Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Acta Biochim Biophys Sin (Shanghai) ; 56(3): 440-451, 2024 03 25.
Article in English | MEDLINE | ID: mdl-38006215

ABSTRACT

Hypertrophic scar (HS) is one of the most common sequelae of patients, especially after burns and trauma. The roles of regulatory long noncoding RNAs (lncRNAs) in mediating HS remain underexplored. Human hypertrophic scar-derived fibroblasts (HSFBs) have been shown to exert more potent promoting effects on extracellular matrix (ECM) accumulation than normal skin-derived fibroblasts (NSFBs) and are associated with enhanced HS formation. The purpose of this study is to search for lncRNAs enriched in HSFBs and investigate their roles and mechanisms. LncRNA MSTRG.59347.16 is one of the most highly expressed lncRNAs in HS detected by lncRNA-seq and qRT-PCR and named as hypertrophic scar fibroblast-associated lncRNA (HSFAS). HSFAS overexpression significantly induces fibroblast proliferation, migration, and myofibroblast trans-differentiation and inhibits apoptosis in HSFBs, while knockdown of HSFAS results in augmented apoptosis and attenuated proliferation, migration, and myofibroblast trans-differentiation of HSFBs. Mechanistically, HSFAS suppresses the expression of A disintegrin and metalloproteinase with thrombospondin motifs 8 (ADAMTS8). ADAMTS8 knockdown rescues downregulated HSFAS-mediated fibroblast proliferation, migration, myofibroblast trans-differentiation and apoptosis. Thus, our findings uncover a previously unknown lncRNA-dependent regulatory pathway for fibroblast function. Targeted intervention in the HSFAS-ADAMTS8 pathway is a potential therapy for HS.


Subject(s)
Cicatrix, Hypertrophic , RNA, Long Noncoding , Humans , Cicatrix, Hypertrophic/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Fibroblasts/metabolism , Apoptosis/genetics , Cell Proliferation/genetics , Cell Transdifferentiation/genetics , ADAMTS Proteins/metabolism
2.
Acta Biochim Biophys Sin (Shanghai) ; 55(8): 1184-1192, 2023 Apr 06.
Article in English | MEDLINE | ID: mdl-37021975

ABSTRACT

Autophagy plays a critical role in the physiology and pathophysiology of hepatocytes. High level of homocysteine (Hcy) promotes autophagy in hepatocytes, but the underlying mechanism is still unknown. Here, we investigate the relationship between Hcy-induced autophagy level and the expression of nuclear transcription factor EB (TFEB). The results show that Hcy-induced autophagy level is mediated by upregulation of TFEB. Silencing of TFEB decreases the level of autophagy-related protein LC3BII/I and increases p62 expression level in hepatocytes after exposure to Hcy. Moreover, the effect of Hcy on the expression of TFEB is regulated by hypomethylation of the TFEB promoter catalyzed by DNA methyltransferase 3b (DNMT3b). In summary, this study shows that Hcy can activate autophagy by inhibiting DNMT3b-mediated DNA methylation and upregulating TFEB expression. These findings provide another new mechanism for Hcy-induced autophagy in hepatocytes.


Subject(s)
Autophagy , DNA Methylation , Hepatocytes , Homocysteine , Autophagy/genetics , DNA , Homocysteine/metabolism , Homocysteine/pharmacology , Humans , DNA Methyltransferase 3B
3.
Acta Biochim Biophys Sin (Shanghai) ; 54(12): 1-9, 2022 Dec 25.
Article in English | MEDLINE | ID: mdl-36514215

ABSTRACT

Long noncoding RNAs (lncRNAs) are increasingly being implicated as key regulators of cell proliferation, apoptosis, and differentiation. However, the molecular mechanisms of specific lncRNAs in the context of hypertrophic scar remain largely unclear. Here, we find that the lncRNA FPASL (fibroblast proliferation-associated LncRNA) is downregulated in HS, and FPASL reduces fibroblast proliferation and colony formation and blocks cell cycle progression. Using GO annotation enrichment analysis along with AZC (a specific inhibitor of DNA methylation), we identify that DNA methylation is responsible for downregulating FPASL in hypertrophic scar. Subsequent studies demonstrate that high expression of DNMT3b inhibits FPASL expression in HS. Mechanistic study reveals a significant increase in fibroblast proliferation after transfection with LNA-FPASL, which is further inhibited by knockdown of DNMT3b. Thus, our study reveals that DNMT3b mediates hypermethylation of the lncRNA FPASL promoter and the downregulation of lncRNA FPASL promotes fibroblast proliferation in hypertrophic scar.


Subject(s)
Cicatrix, Hypertrophic , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cicatrix, Hypertrophic/metabolism , DNA Methylation , Cell Proliferation/genetics , Fibroblasts/metabolism
4.
Mol Ther Nucleic Acids ; 26: 1318-1335, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34853730

ABSTRACT

It has been demonstrated that homocysteine (Hcy) can cause inflammatory diseases. Long noncoding RNAs (lncRNA) and microRNAs (miRNAs) are involved in this biological process, but the mechanism underlying Hcy-induced inflammation remains poorly understood. Here, we found that lncRNA TGFB3-AS1 was highly expressed in macrophages treated with Hcy and the peripheral blood monocytes from cystathionine beta-synthase heterozygous knockout (CBS +/-) mice with a high-methionine diet using lncRNA microarray. In vivo and in vitro experiments further confirmed that TGFB3-AS1 accelerated Hcy-induced inflammation of macrophages through the Rap1a/wnt signaling pathway. Meanwhile, TGFB3-AS1 interacted with Rap1a and reduced degradation of Rap1a through inhibiting its ubiquitination in macrophages treated with Hcy. Rap1a mediated inflammation induced by Hcy and serves as a direct target of miR-144. Moreover, TGFB3-AS1 regulated miR-144 by binding to pri-miR-144 and inhibiting its maturation, which further regulated Rap1a expression. More importantly, we found that high expression of TGFB3-AS1 was positively correlated with the levels of Hcy and proinflammatory cytokines in serum of healthy individuals and patients with HHcy. Our study revealed a novel mechanism by which TGFB3-AS1 promoted inflammation of macrophages through inhibiting miR-144 maturation to stay miR-144 regulated inhibition of functional Rap1a expression.

5.
Aging Cell ; 20(10): e13485, 2021 10.
Article in English | MEDLINE | ID: mdl-34592792

ABSTRACT

Atherosclerosis is a serious age-related disease, which has a tremendous impact on health care globally. Macrophage inflammation is crucial for the initiation and progression of atherosclerosis, and microRNAs (miRNAs) recently have emerged as potent modulators of inflammation, while the underlying mechanisms of its involvement in homocysteine (Hcy)-mediated macrophage inflammation of atherosclerosis remain largely unknown. Here, we demonstrated that elevated Hcy inhibits the expression of miR-195-3p, which in turn enhances IL-31 expression and thereby causes the secretion of macrophages pro-inflammatory factors IL-1ß, IL-6 and TNF-α and accelerate atherosclerosis. Furthermore, we identified that Hcy can induce DNA hypermethylation and H3K9 deacetylation of miR-195-3p promoter due to the increased the binding of DNMT3a and HDAC11 at its promoter. More importantly, Sp1 interacts with DNMT3a suppressed the binding of HDAC11 at miR-195-3p promoter and promoted its transcription. In summary, our results revealed a novel mechanism that transcriptional and epigenetic regulation of miR-195-3p inhibits macrophage inflammation through targeting IL-31, which provides a candidate diagnostic marker and novel therapeutic target in cardiovascular diseases induced by Hcy.


Subject(s)
Atherosclerosis/chemically induced , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Homocysteine/adverse effects , Interleukins/metabolism , Animals , Apoptosis , Humans , Mice
6.
Circ J ; 84(4): 616-625, 2020 03 25.
Article in English | MEDLINE | ID: mdl-32115441

ABSTRACT

BACKGROUND: Ischemic postconditioning (IPostC) is an endogenous protective mechanism to reduce ischemia-reperfusion (I/R) injury. However, whether IPostC protects aged cardiomyocytes against I/R injury is not fully understood. Considering the protective function of microRNA 30a (miR-30a) against ischemia-induced injury in H9C2 cells, its role in the protective effects of IPostC on I/R injury of aged cardiomyocytes was investigated further.Methods and Results:To mimic I/R and IPostC in vitro, the aged cardiomyocyte model for hypoxia postconditioning (HPostC) treatment was established by 9 days of incubation with 8 mg/mL D-galactose and then followed by exposure to hypoxic environment. HPostC significantly alleviated hypoxia/reoxygenation (H/R) injury and reduced autophagy of aged cardiomyocytes, as evidenced by decreased LC3B-II expression and increased p62 by Western blot. Quantified by quantitative real-time polymerase chain reaction (qRT-PCR), miR-30a was increased in aged cardiomyocytes treated with HPostC compared with I/R injury group. Overexpression of miR-30a by LV3-rno-miR-30a mimic promoted cardioprotective effect of HPostC in aged cardiomyocytes by suppressing BECN1-mediated autophagy, all of which was abrogated by knockdown of miR-30a expression. Epigenetic analyses demonstrated that HPostC reduced DNA methyltransferase 3b-mediated DNA hypomethylation levels at miR-30a promoter, leading to upregulation of miR-30a. CONCLUSIONS: HPostC protected aged cardiomyocytes survival against H/R injury via DNMT3b-dependent activation of miR-30a. miR-30a could be a potential therapeutic target for ischemic myocardial infarction.


Subject(s)
Autophagy , Cellular Senescence , DNA Methylation , Epigenesis, Genetic , MicroRNAs/metabolism , Myocardial Reperfusion Injury/prevention & control , Animals , Beclin-1/genetics , Beclin-1/metabolism , Cell Hypoxia , Cell Line , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , MicroRNAs/genetics , Microtubule-Associated Proteins/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Promoter Regions, Genetic , Rats , Sequestosome-1 Protein/metabolism , Signal Transduction , DNA Methyltransferase 3B
7.
Chem Biol Interact ; 311: 108749, 2019 Sep 25.
Article in English | MEDLINE | ID: mdl-31325423

ABSTRACT

PURPOSE: Excessive proliferation, migration and anti-apoptosis of pulmonary artery smooth muscle cells (PASMCs) are the basis for the development of pulmonary vascular remodeling, and it is the driving force for pulmonary arterial hypertension (PAH). 18ß-glycyrrhetinic acid (18ß-GA) is the main active substance extracted from Chinese herbal medicine licorice, with outstanding anti-inflammatory, anti-oxidation and anti-proliferative effects. Our team found in previous studies that 18ß-GA has protective effects on monocrotaline-induced PAH in rats. However, the anti-angiogenic effect of 18ß-GA on PAH remains unclear. Therefore, in order to further investigate whether the beneficial effects of 18ß-GA on PAH are related to its antiproliferative effect, we conducted experiments in vivo and in vitro. METHODS AND RESULTS: In vivo, 18ß-GA relieved mean pulmonary arterial pressure, right ventricular systolic pressure, and right ventricular hypertrophy index, improving pulmonary remodeling. In vitro, 18ß-GA significantly inhibited PDGF-BB-induced proliferation and DNA synthesis of HPASMCs, blocking the progression of G0/G1 to S phase of the cell cycle. Furthermore, after treatment with 18ß-GA, the expression of Rho A, ROCK1, ROCK2 was decreased and ROCK activity was inhibited in HPASMC. In addition, 18ß-GA also attenuated PDGF-induced changes in p27kip1, Bax and Bcl-2. CONCLUSIONS: In summary, these results indicate that 18ß-GA regulates the activity of RhoA-ROCK signaling pathway, inhibits the proliferation of HPASMCs, and has potential value in the treatment of PAH.


Subject(s)
Glycyrrhetinic Acid/analogs & derivatives , Hypertension, Pulmonary/pathology , Protective Agents/pharmacology , Signal Transduction/drug effects , Animals , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation/drug effects , Glycyrrhetinic Acid/pharmacology , Glycyrrhetinic Acid/therapeutic use , Hemodynamics/drug effects , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Male , Monocrotaline/toxicity , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Protective Agents/therapeutic use , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Vascular Remodeling/drug effects , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
8.
J Cell Mol Med ; 23(7): 4611-4626, 2019 07.
Article in English | MEDLINE | ID: mdl-31104361

ABSTRACT

It is well-established that homocysteine (Hcy) is an independent risk factor for atherosclerosis. Hcy can promote vascular smooth muscle cell (VSMC) proliferation, it plays a key role in neointimal formation and thus contribute to arteriosclerosis. However, the molecular mechanism on VSMCs proliferation underlying atherosclerosis is not well elucidated. Mitofusin-2 (MFN2) is an important transmembrane GTPase in the mitochondrial outer membrane and it can block cells in the G0/G1 stage of the cell cycle. To investigate the contribution of aberrant MFN2 transcription in Hcy-induced VSMCs proliferation and the underlying mechanisms. Cell cycle analysis revealed a decreased proportion of VSMCs in G0/G1 and an increased proportion in S phase in atherosclerotic plaque of APOE-/- mice with hyperhomocystinaemia (HHcy) as well as in VSMCs exposed to Hcy in vitro. The DNA methylation level of MFN2 promoter was obviously increased in VSMCs treated with Hcy, leading to suppressed promoter activity and low expression of MFN2. In addition, we found that the expression of c-Myc was increased in atherosclerotic plaque and VSMCs treated with Hcy. Further study showed that c-Myc indirectly regulates MFN2 expression is duo to the binding of c-Myc to DNMT1 promoter up-regulates DNMT1 expression leading to DNA hypermethylation of MFN2 promoter, thereby inhibits MFN2 expression in VSMCs treated with Hcy. In conclusion, our study demonstrated that Hcy-induced hypermethylation of MFN2 promoter inhibits the transcription of MFN2, leading to VSMCs proliferation in plaque formation, and the increased binding of c-Myc to DNMT1 promoter is a new and relevant molecular mechanism.


Subject(s)
Atherosclerosis/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , GTP Phosphohydrolases/genetics , Homocysteine/pharmacology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Proto-Oncogene Proteins c-myc/metabolism , Transcription, Genetic , Animals , Atherosclerosis/pathology , Base Sequence , Cell Proliferation/drug effects , GTP Phosphohydrolases/metabolism , Gene Expression Regulation/drug effects , Humans , Male , Mice, Inbred C57BL , Models, Biological , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic/pathology , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Transcription, Genetic/drug effects
9.
Front Pharmacol ; 10: 13, 2019.
Article in English | MEDLINE | ID: mdl-30723409

ABSTRACT

Pulmonary arterial hypertension (PAH) is a destructive and rare disorder characterized by a progressive increase in pulmonary artery pressure and vasoconstriction, ultimately leading to right ventricular failure and death. 18ß-Glycyrrhetinic acid (18ß-GA) is an active ingredient in the commonly used Chinese herbal medicine radix glycyrrhizae, and it possesses antioxidant, anti-inflammatory, anti-tumor, and other pharmacological properties. This study aimed to determine whether 18ß-GA has protective effects against monocrotaline (MCT)-induced PAH and whether it is associated with oxidative stress. The PAH of rats was induced by MCT (60 mg/kg) and oral administration of 18ß-GA (100, 50, or 25 mg/kg/day), sildenafil (30 mg/kg), or saline for 21 consecutive days. The development of PAH was evaluated by hemodynamic parameters and right ventricular hypertrophy index. Hematoxylin and eosin staining, Masson trichrome staining, and electron microscopy were used to determine the degree of vascular remodeling and proliferation in lung tissue. Moreover, the antioxidant capacity and malondialdehyde levels in the lungs were measured according to the instructions provided by the test kits, and the expression levels of nicotinamide adenine dinucleotide phosphate oxidase-2 (Nox2) and Nox4 were detected through Western blot analysis. Results of our study indicated that 18ß-GA treatment significantly improved the hemodynamic and pathomorphological data of the rats, reduced the changes in oxidative stress biomarkers, and inhibited Nox2 and Nox4 expression. Our research indicated that 18ß-GA has a protective effect against MCT-induced PAH by inhibiting oxidative stress in rats.

10.
Neurochem Res ; 43(8): 1575-1586, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29948728

ABSTRACT

The aim of the study was to elucidate the therapeutic effects of Cytisine (CYT) on cerebral ischemia-reperfusion injury in mice. Male ICR mice were pretreated with reagents (drug), and then subjected to 2 h focal cerebral ischemia and 24 h reperfusion. Morphologically, the histopathological impairment were estimated by the TTC, HE and TUNEL staining. The expression of GluN2B-containing NMDA receptor, phosphorylation of extracellular regulated protein kinases, total ERK, phosphorylation of cAMP-response element binding protein and total CREB were determined by immunofluorescence and Western blot assay, respectively. The mRNA expression of NR2B, ERK and CREB were quantified by the real-time RT-PCR. CYT significantly diminished the infarct size and neuronal apoptosis. Additionally, it ameliorated histopathological lesion dramatically. CYT promoted the phosphorylation of ERK, CREB and their mRNA expression. In contrast, the expression of NR2B was suppressed in concomitant with the down-regulation of genes. The overall results thus far suggest that CYT confers the neuroprotection against cerebral I/R injury by regulating the NR2B-ERK/CREB signal pathway.


Subject(s)
Alkaloids/therapeutic use , Brain Ischemia/prevention & control , Cyclic AMP Response Element-Binding Protein/physiology , MAP Kinase Signaling System/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Reperfusion Injury/prevention & control , Alkaloids/chemistry , Alkaloids/pharmacology , Animals , Azocines/chemistry , Azocines/pharmacology , Azocines/therapeutic use , Brain Ischemia/pathology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred ICR , Neuroprotection/drug effects , Neuroprotection/physiology , Quinolizines/chemistry , Quinolizines/pharmacology , Quinolizines/therapeutic use , Reperfusion Injury/pathology , Signal Transduction/drug effects , Signal Transduction/physiology
11.
Exp Cell Res ; 362(1): 217-226, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29155363

ABSTRACT

Endothelial progenitor cells (EPCs) contribute to neovasculogenesis and reendothelialization of damaged blood vessels to maintain the endothelium. Dysfunction of EPCs is implicated in the pathogenesis of vascular injury induced by homocysteine (Hcy). We aimed to investigate the role of Cyclin A in Hcy-induced EPCs dysfunction and explore its molecular mechanism. In this study, by treatment of EPCs with Hcy, we found that the expression of Cyclin A mRNA and protein were significantly downregulated in a dose-dependent manner. Knockdown of Cyclin A prominently reduced proliferation of EPCs, while over-expression of Cyclin A significantly promoted the cell proliferation, suggesting that Hcy inhibits EPCs proliferation through downregulation of Cyclin A expression. In addition, epigenetic study also demonstrated that Hcy induces DNA hypomethylation of the Cyclin A promoter in EPCs through downregulated expression of DNMT1. Moreover, we found that Hcy treatment of EPCs leads to increased SAM, SAH and MeCP2, while the ratio of SAM/SAH and MBD expression decrease. In summary, our results indicate that Hcy inhibits Cyclin A expression through hypomethylation of Cyclin A and thereby suppress EPCs proliferation. These findings demonstrate a novel mechanism of DNA methylation mediated by DNMT1 in prevention of Hcy associated cardiovascular disease.


Subject(s)
Cell Proliferation/physiology , Cyclin A/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/physiology , Endothelial Progenitor Cells/metabolism , Homocysteine/metabolism , Animals , Cells, Cultured , Down-Regulation/physiology , Epigenesis, Genetic/physiology , Humans , Promoter Regions, Genetic/physiology , Rats
12.
Mol Med Rep ; 16(6): 9181-9188, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29039510

ABSTRACT

Atherosclerosis (AS) is a progressive disease of multifactorial origin, which occurs in response to endothelial injury. Increased homocysteine (Hcy) is considered a major cause of endothelial dysfunction, oxidative stress and DNA methylation; however, the mechanisms remain to be fully elucidated. The aim of the present study was to investigate whether Hcy causes injury to endothelial cells (ECs) by the effect of lectin­like oxidized­low density lipoprotein receptor­1 (LOX­1) DNA methylation through toll­like receptor 4(TLR4)/nuclear factor (NF)­κB/DNA methyltransferase (DNMT)1. The ECs were treated with different concentrations of Hcy, and it was found that Hcy promoted the expression of TLR4, leading to EC injury. The effect of oxidative stress was analyzed by measuring superoxide dismutase, malondialdehyde and hydrogen peroxide in the ECs. In addition, the association between NF­κB and DNMT1 was examined by treatment of the ECs with pyrrolidine dithiocarbamate (PDTC). The results suggested that Hcy induced LOX­1 DNA hypomethyaltion to promote the expression levels of LOX­1. Taken together, Hcy injured the ECs through the effect of methylation and trans­sulfuration metabolism of LOX­1 through TLR4/NF­κB/DNMT1. Following injury to the ECs, lipids, particularly ox­LDL, accumulated in the sub­endothelial layer to promote the formation of AS.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , Endothelial Cells/metabolism , Homocysteine/metabolism , NF-kappa B/metabolism , Oxidative Stress , Scavenger Receptors, Class E/genetics , Toll-Like Receptor 4/metabolism , Biomarkers , Cells, Cultured , Endothelial Cells/drug effects , Gene Expression Regulation , Homocysteine/pharmacology , Humans , Hydrogen Peroxide , Lipoproteins, LDL , Superoxide Dismutase/metabolism , Toll-Like Receptor 4/genetics
13.
Biomed Pharmacother ; 95: 1161-1168, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28926926

ABSTRACT

Pulmonary hypertension (PH) is fatal disease which closely involves Rho A/ Rho kinsase (ROCK) pathway. Aloperine is a main active alkaloid extracted from Sophora alopecuroides, which is a traditional Chinese herbal medicine that has been used widely. However, the effects of this alkaloid on pulmonary hypertension and its mechanisms remain unclear. Therefore, this study is designed to investigate whether aloperine has protective effects on PH induced by monocrotaline, whether these effects may be related to regulation of RhoA/ROCK pathway in rats. Pulmonary hypertension was induced by monocrotaline (60mg/kg), and subsequently oral administration of aloperine (25, 50, 100mg/kg/day) for 21 days. At the end of the experiment, rats were underwent hemodynamic and morphologic assessments. At same time, the expression of Rho A, ROCK1, ROCK2, as well as activities of ROCK in the lung of rat has been detected. Afterwards, the expression of p27kip1, Bax, Bcl-2, which was the downstream proliferation and apoptosis factors of ROCK, were tested. The result indicted that aloperine treatment showed significantly improvement in hemodynamic and pathomorphologic data. Moreover, the reduction in expression of Rho A, ROCK1, ROCK2, and suppression in activities of ROCK were found in rat lungs after aloperine treatment. Furthermore, aloperine also alleviated the MCT-induced changes of p27kip1, Bax and Bcl-2. In summary, this study indicates that aloperine have protective effects on monocrotaline-induced PH. And these effects may be partially related to RhoA/ROCK pathway. Thus, aloperine could be considered a possible therapeutic strategy for PH.


Subject(s)
Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Piperidines/therapeutic use , Protective Agents/therapeutic use , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cardiomegaly/complications , Cardiomegaly/drug therapy , Cardiomegaly/physiopathology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Electrocardiography , Hemodynamics/drug effects , Hypertension, Pulmonary/diagnostic imaging , Hypertension, Pulmonary/physiopathology , Lung/drug effects , Lung/pathology , Lung/physiopathology , Male , Monocrotaline , Piperidines/pharmacology , Proliferating Cell Nuclear Antigen/metabolism , Protective Agents/pharmacology , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Quinolizidines , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Vascular Remodeling/drug effects , bcl-2-Associated X Protein/metabolism , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein/genetics
14.
Cell Tissue Res ; 369(3): 455-465, 2017 09.
Article in English | MEDLINE | ID: mdl-28656471

ABSTRACT

Lycium barbarum polysaccharides (LBP) have been reported to have a wide range of beneficial effects including neuroprotection, anti-aging and anticancer. However, the anti-inflammation mechanism of LBP on primary cultured rat hippocampal neurons injured by oxygen-glucose deprivation/reperfusion (OGD/RP) is incompletely understood. We investigate the neuroprotective effects of LBP on neonatal rat primary cultured hippocampal neurons injured by OGD/RP with different approaches: MTT assay was used to detect cell viability, lactate dehydrogenase leakage was used to detect neuronal damage, formation of reactive oxygen species was determined by using fluorescent probe DCFH-DA. Hoechst 33,342 staining and TUNEL staining were used to determine the cell apoptosis. JC-1 was used to evaluate loss of mitochondrial membrane potential (MMP). The fluorescence intensity of [Ca2+]i in hippocampal neurons was determined by laser scanning confocal microscopy. The expression of various apoptotic markers such as TLR4, IκB, IL-6 and NF-κB were investigated by RT-PCR and western blot analysis. Results from each approach demonstrated that LBP increased the cell abilities and decreased the cell morphologic impairment. Furthermore, LBP increased MMP but inhibited [Ca2+]i elevation and significantly suppressed overexpression of NF-κB, IL-6 TLR4 and increased IκB expression.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Hippocampus/cytology , Neurons/metabolism , Animals , Apoptosis/drug effects , Calcium/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cells, Cultured , Female , Glucose/deficiency , Interleukin-6/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , NF-kappa B/metabolism , Neurons/drug effects , Oxygen , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Reperfusion , Toll-Like Receptor 4/metabolism
15.
Biomed Pharmacother ; 89: 632-641, 2017 May.
Article in English | MEDLINE | ID: mdl-28262616

ABSTRACT

Pulmonary hypertension (PH) is serious, fatal disease which is promoted by oxidative stress. Aloperine have antioxidation effects, which effects on pulmonary arteries remain unclear. Therefore, this study is designed to investigate whether aloperine has protective effects on PH induced by monocrotaline and whether these effects are associated with oxidative stress. PH was induced by monocrotaline (60mg/kg), and subsequently oral administration of aloperine (25, 50, 100mg/kg/day). At the end of the experiment, hemodynamic, pathomorphologic, electrocardiographic and echocardiographic data from the rats were obtained. At same time, oxidative stress biomarkers (superoxide dismutase, malonyldialdehyde, catalase, glutathione peroxidase, total antioxidant capacity) and the protein expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX)-2, NOX-4 in the lung of rat has been detected. The result shows that aloperine treatment showed significantly improvement in hemodynamic, pathomorphologic, electrocardiographic and echocardiographic data. Moreover, aloperine treatment can alleviate the changes of oxidative stress biomarkers and suppress the expression levels of NOX-2, NOX-4. In summary, this study indicates that aloperine have protective effects on monocrotaline-induced PH. And these effects may be related to inhibit oxidative stress.


Subject(s)
Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/prevention & control , Monocrotaline , Piperidines/pharmacology , Protective Agents/pharmacology , Animals , Antioxidants/pharmacology , Biomarkers/analysis , Dose-Response Relationship, Drug , Echocardiography , Electrocardiography , Hypertension, Pulmonary/diagnostic imaging , Hypertrophy, Right Ventricular/diagnostic imaging , Hypertrophy, Right Ventricular/prevention & control , Male , Oxidative Stress/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Pulmonary Circulation/drug effects , Quinolizidines , Rats , Rats, Sprague-Dawley
16.
Brain Res Bull ; 124: 295-305, 2016 06.
Article in English | MEDLINE | ID: mdl-27345710

ABSTRACT

Taurine is an abundant amino acid in the nervous system, which has been proved to possess antioxidation, osmoregulation and membrane stabilization. Previously it has been demonstrated that taurine exerts ischemic brain injury protective effect. This study was designed to investigate whether the protective effect of taurine has the possibility to be applied to treat neonatal hypoxic-ischemic brain damage. Seven-day-old Sprague-Dawley rats were treated with left carotid artery ligation followed by exposure to 8% oxygen to generate the experimental group. The cerebral damage area was measured after taurine post-treatment with 2,3,5-triphenyltetrazolium chloride (TTC) staining, Hematoxyline-Eosin (HE) staining and Nissl staining. The activities of superoxide dismutase (SOD), malondialdehyde (MDA), glutathione peroxidase (GSH-Px), total antioxidant capacity (T-AOC), myeloperoxtidase (MPO), ATP and Lactic Acid productions were assayed with ipsilateral hemisphere homogenates. Western-blot and immunofluorescence assay were processed to detect the expressions of AIF, Cyt C, Bax, Bcl-2 in brain. We found that taurine significantly reduced brain infarct volume and ameliorated morphological injury obviously reversed the changes of SOD, MDA, GSH-Px, T-AOC, ATP, MPO, and Lactic Acid levels. Compared with hypoxic-ischemic group, it showed marked reduction of AIF, Cyt C and Bax expressions and increase of Bcl-2 after post-treatment. We conclude that taurine possesses an efficacious neuroprotective effect after cerebral hypoxic-ischemic damage in neonatal rats.


Subject(s)
Brain Injuries/etiology , Brain Injuries/prevention & control , Hypoxia-Ischemia, Brain/complications , Neuroprotective Agents/therapeutic use , Taurine/therapeutic use , Animals , Animals, Newborn , Apoptosis Inducing Factor/metabolism , Brain Infarction/etiology , Brain Infarction/prevention & control , Female , Glutathione/metabolism , Humans , Lactic Acid/metabolism , Male , Malondialdehyde/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Taurine/pharmacology
17.
Int J Mol Med ; 36(3): 633-44, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26135032

ABSTRACT

Matrine, an active constituent of the Chinese herb, Sophora flavescens Ait., and it is known for its antioxidant, anti-inflammatory and antitumor activities. It has been demonstrated that matrine exerts protective effects against heart failure by decreasing the expression of caspase-3 and Bax, and increasing Bcl­2 levels. In this study, we aimed to determine whether these protective effects of matrine can be applied to cerebral ischemia. Following 7 successive days of treatment with matrine (7.5, 15 and 30 mg/kg) and nimodipine (1 mg/kg) by intraperitoneal injection, male Institute of Cancer Research (ICR) mice were subjected to middle cerebral artery occlusion (MCAO). Following reperfusion, the neurobehavioral score and brain infarct volume were estimated, and morphological changes were analyzed by hematoxylin and eosin (H&E) staining and electron microscopy. The percentage of apoptotic neurons was determined by flow cytometry. The levels of oxidative stress were assessed by measuring the levels of malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and catalase (CAT), and the total antioxidant capacity (T-AOC). Western blot analysis and immunofluorescence staining were used to examine the expression of the apoptosis-related proteins, caspase-3, Bax and Bcl-2. Our results revealed that pre-treatment with matrine significantly decreased the infarct volume and improved the neurological scores. Matrine also reduced the percentage of apoptotic neurons and relieved neuronal morphological damage. Furthermore, matrine markedly decreased the MDA levels, and increased SOD, GSH-Px and CAT activity, and T-AOC. Western blot analysis and immunofluorescence staining revealed a marked decrease in caspase-3 expression and an increase in the Bcl-2/Bax ratio in the group pre-treated with matrine (30 mg/kg) as compared with the vehicle-treated group. The findings of the present study demonstrate that matrine exerts neuroprotective effects against cerebral ischemic injury and that these effects are associated with its antioxidant and anti-apoptotic properties.


Subject(s)
Alkaloids/therapeutic use , Antioxidants/therapeutic use , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Brain/drug effects , Neuroprotective Agents/therapeutic use , Quinolizines/therapeutic use , Alkaloids/isolation & purification , Animals , Antioxidants/isolation & purification , Apoptosis/drug effects , Brain/pathology , Brain Ischemia/metabolism , Male , Mice , Oxidative Stress/drug effects , Quinolizines/isolation & purification , Sophora/chemistry , Matrines
18.
Inflammation ; 38(6): 2259-68, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26178478

ABSTRACT

Oxysophoridine (OSR) is a bioactive alkaloid extracted from the Sophora alopecuroides Linn. Our aim is to explore the potential anti-inflammation mechanism of OSR in cerebral ischemic injury. Mice were intraperitoneally pretreated with OSR (62.5, 125, and 250 mg/kg) or nimodipine (Nim) (6 mg/kg) for 7 days followed by cerebral ischemia. The inflammatory-related cytokines in cerebral ischemic hemisphere tissue were determined by immunohistochemistry staining, Western blot and enzyme-like immunosorbent assay (ELISA). OSR-treated groups observably suppressed the nuclear factor kappa B (NF-κB), intercellular adhesion molecule-1 (ICAM-1), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2). OSR-treated group (250 mg/kg) markedly reduced the inflammatory-related protein prostaglandin E2 (PGE2), tumor necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß), interleukin-6 (IL-6), and interleukin-8 (IL-8). Meanwhile, it dramatically increased the interleukin-10 (IL-10). Our study revealed that OSR protected neurons from ischemia-induced injury in mice by downregulating the proinflammatory cytokines and blocking the NF-κB pathway.


Subject(s)
Alkaloids/pharmacology , Anti-Inflammatory Agents/pharmacology , Brain Ischemia/drug therapy , Brain/drug effects , Neuroprotective Agents/pharmacology , Reperfusion Injury/prevention & control , Animals , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Dinoprostone/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Inflammation Mediators/metabolism , Intercellular Adhesion Molecule-1/metabolism , Male , Mice, Inbred ICR , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/drug effects , Time Factors , Transcription Factor RelA/metabolism
19.
J Nat Med ; 69(4): 575-83, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26142710

ABSTRACT

Aloperine (ALO), one of the alkaloids isolated from Sophora alopecuroides L., is traditionally used for various diseases including neuronal disorders. This study investigated the protective effects of ALO on neonatal rat primary-cultured hippocampal neurons injured by oxygen-glucose deprivation and reperfusion (OGD/RP). Treatment with ALO (25, 50, and 100 mg/l) attenuated neuronal damage (p < 0.01), with evidence of increased cell viability (p < 0.01) and decreased cell morphologic impairment. Furthermore, ALO increased mitochondrial membrane potential (p < 0.01), but inhibited intracellular-free calcium [Ca(2+)] i (p  < 0.01) elevation in a dose-dependent manner at OGD/RP. ALO also reduced the intracellular reactive oxygen species and malondialdehyde production and enhanced the antioxidant enzymatic activities of catalase, superoxide dismutase, glutathione peroxidase and the total antioxidant capacity. The results suggested that ALO has significant neuroprotective effects that can be attributed to anti-oxidative stress.


Subject(s)
Hippocampus/drug effects , Neurons/drug effects , Piperidines/therapeutic use , Reperfusion Injury/metabolism , Animals , Apoptosis , Glucose/metabolism , Neuroprotective Agents/pharmacology , Oxygen , Piperidines/administration & dosage , Quinolizidines , Rats , Rats, Sprague-Dawley
20.
Ann Indian Acad Neurol ; 17(3): 313-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25221402

ABSTRACT

BACKGROUND: Our previous studies demonstrated that oxysophoridine (OSR) had neuroprotective effects on mice through antioxidant and anti-apoptotic mechanisms. In this study, we investigated whether OSR could influence the release of amino acids in ischemic mice brains. MATERIALS AND METHODS: Male ICR mice were scheduled to undergo 2 h middle cerebral artery occlusion (MCAO) and 24 h reperfusion. Before MCAO, mice in corresponding groups were intraperitoneally injected with OSR (62.5, 125 and 250 mg/kg) for seven successive days. After reperfusion, neurological scores were estimated, infarct volume and the brain water content were assessed. The levels of glutamate (Glu), aspartate (Asp), γ-aminobutyric acid (GABA) and Glycine (Gly) were measured by amino acid analyzer. RESULTS: OSR significantly decreased neurological scores, reduced infarct volume and the brain water content. After treatment with OSR of 250 mg/kg, the contents of Glu, Asp, GABA and Gly in mice brains could maintain at a normal level compared with MCAO group mice. The Glu/GABA ratio was significantly decreased in OSR group mice. CONCLUSION: These findings indicate that OSR has a protective effect on cerebral ischemic injury and helps to maintain the amino acids homeostasis after reperfusion for a long time.

SELECTION OF CITATIONS
SEARCH DETAIL
...