Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Heliyon ; 10(3): e24988, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38317912

ABSTRACT

Background: Cancer immunotherapies aimed at activating immune system, especially by blocking immune checkpoints, have become a successful modality for treating patients with advanced cancers. However, its clinical practice is frequently conceded by high outcomes, low initial response rates and severe side effects. New strategies are necessary to complement and advance this biological therapy. Erzhi Pills (EZP) have diverse pharmaceutical effects including immune regulation, anti-tumor and anti-senescence. We hypothesized that EZP could exert its antitumor effect through immunomodulation. Purpose: The aim of this study was to investigate the effects of EZP on anti-tumor activities, and define its molecular mechanisms. Methods: By applying melanoma model with high immune infiltrates, we determined the anti-melanoma effect of EZP. To identify whether this effect was mediated by direct targeting tumor cells, cell viability and apoptosis were examined in vitro. Network pharmacology analysis was used to predict the potential mechanisms of EZP for melanoma via immune response. Flow cytometry, immunohistochemistry (IHC), enzyme-linked immunosorbent assay (ELISA) and crystal violet (CV) experiments were performed to detect T cell infiltrations and functions mediated by EZP. The mechanism of EZP was further investigated by western blotting both in vivo and in vitro. Results: The administration of EZP significantly inhibited tumor weight and volume. EZP extract could only slightly reduce cell viability and induce melanoma apoptosis. Network pharmacology analysis predicted that JAK-STAT signaling pathway and T cell receptor signaling pathway might be involved during EZP treatment. Flow cytometry and IHC analyses showed that EZP increased the number of CD4+ T cells and enhanced the function of CD8+ T cells. In co-culture experiments, EZP elevated killing ability of T cells. Western blotting showed that EZP treatment reduced PD-L1 signaling pathway. Conclusion: These findings indicated that EZP exerted anti-melanoma effects by inducing apoptosis and blocking PD-L1 to activate T cells. EZP might represent a promising candidate drug for cancer immunotherapies.

2.
Phytomedicine ; 104: 154271, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35777120

ABSTRACT

BACKGROUND: Coronary thrombosis and its correlated disorders are main healthcare problems globally. The therapeutic effects of current treatments involving antiplatelet drugs are not fully satisfactory. Danshensu (DSS) is an important monomer obtained from Salvia miltiorrhiza roots that have been widely employed for vascular diseases in medicinal practices. Nonetheless, the underlying mechanisms of DSS are not fully unraveled. PURPOSE: The objective of this study was to penetrate the antithrombotic and antiplatelet mechanisms of DSS. METHODS: Network pharmacology assay was used to forecast the cellular mechanisms of DSS for treating thrombosis. The work focused the impacts of DSS on platelet activation by analyzing aggregation and adhesion in vitro. Flow cytometry, western blotting, CM-H2DCFDA staining and mitochondrial function assays were performed to reveal the molecular mechanisms. The model of common carotid artery thrombus induced by ferric chloride was established. The wet weight of thrombus was measured, and the thrombosis was observed by hematoxylin and eosin (H&E) staining, in order to support the inhibitory effect of DSS on thrombosis. RESULTS: Data mining found the antithrombotic effect of DSS is related to platelet activation and the core target is silent information regulator 1 (SIRT1). We confirmed that DSS dose-dependently inhibited platelet activation in vitro. DSS was further demonstrated to induce the expression of SIRT1 and decreased reactive oxygen species (ROS) burden and thereby prevented mitochondrial dysfunction. Mitochondrial function tests further indicated that DSS prevented mitochondrial DNA (mtDNA) release, which induced activation of platelet in a dendritic cell specific intercellular-adhesion-molecule-3 grabbing non-integrin (DC-SIGN)-dependent manner. In carotid artery injury model induced by ferric chloride, DSS inhibited the development of carotid arterial thrombosis. More encouragingly, in tail bleeding time assay, DSS did not augment bleeding risk. CONCLUSION: These findings indicated that DSS effectively inhibited platelet activation by depressing the collection of ROS and the release of platelet mtDNA without arousing hemorrhage risk. DSS might represent a promising candidate drug for thrombosis and cardiovascular disease therapeutics.


Subject(s)
Sirtuin 1 , Thrombosis , DNA, Mitochondrial , Fibrinolytic Agents/pharmacology , Humans , Lactates , Mitochondria/metabolism , Platelet Activation , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism , Thrombosis/chemically induced , Thrombosis/drug therapy , Thrombosis/prevention & control
3.
J Ethnopharmacol ; 292: 115166, 2022 Jun 28.
Article in English | MEDLINE | ID: mdl-35248678

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Shuxuetong (SXT) injection is formulated by leech and earthworm, has been widely used in the treatment of thrombotic cardiovascular and cerebrovascular diseases with remarkable clinical efficacy. AIM OF THE STUDY: The purpose of this study is to investigate the protective mechanism of SXT injection on the mice model of hindlimb ischemia, and to evaluate the angiogenic effects of SXT injection and its main active substances. MATERIALS AND METHODS: Hindlimb ischemia was induced by left femoral artery ligation. After operation, the mice were injected with saline, 10 mg/kg/d cilostazol, 37.5 mg/kg/d SXT injection, 75 mg/kg/d SXT injection and 150 mg/kg/d SXT injection via tail vein for 4 weeks. Ischemia severity was assessed using laser Doppler perfusion imaging system. Tissue recovery and capillary density were evaluated by histological and immunofluorescent staining. Vascular endothelial growth factor-A (VEGF-A) and platelet-derived growth factor (PDGF-BB) expression were measured by reverse transcription polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) analyses. Human umbilical vein endothelial cells (HUVECs) proliferation was measured using a BrdU kit and the viability of HUVECs was performed by MTT assay. Migration of HUVECs was performed by the wound healing method and a modified transwell assay. Capillary tube formation by HUVECs was examined by using Matrigel assay. Western blotting was used to detect the expressions of p-Cofilin, p-MYPT1, and p-LIMK1. RESULTS: SXT injection treatment significantly restored the blood flow and reduced tissue injury in mouse gastrocnemius muscle. SXT injection treatment increased capillary density and promoted angiogenesis in hindlimb ischemia. Moreover, SXT injection enhanced the expression of VEGF-A and PDGF-BB at both mRNA and protein levels in ischemic tissue of mice. SXT injection and its main active peptides dramatically increased the migration and capillary tube formation of HUVECs. SXT injection and its peptides enhanced protein expressions of the phosphorylation of MYPT1, Cofilin, and LIMK1. DSYVGDEAQSKR, YNELRVAPEEHP, and IQFLPEGSPVTM may act as the active components of SXT injection. CONCLUSION: SXT injection promoted angiogenesis and improved function recovery in hindlimb ischemia mice by regulation of VEGF-A/PDGF-BB. Moreover, SXT injection and its active peptides induced cell migration and tube formation in HUVECs through activating the MYPT1/LIMK1/Cofilin pathway. This study provided experimental basis for SXT injection in the treatment of ischemic diseases and revealed the effective substance of SXT injection in regulating angiogenesis, providing better evidence for the clinical application of SXT injection.


Subject(s)
Neovascularization, Physiologic , Vascular Endothelial Growth Factor A , Actin Depolymerizing Factors/metabolism , Actin Depolymerizing Factors/pharmacology , Animals , Becaplermin , Drugs, Chinese Herbal , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Humans , Ischemia/drug therapy , Ischemia/metabolism , Lim Kinases/metabolism , Mice , Myosin-Light-Chain Phosphatase/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
Ecotoxicol Environ Saf ; 234: 113329, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35255253

ABSTRACT

Copper is a trace element necessary for the normal functioning of organisms, but excessive copper contents may be toxic to the heart. The goal of this study was to investigate the role of excessive copper accumulation in mitochondrial damage and cell apoptosis inhibition. In vivo, the heart copper concentration and cardiac troponin I (c-TnI) and N-terminal forebrain natriuretic peptide (NT-pro-BNP) levels increased in the copper-laden model group compared to those of the control group. Histopathological and ultrastructural observations revealed that the myocardial collagen volume fraction (CVF), perivascular collagen area (PVCA) and cardiomyocyte cross-sectional area (CSA) were markedly elevated in the copper-laden model group compared with the control group. Furthermore, transmission electron microscopy (TEM) showed that the mitochondrial double-layer membrane was incomplete in the copper-laden model groups. Furthermore, cytochrome C (Cyt-C) expression was downregulated in mitochondria but upregulated in the cytoplasm in response to copper accumulation. In addition, Bcl-2 expression decreased, while Bax and cleaved caspase-3 levels increased. These results indicate that copper accumulation in cardiomyocyte mitochondria induces mitochondrial injury, and Cyt-C exposure and induces apoptosis, further resulting in heart damage.

5.
J Ginseng Res ; 45(6): 665-675, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34764721

ABSTRACT

BACKGROUND: Ginsenoside Rg1 (Rg1), an active ingredient in ginseng, may be a potential agent for the treatment of Alzheimer's disease (AD). However, the protective effect of Rg1 on neurodegeneration in AD and its mechanism of action are still incompletely understood. METHODS: Wild type (WT) and APP/PS1 AD mice, from 6 to 9 months old, were used in the experiment. The open field test (OFT) and Morris water maze (MWM) were used to detect behavioral changes. Neuronal damage was assessed by hematoxylin and eosin (H&E) and Nissl staining. Immunofluorescence, western blotting, and quantitative real-time polymerase chain reaction (q-PCR) were used to examine postsynaptic density 95 (PSD95) expression, amyloid beta (Aß) deposition, Tau and phosphorylated Tau (p-Tau) expression, reactive oxygen species (ROS) production, and NAPDH oxidase 2 (NOX2) expression. RESULTS: Rg1 treatment for 12 weeks significantly ameliorated cognitive impairments and neuronal damage and decreased the p-Tau level, amyloid precursor protein (APP) expression, and Aß generation in APP/PS1 mice. Meanwhile, Rg1 treatment significantly decreased the ROS level and NOX2 expression in the hippocampus and cortex of APP/PS1 mice. CONCLUSIONS: Rg1 alleviates cognitive impairments, neuronal damage, and reduce Aß deposition by inhibiting NOX2 activation in APP/PS1 mice.

6.
Phytomedicine ; 91: 153675, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34332285

ABSTRACT

BACKGROUND: Xin-Ji-Er-Kang (XJEK) as a herbal formula of traditional Chinese medicine (TCM) has shown the protective effects on myocardial function as well as renal function in mouse models of myocardial infarction. HYPOTHESIS/PURPOSE: We investigated the effects of XJEK on cardiovascular- and renal-function in a heart failure mouse model induced by high salt (HS) and the associated mechanisms. STUDY DESIGN: For the purpose of assessing the effects of XJEK on a hypertensive heart failure model, mice were fed with 8% high salt diet. XJEK was administered by oral gavage for 8 weeks. Cardiovascular function parameters, renal function associated biomarkers and XJEK's impact on renin-angiotensin-aldosterone system (RAAS) activation were assessed. To determine the underlying mechanism, the calpain1/junctophilin-2 (JP2)/sarcoplasmic reticulum Ca2+ ATPase (SERCA2a) pathway was further studied in AC16 cells after angiotensin II-challenge or after calpastatin small interfering RNA (siRNA) transfection. RESULTS: Mice on HS-diet exhibited hypertensive heart failure along with progressive kidney injury. Similar to fosinopril, XJEK ameliorated hypertension, cardiovascular-and renal- dysfunction in mice of HS-diet group. XJEK inhibited HS-induced activation of RAAS and reversed the abnormal expression pattern of calpain1and JP2 protein in heart tissues. XJEK significantly improved cell viability of angiotensin II-challenged AC16 cells. Moreover, XJEK's impact on calpain1/JP2 pathway was partly diminished in AC16 cells transfected with calpastatin siRNA. CONCLUSION: XJEK was found to exert cardiovascular- and renal protection in HS-diet induced heart failure mouse model. XJEK inhibited HS-diet induced RAAS activation by inhibiting the activity and expression of calpain1 and protected the junctional membrane complex (JMC) in cardiomyocytes.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Heart Failure , Hypertension , Animals , Blood Pressure , Calpain , Heart Failure/drug therapy , Hypertension/drug therapy , Kidney/drug effects , Kidney/physiology , Membrane Proteins , Mice , Muscle Proteins , Oxidative Stress , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Signal Transduction
7.
J Ethnopharmacol ; 279: 114350, 2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34157326

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Astragalus is a medicinal herb used in China for the prevention and treatment of diseases such as diabetes and cancer. As one of the main active ingredients of astragalus, Astragaloside IV (AS-IV) has a wide range of pharmacological effects, including anti-inflammation and anti-cancer effects. AIM OF THE STUDY: Different phosphorylated forms of Smad3 differentially regulate the progression of hepatic carcinoma. The phosphorylation of the COOH-terminal of Smad3 (pSmad3C) and activation of the Nrf2/HO-1 pathway inhibits hepatic carcinoma, while phosphorylation of the linker region of Smad3 (pSmad3L) promotes progression. Thus, pSmad3C/3L and Nrf2/HO-1 pathways are potential targets for drug of anti-cancer development. AS-IV is anti-apoptotic and can inhibit hepatocellular carcinoma cell (HCC) proliferation, invasion, and tumor growth in nude mice. However, it is not clear whether AS-IV has a therapeutic effect on inhibiting the progression of primary liver cancer by regulating the pSmad3C/3L and Nrf2/HO-1 pathway. The purpose of this study is to investigate whether AS-IV inhibits hepatocellular carcinoma by regulating pSmad3C/3L and Nrf2/HO-1 pathway. MATERIALS AND METHODS: primary liver cancer in mice induced by DEN/CCl4/C2H5OH (DCC) and HSC-T6/HepG2 cell models activated by TGF-ß1 was investigated for the mechanisms of AS-IV. In vivo assays included liver biopsy, histopathology and post-mortem analysis included immunohistochemistry, immunofluorescent, and Western blotting analysis, and in vitro assays included immunofluorescent, and Western blotting analysis. RESULTS: AS-IV significantly inhibited the development of primary liver cancer, reflecting improved liver biopsy, histopathology. The incidence and multiplicity of primary liver cancer were markedly decreased by AS-IV treatment at the 20th week. AS-IV had observable effects on the TGF-ß1/Smad and Nrf2/HO-1 expression in vivo, especially up-regulated pSmad3C, pNrf2, HO-1, and NQO1, while it down-regulated pSmad2C, pSmad2L, pSmad3L, PAI-1, and α-SMA at the 12th week and the 20th week. Furthermore, in vitro analysis further confirmed that AS-IV regulated the expression of pSmad3C/3L and Nrf2/HO-1 pathway in HSC-T6 and HepG2 cells activated by TGF-ß1. CONCLUSION: AS-IV administration delays the occurrence of primary liver cancer by continually suppressing the development of fibrosis, the mechanism of the therapeutic effect involving the regulation of the pSmad3C/3L and Nrf2/HO-1 pathways, especially in regulation reversibility and antagonism of pSmad3C and pSmad3L and promoting the phosphorylation of Nrf2.


Subject(s)
Carcinoma, Hepatocellular/prevention & control , Liver Cirrhosis/drug therapy , Liver Neoplasms/prevention & control , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Astragalus Plant/chemistry , Cell Line , Heme Oxygenase-1/metabolism , Hep G2 Cells , Humans , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Nude , NF-E2-Related Factor 2/metabolism , Phosphorylation/drug effects , Rats , Saponins/isolation & purification , Smad3 Protein/metabolism , Triterpenes/isolation & purification
8.
Naunyn Schmiedebergs Arch Pharmacol ; 394(8): 1779-1786, 2021 08.
Article in English | MEDLINE | ID: mdl-34191114

ABSTRACT

Current researches have confirmed that Smads, mediators of TGF-ß signaling, are strictly controlled by domain-specific site phosphorylation in the process of hepatic disease. Usually, Smad3 phospho-isoform pSmad3L and pSmad3C are reversible and antagonistic; pSmad2L/C could act together with pSmad3L by stimulating PAI-1 expression and ECM synthesis to transmit fibrogenic signals. Our recent study found that pSmad3C mutation is supposed to perform a vigorous role on the early phase of liver injury and abates salvianolic acid B's anti-hepatic fibrotic-carcinogenesis. However, whether pSmad3C mutation expedites pSmad2L/C-mediated signaling transduction during hepatic fibrogenesis remains vague. Presently, Smad3 gene C-terminal phosphorylation site mutation heterozygote (pSmad3C+/-) mice were constructed to probe if and how pSmad3C retards CCl4-induced hepatic fibrogenesis by inhibiting pSmad2L/C-mediated signaling transduction. Twelve 6-week-old pSmad3C+/- C57BL/6J mice were intraperitoneally injection with CCl4 for 6 weeks to induce liver fibrogenesis. Results showed that pSmad3C mutation aggravates the relative liver weight, biochemical parameters, collagenous fibers and fibrotic septa formation, contributes to fibrogenesis in HT-CCl4 mice. Furthermore, fibrotic-related proteins TGF-ß1, pSmad2C, pSmad2L, and PAI-1 were also increased in CCl4-induced pSmad3C+/- mice. These results suggest that pSmad3C mutation exacerbates hepatic fibrogenesis which relates to intensifying pSmad2L/C-mediated signaling transduction.


Subject(s)
Liver Cirrhosis/physiopathology , Phosphorylation/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Animals , Carbon Tetrachloride , Disease Models, Animal , Liver Cirrhosis/genetics , Mice , Mice, Inbred C57BL , Mutation , Serpin E2/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta1/metabolism
9.
Exp Ther Med ; 22(1): 782, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34055081

ABSTRACT

Lipopolysaccharide (LPS) is a toxic component of cell walls of Gram-negative bacteria that are widely present in gastrointestinal tracts. Increasing evidence showed that LPS plays important roles in the pathogeneses of neurodegenerative disorders, such as Alzheimer's disease (AD). NADPH oxidase s2 (NOX2) is a complex membrane protein that contributes to the production of reactive oxygen species (ROS) in several neurological diseases. The NLRP1 inflammasome can be activated in response to an accumulation of ROS in neurons. However, it is still unknown whether LPS exposure can deteriorate neuronal damage by activating NOX2-NLRP1 inflammasomes. Ginsenoside Rg1 (Rg1) has protective effects on neurons, although whether Rg1 alleviates LPS-induced neuronal damage by inhibiting NOX2-NLRP1 inflammasomes remains unclear. In the present study, the effect of concentration gradients and different times of LPS exposure on neuronal damage was investigated in HT22 cells, and further observed the effect of Rg1 treatment on NOX2-NLPR1 inflammasome activation, ROS production and neuronal damage in LPS-treated HT22 cells. The results demonstrated that LPS exposure significantly induced NOX2-NLRP1 inflammasome activation, excessive production of ROS, and neuronal damage in HT22 cells. It was also shown that Rg1 treatment significantly decreased NOX2-NLRP1 inflammasome activation and ROS production and alleviated neuronal damage in LPS-induced HT22 cells. The present data suggested that Rg1 has protective effects on LPS-induced neuronal damage by inhibiting NOX2-NLRP1 inflammasomes in HT22 cells, and Rg1 may be a potential therapeutic approach for delaying neuronal damage in AD.

10.
J Ethnopharmacol ; 276: 114187, 2021 Aug 10.
Article in English | MEDLINE | ID: mdl-33957207

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The genus Hypericum are widely distributed in China. Hypericum perforatum L. (genus Hypericum, family Hypericaceae) has a long history as a traditional Chinese medicine, which was traditionally used for the treatment of emotional distress, cardiothoracic depression, and acute mastitis. Hyperoside (Hyp) extracted from Hypericum perforatum L. has been affirmed to exert therapeutic effects on cardiovascular diseases, with widespread existence in plants of genus Hypericum. Hyp could also be extracted from Crataegus pinnatifida Bunge (genus Crataegus pinnatifida Bunge, family Rosaceae), another traditional Chinese medicine that traditionally prevented and treated heart disease in China. The cardioprotection and mechanism of Hyp comprise anti-inflammation, anti-fibrosis, activation of autophagy, and reversal of cardiac remodeling. AIM OF THE STUDY: This study aimed to explore the Hyp effect against MI and its underlying mechanism. MATERIALS AND METHODS: The MI model was constructed in the KM mice via a ligating surgery of the left anterior descending (LAD) coronary artery. Subsequently, the mice were divided into following seven groups: Sham group, MI group, MI + Hyp 9 mg/kg group, MI + Hyp18 mg/kg group, MI + Hyp36 mg/kg group, MI + Fosinopril group, and MI + Hyp-36 mg/kg+3-MA group. Each group was treated with Hyp in different concentrations or positive medicine for two weeks except for the sham group. After two weeks, we examined the cardiac function, electrocardiogram (ECG), myocardial hypertrophy in the non-infarct area, collagen volume fraction (CVF), perivascular collagen area (PVCA) in the infarct area, and several serum cytokines. Autophagy and inflammation in cardiomyocytes were assessed via measuring autophagy-associated proteins and NLRP1 inflammasome pathway related proteins. RESULTS: Hyp reversed LV remodeling and adverse ECG changes through reducing CVF and myocardial hypertrophy. Additionally, Hyp treatment could reduce inflammation levels in cardiomyocytes, compared with those in MI group. Moreover, NLRP1inflammation pathway was activated after MI. Up-regulation of autophagic flux suppressed NLRP1 inflammation pathway after Hyp treatment. However, co-treatment with 3-MA abrogated above effects of Hyp. CONCLUSIONS: Hyp had obvious protective effect on heart injury in MI mice. Echocanrdiographic and histological measurements demonstrated that Hyp treatment improved cardiac function, and ameliorated myocardial hypertrophy and fibrinogen deposition after MI. The partial mechanism is that Hyp could up-regulate autophagy after MI. Furthermore, the promotion of autophagic flux would suppress NLRP1 inflammation pathway induced by MI.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Cardiotonic Agents/pharmacology , Inflammation/prevention & control , Myocardial Infarction/prevention & control , Quercetin/analogs & derivatives , Animals , Body Weight/drug effects , Cardiotonic Agents/therapeutic use , Cytokines/blood , Disease Models, Animal , Electrocardiography/drug effects , Heart Diseases/etiology , Heart Diseases/prevention & control , Hypericum/chemistry , Inflammation/metabolism , Male , Medicine, Chinese Traditional , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/drug effects , Organ Size/drug effects , Quercetin/pharmacology , Quercetin/therapeutic use , Signal Transduction/drug effects , Ventricular Remodeling/drug effects
12.
Food Chem Toxicol ; 147: 111912, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33290806

ABSTRACT

Smad3 phosphorylation is implicated in hepatic fibro-carcinogenesis. Moreover, Smad3 phospho-isoform pSmad3L and pSmad3C are reversible and antagonistic, and the balance could shift from carcinogenesis to tumor-suppression. pSmad3C has recently assigned to perform a preventative effect against primary liver injury. Salvianolic acid B (Sal B), a component derived from Salvia miltiorrhiza, is empirically used for hepatic diseases. Our prior study clarified that Sal B could delay hepatic fibrosis-carcinoma progression by converting pSmad3L/3C in mice. However, the roles of Smad3 phospho-isoform conversion and antagonism in the anti-hepatocarcinogenic effects of Sal B in pSmad3C- or/and pSmad3L-mutated mice/cells remain vague. Currently, corresponding doses/concentrations of Sal B was co-administrated to pSmad3C+/- mutational mice/plasmids-transfected HepG2 cells. Notably, in vivo functional studies revealed that pSmad3C mutation attenuates Sal B-induced ameliorative effects on histopathological characteristics and decreased serological biomarkers, and potential mechanism involves attenuation of increases in pSmad3C/p21 and decreases in pSmad3L/PAI-1/c-Myc expression. Expectedly, in vitro results showed that up-regulating pSmad3C enhances the inhibitory effects on proliferation, migration and contributes to apoptosis accompanied by a shift of pSmad3L/PAI-1/c-Myc oncogenic to pSmad3C/p21 tumour-suppressive signalling; however, opposite effects occur when upregulated pSmad3L. This study is the first to identify pSmad3C as a key target by which Sal B prevents hepatocarcinogenesis.


Subject(s)
Benzofurans/pharmacology , Liver Neoplasms/prevention & control , Smad3 Protein/metabolism , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Mice , Mutation , Phosphorylation , Random Allocation , Smad3 Protein/genetics , Up-Regulation/drug effects
13.
Pharm Biol ; 58(1): 845-853, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32870741

ABSTRACT

CONTEXT: Cryptotanshinone (CT) is a diterpene quinone compound from Salvia miltiorrhiza Bge. Labiatae has been widely used in cardio-cerebral vascular diseases, which could be potentially effective in treating diabetic wounds. OBJECTIVE: This study evaluates the wound healing activity of CT by employing an excisional wound splinting model in db/db mice. MATERIALS AND METHODS: Wounds were induced at the dorsum of non-diabetic (db/+) and diabetic (db/db) mice and treated with sodium carboxymethyl cellulose (CMC-Na) or 300 mg/kg/d CT for 16 days. Wound closure was measured every two days. Body weight, fasting blood glucose, re-epithelialization, granulation, leukocyte infiltration, capillary density, collagen deposition and expressions of CXCL1, CXCL2, VEGF, Ang-1, p-eNOS, eNOS, α-SMA, MMP2 and MMP9 were analysed. Expression of VEGF and tube formation was measured in vitro with human umbilical vein endothelial cells (HUVECs). RESULTS: CT significantly accelerated rate of wound closure, as the contraction ratio increased from 68% (non-treated group) to 83% (CT-treated group) at days 16 post-injury. A significant increase was observed in re-epithelialization and granulation tissue formation. Mechanistically, CT suppressed leukocyte infiltration and CXCL1 and CXCL2 expression. CT treatment also increased blood vessel density and expression level of VEGF, Ang-1 and p-eNOS. In vitro, CT boosted expression of VEGF and tube formation of endothelial cells. Moreover, extracellular matrix (ECM) remodelling was enhanced by CT via promoting fibroblast transformation and inhibiting MMP2 and MMP9. CONCLUSIONS: Our study provides evidence that CT could be developed as a potential therapeutic agent for the treatment of chronic diabetic wound healing.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Phenanthrenes/pharmacology , Wound Healing/drug effects , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/drug therapy , Inflammation/pathology , Male , Mice , Neovascularization, Physiologic/drug effects , Time Factors
14.
Arch Pharm Res ; 43(8): 863-874, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32720163

ABSTRACT

Acute myocardial infarction (AMI) results in irreversible cardiac cell damage or death because of decreased blood flow to the heart. Apoptosis plays an important role in the process of tissue damage after myocardial infarction (MI), which has pathological and therapeutic implications. Ferulic acid (FA) is a phenolic acid endowed with strong antioxidative and cytoprotective activities. The present study aimed to investigate whether FA protects cardiomyocytes from apoptosis by regulating autophagy, which is a cellular self-digestion process, and one of the first lines of defense against oxidative stress. Apoptosis was induced by TNF-α (10 ng/mL) and cycloheximide (CHX, 5 µg/mL) in rat H9c2 cardiomyocytes. FA-inhibited TNF-α/CHX-induced apoptosis was determined by the quantification of TUNEL-positive cells, and the effect was associated with decreased ROS production and inhibited caspase3 activation. FA treatment enhanced autophagy and increased autophagy-associated protein expression, leading to an inhibition of mTOR signaling. When co-treated with 3-methyladenine (3-MA), an autophagy inhibitor, the anti-apoptotic effect of FA was attenuated. In an in vivo mouse MI model, FA treatment decreased the apoptotic cell number, reduced infarct size, and improved cardiac performance, as determined by histological and echocardiographic assessments. Taken collectively, these results suggest that FA could protect cardiomyocytes from apoptosis by enhancing autophagy.


Subject(s)
Apoptosis/drug effects , Coumaric Acids/pharmacology , Myocardial Infarction/drug therapy , Myocytes, Cardiac/drug effects , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagy/drug effects , Cell Line , Cycloheximide/administration & dosage , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/administration & dosage
15.
Pharm Biol ; 58(1): 165-175, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32608342

ABSTRACT

Context: Astragali Radix (AR) and Angelica sinensis Radix (ASR) combinations are used to treat cardiovascular disorders.Objectives: This study investigates the protective effects of different compatibility proportions of AR and ASR on cardiac dysfunction in a C57BL/6 mouse model of myocardial infarction (MI).Materials and methods: MI mice were induced by ligation of the left coronary artery and divided into six groups: sham, vehicle, 10 mg/kg/d simvastatin and combinations of AR and ASR at different ratios, including 1:1 (AR 2.5 g/kg + ASR 2.5 g/kg), 3:1 (AR 3.75 g/kg + ASR 1.25 g/kg) and 5:1 (AR 4.17 g/kg + ASR 0.83 g/kg). Both AR-ASR combinations and simvastatin were dissolved in saline solution and given daily by gavage. The left ventricle function, infarct size, heart tissue injury, apoptosis of cardiomyocytes, leukocyte infiltrates, capillary density and expression of cleaved caspase-3, cleaved caspase-9, Bcl-2, Bax, Bad, IL-1ß, IL-6, VEGF, p-Akt and p-eNOS were analysed.Results: Different combinations of AR and ASR improve cardiac function and reduce infarct size (61.15% vs. 39.3%, 42.65% and 45.5%) and tissue injury through different mechanisms. When AR was combined with ASR at ratio of 1:1, the inflammation and cardiomyocyte apoptosis were suppressed (p < 0.05, p < 0.01). The combination ratio of 3:1 exerted effect in promoting angiogensis (p < 0.05). In the combination of AR and ASR at 5:1 ratio, angiogenesis was significantly improved (p < 0.01) and the apoptosis was inhibited (p < 0.05).Conclusions: Our results reflect the regulation of multiple targets and links in herb pairs and provide an important basis for the use of AR and ASR combinations in the treatment of MI.


Subject(s)
Angelica sinensis , Astragalus Plant , Cardiotonic Agents/therapeutic use , Myocardial Infarction/drug therapy , Plant Extracts/therapeutic use , Plant Roots , Animals , Cardiotonic Agents/isolation & purification , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Plant Extracts/isolation & purification
16.
J Ethnopharmacol ; 227: 121-130, 2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30170078

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Naoxintong (NXT) is a compound preparation that is widely used in patients with cardiovascular and cerebrovascular diseases. AIM OF STUDY: The aim of this study is to investigate the protective mechanism of NXT on the mice model of peripheral vascular disease (PAD). MATERIALS AND METHODS: In the study, hindlimb ischemia was induced by ligation of femoral artery on the right leg of mice. After surgery, the mice were administrated with saline solution, 10 mg/kg/d simvastatin and 700 mg/kg/d NXT for 4 weeks. The blood flow perfusion was measured by laser Doppler perfusion imaging system. Histological and immunofluorescent staining was used to determine muscle recovery, capillary density, tissue vascular endothelial growth factor (VEGF), phosphorylated-Akt (p-Akt) and phosphorylated-endothelial nitric oxide synthase (p-eNOS) expression. Terminal deoxynucleotidyl transferased UTP nick end labeling (TUNEL) was performed to detect the apoptosis of myocytes in hindlimb. The autophagy-associated gene expression and peroxisome proliferator-activated receptors (PPARs) expression were measured by Quantitative Real-Time Reverse Transcription Polymerase Chain Reaction (qRT-PCR). Western blotting was performed to detect the expressions of light-chain 3 (LC3), VEGF, p-Akt, p-eNOS and PPARs. The EMSA experiment was performed to figure out whether PPARδ could directly bind to the predicted PPRE motif of VEGF. RESULTS: NXT treatment significantly accelerated perfusion recovery and reduced tissue injury in mice muscle. Apoptosis and autophagy were decreased within the ischemic muscle of NXT-treated mice. Quantification of vessels in hindlimb muscles provided evidences that NXT promoted angiogenesis in peripheral ischemia. In addition, results from western blotting and immunofluorescent staining suggested NXT induced angiogenesis via VEGF/Akt/eNOS signaling pathway. More interestingly, NXT specifically increased the expression of PPARδ in both mRNA and protein levels. EMSA results showed that PPARδ associated with PPRE site of VEGF promoter, suggesting that NXT-induced VEGF expression is mediated, at least in part, by PPARδ. CONCLUSION: In conclusion, the present study implicated that the restoration of hindlimb blood perfusion and recovery of limb functions were improved in NXT-treated mice with significant improvement of angiogenesis mediated by PPARδ-VEGF-Akt-eNOS axis as well as attenuation of autophagy and apoptosis. These results expand knowledge about the beneficial effects of NXT in angiogenesis and blood flow recovery. It might provide insight into the PPARδ regulating neovascularization in hindlimb ischemia and identify NXT as a potent new compound used for the treatment of peripheral vascular disease.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Ischemia/metabolism , PPAR delta/metabolism , Angiogenesis Inducing Agents/therapeutic use , Animals , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Hindlimb/blood supply , Hindlimb/drug effects , Ischemia/drug therapy , Ischemia/physiopathology , Male , Mice, Inbred C57BL , PPAR delta/genetics , RNA, Messenger/metabolism , Regional Blood Flow/drug effects
17.
J Steroid Biochem Mol Biol ; 172: 149-159, 2017 09.
Article in English | MEDLINE | ID: mdl-28711487

ABSTRACT

Aucubin (AU) is an iridoid glycoside that has been shown to display estrogenic properties and has various pharmacological effects. Herein, we described the angiogenic properties of AU. In the study, hindlimb ischemia was induced by ligation of femoral artery on the right leg of ovariectomized mice. AU treatment significantly accelerated perfusion recovery and reduced tissue injury in mice muscle. Quantification of CD31-positive vessels in hindlimb muscles provided evidences that AU promoted angiogenesis in peripheral ischemia. In addition, results from quantitative PCR and western blot suggested AU induced angiogenesis via vascular endothelial cell growth factor (VEGF)/Akt/endothelial nitric oxide synthase (eNOS) signaling pathway. More interestingly, AU's angiogenic effects could be completely abolished in estrogen receptor beta (ERß) knockout mice. In conclusion, the underlying mechanisms were elucidated that AU produced pro-angiogenic effects through ERß-mediated VEGF signaling pathways. These results expand knowledge about the beneficial effects of AU in angiogenesis and blood flow recovery. It might provide insight into the ERß regulating neovascularisation in hindlimb ischemia and identify AU as a potent new compound used for the treatment of peripheral vascular disease.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Estrogen Receptor beta/genetics , Iridoid Glucosides/pharmacology , Ischemia/drug therapy , Neovascularization, Physiologic/drug effects , Phytoestrogens/pharmacology , Animals , Disease Models, Animal , Estrogen Receptor beta/deficiency , Female , Femoral Artery/surgery , Gene Expression Regulation , Hindlimb , Ischemia/genetics , Ischemia/pathology , Ischemia/surgery , Ligation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Ovariectomy , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Recovery of Function/drug effects , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
18.
Am J Chin Med ; 44(6): 1207-1220, 2016.
Article in English | MEDLINE | ID: mdl-27627919

ABSTRACT

Alcoholic fatty liver (AFL) is early stage of alcoholic liver disease, which can progress to steatohepatitis, fibrosis, and cirrhosis if alcohol consumption is continued. The pathogenesis of AFL is associated with excessive lipid accumulation in hepatocytes. Resveratrol (RES), a dietary polyphenol found in red wines and grapes, has been shown to have a hepatoprotective effect. Autophagy is a crucial physiological process in cellular catabolism that involves the regulation of lipid droplets. Autophagy maintains a balance between protein synthesis, degradation and self-recycling. In the present study, we evaluated the protective effects of RES (10[Formula: see text]mg/kg, 30[Formula: see text]mg/kg, 100[Formula: see text]mg/kg) on AFL mice fed with an ethanol Lieber-DeCarli liquid diet, and HepG2 cells in the presence of oleic acid and alcohol to investigate whether resveratrol could induce autophagy to attenuate lipid accumulation. The results showed that RES (30[Formula: see text]mg/kg and 100[Formula: see text]mg/kg) treatment significantly attenuated hepatic steatosis and lowered the activities of serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), triglyceride (TG), low density lipoprotein cholesterol (LDL-C). H&E staining showed that RES reduced hepatic lipid accumulation. Transmission electron microscopy (TEM) images showed that RES treatment increased the number of autophagosomes and promoted the formation of autophagy. Western blot analysis showed that RES treatment increased the levels of microtubule-associated protein light chain3- II (LC3-II) and Beclin1, decreased expression of p62 protein. In addition, in vitro studies also demonstrated that RES led to the formation of acidic vesicular organelles (AVOs), however, 3-Methyladenine (3-MA), a specific inhibitor of autophagy, obviously inhibited the above effects of RES. In conclusion, RES has protective effects on alcoholic hepatic steatosis, and the potential mechanism might be involved in inducing autophagy.


Subject(s)
Autophagy/drug effects , Fatty Liver, Alcoholic/drug therapy , Fatty Liver, Alcoholic/physiopathology , Hepatocytes/metabolism , Lipid Droplets/metabolism , Stilbenes/pharmacology , Animals , Autophagy/physiology , Disease Models, Animal , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Hep G2 Cells , Humans , Male , Mice, Inbred C57BL , Phytotherapy , Resveratrol , Stilbenes/administration & dosage , Stilbenes/therapeutic use , Vitis , Wine
SELECTION OF CITATIONS
SEARCH DETAIL
...