Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
J Steroid Biochem Mol Biol ; 216: 106041, 2022 02.
Article in English | MEDLINE | ID: mdl-34864206

ABSTRACT

The hypoxic microenvironment of cryptorchidism is an important factor to induce the impairment of the structure and function of Sertoli cells and thus lead to spermatogenesis loss or tumorigenesis. Dihydrotestosterone (DHT), as a potent nonaromatizable 5α-reduced androgen, has both positive and negative effect on pathological fibrosis process. However, it is still unknown whether DHT can regulate hypoxia-induced fibrosis of Sertoli cells. Herein, in this study, we evaluate the DHT level, two 5α-reductase isoforms, 5α-red1 and 5α-red2, as well as HIF-1α expression pattern in canine cryptorchidism and contralateral normal testis. Results showed that the abdominal testes presented low DHT levels and 5α-red1 and 5α-red2 expression, while significantly higher HIF-1α expression and ECM production compared with the scrotum. Moreover, we established a hypoxia-induced fibrosis model in canine Sertoli cells induced by cobalt chloride (CoCl2), and found that DHT inhibited the fibrosis of Sertoli cells in a dose-dependent manner. Meanwhile, DHT interfered with the TGF-ß signaling by reducing the expression of TGF-ßRI and TGF-ßRII and inhibiting the expression and phosphorylation of Smad2 and Smad3, while flutamide (androgen receptor inhibitor) inhibited these effects of DHT. Furthermore, use of LY2109761 (TGF-ß receptor type I/II inhibitor) to interfere with the TGF-ß/Smad pathway showed a similar effect with DHT suppression of the fibrosis in Sertoli cells. Our research data demonstrated that cryptorchidism is located in a hypoxic and DHT deficiency microenvironment. Moreover, supplementing DHT can alleviate the fibrosis process of Sertoli cells caused by hypoxia, which is associated with AR regulating the inhibition of TGF-ß/Smad signaling.


Subject(s)
Cell Hypoxia/physiology , Dihydrotestosterone/pharmacology , Sertoli Cells/drug effects , Animals , Antifibrotic Agents/pharmacology , Cell Hypoxia/drug effects , Cells, Cultured , Dogs , Fibrosis/pathology , Fibrosis/prevention & control , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Scrotum/drug effects , Scrotum/metabolism , Scrotum/pathology , Sertoli Cells/metabolism , Sertoli Cells/pathology , Signal Transduction/drug effects , Smad Proteins/antagonists & inhibitors , Smad Proteins/metabolism , Testis/drug effects , Testis/metabolism , Testis/pathology , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism
2.
Bioorg Med Chem ; 49: 116438, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34610571

ABSTRACT

Liver fibrosis is one of the most common pathological consequences of chronic liver diseases (CLD). To develop effective antifibrotic strategies, a novel class of 1-(substituted phenyl)-1,8-naphthalidine-3-carboxamide derivatives were designed and synthesized. By means of the collagen type I α 1 (COL1A1)-based screening and cytotoxicity assay in human hepatic stellate cell (HSC) line LX-2, seven compounds were screened out from total 60 derivatives with high inhibitory effect and relatively low cytotoxicity for further COL1A1 mRNA expression analysis. It was found that compound 17f and 19g dose-dependently inhibited the expression of fibrogenic markers, including α-smooth muscle actin (α-SMA), matrix metalloprotein 2 (MMP-2), connective tissue growth factor (CTGF) and transforming growth factor ß1 (TGFß1) on both mRNA and protein levels. Further mechanism studies indicated that they might suppress the hepatic fibrogenesis via inhibiting both PI3K/AKT/Smad and non-Smad JAK2/STAT3 signaling pathways. Furthermore, 19g administration attenuated hepatic histopathological injury and collagen accumulation, and reduced fibrogenesis-associated protein expression in liver tissues of bile duct ligation (BDL) rats, showing significant antifibrotic effect in vivo. These findings identified 1,8-naphthalidine derivatives as potent anti-hepatic fibrosis agents, and provided valuable information for further structure optimization.


Subject(s)
1-Naphthylamine/pharmacology , Drug Discovery , Liver Cirrhosis/drug therapy , 1-Naphthylamine/chemical synthesis , 1-Naphthylamine/chemistry , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Molecular Structure , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Smad Proteins/antagonists & inhibitors , Smad Proteins/metabolism , Structure-Activity Relationship
3.
J Interferon Cytokine Res ; 41(8): 271-282, 2021 08.
Article in English | MEDLINE | ID: mdl-34410879

ABSTRACT

Epidural fibrosis after lumbar laminectomy refers to a serious complication, and excessive proliferation of fibroblasts is considered the major factor. Interferon-alpha-2b (IFN-α-2b) can exert antiviral and antiproliferative effects, which has been suggested to effectively prevent several fibrotic diseases. However, the effect of IFN-α-2b on the prevention of epidural fibrosis (EF) and its possible mechanism remain unclear. In this study, in vitro and in vivo experiments were performed to examine the possible mechanism of IFN-α-2b for preventing EF. Cell counting kit-8 (CCK-8), cell cycle test, Edu incorporation, wound healing assay, transwell test, and Western blotting assay were performed to investigate the inhibitory effect of IFN-α-2b on the proliferation and migration of fibroblasts in vitro. As indicated from the results, IFN-α-2b was capable of inhibiting proliferation and migration of fibroblasts and inhibiting the activity of the transforming growth factor ß (TGFß)/Smad signaling pathway. In vivo, the effect of IFN-α-2b on the reduction of EF was determined by performing histological macroscopic evaluation and histological and immunohistochemical staining. As suggested from the results, IFN-α-2b significantly inhibited EF after laminectomy. As revealed from the mentioned results, IFN-α-2b may have a promising application for preventing EF in the future.


Subject(s)
Epidural Space/drug effects , Fibroblasts/drug effects , Fibrosis/drug therapy , Interferon alpha-2/pharmacology , Smad Proteins/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Epidural Space/pathology , Epidural Space/surgery , Fibroblasts/metabolism , Fibrosis/pathology , Fibrosis/surgery , Humans , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
4.
Bioorg Chem ; 114: 105222, 2021 09.
Article in English | MEDLINE | ID: mdl-34375196

ABSTRACT

Seven new diterpenoids, eupholenes A-G (1-7), including two presegetanes (1 and 2), four jatrophanes (3-6), and one paraliane (7), along with 19 known analogues (8-26) were obtained by anti-liver fibrosis bioassay-guided isolation of Euphorbia sieboldiana. Their structures were elucidated by extensive spectroscopic data analyses, chemical methods, ECD calculations, and single-crystal X-ray diffractions. Euphorbesulin A (10), a presegetane diterpenoid (5/9/5 ring system), was identified as a promising anti-liver fibrosis agent that could inhibit the expressions of fibronectin (FN), α-smooth muscle actin (α-SMA), and collagen I in TGF-ß1-stimulated LX-2 cells at a micromolar level. Mechanistic study revealed that 10 suppressed liver fibrosis via inhibition of TGF-ß/Smad signaling pathway, and its potential target was TGF-ß type I receptor. These findings suggested that presegetane diterpenoid could serve as a new type of structural motif in future anti-liver fibrosis drug development.


Subject(s)
Diterpenes/pharmacology , Euphorbia/chemistry , Liver Cirrhosis/drug therapy , Plant Extracts/pharmacology , Smad Proteins/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Cells, Cultured , Diterpenes/chemistry , Diterpenes/isolation & purification , Humans , Plant Components, Aerial/chemistry , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
5.
Biomed Pharmacother ; 141: 111806, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34246190

ABSTRACT

Diabetic nephropathy (DN) is a common disease, and patients often do not have satisfactory treatments. We investigated therapeutic effects of Fuxin Granules(FX) on DN and potential molecular mechanisms. We orally administered doses of FX to db/db mice for 10 weeks and measured total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol. H&E, PAS, Masson, and Oil Red O staining were used to observe the structure of kidneys and calculate indices of kidney function. We used pharmacological analysis to investigate potential mechanisms of FX. Relative mRNA and protein levels in the TGF-ß1/Smad, TGF-ß1/Smad, and VEGF/VEGFR2 pathways were examined. TC, TG, and LDL-C were markedly reduced, lipid accumulation was low, fibrosis reduced, kidney atrophy improved, kidney lipid droplet number significantly reduced, and glomerular filtration function improved by FX treatment. Multi-channel therapeutic effects in DN through the TGF-ß1/Smad and VEGF/VEGFR2 signaling pathways occurred, and FX substantially reduced expression of TGF-ß1 in the glomeruli. FX significantly inhibited TGF-ß1, Smad2/3 total protein levels, Smad2/3 phosphorylation mRNA levels of TGF-ß1, Smad2, and Smad3. eNOS, VEGFA, and VEGFR2 expression was regulated, levels of VEGFA and VEGFR2 were decreased, and FX increased eNOS. FX ameliorated symptoms of DN, resulting in marked improvement in hyperglycemia and hyperlipidemia and optimized structure and function of kidneys in db/db mice. FX efficacy was associated with the TGF-ß1/Smad and VEGF/VEGFR2 signaling pathways. We verified this potential mechanism and hope that this study will provide benefits for the clinical treatment of DN.


Subject(s)
Diabetic Nephropathies/metabolism , Drugs, Chinese Herbal/therapeutic use , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Diabetic Nephropathies/drug therapy , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Network Pharmacology/methods , Signal Transduction/drug effects , Signal Transduction/physiology , Smad Proteins/antagonists & inhibitors , Transforming Growth Factor beta1/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
6.
J Ethnopharmacol ; 275: 114061, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33892065

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The abnormal proliferation and differentiation of cardiac fibroblasts (CFs) are universally regarded as the key process for the progressive development of cardiac fibrosis following various cardiovascular diseases. Huoxin Pill (Concentrated pill, HXP) is a Chinese herbal formula for treating coronary heart disease. However, the cellular and molecular mechanisms of HXP in the treatment of myocardial fibrosis are still unclear. AIM OF THE STUDY: To investigate the effects of HXP on CFs transdifferentiation and collagen synthesis under isoproterenol (ISO) conditions, as well as the potential mechanism of action. MATERIALS AND METHODS: In vivo, we established a rat model of cardiac fibrosis induced by ISO, and administered with low or high dose of HXP (10 mg/kg/day or 30 mg/kg/day). The level of α-SMA was detected by immunohistochemistry examination, and combined with RNA-sequencing analysis to determine the protective effect of HXP on myocardial fibrosis rats. In vitro, by culturing primary rat CFs, we examined the effects of HXP on the proliferation and transdifferentiation of CFs using CCK8, scratch wound healing and immunofluorescence assays. Western blot was used to determine protein expression. RESULTS: The findings revealed that HXP protects against ISO-induced cardiac fibrosis and CFs transdifferentiation in rats. RNA-sequencing and pathway analyses demonstrated 238 or 295 differentially expressed genes (DEGs) and multiple enriched signal pathways, including transforming growth factor-beta (TGF-ß) receptor signaling activates Smads, downregulation of TGF-ß receptor signaling, signaling by TGF-ß receptor complex, and collagen formation under treatment with low or high-dose of HXP. Moreover, HXP also markedly inhibited ISO-induced primary rat CFs proliferation, transdifferentiation, collagen synthesis and the upregulation of TGF-ß1 and phosphorylated Smad2/3 protein expression. CONCLUSION: HXP suppresses ISO-induced CFs transdifferentiation and collagen synthesis, and it may exert these effects in part by inhibiting the activation of the TGF-ß/Smads pathway. This may be a new therapeutic tool for cardiac fibrosis.


Subject(s)
Cardiotonic Agents/pharmacology , Cell Transdifferentiation/drug effects , Collagen/metabolism , Drugs, Chinese Herbal/pharmacology , Fibroblasts/drug effects , Smad Proteins/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Cardiotonic Agents/chemistry , Cardiotonic Agents/therapeutic use , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Fibroblasts/cytology , Fibroblasts/metabolism , Fibrosis/drug therapy , Fibrosis/metabolism , Heart/drug effects , Isoproterenol/toxicity , Male , Myofibroblasts/drug effects , Primary Cell Culture , Rats, Wistar , Signal Transduction/drug effects , Smad Proteins/metabolism , Tablets , Transcriptome/drug effects , Transforming Growth Factor beta/metabolism
7.
Eur J Pharmacol ; 900: 174013, 2021 Jun 05.
Article in English | MEDLINE | ID: mdl-33766620

ABSTRACT

RATIONALE: Higenamine (HG), is one of the main active components in many widely used Chinese herbs, and a common ingredient of health products in Europe and North America. Several groups, including our own, have previously shown the beneficial effects of HG against cardiomyocyte death during acute ischemic damage. However, the effect of HG on chronic cardiac remodeling, such as cardiac fibrosis, remains unknown. OBJECTIVE: Herein, we aim to investigate the role of HG in cardiac fibrosis in vivo as well as its cellular and molecular mechanisms. METHODS AND RESULTS: Chronic pressure overload with transverse aortic constriction (TAC) significantly increased cardiac hypertrophy, fibrosis, and cardiac dysfunction in mice, which were significantly attenuated by HG. Consistently, cardiac fibrosis induced by the chronic infusion of isoproterenol (ISO), was also significantly reduced by HG. Interestingly, our results showed that HG had no effect on adult mouse CM hypertrophy in vitro. However, HG suppressed the activation of cardiac fibroblasts (CFs) in vitro. Furthermore, TGF-ß1-induced expression of ACTA2, a marker of fibroblast activation, was significantly suppressed by HG. Concomitantly, HG inhibited TGF-ß1-induced phosphorylation of Smad2/3 in CFs. HG also reduced the expression of extracellular matrix molecules such as collagen I and collagen III. To our surprise, the inhibitory effect of HG on CFs activation was independent of the activation of the beta2 adrenergic receptor (ß2-AR) that is known to mediate the effect of HG on antagonizing CMs apoptosis. CONCLUSION: Our findings suggest that HG ameliorates pathological cardiac fibrosis and dysfunction at least partially by suppressing TGF-ß1/Smad signaling and CFs activation.


Subject(s)
Alkaloids/pharmacology , Fibrinolytic Agents/pharmacology , Fibroblasts/drug effects , Signal Transduction/drug effects , Smad Proteins/antagonists & inhibitors , Tetrahydroisoquinolines/pharmacology , Transforming Growth Factor beta1/antagonists & inhibitors , Actins/antagonists & inhibitors , Adrenergic beta-Agonists , Animals , Aorta/drug effects , Apoptosis/drug effects , Cardiomegaly/chemically induced , Cardiomegaly/prevention & control , Fibrosis/prevention & control , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Hypertension/chemically induced , Hypertension/drug therapy , Isoproterenol , Mice , Rats , Rats, Sprague-Dawley
8.
Drug Des Devel Ther ; 15: 871-887, 2021.
Article in English | MEDLINE | ID: mdl-33664566

ABSTRACT

PURPOSE: Taxifolin is a kind of dihydroflavone and is usually used as a food additive and health food for its antioxidant, anti-inflammatory, and anti-tumor activities. The purpose of this research is to probe into the hepatoprotective activity and the molecular mechanism of taxifolin. MATERIALS AND METHODS: The liver fibrosis model was established by intraperitoneal injection of 5 mL/kg body weight of CCl4 (20% CCl4 peanut oil solution), and taxifolin was dissolved with 0.9% physiological saline and administered intragastrically to mice. RESULTS: The results indicated that CCl4-induced significantly increased the serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) in mice. Histopathological examination showed severe hepatocyte necrosis and hepatic tissue lesion. Immunohistochemical staining and rt-PCR analysis demonstrated that the expressions of inducible nitric oxide synthetase (iNOS), cyclooxygenase-2 (COX-2), IL-1ß, IL-6, and TNF-α were increased. These changes were significantly reversed when treated with taxifolin. In addition, TUNEL staining and Bcl-2/Bax pathway confirmed that taxifolin significantly inhibited hepatocyte apoptosis. Besides, the research confirmed that taxifolin also inhibited the activation of hepatic stellate cells and the production of extracellular matrix (ECM) by regulating PI3K/AKT/mTOR and TGF-ß1/Smads pathways. CONCLUSION: Taxifolin inhibited inflammation, and attenuated CCl4-induced oxidative stress and cell apoptosis by regulating PI3K/AKT/mTOR and TGF-ß1/Smads pathways, which might in part contributed to taxifolin anti-hepatic fibrosis, further demonstrating that taxifolin may be an efficient hepatoprotective agent.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carbon Tetrachloride/antagonists & inhibitors , Larix/chemistry , Liver Cirrhosis/drug therapy , Quercetin/analogs & derivatives , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Apoptosis/drug effects , Dose-Response Relationship, Drug , Hepatocytes/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred ICR , Molecular Conformation , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Plant Roots/chemistry , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Quercetin/chemistry , Quercetin/isolation & purification , Quercetin/pharmacology , Signal Transduction/drug effects , Smad Proteins/antagonists & inhibitors , Smad Proteins/metabolism , Structure-Activity Relationship , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism
9.
Life Sci ; 271: 119197, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33577847

ABSTRACT

AIMS: To investigate the improvement and mechanisms of silymarin on renal injury in mouse podocytes and streptozotocin (STZ)-induced diabetic nephropathy model (DN) rats. MAIN METHODS: Firstly, the effects of silymarin on the cell viability and cellular injury-related indicators of high-glucose incubated mouse podocytes MPC-5 were assessed by CCK-8 and western blotting (WB) methods, respectively. The STZ-induced diabetic rats with DN were treated with silymarin nanoliposomes at three doses for consecutive 8-week. General metabolic indicators, renal functions and lipid accumulation-related factors were all measured. The renal tissue sections were stained and observed via hematoxylin-eosin (H&E) staining method. Real-time RT-PCR and WB methods were utilized to measure the expression of JAK2/STAT3/SOCS1 and TGF-ß/Smad signaling pathway related factors. KEY FINDINGS: Silymarin significantly improve the high-glucose induced up-regulation of podoxin and nephrin, as well as the expression of inflammatory cytokines IL-6, ICAM-1 and TNF-α, and the cell survival rates were also significantly increased in a dose-dependent manner. Significant improvement on body weight/kidney ratio, renal functions and lipid profiles in renal tissues were observed in STZ-induced diabetic rats after chronic silymarin treatment. The H&E staining exhibited that the pathological damages in renal tissues were obviously improved. Moreover, silymarin nanoliposomes treatment notably suppressed expression levels of inflammation-related proteins as well as IL-6 and ICAM-1, and regulated JAK2/STAT3/SOCS1 and TGF-ß/Smad signaling pathway, thereby exhibited protective effects on kidney of DN model rats. SIGNIFICANCE: Silymarin nanoliposomes ameliorate STZ-induced kidney injury by improving oxidative stress, renal fibrosis, and co-inhibiting JAK2/STAT3/SOCS1 and TGF-ß/Smad signaling pathways in diabetic rats.


Subject(s)
Diabetic Nephropathies/drug therapy , Janus Kinase 2/antagonists & inhibitors , Nanoparticles/administration & dosage , STAT3 Transcription Factor/antagonists & inhibitors , Silymarin/administration & dosage , Suppressor of Cytokine Signaling 1 Protein/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Antioxidants/administration & dosage , Cell Line , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Dose-Response Relationship, Drug , Female , Janus Kinase 2/metabolism , Liposomes , Male , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Smad Proteins/antagonists & inhibitors , Smad Proteins/metabolism , Suppressor of Cytokine Signaling 1 Protein/metabolism , Transforming Growth Factor beta/metabolism
10.
J Ethnopharmacol ; 270: 113838, 2021 Apr 24.
Article in English | MEDLINE | ID: mdl-33460756

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Myocardial fibrosis after myocardial infarction (MI) leads to cardiac remodeling and loss of function. Taohong siwu decoction (THSWD), a well-known traditional Chinese medicinal prescription, has been clinically used to treat various cardiovascular and cerebrovascular diseases, but its potential functions in myocardial fibrosis after MI remain uncharacterized. AIM OF THE STUDY: The purpose of current study was to explore the potential mechanism action and anti-myocardial fibrosis effects of treatment with THSWD in vivo and in vitro. MATERIALS AND METHODS: Mouse underwent ligation of coronary artery to induce MI and divided equally into the sham group, model group and THSWD treatment groups. After 4 weeks, the effects of THSWD treatment on cardiac function were estimated by echocardiography. HE staining was used to detect the pathologic changes and Masson trichrome staining was used to estimate tissue fibrosis. To further explore the regulatory molecular mechanisms of THSWD, transcriptome analysis was performed. Furthermore, in vitro, we investigated the effect of THSWD on cell proliferation and collagen deposition in primary cardiac fibrosis cells and its possible mechanism of action. Overexpression of TGFBR1 was achieved by infection with an adenovirus vector encoding TGFBR1. RESULTS: Treatment with THSWD significantly decreased myocardial fibrosis and recovered cardiac function in the post-MI mouse. The transcriptomics data imply that the TGF-ß pathway might be a target in the anti-fibrosis effect of THSWD. THSWD inhibits TGF-ß1-induced proliferation of primary cardiac fibroblasts. THSWD decreased collagen expression and TGFBR1 and Smad2/3 phosphorylation. Moreover, the inhibitory effect of THSWD on CFs proliferation and collagen deposition, as well as TGFBR1 signaling pathway-associated proteins expression was partially abrogated by overexpression of TGFBR1. CONCLUSION: Collectively, the results implicate that THSWD attenuates myocardial fibrosis by inhibiting fibrosis proliferation and collagen deposition via inhibiting TGFBR1, and might be a potential therapeutic agent for treatment of myocardial fibrosis post-MI.


Subject(s)
Collagen/metabolism , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Fibrosis/drug therapy , Receptor, Transforming Growth Factor-beta Type I/metabolism , Signal Transduction/drug effects , Animals , Cell Proliferation/drug effects , Collagen/antagonists & inhibitors , Disease Models, Animal , Drugs, Chinese Herbal/chemistry , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Male , Mice, Inbred C57BL , Myocardial Infarction/complications , Myocardial Infarction/diagnostic imaging , Myocardium/metabolism , Myocardium/pathology , Primary Cell Culture , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/genetics , Smad Proteins/antagonists & inhibitors , Smad Proteins/metabolism , Transcriptome/drug effects
11.
Pediatr Res ; 89(3): 502-509, 2021 02.
Article in English | MEDLINE | ID: mdl-32365352

ABSTRACT

BACKGROUND: Bronchopulmonary dysplasia remains one of the most common complications of prematurity, despite significant improvements in perinatal care. Functional modeling of human lung development and disease, like BPD, is limited by our ability to access the lung and to maintain relevant progenitor cell populations in culture. METHODS: We supplemented Rho/SMAD signaling inhibition with mTOR inhibition to generate epithelial basal cell-like cell lines from tracheal aspirates of neonates. RESULTS: Single-cell RNA-sequencing confirmed the presence of epithelial cells in tracheal aspirates obtained from intubated neonates. Using Rho/SMAD/mTOR triple signaling inhibition, neonatal tracheal aspirate-derived (nTAD) basal cell-like cells can be expanded long term and retain the ability to differentiate into pseudostratified airway epithelium. CONCLUSIONS: Our data demonstrate that neonatal tracheal aspirate-derived epithelial cells can provide a novel ex vivo human cellular model to study neonatal lung development and disease. IMPACT: Airway epithelial basal cell-like cell lines were derived from human neonatal tracheal aspirates. mTOR inhibition significantly extends in vitro proliferation of neonatal tracheal aspirate-derived basal cell-like cells (nTAD BCCs). nTAD BCCs can be differentiated into functional airway epithelium. nTAD BCCs provide a novel model to investigate perinatal lung development and diseases.


Subject(s)
Epithelial Cells/drug effects , Smad Proteins/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Trachea/cytology , rho-Associated Kinases/antagonists & inhibitors , Base Sequence , Body Fluids/cytology , Bronchopulmonary Dysplasia , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Epithelial Cells/chemistry , Epithelial Cells/cytology , Humans , Infant, Newborn , Primary Cell Culture , Single-Cell Analysis , Sirolimus/pharmacology , Smad Proteins/physiology , Stem Cells/cytology , Stem Cells/drug effects , Suction , TOR Serine-Threonine Kinases/physiology , rho-Associated Kinases/physiology
12.
Biochem Biophys Res Commun ; 534: 279-285, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33288199

ABSTRACT

Emodin, a major component of Chinese herbal rhubarb, delays the progression of chronic renal failure. However, the effect and working mechanisms of Emodin on renal tubulointerstitial fibrosis remains elusive. We hypothesized that emodin inhibits renal tubulointerstitial fibrosis through EZH2, a histone methyltransferase. Our in vivo and in vitro studies demonstrate that emodin reduced extracellular collagen deposition and inhibited Smad3 and CTGF pro-fibrotic signaling pathways, which were correlated with the down-regulation of EZH2 and reduced trimethylation of histone H3 on lysine 27 (H3k27me3) in NRK-49F fibrotic cells and UUO kidneys. Inhibition of EZH2 by 3-DZNeP blocked or attenuated the anti-fibrotic effect of emodin in UUO kidneys and NRK-49F cells. These data indicate that emodin inhibits renal tubulointerstitial fibrosis in obstructed kidneys and this effect is mediated through EZH2.


Subject(s)
Emodin/pharmacology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Renal Insufficiency, Chronic/drug therapy , Animals , Connective Tissue Growth Factor/antagonists & inhibitors , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Enzyme Inhibitors/pharmacology , Fibrosis , In Vitro Techniques , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Signal Transduction/drug effects , Smad Proteins/antagonists & inhibitors , Ureteral Obstruction/drug therapy , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
13.
J Ethnopharmacol ; 262: 113275, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32810620

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Forsythiae Fructuse water extract (FSE) is a water-soluble component extracted from the traditional Chinese medicine Forsythiae Fructuse (The fruit of Forsythia suspensa (Thunb.) Vahl) usually used to treat inflammatory diseases. However, little is known about the therapeutic effect of FSE on liver fibrosis. AIM OF THE STUDY: The purpose of our study was to investigate the therapeutic effect of FSE on liver fibrosis and reveal the underlying mechanism. MATERIALS AND METHODS: Liver fibrosis model was established by subcutaneous injection of olive oil containing 40% CCl4. Rat liver tissue morphologic pathology was investigated by using HE staining, Masson staining and Sirius red staining. Several biochemical markers including liver (ALT, AST, AKP, γ-GT), fibrosis (HA, LN, PC III, Col IV) and inflammation (IL-6, IL-1ß, TNF-α) were determined by using Elisa kits. Immunohistochemistry was used to observe the distribution of α-SMA and COL1 in liver tissue. Effects of FSE on inflammatory pathway (TLR4/MyD88/NF-κB) and fibrotic pathway (TGF-ß/smads) were detected by western blot and qPCR. RESULTS: The results showed that hepatic histopathological injury, abnormal liver function, fibrosis and inflammation induced by CCl4 were improved by FSE (2.5, 5 g/kg). Immunohistochemistry and western blot results indicated that the expression of α-SMA and COL1 in liver tissue was inhibited by FSE (2.5, 5 g/kg). Western blot and qPCR results further proved that FSE (2.5, 5 g/kg) inhibited the transduction of TLR4/MyD88/NF-κB and TGF-ß/smads signaling pathways. CONCLUSION: FSE can inhibit the expression of inflammatory factors and fibrotic cytokines, reduce liver injury, and inhibit the development of liver fibrosis through TLR4/MyD88/NF-κB and TGF-ß/smads signaling pathways.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Forsythia , Liver Cirrhosis/drug therapy , Myeloid Differentiation Factor 88/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Drugs, Chinese Herbal/isolation & purification , Drugs, Chinese Herbal/pharmacology , Female , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Smad Proteins/antagonists & inhibitors , Smad Proteins/metabolism , Toll-Like Receptor 4/metabolism , Transforming Growth Factor beta/metabolism , Water
14.
Sci Rep ; 10(1): 11828, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32678240

ABSTRACT

Glaucoma is a group of progressive optic neuropathies that share common biological and clinical characteristics including irreversible changes to the optic nerve and visual field loss caused by the death of retinal ganglion cells (RGCs). The loss of RGCs manifests as characteristic cupping or optic nerve degeneration, resulting in visual field loss in patients with Glaucoma. Published studies on in vitro RGC differentiation from stem cells utilized classical RGC signaling pathways mimicking retinal development in vivo. Although many strategies allowed for the generation of RGCs, increased variability between experiments and lower yield hampered the cross comparison between individual lines and between experiments. To address this critical need, we developed a reproducible chemically defined in vitro methodology for generating retinal progenitor cell (RPC) populations from iPSCs, that are efficiently directed towards RGC lineage. Using this method, we reproducibly differentiated iPSCs into RGCs with greater than 80% purity, without any genetic modifications. We used small molecules and peptide modulators to inhibit BMP, TGF-ß (SMAD), and canonical Wnt pathways that reduced variability between iPSC lines and yielded functional and mature iPSC-RGCs. Using CD90.2 antibody and Magnetic Activated Cell Sorter (MACS) technique, we successfully purified Thy-1 positive RGCs with nearly 95% purity.


Subject(s)
Cell Differentiation/drug effects , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Smad Proteins/antagonists & inhibitors , Wnt Proteins/antagonists & inhibitors , Computational Biology , Gene Expression Profiling , Humans , Immunohistochemistry , Immunophenotyping , Neurogenesis , Retina/cytology , Signal Transduction
15.
Drug Des Devel Ther ; 14: 2393-2403, 2020.
Article in English | MEDLINE | ID: mdl-32606606

ABSTRACT

INTRODUCTION: Liver disease is common and often life-threatening. Sinomenine (SIN) is an active ingredient extracted from  Sinomenium acutum. This study investigated the protective effect and mechanism of sinomenine (SIN) on acetaminophen (APAP)-induced liver injury from in vitro and in vivo. METHODS: In vivo experiments, mice were randomly divided into six groups (n=10): control group, model group, SIN (25 mg/kg) group, SIN (50 mg/kg) group, SIN (100 mg/kg) group and SIN (100 mg/kg) + SRI-011381 group. Alanine transaminases (ALT), aspartate transaminases (AST) and alkaline phosphatase (ALP) were detected. The pathological lesion was measured by HE staining. Apoptosis was measured by TUNEL staining. In vitro experiments, BRL-3A cells were treated with APAP (7.5 mM) and then subjected to various doses of SIN (10, 50 and 100 µg/mL) at 37°C for 24 h. Inflammatory factors and oxidative stress index were measured by ELISA. The expression of proteins was detected by Western blot. RESULTS: The results showed that compared with the control group, the levels of ALT, AST and ALP in the serum of APAP-induced mice were significantly increased, followed by liver histological damage and hepatocyte apoptosis. Besides, APAP reduced the activity of SOD and GSH-Px, while increasing the content of MDA and LDH. Notably, APAP also promoted the expression of NLRP3, ASC, caspase-1 and IL-1ß. Interestingly, SIN treatment dose-dependently reduced APAP-induced liver injury and oxidative stress, inhibited the activation of NLRP3 inflammasomes, and reduced the levels of inflammatory cytokines. In vitro studies have shown that SIN treatment significantly reduced the viability of BRL-3A cells and oxidative stress and inflammation. In addition, the Western blotting analysis showed that SIN inhibited the activation of TGF-ß/Smad pathway in a dose-dependent manner in vitro and in vivo. These effects were significantly reversed by TGF-ß/Smad activator SRI-011381 or TGF-ß overexpression. DISCUSSION: The study indicates that SIN attenuates APAP-induced acute liver injury by decreasing oxidative stress and inflammatory response via TGF-ß/Smad pathway in vitro and in vivo.


Subject(s)
Acetaminophen/antagonists & inhibitors , Chemical and Drug Induced Liver Injury/drug therapy , Inflammation/drug therapy , Morphinans/pharmacology , Oxidative Stress/drug effects , Smad Proteins/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Dose-Response Relationship, Drug , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Smad Proteins/metabolism , Structure-Activity Relationship , Transforming Growth Factor beta/metabolism
16.
Eur J Gastroenterol Hepatol ; 32(6): 669-677, 2020 06.
Article in English | MEDLINE | ID: mdl-32282548

ABSTRACT

In the 'treat-to-target' era of inflammatory bowel disease (IBD) management, small molecule drugs (SMDs) represent a promising alternative to biomolecular drugs. Moreover, increasing failure rates of anti-tumor necrosis factor α agents have contributed to the development of new molecules with different mechanisms of action and bioavailability. This review focuses on the positioning of new, orally targeted therapies in the treatment algorithm of both Crohn's disease (CD) and ulcerative colitis (UC), with special consideration to their efficacy and safety. We performed a comprehensive search of PubMed and clinical trial registries to identify randomized controlled trials assessing SMDs in adult patients with moderate-to-severe IBD, irrespective of previous exposure to other biologics. In this review, we included 15 double-blind, placebo-controlled trials that assessed the efficacy and safety of Janus kinase inhibitors, sphingosine-1-phosphate modulators (S1P), SMAD blockers, phosphodiesterase 4 inhibitors and α-4 antagonists. The primary endpoints in UC were achieved for tofacitinib in the phase III OCTAVE study and AJM-300, with a favorable safety profile. S1P receptor agonists, such as etrasimod and ozanimod, demonstrated favorable results in induction studies. For CD, filgotinib and upadacitinib also met the primary outcome criteria. Available data have demonstrated so far that SMDs have an advantageous safety and efficacy profile. However, their use in a clinical setting will eventually require a personalized, mechanism-based therapeutic approach.


Subject(s)
Inflammatory Bowel Diseases/drug therapy , Small Molecule Libraries/therapeutic use , Humans , Integrin alpha4/antagonists & inhibitors , Janus Kinase Inhibitors/therapeutic use , Phosphodiesterase 4 Inhibitors/therapeutic use , Smad Proteins/antagonists & inhibitors , Sphingosine 1 Phosphate Receptor Modulators/therapeutic use
17.
FASEB J ; 34(4): 5282-5298, 2020 04.
Article in English | MEDLINE | ID: mdl-32067273

ABSTRACT

Melatonin is a hormone produced by the pineal gland, and it has extensive beneficial effects on various tissue and organs; however, whether melatonin has any effect on cardiac fibrosis in the pathogenesis of diabetic cardiomyopathy (DCM) is still unknown. Herein, we found that melatonin administration significantly ameliorated cardiac dysfunction and reduced collagen production by inhibiting TGF-ß1/Smads signaling and NLRP3 inflammasome activation, as manifested by downregulating the expression of TGF-ß1, p-Smad2, p-Smad3, NLRP3, ASC, cleaved caspase-1, mature IL-1ß, and IL-18 in the heart of melatonin-treated mice with diabetes mellitus (DM). Similar beneficial effects of melatonin were consistently observed in high glucose (HG)-treated cardiac fibroblasts (CFs). Moreover, we also found that lncRNA MALAT1 (lncR-MALAT1) was increased along with concomitant decrease in microRNA-141 (miR-141) in DM mice and HG-treated CFs. Furthermore, we established NLRP3 and TGF-ß1 as target genes of miR-141 and lncR-MALAT1 as an endogenous sponge or ceRNA to limit the functional availability of miR-141. Finally, we observed that knockdown of miR-141 abrogated anti-fibrosis action of melatonin in HG-treated CFs. Our findings indicate that melatonin produces an antifibrotic effect via inhibiting lncR-MALAT1/miR-141-mediated NLRP3 inflammasome activation and TGF-ß1/Smads signaling, and it might be considered a potential agent for the treatment of DCM.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Cardiomyopathies/pathology , Fibrosis/drug therapy , Gene Expression Regulation/drug effects , Heart Diseases/drug therapy , Melatonin/pharmacology , Animals , Antioxidants/pharmacology , Diabetic Cardiomyopathies/epidemiology , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Heart Diseases/etiology , Heart Diseases/metabolism , Heart Diseases/pathology , Inflammasomes/antagonists & inhibitors , Inflammasomes/genetics , Inflammasomes/metabolism , Male , Mice , MicroRNAs/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , RNA, Long Noncoding/genetics , Smad Proteins/antagonists & inhibitors , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
19.
J Cell Physiol ; 235(1): 429-441, 2020 01.
Article in English | MEDLINE | ID: mdl-31230364

ABSTRACT

Colorectal cancer (CRC) is still a fatal health problem around the world. The underlying mechanisms of CRC have not been fully elucidated. N-myc interactor (NMI) acts as an oncogene or a tumor-suppressor gene in several kinds of cancers but CRC. Here, the expression of NMI was found higher in CRC tissues and cells. Higher expression of NMI indicated the poorer prognosis of CRC patients. Moreover, the proliferation of CRC cells was suppressed significantly after we silenced the expression of NMI, while overexpression of NMI promoted CRC cell proliferation. Flow cytometry demonstrated that NMI promoted cell proliferation through facilitating cell transition from the G1 phase to the S phase. Furthermore, it was found that NMI suppressed the phosphorylation of Smad3 by upregulating the expression of STAT1. The effect of NMI depletion on cell proliferation could be reversed by using Smad3 inhibitor SIS3. In summary, our findings demonstrated that NMI promoted cell proliferation via TGFß/Smad pathway and could indicate the prognosis of patients with CRC.


Subject(s)
Colorectal Neoplasms/pathology , Intracellular Signaling Peptides and Proteins/metabolism , STAT1 Transcription Factor/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Isoquinolines/pharmacology , Prognosis , Pyridines/pharmacology , Pyrroles/pharmacology , RNA Interference , RNA, Small Interfering/genetics , STAT1 Transcription Factor/genetics , Smad Proteins/antagonists & inhibitors , Transcriptional Activation/genetics
20.
Acta Pharmacol Sin ; 41(5): 670-677, 2020 May.
Article in English | MEDLINE | ID: mdl-31804606

ABSTRACT

Renal fibrosis is considered as the pathway of almost all kinds of chronic kidney diseases (CKD) to the end stage of renal diseases (ESRD). Ganoderic acid (GA) is a group of lanostane triterpenes isolated from Ganoderma lucidum, which has shown a variety of pharmacological activities. In this study we investigated whether GA exerted antirenal fibrosis effect in a unilateral ureteral obstruction (UUO) mouse model. After UUO surgery, the mice were treated with GA (3.125, 12.5, and 50 mg· kg-1 ·d-1, ip) for 7 or 14 days. Then the mice were sacrificed for collecting blood and kidneys. We showed that GA treatment dose-dependently attenuated UUO-induced tubular injury and renal fibrosis; GA (50 mg· kg-1 ·d-1) significantly ameliorated renal disfunction during fibrosis progression. We further revealed that GA treatment inhibited the extracellular matrix (ECM) deposition in the kidney by suppressing the expression of fibronectin, mainly through hindering the over activation of TGF-ß/Smad signaling. On the other hand, GA treatment significantly decreased the expression of mesenchymal cell markers alpha-smooth muscle actin (α-SMA) and vimentin, and upregulated E-cadherin expression in the kidney, suggesting the suppression of tubular epithelial-mesenchymal transition (EMT) partially via inhibiting both TGF-ß/Smad and MAPK (ERK, JNK, p38) signaling pathways. The inhibitory effects of GA on TGF-ß/Smad and MAPK signaling pathways were confirmed in TGF-ß1-stimulated HK-2 cell model. GA-A, a GA monomer, was identified as a potent inhibitor on renal fibrosis in vitro. These data demonstrate that GA or GA-A might be developed as a potential therapeutic agent in the treatment of renal fibrosis.


Subject(s)
Smad Proteins/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Triterpenes/pharmacology , Ureteral Obstruction/drug therapy , Animals , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Injections, Intraperitoneal , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Triterpenes/administration & dosage , Ureteral Obstruction/metabolism , Ureteral Obstruction/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...