Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Biochem Biophys Res Commun ; 613: 133-139, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35561580

RESUMO

Diabetic retinopathy (DR) is one of the vascular complications associated with diabetes mellitus. Pericyte loss is an early characteristic phenomenon in DR. However, the mechanism by which pericyte apoptosis occurs in DR is not fully understood. We have focused on the increased STAT3 activation in diabetic retinas because STAT3 activation is associated with inflammation, and persistent chronic inflammation is closely related to retinal lesions. In this study, we demonstrated that STAT3 was activated by IFN-γ and IL-6 that highly expressed in diabetic retinas. We identified TNF-α as a potent inducer of pericyte apoptosis in diabetic retinas from the gene expression analysis and found that STAT3 activation in microglia increased TNF-α expression in the diabetic retinas. We also demonstrated that increased TNF-α expression in microglia caused pericyte apoptosis through downregulating AKT/p70S6 kinase signaling. Moreover, we took advantage of mice lacking STAT3 in microglia and demonstrated that STAT3 ablation in microglia reduced the pericyte apoptosis and TNF-α expression in the diabetic retinas. These results suggest that STAT3 activation in microglia plays an important role in pericyte apoptosis in the diabetic retinas through increased TNF-α expression and provide STAT3 activation in microglia as a potential therapeutic target for preventing pericyte loss in DR.


Assuntos
Diabetes Mellitus , Retinopatia Diabética , Animais , Apoptose , Diabetes Mellitus/metabolismo , Retinopatia Diabética/metabolismo , Inflamação/patologia , Camundongos , Microglia/metabolismo , Pericitos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Retina/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
2.
J Cell Physiol ; 236(10): 7058-7070, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33754353

RESUMO

Diabetes mellitus (DM) characterized by hyperglycemia leads to a variety of complications, including cognitive impairment or memory loss. The hippocampus is a key brain area for learning and memory and is one of the regions that is most sensitive to diabetes. However, the pathogenesis of diabetic neuronal lesion is not yet completely understood. We focused on the association of microglia activation and brain lesions in diabetes. In this study, we investigated whether and how signal transducer and activator of transcription 3 (STAT3) activation in microglia affects neuronal lesions in diabetic brains. Using a streptozotocin-induced type 1 DM model, we showed enhanced hippocampal neuronal apoptosis that was associated with increased STAT3 activation. We found that hyperglycemia increased the expression of inflammatory cytokines such as interferon-γ (IFN-γ) and interleukin-6, in the diabetic hippocampus. In particular, IFN-γ induced autocrine activation of microglia, and STAT3 activation is important for this process. We also demonstrated that STAT3 activation in microglia increased tumor necrosis factor-α (TNF-α) expression; subsequently, TNF-α increased neuronal apoptosis by increasing reactive oxygen species (ROS) levels in the neuronal cells. We also took advantage of mice lacking STAT3 in microglia and demonstrated that depletion of microglial STAT3 reduced neuronal apoptosis in the diabetic hippocampus. Taken together, these results suggest that STAT3 activation in microglia plays an important role in hyperglycemia-induced neuronal apoptosis in the diabetic hippocampus and provide a potential therapeutic benefit of STAT3 inhibition in microglia for preventing diabetic neuronal lesions.


Assuntos
Apoptose , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Hipocampo/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Comunicação Autócrina , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Hipocampo/patologia , Humanos , Mediadores da Inflamação/metabolismo , Camundongos Knockout , Microglia/patologia , Neurônios/patologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
3.
Biochem Biophys Res Commun ; 546: 46-53, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33571904

RESUMO

Pericytes play a crucial role in preventing endothelial permeability by maintaining the integrity of tight junctions in endothelial cells; however, early pathological change in diabetic retinopathyis pericyte loss, which can lead to visual impairment by increasing endothelial permeability. Therefore, finding proteins and mechanisms that cause pericyte loss in diabetic retinopathy is beneficial for attenuating vision impairment. The present study focused on the effect of IL-1ß on pericyte loss and endothelial permeability in diabetic retinopathy. It was demonstrated that IL-1ß increased in the diabetic mouse retina and that the source of IL-1ß could be endothelial cells and microglia. IL-1ß induced pericyte apoptosis via NF-κB activation under high glucose conditions, but did not induce endothelial cell apoptosis. Moreover, IL-1ß did not affect permeability in the endothelial cell monolayer; however, when cocultured with pericytes and endothelial cells, it increased endothelial cell permeability by reducing the amount of tight junction protein in endothelial cells. Furthermore, NF-κB inhibitor restored the altered permeability and tight junction protein expression in endothelial cells induced by IL-1ß in cocultures of pericytes and endothelial cells. Collectively, IL-1ß induced pericyte apoptosis via NF-κB activation under high glucose conditions, thereby increasing endothelial permeability in diabetic retinopathy. Blocking IL-1ß/NF-κB signaling could be a promising therapeutic target to prevent pericyte loss in diabetic retinopathy.


Assuntos
Apoptose/efeitos dos fármacos , Retinopatia Diabética/patologia , Interleucina-1beta/farmacologia , NF-kappa B/metabolismo , Pericitos/efeitos dos fármacos , Animais , Permeabilidade Capilar/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Retinopatia Diabética/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Pericitos/citologia
4.
Microvasc Res ; 133: 104103, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33181170

RESUMO

Diabetic retinopathy (DR) is a disease that causes blindness due to vascular leakage or abnormal angiogenesis. Hepatocyte growth factor (HGF) is increased in the serum or vitreous fluid in proliferative diabetic retinopathy (PDR) patients, although the effect of HGF on the blood vessels remains unclear. This study focused on the effect of HGF on pericyte (PC) survival and endothelial cell (EC) permeability. It was demonstrated that HGF was increased in the diabetic mouse retina. However, HGF prevented PC apoptosis caused by TNF-α, which increased in the diabetic retinas both in vitro and in vivo. In addition, HGF was involved in PC survival by increasing the Akt signaling pathway. Moreover, HGF strengthened the EC tight junction in co-cultures of PCs and ECs by promoting PC survival, thereby reducing EC permeability. These results suggest that HGF may play a role in the prevention of increased vascular leakage by inhibiting the PC loss that occurs in DR to some extent. However, careful HGF reduction in DR might avoid an increase in PC loss.


Assuntos
Apoptose/efeitos dos fármacos , Retinopatia Diabética/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Pericitos/efeitos dos fármacos , Vasos Retinianos/efeitos dos fármacos , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Pericitos/metabolismo , Pericitos/patologia , Permeabilidade , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo , Junções Íntimas/patologia
5.
Molecules ; 26(4)2021 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-33546250

RESUMO

As abnormal angiogenesis is associated with exacerbation of various diseases, precise control over angiogenesis is imperative. Vascular endothelial growth factor (VEGF), the most well-known angiogenic factor, binds to VEGF receptor (VEGFR), activates various signaling pathways, and mediates angiogenesis. Therefore, blocking the VEGF-induced angiogenic response-related signaling pathways may alleviate various disease symptoms through inhibition of angiogenesis. Ulmus davidiana is a safe natural product that has been traditionally consumed, but its effects on endothelial cells (ECs) and the underlying mechanism of action are unclear. In the present study, we focused on the effect of a 60% edible ethanolic extract of U. davidiana (U60E) on angiogenesis. U60E inhibited the VEGF-mediated proliferation, tube formation, and migration ability of ECs. Mechanistically, U60E inhibited endothelial nitric oxide synthase activation and nitric oxide production by blocking the protein kinase B signaling pathway activated by VEGF and consequently inhibiting proliferation, tube formation, and migration of ECs. These results suggest that U60E could be a potential and safe therapeutic agent capable of suppressing proangiogenic diseases by inhibiting VEGF-induced angiogenesis.


Assuntos
Inibidores da Angiogênese , Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Patológica/tratamento farmacológico , Extratos Vegetais , Ulmus/química , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacologia , Etanol/química , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia
6.
J Cell Physiol ; 234(4): 3837-3849, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30256387

RESUMO

Transforming growth factor-ß (TGF-ß) is a multifunctional cytokine that is known to modulate various aspects of endothelial cell (EC) biology. Retinal pigment epithelium (RPE) is important for regulating angiogenesis of choriocapillaris and one of the main cell sources of TGF-ß secretion, particularly TGF-ß2. However, it is largely unclear whether and how TGF-ß2 affects angiogenic responses of ECs. In the current study, we demonstrated that TGF-ß2 reduces vascular endothelial growth factor receptor-2 (VEGFR-2) expression in ECs and thereby inhibits vascular endothelial growth factor (VEGF) signaling and VEGF-induced angiogenic responses such as EC migration and tube formation. We also demonstrated that the reduction of VEGFR-2 expression by TGF-ß2 is due to the suppression of JNK signaling. In coculture of RPE cells and ECs, RPE cells decreased VEGFR-2 levels in ECs and EC migration. In addition, we showed that TGF-ß2 derived from RPE cells is involved in the reduction of VEGFR-2 expression and inhibition of EC migration. These results suggest that TGF-ß2 plays an important role in inhibiting the angiogenic responses of ECs during the interaction between RPE cells and ECs and that angiogenic responses of ECs may be amplified by a decrease in TGF-ß2 expression in RPE cells under pathologic conditions.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Fisiológica , Comunicação Parácrina , Epitélio Pigmentado da Retina/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Movimento Celular , Células Cultivadas , Técnicas de Cocultura , Regulação para Baixo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fosforilação , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Via Secretória , Transdução de Sinais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
7.
Glia ; 67(2): 321-331, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30444022

RESUMO

Inner and outer blood-retinal barriers (BRBs), mainly composed of retinal endothelial cells and retinal pigment epithelial (RPE) cells, respectively, maintain the integrity of the retinal tissues. In this study, we aimed to investigate the mechanisms of the outer BRB disruption regarding the interaction between RPE and microglia. In mice with high-fat diet-induced obesity and streptozotocin-induced hyperglycemia, microglia accumulated on the RPE layer, as in those after intravitreal injection of interleukin (IL)-6, which is elevated in ocular fluids of patients with diabetic retinopathy. Although IL-6 did not directly affect the levels of zonula occludens (ZO)-1 and occludin in RPE cells, IL-6 increased VEGFA mRNA in RPE cells to recruit microglial cells. In microglial cells, IL-6 upregulated the mRNA levels of MCP1, MIP1A, and MIP1B, to amplify the recruitment of microglial cells. In this manner, IL-6 modulated RPE and microglial cells to attract microglial cells on RPE cells. Furthermore, IL-6-treated microglial cells produced and secreted tumor necrosis factor (TNF)-α, which activated NF-κB and decreased the levels of ZO-1 in RPE cells. As STAT3 inhibition reversed the effects of IL-6-treated microglial cells on the RPE monolayer in vitro, it reduced the recruitment of microglial cells and the production of TNF-α in RPE tissues in streptozotocin-treated mice. Taken together, IL-6-treated RPE and microglial cells amplified the recruitment of microglial cells and IL-6-treated microglial cells produced TNF-α to disrupt the outer BRB in diabetic retinopathy.


Assuntos
Barreira Hematorretiniana/fisiopatologia , Retinopatia Diabética/patologia , Microglia/fisiologia , Epitélio Pigmentado da Retina/fisiologia , Inibidores da Angiogênese/uso terapêutico , Animais , Antibióticos Antineoplásicos/toxicidade , Barreira Hematorretiniana/efeitos dos fármacos , Retinopatia Diabética/induzido quimicamente , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-6/metabolismo , Interleucina-6/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Piridinas/farmacologia , Retina/patologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Estreptozocina/toxicidade , Tirfostinas/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo
8.
Biochem Biophys Res Commun ; 518(2): 286-293, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31427082

RESUMO

The regulation of endothelial cell (EC) permeability is critical for the physiological homeostasis of blood vessels and tissues. The elevation of pro-inflammatory cytokines is highly associated with lesions, such as the increased vascular permeability of diabetic retinas. We have previously reported that interleukin-6 (IL-6) increases EC permeability through the downregulation of tight junction protein expression. Angiopoietin 1 (Ang1) has an anti-permeability function, but the effect of Ang1 on vascular permeability induced by inflammatory cytokines is unclear. In the present study, we investigated the effect of Ang1 on IL-6-induced EC permeability and its underlying molecular mechanisms. We demonstrated that Ang1 inhibited the IL-6-induced increase in EC permeability by inhibiting the reductions in the levels of tight junction protein ZO-1 and occludin, which was related to the decrease in vascular endothelial growth factor (VEGF) secretion through the inhibition of STAT3 activation by Ang1. Mechanistically, Ang1 induced the dissociation of the tyrosine phosphatase SHP-1 from the Tie2 receptor and increased the binding of SHP-1 to JAK1, JAK2, and STAT3, which are IL-6 downstream signaling proteins. We conclude that SHP-1 plays an important role in the Ang1-induced inhibition of JAK/STAT3 signaling. These results provide evidence for a potential beneficial role of Ang1 in suppressing the vascular permeability induced by the pro-inflammatory cytokine IL-6 in diabetic retinopathy.


Assuntos
Angiopoietina-1/metabolismo , Células Endoteliais/metabolismo , Interleucina-6/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Células Cultivadas , Humanos , Interleucina-6/metabolismo , Permeabilidade
9.
FASEB J ; 32(5): 2324-2338, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29269397

RESUMO

Pericytes (PCs) are crucial in maintaining the quiescence of endothelial cells (ECs) and the integrity of EC tight junctions. Especially in diabetic retinopathy (DR), PC loss is one of the early pathologic changes in capillaries of diabetic retinas. Thus, preventing PC loss is beneficial for attenuating vision impairment in patients with DR. Although many studies have revealed the mechanism of PC loss in retinas, little is known about the mechanisms that increase PC survival. We focused on the effect of ß-adrenergic receptor agonists (ß-agonists) on PC loss in diabetic retinas. In this study, ß-agonists increased the cell viability of PCs by increasing PC survival and proliferation. Mechanistically, ß-agonist-induced protein kinase B activation in PCs reduced PC apoptosis in response to various stimuli. ß2-agonists more potently increased PC survival than ß1-agonists. ß2-Agonist reduced vascular leakage and PC loss in retinas of mice with streptozotocin-induced diabetes. In cocultures of PCs and ECs, ß2-agonists restored the altered permeability and ZO-1 expression in ECs induced by PC loss. We concluded that ß-agonists, especially ß2-agonists, increase PC survival, thereby preventing diabetes-induced PC loss in retinas. These results provide a potential therapeutic benefit of ß-agonists for preventing PC loss in DR.-Yun, J.-H., Jeong, H.-S., Kim, K.-J., Han, M. H., Lee, E. H., Lee, K., Cho, C.-H. ß-Adrenergic receptor agonists attenuate pericyte loss in diabetic retinas through Akt activation.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Retinopatia Diabética/tratamento farmacológico , Pericitos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Retina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Masculino , Camundongos , Pericitos/patologia , Retina/patologia , Proteína da Zônula de Oclusão-1/biossíntese
10.
Biochem Biophys Res Commun ; 503(4): 2792-2799, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30100069

RESUMO

Retinopathy of prematurity (ROP) is an eye disease that causes blindness due to delayed vascular growth, retinal ischemia, and resulting abnormal angiogenesis. Nonselective ß-antagonist propranolol is in clinical trials for the treatment of ROP due to its effect of reducing VEGF expression and inhibiting retinal angiogenesis in oxygen-induced ROP models (OIR), but the mechanism by which propranolol acts on ROP vessels is still unclear. In the present study, we have focused on the effect of propranolol on pericyte survival and vascular permeability. We demonstrated that propranolol increases pericyte apoptosis more sensitively than endothelial cells (ECs), thereby weakening EC tight junctions to increase endothelial permeability in co-cultures of pericytes and ECs. Mechanistically, pericyte apoptosis by propranolol was due to the inhibition of Akt signaling pathway. We also demonstrated that propranolol increases pericyte loss and vascular permeability of retinal vessels in a mouse model of OIR. These results suggest that propranolol may be negative for blood vessels in retinas of OIR, and that the efficacy of propranolol for the treatment of ROP needs to be more thoroughly verified.


Assuntos
Apoptose/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Hiperóxia/induzido quimicamente , Propranolol/farmacologia , Retinopatia da Prematuridade/induzido quimicamente , Vasodilatadores/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/genética , Técnicas de Cocultura , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hiperóxia/genética , Hiperóxia/metabolismo , Hiperóxia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/administração & dosagem , Pericitos/citologia , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neovascularização Retiniana/induzido quimicamente , Neovascularização Retiniana/genética , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/metabolismo , Retinopatia da Prematuridade/patologia , Transdução de Sinais , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo , Junções Íntimas/ultraestrutura , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
J Cell Physiol ; 232(5): 1123-1134, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27580405

RESUMO

Vascular inflammation is characteristic feature of diabetic retinopathy. In diabetic retina, a variety of the pro-inflammatory cytokines are elevated and involved in endothelial dysfunction. STAT3 transcription factor has been implicated in mediating cytokine signaling during vascular inflammation. However, whether and how STAT3 is involved in the direct regulation of the endothelial permeability is currently undefined. Our studies revealed that IL-6-induced STAT3 activation increases retinal endothelial permeability and vascular leakage in retinas of mice through the reduced expression of the tight junction proteins ZO-1 and occludin. In a co-culture model with microglia and endothelial cells under a high glucose condition, the microglia-derived IL-6 induced STAT3 activation in the retinal endothelial cells, leading to increasing endothelial permeability. In addition, IL-6-induced STAT3 activation was independent of ROS generation in the retinal endothelial cells. Moreover, we demonstrated that STAT3 activation downregulates the ZO-1 and occludin levels and increases the endothelial permeability through the induction of VEGF production in retinal endothelial cells. These results suggest the potential importance of IL-6/STAT3 signaling in regulating endothelial permeability and provide a therapeutic target to prevent the pathology of diabetic retinopathy. J. Cell. Physiol. 232: 1123-1134, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Regulação para Baixo , Células Endoteliais/metabolismo , Ocludina/metabolismo , Vasos Retinianos/patologia , Fator de Transcrição STAT3/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo , Animais , Linhagem Celular , Permeabilidade da Membrana Celular , Regulação para Baixo/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Glucose/toxicidade , Humanos , Interleucina-6/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Retina/efeitos dos fármacos , Retina/patologia , Vasos Retinianos/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Biochem Biophys Res Commun ; 454(2): 282-8, 2014 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-25450390

RESUMO

Hemangioma is a benign tumor derived from abnormal blood vessel growth. Unlike other vascular tumor counterparts, a hemangioma is known to proliferate during its early stage but it is followed by a stage of involution where regression of the tumor occurs. The critical onset leading to the involution of hemangioma is currently not well understood. This study focused on the molecular identities of the involution of hemangioma. We demonstrated that a soluble factor released from the involuting phase of hemangioma-derived endothelial cells (HemECs) and identified pigment epithelium-derived factor (PEDF) as an anti-angiogenic factor that was associated with the growth inhibition of the involuting HemECs. The growth inhibition of the involuting HemECs was reversed by suppression of PEDF in the involuting HemECs. Furthermore, we found that PEDF was more up-regulated in the involuting phase of hemangioma tissues than in the proliferating or the involuted. Taken together, we propose that PEDF accelerates the involution of hemangioma by growth inhibition of HemECs in an autocrine manner. The regulatory mechanism of PEDF expression could be a potential therapeutic target to treat hemangiomas.


Assuntos
Vasos Sanguíneos/patologia , Proliferação de Células , Células Endoteliais/patologia , Proteínas do Olho/metabolismo , Hemangioma/metabolismo , Hemangioma/patologia , Fatores de Crescimento Neural/metabolismo , Serpinas/metabolismo , Apoptose , Vasos Sanguíneos/metabolismo , Células Endoteliais/metabolismo , Proteínas do Olho/genética , Hemangioma/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Neovascularização Patológica , Fatores de Crescimento Neural/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Serpinas/genética , Células Tumorais Cultivadas
13.
Front Vet Sci ; 11: 1411886, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39011319

RESUMO

Acute lung injury is an acute inflammation disorder that disrupts the lung endothelial and epithelial barriers. In this study, we investigated the extracellular vesicles (EVs) obtained via priming inflammatory cytokines such as tumor necrosis factor (TNF)-α and interferon (IFN)-γ on canine adipose mesenchymal stem cells in improving their anti-inflammatory and/or immunosuppressive potential, and/or their ability to alleviate lipopolysaccharide-induced lung injury in vitro. We also explored the correlation between epithelial-to-mesenchymal transition and the inflammatory repressive effect of primed EVs. Using small RNA-Seq, we confirmed that miR-16 and miR-502 significantly increased in EVs from TNF-α and IFN-γ-primed canine adipose mesenchymal stem cells. The pro and anti-inflammatory cytokines were analyzed in a lipopolysaccharide-induced lung injury model and we found that the EV anti-inflammatory effect improved on priming with inflammatory cytokines. EVs obtained from primed stem cells effectively suppress endothelial-to-mesenchymal transition in a lung injury model. Our results suggest a potential therapeutic approach utilizing EVs obtained from adipose mesenchymal stem cells primed with TNF-α and IFN-γ against lung inflammation and endothelial to mesenchymal transition.

14.
Am J Cancer Res ; 14(2): 727-743, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38455399

RESUMO

Hypoxia-inducible factors (HIFs) regulate cellular oxygen balance and play a central role in cancer metastasis and angiogenesis. Despite extensive research on HIFs, successful therapeutic strategies remain limited due to the intricate nature of their regulation. In this study, we identified SPATA20, a relatively understudied protein with a thioredoxin-like domain, as an upstream regulator of HIF-1α. Depleting SPATA20 induced HIF-1α expression, suggesting a tumor-suppressive role for SPATA20 in cancer cells. SPATA20 depletion increased HIF-1α protein levels and transcriptional activity without affecting its degradation. It appears that SPATA20 inhibits the de novo synthesis of HIF-1α, possibly by repressing the cap-dependent translation process involving AKT phosphorylation. Additionally, depletion of SPATA20 promoted cancer cell migration and invasion, which can be reversed by pharmacological inhibition of HIF-1α. Clinical data analysis revealed an inverse correlation between SPATA20 expression and colorectal cancer progression, providing evidence of its role as a potential biomarker. Utilizing SPATA20 as an indicator for HIF-1α-targeting therapy may be an attractive strategy for treating patients with hypoxia-driven cancers. In conclusion, this study demonstrates that SPATA20 deficiency promotes cancer progression by activating the HIF-1α signaling pathway.

15.
Biochem Biophys Res Commun ; 436(4): 691-7, 2013 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-23770419

RESUMO

Despite the altered expression of Tie receptors and angiopoietin ligands during hypoxic conditions, the effect of hypoxia on Tie-mediated endothelial responses has not been elucidated. In this study, we found that hypoxia increased Tie receptor expression but attenuated angiopoietin-1 (Ang1)-induced Tie2 activity, including Tie2 phosphorylation, Tie2 downstream signaling activation, and endothelial cell tube formation. However, Ang1 binding to endothelial cells was increased during hypoxic conditions. We demonstrated that Tie1 suppression restored the Tie2 activity and that Tie1-mediated Tie2 suppression was independent of tyrosine phosphatase activity. These results suggest that under hypoxic conditions, Tie1 is critical for reducing Ang1-induced Tie2 activity and angiogenesis.


Assuntos
Angiopoietina-1/metabolismo , Hipóxia Celular , Endotélio Vascular/metabolismo , Receptor de TIE-1/metabolismo , Células Cultivadas , Endotélio Vascular/enzimologia , Humanos , Transdução de Sinais
16.
Acta Pharmacol Sin ; 34(9): 1208-16, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23852087

RESUMO

AIM: Disulfiram is an aldehyde dehydrogenase inhibitor that was used to treat alcoholism and showed anticancer activity, but its anticancer mechanism remains unclear. The aim of this study was to investigate the effects of disulfiram on the hypoxia-inducible factor (HIF)-driven tumor adaptation to hypoxia in vitro. METHODS: Hep3B, Huh7 and HepG2 hepatoma cells were incubated under normoxic (20% O2) or hypoxic (1% O2) conditions for 16 h. The expression and activity of HIF-1α and HIF-2α proteins were evaluated using immunoblotting and luciferase reporter assay, respectively. Semi-quantitative RT-PCR was used to analyze HIF-mediated gene expression. Endothelial tubule formation assay was used to evaluate the anti-angiogenic effect. RESULTS: Hypoxia caused marked expression of HIF-1α and HIF-1α in the 3 hepatoma cell lines, dramatically increased HIF activity and induced the expression of HIF downstream genes (EPO, CA9, VEGF-A and PDK1) in Hep3B cells. HIF-2α expression was positively correlated with the induction of hypoxic genes (CA9, VEGF-A and PDK1). Moreover, hypoxia markedly increased VEGF production and angiogenic potential of Hep3B cells. Disulfiram (0.3 to 2 µmol/L) inhibited hypoxia-induced gene expression and HIF activity in a dose-dependent manner. Disulfiram more effectively suppressed the viability of Hep3B cells under hypoxia, but it did not affect the cell cycle. Overexpression of HIF-2α in Hep3B cells reversed the inhibitory effects of disulfiram on hypoxia-induced gene expression and cell survival under hypoxia. CONCLUSION: Disulfiram deregulates the HIF-mediated hypoxic signaling pathway in hepatoma cells, which may contribute to its anticancer effect. Thus, disulfiram could be used to treat solid tumors that grow in a HIF-dependent manner.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Carcinoma Hepatocelular/metabolismo , Dissulfiram/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Adaptação Fisiológica/efeitos dos fármacos , Adaptação Fisiológica/fisiologia , Carcinoma Hepatocelular/tratamento farmacológico , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/biossíntese
17.
Front Endocrinol (Lausanne) ; 14: 1138676, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37234799

RESUMO

Diabetic retinopathy (DR) is a disease that causes visual deficiency owing to vascular leakage or abnormal angiogenesis. Pericyte apoptosis is considered one of the main causes of vascular leakage in diabetic retina, but there are few known therapeutic agents that prevent it. Ulmus davidiana is a safe natural product that has been used in traditional medicine and is attracting attention as a potential treatment for various diseases, but its effect on pericyte loss or vascular leakage in DR is not known at all. In the present study, we investigated on the effects of 60% edible ethanolic extract of U. davidiana (U60E) and catechin 7-O-ß-D-apiofuranoside (C7A), a compound of U. davidiana, on pericyte survival and endothelial permeability. U60E and C7A prevented pericyte apoptosis by inhibiting the activation of p38 and JNK induced by increased glucose and tumor necrosis factor alpha (TNF-α) levels in diabetic retina. Moreover, U60E and C7A reduced endothelial permeability by preventing pericyte apoptosis in co-cultures of pericytes and endothelial cells. These results suggest that U60E and C7A could be a potential therapeutic agent for reducing vascular leakage by preventing pericyte apoptosis in DR.


Assuntos
Diabetes Mellitus , Retinopatia Diabética , Ulmus , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/prevenção & controle , Retinopatia Diabética/patologia , Pericitos , Células Endoteliais/patologia , Apoptose , Diabetes Mellitus/patologia
18.
Sci Rep ; 12(1): 22484, 2022 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-36577816

RESUMO

Dysregulation of inflammasome activation induces chronic and excess inflammation resulting in several disorders, such as metabolic disorders and cancers. Thus, screening for its regulator derived from natural materials has been conducted progressively. JC2-11 (JC) was designed to enhance the antioxidant activity based on a chalcone, which is abundant in edible plants and a precursor of flavonoids. This study examined the effects of JC on inflammasome activation in human and murine macrophages. JC inhibited the secretion of interleukin (IL)-1ß and lactate dehydrogenases, and the cleavage of caspase-1 and gasdermin D in response to the tested activators (i.e., NLRP3, NLRC4, AIM2, and non-canonical inflammasome triggers). In addition, JC attenuated IL-1ß secretion from lipopolysaccharide (LPS)-injected mice, an inflammasome-mediating disease model. Mechanistically, JC blocked the expression of the inflammasome components during the priming step of the inflammasome, and interrupted the production of mitochondrial reactive oxygen species. In addition, JC inhibited the activity of caspase-1. In conclusion, JC may be a candidate pan-inflammasome inhibitor.


Assuntos
Chalcona , Inflamassomos , Humanos , Animais , Camundongos , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Chalcona/farmacologia , Macrófagos/metabolismo , Caspase 1/metabolismo , Interleucina-1beta/metabolismo , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo
19.
Toxics ; 10(4)2022 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-35448455

RESUMO

We examined the protective effects of esculetin and fucoidan against the neurotoxicity of ZnO NPs in rats. Ninety rats were divided into nine groups and pre-treated with esculetin or fucoidan 1 h before ZnO NP administration on a daily basis for 2 weeks. Serum and brain homogenates were examined by enzyme-linked immunosorbent assay (ELISA), and neurons, microglia, and astrocytes in the hippocampal region were examined with immunohistochemical analysis. The serum levels of interleukin-1-beta (IL-1ß), 3-nitrotyrosine (3-NT), superoxide dismutase (SOD), and 8-hydroxy-2'-deoxyguanosine (8-OHdG) were altered in the ZnO NP treatment groups. Brain IL-1ß and TNF-α levels were elevated after ZnO NP administration, and these effects were inhibited by esculetin and fucoidan. SOD, 8-OHdG, and acetylcholinesterase (AChE) levels in the brain were decreased after ZnO NP administration. The brain levels of beclin-1 and caspase-3 were elevated after ZnO NP treatment, and these effects were significantly ameliorated by esculetin and fucoidan. The number of reactive astrocytes measured by counting glial fibrillary acidic protein (GFAP)-positive cells, but not microglia, increased following ZnO NP treatment, and esculetin and fucoidan ameliorated the changes. Esculetin and fucoidan may be beneficial for preventing ZnO NP-mediated autophagy and apoptosis by the modulation of reactive astrocyte and proinflammatory cytokines in the rat brain.

20.
Front Bioeng Biotechnol ; 9: 673314, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34169066

RESUMO

Alnus sibirica Fisch. ex Turcz (ASFT), belonging to the family of Betulaceae, grows naturally in Asia, Europe, and America. The aims of this study are determining the efficacy of various biomarkers related to hair loss, evaluated by extracting the branch with 60% alcohol, and purely separating diarylheptanoid oregonin, an indicator and active substance, from 60% alcohol extract of the tree. To determine the preventive effects on hair loss, we investigated the anti-oxidative and anti-apoptotic effects on hydrogen peroxide-induced cytotoxicity on human hair dermal papilla cells using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and Western blotting analysis for proving of apoptosis-related marker alteration, respectively. Moreover, we examined the ameliorative effects of 60% alcohol extract of the tree and oregonin against changes of oxidative stress-induced cytokine and testosterone-induced dihydrotestosterone production as crucial pathways of the hair loss mechanism. These results suggest that 60% alcohol extract of the tree and oregonin were available as novel natural materials for maintaining hair health in mammals.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA