Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Am J Physiol Cell Physiol ; 325(5): C1252-C1266, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37694287

RESUMO

Hydrogen sulfide (H2S) promotes microangiogenesis and revascularization after ischemia. Neovascularization starts with the destruction of intercellular junctions and is accompanied by various endothelial cell angiogenic behaviors. Follistatin-like 1 (FSTL1) is a cardiovascular-protective myokine that works against ischemic injury. The present study examined whether FSTL1 was involved in H2S-induced angiogenesis and explored the underlying molecular mechanism. We observed that H2S accelerated blood perfusion after ischemia in the mouse hindlimb ischemia model. Western blot analysis showed that H2S stabilized FSTL1 transcript and increased FSTL1 and Human antigen R (HuR) levels in skeletal muscle. RNA-interference HuR significantly inhibited the H2S-promoted increase in FSTL1 levels. Exogenous FSTL1 promoted the wound-healing migration of human umbilical vein endothelial cells (HUVECs) and increased monolayer endothelial barrier permeability. Immunostaining showed that FSTL1 increased interendothelial gap formation and decreased VE-Cadherin, Occludin, Connexin-43, and Claudin-5 expression. In addition, FSTL1 significantly increased the phosphorylation of Src and VEGFR2. However, the Src inhibitor, not the VEGFR2 inhibitor, could block FSTL1-induced effects in angiogenesis. In conclusion, we demonstrated that H2S could upregulate the expression of FSTL1 by increasing the HuR levels in skeletal muscle, and paracrine FSTL1 could initiate angiogenesis by opening intercellular junctions via the Src signaling pathway.NEW & NOTEWORTHY The myocyte-derived paracrine protein FSTL1 acts on vascular endothelial cells and initiates the process of angiogenesis by opening the intercellular junction via activating Src kinase. H2S can significantly upregulate FSTL1 protein levels in skeletal muscles by increasing HuR expression.

2.
Sheng Li Xue Bao ; 75(3): 317-327, 2023 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-37340641

RESUMO

The present study aimed to investigate the protective effect of S-propargyl-cysteine (SPRC) on atherosclerosis progression in mice. A mouse model of vulnerable atherosclerotic plaque was created in ApoE-/- mice by carotid artery tandem stenosis (TS) combined with a Western diet. Macrophotography, lipid profiles, and inflammatory markers were measured to evaluate the antiatherosclerotic effects of SPRC compared to atorvastatin as a control. Histopathological analysis was performed to assess the plaque stability. To explore the protective mechanism of SPRC, human umbilical vein endothelial cells (HUVECs) were cultured in vitro and challenged with oxidized low-density lipoprotein (ox-LDL). Cell viability was determined with a Cell Counting Kit-8 (CCK-8). Endothelial nitric oxide synthase (eNOS) phosphorylation and mRNA expression were detected by Western blot and RT-qPCR respectively. The results showed that the lesion area quantified by en face photographs of the aortic arch and carotid artery was significantly less, plasma total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) were reduced, plaque collagen content was increased and matrix metalloproteinase-9 (MMP-9) was decreased in 80 mg/kg per day SPRC-treated mice compared with model mice. These findings support the role of SPRC in plaque stabilization. In vitro studies revealed that 100 µmol/L SPRC increased the cell viability and the phosphorylation level of eNOS after ox-LDL challenge. These results suggest that SPRC delays the progression of atherosclerosis and enhances plaque stability. The protective effect may be at least partially related to the increased phosphorylation of eNOS in endothelial cells.


Assuntos
Aterosclerose , Placa Aterosclerótica , Animais , Humanos , Camundongos , Colesterol/metabolismo , Cisteína/farmacologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Lipoproteínas LDL/farmacologia , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia
4.
Sheng Li Xue Bao ; 74(6): 979-992, 2022 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-36594386

RESUMO

Skin wound healing tends to slow down with aging, which is detrimental to both minor wound recovery in daily life and the recovery after surgery. The aim of current study was to explore the effect of histone deacetylase 6 (HDAC6) on wound healing during aging. Cultured human dermal fibroblasts (HDFs) and mouse full-thickness skin wound model were used to explore the functional changes of replicative senescent dermal fibroblasts and the effect of aging on skin wound healing. Scratch wound healing assay revealed significantly decreased migration speed of senescent HDFs, and BrdU incorporation assay indicated their considerably retardant proliferation. The protein expression levels of collagen and HDAC6 were significantly decreased in both senescent HDFs and skin tissues from aged mice. HDAC6 activity inhibition with highly selective inhibitor tubastatin A (TsA) or HDAC6 knockdown with siRNA decreased the migration speed of HDFs and considerably suppressed fibroblast differentiation induced by transforming growth factor-ß1 (TGF-ß1), which suggests the involvement of HDAC6 in regulating fundamental physiological activities of dermal fibroblasts. In vivo full-thickness skin wound healing was significantly delayed in young HDAC6 knockout mice when compared with young wild type mice. In addition, the wound healing was significantly slower in aged wild type mice than that in young wild type mice, and became even worse in aged HDAC6 knockout aged mice. Compared to the aged wild type mice, aged HDAC6 knockout mice exhibited delayed angiogenesis, reduced collagen synthesis, and decreased collagen deposition in skin wounds. Together, these results suggest that delayed skin wound healing in aged mice is associated with impaired fibroblast function. Adequate expression and activity of HDAC6 are required for fibroblasts migration and differentiation.


Assuntos
Pele , Cicatrização , Humanos , Animais , Camundongos , Idoso , Desacetilase 6 de Histona , Movimento Celular , Colágeno/metabolismo , Colágeno/farmacologia , Fibroblastos , Camundongos Knockout , Células Cultivadas
5.
Adv Exp Med Biol ; 1315: 1-16, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34302686

RESUMO

Hydrogen sulfide, a small molecule, produced by endogenous enzymes, such as CTH, CBS, and MPST using L-cysteine as substrates, has been reported to have numerous protective effects. However, the key problem that the target of H2S and how it can affect the structure and activity of biological molecules is still unknown. Till now, there are two main theories of its working mechanism. One is that H2S can modify the free thiol in cysteine to produce the persulfide state of the thiol and the sulfhydration of cysteine can significantly change the structure and activity of target proteins. The other theory is that H2S, as an antioxidant molecule, can directly break the disulfide bond in target proteins, and the persulfide state of thiol can be an intermediate product during the reaction. Both phenomena exit for no doubt since they are both supported by large amounts of experiments. Here, we will summarize both theories and try to discuss which one is the more effective or direct mechanism for H2S and what is the relationship between them. Therefore, we will discover more protein targets of H2S with the mechanism and understand more about the effect of this small molecule.


Assuntos
Sulfeto de Hidrogênio , Cisteína , Proteínas/genética , Compostos de Sulfidrila
6.
Am J Physiol Cell Physiol ; 319(6): C1082-C1096, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32938225

RESUMO

Endogenous hydrogen sulfide (H2S) affects cholesterol homeostasis and liver X receptor α (LXRα) expression. However, whether low-density lipoprotein (LDL) receptor (LDLR), a key player in cholesterol homeostasis, is regulated by exogenous H2S through LXRα signaling has not been determined. We investigated the effects of sodium hydrosulfide (NaHS, H2S donor) on LDLR expression in the presence or absence of LXR agonists, T0901317 or GW3965 in HepG2 cells. We found that H2S strongly accumulated LDLR precursor in the presence of T0901317. Hence, LDLR transcription and the genes involved in LDLR precursor maturation and degradation were studied. T0901317 increased the LDLR mRNA level, whereas H2S did not affect LDLR transcription. H2S had no significant effect on the expression of LXRα and inducible degrader of LDLR (IDOL). H2S and T0901317 altered mRNA levels of several enzymes for N- and O-glycosylation and endoplasmic reticulum (ER) chaperones assisting LDLR maturation, but did not affect their protein levels. H2S decreased proprotein convertase subtilisin/kexin type 9 (PCSK9) protein levels and its mRNA level elevated by T0901317. T0901317 with PCSK9 siRNA also accumulated LDLR precursor as did T0901317 with H2S. High glucose increased PCSK9 protein levels and attenuated LDLR precursor accumulation induced by T0901317 with H2S. Taken together, H2S accumulates LDLR precursor by downregulating PCSK9 expression but not through the LXRα-IDOL pathway, LDLR transcriptional activation, or dysfunction of glycosylation enzymes and ER chaperones. These results also indicate that PCSK9 plays an important role in LDLR maturation in addition to its well-known effect on the degradation of LDLR mature form.


Assuntos
Sulfeto de Hidrogênio/metabolismo , Receptores X do Fígado/metabolismo , Pró-Proteína Convertase 9/metabolismo , Receptores de LDL/metabolismo , Benzoatos/farmacologia , Benzilaminas/farmacologia , Linhagem Celular Tumoral , Colesterol/metabolismo , Retículo Endoplasmático/fisiologia , Glicosilação/efeitos dos fármacos , Células Hep G2 , Homeostase/fisiologia , Humanos , Hidrocarbonetos Fluorados/farmacologia , Receptores X do Fígado/agonistas , Pró-Proteína Convertase 9/genética , Interferência de RNA , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Sulfetos/farmacologia , Sulfonamidas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional/genética
7.
Am J Physiol Cell Physiol ; 318(5): C857-C869, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32186933

RESUMO

Diabetes (especially Type II) is one of the primary threats to cardiovascular health. Wound healing defects and vascular dysfunction are common in diabetic patients, and the primary cause of deterioration is sustained high plasma glucose. microRNA, a noncoding RNA, has regulatory functions that are critical to maintaining homeostasis. MicroRNA (miR)-126-3p is a potential diabetes biomarker and a proangiogenic factor, and its plasma level decreases in diabetic patients. Previous studies have revealed the proangiogenic character of the gasotransmitter hydrogen sulfide (H2S). However, little is known about the relationship between H2S and miR-126-3p when the extracellular glucose level is high, let alone their influences on deteriorated endothelial cell migration, a key component of angiogenesis, which is crucial for wound healing. Human umbilical vein endothelial cells (HUVECs) were treated with high glucose (33.3 mmol/L) or normal glucose (5.5 mmol/L) for 48 h. Affymetrix miRNA profiling and real-time PCR were used to validate the miRNA expression. An H2S probe (HSip-1) was used to detect endogenous H2S. Scratch wound-healing assays were used to evaluate HUVEC migration. The protein levels were quantified by Western blot. Both exogenous and endogenous H2S could upregulate the miR-126-3p levels in HUVECs or muscle tissue. High glucose decreased the H2S level and the protein expression of the H2S-producing enzyme cystathionine γ-lyase (CSE) in HUVECs; however, the DNA methyltransferase 1 (DNMT1) protein level was upregulated. CSE overexpression not only increased the miR-126-3p level by decreasing the DNMT1 protein level but also rescued the deteriorated cell migration in HUVECs treated with high glucose. DNMT1 overexpression decreased the miR-126-3p level and inhibited the migration of HUVECs, whereas silencing DNMT1 improved cell migration. High glucose decreased the endogenous H2S and miR-126-3p levels and increased the DNMT1 expression, thus inducing the migration dysfunction of HUVECs. Treatment with exogenous H2S or the overexpression of the endogenously produced enzyme CSE would rescue this migration dysfunction through H2S-DNMT1-miR-126-3p.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Sulfeto de Hidrogênio/farmacologia , MicroRNAs/genética , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/crescimento & desenvolvimento , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Movimento Celular/efeitos dos fármacos , Cistationina gama-Liase/genética , DNA (Citosina-5-)-Metiltransferase 1/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Glucose/toxicidade , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Sulfeto de Hidrogênio/metabolismo , Camundongos , Neovascularização Fisiológica/genética , Transdução de Sinais/efeitos dos fármacos , Cicatrização/efeitos dos fármacos
8.
J Pharmacol Exp Ther ; 373(3): 463-475, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32238453

RESUMO

Leonurine (LEO) is a bioactive small molecular compound that has protective effects on the cardiovascular system and prevents the early progression of atherosclerosis; however, it is not clear whether LEO is effective for plaque stability. A novel mouse atherosclerosis model involving tandem stenosis (TS) of the right carotid artery combined with western diet (WD) feeding was used. Apolipoprotein E gene-deficient mice were fed with a WD and received LEO administration daily for 13 weeks. TS was introduced 6 weeks after the onset of experiments. We found that LEO enhanced plaque stability by increasing fibrous cap thickness and collagen content while decreasing the population of CD68-positive cells. Enhanced plaque stability by LEO was associated with the nitric oxide synthase (NOS)-nitric oxide (NO) system. LEO restored the balance between endothelial NOS(E)- and inducible NOS(iNOS)-derived NO production; suppressed the NF-κB signaling pathway; reduced the level of the inflammatory infiltration in plaque, including cytokine interleukin 6; and downregulated the expression of adhesion molecules. These findings support the distinct role of LEO in plaque stabilization. In vitro studies with oxidized low-density lipoprotein-challenged human umbilical vein endothelial cells revealed that LEO balanced NO production and inhibited NF-κB/P65 nuclear translocation, thus mitigating inflammation. In conclusion, the restored balance of the NOS-NO system and mitigated inflammation contribute to the plaque-stabilizing effect of LEO. SIGNIFICANCE STATEMENT: LEO restored the balance between endothelial NOS and inducible NOS in NO production and inhibited excessive inflammation in atherosclerotic "unstable" and rupture-prone plaques in apolipoprotein E gene-deficient mice. The protective effect of LEO for stabilizing atherosclerotic plaques was due to improved collagen content, increased fibrous cap thickness, and decreased accumulation of macrophages/foam cells. So far, LEO has passed the safety and feasibility test of phase I clinical trial.


Assuntos
Aterosclerose/tratamento farmacológico , Ácido Gálico/análogos & derivados , Inflamação/tratamento farmacológico , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Placa Aterosclerótica/tratamento farmacológico , Animais , Aterosclerose/metabolismo , Linhagem Celular , Ácido Gálico/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Placa Aterosclerótica/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
J Transl Med ; 15(1): 65, 2017 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-28340574

RESUMO

BACKGROUND: Lung cancer is the leading cause of cancer-related morbidity and mortality worldwide. Patients with chronic respiratory diseases, such as chronic obstructive pulmonary disease (COPD), are exposed to a higher risk of developing lung cancer. Chronic inflammation may play an important role in the lung carcinogenesis among those patients. The present study aimed at identifying candidate biomarker predicting lung cancer risk among patients with chronic respiratory diseases. METHODS: We applied clinical bioinformatics tools to analyze different gene profile datasets with a special focus on screening the potential biomarker during chronic inflammation-lung cancer transition. Then we adopted an in vitro model based on LPS-challenged A549 cells to validate the biomarker through RNA-sequencing, quantitative real time polymerase chain reaction, and western blot analysis. RESULTS: Bioinformatics analyses of the 16 enrolled GSE datasets from Gene Expression Omnibus online database showed myocyte enhancer factor 2D (MEF2D) level significantly increased in COPD patients coexisting non-small-cell lung carcinoma (NSCLC). Inflammation challenge increased MEF2D expression in NSCLC cell line A549, associated with the severity of inflammation. Extracellular signal-regulated protein kinase inhibition could reverse the up-regulation of MEF2D in inflammation-activated A549. MEF2D played a critical role in NSCLC cell bio-behaviors, including proliferation, differentiation, and movement. CONCLUSIONS: Inflammatory conditions led to increased MEF2D expression, which might further contribute to the development of lung cancer through influencing cancer microenvironment and cell bio-behaviors. MEF2D might be a potential biomarker during chronic inflammation-lung cancer transition, predicting the risk of lung cancer among patients with chronic respiratory diseases.


Assuntos
Inflamação/metabolismo , Neoplasias Pulmonares/metabolismo , Fatores de Transcrição MEF2/metabolismo , Células A549 , Carcinoma Pulmonar de Células não Pequenas/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/genética , Inflamação/patologia , Lipopolissacarídeos/farmacologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Fatores de Transcrição MEF2/deficiência , Fatores de Transcrição MEF2/genética , Doença Pulmonar Obstrutiva Crônica/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/genética
10.
Sheng Li Ke Xue Jin Zhan ; 48(1): 12-21, 2017 Feb.
Artigo em Zh | MEDLINE | ID: mdl-29927215

RESUMO

Hydrogen sulfide (H2S)has emerged as pivotal signaling molecules since it is recognized as the third gasotransmitter together with nitric oxide and carbon monoxide. The development of detecting technologies contributed to the research in H2S biology.H2S plays significant roles in human body systems, such as the cardiovascular system, nervous system, respiratory system etc.. Alterations of H2S concentrations have been connected with many diseases. Hypertension, atherosclerosis, neurodegenerative disorder, asthma and many other diseases are found to be related with abnormal H2S metabolism. It has become a potential drug for therapeutic purposes. Understanding the mechanism of H2S biology, including a molecular switch contained in its "receptor", has deepened the research on how small molecules regulate big molecules, as well as providing new strategy for the therapeutic approaches for varies of diseases.


Assuntos
Sulfeto de Hidrogênio/metabolismo , Transdução de Sinais , Asma , Aterosclerose , Monóxido de Carbono , Sistema Cardiovascular , Humanos , Hipertensão , Sistema Nervoso , Doenças Neurodegenerativas , Óxido Nítrico , Sistema Respiratório
11.
Am J Physiol Cell Physiol ; 310(4): C305-17, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26879375

RESUMO

We previously found hydrogen sulfide (H2S) to be a new proangiogenic factor. However, the mechanisms underlying the cardiovascular effect of this small gas molecule remain largely unknown. The aim of the present study was to identify the essential microRNAs (miRNAs) involved in the transduction of H2S signals in vascular endothelial cells (ECs). The expression of miR-640 and its signaling elements, vascular endothelial growth factor receptor 2 (VEGFR2), hypoxia inducible factor 1-α (HIF1A), and mammalian target of rapamycin (mTOR), was measured using quantitative PCR and Western blotting. Overexpression and inhibition of miR-640 were performed to clarify their roles in mediating the effect of H2S. In addition, knockdown of VEGFR2, HIF1A, and mTOR was performed using siRNAs, dominant negative mutants, or inhibitors to examine their roles in the transduction of the H2S signals. miR-640 levels decreased in vascular ECs that were treated with H2S, whereas overexpression of miR-640 blunted the proangiogenic effect of H2S. Knockdown of either VEGFR2 or mTOR blunted the downregulation of miR-640 and the proangiogenic effect induced by H2S. In addition, miR-640 bound to the 3'-UTR of HIF1A mRNA and then inhibited the expression of HIF1A. The inhibition could be recovered by treating cells with H2S. Thus we concluded that miR-640 plays a pivotal role in mediating the proangiogenic effect of H2S; H2S acts through downregulation of the expression of miR-640 and increasing the levels of HIF1A through the VEGFR2-mTOR pathway.


Assuntos
Indutores da Angiogênese/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Sulfeto de Hidrogênio/farmacologia , MicroRNAs/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Regiões 3' não Traduzidas , Sítios de Ligação , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo , 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 , MicroRNAs/genética , Mutação , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética , Transfecção , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
12.
Nitric Oxide ; 46: 114-22, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-25524832

RESUMO

INTRODUCTION: Hydrogen sulfide (H2S) has been reported to inhibit myocardial hypertrophy in a cell model of cardiomyocyte hypertrophy. Our previous study also shows an H2S-induced increase in glucose metabolism in insulin-targeting cells. The present study aims to examine the hypothesis that H2S attenuates myocardial hypertrophy and promotes glucose utilization in cardiomyocytes. METHODS: The cell model of cardiomyocyte hypertrophy was induced by application of phenylephrine and cardiomyocyte hypertrophy was examined using leucine incorporation assay. Protein levels were measured using Western blot analysis. The activity of related enzymes was measured with enzyme-linked immunosorbent assay (ELISA). RESULTS: NaHS (an H2S donor) treatment increased the activity of cultured cardiomyocytes and reduced hypertrophy in cultured cardiomyocytes at concentrations ranging from 25 to 200 µmol/L. NaHS treatment increased glucose uptake and the efficiency of glycolysis and the citric acid cycle. The key enzymes in these reactions, including lactate dehydrogenase and pyruvate kinase and succinate dehydrogenase, were activated by NaHS treatment (100 µmol/L). Some intermediates of glycolysis and the citric acid cycle, including lactic acid, cyclohexylammonium, oxaloacetic acid, succinate, L-dimalate, sodium citrate, cis-aconitic acid, ketoglutarate and DL-isocitric acid trisodium also showed anti-hypertrophic effects in cardiomyocytes. CONCLUSIONS: H2S improves glucose utilization and inhibits cardiomyocyte hypertrophy.


Assuntos
Glucose/metabolismo , Sulfeto de Hidrogênio/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Animais , Crescimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ciclo do Ácido Cítrico/efeitos dos fármacos , Ventrículos do Coração/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fenilefrina , Ratos
13.
Handb Exp Pharmacol ; 230: 137-52, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26162832

RESUMO

Angiogenesis is a physiological process in organ development and also a compensatory response in ischemia. When ischemia occurs, oxygen sensors in vascular endothelial cells sense the decrease in oxygen, thus activating downstream signaling pathways to promote the proliferation, migration, and tube formation of the endothelial cells. The new vasculatures are formed by sprouting from preexisting vessels, in order to maintain oxygen homeostasis in ischemic tissues (Folkman and Shing 1992). Collateral circulation is sometimes established under chronic ischemic conditions such as chronic myocardial ischemia (Banai et al. 1994). However, naturally occurring angiogenesis is usually not sufficient to compensate for ischemia in ischemic tissues. Proangiogenic drugs may be useful to promote angiogenesis in these diseases.


Assuntos
Células Endoteliais/fisiologia , Sulfeto de Hidrogênio/metabolismo , Neovascularização Fisiológica , Animais , Humanos , Isquemia/tratamento farmacológico , Isquemia/etiologia , Receptores Proteína Tirosina Quinases/fisiologia , Transdução de Sinais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia
14.
Antioxid Redox Signal ; 40(1-3): 86-109, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37548532

RESUMO

Significance: As a new important gas signaling molecule like nitric oxide (NO) and carbon dioxide (CO), hydrogen sulfide (H2S), which can be produced by endogenous H2S-producing enzymes through l-cysteine metabolism in mammalian cells, has attracted wide attention for long. H2S has been proved to play an important regulatory role in numerous physiological and pathophysiological processes. However, the deep mechanisms of those different functions of H2S still remain uncertain. A better understanding of the mechanisms can help us develop novel therapeutic strategies. Recent Advances: H2S can play a regulating role through various mechanisms, such as regulating epigenetic modification, protein expression levels, protein activity, protein localization, redox microenvironment, and interaction with other gas signaling molecules such as NO and CO. In addition to discussing the molecular mechanisms of H2S from the above perspectives, this article will review the regulation of H2S on common signaling pathways in the cells, including the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), mitogen-activated protein kinase (MAPK), Janus kinase (JAK)/signal transducer, and activator of transcription (STAT) signaling pathway. Critical Issues: Although there are many studies on the mechanism of H2S, little is known about its direct target molecules. This article will also review the existing reports about them. Furthermore, the interaction between direct target molecules of H2S and the downstream signaling pathways involved also needs to be clarified. Future Directions: An in-depth discussion of the mechanism of H2S and the direct target molecules will help us achieving a deeper understanding of the physiological and pathophysiological processes regulated by H2S, and lay a foundation for developing new clinical therapeutic drugs in the future. Innovation: This review focuses on the regulation of H2S on signaling pathways and the direct target molecules of H2S. We also provide details on the underlying mechanisms of H2S functions from the following aspects: epigenetic modification, regulation of protein expression levels, protein activity, protein localization, redox microenvironment, and interaction with other gas signaling molecules such as NO and CO. Further study of the mechanisms underlying H2S will help us better understand the physiological and pathophysiological processes it regulates, and help develop new clinical therapeutic drugs in the future. Antioxid. Redox Signal. 40, 86-109.


Assuntos
Gasotransmissores , Sulfeto de Hidrogênio , Animais , Sulfeto de Hidrogênio/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/fisiologia , Gasotransmissores/metabolismo , Óxido Nítrico/metabolismo , Mamíferos/metabolismo
15.
Heliyon ; 9(9): e19777, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37809971

RESUMO

In this study, the co-digestion system with Navel orange residues (NOR) and Waste activated sludge (WAS) was established, by pre-treating the NOR and setting different volatile solids (VS) ratios of NOR to WAS to motivate the production of volatile fatty acids (VFA). The pre-treatment method (pH 7 and temperature 70 °C) promoted the release of dissolved organic matter, and the concentration of soluble chemical oxygen demand (SCOD) increased by 45.56% compared with the untreated group (pH 3 and temperature 20 °C). In the co-digestion system, the highest VFA yield (5716.69 mg/L) was obtained at VS ratio of 2. When the VS ratio was increased to 4, the imbalance in proportions of carbon and nitrogen affected VFA production, and the high concentration of essential oils (EO) present in the NOR inhibited the methane production; the cumulative yield of methane gas decreased by 24.10% compared with the yield obtained when the VS ratio was 2. Analysis of microbial community revealed that an increase in the number of VFA-producing microbial populations and the abundance of Methanobacteria resulted in the accumulation of acetic acid. This study demonstrated that co-digestion of NOR with WAS improve VFA production, thus realizing the utilization of solid wastes and reducing environmental pollution.

16.
Cell Rep ; 42(7): 112750, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37421623

RESUMO

The present study examines whether there is a mechanism beyond the current concept of post-translational modifications to regulate the function of a protein. A small gas molecule, hydrogen sulfide (H2S), was found to bind at active-site copper of Cu/Zn-SOD using a series of methods including radiolabeled binding assay, X-ray absorption near-edge structure (XANES), and crystallography. Such an H2S binding enhanced the electrostatic forces to guide the negatively charged substrate superoxide radicals to the catalytic copper ion, changed the geometry and energy of the frontier molecular orbitals of the active site, and subsequently facilitated the transfer of an electron from the superoxide radical to the catalytic copper ion and the breakage of the copper-His61 bridge. The physiological relevance of such an H2S effect was also examined in both in vitro and in vivo models where the cardioprotective effects of H2S were dependent on Cu/Zn-SOD.


Assuntos
Cobre , Sulfeto de Hidrogênio , Cobre/metabolismo , Superóxido Dismutase/metabolismo , Domínio Catalítico , Superóxidos , Zinco/metabolismo
17.
J Cell Physiol ; 227(10): 3518-27, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22252379

RESUMO

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is known to be a "housekeeping" protein; studies in non-cardiomyocytic cells have shown that GAPDH plays pro-apoptotic role by translocating from cytoplasm to the nucleus or to the mitochondria. However, the cardiovascular roles of GAPDH are unknown. We observed that phenylephrine (PE) (100 µM) protected against serum and glucose starvation -induced apoptosis in neonatal rat cardiac myocytes as assessed by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) and mitochondrial membrane potential depolarization. GAPDH glycolysis activity was positively correlated with the antiapoptotic action of PE. GAPDH activity inhibition blunted PE-induced protection of the mitochondrial membrane potential and cardiomyocytes. PE-induced Bcl-2 protein increase, Bax mitochondrial decrease and inhibition of cytochrome C release and Caspase 3 activation, as well as ROS production were blunted by GAPDH activity inhibition. Moreover, GAPDH overexpression provided protection against starvation-induced cardiomyocyte apoptosis in vitro and ischemia-induced cardiac infarction in vivo. Inhibition of Akt prevented PE-induced GAPDH activity increase and cardiomyocytes protection. In conclusion, the present study provides the first direct evidence of an antiapoptotic role of GAPDH in PE-induced cardiomyocytes protection; GAPDH activity elevation mainly affects the mitochondria-induced apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Fenilefrina/farmacologia , Inanição/patologia , Animais , Apoptose/genética , Caspase 3/metabolismo , Células Cultivadas , Citocromos c/metabolismo , DNA Nucleotidilexotransferase/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/biossíntese , Gliceraldeído-3-Fosfato Desidrogenases/genética , Glicólise/efeitos dos fármacos , Glicólise/genética , Marcação In Situ das Extremidades Cortadas/métodos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/genética , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Inanição/enzimologia , Proteína X Associada a bcl-2/metabolismo
18.
Biochem Biophys Res Commun ; 421(2): 164-9, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22503984

RESUMO

Inhibition of oxidative stress has been reported to be involved in the cardioprotective effects of hydrogen sulfide (H(2)S) during ischemia/reperfusion (I/R). However, the mechanism whereby H(2)S regulates the level of cardiac reactive oxygen species (ROS) during I/R remains unclear. Therefore, we investigated the effects of H(2)S on pathways that generate and scavenge ROS. Our results show that pretreating rat neonatal cardiomyocytes with NaHS, a H(2)S donor, reduced the levels of ROS during the hypoxia/reoxygenation (H/R) condition. We found that H(2)S inhibited mitochondrial complex IV activity and increased the activities of superoxide dismutases (SODs), including Mn-SOD and CuZn-SOD. Further studies indicated that H(2)S up-regulated the expression of Mn-SOD but not CuZn-SOD. Using a cell-free system, we showed that H(2)S activates CuZn-SOD. An isothermal titration calorimetry (ITC) analysis indicated that H(2)S directly interacts with CuZn-SOD. Taken together, H(2)S inhibits mitochondrial complex IV and activates SOD to decrease the levels of ROS in cardiomyocytes during I/R.


Assuntos
Citoproteção , Complexo IV da Cadeia de Transporte de Elétrons/antagonistas & inibidores , Sulfeto de Hidrogênio/metabolismo , Mitocôndrias/enzimologia , Miócitos Cardíacos/efeitos dos fármacos , Espécies Reativas de Oxigênio/antagonistas & inibidores , Traumatismo por Reperfusão/enzimologia , Superóxido Dismutase/biossíntese , Animais , Células Cultivadas , Miócitos Cardíacos/enzimologia , Ratos , Ratos Sprague-Dawley , Sulfetos/farmacologia
19.
Sheng Li Xue Bao ; 64(2): 142-8, 2012 Apr 25.
Artigo em Zh | MEDLINE | ID: mdl-22513463

RESUMO

Central urotensin II (UII) may participate in the regulation of cardiovascular functions by stimulating sympathy pathway. However, the central mechanism remained unknown. Recent studies have shown that brain reactive oxygen species (ROS) mediate the sympatho-excitatory effects. In the present study, we tested the hypothesis that ROS mediate central cardiovascular effects of UII. Experiments were conducted in Wistar-Kyoto (WKY) rats and spontaneously hypertensive rats (SHR). Immunocytochemistry, intracerebroventricular (icv) infusion and lucigenin-enhanced chemiluminescence assay were employed to detect UII receptor expression and ROS level, respectively. The following results were obtained: (1) Expressions of UII receptors of rostral ventrolateral medulla (RVLM) and nucleus tractus solitarii (NTS) were increased in SHR rats compared with WKY rats (P < 0.05). (2) UII (icv) significantly increased mean arterial pressure (MAP) (P < 0.05), and the effect of UII was significantly more pronounced in SHR rats than that in WKY rats (P < 0.05); (3) Tempol (a superoxide dismutase mimic) or Urantide (an antagonist of UII receptor) pretreatments eliminated the pressor effect of UII (P < 0.05) in SHR rats; (4) Brain superoxide level was increased in UII-treated SHR rats compared with that in cerebrospinal fluid (CSF)-treated SHR rats (P < 0.05). These results indicate that ROS mediate central cardiovascular effects of UII in SHR rats and provide evidence for a novel relationship between UII and ROS.


Assuntos
Encéfalo/metabolismo , Hipertensão/fisiopatologia , Espécies Reativas de Oxigênio/metabolismo , Urotensinas/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Masculino , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptores Acoplados a Proteínas G/metabolismo , Superóxido Dismutase/metabolismo
20.
Sheng Li Xue Bao ; 64(2): 129-34, 2012 Apr 25.
Artigo em Zh | MEDLINE | ID: mdl-22513461

RESUMO

The purpose of this study was to investigate the molecular mechanisms whereby hydrogen sulfide (H2S) exerts the promoting effect on vascular endothelial cells migration. We used wound healing assay to study the effect of NaHS (H2S donor) on the migration ability of rhesus retinal pigment epithelial cell line, RF/6A cells, under normoxic conditions. Real-time PCR was used to measure hypoxia-inducible factor 1α (HIF-1α) mRNA level. Western blot was used to measure the expression of HIF-1α protein. The probe 2',7'-dichlorofluorescein diacetate (DCFH-DA) was used to measure intracellular reactive oxygen species (ROS) level. The results showed that NaHS (10-100 µmol/L) could significantly promote RF/6A cells migration under normoxic conditions, and this effect could be inhibited by 50 µmol/L HIF-1 inhibitor, CdCl2. NaHS increased the protein level of HIF-1α in a dose- and time-dependent manner, and up-regulated the mRNA level of HIF-1α quickly and continuously. Moreover, NaHS could significantly decrease ROS levels in RF/6A cells under normoxic conditions. These results suggest HIF-1 may mediate the promoting effect of H2S on vascular endothelial cells migration under normoxic conditions. ROS, as an upstream regulator of HIF-1α, may be involved in the migration-promoting effect of H2S.


Assuntos
Movimento Celular/fisiologia , Células Endoteliais/citologia , Sulfeto de Hidrogênio/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Animais , Linhagem Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Macaca mulatta , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Epitélio Pigmentado da Retina/citologia , Sulfetos/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa