Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 327
Filter
Add more filters

Publication year range
1.
Phytother Res ; 38(3): 1367-1380, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38217097

ABSTRACT

Liver fibrosis affects approximately 800 million patients worldwide, with over 2 million deaths each year. Nevertheless, there are no approved medications for treating liver fibrosis. In this study, we investigated the impacts of ginkgetin on liver fibrosis and the underlying mechanisms. The impacts of ginkgetin on liver fibrosis were assessed in mouse models induced by thioacetamide or bile duct ligation. Experiments on human LX-2 cells and primary mouse hepatic stellate cells (HSCs) were performed to explore the underlying mechanisms, which were also validated in the mouse models. Ginkgetin significantly decreased hepatic extracellular matrix deposition and HSC activation in the fibrotic models induced by thioacetamide (TAA) and bile duct ligation (BDL). Beneficial effects also existed in inhibiting hepatic inflammation and improving liver function. In vitro experiments showed that ginkgetin markedly inhibited HSC viability and induced HSC apoptosis dose-dependently. Mechanistic studies revealed that the antifibrotic effects of ginkgetin depend on STAT1 activation, as the effects were abolished in vitro after STAT1 silencing and in vivo after inhibiting STAT1 activation by fludarabine. Moreover, we observed a meaningful cross-talk between HSCs and hepatocytes, in which IL-6, released by ginkgetin-induced apoptotic HSCs, enhanced hepatocyte proliferation by activating STAT3 signaling. Ginkgetin exhibits antifibrotic effects by inducing HSC apoptosis via STAT1 activation and enhances hepatocyte proliferation secondary to HSC apoptosis via the IL-6/STAT3 pathway.


Subject(s)
Biflavonoids , Hepatic Stellate Cells , Thioacetamide , Mice , Animals , Humans , Thioacetamide/metabolism , Thioacetamide/pharmacology , Thioacetamide/therapeutic use , Interleukin-6/metabolism , Liver Cirrhosis/drug therapy , Disease Models, Animal , Apoptosis , Liver/metabolism , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/pharmacology
2.
Calcif Tissue Int ; 112(6): 704-715, 2023 06.
Article in English | MEDLINE | ID: mdl-37032340

ABSTRACT

Osteoporosis, an age-related metabolic bone disease, is mainly caused by an imbalance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption. At present, there are many osteoporosis drugs that can promote bone formation or inhibit bone resorption. However, there were few therapeutic drugs that can simultaneously promote bone formation and inhibit bone resorption. Oridonin (ORI), a tetracyclic diterpenoid compound isolated from Rabdosia rubescens, has been proved to have anti-inflammatory, anti-tumor effects. However, little is known about the osteoprotective effect of oridonin. Thioacetamide (TAA) is a common organic compound with significant hepatotoxicity. Recent studies have found that there was a certain association between TAA and bone injury. In this work, we investigated the effect and mechanism of ORI on TAA-induced osteoclastogenesis and inhibition of osteoblast differentiation. The results showed that TAA could promote the osteoclastogenesis of RAW264.7 by promoting the MAPK/NF-κB pathway, and also promoted p65 nuclear translocation and activated intracellular ROS generation, and ORI can inhibit these effects to inhibit TAA-induced osteoclastogenesis. Moreover, ORI can also promote the osteogenic differentiation pathway and inhibit adipogenic differentiation of BMSCs to promote bone formation. In conclusion, our results revealed that ORI, as a potential therapeutic drug for osteoporosis, could protect against TAA-induced bone loss and TAA-inhibited bone formation.


Subject(s)
Bone Resorption , Osteoporosis , Humans , Osteogenesis , NF-kappa B/metabolism , Thioacetamide/metabolism , Thioacetamide/pharmacology , Thioacetamide/therapeutic use , Core Binding Factor Alpha 1 Subunit/metabolism , Osteoclasts/metabolism , Bone Resorption/metabolism , Osteoporosis/chemically induced , Osteoporosis/drug therapy , Cell Differentiation , RANK Ligand/metabolism
3.
Int J Mol Sci ; 24(22)2023 Nov 20.
Article in English | MEDLINE | ID: mdl-38003735

ABSTRACT

The insulin resistance caused by impaired glucose metabolism induces ovarian dysfunction due to the central importance of glucose as a source of energy. However, the research on glucose metabolism in the ovaries is still lacking. The objectives of this study were to analyze the effect of PD-MSCs on glucose metabolism through IGFBP2-AMPK signaling and to investigate the correlation between glucose metabolism and ovarian function. Thioacetamide (TAA) was used to construct a rat injury model. PD-MSCs were transplanted into the tail vein (2 × 106) 8 weeks after the experiment started. The expression of the IGFBP2 gene and glucose metabolism factors (e.g., AMPK, GLUT4) was significantly increased in the PD-MSC group compared to the nontransplantation (NTx) group (* p < 0.05). The levels of follicular development markers and the sex hormones AMH, FSH, and E2 were also higher than those in the TAA group. Using ex vivo cocultivation, the mRNA and protein expression of IGFBP2, AMPK, and GLUT4 were significantly increased in the cocultivation with the PD-MSCs group and the recombinant protein-treated group (* p < 0.05). These findings suggest that the increased IGFBP2 levels by PD-MSCs play an important role in glucose metabolism and ovarian function through the IGFBP2-AMPK signaling pathway.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Rats , Animals , Thioacetamide/pharmacology , AMP-Activated Protein Kinases/metabolism , Mesenchymal Stem Cells/metabolism , Signal Transduction , Glucose/metabolism
4.
Molecules ; 28(22)2023 Nov 15.
Article in English | MEDLINE | ID: mdl-38005330

ABSTRACT

The protective effect of biochanin A (BCA) on the histopathology, immunohistochemistry, and biochemistry of thioacetamide (TAA)-induced liver cirrhosis in vivo was investigated. There was a significant reduction in liver weight and hepatocyte propagation, with much lower cell injury in rat groups treated with BCA (25 mg/kg and 50 mg/kg) following a TAA induction. These groups had significantly lower levels of proliferating cell nuclear antigen (PCNA) and α-smooth muscle actin (α-SMA). The liver homogenates showed increased antioxidant enzyme activity of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx), as well as decreased malondialdehyde (MDA) levels. The serum biomarkers associated with liver function, namely alkaline phosphatase (ALP), alanine aminotransferase (ALT), aspartate aminotransferase (AST), and gamma glutamyl transaminase (GGT), returned to normal levels, comparable to those observed in both the normal control group and the reference control group. Taken together, the normal microanatomy of hepatocytes, the inhibition of PCNA and α-SMA, improved antioxidant enzymes (SOD, CAT, and GPx), and condensed MDA with repairs of liver biomarkers validated BCA's hepatoprotective effect.


Subject(s)
Antioxidants , Chemical and Drug Induced Liver Injury , Rats , Animals , Antioxidants/pharmacology , Thioacetamide/pharmacology , Proliferating Cell Nuclear Antigen , Oxidative Stress , Rats, Wistar , Liver , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Alanine Transaminase , Superoxide Dismutase/pharmacology , Chemical and Drug Induced Liver Injury/pathology , Aspartate Aminotransferases
5.
Metab Brain Dis ; 37(5): 1541-1556, 2022 06.
Article in English | MEDLINE | ID: mdl-35298730

ABSTRACT

This study was planned to assess the potential protective effects of taxifolin against thioacetamide-induced hepatic encephalopathy and subsequently to portray its behavioural results. The experimental model was induced with three doses of (200 mg/kg i.p.) thioacetamide and taxifolin (50 and 100 mg/kg, p.o.) was administered for fourteen days. Taxifolin effectively attenuated hepatic encephalopathy through decrease in AST, ALT, ALP and LDH concentrations and improvement of hyperammonemia, and increase in antioxidant capacity by decreasing MDA, ROS, and increasing CAT and GSH. In addition, the expressions of NF-κB, TNF-α, IL-1ß, caspase-3 and Bax was down-regulated while IL-10 and Bcl-2 expressions were up-regulated with taxifolin treatment. The recovery was confirmed by downregulation of iNOS and 8-OHdG expressions in our immunohistochemical analysis. Taxifolin treatment reduced the disrupting role of thioacetamide as seen by corrected hyperammonemia as well as preservation of astrocyte and hepatocyte structure. Elevated plus maze and locomotor activity tests also proved that taxifolin might repeal the neurobehavioral disabilities. In conclusion, taxifolin has shown hepatoprotective and neuroprotective roles with antioxidant and anti-inflammatory effects, as well as suppressing the excessive release of ammonia, and it eventually reversed neurobehavioral impairments.


Subject(s)
Hepatic Encephalopathy , Hyperammonemia , Neuroprotective Agents , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Hepatic Encephalopathy/metabolism , Hyperammonemia/drug therapy , Hyperammonemia/metabolism , Liver/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress , Quercetin/analogs & derivatives , Rats , Rats, Wistar , Thioacetamide/pharmacology
6.
Molecules ; 27(14)2022 Jul 07.
Article in English | MEDLINE | ID: mdl-35889233

ABSTRACT

Oxidative stress induced by well-known toxins including ferric nitrilotriacetate (Fe-NTA), carbon tetrachloride (CCl4) and thioacetamide (TAA) has been attributed to causing tissue injury in the liver and kidney. In this study, the effect of glyceryl trinitrate (GTN), a donor of nitric oxide and NG-nitroarginine methyl ester (l-NAME), a nitric oxide inhibitor on TAA-induced hepatic oxidative stress, GSH and GSH-dependent enzymes, serum transaminases and tumor promotion markers such as ornithine decarboxylase (ODC) activity and [3H]-thymidine incorporation in rats were examined. The animals were divided into seven groups consisting of six healthy rats per group. The six rats were injected intraperitoneally with TAA to evaluate its toxic effect, improvement in its toxic effect if any, or worsening in its toxic effect if any, when given in combination with GTN or l-NAME. The single necrogenic dose of TAA administration caused a significant change in the levels of both hepatic and serum enzymes such as glutathione S-transferase (GST), glutathione reductase (GR), glutathione peroxidase (GPx), γ-glutamyl transpeptidase (GGT), glucose 6-phosphate dehydrogenase (G6PD), alanine aminotransferase (AST) and aspartate aminotransferase (ALT). In addition, treatment with TAA also augmented malondialdehyde (MDA), ornithine decarboxylase (ODC) activity and [3H]-thymidine incorporation in rats liver. Concomitantly, TAA treatment depleted the levels of GSH. However, most of these changes were alleviated by the treatment of animals with GTN dose-dependently. The protective effect of GTN against TAA was also confirmed histopathologically. The present data confirmed our earlier findings with other oxidants including Fe-NTA and CCl4. The GTN showed no change whatsoever when administered alone, however when it was given along with TAA then it showed protection thereby contributing towards defending the role against oxidants-induced organ toxicity. Overall, GTN may contribute to protection against TAA-induced oxidative stress, toxicity, and proliferative response in the liver, according to our findings.


Subject(s)
Nitric Oxide Donors , Nitroglycerin , Animals , Glutathione/metabolism , Liver , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Donors/metabolism , Nitric Oxide Donors/pharmacology , Nitroglycerin/pharmacology , Ornithine Decarboxylase/metabolism , Oxidants/pharmacology , Oxidative Stress , Rats , Rats, Wistar , Thioacetamide/pharmacology , Thymidine/metabolism
7.
Zhongguo Zhong Yao Za Zhi ; 47(21): 5882-5889, 2022 Nov.
Article in Zh | MEDLINE | ID: mdl-36472007

ABSTRACT

This study aims to investigate the therapeutic effect of icariin(ICA) on thioacetamide(TAA)-induced femoral osteolysis in rats. RAW264.7 cells were treated with TAA and ICA. Cell counting kit-8(CCK-8) assay was used to detect cell proliferation, and tartrate-resistant acid phosphatase(TRAP) staining to examine the formation of osteoclasts. The expression of TRAP, cathepsin K, c-FOS, and NFATc1 in RAW264.7 cells was determined by Western blot and immunofluorescence method. Thirty-two SD rats were randomized into the control group, TAA group(intraperitoneal injection of TAA at 300 mg·kg~(-1)), ICA group(gavage of ICA at 600 mg·kg~(-1)) and TAA + ICA group(intraperitoneal injection of TAA at 300 mg·kg~(-1) and gavage of ICA at 600 mg·kg~(-1)). Administration was performed every other day for 6 weeks. Body weight and length of femur were recorded at execution. Pathological injury and osteoclast differentiation of femur were observed based on hematoxylin-eosin(HE) staining and TRAP staining, and the changes of bone metabolism-related indexes alkaline phosphatase(ALP), calcium(Ca), phosphorus(P), magnesium(Mg), and cross-linked N-telopeptide of type Ⅰ collagen(NTX-Ⅰ) in serum were detected. Three-point bending test and micro-CT were applied to evaluate the quality of femur, and Western blot to detect the levels of osteoclast-related proteins TRAP, cathepsin K, RANK, RANKL, p38, p-p38, ERK, p-ERK, JNK, p-JNK, c-Fos, and NFATc1. The results showed ICA could inhibit TAA-induced production of TRAP-positive cells, the expression of osteoclast-related proteins, and nuclear translocation of NFATc1. ICA alleviated the weight loss, reduction of femur length, and growth inhibition induced by TAA in SD rats. ICA ameliorated the decline of femur elastic modulus caused by TAA and significantly restored trabecular bone mineral density(BMD), trabecular pattern factor(Tb.Pf), trabecular number(Tb.N), trabecular thickness(Tb.Th), and structure model index(SMI), thus improving bone structure. Western blot results showed ICA suppressed femoral osteoclast differentiation induced by TAA through RANKL-p38/ERK-NFATc1 signaling pathway. ICA inhibits osteoclast differentiation and prevents TAA-induced osteolysis by down-regulating RANKL-p38/ERK-NFAT signaling pathway.


Subject(s)
Bone Resorption , Osteolysis , Rats , Animals , Osteoclasts , Cathepsin K/genetics , Cathepsin K/metabolism , Cathepsin K/pharmacology , Thioacetamide/metabolism , Thioacetamide/pharmacology , Bone Resorption/metabolism , Bone Resorption/pathology , Osteolysis/metabolism , Osteolysis/pathology , Cell Differentiation , Rats, Sprague-Dawley , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism
8.
Pharmazie ; 76(5): 232-238, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33964998

ABSTRACT

Liver fibrosis is the result of long-term liver injury and has a high incidence worldwide. Protocatechuic acid (PCA) is ubiquitous in vegetables, nuts, brown rice and herbal medicines, which is reported to possess anti-asthmatic, anti-cancer, and anti-oxidation properties. Our research aimed to investigate the effect of PCA on liver fibrosis. In vitro, TNF-α-induced hepatic stellate cell (HSC) model was used to assess the anti-fibrosis effects of PCA. In vivo, mice were treated with thioacetamide (TAA) to develop liver fibrosis. Body weight, organ index, histological changes, and proteins alteration of factors associated with TGF-ß signaling pathway were used to assess the anti-fibrosis effects of PCA. Our results showed that PCA not only inhibited cell viability in TNF-α activated HSC-T6 cells in vitro, but also efficiently mitigated TAA-induced liver damage and fibrosis in vivo. Further experiments indicated that PCA played a protective role in liver fibrosis through regulation of the TGF-ß signaling pathway downregulating the protein expression of p-Smad2, p-ERK, c-Jun. In summary, our findings provide a pharmacological justification for the clinical application of PCA in preventing or treating liver fibrosis.


Subject(s)
Hydroxybenzoates/pharmacology , Liver Cirrhosis/drug therapy , Animals , Cell Line , Hepatic Stellate Cells/drug effects , Inflammation/metabolism , Kidney/drug effects , Liver/drug effects , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Models, Animal , Rats , Signal Transduction/drug effects , Spleen/drug effects , Thioacetamide/pharmacology , Transaminases/blood , Transaminases/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/pharmacology
9.
Cell Biol Int ; 44(12): 2459-2472, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32827326

ABSTRACT

Adult stem cells, such as bone marrow mesenchymal stem cells (BMSCs), are postdevelopmental cells found in many bone tissues. They are capable of multipotent differentiation and have low immune-rejection characteristics. Hepatocytes may become inflamed and produce a large number of free radicals when affected by drugs, poisoning, or a viral infection. The excessive accumulation of free radicals in the extracellular matrix (ECM) eventually leads to liver fibrosis. This study aims to investigate the restorative effects of mouse bone marrow mesenchymal stem cells (mBMSCs) on thioacetamide (TAA)-induced damage in hepatocytes. An in vitro transwell co-culture system of HepG2 cells were co-cultured with mBMSCs. The effects of damage done to TAA-treated HepG2 cells were reflected in the overall cell survival, the expression of antioxidants (SOD1, GPX1, and CAT), the ECM (COL1A1 and MMP9), antiapoptosis characteristics (BCL2), and inflammation (TNF) genes. The majority of the damage done to HepG2 by TAA was significantly reduced when cells were co-cultured with mBMSCs. The signal transducer and activator of transcription 3 (STAT3) and its phosphorylated STAT3 (p-STAT3), as related to cell growth and survival, were detected in this study. The results show that STAT3 was significantly decreased in the TAA-treated HepG2 cells, but the STAT3 and p-STAT3 of HepG2 cells were significantly activated when the TAA-treated HepG2 co-cultured with mBMSCs. Strong expression of interleukin (Il6) messenger RNA in co-cultured mBMSCs/HepG2 indicated mBMSCs secret the cytokines IL-6, which promotes cell survival through downstream STAT3 activation and aid in the recovery of HepG2 cells damaged by TAA.


Subject(s)
Bone Marrow Cells/metabolism , Hepatocytes/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Cell Proliferation , China , Coculture Techniques , Hep G2 Cells , Hepatocytes/drug effects , Humans , Liver Cirrhosis/pathology , Mice , Mice, Mutant Strains , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Thioacetamide/adverse effects , Thioacetamide/pharmacology
10.
Biosci Biotechnol Biochem ; 84(1): 171-177, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31476130

ABSTRACT

We tested the hypothesis that α-lactalbumin inhibits the disruption of intestinal barrier function and liver cirrhosis by restoring gut-liver axis function in thioacetamide (TAA) -treated rats. Rat diets were supplemented with α-lactalbumin replacing 50% of dietary protein. After consuming α-lactalbumin for one week, rats were intraperitoneally injected with TAA twice a week for 14 weeks. The α-lactalbumin-enriched diet significantly inhibited the elevation of plasma alanine aminotransferase, aspartate aminotransferase, and hyaluronic acids. The supplement significantly reduced plasma lipopolysaccharide levels and increased occludin mRNA level. Hepatic fibrosis and regenerative nodules was developed and intestinal villi were shortened by TAA; α-Lactalbumin attenuated these histopathological changes. These results indicated that α-lactalbumin improved intestinal barrier function, suppressing endotoxin levels. These data also suggested that α-lactalbumin ameliorated the impairment of the gut-liver axis by TAA, inhibiting the development of liver cirrhosis.


Subject(s)
Dietary Supplements , Gastrointestinal Tract/drug effects , Lactalbumin/therapeutic use , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/diet therapy , Liver/drug effects , Protective Agents/therapeutic use , Thioacetamide/pharmacology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Fibrosis/drug therapy , Gastrointestinal Tract/metabolism , Gene Expression/drug effects , Hyaluronic Acid/blood , Injections, Intraperitoneal , Lipopolysaccharides/blood , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/prevention & control , Male , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Thioacetamide/administration & dosage , Tight Junction Proteins/genetics
11.
Int J Mol Sci ; 21(23)2020 Nov 24.
Article in English | MEDLINE | ID: mdl-33255464

ABSTRACT

Small biomolecules, such as coenzyme A (CoA) and acetyl coenzyme A (acetyl-CoA), play vital roles in the regulation of cellular energy metabolism. In this paper, we evaluated the delayed effect of the potent hepatotoxin thioacetamide (TAA) on the concentrations of CoA and acetyl-CoA in plasma and in different rat tissues. Administration of TAA negatively affects liver function and leads to the development of hepatic encephalopathy (HE). In our experiments, rats were administered a single intraperitoneal injection of TAA at doses of 200, 400, or 600 mg/kg. Plasma, liver, kidney, and brain samples were collected six days after the TAA administration, a period that has been suggested to allow for restoration of liver function. The concentrations of CoA and acetyl-CoA in the group of rats exposed to different doses of TAA were compared to those observed in healthy rats. The results obtained indicate that even a single administration of TAA to rats is sufficient to alter the physiological balance of CoA and acetyl-CoA in the plasma and tissues of rats for an extended period of time. The initial concentrations of CoA and acetyl-CoA were not restored even after the completion of the liver regeneration process.


Subject(s)
Acetyl Coenzyme A/blood , Coenzyme A/blood , Hepatic Encephalopathy/blood , Thioacetamide/pharmacology , Acetyl Coenzyme A/genetics , Animals , Brain/drug effects , Brain/metabolism , Coenzyme A/genetics , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/pathology , Humans , Injections, Intraperitoneal , Liver/drug effects , Liver/pathology , Liver Regeneration/genetics , Rats , Thioacetamide/toxicity
12.
Pak J Pharm Sci ; 33(4): 1519-1525, 2020 Jul.
Article in English | MEDLINE | ID: mdl-33583782

ABSTRACT

The present research work was designed to evaluate the effects of curcumin supplementation on various biochemical parameters in rats with thioacetamide (TAA) induced liver cirrhosis. For this purpose 24 male Albino Wistar rats were randomly distributed into four groups (n=6).Group I served as control, Group II and Group III received thioacetamide 200mg/kg b.w, i.p, twice a week for 12 weeks in first phase. In second phase Group II received saline and Group III received curcumin 50mg/kg b.w/day, i.p for 12 weeks, in second phase, Group IV received curcumin 50mg/kg b.w/day, i.p, for 12 weeks, in first phase and saline in second phase. Evaluation of histopathological and biochemical parameters was carried out by liver histopathology and estimation of total and direct bilirubin, liver specific enzymes, antioxidant enzymes, MDA level, plasma and intraerythrocyte sodium and potassium respectively. Histopathology of liver showed highest degree of fibrosis and nodule formation, significant alteration in biochemical parameters indicated development of severe liver cirrhosis. Curcumin treatment showed reduced amount of fibrosis and significant reduction in level of liver biomarkers, reversal of antioxidant enzymes (SOD and GSH), MDA level, catalase activity and regain of electrolyte homeostasis. These findings confirm the protective role of curcumin in liver cirrhosis.


Subject(s)
Curcumin/pharmacology , Liver Cirrhosis/drug therapy , Liver/drug effects , Protective Agents/pharmacology , Animals , Antioxidants/metabolism , Biomarkers/metabolism , Homeostasis/drug effects , Liver/metabolism , Liver Cirrhosis/metabolism , Male , Oxidative Stress/drug effects , Rats , Rats, Wistar , Thioacetamide/pharmacology
13.
Toxicol Appl Pharmacol ; 379: 114673, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31323263

ABSTRACT

BACKGROUND AND AIM: Tadalafil displays important neuroprotective effects in experimental models of neurodegenerative diseases, however its mechanisms of action remain poorly understood. The aim of the present study was to investigate the action of Tadalafil on learning and memory, neuroinflammation, glial cell activation and neuroprotection in the experimental model of hepatic encephalopathy (HE) induced by Thioacetamide (TAA) in mice. METHODS: Mice received intraperitoneal injections of TAA, for 3 consecutive days, reaching the final dose of 600 mg/kg. Tadalafil 15 mg/kg body weight was administered by gavage during 15 days after TAA induction. Mice underwent a Barnes maze for learning and memory evaluation. RESULTS: Animals with hepatic encephalopathy showed reduced learning and spatial memory in the Barnes Maze, presented astrocyte and microglia activation and increased neuroinflammatory markers such as TNF-α, IL-1ß, IL-6, p-p38, p-ERK and p-NF-kB. In addition, the signaling pathway PKA/PKG/CREB/BDNF/NeuN/synaptophysin and glutamate receptors were deregulated by TAA. Tadalafil treatment regulated the inflammation signaling pathways restoring learning and spatial memory. CONCLUSION: Tadalafil significantly reduced neuroinflammation, promoted neuroprotection and plasticity, regulated the expression of hippocampal glutamate receptor and restored spatial learning ability and memory.


Subject(s)
Hepatic Encephalopathy/complications , Memory Disorders/drug therapy , Memory, Long-Term/drug effects , Neuronal Plasticity/drug effects , Neuroprotective Agents/therapeutic use , Tadalafil/therapeutic use , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cytokines/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/psychology , Injections, Intraperitoneal , Memory Disorders/etiology , Mice , Microglia/drug effects , Microglia/metabolism , Neuroprotective Agents/administration & dosage , Nitric Oxide/metabolism , Spatial Learning/drug effects , Tadalafil/administration & dosage , Thioacetamide/pharmacology
14.
Bioorg Med Chem Lett ; 29(1): 22-27, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30448234

ABSTRACT

Based on our previous research, three series of new triazolylthioacetamides possessing 3,4,5-trimethoxyphenyl moiety were synthesized, and evaluated for antiproliferative activities and inhibition of tubulin polymerization. The most promising compounds 8b and 8j demonstrated more significant antiproliferative activities against MCF-7, HeLa, and HT-29 cell lines than our lead compound 6. Moreover, analogues 8f, 8j, and 8o manifested more potent antiproliferative activities against HeLa cell line with IC50 values of 0.04, 0.05 and 0.16 µM, respectively, representing 100-, 82-, and 25-fold improvements of the activity compared to compound 6. Furthermore, the representative compound, 8j, was found to induce significant cell cycle arrest at the G2/M phase in HeLa cell lines via a concentration-dependent manner. Meanwhile, compound 8b exhibited the most potent tubulin polymerization inhibitory activity with an IC50 value of 5.9 µM, which was almost as active as that of CA-4 (IC50 = 4.2 µM). Additionally, molecular docking analysis suggested that 8b formed stable interactions in the colchicine-binding site of tubulin.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Thioacetamide/pharmacology , Tubulin Modulators/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HEK293 Cells , Humans , Molecular Docking Simulation , Molecular Structure , Polymerization/drug effects , Structure-Activity Relationship , Thioacetamide/chemical synthesis , Thioacetamide/chemistry , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
15.
Mar Drugs ; 17(6)2019 May 30.
Article in English | MEDLINE | ID: mdl-31151157

ABSTRACT

Thioacetamide (TAA) is known to induce lipid accumulation in the liver. In the present study, we investigated the effects of magma seawater (MS) rich in minerals on hepatic lipid metabolism by evaluating lipogenic enzymes regulated by sterol regulatory element-binding proteins (SREBPs). Rats (n = 10 per group) were intraperitoneally injected with TAA (200 mg/kg bw) thrice a week for seven weeks in combination with a respective experimental diet. Rats in the TAA-treated group received either a chow diet (Control group) or a chow diet containing MS (TMS group, 2.05%) or silymarin (TSM group, 0.05%). Rats in the normal group were injected with PBS as a vehicle and received a chow diet. Rats in the TMS group showed significantly lower hepatic lipid concentrations than rats in the control group (p < 0.05). Hepatic protein expression levels of fatty acid synthase, SREBP-1, 3-hydroxy-3-methylglutaryl-coenzyme A reductase, and SREBP-2 were significantly downregulated in the TMS group, whereas carnitine palmitoyltransferase 1 levels were upregulated (p < 0.05). Hepatic thiobarbituric acid reactive substances levels were lower in the TMS group, whereas protein levels of glutathione peroxidase and catalase were elevated (p < 0.05). The effects of MS were comparable to those of silymarin. Our results evidently showed that MS inhibits hepatic lipid accumulation by suppressing lipid synthesis, accompanied by lipid oxidation and elevation of antioxidative status.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Lipid Metabolism/drug effects , Liver , Minerals/pharmacology , Seawater/chemistry , Sterol Regulatory Element Binding Proteins/metabolism , Thioacetamide/pharmacology , Animals , Diet , Liver/chemistry , Liver/drug effects , Liver/enzymology , Male , Minerals/administration & dosage , Oxidoreductases/genetics , Oxidoreductases/metabolism , Rats , Rats, Sprague-Dawley , Weight Gain/drug effects
16.
Int J Mol Sci ; 20(24)2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31847135

ABSTRACT

Tumor necrosis factor-α (TNF-α)-driven inflammatory reaction plays a crucial role in the initiation of liver fibrosis. We herein attempted to design genetically engineered adipose-derived stem cells (ASCs) producing etanercept (a potent TNF-α inhibitor), and to determine the anti-fibrotic potential of the secretome released from the etanercept-synthesizing ASCs (etanercept-secretome). First, we generated the etanercept-synthesizing ASCs by transfecting the ASCs with mini-circle plasmids containing the gene insert encoding for etanercept. We subsequently collected the secretory material released from the etanercept-synthesizing ASCs and determined its anti-fibrotic effects both in vitro (in thioacetamide [TAA]-treated AML12 and LX2 cells) and in vivo (in TAA-treated mice) models of liver fibrosis. We observed that while etanercept-secretome increased the viability of the TAA-treated AML12 hepatocytes (p = 0.021), it significantly decreased the viability of the TAA-treated LX2 HSCs (p = 0.021). In the liver of mice with liver fibrosis, intravenous administration of the etanercept-secretome induced significant reduction in the expression of both fibrosis-related and inflammation-related markers compared to the control group (all Ps < 0.05). The etanercept-secretome group also showed significantly lower serum levels of liver enzymes as well as pro-inflammatory cytokines, such as TNF-α (p = 0.020) and IL-6 (p = 0.021). Histological examination of the liver showed the highest reduction in the degree of fibrosis in the entanercept-secretome group (p = 0.006). Our results suggest that the administration of etanercept-secretome improves liver fibrosis by inhibiting TNF-α-driven inflammation in the mice with liver fibrosis. Thus, blocking TNF-α-driven inflammation at the appropriate stage of liver fibrosis could be an efficient strategy to prevent fibrosis.


Subject(s)
Adipose Tissue/metabolism , Etanercept/metabolism , Liver Cirrhosis/prevention & control , Stem Cells/metabolism , Adipose Tissue/pathology , Cell Line , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Stem Cells/pathology , Thioacetamide/adverse effects , Thioacetamide/pharmacology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
17.
Molecules ; 24(6)2019 Mar 25.
Article in English | MEDLINE | ID: mdl-30934584

ABSTRACT

Metallo-ß-lactamases (MßLs) are the target enzymes of ß-lactam antibiotic resistance, and there are no effective inhibitors against MßLs available for clinic so far. In this study, thirteen halogen-substituted triazolethioacetamides were designed and synthesized as a potent skeleton of MßLs inhibitors. All the compounds displayed inhibitory activity against ImiS with an IC50 value range of 0.032⁻15.64 µM except 7. The chlorine substituted compounds (1, 2 and 3) inhibited NDM-1 with an IC50 value of less than 0.96 µM, and the fluorine substituted 12 and 13 inhibited VIM-2 with IC50 values of 38.9 and 2.8 µM, respectively. However, none of the triazolethioacetamides exhibited activity against L1 at inhibitor concentrations of up to 1 mM. Enzyme inhibition kinetics revealed that 9 and 13 are mixed inhibitors for ImiS with Ki values of 0.074 and 0.27µM using imipenem as the substrate. Docking studies showed that 1 and 9, which have the highest inhibitory activity against ImiS, fit the binding site of CphA as a replacement of ImiS via stable interactions between the triazole group bridging ASP120 and hydroxyl group bridging ASN233.


Subject(s)
Halogens/chemistry , Thioacetamide/chemistry , Thioacetamide/pharmacology , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/chemistry , Binding Sites , Dose-Response Relationship, Drug , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Binding , Structure-Activity Relationship , Thioacetamide/analogs & derivatives
18.
Bioorg Chem ; 80: 288-295, 2018 10.
Article in English | MEDLINE | ID: mdl-29980114

ABSTRACT

A novel series of acridine linked to thioacetamides 9a-o were synthesized and evaluated for their α-glucosidase inhibitory and cytotoxic activities. All the synthesized compounds exhibited excellent α-glucosidase inhibitory activity in the range of IC50 = 80.0 ±â€¯2.0-383.1 ±â€¯2.0 µM against yeast α-glucosidase, when compared to the standard drug acarbose (IC50 = 750.0 ±â€¯1.5 µM). Among the synthesized compounds, 2-((6-chloro-2-methoxyacridin-9-yl)thio)-N-(p-tolyl) acetamide 9b displayed the highest α-glucosidase inhibitory activity (IC50 = 80.0 ±â€¯2.0 µM). The in vitro cytotoxic assay of compounds 9a-o against MCF-7 cell line revealed that only the compounds 9d, 9c, and 9n exhibited cytotoxic activity. Cytotoxic compounds 9d, 9c, and 9n did not show cytotoxic activity against the normal human cell lines HDF. Kinetic study revealed that the most potent compound 9b is a competitive inhibitor with a Ki of 85 µM. Furthermore, the interaction modes of the most potent compounds 9b and 9f with α-glucosidase were evaluated through the molecular docking studies.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Drug Design , Glycoside Hydrolase Inhibitors/chemical synthesis , Thioacetamide/chemistry , alpha-Glucosidases/metabolism , Acridines/chemistry , Binding Sites , Catalytic Domain , Cell Line , Cell Survival/drug effects , Diabetes Mellitus, Type 2/metabolism , Glycoside Hydrolase Inhibitors/pharmacology , Glycoside Hydrolase Inhibitors/therapeutic use , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Kinetics , Molecular Docking Simulation , Structure-Activity Relationship , Thioacetamide/pharmacology , Thioacetamide/therapeutic use , alpha-Glucosidases/chemistry
19.
Can J Physiol Pharmacol ; 96(12): 1308-1317, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30398909

ABSTRACT

Liver fibrosis is a health concern that leads to organ failure mediated via production of inflammatory cytokines and fibrotic biomarkers. This study aimed to explore the protective effect of tadalafil, a phosphodiesterase-5 inhibitor, against thioacetamide (TAA)-induced liver fibrosis. Fibrosis was induced by administration of TAA (200 mg/kg, i.p.) twice weekly for 6 weeks. Serum transaminases activities, liver inflammatory cytokines, fibrotic biomarkers, and liver histopathology were assessed. TAA induced marked histopathological changes in liver tissues coupled with elevations in serum transaminases activities. Furthermore, hepatic content of nitric oxide and tumor necrosis factor-alpha, interleukin-6, and interleukin-1 beta were elevated, together with a reduction of interleukin-10 in the liver. In addition, TAA increased hepatic contents of transforming growth factor-beta, hydroxyproline, alpha-smooth muscle actin, and gene expression of collagen-1. Pretreatment with tadalafil protected against TAA-induced liver fibrosis, in a dose-dependent manner, as proved by the alleviation of inflammatory and fibrotic biomarkers. The effects of tadalafil were comparable with that of silymarin, a natural antioxidant, and could be assigned to its anti-inflammatory and anti-fibrotic properties.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation/drug therapy , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Tadalafil/pharmacology , Thioacetamide/pharmacology , Actins/metabolism , Animals , Biomarkers/metabolism , Collagen Type I/metabolism , Cytokines/metabolism , Gene Expression/drug effects , Hydroxyproline/metabolism , Inflammation/metabolism , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Liver/drug effects , Liver/metabolism , Liver Cirrhosis/metabolism , Male , Nitric Oxide/metabolism , Rats , Rats, Wistar , Transaminases/metabolism , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
Clin Exp Pharmacol Physiol ; 45(12): 1245-1256, 2018 12.
Article in English | MEDLINE | ID: mdl-30019784

ABSTRACT

Choline deprivation is a recognized experimental approach to nonalcoholic steatohepatitis, while thioacetamide (TAA)-induced liver fibrosis resembles alcoholic liver fibrogenesis. In order to elucidate the effect of TAA on liver extracellular matrix composition under choline deprivation due to choline-deficient diet (CDD) administration, we evaluated the transcriptional and immunohistochemical (IHC) pattern of major hepatic matrix metalloproteinases (namely, MMP-2, -9) and their tissue inhibitors (TIMP-1, -2) in adult male albino Wistar rats at 30, 60 and 90 days. In the CDD+TAA group, IHC showed an early progressive increase in MMP-2 expression, while MMP-9 initially exhibited a significant increase followed by a gradual decrease; TIMP-1 and TIMP-2 IHC expressions showed gradual increase throughout the experiment. The MMPs-TIMPs regulation at the transcriptional level was found to be increased in all groups throughout the experiment. The increased MMP-2/TIMP-2 and suppressed MMP-9/TIMP-1 ratios in IHC and in real-time polymerase chain reaction (RT-PCR) seemed to correlate with the degree of liver fibrosis. These results support the important role of MMPs and TIMPs in controlling the hepatic pathogenesis and shed more light on the recently described experimental approach to liver disease (steatohepatitis) under the impact of two insults (TAA and CDD).


Subject(s)
Choline/analysis , Diet , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Liver/cytology , Thioacetamide/pharmacology , Animals , Gene Expression Regulation, Enzymologic/drug effects , Liver/drug effects , Male , Matrix Metalloproteinases/genetics , Rats , Rats, Wistar , Thioacetamide/administration & dosage , Tissue Inhibitor of Metalloproteinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL