Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Chem Biol Interact ; 366: 110139, 2022 Oct 01.
Article En | MEDLINE | ID: mdl-36057361

Gentamicin (GEN) possesses a broad range of antimicrobial effects. However, it belongs to the aminoglycosides, and has the greatest potential for nephrotoxic effect above all other antibiotics from this group. This study aims to evaluate the possible protective effect of lipoxin A4 (LXA4) against GEN-induced nephrotoxicity in rats. Nephrotoxicity was induced in male Wistar rats by injection of GEN (80 mg/kg/day, i.p.) for 6 days starting from day 7 of the experiment. Rats were treated with either LXA4 (10 µg/kg/day, i.p.) or LXA4 (50 µg/kg/day, i.p.) for 14 days starting from day 1 of the experiment, along with GEN as the previous schedule. GEN resulted in a significantly elevated renal function in the form of higher serum creatinine and urea levels. Further, GEN induced abnormal renal histopathology including degenerated glomeruli and tubules, with perivascular inflammation and hemorrhage. All of these findings were significantly decreased by the LXA4 administration. Additionally, LXA4 remarkably reduced the increased serum lipid biomarkers. Moreover, LXA4 significantly inhibited the GEN-induced oxidative stress in the kidneys by decreasing the elevated levels of renal malondialdehyde (MDA) and total nitrite while raising the suppressed levels of renal superoxide dismutase (SOD) and serum total antioxidant capacity (TAC). LXA4 inhibited the up-regulated inflammatory mediators ICAM-1, TGFß 1 protein levels, and TNF-α protein expression. Finally, LXA4 suppressed the elevated apoptotic mediators; p-JNK and cleaved caspase-3 expression. Our results proved for the first time that LXA4 ameliorated GEN-induced nephrotoxicity through its antioxidant, anti-inflammatory and anti-apoptotic properties.


Anti-Infective Agents , Lipoxins , Aminoglycosides , Animals , Anti-Bacterial Agents/toxicity , Antioxidants , Caspase 3 , Creatinine , Gentamicins/toxicity , Inflammation Mediators , Intercellular Adhesion Molecule-1 , Male , Malondialdehyde/metabolism , Nitrites , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Transforming Growth Factor beta , Tumor Necrosis Factor-alpha , Urea
2.
Biomed Pharmacother ; 145: 112374, 2022 Jan.
Article En | MEDLINE | ID: mdl-34915671

Hepatic ischemia-reperfusion (I/R) is an important cause of liver damage in many clinical situations. Toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) is an inflammatory pathway activated in hepatic I/R injury. Telmisartan, a selective angiotensin II type 1 receptor antagonist and peroxisome proliferator-activated receptor-gamma (PPARγ) partial agonist, can inhibit the expression of pro-inflammatory cytokines. The present work investigated the possible protective effect of telmisartan against hepatic I/R injury and explored its possible mechanisms in rats. Rats were divided into four equal groups: sham-operated control, telmisartan-treated sham-operated control, I/R untreated, and I/R telmisartan-treated groups. Hepatic injury was evaluated biochemically by serum activity of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and histopathological examination. Hepatic oxidative stress biomarkers, myeloperoxidase level, PPARγ and TLR4 mRNA expression, and NF-κB and active caspase 3 immunoexpression were determined. The study showed that telmisartan attenuated hepatic I/R, as evidenced by decreased serum ALT and AST activities and confirmed by improvement of the histopathological changes. The protective effect of telmisartan was associated with modulation of oxidative stress parameters, myeloperoxidase level, PPARγ and TLR4 mRNA expression, and NF-κB and caspase 3 immunoexpression. Taken together, the current study showed that telmisartan could protect the rat liver from I/R injury. This hepatoprotective effect was attributed to, at least in part, increase in PPARγ expression and suppression of TLR4/NF-κB pathway.


Liver/metabolism , NF-kappa B/metabolism , PPAR gamma/metabolism , Reperfusion Injury/prevention & control , Telmisartan/pharmacology , Toll-Like Receptor 4/metabolism , Alanine Transaminase/blood , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Aspartate Aminotransferases/blood , Caspase 3/metabolism , Cytokines/metabolism , Male , Oxidative Stress , Rats , Rats, Wistar
3.
Biomed Pharmacother ; 141: 111902, 2021 Sep.
Article En | MEDLINE | ID: mdl-34328119

BACKGROUND: Inflammatory bowel disease is defined as chronic noninfectious inflammation of the gastrointestinal tract, including ulcerative colitis and Crohn's disease. Its incidence and predominance have increased globally, with no effective agents for preventing its recurrence or treatment until now. AIM: The current study aimed to investigate the possible role of canagliflozin (CANA), a sodium-glucose co-transporter-2 inhibitor (SGLT-2), to prevent and treat acetic acid (AA)-induced colitis in a rat model. METHODS: Colitis was induced in male Wistar rats by intrarectal instillation of 1 ml of 4% (v/v) AA. Rats were treated orally with either CANA (30 mg/kg/day, p.o.) for 10 days before or after colitis induction or sulfasalazine (360 mg/kg/day, p.o.) for 10 days before colitis induction. RESULTS: AA resulted in a significant increase in disease activity index, colonic weight over length ratio, colon macroscopic damage score, and histological signs of colitis. All of these effects were significantly decreased by CANA administration. Additionally, CANA markedly inhibited AA-induced oxidative stress and inflammatory responses by significantly reducing the up-regulated levels in malondialdehyde, total nitrite, NF-κB, interleukin-1ß, and TNF-α, and significantly increasing the down-regulated levels in reduced glutathione, superoxide dismutase, and interleukin-10. CANA significantly inhibited caspase-3 level while rescued survivin expression in colons. Finally, CANA reduced the elevated levels of pyruvic acid and G6PDH activity, as well as the levels of p22phox and NOX2 in the AA-induced colitis. CONCLUSION: Our findings provide novel evidence that CANA has protective and therapeutic effects against AA-induced colitis by the impact of its antioxidant, anti-inflammatory, and anti-apoptotic effects.


Canagliflozin/therapeutic use , Colitis/drug therapy , Glucose/metabolism , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Acetic Acid , Animals , Antioxidants/metabolism , Colitis/chemically induced , Colitis/metabolism , Colon/pathology , Male , NADPH Oxidase 2/metabolism , Organ Size , Oxidative Stress/drug effects , Rats , Rats, Wistar , Sulfasalazine/therapeutic use
4.
Arch Pharm Res ; 43(5): 514-525, 2020 May.
Article En | MEDLINE | ID: mdl-32436127

Ischemic stroke is a serious condition associated with severe functional disability and high mortality, however; effective therapy remains elusive. Empagliflozin, a sodium-glucose cotransporter 2 inhibitor, has been shown to exert additional non-glycemic benefits including anti-apoptotic effects in different disease settings. Thereby, this study was designed to investigate the ameliorative effect of empagliflozin on the neuronal apoptosis exhibited in cerebral ischemia/reperfusion (I/R) in a rat model targeting HIF-1α/VEGF signaling which is involved in this insult. Global cerebral I/R injury was induced in male Wistar rats through occlusion of the bilateral common carotid arteries for 30 min followed by one-hour reperfusion. Empagliflozin doses of 1 and 10 mg/kg were administered 1 and 24 h after reperfusion. In I/R-injured rats, empagliflozin treatments significantly reduced infarct size and enhanced neurobehavioral functions in a dose-dependent manner. The drug alleviated neuronal death and cerebral injury inflicted by global ischemia as it suppressed neuronal caspase-3 protein expression. In parallel, protein expressions of HIF-1α and its downstream mediator VEGF were upregulated in the ischemic brain following empagliflozin treatment. The results indicated that empagliflozin attenuates cerebral I/R-induced neuronal death via the HIF-1α/VEGF cascade.


Apoptosis/drug effects , Benzhydryl Compounds/pharmacology , Brain Ischemia/drug therapy , Glucosides/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Reperfusion Injury/drug therapy , Signal Transduction/drug effects , Vascular Endothelial Growth Factors/metabolism , Administration, Oral , Animals , Benzhydryl Compounds/administration & dosage , Brain Ischemia/metabolism , Brain Ischemia/pathology , Disease Models, Animal , Glucosides/administration & dosage , Male , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
5.
Mediators Inflamm ; 2020: 8641026, 2020.
Article En | MEDLINE | ID: mdl-32104151

Methotrexate (MTX) is a commonly used chemotherapeutic agent. Oxidative stress and inflammation have been proved in the development of MTX toxicity. Paeonol is a natural phenolic compound with various pharmacological activities including antioxidant and anti-inflammatory properties. The aim of the present study was to evaluate the protective effect of paeonol against MTX-induced cardiac toxicity in rats and to evaluate the various mechanisms that underlie this effect. Paeonol (100 mg/kg) was administered orally for 10 days. MTX cardiac toxicity was induced at the end of the fifth day of the experiment, with or without paeonol pretreatment. MTX-induced cardiac damage is evidenced by a distortion in the normal cardiac histological structure, with significant oxidative and nitrosative stress shown as a significant increase in NADPH oxidase-2, malondialdehyde, and nitric oxide levels along with a decrease in reduced glutathione concentration and superoxide dismutase activity compared to the control group. MTX-induced inflammatory effects are evidenced by the increased cardiac toll-like receptor 4 (TLR4) mRNA expression and protein level as well as increased cardiac tumor necrosis factor- (TNF-) α and interleukin- (IL-) 6 levels along with increased nuclear factor- (NF-) κB/p65 immunostaining. MTX increased apoptosis as shown by the upregulation of cardiac caspase 3 immunostaining. Paeonol was able to correct the oxidative and nitrosative stress as well as the inflammatory and apoptotic parameters and restore the normal histological structure compared to MTX alone. In conclusion, paeonol has a protective effect against MTX-induced cardiac toxicity through inhibiting oxidative and nitrosative stress and suppressing the TLR4/NF-κB/TNF-α/IL-6 inflammatory pathway, as well as causing an associated reduction in the proapoptotic marker, caspase 3.


Acetophenones/therapeutic use , Cardiotoxicity/drug therapy , Cardiotoxicity/metabolism , Methotrexate/toxicity , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Animals , Caspase 3/metabolism , Immunohistochemistry , Male , Oxidative Stress/drug effects , Rats , Rats, Wistar , Signal Transduction/drug effects , Transcription Factor RelA/metabolism
6.
Fundam Clin Pharmacol ; 34(5): 548-558, 2020 Oct.
Article En | MEDLINE | ID: mdl-32068294

Hyperglycemia is one of the ischemic neuronal damage triggers that exacerbate the response to oxidative stress, inflammation, and apoptosis induced by cerebral ischemia/reperfusion (I/R) injury. Empagliflozin, a sodium-glucose cotransporter 2 (SGLT 2) inhibitor, was shown to effectively reduce hyperglycemia and glucotoxicity besides improving glycemic control in diabetics. Therefore, the present study was conducted to investigate the neuroprotective effect of empagliflozin against cerebral I/R injury in hyperglycemic rats. Hyperglycemia was induced by streptozotocin (55 mg/kg), and transient cerebral I/R was induced by bilateral common carotid occlusion for 30 min followed by 24-h reperfusion. Either empagliflozin (10 mg/kg; i.p.) or gliclazide (2 mg/kg, p.o.) was administered at 1 and 24 h after reperfusion. Treatment with empagliflozin showed a significant amelioration of behavioral/neurological functions and histopathological changes observed in brain tissues of hyperglycemic rats subjected to cerebral I/R injury. Comparable to gliclazide, empagliflozin decreased cerebral infarct volume along with suppression of cerebral oxidative stress, inflammatory, and apoptotic markers in brain tissues of hyperglycemic I/R-injured rats. These findings suggested that empagliflozin can significantly alleviate neuronal damage resulting from global I/R injury induced in hyperglycemic rats. The proposed neuroprotective effect of empagliflozin may be attributed to its glycemic control effect and related antioxidant, anti-inflammatory, and antiapoptotic effects.


Benzhydryl Compounds/therapeutic use , Glucosides/therapeutic use , Hyperglycemia , Ischemic Attack, Transient/prevention & control , Neuroprotective Agents/therapeutic use , Animals , Apoptosis/drug effects , Benzhydryl Compounds/pharmacology , Disease Models, Animal , Gliclazide/pharmacology , Gliclazide/therapeutic use , Glucosides/pharmacology , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Inflammation/metabolism , Male , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Rats , Rats, Wistar
7.
Fundam Clin Pharmacol ; 34(3): 302-312, 2020 Jun.
Article En | MEDLINE | ID: mdl-31854010

Ischemic heart disease is a common cardiac health problem. Despite the significant advances in prevention and treatment of this disorder, its incidences and complications are very serious. So, the search for more antioxidants and anti-inflammatory agents with cardioprotective effects is an urgent task. We aimed to evaluate the effects of a heme oxygenase 1 (HO1) inducer, hemin (HEM), on isoprenaline (ISO)-induced myocardial damage. Forty-five Wistar albino rats were used. Animals were treated with HEM (25 mg/kg/day) i.p. for 5 days and injected with ISO (150 mg/kg/day) i.p. on 4th and 5th day of the experiment. Detection of the role of ATP-sensitive potassium channel (KATP ) was performed by administration of glibenclamide (GP) (5 mg/kg/day) orally 2 h before HEM. Moreover, the role of endothelial nitric oxide synthase (eNOS) was detected by coadministration of Nitro- ω-L-arginine (L-NNA) (25 mg/kg/day) for 5 days. The ISO group showed increase in heart weight, cardiac enzymes, tumor necrosis factor alpha (TNFα), and malondialdehyde (MDA) with decrease in reduced glutathione (GSH), HO1, and total antioxidant capacity (TAC). In addition, there were increases in Bcl-2 associated X protein (Bax) and cleaved caspase-3, but decreases in B-cell lymphoma-2 (Bcl-2) and eNOS. Moreover, the histopathological examination of the ISO group showed degeneration of the cardiac muscle fibers and marked infiltration of the inflammatory cells. The biochemical and histopathological changes induced by ISO were markedly ameliorated in the HEM plus ISO group. This protective effect was diminished with coadministration of GP or L-NNA; thus, KATP and eNOS might mediate HEM cardioprotection.


Cardiotonic Agents/pharmacology , Hemin/pharmacology , KATP Channels/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Nitric Oxide Synthase Type III/metabolism , Animals , Antioxidants/metabolism , Caspase 3/metabolism , Heme Oxygenase-1/metabolism , Isoproterenol/pharmacology , Male , Malondialdehyde/metabolism , Myocardial Infarction/chemically induced , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Rats , Rats, Wistar
8.
Fundam Clin Pharmacol ; 32(6): 617-626, 2018 Dec.
Article En | MEDLINE | ID: mdl-29862557

Nearly all men who reach average life expectancy have prostate disease. The most common is benign prostatic hyperplasia (BPH). Peroxisome proliferator-activated receptor alpha (PPARα) had protective effect in different models, but still, there are no studies explain its role in BPH. So that we investigated the effect of fenofibrate (FEN) on induced BPH by testosterone propionate (TP) (3 mg/kg/day for 4 weeks) subcutaneous injection followed by FEN (300 mg/kg/day) was given orally for 4 weeks. We measured prostate weights changes, prostatic tissue superoxide dismutase (SOD), and malondialdehyde (MDA) levels. Prostate-specific antigen (PSA), dihydrotestosterone (DHT), and total antioxidant capacity (TAC) in serum were determined. The mRNA gene expressions of proliferating cell nuclear antigen (PCNA), PPARα, and glutathione peroxidase (GPx) in prostatic tissue were also measured by quantitative real-time polymerase chain reaction. In addition, the histopathological changes and activated caspase3 immunoexpression were evaluated. Our results showed that TP succeeded in induction of BPH, which was detected by significant increase in prostate weights, prostatic tissue MDA, serum levels of DHT, PSA, and mRNA gene expression of PCNA but significant decrease in PPARα and GPx gene expression. Moreover, TAC in serum and SOD level in prostate tissue decreased. The histopathological examination showed typical changes of BPH with dysplastic changes with marked decrease in activated caspase3 immunoexpression indicating marked suppression of the apoptotic process. FEN significantly improved all disturbed parameters of BPH model. Moreover, there are no dysplastic changes with co-administration of FEN to BPH induced group.


Fenofibrate/pharmacology , PPAR alpha/agonists , Prostate/drug effects , Prostatic Hyperplasia/drug therapy , Animals , Antioxidants/metabolism , Dihydrotestosterone/metabolism , Glutathione Peroxidase/metabolism , Male , Malondialdehyde/metabolism , Prostate/metabolism , Prostate-Specific Antigen/metabolism , Prostatic Hyperplasia/metabolism , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Testosterone/metabolism
9.
J Toxicol ; 2017: 9815853, 2017.
Article En | MEDLINE | ID: mdl-29279713

Tramadol is a synthetic, centrally acting analgesic. It is the most consumed narcotic drug that is prescribed in the world. Tramadol abuse has dramatically increased in Egypt. Long term use of tramadol can induce endocrinopathy. So, the aim of this study was to analyze the adrenal insufficiency induced by long term use of tramadol in experimental animals and also to assess its withdrawal effects through histopathological and biochemical genetic study. Forty male albino rats were used in this study. The rats were divided into 4 groups (control group, tramadol-treated group, and withdrawal groups). Tramadol was given to albino rats at a dose of 80 mg/kg body weight for 3 months and after withdrawal periods (7-15 days) rats were sacrificed. Long term use of tramadol induced severe histopathological changes in adrenal glands. Tramadol decreased the levels of serum cortisol and DHEAS hormones. In addition, it increased the level of adrenal MDA and decreased the genetic expression of glutathione peroxidase and thioredoxin reductase in adrenal gland tissues. All these changes started to return to normal after withdrawal of tramadol. Thus, it was confirmed that long term use of tramadol can induce severe adrenal insufficiency.

10.
J Toxicol ; 2014: 489382, 2014.
Article En | MEDLINE | ID: mdl-25120567

Gentamicin, an aminoglycoside antibiotic, is used for the treatment of serious Gram-negative infections. However, its usefulness is limited by its nephrotoxicity. Sildenafil, a selective phosphodiesterase-5 inhibitor, was reported to prevent or decrease tissue injury. The aim of this study is to evaluate the potential protective effects of sildenafil on gentamicin-induced nephrotoxicity in rats. Male Wistar rats were injected with gentamicin (100 mg/kg/day, i.p.) for 6 days with and without sildenafil. Sildenafil administration resulted in nephroprotective effect in gentamicin-intoxicated rats as it significantly decreased serum creatinine and urea, urinary albumin, and renal malondialdehyde and nitrite/nitrate levels, with a concomitant increase in renal catalase and superoxide dismutase activities compared to gentamicin-treated rats. Moreover, immunohistochemical examination revealed that sildenafil treatment markedly reduced inducible nitric oxide synthase (iNOS) expression, while expression of endothelial nitric oxide synthase (eNOS) was markedly enhanced. The protective effects of sildenafil were verified histopathologically. In conclusion, sildenafil protects rats against gentamicin-induced nephrotoxicity possibly, in part, through its antioxidant activity, inhibition of iNOS expression, and induction of eNOS production.

11.
PPAR Res ; 2014: 626319, 2014.
Article En | MEDLINE | ID: mdl-24803924

Hepatoprotective potential of peroxisome proliferator activator receptor (PPAR)- α and - γ agonists, fenofibrate (FEN), and pioglitazone (PIO), respectively, against cyclophosphamide (CP)-induced toxicity has been investigated in rat. FEN and PIO (150 and 10 mg/kg/day, resp.) were given orally for 4 weeks. In separate groups, CP (150 mg/kg, i.p.) was injected as a single dose 5 days before the end of experiment, with or without either PPAR agonist. CP induced hepatotoxicity, as it caused histopathological alterations, with increased serum alanine and aspartate transaminases, total bilirubin, albumin, alkaline phosphatase and lactate dehydrogenase. CP caused hepatic oxidative stress, indicated by decrease in tissue reduced glutathione, with increase in malondialdehyde and nitric oxide levels. CP also caused decrease in hepatic antioxidant enzyme levels, including catalase, superoxide dismutase, glutathione peroxidase, and glutathione S-transferase. Furthermore, CP increased serum and hepatic levels of the inflammatory marker tumor necrosis factor (TNF)- α , evaluated using ELISA. Preadministration of PIO, but not FEN, prior to CP challenge improved hepatic function and histology, and significantly reversed oxidative and inflammatory parameters. In conclusion, activation of PPAR- γ , but not PPAR- α , conferred protection against CP-induced hepatotoxicity, via activation of antioxidant and anti-inflammatory mechanisms, and may serve as supplement during CP chemotherapy.

12.
Environ Toxicol Pharmacol ; 37(2): 772-81, 2014 Mar.
Article En | MEDLINE | ID: mdl-24632013

The anticancer drug; doxorubicin (DOX), causes testicular toxicity as an adverse effect. P-glycoprotein (P-gp) is a multidrug resistance efflux transporter expressed in blood-testis barrier, which extrudes DOX from the testis. We investigated whether DOX-induced gonadal injury could be prevented by the use of antioxidant; coenzyme-Q10 (CoQ10). The involvement of P-gp expression, as a possible protective mechanism, was also investigated. CoQ10 was administered orally for 8 days, and DOX toxicity was induced via a single i.p. dose of 15 mg/kg at day 4. Concomitant administration of CoQ10 with DOX significantly restored testicular oxidative stress parameters and the distorted histopathological picture, reduced the up-regulation of caspase 3 caused by DOX, and increased P-gp expression. We show for the first time that CoQ10 up-regulates P-gp as a novel mechanism for gonadal protection. In conclusion, CoQ10 protects against DOX-induced testicular toxicity in rats via ameliorating oxidative stress, reducing apoptosis and up-regulating testicular P-gp.


ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Protective Agents/pharmacology , Testis/drug effects , Ubiquinone/analogs & derivatives , Animals , Apoptosis/drug effects , Body Weight/drug effects , Catalase/metabolism , Glutathione/metabolism , Male , Malondialdehyde/metabolism , Nitrates/metabolism , Nitrites/metabolism , Organ Size/drug effects , Rats, Wistar , Testis/metabolism , Testis/pathology , Ubiquinone/pharmacology , Up-Regulation
13.
Adv Pharmacol Sci ; 2013: 387071, 2013.
Article En | MEDLINE | ID: mdl-24381587

Methotrexate is an effective anticancer and immunosuppressive agent. However, nephrotoxicity is one of the complications of its use. On the other hand, curcumin, a naturally occurring polyphenolic compound, is reported to have antioxidant and anti-inflammatory properties. Those two properties are likely to prevent methotrexate-induced nephrotoxicity. The aim of this study is to evaluate the possible protective effect of curcumin against methotrexate-induced nephrotoxicity and delineate various mechanism(s) underlies this effect in rats. Nephrotoxicity was induced in Wistar rats by intraperitoneal administration of methotrexate (7 mg/kg/day) for three consecutive days. Curcumin administration in methotrexate-intoxicated rats resulted in nephroprotective effects as evidenced by the significant decrease in levels of serum creatinine and urea as well as renal malondialdehyde, nitric oxide, and tumor necrosis factor- α with a concurrent increase in renal glutathione peroxidase and superoxide dismutase activities compared to nephrotoxic untreated rats. Additionally, immunohistochemical analysis demonstrated that curcumin treatment markedly reduced cyclooxygenase-2 expression. Histopathological examination confirmed the protective effects of curcumin. In conclusion, curcumin protected rats from methotrexate nephrotoxicity, at least in part, through its antioxidant and anti-inflammatory activities.

14.
Adv Pharmacol Sci ; 2012: 981461, 2012.
Article En | MEDLINE | ID: mdl-23346106

Nephrotoxicity is one of the limiting factors for using doxorubicin (Dox) as an anticancer chemotherapeutic. Here, we investigated possible protective effect of coenzyme-Q10 (CoQ10) on Dox-induced nephrotoxicity and the mechanisms involved. Two doses (10 and 100 mg/kg) of CoQ10 were administered orally to rats for 8 days, in the presence or absence of nephrotoxicity induced by a single intraperitoneal injection of Dox (15 mg/kg) at day 4 of the experiment. Our results showed that the low dose of CoQ10 succeeded in reversing Dox-induced nephrotoxicity to control levels (e.g., levels of blood urea nitrogen and serum creatinine, concentrations of renal reduced glutathione (GSH) and malondialdehyde, catalase activity and caspase 3 expression, and renal histopathology). Alternatively, the high dose of CoQ10 showed no superior nephroprotection over the low dose, as there were no significant improvements in renal histopathology, catalase activity, or caspase 3 expression compared to the Dox-treated group. Interestingly, the high dose of CoQ10 alone significantly decreased renal GSH level as well as catalase activity and caused a mild induction of caspase 3 expression compared to control, probably due to a prooxidant effect at this dose of CoQ10. We conclude that CoQ10 protects from Dox-induced nephrotoxicity with a precaution to dosage adjustment.

...