Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Hum Exp Toxicol ; 40(12_suppl): S573-S582, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34802289

ABSTRACT

Inflammation and oxidative stress are the major pathways involved in ischemia-reperfusion (I/R)-induced renal injury. This study was designed to evaluate the potential effect of pomegranate against I/R-induced renal injury. I/R injury was induced in nephrectomized rats by unilateral occlusion of the left renal pedicle for 45 min followed by 24 h of perfusion. Pomegranate succeeded to decrease serum levels of creatinine, potassium, and urea nitrogen, along with increasing creatinine clearance. Pomegranate also decreased I/R-induced changes in histopathological examination. Pomegranate attenuated the renal inflammatory response reflected by the suppression of nuclear factor κB p65 DNA binding activity, the upregulation of inhibitory protein kappa B-alpha mRNA expression, the downregulation of mRNA and protein expression of tumor necrosis factor α, in addition to the reduced myeloperoxidase activity and mRNA expression. Additionally, pomegranate attenuated oxidative stress likely through the modulation of lipid peroxidation and antioxidant levels reflected by the decreased MDA content and the increased glutathione level and superoxide dismutase activity. Results confirm the potential protective effect of pomegranate against I/R-induced renal injury through its anti-inflammatory and anti-oxidant effects mediated through the upregulation of inhibitory protein kappa B-alpha, the inhibition of NF-κB activity, and the associated TNF-α release, neutrophil infiltration, and oxidative stress.


Subject(s)
Kidney/blood supply , NF-kappa B/metabolism , Plant Extracts/pharmacology , Pomegranate/chemistry , Reperfusion Injury/prevention & control , Animals , Rats
2.
J Biochem Mol Toxicol ; 35(5): e22745, 2021 May.
Article in English | MEDLINE | ID: mdl-33749060

ABSTRACT

Liver fibrosis is a public health burden that is highly associated with morbidity and mortality. Therefore, this study aims to explore the anti-fibrotic effects of low dose of paclitaxel (PTX) against thioacetamide (TAA)-induced liver fibrosis in rats and the possible mechanisms involved. TAA was administered at a dose of 200 mg/kg twice weekly for 6 weeks in rats to induce liver fibrosis similar to that in humans. Liver dysfunction was shown by increased alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, and γ-glutamyl transferase, along with histopathological changes. Liver fibrosis was confirmed by Masson's Trichome staining, increased collagen content, and elevated α-smooth muscle actin (α-SMA) protein expression. In addition, TAA induced liver apoptosis as indicated by the increased terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells in liver tissues. This study demonstrated that the administration of PTX (0.3 mg/kg/i.p.) three times a week for 6 weeks significantly alleviated functional and biochemical changes induced by TAA in addition to improving the liver architecture. PTX attenuated liver fibrosis as reflected by the decreased collagen content and α-SMA protein expression. Additionally, PTX attenuated liver apoptosis as indicated by the decreased TUNEL-positive cells. Moreover, PTX prevented TAA-induced elevation of transforming growth factor-ß1 (TGF-ß1), platelet-derived growth factor-BB (PDGF-BB), and tissue inhibitor of metalloproteinase 1 (TIMP-1) levels in liver tissues. These findings suggest that the low dose of PTX prevented TAA-induced liver fibrosis in rats, possibly by inhibiting the expression of TGF-ß1 and PDGF-BB and subsequently suppressing the apoptosis and the expression of TIMP-1.


Subject(s)
Liver Cirrhosis , Paclitaxel/pharmacology , Thioacetamide/toxicity , Animals , Disease Models, Animal , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Cirrhosis/prevention & control , Male , Rats , Rats, Sprague-Dawley
3.
Acta Pharm ; 70(4): 515-525, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32412432

ABSTRACT

Cisplatin-induced nephrotoxicity limits its anticancer effectiveness, thus this study's aim was to assess the potential modulatory effect of perindopril on cisplatin-induced nephrotoxicity and to elucidate the possible underlying mechanisms. Renal dysfunction was induced in mice by a single injection of cisplatin (10 mg kg-1, i.p.) and perindopril was administered orally (2 mg kg-1, once daily) for 5 days. Perindopril remarkably ameliorated cisplatin-induced perturbations in renal histology, renal levels of tumor necrosis factor-alpha, interleukin-6 and interleukin-10, apoptosis-regulating protein expressions (Bax and Bcl2), and partially normalized Bax to Bcl2 ratio and active caspase 3 protein expression. Conversely, perindopril had no significant effect on cisplatin-induced elevations in serum creatinine and urea, microalbuminuria, kidney to body weight ratio, lipid peroxidation marker, superoxide dismutase and catalase activities and reduced glutathione content. In conclusion, perindopril may be safely used with cisplatin in mice since it ameliorated cisplatin-induced histopathological changes, inflammation and apoptosis without affecting renal biomarkers or oxidative stress.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Antineoplastic Agents/toxicity , Caspase 3/physiology , Cisplatin/antagonists & inhibitors , Cisplatin/toxicity , Cytokines/physiology , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Perindopril/pharmacology , Albuminuria/chemically induced , Albuminuria/prevention & control , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Inflammation/pathology , Kidney Function Tests , Lipid Peroxidation/drug effects , Male , Mice , Oxidative Stress/drug effects
4.
Naunyn Schmiedebergs Arch Pharmacol ; 393(7): 1161-1171, 2020 07.
Article in English | MEDLINE | ID: mdl-31950223

ABSTRACT

The current study was designed to evaluate the potential abatement effect of pantoprazole against cisplatin-induced nephrotoxicity and establishing the possible protective mechanisms. Thirty-two male mice were allocated for treatment with saline, single dose of cisplatin (10 mg/kg/i.p), pantoprazole (30 mg/kg/once daily) for 5 days or combination of pantoprazole and cisplatin for 5 days. Urine, blood, and both kidneys were collected for further evaluations. Pantoprazole significantly countermand cisplatin-induced disfigurement of renal histology, kidney weight to body weight ratio, serum levels of creatinine and urea, and microalbuminuria. Furthermore, pantoprazole mostly normalized cisplatin-induced distortion of renal levels of inflammatory cytokines (tumor necrosis factor-alpha, interleukin-6, interleukin-10) and renal content of apoptosis regulating protein expressions (Bax, Bcl2, and active caspase 3). In addition, pantoprazole significantly subsided cisplatin-induced distortion of renal lipid peroxidation marker (MDA), renal superoxide dismutase, and catalase activities and renal reduced glutathione content. This study provides an evidence for the protective utility of short-term pantoprazole against cisplatin-induced nephrotoxicity in mice. The protective mechanism of pantoprazole could be through diminution of cisplatin-induced inflammation, oxidative stress, and their subsequent apoptotic renal cell death via abatement of apoptosis regulating protein expressions (Bax, Bcl2, and active caspase3).


Subject(s)
Cisplatin/toxicity , Kidney Diseases/prevention & control , Oxidative Stress/drug effects , Pantoprazole/pharmacology , Animals , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Caspase 3/metabolism , Cytokines/metabolism , Inflammation/chemically induced , Inflammation/prevention & control , Kidney Diseases/chemically induced , Lipid Peroxidation/drug effects , Male , Mice , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism
5.
Toxicol Appl Pharmacol ; 380: 114697, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31394158

ABSTRACT

Liver cholestasis is a complex disease of broad etiologies. Hedgehog (Hh) signaling has been shown to play a pivotal role in modulating liver repair post cholestatic liver injury. We here investigated the role of vitamin D in experimental liver cholestasis induced by bile-duct ligation (BDL) in rats. Male Sprague Dawley rats underwent BDL surgery and two weeks post-surgery; vitamin D was administered daily for two weeks. Serum markers of hepatocellular integrity were measured and fibrosis was detected by measuring α-SMA and hepatic TGF-ß1 as well as hepatic collagen content. Hh signaling was evaluated by measuring Smo and Gli1 levels. Histopathological examination of hepatic tissue was performed. Vitamin D alleviated obstructive cholestatic damage as illustrated by total bilirubin as well as gamma glutamyl transferase (γ-GT) serum levels. It also alleviated hepatocellular damage as suggested by reducing elevated serum aminotransferases induced by BDL. Antifibrotic activity of vitamin D was confirmed by decrease in mRNA and protein expression of α-SMA, as well as collagen content in hepatic tissue. Furthermore, vitamin D suppressed BDL-induced elevated hepatic TGF-ß1 mRNA and protein levels. BDL activated Hh signaling and upregulated its upstream component smoothened (Smo) and downstream target gene Gli1. Treatment with vitamin D reduced Smo mRNA levels and suppressed hepatic Gli1 mRNA and protein levels. Molecular docking of vitamin D to Smo revealed that vitamin D binds similarly to its endogenous cholesterol ligand and blocks its activation. These results demonstrate that vitamin D mitigated cholestatic liver injury through inhibiting Hh signaling.


Subject(s)
Cholecalciferol/therapeutic use , Cholestasis/drug therapy , Liver Cirrhosis/drug therapy , Vitamins/therapeutic use , Animals , Bile Ducts , Cholecalciferol/pharmacology , Cholestasis/genetics , Cholestasis/metabolism , Cholestasis/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Hedgehog Proteins/metabolism , Ligation , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Molecular Docking Simulation , Rats, Sprague-Dawley , Signal Transduction , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vitamins/pharmacology , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
6.
Naunyn Schmiedebergs Arch Pharmacol ; 392(12): 1569-1576, 2019 12.
Article in English | MEDLINE | ID: mdl-31367862

ABSTRACT

Liver fibrosis is a challenging global health problem resulting from chronic liver injury with no treatment currently available. It has been shown that activators for different peroxisome proliferator-activated receptor (PPAR) isoforms (α, γ, and δ) can affect different pathways in liver fibrosis. To evaluate the effects of the dual PPAR-α/γ agonist saroglitazar (SGZ) against thioacetamide (TAA)-induced fibrosis in rats, SGZ was administered for 6 weeks together with TAA injection. Administration of SGZ ameliorated TAA-induced elevation in hepatic biomarkers. SGZ was able to inhibit periportal and intralobular fibrous connective tissue proliferation, to decrease hydroxyproline content, and to lower alpha smooth muscle actin (α-SMA) protein expression. To unearth the antifibrotic mechanism of SGZ, the role of several fibrotic markers was studied. SGZ possesses inhibitory effect on protein levels of leptin, transforming growth factor-beta 1 (TGF-ß1) and platelet-derived growth factor-BB (PDGF-BB). Furthermore, SGZ rectified matrix degradation through decreasing tissue inhibitor of metalloproteinases-1 (TIMP-1). This study suggests that SGZ could have a possible antifibrotic effect via suppression of leptin that can repress TGF-ß1 and PDFG-BB, with subsequent inhibition of TIMP-1.


Subject(s)
Liver Cirrhosis/drug therapy , PPAR alpha/agonists , PPAR gamma/agonists , Phenylpropionates/therapeutic use , Pyrroles/therapeutic use , Actins/metabolism , Animals , Becaplermin/metabolism , Leptin/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Phenylpropionates/pharmacology , Pyrroles/pharmacology , Rats, Sprague-Dawley , Thioacetamide , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta1/metabolism
7.
J Biochem Mol Toxicol ; 32(11): e22214, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30194743

ABSTRACT

Acute lung injury (ALI) is a universal cause of respiratory failure and death especially after sepsis. The current study evaluates the protective efficacy of acylated catalpol diglycoside (ACD), a plant iridoid glycoside, against lipopolysaccharides (LPS)-induced ALI in rats. ACD prevented LPS-induced elevations in total and differential cell counts and total protein content in bronchoalveolar lavage fluid, lung malondialdehyde content, and serum lactate dehydrogenase activity. Moreover, ACD significantly increased lung glutathione and superoxide dismutase and improved lung histopathology with significant reduction in lung tumor necrosis factor-α (TNF-α) content mediated mainly via inhibition of TNF-α messenger RNA (mRNA) expression. In addition, ACD significantly reduced lung total nitrate concentration content through downregulation of inducible nitric oxide synthase (iNOS) mRNA expression, negating the excessive undesired vasodilatation characteristic to sepsis. In conclusion, the reduction of oxidative stress, amelioration of inflammatory cytokines expression, and antagonism of sepsis associated-excessive vasodilatation are main contributors in ACD's protective effect in LPS-induced ALI in rats.


Subject(s)
Acute Lung Injury/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/therapeutic use , Lung/drug effects , Nitric Oxide Synthase Type II/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acylation , Administration, Inhalation , Aerosols , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Antioxidants/chemistry , Antioxidants/isolation & purification , Biomarkers/metabolism , Gene Expression Regulation/drug effects , Lipid Peroxidation/drug effects , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/toxicity , Lung/immunology , Lung/metabolism , Lung/pathology , Molecular Structure , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
Life Sci ; 211: 245-251, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-30243650

ABSTRACT

Liver fibrosis is a global health issue that causes morbidity and mortality with no currently available treatment. It has been shown that low dose paclitaxel (PTX) can stabilize microtubules and inhibit the profibrotic transforming growth factor-beta 1 (TGF-ß1) signaling pathway. In this study the effect of treatment with low dose PTX was examined using a model of cholestatic liver fibrosis. Bile-duct ligation (BDL) was induced in rats for 2 weeks then PTX (0.3 mg/kg/ip) was administered three times a week for 2 weeks. Administration of PTX ameliorated BDL-induced elevation in biomarkers of hepatocellular damage (alanine transaminase; ALT and aspartate transaminase; AST) and obstructive cholestatic injury (total bilirubin and gamma glutamyl transferase; γ-GT). PTX was able to correct the increase in liver weight to body weight ratio and the bile duct proliferation induced by BDL. Additionally, PTX treatment corrected the BDL-induced fibrosis of portal tracts, elevation of hydroxyproline content and increased alpha smooth muscle actin (α-SMA) mRNA and protein expression. This antifibrotic effect of PTX was further examined through its inhibitory effect on TGF-ß1 mRNA and protein expression in addition to c-Myc mRNA expression. Furthermore, PTX rectified the BDL-induced decrease in interleukin-10 (IL-10) mRNA and protein expression. In conclusion, this study suggests that PTX at low dose has the potential to treat BDL-induced liver fibrosis in rats possibly through suppression of TGF-ß1 and c-Myc and activation of IL-10 pathways.


Subject(s)
Bile Ducts/surgery , Gene Expression Regulation/drug effects , Interleukin-10/metabolism , Liver Cirrhosis/prevention & control , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Interleukin-10/genetics , Ligation , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Proto-Oncogene Proteins c-myc/genetics , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/genetics
9.
Naunyn Schmiedebergs Arch Pharmacol ; 391(11): 1203-1219, 2018 11.
Article in English | MEDLINE | ID: mdl-30058017

ABSTRACT

The effects of cinnamaldehyde (CIN), a commonly consumed food flavor, against high-cholesterol diet (HCD)-induced vascular damage in rabbits were evaluated. Male New Zealand rabbits (n = 24) were allocated to four groups at random: control, fed with standard rabbit chow; CIN, fed with standard diet and administered CIN; HCD, fed with 1% cholesterol-enriched diet; and HCD-CIN, fed with HCD and treated with CIN. CIN was orally given at a dose of (10 mg/kg/day) concomitantly with each diet type from day 1 until the termination of the experimental protocol (4 weeks). HCD elicited significant elevations in serum levels of total cholesterol (TC), triglycerides (TGs), and high- and low-density lipoprotein cholesterol (HDL-C and LDL-C, respectively) compared with control rabbits. Moreover, aortic levels of nitric oxide metabolites (NOx) and antioxidant enzyme activities were significantly lower, while aortic levels of malondialdehyde (MDA) and myeloperoxidase (MPO) activity were significantly higher, in HCD-fed rabbits relative to control animals. CIN administration mitigated or completely reversed HCD-induced metabolic alterations, vascular oxidative stress, and inflammation. Moreover, CIN ameliorated HCD-induced vascular functional and structural irregularities. Aortic rings from HCD-CIN group showed improved relaxation to acetylcholine compared to aortas from HCD group. Moreover, CIN decreased atherosclerotic lipid deposition and intima/media (I/M) ratio of HCD aortas. CIN-mediated effects might be related to its ability to attenuate the elevated aortic mRNA expression of cholesteryl ester transfer protein (CETP) and MPO in HCD group. Interestingly, the vasculoprotective effects of CIN treatment in the current study do not seem to be mediated via Nrf2-dependent mechanisms. In conclusion, CIN may mitigate the development of atherosclerosis in hypercholestrolemic rabbits via cholesterol-lowering, antiinflammatory and antioxidant activities.


Subject(s)
Acrolein/analogs & derivatives , Hypercholesterolemia/drug therapy , Protective Agents/therapeutic use , Acrolein/pharmacology , Acrolein/therapeutic use , Animals , Aorta/drug effects , Aorta/pathology , Aorta/physiology , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cholesterol/blood , Cholesterol Ester Transfer Proteins/genetics , Cholesterol, Dietary/administration & dosage , Gene Expression Regulation , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Male , Nitric Oxide/metabolism , Peroxidase/genetics , Peroxidase/metabolism , Protective Agents/pharmacology , Rabbits , Triglycerides/blood
10.
Environ Toxicol Pharmacol ; 55: 74-80, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28837867

ABSTRACT

Vascular dysfunction leading to hypotension is a major complication in patients with septic shock. Inducible nitric oxide synthase (iNOS) together with oxidative stress play an important role in development of vascular dysfunction in sepsis. Searching for an endogenous, safe and yet effective remedy was the chief goal for this study. The current study investigated the effect of agmatine (AGM), an endogenous metabolite of l-arginine, on sepsis-induced vascular dysfunction induced by lipopolysaccharides (LPS) in rats. AGM pretreatment (10mg/kg, i.v.) 1h before LPS (5mg/kg, i.v.) prevented the LPS-induced mortality and elevations in serum creatine kinase-MB isoenzyme (CK-MB) activity, lactate dehydrogenase (LDH) activity, C-reactive protein (CRP) level and total nitrite/nitrate (NOx) level after 24h from LPS injection. The elevation in aortic lipid peroxidation illustrated by increased malondialdehyde (MDA) content and the decrease in aortic glutathione (GSH) and superoxide dismutase (SOD) were also ameliorated by AGM. Additionally, AGM prevented LPS-induced elevation in mRNA expression of iNOS, while endothelial NOS (eNOS) mRNA was not affected. Furthermore AGM prevented the impaired aortic contraction to KCl and phenylephrine (PE) and endothelium-dependent relaxation to acetylcholine (ACh) without affecting endothelium-independent relaxation to sodium nitroprusside (SNP). IN CONCLUSION: AGM may represent a potential endogenous therapeutic candidate for sepsis-induced vascular dysfunction through its inhibiting effect on iNOS expression and oxidative stress.


Subject(s)
Agmatine/administration & dosage , Antioxidants/administration & dosage , Endothelium, Vascular/drug effects , Endotoxemia/prevention & control , Lipopolysaccharides/adverse effects , Nitric Oxide Synthase Type II/genetics , Agmatine/pharmacology , Animals , Antioxidants/pharmacology , Disease Models, Animal , Down-Regulation , Endothelium, Vascular/physiopathology , Endotoxemia/chemically induced , Endotoxemia/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Lipid Peroxidation/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley
11.
Environ Toxicol Pharmacol ; 50: 175-182, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28189063

ABSTRACT

Acute hepatic dysfunction associating sepsis is mediated mainly by toll-like receptor-4 (TLR-4)/nuclear factor kappa-B (NF-κB) inflammatory pathway. This study explores potential hepatoprotective effect of the NF-κB inhibitor celastrol in cecal ligation and puncture (CLP) model in rats. Protective effect of celastrol (1mg/kg, i.p., 1h before CLP) was illustrated after 24h by preventing CLP-induced hepatic histopathological changes and elevation in serum hepatic biomarkers [alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TB) and gamma aminotransferase (γ-GT)] without affecting mortality. Celastrol anti-inflammatory effect was illustrated by inhibiting increased serum and hepatic mRNA expression of interleukin-6 (IL-6) without affecting IL-10 elevation. Furthermore, celastrol inhibited CLP-induced elevations in hepatic mRNA expression of nuclear factor inhibitory protein kappa-B alpha (NFκBia), TLR-4, 5-lipoxygenase (5-LOX) and prevented NF-κB/p65 nuclear translocation and activation. In conclusion, celastrol prevented CLP-induced acute hepatic dysfunction through its anti-inflammatory effect by attenuating NF-κB activation, TLR-4 and 5-LOX expression with subsequent reduction in pro-inflammatory IL-6.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Liver Diseases/prevention & control , NF-kappa B/antagonists & inhibitors , Triterpenes/administration & dosage , Animals , Anti-Inflammatory Agents/pharmacology , Cecum/surgery , Disease Models, Animal , Gene Expression Regulation/drug effects , Interleukin-10/blood , Interleukin-10/genetics , Interleukin-6/blood , Interleukin-6/genetics , Liver Diseases/etiology , Liver Diseases/genetics , Liver Diseases/metabolism , Male , Pentacyclic Triterpenes , Rats , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Triterpenes/pharmacology
12.
Naunyn Schmiedebergs Arch Pharmacol ; 390(4): 361-368, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28035464

ABSTRACT

Sepsis is a systemic inflammatory response associating severe infection leading to multi-organ failure, such as hepatic dysfunction. This study investigates the possible hepatoprotective effect of the lipoxin A4 agonist (BML-111) in cecal ligation and puncture (CLP) model in rats. Pretreatment with BML-111 (1 mg/kg, i.p., 1 h before CLP) protected against CLP-induced mortality after 24 h. BML-111 prevented marked inflammatory cells in liver tissues and decreased elevation in serum hepatic biomarkers [alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TB), gamma-glutamyl transferase (γ-GT)] induced by CLP. Additionally, BML-111 attenuated elevated serum level of interleukin-6 (IL-6) and downregulated hepatic IL-6 mRNA expression. Meanwhile, BML-111 further increased serum IL-10 and upregulated hepatic IL-10 mRNA expression, while it downregulated hepatic mRNA expression of nuclear factor inhibitory protein kappa-B alpha (NFκBia), toll-like receptor-4 (TLR-4), and 5-lipooxygenase (5-LOX). Moreover, BML-111 prevented NF-κB/p65 nuclear translocation and activation. In conclusion, BML-111 attenuated CLP-induced acute hepatic dysfunction through its anti-inflammatory effect by decreasing NF-κB activity, TLR-4, and 5-LOX expression with subsequent decrease in pro-inflammatory IL-6 and elevation in anti-inflammatory IL-10.


Subject(s)
Heptanoic Acids/therapeutic use , Lipoxins/agonists , Liver Diseases/drug therapy , Protective Agents/therapeutic use , Alanine Transaminase/blood , Animals , Arachidonate 5-Lipoxygenase/genetics , Aspartate Aminotransferases/blood , Bilirubin/blood , Cecum/injuries , Cecum/surgery , Heptanoic Acids/pharmacology , Interleukin-10/blood , Interleukin-10/genetics , Interleukin-6/blood , Interleukin-6/genetics , Ligation , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Diseases/etiology , Liver Diseases/metabolism , Liver Diseases/pathology , Male , NF-kappa B/genetics , NF-kappa B/metabolism , Protective Agents/pharmacology , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Toll-Like Receptor 4/genetics , gamma-Glutamyltransferase/blood
13.
Environ Toxicol Pharmacol ; 43: 182-92, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27011232

ABSTRACT

Acute liver injury secondary to sepsis is a major challenge in intensive care unit. This study was designed to investigate potential protective effects of pomegranate against sepsis-induced acute liver injury in rats and possible underlying mechanisms. Pomegranate was orally given (800mg/kg/day) for two weeks before sepsis induction by cecal ligation and puncture (CLP). Pomegranate improved survival and attenuated liver inflammatory response, likely related to downregulation of mRNA expression of toll like recptor-4, reduced nuclear translocation and DNA binding activity of proinflammatory transcription factor NF-κB subunit p65, decreased mRNA and protein expression of tumor necrosis factor-alpha and reduction in myeloperoxidase activity and mRNA expression. Pomegranate also decreased CLP-induced oxidative stress as reflected by decreased malondialdehyde content, and increased reduced glutathione level and superoxide dismutase activity. These results confirm the antiinflammatory and antioxidant effects of pomegranate in CLP-induced acute liver injury mediated through inhibiting TLR4/NF-κB pathway, lipid peroxidation and neutrophil infiltration.


Subject(s)
Lythraceae , NF-kappa B/metabolism , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Protective Agents/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Cecum/surgery , Inflammation/metabolism , Ligation , Liver/metabolism , Liver/physiology , NF-kappa B/antagonists & inhibitors , Punctures , Rats
14.
Can J Physiol Pharmacol ; 94(5): 488-97, 2016 May.
Article in English | MEDLINE | ID: mdl-26871756

ABSTRACT

This study investigates the effect of the ergogenic supplement ß-hydroxy-ß-methylbutyrate (HMB) on insulin resistance induced by high-fructose diet (HFD) in rats. Male Sprague Dawley rats were fed 60% HFD for 12 weeks and HMB (320 mg·kg(-1)·day(-1), orally) for 4 weeks. HFD significantly increased fasting insulin, fasting glucose, glycosylated hemoglobin (HBA1C), liver glycogen content, and homeostasis model assessment of insulin resistance (HOMA-IR) index, while it decreased glucose and insulin tolerance. Furthermore, HFD significantly increased serum triglycerides (TG), low density lipoprotein cholesterol (LDL-C), and very low density lipoprotein cholesterol (VLDL-C) levels, while it significantly decreased high density lipoprotein cholesterol (HDL-C). Moreover, HFD significantly increased mRNA expression of glucose transporter type-2 (GLUT-2), the mammalian target of rapamycin (mTOR), and sterol regulatory element-binding protein-1c (SREBP-1c) but decreased peroxisome proliferator-activated receptor-alpha (PPAR-α) in liver. Aortic relaxation to acetylcholine (ACh) was impaired and histopathology showed severe hepatic steatosis. HMB significantly increased insulin tolerance and decreased fasting insulin, HOMA-IR, HBA1C, hepatic glycogen content, serum TG, LDL-C, and VLDL-C. Additionally, HMB enhanced ACh-induced relaxation, ameliorated hepatic steatosis, and decreased mRNA expression of GLUT-2. In conclusion, HMB may attenuate insulin resistance and hepatic steatosis through inhibiting GLUT-2 in liver.


Subject(s)
Dietary Supplements , Glucose Transporter Type 2/antagonists & inhibitors , Insulin Resistance , Liver/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Performance-Enhancing Substances/therapeutic use , Valerates/therapeutic use , Animals , Dietary Carbohydrates/adverse effects , Endothelium, Vascular/physiopathology , Fructose/adverse effects , Gene Expression Regulation , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Glucose Intolerance/physiopathology , Glucose Intolerance/prevention & control , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Hyperinsulinism/physiopathology , Hyperinsulinism/prevention & control , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Hyperlipidemias/physiopathology , Hyperlipidemias/prevention & control , Liver/pathology , Male , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/physiopathology , PPAR alpha/metabolism , Random Allocation , Rats, Sprague-Dawley , Sterol Regulatory Element Binding Protein 1/metabolism , TOR Serine-Threonine Kinases/metabolism , Vascular Resistance
15.
Can J Physiol Pharmacol ; 94(2): 140-146, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26544923

ABSTRACT

In this study, we studied the effect of lipopolysaccharide from Rhodobacter sphaeroides (LPS-RS), an inhibitor of Toll-like receptor 4 (TLR4), in LPS-induced acute lung injury (ALI). Male Sprague-Dawley rats were treated with LPS-RS (0.1 mg/kg body mass, by intraperitoneal (i.p.) injection) 1 h before LPS injection (10 mg/kg, i.p.). Bronchoalveolar lavage fluid (BALF) and lung tissues were collected 24 h later to determine total and differential cell count, total protein content, levels of lactate dehydrogenase (LDH), histopathological changes, markers of oxidative stress, and mRNA expression of the inhibitory protein nuclear factor kappaB-α (NFκBIA) and TLR4. Additionally, rings of pulmonary artery were isolated for measuring vascular reactivity. LPS-induced ALI was indicated by increases in total and differential cell count, total protein, and LDH in BALF, and increased lung levels of malondialdehyde (MDA), as well as decreased activity of reduced glutathione (GSH) and superoxide dismutase (SOD). Moreover, LPS increased pulmonary artery contraction in response to phenylephrine (PE). Additionally, LPS downregulated mRNA expression of NFκBIA and upregulated mRNA expression of TLR4. LPS caused a marked inflammation in the lung tissue, with tubercular granuloma and numerous neutrophils. Pretreatment with LPS-RS protected against LPS-induced ALI by decreasing total and differential cell count, total protein, and LDH in BALF, and increased pulmonary GSH content and SOD activity without affecting MDA content. Additionally, it decreased the elevated PE-induced pulmonary artery contraction. LPS-RS upregulated mRNA expression of NFκBIA and downregulated mRNA expression of TLR4. Moreover, LPS-RS prevented inflammation in lung tissues. In conclusion, pretreatment with LPS-RS protects against LPS-induced ALI in rats through its anti-inflammatory effects, possibly by decreasing the mRNA expression of TLR4 and increasing that of NFκBIA.

16.
Naunyn Schmiedebergs Arch Pharmacol ; 389(1): 45-56, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26449613

ABSTRACT

Insulin resistance is a serious health condition worldwide; however, its exact mechanisms are still unclear. This study investigates agmatine (AGM; an endogenous metabolite of L-arginine) effects on insulin resistance induced by high fructose diet (HFD) in rats and the possible involved mechanisms. Sprague Dawley rats were fed 60% HFD for 12 weeks, and AGM (10 mg/kg/day, orally) was given from week 9 to 12. AGM significantly reduced HFD-induced elevation in fasting insulin level, homeostasis model assessment of insulin resistance (HOMA-IR) index and liver glycogen content from 3.44-, 3.62- and 2.07- to 2.59-, 2.78- and 1.3-fold, respectively, compared to the control group, while it increased HFD-induced reduction in glucose tolerance. Additionally, AGM significantly decreased HFD-induced elevation in serum triglycerides, low density lipoprotein cholesterol and very low density lipoprotein cholesterol levels from 3.18-, 2.97- and 4.75- to 1.25-, 1.25- and 1.07-fold, respectively, compared to control group. Conversely, AGM had no significant effect on HFD-induced changes in fasting glucose, glycosylated hemoglobin, insulin tolerance and high density lipoprotein cholesterol. Furthermore, AGM significantly reduced HFD-induced elevation in mRNA expression of glucose transporter type-2 (GLUT-2), mammalian target of rapamycin (mTOR) and sterol regulatory element-binding protein-1c (SREBP-1c) without affecting that of peroxisome proliferator-activated receptor-alpha (PPAR-α) in the liver. Additionally, AGM enhanced ACh-induced aortic relaxation and attenuated liver steatosis induced by HFD. In conclusion, AGM may have a therapeutic potential in insulin resistance through suppressing SREBP-1c, mTOR and GLUT-2 in liver.


Subject(s)
Agmatine/pharmacology , Glucose Transporter Type 2/genetics , Insulin Resistance , Sterol Regulatory Element Binding Protein 1/genetics , TOR Serine-Threonine Kinases/genetics , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Blood Glucose/analysis , Diet , Fructose , Glucose Tolerance Test , Glycated Hemoglobin/analysis , Glycogen/metabolism , In Vitro Techniques , Insulin/blood , Lipids/blood , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , PPAR alpha/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley
17.
Int Immunopharmacol ; 30: 163-170, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26687059

ABSTRACT

Acute lung injury (ALI) is a serious clinical syndrome with a high rate of mortality. In this study, the effects of apocynin, a NADPH-oxidase (NOX) inhibitor on lipopolysaccharide (LPS)-induced ALI in rats were investigated. Male Sprague-Dawley rats were treated with apocynin (10mg/kg) intraperitoneally (i.p.) 1h before LPS injection (10mg/kg, i.p.). The results revealed that apocynin attenuated LPS-induced ALI as it decreased total protein content, lactate dehydrogenase (LDH) activity and the accumulation of the inflammatory cells in the bronchoalveolar lavage fluid (BALF), In addition, apocynin significantly increased superoxide dismutase (SOD) and reduced glutathione (GSH) activities with significant decrease in the lung malondialdehyde (MDA) content as compared to LPS group in lung tissue and decreased pulmonary artery contraction induced by LPS. It also upregulated mRNA expression of inhibitory protein kappaB-alpha (NFκBia) and downregulated mRNA expression of Toll-Like receptor 4 (TLR4) and decreased inflammation observed in lung tissues. Collectively, these results demonstrate the protective effects of apocynin against the LPS-induced ALI in rats through its antioxidant and antiinflammatory effect that may be attributed to the decrease in mRNA expression of TLR4 and increasing that of NFκBia.


Subject(s)
Acetophenones/administration & dosage , Acute Lung Injury/drug therapy , Anti-Inflammatory Agents/administration & dosage , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Acute Lung Injury/chemically induced , Animals , Endotoxins/immunology , L-Lactate Dehydrogenase/metabolism , Lipopolysaccharides/immunology , Lung/metabolism , Male , NF-kappa B/genetics , Oxidation-Reduction/drug effects , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Toll-Like Receptor 4/genetics
18.
Can J Physiol Pharmacol ; 92(3): 175-80, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24593781

ABSTRACT

Cyclic nucleotides, such as cAMP and cGMP, play a protective role in the modulation of the activity of some inflammatory cells in allergic disorders. Their intracellular concentrations are tightly regulated by the phosphodiesterases (PDEs). The protective efficacy of the selective PDE5 inhibitor vardenafil against mast-cell-mediated allergic reactions in murine models has been investigated. Compound 48/80 was used as a direct mast cell degranulator to induce anaphylaxis. Vardenafil (administered orally at 5, 10, 20, 40, and 80 mg/kg body mass) significantly (P < 0.05, n = 12) increased protection against compound-48/80-induced anaphylaxis in mice to 33.33%, 66.67%, 66.67%, 83.33%, and 66.67% respectively compared with the control (vehicle). In passive cutaneous anaphylaxis (PCA) in rats, vardenafil (10 mg/kg body mass) significantly (P < 0.05, n = 6) decreased Evans' blue dye extravasation (4.6-fold). Pre-incubation of isolated rat peritoneal mast cells (RPMCs) with vardenafil (10 and 100 µmol/L) significantly (P < 0.05, n = 6) reduced compound-48/80-induced histamine release by 2.8- and 3-fold, respectively. Moreover, histamine release by immunogenic stimulation of sensitized RPMCs by egg albumin significantly declined following pre-incubation with vardenafil (10 and 100 µmol/L) by 1.94- and 1.99-fold, respectively. In conclusion, inhibition of PDE5 by vardenafil ameliorated immunologic and non-immunologic mast-cell-mediated allergic reactions and reduced histamine release, providing evidence for the potential anti-allergic properties of vardenafil.


Subject(s)
Anti-Allergic Agents/pharmacology , Hypersensitivity/drug therapy , Imidazoles/pharmacology , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Anaphylaxis/drug therapy , Anaphylaxis/immunology , Animals , Anti-Allergic Agents/therapeutic use , Cell Degranulation , Histamine Release , Hypersensitivity/immunology , Imidazoles/therapeutic use , Male , Mast Cells/drug effects , Mast Cells/immunology , Mice , Passive Cutaneous Anaphylaxis/drug effects , Peritoneal Cavity/cytology , Phosphodiesterase 5 Inhibitors/therapeutic use , Piperazines/therapeutic use , Rats , Sulfones/pharmacology , Sulfones/therapeutic use , Triazines/pharmacology , Triazines/therapeutic use , Vardenafil Dihydrochloride , p-Methoxy-N-methylphenethylamine/immunology
19.
J. physiol. biochem ; 70(1): 247-254, mar. 2014.
Article in English | IBECS | ID: ibc-121623

ABSTRACT

While propolis is known to have abundant bioactive constituents and a variety of biological activities, it is not clear whether propolis has beneficial effects on high glucose-mediated vascular endothelial impairment. The aim of the present study was to investigate the potential protective effect of propolis extract against the acute vascular endothelial dysfunction resulting from exposure to high glucose load and to elucidate its underlying mechanism. Rat aortic rings were incubated with normal glucose (11 mM), high glucose (44 mM), or mannitol (44 mM) for 3 h with or without propolis extract (400 ìg/ml). Contraction to phenylephrine (Phe, 10−9–10−5 M) and relaxation to acetylcholine (ACh, 10−9–10−5 M) and sodium nitroprusside (SNP, 10−9–10−5 M) were measured before and after incubation. Changes in malondialdehyde (MDA), reduced glutathione (GSH), and superoxide dismutase (SOD) were also measured. Phe-induced contraction was impaired by high glucose as the E max decreased from 138.87 ± 11.43 to 103.65 ± 11.5 %. In addition, ACh-induced relaxation was impaired as the E max decreased from 99.80 ± 7.25 to 39.20 ± 6.5 %. SNP-induced relaxation was not affected. Furthermore, high glucose decreased the levels of both SOD (by 6 U/ml) and GSH (by 68 %) and increased levels of MDA (by 85 %). Propolis extract prevented high glucose-induced impairment of Phe and ACh responses and increased both SOD and GSH, leading to decreased MDA levels. In conclusion, propolis can protect against high glucose-induced vascular dysfunction by reducing oxidative stress (AU)


Subject(s)
Animals , Rats , Propolis/pharmacokinetics , Endothelium, Vascular/physiopathology , Diabetic Angiopathies/prevention & control , Protective Agents/pharmacokinetics , Disease Models, Animal , Oxidative Stress
20.
J Pharm Pharmacol ; 66(6): 835-43, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24393128

ABSTRACT

OBJECTIVES: The aim of this work was to explore possible effects of agmatine, an endogenous inhibitor of inducible nitric oxide synthase (iNOS), against hypercholesterolemia-induced lipid profile changes and endothelial dysfunction. METHODS: Hypercholesterolemia was induced by feeding rabbits with a high-cholesterol diet (HCD, 0.5%) for 8 weeks. Another HCD-fed group was orally administered agmatine (10 mg/kg/day) during weeks 5 through 8. Serum lipid profile, malondialdehyde (MDA), nitric oxide (NO) and lactate dehydrogenase (LDH) were determined. Aorta was isolated to analyse vascular reactivity, atherosclerotic lesions and intima/media (I/M) ratio. KEY FINDINGS: HCD induced a significant increase in serum total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), triglycerides and high-density lipoprotein cholesterol (HDL-C). Agmatine administration significantly decreased HCD-induced elevations in serum TC and LDL-C, MDA, LDH and NO while significantly increased HDL-C levels. Additionally, agmatine significantly protected against HCD-induced attenuation of rabbit aortic endothelium-dependent relaxation to acetylcholine. HCD and agmatine did not significantly influence aortic endothelium-independent relaxation to sodium nitroprusside. Moreover, agmatine significantly reduced the elevation in aortic atherosclerotic lesion area and I/M ratio. CONCLUSIONS: This study is the first to reveal that agmatine has the ability to ameliorate hypercholesterolemia-induced lipemic-oxidative and endothelial function injuries possibly by its antioxidant potential and/or iNOS inhibition.


Subject(s)
Agmatine/therapeutic use , Atherosclerosis/drug therapy , Endothelium, Vascular/drug effects , Animals , Atherosclerosis/pathology , Body Weight/drug effects , Cholesterol/administration & dosage , Disease Progression , Endothelium, Vascular/physiology , L-Lactate Dehydrogenase/blood , Lipids/blood , Male , Nitric Oxide/physiology , Oxidative Stress , Rabbits , Vasoconstriction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...