Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
2.
Am J Transl Res ; 11(5): 2877-2886, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31217861

RESUMO

Statins and therapeutic ultrasound (TUS) have been shown to ameliorate angiogenesis on ischemic hindlimb animals and promote human umbilical vein endothelial cells (HUVECs) tube formation and proliferation. Here, we evaluate the therapeutic effect of TUS in combination with atorvastatin (Ator) therapy on angiogenesis in hindlimb ischemia and HUVECs. After subjecting excision of the left femoral artery, all mice were randomly distributed to one of four groups: Control; Ator treated mice (Ator); TUS treated mice (TUS); and Ator plus TUS treated mice (Ator+TUS). At day 14 post-surgery, the Ator plus TUS treatment cohort had the greatest blood perfusion, accompanied by elevated capillary density. In vitro, Ator plus TUS augmented tube formation, migration and proliferative capacities of HUVECs. Additionally, the united administration upregulated expression of angiogenic factors phosphorylated Akt (p-Akt), phosphorylated endothelial nitric oxide synthase (p-eNOS), as well as vascular endothelial growth factor (VEGF), both in vivo and in vitro. These benefits could be blocked by either phosphoinositide 3-kinase (PI3K) or eNOS inhibitor. Our data indicated that the united administration could significantly enhance ischemia-mediated angiogenesis and exert a protective effect against ischemic/hypoxia induced damage among HUVECs through up-regulating VEGF expression and activating the PI3K-Akt-eNOS pathway.

3.
Am J Transl Res ; 10(7): 2068-2077, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30093944

RESUMO

Nanostructured calcium phosphate (CaP) and magnesium phosphate (MgP) are promising for the application as the nanocarriers in drug delivery. However, the difference between CaP and MgP nanocarriers in drug delivery is rarely investigated. In this work, we comparatively investigated nanostructured CaP, MgP and calcium magnesium phosphate (CMP) for the delivery of SRT1720, which is a silent information regulator (SIRT1) specific activator with pro-angiogenic and anti-aging properties in response to hydrogen peroxide (H2O2)-induced endothelial senescence. The protection of SRT1720-loaded CaP nanospheres, MgP nanosheets and CMP microspheres on the H2O2-induced senescent endothelium was examined by using human umbilical vein endothelial cells (HUVECs), demonstrating the improved cell viability, anti-aging, tube formation and migration. In addition, the SRT1720-loaded CaP nanospheres, MgP nanosheets and CMP microspheres can rescue the impaired angiogenic potential of HUVECs via activation of Akt/eNOS/VEGF pathway. The SRT1720-loaded MgP nanosheets and CMP microspheres have a similar protective effect compared with the pure SRT1720, while the SRT1720-loaded CaP nanospheres decrease the protective capability of SRT1720. These results lead us to figure out both MgP nanosheets and CMP microspheres are suitable and effective delivery for SRT1720 and this system can be further applied in vivo treatment.

5.
Am J Transl Res ; 9(9): 4184-4194, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28979692

RESUMO

The objective of this investigation was to evaluate the therapy effect of combined therapeutic ultrasound (TUS) treatment and pulsed electromagnetic field (PEMF) therapy on angiogenesis in hypertension-related hindlimb ischemia. After subjecting excision of the left femoral artery, spontaneously hypertensive rats (SHRs) were randomly distributed to one of four groups: SHR; TUS treated SHR (SHR-TUS); PEMF treated SHR (PEMF-TUS); and TUS plus PEMF treated SHR (SHR-TUS-PEMF). Wistar-Kyoto rats (WKYs) with femoral artery excision were regarded as a control group. At day 14 after surgery, the TUS plus PEMF united administration had the greatest blood perfusion accompanied by elevated capillary density and the lowest TUNEL index. Interestingly, the united administration up-regulated the angiogenic factors expression of phosphorylated Akt (p-Akt), phosphorylated endothelial nitric oxide synthase (p-eNOS), vascular endothelial growth factor (VEGF), anti-apoptotic protein of Bcl-2 and down-regulated pro-apoptotic protein levels of Bax and Cleaved caspase-3 in vivo. Our results demonstrated that the united administration could significantly rescue hypertension-related inhibition of ischemia-induced neovascularization partly by promoting angiogenesis and inhibiting apoptosis.

6.
Exp Ther Med ; 14(2): 1157-1162, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28810573

RESUMO

Patients with acute limb ischemia, deep venous thrombosis and pulmonary artery embolism may be treated with warfarin. The dose-response interaction of warfarin is associated with numerous factors, depending on which an uncommon life-threatening bleeding may occur. The present case study reported on a patient with acute limb ischemia and a history of warfarin-induced bleeding ten years previously and who again developed life threatening bleeding associated with warfarin treatment and received vascular surgery. In this patient, a cytochrome P450 3A4 loss-of-function mutation decreased the effective dose of warfarin. Although this was a rare case, clinicians should be alert to the bleeding risk associated with such rare genetic mutations.

7.
J Am Heart Assoc ; 6(7)2017 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-28710180

RESUMO

BACKGROUND: DPP4 (Dipeptidyl peptidase-4)-GLP-1 (glucagon-like peptide-1) and its receptor (GLP-1R) axis has been involved in several intracellular signaling pathways. The Adrß3 (ß3-adrenergic receptor)/CXCL12 (C-X-C motif chemokine 12) signal was required for the hematopoiesis. We investigated the novel molecular requirements between DPP4-GLP-1/GLP-1 and Adrß3/CXCL12 signals in bone marrow (BM) hematopoietic stem cell (HSC) activation in response to chronic stress. METHODS AND RESULTS: Male 8-week-old mice were subjected to 4-week intermittent restrain stress and orally treated with vehicle or the DPP4 inhibitor anagliptin (30 mg/kg per day). Control mice were left undisturbed. The stress increased the blood and brain DPP4 levels, the plasma epinephrine and norepinephrine levels, and the BM niche cell Adrß3 expression, and it decreased the plasma GLP-1 levels and the brain GLP-1R and BM CXCL12 expressions. These changes were reversed by DPP4 inhibition. The stress activated BM sca-1highc-KithighCD48lowCD150high HSC proliferation, giving rise to high levels of blood leukocytes and monocytes. The stress-activated HSC proliferation was reversed by DPP4 depletion and by GLP-1R activation. Finally, the selective pharmacological blocking of Adrß3 mitigated HSC activation, accompanied by an improvement of CXCL12 gene expression in BM niche cells in response to chronic stress. CONCLUSIONS: These findings suggest that DPP4 can regulate chronic stress-induced BM HSC activation and inflammatory cell production via an Adrß3/CXCL12-dependent mechanism that is mediated by the GLP-1/GLP-1R axis, suggesting that the DPP4 inhibition or the GLP-1R stimulation may have applications for treating inflammatory diseases.


Assuntos
Encéfalo/enzimologia , Diferenciação Celular , Proliferação de Células , Dipeptidil Peptidase 4/metabolismo , Células-Tronco Hematopoéticas/enzimologia , Estresse Psicológico/enzimologia , Antagonistas de Receptores Adrenérgicos beta 3/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Doença Crônica , Dipeptidil Peptidase 4/deficiência , Dipeptidil Peptidase 4/genética , Inibidores da Dipeptidil Peptidase IV/farmacologia , Modelos Animais de Doenças , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Ratos Endogâmicos F344 , Ratos Transgênicos , Receptores Adrenérgicos beta 3/metabolismo , Restrição Física/psicologia , Transdução de Sinais , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/fisiopatologia , Estresse Psicológico/psicologia , Fatores de Tempo
8.
Aging Dis ; 8(3): 287-300, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28580185

RESUMO

The mechanism by which angiogenesis declines with aging is not fully understood. Soluble vascular endothelial growth factor receptor 1 (VEGFR1) form (sFlt1) contributes to endothelial dysfunction in pathological conditions. However, the roles of sFlt1 in ischemia-induced neovascularizationof aged animals have not been investigated. To study aging-related sFlt1 change and its impact on ischemia-induced neovascularization, a hindlimb ischemia model was applied to young and aged mice. Blood flow imaging assay revealed that the blood flow recovery remained impaired throughout the follow-up period. At day 14, immunostaining showed lesser capillary formation in the aged mice. An ELISA showed that the aged mice had increased plasma sFlt-1 levels at indicated time points after surgery. On operative day 4, the aged ischemic muscles had decreased levels of p-VEGFR2 and p-Akt and increased levels of sFlt-1, Wnt5a, and SC35 genes or/and protein as well as increased numbers of inflammatory cells (macrophages and leucocytes) and matrix metalloproteinase-9 activity. Immnunofluorescence showed that Flt-1 was co-localized with CD11b+ macrophages of aged ischemic muscles. Hypoxia stimulated sFlt1 expression in CD11b+ cells of aged bone-marrow (BM), and this effect was diminished by siWnt5a. The cultured medium of aged mice BM-derived CD11b+ cells suppressed human endothelial cell (EC) and endothelial progenitor cell (EPC) angiogenic actions induced by VEGF, and these decreases were improved by treatment with siWnt5a-conditioned medium. Thus, aging appears to decline neovascularization in response to ischemic stress via the VEGFR2/Akt signaling inactivation in ECs and ECPs that is mediated by Wnt5a/SC35 axis activated macrophages-derived sFlt1 production in advanced age.

9.
Am J Transl Res ; 9(4): 1990-1999, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28469805

RESUMO

Background: Previous studies have demonstrated that therapeutic ultrasound (TUS) ameliorates angiogenesis on ischemic hind limb animals and also promotes human umbilical vein endothelial cells (HUVECs) tube formation. Apoptosis plays a key role in post-ischemic angiogenesis pathogenesis. However, the mechanisms underlying the anti-apoptotic effects of TUS are not clear. Therefore we put forward the hypothesis that TUS might promote angiogenesis during ischemia/hypoxia (I/H) by decreasing apoptosis. Methods: We investigated the cytoprotective role of TUS and the underlying mechanisms in I/H-induced HUVEC apoptosis. HUVECs were treated under hypoxic serum-starved conditions for 36 h and then treated with or without TUS (9 minutes, 1 MHz, 0.3 W/cm2). The cell viability was examined by the CCK-8 assay, apoptosis cell rate was determined by TUNEL staining and flow cytometry assay. In addition, the mitochondrial-dependent apoptosis pathway was evaluated by the protein activity of Bax, Bcl-2 and Caspase-3. Results: 1) apoptosis could be induced by I/H in HUVECs. 2) TUS attenuates HUVECs cell apoptosis induced by I/H. 3) TUS inhibits the protein expression of apoptosis modulators and effectors that regulate the mitochondrial pathway of apoptosis in HUVECs. 4) TUS increases the phosphorylation of Akt, which demonstrates the activation of the phosphoinositide 3-kinase (PI3K)- serine/threonine kinase (Akt) signal pathway. Conclusions: The present study indicates that exposure to TUS exerts a protective effect against I/H-induced apoptosis among HUVECs and that this process is mediated through the mitochondrial-dependent intrinsic apoptotic pathway. We also confirm that the PI3K-Akt signal cascade may be taken part in the TUS effects on apoptosis.

10.
Am J Transl Res ; 8(9): 3666-3677, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27725849

RESUMO

Therapeutic ultrasound (TUS) has been demonstrated to improve endothelial nitric oxide synthase (eNOS) activity, which played a crucial role in the regulation of angiogenesis. Diabetes Mellitus (DM) impairs eNOS activity. We tested the hypothesis that DM may retard unilateral hindlimb ischemia-induced angiogenesis by inhibiting eNOS in high-fat diet (HFD)/streptozocin (STZ) induced diabetic mice, and that TUS may reverse DM-related impairment of angiogenesis. C57BL/6 mice were allocated to four groups: (A) mice were fed standard diet (control); (B) mice were fed standard diet and treated with TUS (control+TUS); (C) type-2 DM mice were induced by HFD/STZ (diabetic); and (D) type-2 DM mice and treated with TUS (dabetic+TUS). All mice were surgically induced unilateral limb ischemia. The ischemic skeletal muscles in groups B and D were irradiated with extracorporeal TUS for 9 minutes/day (frequency of 1 MHz, intensity of 0.3 W/cm2) for 14 consecutive days. The result showed that TUS augmented the blood perfusion, increased capillary density accompanied by an upregulation of angiogenic factors and a downregulation of apoptotic proteins in group D relative to group C. In vitro, TUS inhibited the apoptosis, promoted tubule formation, proliferation and migration capacities, increased angiogenic factors expression and reduced apoptotic protein levels in human umbilical vein endothelial cells (HUVECs). Furthermore, TUS can robust reverse the inhibiting effect induced by high glucose (HG) on HUVECs, and these benefits could be blocked by phosphoinositide 3-kinase (PI3K) inhibitor (LY294002) or eNOS inhibitor (L-NAME). Together, TUS restored type-2 DM-mediated inhibition of ischemia-induced angiogenesis, partially via PI3K-Akt-eNOS signal pathway.

11.
Am J Transl Res ; 8(7): 2876-88, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27508009

RESUMO

Silent information regulator 1 (SIRT1) plays a critical role in maintaining vascular homeostasis via modulating senescent-related signal pathway, however, the molecular mechanism remains modest clarified. The purpose of this study was to examine whether SIRT1 specific activator SRT1720 would exhibit pro-angiogenic and anti-aging properties in response to hydrogen peroxide (H2O2)-induced endothelial senescence, and determine the underlying mechanisms. We pre-treated senescent human umbilical vein endothelial cells (HUVECs) with SRT1720, senescence-associated beta-galactosidase activity, apoptosis, migration, tube formation, proliferation and angiogenic factors were quantitatively examined. The results revealed that pharmacologic activation of SIRT1 by SRT1720 rescued apoptotic HUVECs and upregulated angiogenic response through reinforcing the protein expressions of angiogenic and survival factors in vitro. Furthermore, we confirmed that the expressions of endothelial nitric oxide synthase (eNOS), vascular endothelial growth factor (VEGF) and phosphoryl-Akt were augmented in SRT1720-treated senescent HUVECs. In conclusion, our data indicated that SRT1720 could protect against endothelial senescence and maintain cell function via Akt/eNOS/VEGF axis.

12.
Am J Transl Res ; 8(7): 3087-96, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27508029

RESUMO

We examined the hypothesis that therapeutic ultrasound (TUS) treatment would rescue the hypertension-related inhibition of ischemia-induced angiogenesis. TUS protects against endothelial dysfunction, but it is little known that the effect of TUS treatment on angiogenesis inhibited by hypertension. 20-week-old male spontaneously hypertensive rats (SHRs) and Wistar-Kyoto rats (WKYs) were randomly allocated to 4 groups: SHR; TUS treated SHR (SHR-TUS); WKY and TUS treated WKY (WKY-TUS). After undergoing excision of the left femoral artery, the ischemic skeletal muscles were treated with extracorporeal TUS for 9 minutes of daily exposure (frequency of 1 MHz, intensity of 0.3 W/cm(2)) for 14 consecutive days. We found that TUS normalized the blood perfusion in SHR-TUS accompanied by elevated capillary density. Similar results were found in the protein expression of angiogenic factors. TUS treatment also enhanced peripheral capillary density in WKY rats and restored the capillary rarefaction in hypertension by elevating the protein levels of endothelial nitric oxide synthase (eNOS), hypoxic inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF) and phosphorylated Akt (p-Akt) in vivo. Our data demonstrated that TUS treatment ameliorated hypertension-related inhibition of ischemia-induced angiogenesis, at least in part, via an NO-dependent manner.

13.
Psychoneuroendocrinology ; 73: 186-195, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27509090

RESUMO

BACKGROUND: Stress evokes lipolytic release of free fatty acid (FFA) and low-grade inflammation in visceral adipose tissue, mediated by increased adipokine secretion, and contributes to glucose metabolism disorder and prothrombotic state. We tested the hypothesis that alogliptin, a dipeptidyl peptidase-4 inhibitor, can ameliorate the biological effects of chronic stress in mice. METHOD AND RESULTS: C57BL/6J mice were subjected to 2-week intermittent restraint stress and orally treated with vehicle or alogliptin (dose: 15 or 45mg/kg/day). Plasma levels of lipids, proinflammatory cytokines (monocyte chemoattractant protein-1, tumor necrosis factor-α, and interleukin-6), and 8-hydroxydeoxyguanosine were measured with enzyme-linked immunosorbent assay. Monocyte/macrophage accumulation in inguinal white adipose tissue (WAT) was examined by CD11b-positive cell count and mRNA expression of CD68 and F4/80 was examined by immunohistochemistry and RT-PCR, respectively. The mRNA levels of the above-mentioned proinflammatory cytokines, NADPH oxidase 4, adiponectin, and coagulation factors (plasminogen activation inhibitor-1 and tissue factor) in WAT were also assessed with RT-PCR. Glucose metabolism was assessed by glucose and insulin tolerance tests, plasma levels of DPP-4 activity, glucagon-like peptide-1, expression of DPP-4, insulin receptor substrate-1 and glucose transporter 4 in WAT and skeletal muscle. Alogliptin administration suppressed stress-induced FFA release, oxidative stress, adipose tissue inflammation, DPP-4 activation, and prothrombotic state in a dose-dependent manner, and improved insulin sensitivity in stressed mice. CONCLUSIONS: The results indicate that alogliptin improves stress-induced prothrombotic state and insulin resistance; suggesting that alogliptin could have beneficial therapeutic effects against cardiovascular complications in diabetic patients under stress.


Assuntos
Tecido Adiposo/metabolismo , Dipeptidil Peptidase 4/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Resistência à Insulina , Piperidinas/farmacologia , Estresse Psicológico/complicações , Trombofilia/tratamento farmacológico , Uracila/análogos & derivados , Animais , Modelos Animais de Doenças , Inflamação/sangue , Inflamação/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Trombofilia/etiologia , Uracila/farmacologia
15.
Am J Transl Res ; 7(6): 1106-15, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26279754

RESUMO

Therapeutic angiogenic effects of low-intensity ultrasound have been reported in endothelial cells and animal models of hind limb ischemia. It has been shown that the proliferation, migration, and tube formation of endothelial cells play critical roles in angiogenesis. The purpose of this study was to determine the underlying mechanism of low-intensity continuous therapeutic ultrasound on angiogenesis in endothelial cells. In the present study, human umbilical vein endothelial cells (HUVECs) were simulated of low-intensity therapeutic ultrasound (TUS, 1 MHz, 0.3 W/cm(2), 9 minute per day) for 3 days, and we observed migration, tube formation, and expression of endothelial nitric oxide synthase (eNOS) and serine/threonine kinase (Akt) in HUVECs. Specific inhibitors of eNOS and phosphoinositide 3-kinase (PI3K) were added to the culture medium and TUS-induced changes in the pathways that mediate angiogenesis were investigated. After exposure to TUS, HUVECs tube formation and migration were significantly promoted, which was blocked by the eNOS inhibitor Immunofluorescence assay and Western blotting analysis demonstrated that eNOS expression in the HUVECs was significantly increased after TUS exhibition. Proteins of phosphorylated eNOS and Akt were both up-regulated after TUS stimulation. However, the specific inhibitor of PI3K not only significantly decreased the expression of p-Akt, but also down-regulated the p-eNOS. This suggested that the PI3K/Akt signal pathway might participate in modulating the activity of eNOS. In short, TUS therapy promotes angiogenesis through activation of the PI3K-Akt-eNOS signal cascade in HUVECs.

16.
Am J Transl Res ; 7(3): 430-44, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26045885

RESUMO

Pulsed electromagnetic fields (PEMF) have been shown to promote proliferation and regeneration in the damaged tissue. Here, we examined whether PEMF therapy improved postnatal neovascularization using murine model of hindlimb ischemia, and the underlying cellular/molecular mechanisms were further investigated. Hindlimb ischemia was induced by unilateral femoral artery resection using 6-8 week-old male C57BL6 mice. Then, mice were exposed to extracorporeal PEMF therapy (4 cycles, 8min/cycle, 30 ± 3 Hz, 5 mT) every day until day 14. Our data demonstrated that PEMF therapy significantly accelerated wound healing, decreased prevalence of gangrene and increased postnatal neovascularization. Moreover, the levels of vascular endothelial growth factor (VEGF), endothelial nitric oxide synthase (eNOS) and Akt phosphorylation in ischemic muscles were markedly enhanced following PEMF therapy. In vitro, PEMF inhibited the process of hypoxia-induced apoptosis and augmented tube formation, migration and proliferative capacities of human umbilical vein endothelial cells (HUVECs). Additionally, PEMF exposure increased VEGF secretion, as well as the eNOS and Akt phosphorylation, and these benefits could be blocked by either phosphoinositide 3-kinase (PI3K) or eNOS inhibitor. In conclusion, our data indicated that PEMF therapy enhanced ischemia-mediated angiogenesis, through up-regulating VEGF expression and activating the PI3K-Akt-eNOS pathway. Therefore, PEMF should be a valuable treatment for the patients with critical limb ischemia.

17.
Int J Cardiol ; 183: 198-208, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25668148

RESUMO

BACKGROUND: Evidence from human and animal studies has demonstrated elevated levels of the cysteine protease cathepsin S (CatS) in hypoxic atherosclerotic lesions. We hypothesized that silencing of CatS gene would suppress ischemia-induced angiogenic action. METHODS AND RESULTS: Left femoral artery ligation-induced ischemia in mice showed the increased expression and activity of CatS in the ischemic muscle. The CatS-deficiency (CatS(-/-)) mice showed impaired functional recovery following hindlimb ischemia and reduced levels of peroxisome proliferator-activated receptor-γ (PPAR-γ), phospho-Akt (p-Akt), p-endothelial nitric oxide synthase, p-extracellular signal-regulated kinase1/2 (Erk1/2), p-p38 mitogen-activated protein kinase, and vascular endothelial growth factor (VEGF) proteins, as well as reduced levels of matrix metalloproteinase-9 and macrophage infiltration in the ischemic muscles. In vitro, CatS silencing reduced the levels of these targeted essential molecules for angiogenesis and vasculogenesis. Together, the results indicated that the effects of CatS knockdown led to defective endothelial cell invasion, proliferation, and tube formation. This notion was reinforced by the finding that CatS inhibition led to a decreased PPAR-γ level and VEGF/Erk1/2 signaling activation in response to ischemia. CatS(-/-) resulted in decreased circulating EPC-like CD31(+)/c-Kit(+) cells, accompanied by the reduction of the cellular levels of PPAR-γ, p-Akt, and VEGF induced by ischemic stress. Transplantation of bone-marrow-derived mononuclear cells from CatS(+/+) mice restored neovascularization in CatS(-/-) mice. CONCLUSIONS: CatS activity controls ischemia-induced neovascularization partially via the modulation of PPAR-γ and VEGF/Akt signaling activation.


Assuntos
Catepsinas/metabolismo , Isquemia/metabolismo , Músculo Esquelético/irrigação sanguínea , Animais , Células Endoteliais/citologia , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Artéria Femoral/citologia , Artéria Femoral/enzimologia , Artéria Femoral/metabolismo , Membro Posterior/irrigação sanguínea , Humanos , Imuno-Histoquímica , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Neovascularização Patológica/enzimologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Óxido Nítrico Sintase Tipo III/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células-Tronco/citologia , Células-Tronco/enzimologia , Células-Tronco/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
Int J Cardiol ; 178: 253-5, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25464264

RESUMO

The acetylcholinesterase inhibitor (AChEI)-based therapeutic strategies have been shown to have vasculoprotective properties in the animal model of hindlimb ischemia due to its activation of the endothelial cholinergic system. However, little is know about whether other cell types (myocytes, immunocytes) are involved in the AChEI-related therapeutic benefits in peripheral artery disease. Therefore, we review the multiple cell-targeted effects of AChEI on the animal model of hindlimb ischemia and explore its clinical application in angiomyogenesis.


Assuntos
Inibidores da Colinesterase/uso terapêutico , Doença Arterial Periférica/tratamento farmacológico , Animais , Sistema Nervoso Autônomo/efeitos dos fármacos , Sistema Nervoso Autônomo/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Inibidores da Colinesterase/farmacologia , Humanos , Doença Arterial Periférica/diagnóstico , Doença Arterial Periférica/enzimologia
19.
Atherosclerosis ; 237(2): 739-47, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25463114

RESUMO

OBJECTIVE: The interaction between the renin-angiotensin system and toll-like receptors (TLRs) in the pathogenesis of advanced atherosclerotic plaques is not well understood. We studied the effects of the renin inhibitor aliskiren on the progression of advanced atherosclerotic plaque in apolipoprotein E-deficient (ApoE(-/-)) mice with a special focus on plaque neovessel formation. METHODS AND RESULTS: Four-wk-old ApoE(-/-) mice were fed a high-fat diet for 8 wks, and the mice were randomly assigned to one of three groups and administered a vehicle, hydralazine, or aliskiren for an additional 12 wks. Aliskiren reduced the atherosclerotic plaque area and plaque neovessel density. It increased the plaque collagen and elastin contents, and reduced plasma angiotensin II levels and plaque macrophage infiltration and cathepsin S (CatS) protein. Aliskiren also decreased the levels of AT1R, gp91phox, TLR2, monocyte chemotactic protein-1, and CatS mRNAs in the aortic roots. Hydralazine had no beneficial vascular effects, although its administration resulted in the same degree of blood pressure reduction as aliskiren. CatS deficiency mimicked the aliskiren-mediated vasculoprotective effect in the ApoE(-/-) mice, but aliskiren showed no further benefits in ApoE(-/-) CatS(-/-) mice. In vitro, TLR2 silencing reduced CatS expression induced by angiotensin II. Moreover, aliskiren or the inhibition of CatS impaired the endothelial cell angiogenic action in vitro or/and ex vivo. CONCLUSION: Renin inhibition appears to inhibit advanced plaque neovessel formation in ApoE(-/-) mice and to decrease the vascular inflammatory action and extracellular matrix degradation, partly by reducing AT1R/TLR2-mediated CatS activation and activity, thus regressing advanced atherosclerosis.


Assuntos
Amidas/farmacologia , Aterosclerose/sangue , Aterosclerose/tratamento farmacológico , Fumaratos/farmacologia , Renina/antagonistas & inibidores , Angiotensina II/metabolismo , Animais , Aorta/metabolismo , Apolipoproteínas E/genética , Pressão Sanguínea , Proteínas de Transporte/metabolismo , Catepsinas/metabolismo , Quimiocina CCL2/metabolismo , Matriz Extracelular/metabolismo , Inativação Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Hidralazina/farmacologia , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica , Proteínas Nucleares , Placa Aterosclerótica/patologia , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor 2 Toll-Like/metabolismo , Resultado do Tratamento , Molécula 1 de Adesão de Célula Vascular/metabolismo
20.
Am J Transl Res ; 6(3): 281-90, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24936220

RESUMO

Extracorporeal pulsed electromagnetic field (PEMF) has been shown the ability to improve regeneration in various ischemic episodes. Here, we examined whether PEMF therapy facilitate cardiac recovery in rat myocardial infarction (MI), and the cellular/molecular mechanisms underlying PEMF-related therapy was further investigated. The MI rats were exposed to active PEMF for 4 cycles per day (8 minutes/cycle, 30 ± 3 Hz, 5 mT) after MI induction. The data demonstrated that PEMF treatment significantly inhibited cardiac apoptosis and improved cardiac systolic function. Moreover, PEMF treatment increased capillary density, the levels of vascular endothelial growth factor (VEGF) and hypoxic inducible factor-1α in infarct border zone. Furthermore, the number and function of circulating endothelial progenitor cells were advanced in PEMF treating rats. In vitro, PEMF induced the degree of human umbilical venous endothelial cells tubulization and increased soluble pro-angiogenic factor secretion (VEGF and nitric oxide). In conclusion, PEMF therapy preserves cardiac systolic function, inhibits apoptosis and trigger postnatal neovascularization in ischemic myocardium.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...