Review Article

Potential Protective Effects of Antioxidants against Cyclophosphamide-Induced Nephrotoxicity

Table 1

Protective role of antioxidants against cyclophosphamide-induced nephrotoxicity.

ReferencesAnimal UsedMethod and InterventionMajor Findings

[31]Male Wistar rats (n = 35)Rats were pretreated with naringin (NG) (50 and 100 mg/kg/day), for 7 days. Then, single dose of CP (200 mg/kg b.w.) was administering on the seventh day.(i) ↓ levels of serum urea and creatinine
(ii) ↓ levels of renal iNOS, COX-2, LC3B and significantly modulated renal NF-κB, TNF-α, IL-1β, and IL-6
(iii) ↑ levels of renal SOD, CAT, GPx, GSH
(iv) The protective effects of NG is possibly due to the mitigation of apoptosis, autophagy, inflammation, oxidative stress, and oxidative DNA damage.
[32]Male Swiss albino mice (n = 25)The mice were pretreated with ellagic acid (EA), orally at a dose of 50 and 100 mg/kg for 7 consecutive days before the administration of a single intraperitoneal (i.p.) injection of CP at 50 mg/kg.(i) EA pretreatment restored the activities of antioxidant enzymes
(ii) ↓ micronuclei formation, and DNA fragmentation
(iii) ↓ levels of serum BUN, creatinine and LDH
[33]Female Wistar albino rats (n = 18)Spirulina was administered orally (1000 mg/kg bw/day) for 7 days, then a single dose of CP was injected i.p. (150 mg/kg) on the seventh day of the experiment.(i) ↑ Tissue levels of SOD and CAT
(ii) ↓ MDA levels and apoptosis
(iii) The protective effect of spirrulina might be due to its antioxidant and antiapoptotic properties.
[34]Male Wistar rats (n = 40)Rats were pretreated with vitamin E (100 mg/kg) for 3 days and then, a single dose of CP (150 mg/kg, i.p) was administered.(i) ↓ TNF-α and MDA production
(ii) ↑ GPx and MDA levels in the renal tissues
(iii) The protective effect might be due to the antioxidant activity of vitamin E.
[35]Male Wistar rats (n = 36)To induce nephrotoxicity, CP (150 mg/kg) was administered through i.p. route.
Murraya koenigii extract (100 mg/kg and 200 mg/kg i.p.) was administered and oxidative stress parameters were measured.
(i) ↓ BUN, creatinine and LPO levels
(ii) ↑ SOD, GSH levels
(iii) Murraya koenigii extract showed protective effect against CP-induced nephrotoxicity
[36]Male Swiss albino mice (n = 40)Mice were treated with Capparis spinosa extract (CSE) orally in doses of 100, 200, and 400 mg/kg, for 5 consecutive days, and CP (200 mg/kg, i.p) was administered on the fifth day, 1 hour after the last dose of extract administration.(i) ↓ BUN, creatinine and MDA levels.
(ii) ↑ GSH levels
(iii) Histological examination also confirmed the protective effect of CSE.
[37]Male Swiss albino mice (n = 50)Rats were pretreated with different doses of melatonin (MEL) (5, 10, and 20 mg/kg/day, i.p) for 5 days, then CP (200 mg/kg, i.p) was administered 1 hour after the last MEL on day 5.(i) ↓ BUN and Creatinine levels
(ii) ↓ MDA, PC, NO levels and MPO activity
(iii) ↑ GSH, SOD, GPx and CAT activity
(iv) Attenuated CP-induced histological changes of the kidney tissues
[38]Male Swiss albino mice (n = 30)Rats were treated with Elaeagnus angustifolia fruit extract (EAFE) orally at doses of 100, 200, and 400 mg/kg, respectively, for 5 consecutive days and CP (200 mg/kg, i.p.) on the 5th day, 1 hour after the last dose of the extract.(i) ↓ MDA, creatinine, and BUN levels
(ii) ↑ GSH level
(iii) Histological evaluation of the kidneys also showed protective effect
[39]Male BALB/c mice (n = 32)Rats were administered with cerium oxide nanoparticles (NC) at a dose of 100 μg/kg i.p. for 3 consecutive days. And, a single dose of CP (200 mg/kg, i.p) was administered on the third day. On the sixth day of the experiment, various parameters were evaluated.(i) ↓ MDA, urea and creatinine levels• ↑ GSH level
(ii) In addition, NC pre-treatment alleviated immunoreactivity of caspase-3and it showed strong antioxidant activity.
[40]Male Parke’s strain mice (n = 24)CP (200 mg/kg, i.p) was administered (once in a week) for five weeks and also aqueous extract of Phyllanthus fraternus (AEPF) (400 mg/kg) for 5 weeks (once in a week) orally.(i) ↑ SOD and CAT activity
(ii) ↓ MDA levels
(iii) Restored the percentage of the kidney somatic index
[41]Male Wistar rats (n = 30)Rats were treated with 100 and 200 mg/kg Olea europaea leaf extract (OLE) orally for 15 days and a single i.p injection of 150 mg/kg CP at day 16.(i) ↓ BUN and creatinine levels
(ii) ↑ Bcl-2 expression
(iii) ↓ NF-κB, Bax, cytochrome c and caspase-3
(iv) Upregulated Nrf2/ARE/HO-1 signaling
(v) Enhanced the antioxidant activity, attenuating inflammation and apoptosis.
[42]Male Swiss albino mice (n = 35)Mice were pretreated with Lavandula officinalis L extract (LOE) orally at 100, 200, and 400 mg/kg for five consecutive days and CP (200 mg/kg, i.p.) was administrated on the fifth day, 1 hour after the last dose of the extract.(i) ↓ MDA, BUN, and creatinine levels
(ii) ↑ GSH, SOD, and CAT activity
(iii) Histological examination of the kidneys also revealed the protective effect of LOE.
[43]Male Sprague-Dawley rats (n = 42)Rats were treated with carvacrol (CAR) at 5 and 10 mg/kg for 6 consecutive days, and CP (100 mg/kg) was administered at the fourth day.(i) ↓ MDA, TOS, and OSI levels
(ii) ↑ GSH, SOD, CAT, and TAC levels
(iii) Lower tissue damage
[44]Male Swiss Albino mice (n = 48, for each models)To induce acute kidney injury, single dose of CP was administered at the dose of 75 mg/kg i.p., whereas the subacute kidney injury was induced by daily injection of CP (50 mg/kg i.p) for 1 week. Mice were treated with tranilast (300 mg/kg, orally) for 8 days in acute injury. In subacute kidney injury, mice were treated with tranilast for the first 7 days and then tranilast (300 mg/kg, orally) + CP (50 mg/kg, I.P.) for the successive 7 days.(i) ↓ BUN, creatinine, TNF-α, LDH, total kidney protein contents, and lipid peroxidation
(ii) ↑ SOD activity and GSH content
(iii) This protective effect might be through enhancing antioxidant defense mechanisms, decreasing cytotoxicity, and decreasing expression of inflammatory cytokines.
[45]Male albino Wistar rats (n = 35)Rats were treated with Mangifera indica L. (MPS) (500, 1000 mg/kg, p.o.) and/or silymarin (150 mg/kg, p.o.) for 10 days. In the last 5 days of treatment, rats were administered CP (150 mg/kg, i.p).(i) ↑ GSH and SOD activity
(ii) ↓ Total MDA and GST
(iii) The antioxidant effect of MPS and/or silymarin might be responsible for the kidney protection.
[46]Male Sprague-Dawley rats (n = 66)Rats were pretreated with single dose of Wuzhi capsule (WZC) (300 mg/kg), 15 minutes before receiving CP injection. One hour after the injections, all rats were injected with MESNA (420 mg/kg) to prevent possible bladder injuries.(i) ↑ GSH, GPx, and SOD contents/or activities in both tissues and plasma
(ii) ↓ BUN, creatinine, and MDA
(iii) Improved morphology and pathology of the kidneys
[47]Male Wistar albino rats (n = 40)To induce renal toxicity, CP (200 mg/kg, i.p.) was administered as a single dose on first day of the experimental period, followed by the administration of taurine (200 mg/kg, i.p.) daily for 3 weeks.(i) Serum activities of creatine kinase, creatine kinase isoenzyme, LDH, creatinine as well as BUN disturbances were attenuated
(ii) CP-induced ECG changes were significantly reversed
[48]Either sex Wistar rats (n = 30)Rats were treated with daidzein, (20 and 40 mg/kg, p.o.) for 10 days and administered with CP (150 mg/kg, i.p.) in the last 5 days.(i) ↓ MDA level
(ii) ↑ GSH, SOD, and CAT levels
(iii) Improved the structural architecture of renal profiles
[49]Male Sprague-Dawley rats (n = 42)Rats were administered with 0.5 or 1 mg/kg selenium for 6 consecutive days and then a single dose of CP (150 mg/kg, i.p.) was administered on the sixth day.(i) Decreased creatinine levels in a dose-dependent manner. But, creatinine levels remained high relative to the control group, which indicates that selenium cotreatment might be partially effective.
(ii) Cotreatment with selenium 1 mg/kg resulted in a better improvement of oxidative stress markers
[50]Male Wistar albino rats (n = 35)The rats were pretreated with chrysin (CH) orally in doses of 25 and 50 mg/kg for 7 consecutive days, and CP (200-mg/kg, i.p.) was administrated on the 7th day, 1 h after the last dose of CH.(i) ↓ Urea, creatinine, MDA, and renal deterioration
(ii) ↑ SOD, CAT, GPx, and GSH levels
(iii) Furthermore, CH reversed the changes in levels of inflammatory, apoptotic, and autophagic parameters such as NF-κB, TNF-α, IL-1β, IL-6, iNOS, COX-2, Bax, Bcl-2, and LC3B in kidney tissues.
[51]Albino rats (n = 60)Rats were pretreated with N-acetylcysteine (NAC) (10 mg/kg/d), melatonin (MT) (10 mg/kg/d), alpha-lipoic acid (ALA) (10 mg/kg/d) and MT + ALA, i.p. for 5 days before treatment with CP (150 mg/kg, i.p.) on day 5.(i) ↓ Serum creatinine, urea, uric acid, potassium, sodium, chloride bicarbonate, and oxidative markers
(ii) Combined administration of MT and ALA was found to be more effective
[52]Male Swiss albino mice (n = 48)Mice were administered with a single dose of CP (200 mg/kg, i.p.), and then followed by propolis (100 mg/kg) for 7 consecutive days.(i) Improved the levels of urea and creatinine.
(ii) Moreover, the histological picture of the kidney was significantly improved.
(iii) In addition, propolis prevented liver toxicity and immunosuppression.
[53]Male Wistar rats (n = 40)Animals were pretreated with oral whey protein isolate (WPI) (75, 150 or 300 mg/kg/day), respectively, for 15 days before CP (200 mg/kg, i.p.) treatment on day 15.(i) ↓ Renal MDA, NOx, MPO and IL-1β levels
(ii) Treatment with WPI significantly protected against CP-induced damage showing marked dose-dependent antioxidant and anti-inflammatory properties.
[54]Male albino mice (n = 40)Hesperidin (HSN) was administered for 10 consecutive days at a dose of 100 and 200 mg/kg/day, orally. While, CP (200 mg/kg, i.p.) was administered on the fifth day, after starting HSN.(i) ↓ Serum creatinine and cystatin C
(ii) ↓ MDA, nitric oxide, Bax/Bcl-2 ratio, and caspase-3 levels
(iii) HSN prevented CP-induced nephrotoxicity by tackling oxidative/nitrative stress, inflammation, and apoptosis.
[55]Male albino rats (n = 46)Rats were treated with fennel oil, an oil extracted from the seeds of Foeniculum vulgare (1 ml/kg, once a week for 6 weeks) orally. CP was administered orally at a dose of 15 mg/kg once a week for six weeks.(i) ↓ creatinine, urea, PCNA, caspase-3, and α-SMA
(ii) Improved the histological structure of the kidney
(iii) In general, the ameliorative effect of fennel oil might be due to its antioxidant activity
[56]Male Swiss albino mice (n = 36)Iridoid glycosides enriched fraction (IGs), obtained from Picrorhiza kurroa was administered daily at 25, 50, and 100 mg/kg; orally for 21 days. Followed by CP (200 mg/kg, i.p.) intoxication for consecutive two days. To evaluate the role of PPAR-γ receptors for the protective effect of IGs, additional mice were pretreated with PPAR-γ antagonist (BADGE 5 mg/kg, i.p) followed by IGs (100 mg/kg; p.o.) for 21 days before CP intoxication.(i) Treatment with IGs prevented renal tubular swelling, granular degeneration and glomerular damage.
(ii) Improved the altered expressions of NF-kB, IL-1β and TNF-α
(iii) The antiapoptotic effect of IGs was showed by the Bax/Bcl-2 expressions and caspase 3/9 activity in renal tissues.
(iv) Improved the PPAR-γ expression in the kidney tissues
[57]Male Sprague-Dawley rats (n = 24)Rats were treated with boric acid (BA) for 6 days and CP (200 mg/kg) with BA (200 mg/kg) on the fourth day of the experiment.(i) ↓ Serum creatinine, BUN, MDA, and NO levels
(ii) ↑ CAT, GSH, and GPx levels
(iii) BA-induced renoprotection might be due to an increase in the activity of the antioxidant protection system and also inhibition of lipid peroxidation.
[58]Sprague-Dawley rats (n = 18)N-acetylcysteine (NAC) (100 mg/Kg) was administered i.p., once daily for 5 consecutive days and followed by a single dose of CP (200 mg/Kg), 1 hour after the last dose.(i) NAC re-established the GSH pool and preserved the normal histoarchitecture of the kidney.
(ii) This might be due the antioxidant properties of NAC.
[59]Male Wistar rats (n = 33)Animals were treated with Echinodorus macrophyllus (ECM) (2 g/kg) by oral gavage once a day for 5 days, and followed by single dose of CP (150 mg/kg, i.p.), in the fifth day of the experiment.(i) ↑ creatinine clearance and levels of thiol in the kidney tissue
(ii) ↓ Excretion of urinary peroxides
(iii) ↓ TBARS
[60]Male albino rats (n = 20)Rats were coadministered orally with CP (20 mg/kg) and aqueous leaf extract of Acalypha wilkesiana (110, 220, and 440 mg/kg) leaf extract daily for 7 days.(i) ↓ Plasma creatinine, urea, and uric acid
(ii) ↑ Plasma SOD, CAT, GST, and the level of GSH in a dose-dependent manner.
[61]Male Swiss albino mice (n = 30)Rats were treated with Eucalyptus globulus (EG) (50 and 100 mg/kg) once daily for 15 days along with CP (200 mg/kg, on 12th and 13th day). All treatments were administered intraperitoneally.(i) ↓ creatinine, BUN, and liver enzymes.
(ii) ↓ MDA levels
(iii) ↑ GSH contents
(iv) This protective effect might be partially through induction of Nrf2/HO-1 signaling with attenuation of excessive inflammatory responses as well as apoptosis in renal tissues.
[62]Male Wistar rats (n = 5–7 rats per group, 4 groups in total)Rats were pretreated with aminoguanidine (AG) at a dose of 200 mg/kg i.p. 1 hour before the administration of CP at a dose of 150 mg/kg.(i) AG prevented lipid peroxidation, protein oxidation, depletion of reduced GSH, and loss of activities of the antioxidant enzymes, including GPx, catalase, and GSTase and also MPO activity.
(ii) This effect might be through inhibiting oxidative stress.
[63]Either sex of SD rats (n = 66)Nephrotoxicity was induced with a single administration of CP 200 mg/kg, i.p., on the first day. Then, followed by the treatment of Croton macrostachyus (CM) crude extract and solvent fractions orally for 10 days.(i) ↓ Serum creatinine and BUN
(ii) Histopathological results also confirmed the protective effect of the crude extract and solvent fractions of CM.
[64]Male ICR mice (n = 40)Animals were treated once daily with CP (80 mg/kg/day) for 5 days and pyrroloquinoline quinone (PQQ) (5, 10, and 20 mg/kg/day) for another 14 days.(i) ↓ Serum levels of creatinine and urea
(ii) ↓ MDA, IL-1β, IL-6, and TNF-α levels
(iii) PQQ prevented nephrotoxicity probably by activating the Nrf2-mediated antioxidant pathway and through inhibiting the NLRP3 inflammatory pathway.
[65]Male swiss albino mice (n = 30)Animals were treated with rutin (40 mg/kg and 80 mg/kg) after the administration of CP (25 mg/kg), respectively, for 14 days.(i) Rutin treatment significantly reversed the status serum biomarkers, hematological variables, and antioxidant markers
[66]Male SD rats (n = 70)Rats were administered with single injection of CP (150 mg/Kg, i.p) and followed by garcinol treatment (10 mg/Kg, orally/daily) for 4 weeks.(i) ↓ IL-1b, IL-6, monocyte chemotactic protein-1(MCP-1), macrophage inducible c type lectin (mincle), spleen tyrosine kinase (Syk), transcriptional factor (NF-kB), and toll-like receptor (TLR-4).
(ii) This protective effect might be through inhibiting mincle expression, and prevents Syk and TLR-4/NF-kB activation.
[67]Male albino rats (n = 18)Animals were intraperitoneally injected with a single dose of CP (200 mg/kg) and oral administration of Sargassum cinereum extract at dose of 180 mg/kg for consecutive 20 days.(i) Ameliorated hematological, biochemical, oxidative damages and histopathological changes induced by CP-injection.
[68]Female Balb/c mice (n = 90)Mice were administered with CP (25 mg/kg, i.p.) for 10 consecutive days and Pithecellobium dulce fruit extract (P. dulce) (40 mg/kg) for 10 consecutive days orally, starting from the same day of CP administration.(i) ↓ Serum urea and creatinine levels.
(ii) CP-induced immunosuppression accompanied with urotoxicity, hepatotoxicity, and nephrotoxicity were ameliorated.
[41]Male Wistar rats (n = 30)Rats were treated with Olive leaf extract (OLE) 100 or 200 mg/kg body weight for 15 days and a single injection of 150 mg/kg CP at day 16.(i) ↑ antioxidant defenses and Bcl-2 expression
(ii) ↓ Proinflammatory and proapoptotic markers NF-κB, Bax, cytochrome c, and caspase-3.
(ii) ↑ Nrf2/ARE/HO-1 signaling, enhanced the antioxidant activity and attenuated inflammation and apoptosis.
[69]Male albino Wistar rats (n = 24)Rats were pretreated with Formononetin (FOR) (40 mg/kg/day) 15 days followed by CP-injection on the 16th day.(i) Enhanced the level of antioxidants and suppressed oxidative stress
(ii) Proinflammatory mediators and apoptosis were also suppressed due to FOR treatment.
(iii) Protective effect might be due to attenuation of the oxidative damage and inflammation.

Tumor necrosis factor-α (TNF-α), nuclear factor kappa B (NF-κB), interleukin-6 (IL-6), interleukin-1β (IL-1β), inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), light chain 3B (LC3B), malondialdehyde (MDA), number of animals used (n), gluthathione (GSH), superoxide dismutase (SOD), catalase (CAT), terminal deoxynucleotidyl transferase-mediated dUTP-biotin Nick end labeling assay (TUNEL), glutathion peroxidase (GPx), protein carbonyl (PC), total oxidant state (TOS), oxidative stress indexes (OSI), total antioxidant capacity (TAC), and glutathione-S-transferase (GST).