRA treatment led to a marked decrease of the transcript levels of Ang-1, Ang-2, Tie-2, VEGF, and VEGF receptors, as assessed by real-time RT-PCR. Importantly, the protein levels of Ang-1, Ang-2 and Tie-2 were reduced by ATRA treatment. In vivo, we found ATRA treatment suppressed the tumor growth and improved the cachexia of mice. Importantly, ATRA treatment decreased the expression of CD31, Ang-1, Ang-2 and Tie-2 in subcutaneous tumors of EC1 cells. Collectively, our findings demonstrate that ATRA exhibits a dose- and temporal-dependent effect on the metastatic behavior, suppresses the angiopoietin-Tie2 pathway and inhibits angiogenesis and the progression of xenograft tumors of EC1 cells.


Assuntos
Angiopoietina-1/biossíntese , Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias Esofágicas/tratamento farmacológico , Metástase Neoplásica/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Receptor TIE-2/biossíntese , Tretinoína/farmacologia , Proteínas de Transporte Vesicular/biossíntese , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Carcinoma de Células Escamosas do Esôfago , Feminino , Fluoruracila/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Receptores de Fatores de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese
17.
Neurosci Lett ; 644: 62-66, 2017 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-28219791

RESUMO

Our previous studies have found that stem cells conditioned medium (CM) facilitated functional recovery after stroke in non-diabetics. However, whether bone marrow stromal cells conditioned medium (BMSCs-CM) treatment after stroke in type 2 diabetic (T2DM) rats improve functional outcomes remains unclear. T2DM rats were induced and subjected to stroke then treated with or without BMSCs-CM. Functional outcomes and blood-brain barrier (BBB) leakage were performed, the expression of Angiopoietin (Ang) 1 and tyrosine kinase (Tie) 2 were also assessed. Our results showed that BMSCs-CM treatment significantly improved functional outcomes, decreased BBB leakage and the expression of Ang1 and Tie2 were also changed after BMSCs-CM treatment in type 2 diabetes after stroke. In conclusion, enhanced expression of Ang1 and Tie2 in ischemic brain after BMSCs-CM treatment of stroke may contribute to the improved functional recovery after stroke in type 2 diabetic rats.


Assuntos
Diabetes Mellitus Experimental/patologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Recuperação de Função Fisiológica , Acidente Vascular Cerebral/patologia , Remodelação Vascular , Angiopoietina-1/biossíntese , Animais , Células da Medula Óssea , Encéfalo/metabolismo , Encéfalo/patologia , Meios de Cultivo Condicionados/farmacologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2 , Masculino , Transplante de Células-Tronco Mesenquimais , Ratos , Ratos Wistar , Receptor TIE-2/biossíntese , Recuperação de Função Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/metabolismo
18.
Leuk Res ; 55: 49-54, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28122283

RESUMO

Angiopoietins play an important role in vascular endothelial function. Endothelial damage is an important pathogenesis relating with acute graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT), protecting endothelial cells (ECs) from damage may be a potent prophylaxis and therapeutic strategy of acute GVHD (aGVHD). In this study, we explored changes in Angiopoietin-1 (Ang-1) and Ang-2 expression in a aGVHD mouse model and determined whether simvastatin prevents GVHD through regulating Ang-1 and Ang-2 expression. In vitro simvastatin administration increased Ang-1 production and release but conversely inhibited Ang-2 release from EA.hy926 ECs. Simvastatin improved the survival of aGVHD mice, attenuated the histopathological GVHD grades and plasma levels of Ang-2, and elevated the plasma levels of Ang-1 as well as the aortic endothelial levels of Ang-1 and Ang-2. In summary, simvastatin represents a novel approach to combat GVHD by increasing Ang-1 production while suppressing Ang-2 release to stabilize endothelial cells.


Assuntos
Doença Enxerto-Hospedeiro/prevenção & controle , Sinvastatina/farmacologia , Angiopoietina-1/análise , Angiopoietina-1/biossíntese , Angiopoietina-1/sangue , Angiopoietina-2/análise , Angiopoietina-2/biossíntese , Angiopoietina-2/sangue , Animais , Aorta/citologia , Modelos Animais de Doenças , Células Endoteliais/química , Células Endoteliais/patologia , Expressão Gênica/efeitos dos fármacos , Doença Enxerto-Hospedeiro/tratamento farmacológico , Camundongos , Sinvastatina/uso terapêutico
19.
Cardiovasc Res ; 113(1): 81-89, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28069704

RESUMO

AIMS: Atherosclerosis is a chronic inflammatory disease occurring within the artery wall. A crucial step in atherogenesis is the infiltration and retention of monocytes into the subendothelial space of large arteries induced by chemokines and growth factors. Angiopoietin-1 (Ang-1) regulates angiogenesis and reduces vascular permeability and has also been reported to promote monocyte migration in vitro. We investigated the role of Ang-1 in atherosclerosis-prone apolipoprotein-E (Apo-E) knockout mouse. METHODS AND RESULTS: Apo-E knockout (Apo-E-/-) mice fed a western or normal chow diet received a single iv injection of adenovirus encoding Ang-1 or control vector. Adenovirus-mediated systemic expression of Ang-1 induced a significant increase in early atherosclerotic lesion size and monocyte/macrophage accumulation compared with control animals receiving empty vector. Ang-1 significantly increased plasma MCP-1 and VEGF levels as measured by ELISA. FACS analysis showed that Ang-1 selectively increased inflammatory Gr1+ monocytes in the circulation, while the cell-surface expression of CD11b, which mediates monocyte emigration, was significantly reduced. CONCLUSIONS: Ang-1 specifically increases circulating Gr1+ inflammatory monocytes and increases monocyte/macrophage retention in atherosclerotic plaques, thereby contributing to development of atherosclerosis.


Assuntos
Angiopoietina-1/biossíntese , Antígenos Ly/metabolismo , Aorta Torácica/metabolismo , Doenças da Aorta/metabolismo , Aterosclerose/metabolismo , Monócitos/metabolismo , Placa Aterosclerótica , Adenoviridae/genética , Angiopoietina-1/genética , Animais , Aorta Torácica/patologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Antígeno CD11b/sangue , Quimiocina CCL2/sangue , Dieta Hiperlipídica , Modelos Animais de Doenças , Predisposição Genética para Doença , Vetores Genéticos , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/patologia , Fenótipo , Transdução de Sinais , Técnicas de Cultura de Tecidos , Fator A de Crescimento do Endotélio Vascular/sangue
20.
Pol J Pathol ; 68(4): 326-329, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29517203

RESUMO

The aim of the study was to investigate how an intramuscular injection of plasmids with genes coding various pro-angiogenic factors: angiopoetin-1 (ANGPT1), vascular endothelial growth factor (VEGF165) and hepatic growth factor (HGF), influences the production of ANGPT1. 40 Healthy Fisher rats received i.m. injections containing plasmids encoding pro-angiogenic genes in thigh muscles. They were divided into four equal groups. The first group received the plANGPT1 plasmid and the second group- the pIRES/ANGPT1/VEGF165 bicistronic plasmid. The pIRES/VEGF165/HGF bicistronic plasmid was administered to the third group and an empty plasmid (control group) to the fourth group. The animals were euthanized after 12 weeks. In each group, the number of vessels stained with the anti-ANGPT1 antibody was assessed under an optical microscope. The anti-ANGPT1 antibodies stained the vessels in all the groups. There were on average 14.1 ±2.3 vessels in the the plANGPT1 group, 32.5 ±10.5 in the pl/RESANGPT1/VEGF group and 30.8 ±13.3 in the plRES/HGV/VEGF group. There were on average 7.3 ±2.3 stained vessels (p < 0.0001) in the control group . The VEGF plays a role in the induction of the production of ANGPT1. The administration of plasmids only encoding ANGPT1 does not induce its production.


Assuntos
Angiopoietina-1/biossíntese , Vasos Sanguíneos/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/biossíntese , Angiopoietina-1/genética , Animais , Técnicas de Transferência de Genes , Fator de Crescimento de Hepatócito/biossíntese , Fator de Crescimento de Hepatócito/genética , Humanos , Injeções Intramusculares , Ratos Endogâmicos F344 , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA