Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Am J Pathol ; 192(11): 1633-1646, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36029802

RESUMO

Retinopathy of prematurity (ROP) is characterized by pathologic angiogenesis in retina, and remains a leading cause of blindness in children. Although enhanced extracellular adenosine is markedly increased in response to retinal hypoxia, adenosine acting at the A1 and A2A receptors has the opposite effect on pathologic angiogenesis. Herein, the oxygen-induced retinopathy (OIR) model of ROP was used to demonstrate that pharmacologic and genetic inactivation of CD73 (the key 5'-ectonucleotidase for extracellular generation of adenosine) did not affect normal retinal vasculature development but exacerbated intravitreal neovascularization at postnatal day (P) 17 and delayed revascularization at P21 of OIR. This exacerbated damage to retinal vessels by CD73 inactivation was associated with increased cellular apoptosis and microglial activation but decreased astrocyte function at P17 of OIR. Furthermore, pharmacologic blockade of equilibrative nucleoside transporter 1/2 (ENT1/2; bidirectional transport for controlling the balance of intracellular and extracellular adenosine) by 6-nitrobenzylthioinosine aggravated pathologic angiogenesis at P17 of OIR. Pharmacologic blockade of ENT1/2 and genetic inactivation of CD73 also aggravated avascular areas at the hyperoxia phase (P12) of OIR. Thus, disruption of CD73-derived extracellular adenosine or ENT1/2-mediated transport of adenosine flux across membrane aggravated the damage to retinal vessels. These findings support the role of adenosine as an endogenous protective regulator that limits oxygen-induced retinopathy. Thus, enhancing extracellular adenosine signaling represents a novel neuroprotection strategy for ROP by targeting CD73 and ENT1/2 activities.

2.
Molecules ; 27(10)2022 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-35630603

RESUMO

The Cassia (Leguminosae) genus has attracted a lot of attention as a prolific source of alkaloids and chromones with diverse structures and biological properties. The aim of this study is to screen the antiviral compounds from Cassia alata. The extract of the stem bark of this plant was separated using silica gel, MCI, ODS C18, and Sephadex LH-20 column chromatography, as well as semi-preparative HPLC. As a result, three new indole alkaloids, alataindoleins A-C (1-3); one new chromone, alatachromone A (4); and a new dimeric chromone-indole alkaloid, alataindolein D (5) were isolated. Their structures were determined by means of HRESIMS and extensive 1D and 2D NMR spectroscopic studies. Interestingly, alataindolein D (5) represents a new type of dimeric alkaloid with an unusual N-2-C-16' linkage, which is biogenetically derived from a chromone and an indole alkaloid via an intermolecular nucleophilic substitution reaction. Compounds 1-5 were tested for their anti-tobacco mosaic virus (TMV) and anti-rotavirus activities, and the results showed that compounds 2-4 showed high anti-TMV activities with inhibition rates of 44.4%, 66.5%, and 52.3%, respectively. These rates were higher than those of the positive control (with inhibition rate of 32.8%). Compounds 1 and 5 also showed potential anti-TMV activities with inhibition rates of 26.5% and 31.8%, respectively. In addition, compounds 1-5 exhibited potential anti-rotavirus activities with therapeutic index (TI) values in the range of 9.75~15.3. The successful isolation and structure identification of the above new compounds provided materials for the screening of antivirus drugs, and contributed to the development and utilization of C. alata.


Assuntos
Alcaloides , Cassia , Senna , Vírus do Mosaico do Tabaco , Alcaloides/farmacologia , Antivirais/química , Cassia/química , Cromonas/química , Alcaloides Indólicos , Casca de Planta
3.
Mol Med ; 24(1): 41, 2018 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-30134834

RESUMO

BACKGROUND: Retinopathy of prematurity (ROP) remains a major cause of childhood blindness and current laser photocoagulation and anti-VEGF antibody treatments are associated with reduced peripheral vision and possible delayed development of retinal vasculatures and neurons. In this study, we advanced the translational potential of adenosine A2A receptor (A2AR) antagonists as a novel therapeutic strategy for selectively controlling pathological retinal neovascularization in oxygen-induced retinopathy (OIR) model of ROP. METHODS: Developing C57BL/6 mice were exposed to 75% oxygen from postnatal (P) day 7 to P12 and to room air from P12 to P17 and treated with KW6002 or vehicle at different postnatal developmental stages. Retinal vascularization was examined by whole-mount fluorescence and cross-sectional hematoxylin-eosin staining. Cellular proliferation, astrocyte and microglial activation, and tip cell function were investigated by isolectin staining and immunohistochemistry. Apoptosis was analyzed by TUNEL assay. The effects of oxygen exposure and KW6002 treatment were analyzed by two-way ANOVA or Kruskal-Wallis test or independent Student's t-test or Mann-Whitney U test. RESULTS: The A2AR antagonist KW6002 (P7-P17) did not affect normal postnatal development of retinal vasculature, but selectively reduced avascular areas and neovascularization, with the reduced cellular apoptosis and proliferation, and enhanced astrocyte and tip cell functions in OIR. Importantly, contrary to our prediction that A2AR antagonists were most effective at the hypoxic phase with aberrantly increased adenosine-A2AR signaling, we discovered that the A2AR antagonist KW6002 mainly acted at the hyperoxic phase to confer protection against OIR as KW6002 treatment at P7-P12 (but not P12-P17) conferred protection against OIR; this protection was observed as early as P9 with reduced avascular areas and reduced cellular apoptosis and reversal of eNOS mRNA down-regulation in retina of OIR. CONCLUSIONS: As ROP being a biphasic disease, our identification of the hyperoxic phase as the effective window, together with selective and robust protection against pathological (but not physiological) angiogenesis, elevates A2AR antagonists as a novel therapeutic strategy for ROP treatment.


Assuntos
Antagonistas do Receptor A2 de Adenosina/uso terapêutico , Hiperóxia , Purinas/uso terapêutico , Neovascularização Retiniana/tratamento farmacológico , Retinopatia da Prematuridade/tratamento farmacológico , Animais , Camundongos Endogâmicos C57BL
4.
FASEB J ; 31(8): 3334-3348, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28420694

RESUMO

Retinopathy of prematurity (ROP) is the leading cause of childhood blindness, but current anti-VEGF therapy is concerned with delayed retinal vasculature, eye, and brain development of preterm infants. The clinical observation of reduced ROP severity in premature infants after caffeine treatment for apnea suggests that caffeine may protect against ROP. Here, we demonstrate that caffeine did not interfere with normal retinal vascularization development but selectively protected against oxygen-induced retinopathy (OIR) in mice. Moreover, caffeine attenuated not only hypoxia-induced pathologic angiogenesis, but also hyperoxia-induced vaso-obliteration, which suggests a novel protection window by caffeine. At the hyperoxic phase, caffeine reduced oxygen-induced neural apoptosis by adenosine A2A receptor (A2AR)-dependent mechanism, as revealed by combined caffeine and A2AR-knockout treatment. At the hypoxic phase, caffeine reduced microglial activation and enhanced tip cell formation by A2AR-dependent and -independent mechanisms, as combined caffeine and A2AR knockout produced additive and nearly full protection against OIR. Together with clinical use of caffeine in neonates, our demonstration of the selective protection against OIR, effective therapeutic window, adenosine receptor mechanisms, and neuroglial involvement provide the direct evidence of the novel effects of caffeine therapy in the prevention and treatment of ROP.-Zhang, S., Zhou, R., Li, B., Li, H., Wang, Y., Gu, X., Tang, L., Wang, C., Zhong, D., Ge, Y., Huo, Y., Lin, J., Liu, X.-L., Chen, J.-F. Caffeine preferentially protects against oxygen-induced retinopathy.


Assuntos
Cafeína/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Oxigênio/toxicidade , Vasos Retinianos/crescimento & desenvolvimento , Retinopatia da Prematuridade/prevenção & controle , Animais , Animais Recém-Nascidos , Endotélio Vascular/efeitos dos fármacos , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptor A1 de Adenosina/genética , Receptor A1 de Adenosina/metabolismo , Receptores A2 de Adenosina/genética , Receptores A2 de Adenosina/metabolismo , Vasos Retinianos/efeitos dos fármacos , Retinopatia da Prematuridade/etiologia
5.
Dalton Trans ; 52(43): 15767-15774, 2023 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-37847404

RESUMO

Hydrogen economy has emerged as a promising alternative to the current hydrocarbon economy. It involves harvesting renewable energy to split water into hydrogen and oxygen and then further utilising clean hydrogen fuel for various applications. The rational exploration of advanced non-precious metal bifunctional electrocatalysts for the oxygen evolution reaction (OER) and hydrogen evolution reaction (HER) is critical for efficient water splitting. Herein, an ultralow Ru-modified cobalt metal-organic framework (CoRu0.06-MOF/NF) two-dimensional nanosheet array bifunctional catalyst was fabricated through a strategy under mild experimental conditions. The obtained CoRu0.06-MOF/NF exhibited excellent bifunctional electrocatalytic activity and stability in alkaline media, with low overpotentials of 37 and 181 mV and significant durability for more than 95 and 110 h toward the HER and OER at 10 mA cm-2, respectively. The experimental results showed that the two-dimensional nanoarray structure had a large specific surface area and abundant exposed active sites. Additionally, ultralow Ru modification optimized the electronic structure and improved the conductivity of the cobalt metal-organic frameworks, thereby reducing the energy barrier of the rate-limiting step and accelerating the water splitting reaction.

6.
Dalton Trans ; 51(24): 9486-9494, 2022 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-35678597

RESUMO

The advancement of a bifunctional electrocatalyst consisting of Earth's rich elements and exhibiting high efficiency is the key to obtain hydrogen fuel by overall water splitting (OWS). Here, a facile and extensible hydrothermal synthesis of an electrocatalyst on iron foam (MoOx/Fe1-xS/IF) as a robust bifunctional catalyst with excellent catalytic activity is designed for the hydrogen evolution reaction (HER) with an overpotential of 142 mV at 100 mA cm-2, and for the OER with lower overpotentials of 300 and 500 mV at 100 and 1000 mA cm-2. The good activity is ascribed to the controllable morphology, stronger bonding of the catalyst to a substrate and optimized electronic configuration. When used as bifunctional electrocatalysts toward alkaline overall water splitting, MoOx/Fe1-xS/IF delivers a current density of 10 mA cm-2 at a low cell voltage of 1.56 V for 110 h. Such high performance coupled with low-cost iron-based materials suggests that the present strategy may open new avenues for the rational design of electrocatalysts and for use in practical water splitting.

7.
Nat Commun ; 8(1): 584, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28928465

RESUMO

Adenosine/adenosine receptor-mediated signaling has been implicated in the development of various ischemic diseases, including ischemic retinopathies. Here, we show that the adenosine A2a receptor (ADORA2A) promotes hypoxia-inducible transcription factor-1 (HIF-1)-dependent endothelial cell glycolysis, which is crucial for pathological angiogenesis in proliferative retinopathies. Adora2a expression is markedly increased in the retina of mice with oxygen-induced retinopathy (OIR). Endothelial cell-specific, but not macrophage-specific Adora2a deletion decreases key glycolytic enzymes and reduces pathological neovascularization in the OIR mice. In human primary retinal microvascular endothelial cells, hypoxia induces the expression of ADORA2A by activating HIF-2α. ADORA2A knockdown decreases hypoxia-induced glycolytic enzyme expression, glycolytic flux, and endothelial cell proliferation, sprouting and tubule formation. Mechanistically, ADORA2A activation promotes the transcriptional induction of glycolytic enzymes via ERK- and Akt-dependent translational activation of HIF-1α protein. Taken together, these findings advance translation of ADORA2A as a therapeutic target in the treatment of proliferative retinopathies and other diseases dependent on pathological angiogenesis.Pathological angiogenesis in the retina is a major cause of blindness. Here the authors show that adenosine receptor A2A drives pathological angiogenesis in the oxygen-induced retinopathy mouse model by promoting glycolysis in endothelial cells via the ERK/Akt/HIF-1α pathway, thereby suggesting new therapeutic targets for disease treatment.


Assuntos
Células Endoteliais/metabolismo , Receptor A2A de Adenosina/metabolismo , Doenças Retinianas/metabolismo , Neovascularização Retiniana/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Glicólise , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor A2A de Adenosina/genética , Retina/metabolismo , Retina/patologia , Doenças Retinianas/genética , Doenças Retinianas/patologia , Neovascularização Retiniana/genética , Neovascularização Retiniana/patologia
8.
Invest Ophthalmol Vis Sci ; 56(13): 8108-19, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26720463

RESUMO

PURPOSE: We critically evaluated the role of the adenosine A1 receptor (A1R) in normal development of retinal vasculature and pathogenesis of retinopathy of prematurity (ROP) by using the A1R knockout (KO) mice and oxygen-induced retinopathy (OIR) model. METHODS: Mice deficient in A1Rs and their wild-type (WT) littermates were examined during normal postnatal development or after being subjected to 75% oxygen from postnatal day (P) 7 to P12 and to room air from P12 to P17 (OIR model of ROP). Retinal vascularization was examined by whole-mount fluorescence and cross-sectional hematoxylin-eosin staining. Cellular proliferation, astrocyte and microglial activation, and tip cell function were determined by isolectin staining and immunohistochemistry. Apoptosis was determined by TUNEL assay. RESULTS: Genetic deletion of the A1R did not affect normal retinal vascularization during postnatal development with indistinguishable three-layer vascularization patterns in retina between WT and A1R KO mice. In the OIR model, genetic deletion of the A1R resulted in stage-specific effects: reduced hyperoxia-induced retinal vaso-obliteration at P12, but reduced avascular area and attenuated hypoxia-induced intraretinal revascularization without affecting intravitreal neovascularization at P17 and reduced avascular areas in retina at P21. These distinct effects of A1Rs on OIR were associated with A1R control of apoptosis mainly in inner and outer nuclear layers at the vaso-obliterative phase (P12) and the growth of endothelium tip cells at the vasoproliferative phase (P17), without modification of cellular proliferation, astrocytic activation, and tissue inflammation. CONCLUSIONS: Adenosine A1 receptor activity is not required for normal postnatal development of retinal vasculature but selectively controls hyperoxia-induced vaso-obliteration and hypoxia-driven revascularization by distinct cellular mechanisms.


Assuntos
Hiperóxia/fisiopatologia , Hipóxia/fisiopatologia , Receptor A1 de Adenosina/fisiologia , Retinopatia da Prematuridade/fisiopatologia , Animais , Estudos Transversais , Modelos Animais de Doenças , Humanos , Recém-Nascido , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxigênio/efeitos adversos , Neovascularização Retiniana/etiologia , Neovascularização Retiniana/patologia , Neovascularização Retiniana/fisiopatologia , Vasos Retinianos/fisiologia , Retinopatia da Prematuridade/etiologia , Retinopatia da Prematuridade/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA