Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Pharmacol Rep ; 76(1): 98-111, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38214881

ABSTRACT

BACKGROUND: Obstructive nephropathy is a condition often caused by urinary tract obstruction either anatomical (e.g., tumors), mechanical (e.g., urolithiasis), or compression (e.g., pregnancy) and can progress to chronic kidney disease (CKD). Studies have shown sexual dimorphism in CKD, where males were found to have a more rapid decline in kidney function following kidney injury compared to age-matched females. Protocatechuic acid (PCA), an anti-oxidant and anti-inflammatory polyphenolic compound, has demonstrated promising effects in mitigating drug-induced kidney injuries. The current study aims to explore sexual dimorphism in kidney injury after unilateral ureteral obstruction (UUO) and assess whether PCA treatment can mitigate kidney injury in both sexes. METHODS: UUO was induced in 10-12 weeks old male and female C57BL/6J mice. Mice were categorized into four groups (n = 6-8/group); Sham, Sham plus PCA (100 mg/kg, I.P daily), UUO, and UUO plus PCA. RESULTS: After 2 weeks of induction of UUO, markers of kidney oxidative stress (TBARs), inflammation (IL-1α and IL-6), tubular injury (neutrophil gelatinase-associated lipocalin, NGAL and urinary kidney injury molecule-1, KIM-1), fibrosis (Masson's trichrome staining, collagen IV expression, MMP-2 and MMP-9) and apoptosis (TUNEL+ cells, active caspase-1 and caspase-3) were significantly elevated in both males and females relative to their sham counterparts. Males exhibited significantly greater kidney oxidative stress, inflammation, fibrosis, and apoptosis after induction of UUO when compared to females. PCA treatment significantly attenuated UUO-induced kidney injury, inflammation, fibrosis, and apoptosis in both sexes. CONCLUSION: Our findings suggest a differential gender response to UUO-induced kidney injury with males being more sensitive to UUO-induced kidney inflammation, fibrosis, and apoptosis than age-matched females. Importantly, PCA treatment reduced UUO-induced kidney injury in a sex-independent manner which might be attributed to its anti-oxidant, anti-inflammatory, anti-fibrotic, and anti-apoptotic properties.


Subject(s)
Hydroxybenzoates , Kidney Diseases , Renal Insufficiency, Chronic , Ureteral Obstruction , Female , Mice , Male , Animals , Ureteral Obstruction/complications , Ureteral Obstruction/drug therapy , Sex Characteristics , Antioxidants/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Kidney , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Kidney Diseases/prevention & control , Renal Insufficiency, Chronic/metabolism , Apoptosis , Inflammation/metabolism , Fibrosis , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
2.
Int Immunopharmacol ; 128: 111587, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38286073

ABSTRACT

BACKGROUND: Ulcerative colitis (UC), an ongoing inflammatory disorder of the colon, is marked by persistent mucosal surface irritation extending from the rectum to the near-proximal colon. Tiron is a synthetic analogue of vitamin E which is known to have antioxidant and anti-inflammatory effects in various animal models, so the goal of this study was to find out whether Tiron had any preventive impacts on UC inflicted by acetic acid (A.A) exposure in rats. METHOD: Tiron (235 and 470 mg/kg) was administered intra-peritoneally for 2 weeks, and A.A (2 ml, 3 % v/v) was injected intra-rectally to cause colitis. Colon tissues and blood samples were then collected for measurement of various inflammatory and oxidative stress biomarkers. RESULTS: Tiron administration significantly diminished lactate dehydrogenase (LDH), C-reactive protein (CRP), colon weight, and the weight/length ratio of the colon as compared to A.A-injected rats. Additionally, Tiron attenuated oxidative stress biomarkers. Tiron also enforced the levels of Glucagon-like peptide-1 (GLP-1) and trefoil factor-3 (TFF-3), while it greatly lowered the expression of nuclear factor kappa B (NF-κB), interleukin-6 (IL-6), interferon-γ (IFN-γ), and transforming growth factor-1(TGF-ß1), phosphorylated epidermal growth factor receptor (P-EGFR), phosphatidylinositol-3-kinase (PI3K) and protein kinase B (AKT) expression in colonic cellular structures. Furthermore, colonichistopathologic damages, revealed by hematoxylin and eosin (H&E) and Alcian Blue stain, were significantly decreased upon Tiron administration. CONCLUSION: Tiron prevented A.A-induced colitis in rats via modulating inflammatory pathway TGF-ß1/P-EGFR/PI3K/AKT/NF-κB, alongside managing the oxidant/antioxidant equilibrium, and boosting the reliability of the intestinal barrier.


Subject(s)
Colitis, Ulcerative , Colitis , Rats , Animals , NF-kappa B/metabolism , Transforming Growth Factor beta1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/metabolism , 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Antioxidants/pharmacology , Acetic Acid/metabolism , Transforming Growth Factor beta/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Reproducibility of Results , Colon/pathology , Signal Transduction , Colitis/pathology , Colitis, Ulcerative/chemically induced , ErbB Receptors/metabolism , Biomarkers/metabolism
3.
Life Sci ; 327: 121820, 2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37263490

ABSTRACT

BACKGROUND: Ulcerative colitis (UC) is a chronic mucosal inflammation of the large intestine that mostly affects the rectum and colon. The absence of safe and effective therapeutic agents encourages the discovery of novel therapeutic agents to effectively treat UC and its complications. The purpose of this research was to examine the protective impact of Eicosapentaenoic acid (EPA) in rats with UC induced by acetic acid (AA). METHOD: AA (2 ml, 3 % v/v) was injected intrarectally to cause UC. Before administering AA, EPA (300 and 1000 mg/kg) was given orally for 28 days. RESULTS: EPA inhibited AA-induced UC by enhancing colonic histopathological changes like inflammation, goblet cell loss, glandular hyperplasia and mucosal ulceration, concomitant with a reduction in colon weight, colon weight/length ratio, C-reactive protein (CRP), and serum lactate dehydrogenase (LDH). EPA also effectively restored the imbalance between oxidants and antioxidants caused by AA. In addition, EPA increased the levels of trefoil factor-3 (TFF-3) and glucagon-like peptide-1 (GLP-1), while significantly reducing the expression of nuclear factor kappa B (NF-κB), interferon-γ (IFN-γ), and interleukin-6 (IL-6), transforming growth factor-1(TGF-ß1), and phosphorylated epidermal growth factor receptor (P-EGFR), phosphatidylinositol-3-kinase (PI3K) and protein kinase B (AKT) expression in colonic tissues. CONCLUSION: EPA inhibited AA-induced UC in rats by modulating the TGF-ß/P-EGFR and NF-κB inflammatory pathways, regulating the oxidant/antioxidant balance, and enhancing the colon barrier integrity.


Subject(s)
Colitis, Ulcerative , Rats , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , NF-kappa B/metabolism , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/therapeutic use , Acetic Acid , Transforming Growth Factor beta/metabolism , Signal Transduction , Colon/metabolism , Inflammation/pathology , Antioxidants/pharmacology , ErbB Receptors/metabolism
4.
Anal Biochem ; 673: 115181, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37247750

ABSTRACT

Arginine Vasopressin (AVP) is one of the key hormones in the human body. AVP is clinically important because it maintains body fluid balance and vascular tone. Unfortunately, AVP laboratory measurements are always difficult and with low accuracy. Copeptin, the C-terminal of the AVP precursor, is released in equal amounts with AVP, making it a sensitive marker of AVP release. Despite being a non-specific biomarker, copeptin earned a lot of attention as a novel biomarker due to easy and quick laboratory measurements. Recent studies have reported the critical role of copeptin as a clinical indicator, especially in the diagnosis and prognosis of many diseases. Besides, it was reported that the combination between copeptin and gold standard biomarkers improved the prognostic values of those biomarkers. In this review, the role of copeptin as a new predictive diagnostic and prognostic biomarker of various diseases is highlighted according to the most recent studies. In addition, the importance of using copeptin as a marker in different medical departments and the impact of this on improving healthcare service was discussed.


Subject(s)
Arginine Vasopressin , Glycopeptides , Humans , Prognosis , Biomarkers
5.
Life Sci ; 322: 121654, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37023955

ABSTRACT

Thioacetamide (TAA), a classic liver toxic compound, is used to establish experimental models of liver injury via induction of inflammation and oxidative stress. The current study was employed to explore the effects of canagliflozin (CANA), a sodium glucose cotransporter 2 (SGLT-2) inhibitor and antidiabetic agent, on TAA-induced acute liver injury. METHODS: A rat model of acute hepatic injury was established using single intraperitoneal injection of TAA (500 mg/kg) and rats received CANA (10 and 30 mg/kg, orally) once daily for 10 days prior to TAA challenge. Liver function, oxidative stress, and inflammatory parameters were measured in serum and hepatic tissues of rats. RESULTS: Elevated levels of liver enzymes, hepatic malondialdehyde (MDA), and serum lactate dehydrogenase (LDH) were significantly attenuated by CANA. CANA also increased hepatic superoxide dismutase (SOD) and glutathione (GSH). Hepatic levels of high-mobility group box 1 (HMGB1), toll like receptor4 (TLR4), receptor for advanced glycation end products (RAGE), and pro-inflammatory cytokines (IL-6, and IL-1ß) were normalized with CANA. Additionally, Hepatic expression of p-JNK/p-p38 MAPK was significantly attenuated by CANA compared to TAA-treated rats. CANA also decreased hepatic immunoexpression of NF-κB and TNF-α and attenuated hepatic histopathological alterations via reduction of inflammation and necrosis scores and collagen deposition. Moreover, mRNA expression levels of TNF-α and IL-6 were reduced upon CANA treatment. CONCLUSION: CANA attenuates TAA-prompted acute liver damage, via suppressing HMGB1/RAGE/TLR4 signaling, regulation of oxidative stress and inflammation pathways.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , HMGB1 Protein , Rats , Animals , Thioacetamide/toxicity , Receptor for Advanced Glycation End Products/metabolism , Toll-Like Receptor 4/metabolism , Canagliflozin/pharmacology , Tumor Necrosis Factor-alpha/metabolism , HMGB1 Protein/metabolism , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Interleukin-6/metabolism , Liver/metabolism , Signal Transduction , NF-kappa B/metabolism , Inflammation/pathology , Oxidative Stress , Glutathione/metabolism
6.
Neurotoxicology ; 92: 110-121, 2022 09.
Article in English | MEDLINE | ID: mdl-35961375

ABSTRACT

RATIONALE: Hepatic encephalopathy (HE) is a neuropsychiatric disorder that results from either acute or chronic liver failure. CXCR2 plays an essential role in the pathophysiology of liver and brain diseases. In the present study, the potential beneficial effects of SB332235, a selective inhibitor of CXCR2, against HE were evaluated. METHODS: HE was induced in male rats by thioacetamide injection (200 mg/kg, i.p.) at three alternative days. SB332235 was injected in rats 1 h before TAA at a dose of 1 and 3 mg/kg i.p. RESULTS: SB332235 alleviated oxidative stress as shown by the decreased serum NO and reduced MDA, elevated GSH and SOD levels, and reduced TNF-α and NF-κB levels in both brain and liver tissues of rats. Additionally, SB332235 suppressed brain ASK-1, JNK, IL-8, and caspase-3 expression, and activated PI3K/AKT expression in brain tissues. Markers of brain dysfunction, such as ammonia, and markers of hepatic injury, such as LDH, albumin, bilirubin, γGT, AST, ALT, and ALP, were significantly ameliorated. Also, the protective effect of SB332235 was confirmed by histological examination of both brain and liver tissues. CONCLUSIONS: Both doses (1 and 3 mg/kg) of SB332235 revealed significant hepatic/neuroprotective effects due to their anti-inflammatory, antioxidant, and antiapoptotic activities via activation of the PI3K/AKT pathway. Between the two, the 1 mg/kg dose provided significantly improved outcomes.


Subject(s)
Hepatic Encephalopathy , Neuroprotective Agents , Albumins/pharmacology , Ammonia , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Bilirubin , Caspase 3/metabolism , Hepatic Encephalopathy/chemically induced , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/metabolism , Interleukin-8/metabolism , Liver/metabolism , Male , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Superoxide Dismutase/metabolism , Thioacetamide/metabolism , Thioacetamide/toxicity , Tumor Necrosis Factor-alpha/metabolism
7.
Arch Pharm Res ; 45(3): 142-158, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35244883

ABSTRACT

The current study aims at repurposing the anti-arthritic drug diacerein (DCN) for the treatment of acetaminophen hepatotoxicity and investigating the potential underlying mechanisms. Mice were randomly divided into six groups receiving either no treatment (control group), 20 mg/kg DCN i.p, 400 mg/kg acetaminophen i.p, DCN 4 h before acetaminophen, DCN 2 h after acetaminophen, or 400 mg/kg N-acetylcysteine (NAC) i.p, 2 h after acetaminophen. Biomarkers of liver dysfunction, oxidative stress, and apoptosis were assessed. Hepatic necroinflammatory changes were evaluated along with hepatic expression of NF-κB and caspase-1. The levels of NLRP3, IL-1ß, IL-4, MCP-1, and TNF-α in the liver, as well as CYP2E1 mRNA expression, were measured. Diacerein significantly reduced biomarkers of liver dysfunction, oxidative stress, hepatocyte necrosis, and infiltration of neutrophils and macrophages whether administered 4 h before or 2 h after acetaminophen. Further, the effects were comparable to those of NAC. Diacerein also counteracted acetaminophen-induced hepatocellular apoptosis by increasing Bcl-2 and decreasing Bax and caspase-3 expression levels. Moreover, DCN normalized hepatic TNF-α and significantly decreased NF-κB p65 expression. Accordingly, DCN can prevent or reverse acetaminophen hepatotoxicity in mice, suggesting potential utility as a repurposed drug for clinical treatment.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Acetaminophen/metabolism , Acetaminophen/toxicity , Animals , Anthraquinones , Caspase 1/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Interleukin-4/metabolism , Liver/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress , Signal Transduction
8.
Environ Toxicol Pharmacol ; 91: 103833, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35218923

ABSTRACT

BACKGROUND: Ulcerative colitis (UC) is a chronic colon inflammation that is linked to exposure to environmental factors leading to improper immune responses to enteric microbes in genetically susceptible individuals. This study was designed to explore the possible protective impact of Empagliflozin (EMPA), an anti-diabetic sodium-glucose cotransporter-2 (SGLT2) inhibitor, on acetic acid (AA)-induced UC in rats. METHOD: Intrarectal instillation of AA (2 ml, 3% v/v) was used to induce UC. EMPA (10 & 30 mg/kg) was administered orally for 11 days. RESULTS: EMPA successfully counteracted AA-induced UC that was manifested by improving colonic histopathological architecture concomitant with a marked decrease in disease activity index (DAI), colon weight, weight/length ratio, serum lactate dehydrogenase (LDH) activity, and C-reactive protein (CRP) level. Additionally, EMPA successfully restored the disrupted oxidant/antioxidants balance induced by AA. Moreover, EMPA significantly induced silent information regulator-1(SIRT-1) expression along with a significant reduction in phosphatidylinositol-3-Kinase (PI3K), Protein Kinase B (AKT), nuclear factor kappa B (NF-κB), tumor necrosis factor (TNF)-α and interleukins (IL-1ß and IL-6) expression in colonic tissues. Furthermore, EMPA successfully improved the colonic barrier that was appeared from the marked induction of tight junction proteins level (occludin and claudin-1). CONCLUSION: EMPA successfully counteracted AA-induced UC in rats via the modulation of SIRT1/PI3K/AKT/NF-κB inflammatory pathway, normalizing oxidant/antioxidants balance, and improving the integrity of colon barrier.


Subject(s)
Colitis, Ulcerative , Sirtuins , Acetic Acid , Animals , Benzhydryl Compounds , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/prevention & control , Colon/metabolism , Dextran Sulfate/adverse effects , Dextran Sulfate/metabolism , Disease Models, Animal , Glucosides , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Sirtuins/metabolism
9.
Eur J Pharmacol ; 906: 174279, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34197778

ABSTRACT

Acute pancreatitis (AP) is a common pancreatic inflammation associated with substantial morbidity and mortality. AP may be mild or severe which can spread systemically causing multiple organs failure (MOF) and even death. In the current study, protocatechuic acid (PCA), a natural phenolic acid, was investigated for its possible protective potential against L-arginine induced AP and multiple organs injury (MOI) in rats. AP was induced by L-arginine (500 mg/100 g, ip). Two dose levels of PCA were tested (50 and 100 mg/kg, oral, 10 days before L-arginine injection). PCA successfully protected against L-arginine induced AP and MOI that was manifested by normalizing pancreatic, hepatic, pulmonary, and renal tissue architecture and restoring the normal values of pancreatic enzymes (amylase and lipase), serum total protein, liver enzymes (alanine transaminase (ALT) and aspartate transaminase (AST)) and kidney function biomarkers (blood urea nitrogen (BUN) and serum creatinine (Cr)) that were significantly elevated upon L-arginine administration. Additionally, PCA restored balanced oxidant/antioxidants status that was disrupted by L-arginine and normalized pancreatic levels of inducible nitric oxide synthase (iNOS) and nitric oxide (NO) content. Moreover, PCA significantly decreased L-arginine induced elevation in pancreatic high motility group box protein 1 (HMGB1), toll like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), nuclear factor kappa B (NF-κB), tumor necrosis factor- α (TNF-α), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6) expression. PCA significantly ameliorated L-arginine-induced AP and MOI through its anti-inflammatory and antioxidant effects. HMGB1/TLR4/NF-κB was the major pathway involved in the observed protective potential.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hydroxybenzoates/pharmacology , Multiple Organ Failure/prevention & control , Pancreatitis/prevention & control , Animals , Anti-Inflammatory Agents/therapeutic use , Arginine/administration & dosage , Arginine/toxicity , Disease Models, Animal , HMGB1 Protein/metabolism , Humans , Hydroxybenzoates/therapeutic use , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Liver/drug effects , Liver/immunology , Liver/pathology , Male , Multiple Organ Failure/chemically induced , Multiple Organ Failure/immunology , Multiple Organ Failure/pathology , NF-kappa B/metabolism , Pancreas/drug effects , Pancreas/immunology , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/immunology , Pancreatitis/pathology , Rats , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism
10.
J Biochem Mol Toxicol ; 35(6): 1-13, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33710703

ABSTRACT

Cyclosporine-A (CsA) is a powerful immunosuppressive agent and hepatotoxicity results from CsA treatment. This study aimed to elucidate the effectiveness of tyrosine kinase inhibitor nilotinib against CsA-induced hepatotoxicity and the underlying molecular mechanisms. Male Sprague-Dawley rats were allocated into four groups and received drugs for 28 days as follows: Control group: received vehicle, Nilotinib group: received nilotinib (20 mg/kg orally), CsA group: received CsA by subcutaneous injection (20 mg/kg daily), CsA-nilotinib: received nilotinib and CsA. Serum lactate dehydrogenase (LDH), liver function biomarkers, hepatic levels of oxidative stress biomarkers, nuclear factor erythroid-2 like-2 (Nrf2), total antioxidant capacity (TAC), interleukin-2 (IL-2), IL-1ß, IL-6, and cytochrome-C were assessed. Additionally, the protein levels and mRNA expression of Bcl2 associated X protein (Bax), caspase-3, nuclear factor-κB (NF-κB), hemoxygenase-1 (HO-1) were measured. Moreover, liver tissues were assessed histopathologically using hematoxylin-eosin and Masson trichrome stain. Nilotinib treatment decreased serum LDH, alanine aminotransferase, aspartate aminotransferase, and γ-glutamyltransferase (γ-GT), hepatic malondialdehyde, and cytochrome-C. It also increased superoxide dismutase, reduced glutathione, glutathione reductase, glutathione peroxidase, glutathione-S-transferase (GST), TAC, and Nrf2 compared to CsA-injected rats. In addition, nilotinib decreased NF-κB, IL-1ß, IL-6, Bax, and caspase-3, while elevated IL-2 and immunoexpression of HO-1. Additionally, mRNA expression of Bax and caspase-3 was elevated and that of HO-1 and inhibitory protein κB-α was reduced in the nilotinib-treated group. Moreover, nilotinib significantly attenuated CsA-induced histopathological alterations. Nilotinib may have a promising role as a hepato-protective through its antiapoptotic, antioxidant, and anti-inflammatory effects.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Chemical and Drug Induced Liver Injury/drug therapy , Cyclosporine/adverse effects , Cytochromes c/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism , Animals , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cyclosporine/pharmacology , Male , Rats , Rats, Sprague-Dawley
11.
Environ Toxicol Pharmacol ; 83: 103577, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33383195

ABSTRACT

This work explored influences of protocatechuic acid (PCA) on type 2 diabetes (T2D)-associated hepatic insulin resistance and other metabolic, hepatic and vascular irregularities using the rat model of high fat diet (HFD)+high fructose+low dose streptozotocin (STZ). Twenty-four male Wister rats were used. Twelve rats were ad libitum supplied with HFD and high fructose drinking water (25 % w/v) for 60 days. On day 30, they received a single injection of STZ (35 mg/kg, i.p). On day 32, they were divided into two subgroups (n = 6/each): T2D + PCA, received PCA (100 mg/kg/day, orally) and T2D, received PCA vehicle till the end of experiment. Rats provided with regular diet and fructose-free drinking water, with or without PCA treatment, served as PCA and control groups (n = 6/each), respectively. PCA treatment significantly reduced the elevated levels of fasting glycemia and insulin, AUCOGTT, AUCITT, and HOMA-IR index, while it boosted HOMA-ß and insulinogenic index values in T2D rats. PCA ameliorated serum lipid levels and hepatic function parameters and mitigated hepatosteatosis in T2D rats. Mechanistically, PCA mitigated hepatic lipid peroxidation and restored reduced glutathione (GSH) and superoxide dismutase (SOD) to near-normal levels. Moreover, PCA enhanced hepatic protein levels of P-AKTser473 and hepatic mRNA expression of insulin receptor substrate 1 (IRS1), phosphatidylinositol 3 kinase (PI3K)-p85 and AKT2. Furthermore, PCA ameliorated aortic oxidative stress in T2D rats, possibly via reducing serum levels of advanced glycation end products (AGEs) and diminishing vascular expression of RAGE and NOX4 mRNA. Collectively, PCA may improve hepatic insulin resistance and vascular oxidative status by modulating IRS1/PI3K/AKT2 and AGE-RAGE-NOX4 pathways, respectively.


Subject(s)
Aorta, Thoracic/drug effects , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Hydroxybenzoates/pharmacology , Insulin Resistance , Liver/drug effects , Animals , Aorta, Thoracic/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Glycation End Products, Advanced/blood , Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , Liver/metabolism , Male , NADPH Oxidase 4/genetics , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Rats, Wistar , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/drug effects
12.
Chem Biol Interact ; 315: 108863, 2020 Jan 05.
Article in English | MEDLINE | ID: mdl-31628940

ABSTRACT

Gentamicin-induced nephrotoxicity has been well documented, although the causing mechanisms and preventative measures need further investigation. The current study aimed to explore the potential protective impacts of celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor, on gentamicin-induced nephrotoxicity and the potential mechanisms in rats. Rats were randomly divided into four groups as follows: group1: normal control, group 2: received gentamicin only (100 mg/kg intraperitoneally), group 3: concurrently received gentamicin and celecoxib (30 mg/kg, orally) and group 4: received celecoxib. Celecoxib administration decreased gentamicin-induced rise in kidney weight, renal somatic index (RSI), blood urea nitrogen (BUN), serum creatinine (Cr), protein in urine, lactate dehydrogenase (LDH), nitric oxide (NOx), meanwhile, it increased serum albumin, urine Cr level and creatinine clearance (CCr), increased the renal endogenous antioxidant status, revealed by decreased malondialdehyde (MDA) and increased superoxide dismutase (SOD) and reduced glutathione (GSH). Gentamicin-induced elevated nuclear factor kappa B-P65 subunit (NFκB-p65), tumor necrosis factor-alpha (TNF-α) and apoptotic markers (tumor suppressor protein (p53) and caspase-3) protein levels were significantly decreased upon celecoxib treatment. Moreover, celecoxib suppressed renal myeloperoxidase (MPO) activity and posed improvement of histological features. In immunohistochemistry, celecoxib-treated rats showed decreased immunoreactivity against COX-2 in tubular cells and a mild positive immunoreactivity against heat shock protein 70 (HSP70) in renal interstitial cells. These findings propose that celecoxib treatment mitigates renal dysfunction via decreasing renal inflammation, oxidative/nitrosative stress, and apoptosis.


Subject(s)
Caspase 3/metabolism , Celecoxib/pharmacology , Kidney Diseases/drug therapy , Kidney/drug effects , NF-kappa B/metabolism , Protective Agents/pharmacology , Signal Transduction/drug effects , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Blood Urea Nitrogen , Creatinine/metabolism , Cyclooxygenase 2/metabolism , Gentamicins/pharmacology , Glutathione/metabolism , HSP70 Heat-Shock Proteins/metabolism , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Male , Malondialdehyde/metabolism , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Transcription Factor RelA/metabolism
13.
Naunyn Schmiedebergs Arch Pharmacol ; 392(2): 243-258, 2019 02.
Article in English | MEDLINE | ID: mdl-30460386

ABSTRACT

Type 2 diabetes mellitus (T2D) is a chronic metabolic disorder considered to be the most predominant form of diabetes throughout the world. This study aimed to investigate the possible effects of cinnamaldehyde (CIN) on insulin signaling pathways in STZ-induced T2D rat model. T2D was originated by feeding rats with a high-fat diet (HFD) plus 25% fructose solution plus streptozotocin (STZ) (35 mg/kg, i.p.). CIN effects were investigated on fasting blood glucose, insulin, oral glucose tolerance test (OGTT), insulin tolerance test (ITT), liver biomarkers, lipid profile, oxidative stress biomarkers, serum advanced glycation end products (AGEs) and its receptors (RAGE) in the aorta, and histopathology of the liver and aorta. Additionally, the mRNA expression of hepatic insulin signaling pathway genes, phosphorylated AKT (serine 473) (P-AKT ser473) level, and aortic nitric oxide synthase3 (eNOS) and NADPH oxidase4 (NOX4) were determined. CIN treatment for 30 days significantly decreased OGTT, ITT, fasting blood glucose, insulin, and HOMA-IR and increased HOMA-ß index when compared to diabetic rats. CIN also improved lipid profile and decreased serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activity, serum AGEs, and aortic RAGE. Additionally, CIN treatment significantly decreased hepatic malondialdehyde (MDA), increased hepatic and aortic glutathione (GSH) and superoxide dismutase (SOD), and decreased steatosis and inflammation observed in liver tissue of rats. Additionally, significant elevation in mRNA expression of insulin receptor substrate1 (IRS1), phosphatidylinositol 3-kinase regulatory subunit1 (PI3K-P85 subunit), and AKT serine/threonine kinase2 (AKT2); increased levels of P-AKT ser473 and aortic eNOS; and decrease in mRNA expression of NOX4 were detected in CIN-treated group when compared to diabetic group. This study suggests antidiabetic and antioxidant effects of CIN probably through upregulation of eNOS and IRS1/PI3K/AKT2 signaling pathway and alleviating AGEs, RAGE, and NOX4 elevation.


Subject(s)
Acrolein/analogs & derivatives , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/therapeutic use , Acrolein/pharmacology , Acrolein/therapeutic use , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat , Fructose/administration & dosage , Gene Expression Regulation/drug effects , Glycation End Products, Advanced/blood , Hypoglycemic Agents/pharmacology , Insulin Receptor Substrate Proteins/genetics , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Nitric Oxide Synthase Type III/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Rats, Wistar , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/drug effects
14.
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.

15.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...