Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Invest Ophthalmol Vis Sci ; 62(7): 6, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34086044

RESUMO

Purpose: To investigate the expression of angiotensin-converting enzyme 2 (ACE2), the receptor for SARS-CoV-2 in human retina. Methods: Human post-mortem eyes from 13 non-diabetic control cases and 11 diabetic retinopathy cases were analyzed for the expression of ACE2. To compare the vascular ACE2 expression between different organs that involve in diabetes, the expression of ACE2 was investigated in renal specimens from nondiabetic and diabetic nephropathy patients. Expression of TMPRSS2, a cell-surface protease that facilitates SARS-CoV-2 entry, was also investigated in human nondiabetic retinas. Primary human retinal endothelial cells (HRECs) and primary human retinal pericytes (HRPCs) were further used to confirm the vascular ACE2 expression in human retina. Results: We found that ACE2 was expressed in multiple nonvascular neuroretinal cells, including the retinal ganglion cell layer, inner plexiform layer, inner nuclear layer, and photoreceptor outer segments in both nondiabetic and diabetic retinopathy specimens. Strikingly, we observed significantly more ACE2 positive vessels in the diabetic retinopathy specimens. By contrast, in another end-stage organ affected by diabetes, the kidney, ACE2 in nondiabetic and diabetic nephropathy showed apical expression of ACE2 tubular epithelial cells, but no endothelial expression in glomerular or peritubular capillaries. Western blot analysis of protein lysates from HRECs and HRPCs confirmed expression of ACE2. TMPRSS2 expression was present in multiple retinal neuronal cells, vascular and perivascular cells, and Müller glia. Conclusions: Together, these results indicate that retina expresses ACE2 and TMPRSS2. Moreover, there are increased vascular ACE2 expression in diabetic retinopathy retinas.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Retinopatia Diabética/enzimologia , Receptores Virais/metabolismo , Retina/enzimologia , SARS-CoV-2/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Sítios de Ligação , Western Blotting , Células Cultivadas , Nefropatias Diabéticas/enzimologia , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/virologia , Retinopatia Diabética/patologia , Retinopatia Diabética/virologia , Endotélio Vascular/enzimologia , Endotélio Vascular/virologia , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Pericitos/enzimologia , Pericitos/virologia , Vasos Retinianos/enzimologia , Vasos Retinianos/patologia , Vasos Retinianos/virologia , Serina Endopeptidases/metabolismo
2.
Exp Eye Res ; 207: 108559, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33848522

RESUMO

Diabetic retinopathy is a multifactorial microvascular complication, and its pathogenesis hasn't been fully elucidated. The irreversible oxidation of cysteine 674 (C674) in the sarcoplasmic/endoplasmic reticulum calcium ATPase 2 (SERCA2) was increased in the type 1 diabetic retinal vasculature. SERCA2 C674S knock-in (SKI) mouse line that half of C674 was replaced by serine 674 (S674) was used to study the effect of C674 inactivation on retinopathy. Compared with wild type (WT) mice, SKI mice had increased number of acellular capillaries and pericyte loss similar to those in type 1 diabetic WT mice. In the retina of SKI mice, pro-apoptotic proteins and intracellular Ca2+-dependent signaling pathways increased, while anti-apoptotic proteins and vessel density decreased. In endothelial cells, C674 inactivation increased the expression of pro-apoptotic proteins, damaged mitochondria, and induced cell apoptosis. These results suggest that a possible mechanism of retinopathy induced by type 1 diabetes is the interruption of calcium homeostasis in the retina by oxidation of C674. C674 is a key to maintain retinal health. Its inactivation can cause retinopathy similar to type 1 diabetes by promoting apoptosis. SERCA2 might be a potential target for the prevention and treatment of diabetic retinopathy.


Assuntos
Cisteína/genética , Retinopatia Diabética/enzimologia , Retículo Endoplasmático/enzimologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/enzimologia , Adenoviridae , Animais , Apoptose , Western Blotting , Calcineurina/metabolismo , Capilares/enzimologia , Capilares/patologia , Cisteína/metabolismo , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/enzimologia , Diabetes Mellitus Tipo 1/genética , Retinopatia Diabética/patologia , Técnica Indireta de Fluorescência para Anticorpo , Técnicas de Introdução de Genes , Inativação Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Imuno-Histoquímica , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Mitocôndrias/metabolismo , Oxirredução , Reação em Cadeia da Polimerase em Tempo Real , Vasos Retinianos/enzimologia , Vasos Retinianos/patologia , Transdução de Sinais , Estreptozocina
3.
FASEB J ; 35(1): e21152, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33151576

RESUMO

Vitreous has been reported to prevent tumor angiogenesis, but our previous findings indicate that vitreous activate the signaling pathway of phosphoinositide 3-kinase (PI3K)/Akt, which plays a critical role in angiogenesis. The goal of this research is to determine which role of vitreous plays in angiogenesis-related cellular responses in vitro. We found that in human retinal microvascular endothelial cells (HRECs) vitreous activates a number of receptor tyrosine kinases including Anexelekto (Axl), which plays an important role in angiogenesis. Subsequently, we discovered that depletion of Axl using CRISPR/Cas9 and an Axl-specific inhibitor R428 suppress vitreous-induced Akt activation and cell proliferation, migration, and tuber formation of HRECs. Therefore, this line of research not only demonstrate that vitreous promotes angiogenesis in vitro, but also reveal that Axl is one of receptor tyrosine kinases to mediate vitreous-induced angiogenesis in vitro, thereby providing a molecular basis for removal of vitreous as cleanly as possible when vitrectomy is performed in treating patients with proliferative diabetic retinopathy.


Assuntos
Neovascularização Patológica/enzimologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Vasos Retinianos/enzimologia , Corpo Vítreo/enzimologia , Animais , Benzocicloeptenos/farmacologia , Sistemas CRISPR-Cas , Retinopatia Diabética/enzimologia , Retinopatia Diabética/genética , Retinopatia Diabética/patologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Células HEK293 , Humanos , Camundongos , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/genética , Vasos Retinianos/patologia , Triazóis/farmacologia , Vitreorretinopatia Proliferativa/enzimologia , Vitreorretinopatia Proliferativa/genética , Vitreorretinopatia Proliferativa/patologia , Corpo Vítreo/patologia , Receptor Tirosina Quinase Axl
4.
Biosci Rep ; 40(5)2020 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-32319515

RESUMO

Diabetic retinopathy (DR) is the most common microvascular complication of diabetes and is characterized by visible microvascular alterations including retinal ischemia-reperfusion injury, inflammation, abnormal permeability, neovascularization and macular edema. Despite the available treatments, some patients present late in the course of the disease when treatment is more difficult. Hence, it is crucial that the new targets are found and utilized in the clinical therapy of DR. In the present study, we constructed a DR animal model and a model in HRMECs to investigate the relationship between p38 and RUNX1 in retinal micro-angiogenesis in diabetic retinopathy. We found that p38 could promote retinal micro-angiogenesis by up-regulating RUNX1 expression in diabetic retinopathy. This suggested that the p38/ RUNX1 pathway could become a new retinal micro-angiogenesis target in DR treatment.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Retinopatia Diabética/enzimologia , Células Endoteliais/enzimologia , Neovascularização Retiniana/enzimologia , Vasos Retinianos/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Glicemia/metabolismo , Linhagem Celular , Proliferação de Células , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/complicações , Retinopatia Diabética/etiologia , Retinopatia Diabética/genética , Retinopatia Diabética/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Glucose/toxicidade , Humanos , Masculino , Camundongos Endogâmicos C57BL , Neovascularização Retiniana/etiologia , Neovascularização Retiniana/genética , Neovascularização Retiniana/patologia , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/patologia , Transdução de Sinais , Regulação para Cima
5.
Microvasc Res ; 121: 14-23, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30189210

RESUMO

PURPOSE: Diabetic retinopathy (DR) is characterized by pro-inflammatory, pro-angiogenic and pro-fibrotic environment during the various stages of the disease progression. Basement membrane changes in the retina and formation of fibrovascular membrane are characteristically seen in DR. In the present study the effect of Alcoholic (AlE) extracts of Triphala an ayurvedic herbal formulation and its chief compounds, Chebulagic (CA), Chebulinic (CI) and Gallic acid (GA) were evaluated for TGFß1-induced anti-fibrotic activity in choroid-retinal endothelial cells (RF/6A). METHOD: RF/6A cells were treated with TGFß1 alone or co-treated with AlE, CA, CI or GA. The mRNA and protein expression of fibrotic markers (αSMA, CTGF) were assessed by qPCR and western blot/ELISA. Functional changes were assessed using proliferation assay and migration assay. To deduce the mechanism of action, downstream signaling was assessed by western blot analysis along with in silico docking studies. RESULT: AlE (50 µg/ml) CA and CI at 10 µM reduced the expression of pro-fibrotic genes (αSMA and CTGF) induced by TGFß1, by inhibiting ERK phosphorylation. GA did not inhibit TGFß1 mediated changes in RF/6A cells. In silico experiments shows that CA and CI has favourable binding energy to bind with TGFß receptor and inhibit the downstream signaling, while GA did not. CONCLUSION: Hence this study identifies Triphala and its chief compounds CA and CI as potential adjuvants in the management of DR.


Assuntos
Benzopiranos/farmacologia , Corioide/irrigação sanguínea , Retinopatia Diabética/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glucosídeos/farmacologia , Taninos Hidrolisáveis/farmacologia , Extratos Vegetais/farmacologia , Vasos Retinianos/efeitos dos fármacos , Fator de Crescimento Transformador beta1/toxicidade , Animais , Benzopiranos/metabolismo , Sítios de Ligação , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Retinopatia Diabética/enzimologia , Retinopatia Diabética/patologia , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Fibrose , Glucosídeos/metabolismo , Taninos Hidrolisáveis/metabolismo , Macaca mulatta , Simulação de Acoplamento Molecular , Neovascularização Patológica , Fosforilação , Ligação Proteica , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Vasos Retinianos/enzimologia , Vasos Retinianos/patologia , Transdução de Sinais/efeitos dos fármacos
6.
Sci Rep ; 8(1): 10581, 2018 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-30002415

RESUMO

Nucleoside diphosphate kinase B (NDPK-B) acts as a protective factor in the retinal vasculature. NDPK-B deficiency leads to retinal vasoregression mimicking diabetic retinopathy (DR). Angiopoetin 2 (Ang-2), an initiator of retinal vasoregression in DR, is upregulated in NDPK-B deficient retinas and in NDPK-B depleted endothelial cells (ECs) in vitro. We therefore investigated the importance of Ang-2 in NDPK-B deficient retinas and characterized the mechanisms of Ang-2 upregulation upon NDPK-B depletion in cultured ECs. The crucial role of retinal Ang-2 in the initiation of vasoregression was verified by crossing NDPK-B deficient with Ang-2 haplodeficient mice. On the molecular level, FoxO1, a transcription factor regulating Ang-2, was upregulated in NDPK-B depleted ECs. Knockdown of FoxO1 abolished the elevation of Ang-2 induced by NDPK-B depletion. Furthermore O-GlcNAcylated FoxO1 was found preferentially in the nucleus. An increased O-GlcNAcylation of FoxO1 was revealed upon NDPK-B depletion. In accordance, the inhibition of protein O-GlcNAcylation normalized NDPK-B depletion induced Ang-2 upregulation. In summary, we demonstrated that the upregulation of Ang-2 upon NDPK-B deficiency is driven by O-GlcNAcylation of FoxO1. Our data provide evidence for a central role of protein O-GlcNAcylation in NDPK-B associated vascular damage and point to the hexosamine pathway as an important target in retinal vasoregression.


Assuntos
Angiopoietina-2/genética , Retinopatia Diabética/patologia , Proteína Forkhead Box O1/metabolismo , Nucleosídeo NM23 Difosfato Quinases/deficiência , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Retina/patologia , Acetilglucosamina/metabolismo , Angiopoietina-2/metabolismo , Animais , Núcleo Celular/metabolismo , Retinopatia Diabética/genética , Modelos Animais de Doenças , Endotélio Vascular/citologia , Endotélio Vascular/enzimologia , Endotélio Vascular/patologia , Proteína Forkhead Box O1/genética , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Knockout , Nucleosídeo NM23 Difosfato Quinases/genética , Cultura Primária de Células , RNA Interferente Pequeno/metabolismo , Retina/citologia , Retina/enzimologia , Vasos Retinianos/citologia , Vasos Retinianos/enzimologia , Vasos Retinianos/patologia , Regulação para Cima
7.
Invest Ophthalmol Vis Sci ; 59(5): 2042-2053, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29677366

RESUMO

Purpose: Loss of retinal capillary endothelial cells and pericytes through apoptosis is an early event in diabetic retinopathy (DR). Inflammatory pathways play a role in early DR, yet the biochemical mechanisms are poorly understood. In this study, we investigated the role of indoleamine 2,3-dioxygenase (IDO), an inflammatory cytokine-inducible enzyme, on retinal endothelial apoptosis and capillary degeneration in the diabetic retina. Methods: IDO was detected in human and mouse retinas by immunohistochemistry or Western blotting. Interferon-γ (IFN-γ) levels were measured by ELISA. IDO levels were measured in human retinal capillary endothelial cells (HREC) cultured in the presence of IFN-γ ± 25 mM D-glucose. Reactive oxygen species (ROS) were measured using CM-H2DCFDA dye and apoptosis was measured by cleaved caspase-3. The role of IDO in DR was determined in IDO knockout (IDO-/-) mice with streptozotocin-induced diabetes. Results: The IDO and IFN-γ levels were higher in human diabetic retinas with retinopathy relative to nondiabetic retinas. Immunohistochemical data showed that IDO is present in capillary endothelial cells. IFN-γ upregulated the IDO and ROS levels in HREC. The blockade of either IDO or kynurenine monooxygenase led to inhibition of ROS in HREC. Apoptosis through this pathway was inhibited by an ROS scavenger, TEMPOL. Capillary degeneration was significantly reduced in diabetic IDO-/- mice compared to diabetic wild-type mice. Conclusions: The results suggest that the kynurenine pathway plays an important role in the inflammatory damage in the diabetic retina and could be a new therapeutic target for the treatment of DR.


Assuntos
Retinopatia Diabética/complicações , Células Endoteliais/patologia , Indolamina-Pirrol 2,3,-Dioxigenase/deficiência , Degeneração Retiniana/prevenção & controle , Vasos Retinianos/patologia , Idoso , Animais , Western Blotting , Células Cultivadas , Diabetes Mellitus Experimental/complicações , Eletroforese em Gel de Poliacrilamida , Células Endoteliais/enzimologia , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Espécies Reativas de Oxigênio/metabolismo , Degeneração Retiniana/enzimologia , Vasos Retinianos/enzimologia
8.
Vascul Pharmacol ; 108: 23-35, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29678603

RESUMO

Tumor necrosis factor-α (TNFα) a pleiotropic cytokine induces pro-inflammatory and pro-angiogenic changes in conditions such as diabetic retinopathy (DR) and neovascular age related macular degeneration (NV-AMD). Hence, inhibition of TNFα mediated changes can benefit the management of DR and NV-AMD. Triphala, an ayurvedic herbal preparation is known to have immunomodulatry functions. In this study we evaluated the alcoholic extract of triphala (AlE) and its compounds Chebulagic acid (CA), Chebulinic acid (CI) and Gallic acid (GA) for their anti-TNFα activity. TNFα induced pro-inflammatory and pro-angiogenic changes in the retinal-choroid microvascular endothelial cells (RF/6A). Treatment with CA/CI/GA and the whole Triphala extract showed characteristic inhibition of MMP-9, cell proliferation/migration and tube formation as well the expression of IL-6, IL-8 and MCP-1 without affecting cell viability. This was mediated by inhibition of p38, ERK and NFκB phosphorylation. Ex vivo angiogenesis assay using chick chorioallantoic membrane (CAM) model also showed that TNFα-induced angiogenesis and it was inhibited by AlE and its active principles. Further, in silico studies revealed that CA, CI and GA are capable of binding the TNFα-receptor-1 to mediate anti-TNFα activity. This study explains the immunomodulatory function of Triphala, evaluated in the context of retinal and choroid vasculopathies in vitro and ex vivo; which showed that CA, CI and GA can be a potential pharmacological agents in the management of DR and NV-AMD.


Assuntos
Inibidores da Angiogênese/farmacologia , Anti-Inflamatórios/farmacologia , Benzopiranos/farmacologia , Células Endoteliais/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Ácido Gálico/farmacologia , Glucosídeos/farmacologia , Taninos Hidrolisáveis/farmacologia , NF-kappa B/metabolismo , Extratos Vegetais/farmacologia , Neovascularização Retiniana/prevenção & controle , Vasos Retinianos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/toxicidade , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Inibidores da Angiogênese/metabolismo , Animais , Anti-Inflamatórios/metabolismo , Benzopiranos/metabolismo , Linhagem Celular , Embrião de Galinha , Relação Dose-Resposta a Droga , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Ácido Gálico/metabolismo , Glucosídeos/metabolismo , Taninos Hidrolisáveis/metabolismo , Mediadores da Inflamação/metabolismo , Macaca mulatta , Metaloproteinase 9 da Matriz/metabolismo , Simulação de Acoplamento Molecular , Neovascularização Fisiológica/efeitos dos fármacos , Fosforilação , Ligação Proteica , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Neovascularização Retiniana/enzimologia , Neovascularização Retiniana/patologia , Vasos Retinianos/enzimologia , Vasos Retinianos/patologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
9.
Biosci Biotechnol Biochem ; 82(8): 1366-1376, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29658404

RESUMO

The aim of the present study is to investigate the role of miR-21-5p in angiogenesis of human retinal microvascular endothelial cells (HRMECs). HRMECs were incubated with 5 mM glucose, 30 mM glucose or 30 mM mannitol for 24 h, 48 h or 72 h. Then, HRMECs exposed to 30 mM glucose were transfected with miR-21-5p inhibitor. We found that high glucose increased the expression of miR-21-5p, VEGF, VEGFR2 and cell proliferation activity. Inhibition of miR-21-5p reduced high glucose-induced proliferation, migration, tube formation of HRMECs, and reversed the decreased expression of maspin as well as the abnormal activation of PI3K/AKT and ERK pathways. Down-regulation of maspin by siRNA significantly increased the activities of PI3K/AKT and ERK pathways. In conclusion, inhibition of miR-21-5p could suppress high glucose-induced proliferation and angiogenesis of HRMECs, and these effects may partly dependent on the regulation of PI3K/AKT and ERK pathways via its target protein maspin.


Assuntos
Proliferação de Células/efeitos dos fármacos , Glucose/farmacologia , Sistema de Sinalização das MAP Quinases/fisiologia , MicroRNAs/antagonistas & inibidores , Neovascularização Patológica/prevenção & controle , Proteínas Proto-Oncogênicas c-akt/metabolismo , Vasos Retinianos/efeitos dos fármacos , Serpinas/metabolismo , Células Cultivadas , Retinopatia Diabética/patologia , Regulação para Baixo , Glucose/antagonistas & inibidores , Humanos , MicroRNAs/metabolismo , Vasos Retinianos/citologia , Vasos Retinianos/enzimologia , Vasos Retinianos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 38(2): 373-385, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29242271

RESUMO

OBJECTIVE: SNRK (sucrose nonfermenting 1-related kinase) is a novel member of the AMPK (adenosine monophosphate-activated protein kinase)-related superfamily that is activated in the process of angiogenesis. Currently, little is known about the function of SNRK in angiogenesis in the physiological and pathological conditions. APPROACH AND RESULTS: In this study, in Snrk global heterozygous knockout mice, retina angiogenesis and neovessel formation after hindlimb ischemia were suppressed. Consistently, mice with endothelial cell (EC)-specific Snrk deletion exhibited impaired retina angiogenesis, and delayed perfusion recovery and exacerbated muscle apoptosis in ischemic hindlimbs, compared with those of littermate wide-type mice. Endothelial SNRK expression was increased in the extremity vessel samples from nonischemic human. In ECs cultured in hypoxic conditions, HIF1α (hypoxia inducible factor 1α) bound to the SNRK promoter to upregulate SNRK expression. In the nuclei of hypoxic ECs, SNRK complexed with SP1 (specificity protein 1), and together, they bound to an SP1-binding motif in the ITGB1 (ß1 integrin) promoter, resulting in enhanced ITGB1 expression and promoted EC migration. Furthermore, SNRK or SP1 deficiency in ECs ameliorated hypoxia-induced ITGB1 expression and, consequently, inhibited EC migration and angiogenesis. CONCLUSIONS: Taken together, our data have revealed that SNRK/SP1-ITGB1 signaling axis promotes angiogenesis in vivo.


Assuntos
Células Endoteliais/enzimologia , Isquemia/enzimologia , Pulmão/irrigação sanguínea , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Proteínas Serina-Treonina Quinases/metabolismo , Vasos Retinianos/enzimologia , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Apoptose , Velocidade do Fluxo Sanguíneo , Caderinas/genética , Caderinas/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Regulação Enzimológica da Expressão Gênica , Membro Posterior , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Integrina beta1/genética , Integrina beta1/metabolismo , Isquemia/genética , Isquemia/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Fluxo Sanguíneo Regional , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo
11.
J Endocrinol ; 235(1): 39-48, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28751454

RESUMO

Cysteamine (an aminothiol), which is derived from coenzyme A degradation and metabolized into taurine, has beneficial effects against cystinosis and neurodegenerative diseases; however, its role in diabetic complications is unknown. Thus, we sought to determine the preventive effect of cysteamine against hyperglycemia-induced vascular leakage in the retinas of diabetic mice. Cysteamine and ethanolamine, the sulfhydryl group-free cysteamine analogue, inhibited vascular endothelial growth factor (VEGF)-induced stress fiber formation and vascular endothelial (VE)-cadherin disruption in endothelial cells, which play a critical role in modulating endothelial permeability. Intravitreal injection of the amine compounds prevented hyperglycemia-induced vascular leakage in the retinas of streptozotocin-induced diabetic mice. We then investigated the potential roles of reactive oxygen species (ROS) and transglutaminase (TGase) in the cysteamine prevention of VEGF-induced vascular leakage. Cysteamine, but not ethanolamine, inhibited VEGF-induced ROS generation in endothelial cells and diabetic retinas. In contrast, VEGF-induced TGase activation was prevented by both cysteamine and ethanolamine. Our findings suggest that cysteamine protects against vascular leakage through inhibiting VEGF-induced TGase activation rather than ROS generation in diabetic retinas.


Assuntos
Cisteamina/administração & dosagem , Retinopatia Diabética/prevenção & controle , Retina/metabolismo , Vasos Retinianos/enzimologia , Transglutaminases/metabolismo , Animais , Retinopatia Diabética/enzimologia , Retinopatia Diabética/genética , Retinopatia Diabética/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Retina/efeitos dos fármacos , Retina/enzimologia , Vasos Retinianos/efeitos dos fármacos , Transglutaminases/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Invest Ophthalmol Vis Sci ; 58(1): 230-241, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28114584

RESUMO

Purpose: Endothelial nitric oxide synthase (eNOS)-derived nitric oxide (NO) has important vasoprotective functions that are compromised in the vasodegenerative phase of retinopathy of prematurity, owing to hyperoxia-induced depletion of the essential NOS cofactor BH4. Because modulating eNOS function can be beneficial or detrimental, our aim was to investigate the effect of BH4 supplementation on eNOS function and vascular regression in hyperoxia. Methods: Endothelial-specific eNOS-green fluorescent protein (GFP) overexpressing mice at postnatal day 7 (P7) were exposed to hyperoxia for 48 hours in the presence or absence of supplemental BH4, achieved by administration of sepiapterin, a stable BH4 precursor. Tissue was collected either for retinal flat mounts that were stained with lectin to determine the extent of vessel coverage or for analysis of BH4 by high-performance liquid chromatography, nitrotyrosine (NT) marker by Western blotting, VEGF expression by ELISA, and NOS activity by arginine-to-citrulline conversion. Primary retinal microvascular endothelial cells (RMEC) were similarly treated, and hyperoxia-induced damage was determined. Results: Sepiapterin effectively enhanced BH4 levels in hyperoxia-exposed retinas and brains, elevated NOS activity, and reduced NT-modified protein, leading to reversal of the exacerbated vasoregression observed in the presence of eNOS overexpression. In RMECs, hyperoxia-mediated depletion of BH4 dysregulated the redox balance by reducing nitrite and elevating superoxide and impaired proliferative ability. BH4 supplementation restored normal RMEC proliferation in vitro and also in vivo, providing a mechanistic link with the enhanced vascular coverage in eNOS-GFP retinas. Conclusions: These results demonstrate that BH4 supplementation corrects hyperoxia-induced RMEC dysfunction and preserves vascular integrity by enhancing eNOS function.


Assuntos
Biopterinas/análogos & derivados , Endotélio Vascular/enzimologia , Hiperóxia/prevenção & controle , Óxido Nítrico Sintase Tipo III/metabolismo , Vasos Retinianos/enzimologia , Retinopatia da Prematuridade/prevenção & controle , Animais , Animais Recém-Nascidos , Biopterinas/farmacologia , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Ensaio de Imunoadsorção Enzimática , Humanos , Hiperóxia/complicações , Hiperóxia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo III/efeitos dos fármacos , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/patologia , Retinopatia da Prematuridade/etiologia , Retinopatia da Prematuridade/patologia
13.
Planta Med ; 83(3-04): 318-325, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27690380

RESUMO

In our ongoing efforts to identify effective naturally sourced agents for the treating of diabetic complications, two new (1 and 2) and 11 known phenolic compounds (3-13) were isolated from an 80 % ethanol extract of Litsea japonica leaves. The structures of the new compounds were established by spectroscopic and chemical studies. These isolates (1-13) were subjected to an in vitro bioassay evaluating their inhibitory activity on advanced glycation end products formation and rat lens aldose reductase activity. Of the compounds evaluated, the flavonoids (3, 4, 6-8, 11, and 12) markedly inhibited advanced glycation end products formation, with IC50 values of 7.4-72.0 µM, compared with the positive control, aminoguanidine (IC50 = 975.9 µM). In the rat lens aldose reductase assay, consistent with the inhibition of advanced glycation end products formation, the flavonoids (3, 4, 6-8, 11, and 12) exhibited considerable inhibition of rat lens aldose reductase activity, with IC50 values of 1.1-12.5 µM. In addition, the effects of kaempferol (4) and tiliroside (7) on the dilation of hyaloid-retinal vessels induced by high glucose in larval zebrafish were investigated. Only kaempferol significantly reduced the diameters of high glucose-induced hyaloid-retinal vessels, by 52.2 % at 10 µM, compared with those in the high glucose-treated control group.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Flavonoides/farmacologia , Produtos Finais de Glicação Avançada/antagonistas & inibidores , Cristalino/enzimologia , Litsea/química , Aldeído Redutase/metabolismo , Animais , Angiopatias Diabéticas/induzido quimicamente , Modelos Animais de Doenças , Flavonoides/química , Flavonoides/isolamento & purificação , Produtos Finais de Glicação Avançada/metabolismo , Guanidinas/farmacologia , Técnicas In Vitro , Concentração Inibidora 50 , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação , Extratos Vegetais/farmacologia , Folhas de Planta/química , Ratos , Ratos Sprague-Dawley , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/enzimologia , Vasos Retinianos/metabolismo , Peixe-Zebra
14.
Cell Death Dis ; 7(11): e2483, 2016 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-27882947

RESUMO

Retinal ischemia is a major cause of visual impairment and blindness and is involved in various disorders including diabetic retinopathy, glaucoma, optic neuropathies and retinopathy of prematurity. Neurovascular degeneration is a common feature of these pathologies. Our lab has previously reported that the ureahydrolase arginase 2 (A2) is involved in ischemic retinopathies. Here, we are introducing A2 as a therapeutic target to prevent neurovascular injury after retinal ischemia/reperfusion (I/R) insult. Studies were performed with mice lacking both copies of A2 (A2-/-) and wild-type (WT) controls (C57BL6J). I/R insult was conducted on the right eye and the left eye was used as control. Retinas were collected for analysis at different times (3 h-4 week after injury). Neuronal and microvascular degeneration were evaluated using NeuN staining and vascular digests, respectively. Glial activation was evaluated by glial fibrillary acidic protein expression. Necrotic cell death was studied by propidium iodide labeling and western blot for RIP-3. Arginase expression was determined by western blot and quantitative RT-PCR. Retinal function was determined by electroretinography (ERG). A2 mRNA and protein levels were increased in WT I/R. A2 deletion significantly reduced ganglion cell loss and microvascular degeneration and preserved retinal morphology after I/R. Glial activation, reactive oxygen species formation and cell death by necroptosis were significantly reduced by A2 deletion. ERG showed improved positive scotopic threshold response with A2 deletion. This study shows for the first time that neurovascular injury after retinal I/R is mediated through increased expression of A2. Deletion of A2 was found to be beneficial in reducing neurovascular degeneration after I/R.


Assuntos
Arginase/metabolismo , Degeneração Neural/enzimologia , Degeneração Neural/patologia , Traumatismo por Reperfusão/enzimologia , Vasos Retinianos/enzimologia , Vasos Retinianos/patologia , Animais , Arginase/genética , Morte Celular , Sobrevivência Celular , Deleção de Genes , Camundongos Endogâmicos C57BL , Microvasos/patologia , Modelos Biológicos , Neuroglia/patologia , Neurônios/enzimologia , Neurônios/patologia , Neuroproteção , Estresse Oxidativo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Traumatismo por Reperfusão/patologia
15.
Arterioscler Thromb Vasc Biol ; 36(4): 707-17, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26821948

RESUMO

OBJECTIVE: To determine the role of Gja5 that encodes for the gap junction protein connexin40 in the generation of arteriovenous malformations in the hereditary hemorrhagic telangiectasia type 2 (HHT2) mouse model. APPROACH AND RESULTS: We identified GJA5 as a target gene of the bone morphogenetic protein-9/activin receptor-like kinase 1 signaling pathway in human aortic endothelial cells and importantly found that connexin40 levels were particularly low in a small group of patients with HHT2. We next took advantage of the Acvrl1(+/-) mutant mice that develop lesions similar to those in patients with HHT2 and generated Acvrl1(+/-); Gja5(EGFP/+) mice. Gja5 haploinsufficiency led to vasodilation of the arteries and rarefaction of the capillary bed in Acvrl1(+/-) mice. At the molecular level, we found that reduced Gja5 in Acvrl1(+/-) mice stimulated the production of reactive oxygen species, an important mediator of vessel remodeling. To normalize the altered hemodynamic forces in Acvrl1(+/-); Gja5(EGFP/+) mice, capillaries formed transient arteriovenous shunts that could develop into large malformations when exposed to environmental insults. CONCLUSIONS: We identified GJA5 as a potential modifier gene for HHT2. Our findings demonstrate that Acvrl1 haploinsufficiency combined with the effects of modifier genes that regulate vessel caliber is responsible for the heterogeneity and severity of the disease. The mouse models of HHT have led to the proposal that 3 events-heterozygosity, loss of heterozygosity, and angiogenic stimulation-are necessary for arteriovenous malformation formation. Here, we present a novel 3-step model in which pathological vessel caliber and consequent altered blood flow are necessary events for arteriovenous malformation development.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Receptores de Ativinas Tipo I/metabolismo , Malformações Arteriovenosas/enzimologia , Conexinas/metabolismo , Células Endoteliais/enzimologia , Vasos Retinianos/enzimologia , Telangiectasia Hemorrágica Hereditária/enzimologia , Receptores de Ativinas Tipo I/genética , Receptores de Activinas Tipo II/genética , Animais , Malformações Arteriovenosas/genética , Malformações Arteriovenosas/patologia , Células Cultivadas , Conexinas/genética , Modelos Animais de Doenças , Predisposição Genética para Doença , Haploinsuficiência , Humanos , Camundongos Mutantes , Camundongos Transgênicos , Neovascularização Patológica , Fenótipo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Vasos Retinianos/patologia , Transdução de Sinais , Telangiectasia Hemorrágica Hereditária/genética , Telangiectasia Hemorrágica Hereditária/patologia , Transfecção , Remodelação Vascular , Proteína alfa-5 de Junções Comunicantes
16.
Prostaglandins Other Lipid Mediat ; 122: 69-72, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26791393

RESUMO

AIM: To assess the role of plasma platelet activating factor acetylhydrolase (PAF-AH) in pathogenesis and progression of diabetic retinopathy (DR). MATERIALS AND METHODS: Sixty eight diabetics and 23 age-frequency-matched non-diabetic patients underwent blood sampling and the plasma PAF-AH activity was calculated. The diabetic patients were further classified into two groups, according to the Early Treatment Diabetic Retinopathy Study (ETDRS) classification, based on indirect fundoscopy and fluorescein angiography. Thirty seven patients with non-proliferative DR (NPDR) and 31 patients with proliferative DR (PDR) were finally included in the study. RESULTS: The plasma PAF-AH activity was increased in diabetic patients with PDR (0.206 µmol/min/ml) compared to control group (0.114 µmol/min/ml, post-hoc Bonferroni comparison test: p<0.0001) and to NPDR group (0.147 µmol/min/ml, post-hoc Bonferroni comparison test: p=0.012). CONCLUSIONS: The activity of PAF-AH in the plasma increases in parallel with DR severity.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/sangue , 1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Retinopatia Diabética/sangue , Retinopatia Diabética/enzimologia , Idoso , Retinopatia Diabética/fisiopatologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Inflamação/sangue , Inflamação/enzimologia , Inflamação/fisiopatologia , Masculino , Pessoa de Meia-Idade , Retina/enzimologia , Retina/patologia , Retina/fisiopatologia , Vasos Retinianos/enzimologia , Vasos Retinianos/patologia , Vasos Retinianos/fisiopatologia , Fatores de Risco , Índice de Gravidade de Doença , Acuidade Visual
18.
J Cell Physiol ; 231(8): 1709-18, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26599598

RESUMO

Increase in matrix metalloproteinase-9 (MMP-9) is implicated in retinal capillary cell apoptosis, a phenomenon which precedes the development of diabetic retinopathy. MMP-9 promoter has multiple sites for binding the transcriptional factors, including two for activator protein 1 (AP-1). The binding of AP-1, a heterodimer of c-Jun and c-Fos, is regulated by posttranslational modifications, and in diabetes, deacetylating enzyme, Sirt1, is inhibited. Our aim, is to investigate the molecular mechanism of MMP-9 transcriptional regulation in diabetes. Binding of AP-1 (c-Jun, c-Fos) at the MMP-9 promoter, and AP-1 acetylation were analyzed in retinal endothelial cells incubated in normal or high glucose by chromatin-immunoprecipitation and co-immunoprecipitation respectively. Role of AP-1 in MMP-9 regulation was confirmed by c-Jun or c-Fos siRNAs, and that of its acetylation, by Sirt1 overexpression. In vitro results were validated in the retina from diabetic mice overexpressing Sirt1, and in the retinal microvessels from human donors with diabetic retinopathy. In experimental models, AP-1 binding was increased at the proximal and distal sites of the MMP-9 promoter, and similar phenomenon was confirmed in the retinal microvessels from human donors with diabetic retinopathy. Silencing of AP-1, or overexpression of Sirt1 ameliorated glucose-induced increase in MMP-9 expression and cell apoptosis. Thus, in diabetes, due to Sirt1 inhibition, AP-1 is hyperacetylated, which increases its binding at MMP-9 promoter, and hence, activation of Sirt1 could inhibit the development of diabetic retinopathy by impeding MMP-9-mediated mitochondrial damage. J. Cell. Physiol. 231: 1709-1718, 2016. © 2015 Wiley Periodicals, Inc.


Assuntos
Retinopatia Diabética/enzimologia , Células Endoteliais/enzimologia , Metaloproteinase 9 da Matriz/metabolismo , Vasos Retinianos/enzimologia , Transcrição Gênica , Acetilação , Idoso , Animais , Apoptose , Sítios de Ligação , Glicemia/metabolismo , Bovinos , Células Cultivadas , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/complicações , Retinopatia Diabética/sangue , Retinopatia Diabética/etiologia , Retinopatia Diabética/genética , Retinopatia Diabética/patologia , Células Endoteliais/patologia , Regulação Enzimológica da Expressão Gênica , Humanos , Metaloproteinase 9 da Matriz/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Regiões Promotoras Genéticas , Interferência de RNA , Vasos Retinianos/patologia , Sirtuína 1/genética , Sirtuína 1/metabolismo , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Transfecção
19.
Arterioscler Thromb Vasc Biol ; 36(2): 350-60, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26634655

RESUMO

OBJECTIVE: We recently demonstrated that low-density lipoprotein receptor-related protein 1 (LRP1) is required for cardiovascular development in zebrafish. However, what role LRP1 plays in angiogenesis remains to be determined. To better understand the role of LRP1 in endothelial cell function, we investigated how LRP1 regulates mouse retinal angiogenesis. APPROACH AND RESULTS: Depletion of LRP1 in endothelial cells results in increased retinal neovascularization in a mouse model of oxygen-induced retinopathy. Specifically, retinas in mice lacking endothelial LRP1 have more branching points and angiogenic sprouts at the leading edge of the newly formed vasculature. Increased endothelial proliferation as detected by Ki67 staining was observed in LRP1-deleted retinal endothelium in response to hypoxia. Using an array of biochemical and cell biology approaches, we demonstrate that poly(ADP-ribose) polymerase-1 (PARP-1) directly interacts with LRP1 in human retinal microvascular endothelial cells. This interaction between LRP1 and PARP-1 decreases under hypoxic condition. Moreover, LRP1 knockdown results in increased PARP-1 activity and subsequent phosphorylation of both retinoblastoma protein and cyclin-dependent kinase 2, which function to promote cell cycle progression and angiogenesis. CONCLUSIONS: Together, these data reveal a pivotal role for LRP1 in endothelial cell proliferation and retinal neovascularization induced by hypoxia. In addition, we demonstrate for the first time the interaction between LRP1 and PARP-1 and the LRP1-dependent regulation of PARP-1-signaling pathways. These data bring forth the possibility of novel therapeutic approaches for pathological angiogenesis.


Assuntos
Proliferação de Células , Células Endoteliais/enzimologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Receptores de LDL/metabolismo , Neovascularização Retiniana/enzimologia , Vasos Retinianos/enzimologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Ciclo Celular , Hipóxia Celular , Quinase 2 Dependente de Ciclina/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Genótipo , Células HEK293 , Humanos , Hipóxia/complicações , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Camundongos Knockout , Fenótipo , Fosforilação , Poli(ADP-Ribose) Polimerase-1 , Interferência de RNA , Receptores de LDL/deficiência , Receptores de LDL/genética , Neovascularização Retiniana/etiologia , Neovascularização Retiniana/genética , Neovascularização Retiniana/patologia , Vasos Retinianos/patologia , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais , Fatores de Tempo , Transfecção , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
20.
Sci Rep ; 5: 12796, 2015 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-26242473

RESUMO

Neural vascular barrier is essential for the life of multicellular organisms, and its impairment by tissue hypoxia is known to be a central of pathophysiology accelerating the progression of various intractable neural diseases. Therefore, the molecules involved in hypoxia-induced impairment of vascular barrier can be the targets to establish new therapies for intractable diseases. Here, we demonstrate that a disintegrin and metalloproteinases (ADAMs) 12 and 17 expressed in endothelial cells are the molecules responsible for the impairment of neural vascular barrier by hypoxia. Brain microvascular endothelial cells in vitro lost their barrier properties immediately after hypoxic stimulation through diminished localization of claudin-5, a tight junction molecule, on cell membranes. Hypoxic disappearance of claudin-5 from cell membranes and the consequent loss of barrier properties were completely suppressed by inhibition of the metalloproteinase activity which was found to be attributed to ADAM12 and ADAM17. Inhibition of either ADAM12 or ADAM17 was sufficient to rescue the in vivo neural vasculature under hypoxia from the loss of barrier function. This is the first report to specify the molecules which are responsible for hypoxia-induced impairment of neural vascular barrier and furthermore can be the targets of new therapeutic strategies for intractable neural diseases.


Assuntos
Proteínas ADAM/fisiologia , Células Endoteliais/enzimologia , Proteína ADAM12 , Proteína ADAM17 , Animais , Barreira Hematorretiniana/citologia , Barreira Hematorretiniana/enzimologia , Hipóxia Celular , Linhagem Celular , Membrana Celular/metabolismo , Claudina-5/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Retina/enzimologia , Vasos Retinianos/citologia , Vasos Retinianos/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA