Skip to main content

Pharmacological agents for the prevention of colistin-induced nephrotoxicity

Abstract

Background

Colistin is a polymyxin antibiotic which has been used for treatment of Gram-negative infections, but it was withdrawn due to its nephrotoxicity. However, colistin has gained its popularity in recent years due to the reemergence of multidrug resistant Gram-negative infections and drug-induced toxicity is considered as the main obstacle for using this valuable antibiotic.

Results

In total, 30 articles, including 29 animal studies and one clinical trial were included in this study. These compounds, including aged black garlic extract, albumin fragments, alpha lipoic acid, astaxanthin, baicalein, chrysin, cilastatin, colchicine, curcumin, cytochrome c, dexmedetomidine, gelofusine, grape seed proanthocyanidin extract, hesperidin, luteolin, lycopene, melatonin, methionine, N-acetylcysteine, silymarin, taurine, vitamin C, and vitamin E exhibited beneficial effects in most of the published works.

Conclusions

In this review, the authors have attempted to review the available literature on the use of several compounds for prevention or attenuation of colistin-induced nephrotoxicity. Most of the studied compounds were potent antioxidants, and it seems that using antioxidants concomitantly can have a protective effect during the colistin exposure.

Background

Polymyxins are a group of polypeptide antibiotics consisting of five drugs; i.e., polymyxin A–E. However, only polymyxin B and polymyxin E or colistin have therapeutic applications. Colistin was discovered in 1949. It is produced by a specific subspecies of Bacillus polymyxa called colistinus [1]. Colistin became available for clinical use in the 1969s. Approximately one decade later, however, it was replaced with less toxic antibiotics due to concerns for its adverse effects, particularly nephrotoxicity [2, 3]. Nonetheless, in recent years, the emergence of multidrug resistant Gram-negative bacilli (MDR-GNB), particularly Pseudomonas aeruginosa, Acinetobacter baumannii, and Klebsiella pneumonia, as well as the lack of development of new antibiotics have led to the increased use of colistin worldwide [4]. The most common side effects that limit the use of colistin are nephrotoxicity and neurotoxicity. Both side effects are dose-dependent and reversible, and permanent kidney damage has rarely been seen [5]. The incidence of colistin-induced nephrotoxicity has been found to range from 20 to 76% in different published studies [6]. Although the exact mechanism of colistin-induced nephrotoxicity is not known, it has been demonstrated that colistin use is associated with increased membrane permeability, oxidative injuries, and subsequently acute tubular necrosis [7]. Increased tubular epithelial cell membrane permeability results in cations, anions and water influx and subsequently cell swelling and lysis. Other mechanisms of nephrotoxicity include oxidative stress, apoptosis (via mitochondrial, death receptor, and endoplasmic reticulum pathways), cell cycle arrest, autophagy, altered nitric oxide balance, mitochondrial dysfunction and oxidative stress [6, 8, 9]. The factors affecting the increased risk of nephrotoxicity include age, sex, hypoalbuminemia, hyperbilirubinemia, concurrent nephrotoxic agents, various comorbidities, and high dose and long-term use of colistin [6]. Also, the administration of loading doses has increased the risk of colistin nephrotoxicity in some studies [10,11,12]. Colistin-induced nephrotoxicity has been associated with an increase in adverse outcomes in critically ill patients [13]. Considering the increasing incidence of MDR infections, a dearth of options of antimicrobial agents against MDR-GNB infection and the slow rate of antibiotic discovery, colistin remains an important option for severe MDR-GNB infections. However, its nephrotoxicity is a major clinical problem, affecting negatively its therapeutic use [14]. Thus, recognizing pharmacological interventions used to prevent or attenuate colistin-induced nephrotoxicity has gained special interest among clinicians, including physicians, pharmacists and other healthcare professionals in recent years.

The present study aims to review the available literature on the pharmacological interventions used to prevent or attenuate colistin-induced nephrotoxicity. These agents include antioxidant compounds, chemical agents, synthetic drugs, hormones, vitamins, and minerals.

Methods

A literature review was conducted using the following electronic relevant databases: Scopus, PubMed, Medline, Embase, Cochrane Central Register of Controlled Trials, ISI Web of Knowledge, Cochrane Database of Systematic Reviews, and Google Scholar. The searched keywords included ‘Colistin, ‘Polymyxin E, ‘Nephrotoxicity’, ‘Kidney injury’, ‘Renal dysfunction’, ‘Renal impairment’, ‘Nephroprotective Agents’, ‘Nephroprotection’, ‘Nephroprotective effects’, ‘Nephroprotective activity’, and ‘Reno protective effects’. The references of published articles were also screened to find further relevant studies. Searches were performed by two authors independently to confirm consistency and accuracy of results. No time limitation regarding publication date was considered. Non-English language articles and congress abstracts were not eligible for the study.

In total, 28 articles have been included in this study. Both experimental and clinical studies evaluating the effects of the potential nephroprotective agents against colistin-induced nephrotoxicity are summarized in Table 1.

Table 1 Summary of the experimental and clinical studies regarding the effects of potential nephroprotective agents against colistin-induced nephrotoxicity

Aged black garlic extract

Aged black garlic extract (ABGE) is produced by storing fresh garlic in an aqueous ethanol solution for up to 20 months and contains antioxidant compounds that prevent oxidative damage. The process of prolonged extraction modifies unstable compounds such as allicin and converts them to more stable and bioavailable water-soluble organosulfur phytochemicals such as S-allylcysteine and S-allylmercaptocysteine [15]. ABGE has the highest pharmacological and antioxidant activities in comparison to other garlic preparations [16]. Currently, therapeutic and health-promoting effects of ABGE such as antioxidant activities, reactive oxygen species (ROS) scavenging properties, lipid peroxidation inhibition, endothelial cells protection, inhibition of the activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), antihypertensive, anti-thrombotic, neuroprotective, antiaging, cholesterol lowering, and anti-diabetes properties, prevention of deoxyribonucleic acid (DNA) damage and mutagenesis, anti-cancer activity, and immunity modulation have attracted considerable interest [17, 18]. The beneficial effects of ABGE on the amelioration of the toxic effects of cisplatin, doxorubicin, acrylamide, carbon tetrachloride, acetaminophen, and cadmium have been previously proven [17, 19]. Shin et al. investigated the reno protective effect of ABGE against colistin toxicity in rats, which was attributed to its antioxidant and anti-inflammatory properties. The results indicated that 1% ABGE (100 µL per individual) injection intragastrically 30 min prior to 10 mg/kg colistin administration for six consecutive days prevented the elevation of the serum levels of blood urea nitrogen (BUN) and creatinine (Cr). Pretreatment with ABGE could also alleviate tubular damage including vacuolation and necrosis, reduce terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and CD68 positive cells, suppress oxidative stress and inflammatory biomarkers including 8-hydroxydeoxyguanosine (8-OHdG), malondialdehyde (MDA), NF-κB, inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), transforming growth factor-beta 1 (TGF-β1), interleukin (IL)-1β, and tumor necrosis factor-alpha (TNF-α), and restore antioxidant levels such as renal superoxide dismutase (SOD), catalase (CAT), and reduced glutathione [20].

Albumin fragments

The membrane-associated endocytic receptor megalin is an extremely large glycoprotein (≈ 600 kda), belonging to the low-density lipoprotein receptor superfamily. Megalin is heavily expressed on the apical membrane of proximal tubule epithelial cells (PTECs) and plays an important role in the receptor-mediated endocytosis and the subsequent metabolization of proteins and nutrients [21]. Megalin mediates the proximal tubular uptake of different groups of ligands including plasma proteins, peptides, enzymes, vitamin-binding proteins, hormones, hormone-binding proteins, drugs, and toxins. Albumin, myoglobin, hemoglobin, vitamin D-binding protein, retinol-binding protein, β2-microglobulin, lactoferrin, and insulin have been identified as megalin ligands. Additionally, some nephrotoxic drugs such as aminoglycosides, cisplatin, vancomycin, polymyxin B, and colistin are absorbed through megalin receptors [22, 23].

Albumin, a protein with a molecular weight of 67 kDa, is a ligand for megalin and its reabsorption in renal proximal tubules is mediated through megalin. Thus, it has been assumed that it can be used for the inhibition of the renal accumulation of colistin [24]. Considering the fact that only a small fraction of albumin passes the glomerular membrane, albumin fragments can be a more appropriate alternative for the inhibition of colistin accumulation in the kidneys [25]. The results of the research by Suzuki et al. indicated that the co-administration of albumin fragments (with molecular masses of less than 50 kDa) at the dose of 50 mg with 1.0 mg/kg colistin resulted in a significant decrease in urinary N-acetyl-β-D glucosaminidase (NAG) excretion as well as an increase in urinary colistin excretion [26].

Alpha-lipoic acid

Alpha-lipoic acid (ALA), also known as thioctic acid, is an organosulfur compound commonly found in mitochondria, which is necessary for different enzymatic functions. Accumulating evidence has suggested that ALA has multiple pharmacological effects including antidiabetic, anti-dementia or anti-Alzheimer’s disease (AD), anti-ageing, metal chelating and detoxifying, anti-inflammatory, anti-cancer, cardiovascular protective, cognitive protective, and neuroprotective properties [27]. It has also been discovered in various clinical trials that ALA can be efficient in particular diseases including diabetic neuropathy, obesity, schizophrenia, multiple sclerosis (MS), pregnancy complications, and organ transplantation [27]. The nephroprotective actions of ALA against nephrotoxicity induced by cisplatin, chloroquine, methotrexate, and iron overload have been shown in different studies, as well [28,29,30,31,32,33]. Moreover, the great antioxidant potential and scavenging activity of ALA can protect against oxidative injuries [34].

Oktan et al. investigated ALA’s ability to protect against colistin-induced nephrotoxicity in rats. In that study, the rats received 100 mg/kg ALA 30 min before the administration of 450,000 IU/kg colistimethate sodium (CMS) for 10 days. The results demonstrated that ALA could reverse the effects of colistin-induced nephrotoxicity by reducing oxidative stress and renal tubular apoptosis, partly through its suppressing effect on NADPH oxidase 4 (Nox4) and caspase-3. In addition, messenger ribonucleic acid (mRNA) expression of kidney injury molecule-1 (KIM-1), Nox4, and p22phox in the kidney as well as kidney active caspase-3 protein expression were reduced by the administration of ALA. ALA administration also resulted in a significant reduction in the level of urine KIM-1, as a strong biomarker for tubular injury. However, there were no significant changes in renal MDA, SOD, TNF-α, urine microalbumin/Cr, and serum Cr [35].

Astaxanthin

Astaxanthin, a naturally occurring lipid-soluble and red–orange oxycarotenoid pigment, is found in several species of bacteria and yeasts and a wide variety of aquatic organisms such as microalgae, fish, and crustaceans such as shrimps [36]. Its potential pharmacological effects including antioxidant properties, DNA repair, cell regeneration, and neuroprotective, immunomodulatory, antiproliferative, anti-inflammatory, anti-apoptotic, antidiabetic, anticancer, photoprotective, and skin-protective effects have been established in various investigations [37,38,39]. Furthermore, it can prevent oxidative damage to fatty acids and biological membranes by scavenging lipid radicals and destroying peroxides [40]. Animal studies have also revealed its nephroprotective effects against diabetic nephropathy and mercuric chloride-induced nephrotoxicity [41, 42].

An experimental study conducted by Ghlissia et al. exhibited that the co-treatment of astaxanthin (20 mg/kg/day given by oral gavages) and colistin (300,000 or 450,000 IU/kg/day of intramuscular (IM) CMS) for 7 days provided nephroprotection against colistin evidenced by biochemical, histological, and oxidative stress parameters. Besides, astaxanthin prevented change in antioxidant parameters including SOD, glutathione peroxidase (GPx), CAT, reduced glutathione (GSH), and MDA, induced by colistin. It also attenuated the plasma levels of Cr and urine Gamma-Glutamyl Transferase (GGT) and partially diminished the degree of renal tissue damage induced by colistin [43].

Baicalein

Baicalein is a flavone and an active component in the root of Scutellaria baicalensis [44]. There is a large body of evidence on its beneficial pharmacological activities including anti-inflammatory, antioxidative, antiviral, antibacterial, immuno-regulatory, anti-cancer, cardioprotective, neuroprotective, and hepatoprotective effects [45,46,47]. Due to its anti-inflammatory, anti-apoptotic, and antioxidant effects, baicalein was used as a nephroprotective agent against cisplatin and myocardial ischemia-induced nephrotoxicity in the previous studies [47, 48].

In an experimental study, Dai et al. assessed the effects of baicalein on the colistin-induced changes in the renal tissue of a mouse model exposed to 18 mg/kg/day colistin. The results indicated that the 7-day treatment with oral baicalein at 25, 50, and 100 mg/kg/day doses could attenuate colistin-induced tissue damages, decrease BUN, SCr, IL-1β, and TNF-a levels, attenuate all the colistin-induced biomarkers of oxidative stress including MDA, iNOS, nitric oxide (NO), SOD, and CAT, upregulate the expression of nuclear factor-erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) mRNAs, and downregulate the expression of NF-κB mRNA in a dose-dependent manner [49].

Chrysin

Chrysin, as a natural flavonoid, is considered one of the main metabolites of medicinal plants and is found in many fruits, vegetables, and plant extracts such as honey, blue passion flower, Passiflora caerulea, and propolis [50]. The antioxidant, anti-inflammatory, anticancer, antivirus, anti-diabetogenic, anti-anxiolytic, and anti-autophagic effects of chrysin have been proved in various studies [51,52,53]. Its beneficial effects have also been reported in paracetamol-induced nephrotoxicity and doxorubicin-induced cardiotoxicity [54, 55]. Moreover, the beneficial effect of chrysin on the prevention of colistin-induced renal injury was shown in an animal study. In that study, a cumulative dose of 73 mg/kg of colistin was administered to rats during 7 days. Then, oral chrysin was administered at 25 and 50 mg/kg doses for 7 days. The results demonstrated that chrysin could alleviate colistin-induced renal inflammation by significantly decreasing the levels of TNF-α, IL-6, IL-1β, and MDA as well as increasing the levels of SOD, CAT, GPx, GSH, cystatin C (Cys C), and calbindin D28K. Additionally, histopathological examination revealed an improvement in colistin-induced interstitial nephritis in chrysin-treated rats [56].

Cilastatin

Cisplatin, an inhibitor of renal dehydropeptidase-I (DHP-I), has been co-administered with imipenem to prevent its metabolism through DHP-I of proximal renal tubules, increase its stability, and suppress the tubular injury. Its nephroprotective effects have also been proved against nephrotoxicity induced by other drugs such as vancomycin, cisplatin, cyclosporine, tacrolimus, diclofenac, and gentamycin [57]. Considering the fact that megalin and DHP-1 are both located at the brush border membranes of [58], cilastatin may encounter megalin in vivo [23].

Hori et al. emphasized the protective role of colchicine against colistin-induced nephrotoxicity, which was achieved through megalin blockade. Importantly, subcutaneous (SQ) injection of 100 mg/kg cilastatin with 30 mg/kg colistin once daily for 4 days decreased the urinary excretion of NAG and the expression of (KIM-1 in proximal tubule epithelial cells and suppressed colistin-induced tubulointerstitial injury including tubular vacuolization, tubular dilation or atrophy, brush border loss, tubular cell lysis, and cast formation [23].

Colchicine

Previous studies have demonstrated that microtubules are involved in the apical accumulation of megalin-containing vacuoles [59]. Colchicine, a microtubule-depolymerizing agent, interferes with intracellular transport of megalin from the cell membrane and causes the internalization of megalin from the brush border membrane into intracellular vesicles [60]. In a study carried out on colistin-treated rats, the effect of colchicine, a microtubule-depolymerizing agent, was assessed on colistin-induced nephrotoxicity. The results demonstrated that colchicine administration (3.5 mg/kg, IP) before the injection of 1.0 mg/kg IV colistin led to a significant decrease in urinary NAG excretion [26].

Curcumin

Curcumin, a polyphenol compound, is found within turmeric and is extracted from the rhizome of Curcuma longa Linn. Curcumin has broad biological activities including anti-inflammatory, antioxidant, and anticancer effects, inhibition of angiogenesis and metastasis, radio sensitization in cancer cells, radioprotection in normal cells, and anti-human immunodeficiency virus, antibacterial, nematocidal, and hepatoprotective functions [61]. Its renoprotective effects have also been proved in several experiments including diabetic nephropathy, chronic renal failure, renal injury induced by ischemia and reperfusion, shock-wave lithotripsy, and nephrotoxicity induced by compounds such as cisplatin, methotrexate, gentamicin, adriamycin, chloroquine, iron nitrilotriacetate, sodium fluoride, heavy metals, and triiodothyronine [62,63,64]. It seems that curcumin’s pleiotropic activities are related to its free radical scavenging, hydrogen donation, and antioxidant properties as well as to the activation of signaling pathways such as NF-kB, protein kinase B (Akt), and Nrf2 [62].

Edrees et al. evaluated the nephroprotective effects of the oral administration of curcumin (200 mg/kg/day for 6 days) on antioxidant, inflammatory, and apoptotic markers as well as on colistin-induced renal damage in rats treated with 300,000 IU colistin/kg/day. They found that curcumin treatment could partially restore the altered biochemical markers such as increased expression levels of SCr, serum urea, serum uric acid (UA), MDA, NO, TNF-α, IL-6, and caspase-3 as well as decreased expression levels of CAT, GSH, and B-cell lymphoma 2 (Bcl-2). Furthermore, co-treatment with curcumin could alleviate the renal histopathological findings. Curcumin was found to exert its protective effects on the renal tissue via its antioxidant, anti-inflammatory, and anti-apoptotic activities [65].

Cytochrome C

Cytochrome C (cyt c) is a small mitochondrial protein (MW = 12 kDa), which is involved in the mitochondrial respiratory chain and acts downstream of p53 in the apoptotic pathway. It has been used for the treatment of carbon monoxide poisoning, hypnotic intoxication, severe hypoxia in shock, dyspnea, myocardial anoxia, and cancers [66].

A study conducted by Suzuki et al. evaluated the effect of cyt c, as a megalin ligand, on colistin-induced nephrotoxicity as well as on the binding of colistin to megalin. Co-administration of 1.0 mg/kg colistin with 100 mg/kg cyt c significantly decreased the urinary NAG excretion, a marker of renal tubular damage, and increased the urinary excretion of colistin. Furthermore, cyt c inhibited the binding of colistin to megalin competitively [26].

Dexmedetomidine

Dexmedetomidine is a potent and highly selective α2-adrenoceptor agonist with sedative, analgesic, sympatholytic, hemodynamic stabilizing, anti-inflammatory, and diuretic activities [67]. In recent years, numerous studies have been focused on the organoprotective properties of dexmedetomidine in brain, heart, and kidneys. The α2 adrenoceptors are distributed widely in proximal and distal tubules, as well as in peritubular vasculature. The underlying mechanism of its nephroprotection may be attributed to its anti-inflammatory, anti-oxidative stress, and anti-apoptotic actions. Moreover, it causes renal artery vasodilatation and an increase in renal blood flow and urine output through the activation of adrenoreceptors, inhibition of renal sympathetic nerves, reduction of the norepinephrine level and vasopressin, increase of atrial natriuretic peptide secretion, and inhibition of renin release [68]. The renoprotective effects of dexmedetomidine have been supported in animal and human models of renal injury induced by ischemia–reperfusion or sepsis and following cardiac surgery [69, 70].

In a study by Talih et al. the effect of dexmedetomidine on colistin nephrotoxicity was assessed in rats by determining the plasma levels of BUN, Cr, KIM-1, total oxidative stress (TOS), and total anti-oxidative stress (TAS). The altered parameters were improved using 10 and 20 mcg/kg IP dexmedetomidine 20 min before the administration of 10 mg/kg IP colistin twice a day for 7 days. Moreover, the apoptotic activity was evaluated via caspase-3 immunostaining, which indicated that the caspase-3 staining rate was lower in the dexmedetomidine group compared to the colistin group, but the difference was not statistically significant [71].

Gelofusine

Gelofusine is a 4% w/v solution of succinylated bovine gelatin (modified fluid gelatin) with a mean molecular mass of 30,000 Da, which is used for volume replacement. It has been demonstrated that gelofusine can competitively inhibit the renal tubular reabsorption of proteins and peptides such as albumin, β2-microglobulin, nephrotoxic radiolabeled integrin, and somatostatin peptides [72,73,74]. The exact mechanism for this inhibition is not completely understood, but it is proposed to be related to the megalin receptor. Considering the role of megalin in the renal reabsorption of colistin, it has been proposed that gelofusine can competitively inhibit its renal reabsorption and endosomal/lysosomal trafficking [75].

Sivanesan et al. evaluated the nephroprotective effects of gelofusine in a mouse model of colistin-induced nephrotoxicity using a fixed dose of colistin (cumulative dose, 84 mg/kg SC) and four different gelofusine dosage regimens (cumulative doses of 450, 900, 1800, and 3600 mg/kg IP). The results indicated that gelofusine could serve as a safe adjunct for ameliorating nephrotoxicity and increasing the therapeutic index of polymyxins at the histological level in a dose-dependent manner. The dose-dependent nephroprotective effect of gelofusine could be associated with competition between gelofusine and colistin for uptake by proximal tubular cells [75].

Grape seed proanthocyanidin extract

Grape seed proanthocyanidin extract (GSPE) is a natural flavonoid polyphenolic molecule obtained from black grape seeds. GSPE is one of the most potent plant antioxidants and free radical scavengers whose pharmacological benefits including antioxidant, antiapoptotic, antimicrobial, anticarcinogenic, vasodilator, anti-fatigue, and anti-inflammatory properties have been demonstrated in numerous investigations [76]. GSPE has been found to be effective in a broad spectrum of pathological conditions including cardiovascular diseases, acute and chronic stress, gastrointestinal distress, neurological disorders, pancreatitis, neoplastic processes, and carcinogenesis, drug and chemical-induced multi-organ toxicity, metabolic syndrome-related disorders, and obesity. Its nephroprotective effects have also been proved in renal injury induced by cadmium, contrast agents, carboplatin, thalidomide, amikacin, cyclosporine A, and diabetes [77,78,79].

Ozkan et al. reported the protective role of GSPE (100 mg/kg/day orally along with 300,000 IU/kg/day IP colistimethate sodium for 7 days) against colistin-induced nephrotoxicity, including a significant decrease in BUN and Cr levels, renal histopathological scores, TUNEL, caspase 1 and 3, calpain 1, iNOS, and endothelial NO synthase [80] staining. The results suggested that GSPE exerted its renoprotective effects via reducing oxidative damage, caspase mediated apoptosis, and iNOS, which might be involved in the pathogenesis of colistin-associated nephropathy [79].

Hesperidin

Hesperidin is a flavonoid glycoside, which is present in high concentrations in some citrus species such as lemon, sweet orange, and grapefruit [81]. Previous studies indicated that hesperidin had antioxidant, anti-inflammatory, antihypertensive, lipid-lowering, insulin sensitivity, bone loss inhibitory, and neuroprotective effects [81,82,83,84]. Hesperidin has also shown promising results in acetaminophen toxicity, cisplatin-induced acute kidney injury, and lipopolysaccharide-caused lung injury in animal studies [85,86,87]. Hesperidin’s pharmacologic effects have been attributed to its significant free radical scavenging and anti-oxidation activities [88].

In an animal study, treatment with hesperidin was associated with protective effects on renal injury induced by colistin in rats. Based on the results, oral hesperidin administration at 200 and 300 mg/kg/day doses for 7 days after colistin administration at a cumulative dose of 73 mg/kg could reduce TNF-α, IL-6 IL-1β, and MDA levels. It also increased the activities of SOD, CAT, and GPx as well as the level of GSH. Furthermore, Cys C and calbindin D28K immunopositivities were significantly improved through hesperidin treatment [56].

Luteolin

Luteolin, a 3,4,5,7-tetrahydroxyflavone, is the main member of the flavonoid family that is found in numerous plants, fruits, and vegetables. Luteolin has major biological and pharmacological properties including antioxidant, anti-inflammatory, antiapoptosis, neuroprotective, and cardioprotective activities [89]. Its notable defensive and preventive effects against pathological conditions such as chronic inflammatory diseases, atherosclerosis, drug-induced liver injury, diabetes, cancer, and allergies have also attracted attention in clinical practice [90]. Regarding its nephroprotective effects, luteolin has been found to protect against cisplatin, bisphenol-A, lead, chromium, mercury-induced nephrotoxicity, diabetic nephropathy, lipopolysaccharide (LPS) induced acute renal injury, and so forth [91,92,93].

In an animal experiment, Arslan et al. studied the potential effects of luteolin (10 mg/kg) on the biomarkers of nephrotoxicity in rats receiving 480,000 IU/kg/day colistin IP for 7 days. The findings showed that the level of post-treatment Cr was significantly increased in the rats receiving colistin alone, while no significant difference was observed in pretreatment and post-treatment Cr levels in the group receiving colistin + luteolin. Besides, the number of apoptotic cells and renal histological damage score were significantly higher in the colistin group compared to the rats receiving colistin in combination with luteolin [94].

Lycopene

Lycopene is a lipophilic pigment, which belongs to the carotenoid family and is responsible for the red color of tomato and its related products. It is considered as an important natural antioxidant, anti-inflammatory agent, and free radical scavenger, which protects cell biomolecules including lipids, lipoproteins, and DNA against oxidation [95]. Up to now, numerous studies have evaluated the effects of lycopene in chronic diseases such as certain types of cancer (e.g., lung and prostate), male infertility, AD, chronic obstructive pulmonary diseases (COPD), osteoporosis, and neuropathic pain [96]. The beneficial roles of lycopene in the prevention of nephrotoxicity induced by gentamicin, vancomycin, adriamycin, cisplatin, mercuric chloride, combined use of isoniazid and rifampicin, furan, and ischemic reperfusion have been reported [97,98,99].

Dai et al. studied the effect of lycopene on colistin-induced nephrotoxicity in a mouse model. Lycopene pretreatment (5 or 20 mg/kg/day orally) 2 h before the administration of colistin (15 mg/kg/day intravenously) for 7 days attenuated colistin-induced nephrotoxicity through increasing the levels of endogenous antioxidant biomarkers including GSH, SOD, and CAT as well as decreasing the concentrations of BUN and SCr, caspase-dependent tubular apoptosis/necrosis, MDA, NO, iNOS, and HO-1 activity. Furthermore, lycopene, especially in the colistin plus lycopene (20 mg/kg) group, was found to significantly downregulate the expression of NF-κB mRNA and upregulate the expressions of Nrf2 and HO-1 mRNA, which seem to be the main reason for its preventive effects on colistin-induced nephrotoxicity. This study was the first to reveal that the Nrf2/HO-1 pathway plays a protective role in colistin-induced nephrotoxicity in mice [97].

Melatonin

Melatonin, N-acetyl-5-methoxytryptamine, is mainly synthesized within the pineal cells and is released into the circulation in a circadian rhythm. It plays important roles in complex physiological and pathological processes through its receptors, which are located in most central and peripheral tissues and organs including kidneys. Melatonin and its metabolites have antioxidant and direct free radical scavenging activities, resulting in the augmentation of antioxidant enzymes in oxidative stress. Its beneficial effects also result from its ability to inhibit principle pro-inflammatory or apoptotic cytokines such as TNF-α and NF-κB [100]. Melatonin was first used for the treatment of sleep disorders and jet lags. Recently, however, researchers have found new applications for this hormone including cancer, neurodegenerative and mental disorders (e.g., AD), immune disorders (including rheumatoid arthritis), endocrine and metabolic disorders (including type II diabetes), mental and neurodegenerative disorders, and cardiovascular diseases [101]. The nephroprotective capacity of melatonin has also been elucidated in different studies, revealing it as a promising agent for the pathogenetic correction of diabetic nephropathy, chronic kidney disease progression, idiopathic membranous nephropathy, ischemia–reperfusion, and rhabdomyolysis-induced renal injury as well as the eradication of renal oxidative stress induced by different agents such as anticancers (cisplatin, ifosfamide, doxorubicin, methotrexate, and mechlorethamine), antibiotics (gentamicin, amikacin, ciprofloxacin, vancomycin, and tenofovir), immunosuppressants (cyclosporine and tacrolimus), antipyretics (acetaminophen), and other toxic compounds (mercuric chloride, carbon tetrachloride, uranium, cadmium, and chlorpryfos-ethyl) [100, 102].

Melatonin at the dose of 10 mg/kg/day along with colistin at the cumulative dose of 36.5 mg/kg could significantly mitigate the consequences of colistin-induced renal injury including increased urinary excretion of NAG and plasma Cr level as well as renal histological abnormalities. Melatonin also decreased the total body clearance and increased the half-life of colistin [103].

N-Acetyl cysteine

N-Acetylcysteine (NAC), as a sulfhydryl-containing compound, is a precursor to the amino acid L-cysteine and consequently the antioxidant glutathione. NAC is found naturally in some vegetables and fruits, mostly in plants of the Allium species. It has mucolytic, antioxidant, anti-inflammatory, anticarcinogenic, and vasodilatory effects [104]. Due to the sulfhydryl group (–SH) within NAC, this molecule is able to scavenge free radicals, modulate cytokine synthesis through the inhibition of NF-κB, stabilize proteins and DNA structures, and chelate metals. NAC has been widely used in cystic fibrosis, acetaminophen overdose, and chronic obstructive lung disease. Previous studies have demonstrated the beneficial impact of NAC on non-alcoholic steatohepatitis, diabetic neuropathy, retinopathy, nephropathy, various types of cancer, infectious diseases (Acquired Immunodeficiency Syndrome (AIDS), tuberculosis, influenza, and diseases caused by respiratory syncytial virus, Helicobacter pylori, and SARS-CoV-2), stroke, Parkinson’s disease, dementia, recurrent unexplained pregnancy loss, male infertility, polycystic ovary syndrome, psychiatric disorders (schizophrenia, bipolar disorder, obsessive compulsive disorder, and addiction behavior), metal toxicity, age-related macular degeneration, cataract, and dry eye syndrome [105].

Recently, special attention has been drawn to NAC as a drug to attenuate contrast-induced nephropathy. Several studies have been conducted in this field, but the results are too conflicting to draw definite conclusions.[106]. The effect of NAC on renal function has also been evaluated in renal toxicity caused by cisplatin, ifosfamide, and renal ischemia–reperfusion injury [107,108,109].

Ozyilmaz et al. disclosed that the simultaneous administration of 150 mg/kg IP NAC with 300,000 IU/kg/day IP colistin for six consecutive days did not exert any significant effects on biochemical parameters including plasma BUN and Cr levels, creatinine clearance (ClCr), urine Cr and protein levels, TNF-alpha plasma level, and MDA tissue level. In contrast, reduction of elevated SOD levels and reversal of the increase in the tubular immunohistochemical expression of i-NOS and neurotrophin-3 were observed in NAC-treated rats [110].

In another study conducted by Ceylan et al. in 2018, NAC-induced protective and repairing effects against colistin nephrotoxicity were evaluated. The researchers found that the animals treated with 300,000 IU/kg/day CMS for 10 days showed marked elevations in Cr and urine NAG levels, while no significant changes were detected in serum Cr in the NAC + colistin group. Moreover, urine NAG levels were significantly higher in the CMS group compared to the other group on the 10th day of treatment. More interestingly, NAC could reverse colistin-induced negative effects, as determined by increased apoptosis index and renal histological damage score as well as by decreased renal expression levels of eNOS, SOD2, and matrix metalloproteinase (MMP). However, no significant differences were observed between the study groups regarding the renal total antioxidant and oxidant capacity. Thus, the researchers proposed that NAC exerted its beneficial effects through the activation of SOD2 and eNOS expression levels as well as MMP3 [111].

Silymarin

Silymarin is extracted from the fruit and seeds of the Silybum marianum (milk thistle) and is a complex of other components such as a family of flavonolignans (silybin, isosilybin, silychristin, isosilychristin, and silydianin) and a flavonoid (taxifolin). Silybin is considered the major biologically active compound of the extract [112]. Silymarin has exhibited a number of pharmacological activities including antioxidant, immunomodulatory, antifibrotic, antiproliferative, anticancer, and antiviral properties. The cytoprotection activities of silymarin have been attributed to its antioxidant and radical scavenging effects. Silymarin has been used for centuries as a liver tonic and for the treatment of various liver diseases. Moreover, studies have demonstrated its protective effects in the fields of nephrotoxicity, hepatotoxicity, viral hepatitis, cancer, in vitro fertilization, neurotoxicity, depression, environmental toxins exposure, and lung, prostate, pancreas, and other diseases [113]. Silymarin has also shown protective roles in nephropathic processes including hemodialysis, end-stage diabetic nephropathy, contrast-induced nephropathy, renal injury caused by chemicals such as arsenic and ferric nitrilotriacetate, and drug-induced nephrotoxicity such as paracetamol, aminoglycosides, vancomycin, polymyxin, isoniazid, doxorubicin, cisplatin, cyclophosphamide, methotrexate, and cyclosporin [64, 114,115,116].

Hassan et al. evaluated the effect of silybin on the damages produced by polymyxin E on the rat kidney using histological, ultrastructural, and morphometric analyses. According to their findings, treatment with 50 mg/kg IV silybin in its active form (silymarin) twice a day for seven days two hours before each of the two daily doses of polymyxin E (at a cumulative dose of 36.5 mg/kg) alleviated the degenerative changes on rat kidney induced by polymyxin E. Interestingly, they observed a minimal residual pathology in animals treated with polymyxin + silybin, which suggested that the concomitant administration of silybin might not completely reverse the rapid process, by which polymyxin E induced nephrotoxicity [117]. Similarly, administration of 50 mg/kg IV silybin in the form of silymarin twice daily before IV polymyxin E (at the collective dose of 36.5 mg/kg) ameliorated the biochemical changes induced by polymyxin E in rats including elevated urinary NAG and serum levels of urea, Cr, UA, sodium, and potassium. However, the differences between the polymyxin and polymyxin + silybin groups were statistically significant only for NAG [118].

Recently, Dumludag et al. indicated that the co-administration of 750,000 IU/kg/day colistin and 100 mg/kg/day silymarin for 7 days led to a significant increase in GPx and SOD levels. In contrast, the study groups were not different in terms of serum MDA, Cys C, and Cr levels. Moreover, the results of histological examination demonstrated that silymarin led to some improvements in tubular necrosis. However, no significant changes were observed in the activity scores of tubular injury, medullar congestion, and interstitial inflammation and the apoptotic index [119]. The authors proposed that more pronounced nephroprotective effects were expected when silymarin was used at higher doses or in longer treatment courses.

Taurine

Taurine is an amino acid found abundantly in most cells of the human body. Taurine has been confirmed to be a promising therapeutic agent against various disorders and pathological conditions due to its cytoprotective properties [120]. Various studies have been performed on the antioxidant activity of taurine in lung, liver, heart, and kidney. Studies have also emphasized the preventive and therapeutic effects of taurine therapy on the central nervous system (stroke, neurodegenerative diseases such as AD, Huntington, and Parkinson’s disease, epilepsy, and retinal degeneration), cardiovascular system (congestive heart failure, hypertension, atherosclerosis, ischemia–reperfusion injury, myocardial arrhythmias, metabolic diseases including mitochondrial diseases, diabetes, and arthritis), and muscles (sarcopenia, Duchenne muscular dystrophy, and myotonic dystrophy)[121]. Moreover, several studies have reported the beneficial effects of taurine on the kidney tissue in diabetic nephropathy, Fanconi anemia, cystinosis, chronic kidney disease, and acute kidney injury caused by lead, arsenic, acetaminophen, gentamycin, and radiation [122].

In a study carried out on colistin-treated mice (15 mg/kg/day IV for seven consecutive days), the effect of taurine was assessed on various plasma biomarkers of nephrotoxicity, kidney tissue markers of oxidative stress, and kidney mitochondrial indices. The results indicated that taurine administration (500 and 1000 mg/kg/day) intraperitoneally in combination with colistin for 7 days led to a significant decrease in colistin-induced elevation in the plasma levels of Cr and BUN. Taurine could also alleviate colistin-induced oxidative stress and mitochondrial dysfunction in the kidney tissue. Furthermore, taurine treatment reversed colistin-induced negative effects such as increased kidney ROS, lipid peroxidation (LPO), tissue oxidized glutathione (GSSG) level, mitochondrial LPO, permeabilization, and GSSG content, decreased renal tissue antioxidant capacity, and reduced glutathione stores, mitochondrial dehydrogenase activity, membrane potential, GSH, and ATP. Kidney tissue histopathological alterations were not evident in colistin-taurine groups. The regulation of mitochondrial function and decreased oxidative stress seem to be a fundamental mechanism of the positive effects of taurine on colistin nephrotoxicity [123].

Vitamin C

Vitamin C is a water-soluble essential vitamin, which exists in the body primarily in its reduced form; i.e., ascorbic acid. Vitamin C is potentially involved in the biosynthesis of collagen and is a co-factor in the biosynthesis of catechol amines, L-carnitine, cholesterol, amino acids, and some peptide hormones. It is a chain-breaking antioxidant and free radical scavenger due to its ability to donate electrons and be readily converted back to its reduced form [124]. Vitamin C has shown beneficial effects on the prevention and treatment of several health conditions including scurvy, common cold, male and female infertility, atherosclerosis, cardiovascular diseases, cancers, diabetes, heavy metal toxicity (lead, arsenic, and cadmium), neurological and neurodegenerative disorders (schizophrenia, Parkinson’s disease, and AD), asthma, respiratory conditions, and ocular diseases (cataract and diabetic retinopathy). Moreover, this micronutrient can accelerate wound and burn healing, enhance immune system, and improve outcomes in critically ill patients [125]. Studies have also demonstrated the nephroprotective effects of vitamin C against nephrotoxicity caused by gentamicin, vancomycin, ampicillin, cisplatin, and contrast agents [126,127,128].

Yousef et al. conducted a dual in vivo and in vitro study to determine the protective effects of ascorbic acid against colistin-induced nephrotoxicity and apoptosis. In that study, ascorbic acid was administered at a dose of 50 or 200 mg/kg twice daily 20 min before each colistin dose (cumulative dose of 36.5 mg/kg) for 7 days. The results showed that urinary NAG excretion was significantly lower in the colistin/ascorbic acid 200 mg/kg group than in the colistin group. Additionally, no significant increase was observed in the plasma level of Cr in the colistin/ascorbic acid 200 mg/kg group compared to the colistin group. However, no significant differences were observed between the two groups in terms of SOD activity. Cell culture studies demonstrated that ascorbic acid had a dose-dependent inhibitory effect on colistin-induced apoptosis. Besides, histological examinations indicated the nephroprotective effect of ascorbic acid, at both low and high doses. More interestingly, ascorbic acid was able to decrease the total body clearance of colistin [128].

In an open-label, non-placebo, randomized controlled trial, Sirijatuphat et al. studied the potential nephroprotective effect of IV ascorbic acid against colistin-associated nephrotoxicity. In that study, 15 patients were given colistin alone (CMS at a loading dose of 300 mg of colistin base activity (CBA) followed by renally adjusted maintenance doses every 12 h), while 13 patients received a combination of colistin and IV ascorbic acid (2 g every 12 h, 20 min before colistin). The findings revealed no significant differences in the incidence of nephrotoxicity and the urinary excretion rates of urinary neutrophil gelatinase-associated lipocalin and NAG at various time points during colistin treatment, plasma colistin concentrations, patients’ clinical and microbiological outcomes, and mortality [129].

Vitamin E

Vitamin E is a family of eight fat soluble compounds including a-, b-, g-, and d-tocopherol and a-, b-, g-, and d-tocotrienol, with α-tocopherol being the most widely known analog and being used most preferentially by the body. Vitamin E, primarily located in the cell and organelle membranes, can be found ubiquitously in various foods and different natural sources. It is considered a potent antioxidant, which can effectively scavenge free radicals and prevent LPO [130]. Vitamin E has been proven to exert excellent antioxidant, anti-inflammatory, anti-diabetic, anti-atherogenic, bone and joint protective, neuroprotective, skin protective, anti-obesity, immune-enhancing, and platelet aggregation inhibitory effects in different in vivo and in vitro studies. Potential therapeutic applications have also been suggested for vitamin E including Parkinson’s disease, AD, obesity, diabetes, atherosclerosis, osteoporosis, AIDS, rheumatoid arthritis, osteoarthritis, radioprotection, and different types of cancer [131]. Moreover, vitamin E has demonstrated promising properties in ischemia/reperfusion, cadmium, phenol, paraquat, contrast agents, aminoglycosides, cisplatin, doxorubicin, vancomycin-induced nephrotoxicity, and diabetic nephropathy [132].

The effect of vitamin E on colistin-induced nephrotoxicity was assessed by Ghlissi’s et al. Based on the results, treatment of rats with CMS at 300,000 IU/kg/day IM for 7 days led to a slight focal tubular dilatation, whereas the severity of renal damage was more prominent in the group receiving 450,000 IU/kg/day of CMS, causing acute tubular necrosis. The results also revealed a significant increase in the plasma levels of Cr, urine GGT, and MDA and a decline in the antioxidants’ activities including SOD, CAT, GPx, and GSH in the renal tissue following colistin treatment. Co-treatment with vitamin E at a daily dose of 100 mg/kg/day could partially prevent renal damage as well as changes in antioxidant parameters and LPO markers. These effects might be due to the scavenging activity of vitamin E against free radicals and the protection of cellular biomembrane from oxidative attack [43].

Ghlissi et al. conducted another study to determine the spontaneous renal recovery after stopping the low and high doses of colistin (300,000 IU/kg/day and 450,000 IU/kg/day of CMS for 7 days, respectively) as well as the curative effect of oral vitamin E (100 mg/kg/day for 2 weeks after colistin discontinuation) on antioxidant status and colistin-induced renal damage in colistin-treated rats. They found that vitamin E treatment improved tubular regeneration and normalized the altered biomarkers (including increased NAG and MDA levels and decreased SOD and GSH activities) in the colistin-treated rats. On the contrary, stopping CMS alone did not cause significant renal recovery [133].

The authors of the previous studies conducted another research and assessed the synergistic effect of vitamins E and C on colistin-induced nephrotoxicity in rats. The rats were exposed to 450,000 IU/kg/day of CMS for 7 days, which induced acute tubular necrosis, increased the urine NAG and GGT levels as well as renal tissue MDA levels, and reduced the plasma levels of vitamins E and C and renal tissue activities of SOD, CAT, and GPx. The findings demonstrated that the co-administration of vitamins E and C (at 100 mg/kg each) could improve the histopathological damage and restore all the mentioned biochemical parameters. The authors concluded that the combination therapy of vitamins E and C had a better antioxidant effect against colistin-induced tubular damage than either of the two vitamins given alone, which was attributed to their different subcellular locations as well as to their synergistic action [134].

In a pilot animal study, the nephroprotective effect of alpha-tocopherol, as the most common and biologically active form of vitamin E, was tested in colistin-treated rabbits. In that study, colistin was administered in low (an IV loading dose of 80 mg/kg CMS followed by IM maintenance doses of 10 mg/kg colistin sulphate for 6 days) and high (an IV loading dose of 120 mg/kg CMS followed by IM maintenance doses of 30 mg/kg colistin sulphate for 6 days) doses [135]. Oral α-tocopherol was administered at a dose of 200 mg for 1 week before colistin treatment and half an hour prior to exposing the animals to colistin in the subsequent week. The results showed a significant decrease in the serum level of Cr, as well as the attenuation of the pathological lesions in the kidneys following α-tocopherol administration. Tocopherol also significantly reduced the serum level of urea in the low-dose group [136].

A randomized clinical trial is being conducted in Loghman-Hakim hospital, a referral tertiary teaching medical center in Iran, to investigate the effect of a daily dose of 400 mg alpha-tocopherol on colistin nephrotoxicity [137].

Conclusion

This review aimed to present an overview of different compounds used in experimental and clinical studies for preventing or attenuating colistin nephrotoxicity. These compounds including ABGE, albumin fragments, ALA, astaxanthin, baicalein, chrysin, cilastatin, colchicine, curcumin, cyt c, dexmedetomidine, gelofusine, GSPE, hesperidin, luteolin, lycopene, melatonin, NAC, silymarin, taurine, vitamin C, and vitamin E exhibited beneficial effects in most of the published works. These effects were attributed to their antioxidant, anti-inflammatory, and anti-apoptotic activities. However, the clinical experience about these agents was limited to only one clinical trial, in which the efficacy of IV ascorbic acid was evaluated in patients and the other studies were all animal-based studies. Thus, further clinical studies are required to elucidate the potential usefulness of these agents against nephropathy induced by colistin. Considering the availability in clinical practice, minimal adverse effect profiles, and positive results in animal studies, curcumin, melatonin, NAC, silymarin, and vitamin E can be suggested to be used in clinical trials.

There may be some possible limitations in this study. First, only English-language articles with full text were included in this searching strategy. Second, the majority of studies included in this review were animal studies, and the clinical use of nephroprotective agents was not evaluated. Another limitation is that the quality, number of sample sizes and studied parameters were various in the reviewed studies. Therefore, it is difficult to compare the treatment effects among these interventions.

Availability of data and materials

Not applicable.

Abbreviations

AKI:

Acute kidney injury

BUN:

Blood urea nitrogen

CAT:

Catalase

CBA:

Colistin base activity

CMS:

Colistimethate sodium

eNOS:

Endothelial nitric oxide synthase

FRAP:

Ferric reducing antioxidant power

GGT:

Gamma-glutamyl transferase

GSH:

Reduced glutathione

GSSG:

Oxidized glutathione

HO-1:

Heme oxygenase-1

iNOS:

Inducible NO synthase

GABA:

Gamma amino butyric acid

GPx:

Glutathione peroxidase

IL:

Interleukin

I.M.:

Intramuscularly

I.P.:

Intraperitoneally

K:

Potassium

KIM-1:

Kidney injury molecule-1

LPO:

Lipid peroxidation

Na:

Sodium

NAG:

N-Acetyl-b-D-glucosaminidase

NGAL:

Neutrophil gelatinase-associated lipocalin

NO:

Nitric oxide

NF-κβ:

Nuclear factor-κβ

Nrf2:

Nuclear factor-erythroid 2-related factor 2

NT-3:

Neurotrophin-3

OSI:

Oxidative stress index

PCr:

Plasma creatinine

ROS:

Reactive oxygen species

S.C.:

Subcutaneous

SCr:

Serum creatinine

SOD:

Superoxide dismutase

TAS:

Total anti-oxidative stress

TNF-α:

Tumor necrosis factor-α

TOS:

Total oxidative stress

UA:

Uric acid

UCr:

Urine creatinine

8-OHdG:

8-Hydroxydeoxyguanosine

AIDS:

Acquired immunodeficiency syndrome

ALA:

Alpha-lipoic acid

AD:

Alzheimer’s disease

ABGE:

Aged black garlic extract

Bcl-2:

B-cell lymphoma 2

COPD:

Chronic obstructive pulmonary diseases

CBA:

Colistin base activity

ClCr:

Creatinine clearance

COX-2:

Cyclooxygenase-2

Cys C:

Cystatin C

cyt c:

Cytochrome C

DHP-I:

Dehydropeptidase-I

DNA:

Deoxyribonucleic acid

iNOS:

Inducible nitric oxide synthase

IL:

Interleukin

LPS:

Lipopolysaccharide

MDA:

Malondialdehyde

MMP:

Matrix metalloproteinase

mRNA:

Messenger ribonucleic acid

MS:

Multiple sclerosis

NAC:

N-Acetylcysteine

NAG:

N-Acetyl-β-D glucosaminidase

Nox4:

NADPH oxidase 4

NO:

Nitric oxide

NF-κB:

Nuclear factor kappa-light-chain-enhancer of activated B cells

Akt:

Protein kinase B

PTECs:

Proximal tubule epithelial cells

TUNEL:

Terminal deoxynucleotidyl transferase dUTP nick end labeling

TGF-β1:

Transforming growth factor-beta 1

TNF-α:

Tumor necrosis factor-alpha

TAS:

Total anti-oxidative stress

–SH:

Sulfhydryl group

References

  1. Falagas ME, Kasiakou SK. Colistin: the revival of polymyxins for the management of multidrug-resistant gram-negative bacterial infections. Clin Infect Dis. 2005;40(9):1333–41.

    Article  CAS  PubMed  Google Scholar 

  2. Nation RL, Li J. Colistin in the 21st century. Curr Opin Infect Dis. 2009;22(6):535–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Li J, Nation RL, Turnidge JD, Milne RW, Coulthard K, Rayner CR, et al. Colistin: the re-emerging antibiotic for multidrug-resistant Gram-negative bacterial infections. Lancet Infect Dis. 2006;6(9):589–601.

    Article  CAS  PubMed  Google Scholar 

  4. Lim LM, Ly N, Anderson D, Yang JC, Macander L, Jarkowski A 3rd, et al. Resurgence of colistin: a review of resistance, toxicity, pharmacodynamics, and dosing. Pharmacotherapy. 2010;30(12):1279–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Falagas ME, Fragoulis KN, Kasiakou SK, Sermaidis GJ, Michalopoulos A. Nephrotoxicity of intravenous colistin: a prospective evaluation. Int J Antimicrob Agents. 2005;26(6):504–7.

    Article  CAS  PubMed  Google Scholar 

  6. Ordooei Javan A, Shokouhi S, Sahraei Z. A review on colistin nephrotoxicity. Eur J Clin Pharmacol. 2015;71(7):801–10.

    Article  CAS  PubMed  Google Scholar 

  7. Ciftci A, Izdes S, Altintas ND. Factors determining nephrotoxicity and mortality in critical care patients receiving colistin. J Infect Dev Ctries. 2018;11(12):912–8.

    Article  PubMed  CAS  Google Scholar 

  8. Heybeli C, Oktan MA, Çavdar Z. Rat models of colistin nephrotoxicity: previous experimental researches and future perspectives. Eur J Clin Microbiol Infect Dis. 2019;38(8):1387–93.

    Article  CAS  PubMed  Google Scholar 

  9. Zavascki AP, Nation RL. Nephrotoxicity of polymyxins: is there any difference between colistimethate and polymyxin B? Antimicrob Agents Chemother. 2017. https://doi.org/10.1128/AAC.02319-16.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Katip W, Yoodee J, Uitrakul S, Oberdorfer P. Efficacy of loading dose colistin versus carbapenems for treatment of extended spectrum beta lactamase producing Enterobacteriaceae. Sci Rep. 2021;11(1):18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Katip W, Uitrakul S, Oberdorfer P. Clinical outcomes and nephrotoxicity of colistin loading dose for treatment of extensively drug-resistant Acinetobacter baumannii in cancer patients. Infect Drug Resist. 2017;10:293–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Katip W, Uitrakul S, Oberdorfer P. Clinical efficacy and nephrotoxicity of the loading dose colistin for the treatment of carbapenem-resistant Acinetobacter baumannii in critically ill patients. Pharmaceutics. 2021;14(1):31.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Gunay E, Kaya S, Baysal B, Yuksel E, Arac E. Evaluation of prognosis and nephrotoxicity in patients treated with colistin in intensive care unit. Ren Fail. 2020;42(1):704–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Gai Z, Samodelov SL, Kullak-Ublick GA, Visentin M. Molecular mechanisms of colistin-induced nephrotoxicity. Molecules (Basel, Switzerland). 2019;24(3):653.

    Article  CAS  PubMed Central  Google Scholar 

  15. Xu C, Mathews AE, Rodrigues C, Eudy BJ, Rowe CA, O’Donoughue A, et al. Aged garlic extract supplementation modifies inflammation and immunity of adults with obesity: a randomized, double-blind, placebo-controlled clinical trial. Clin Nutr ESPEN. 2018;24:148–55.

    Article  PubMed  Google Scholar 

  16. Kasuga S, Uda N, Kyo E, Ushijima M, Morihara N, Itakura Y. Pharmacologic activities of aged garlic extract in comparison with other garlic preparations. J Nutr. 2001;131(3s):1080s-s1084.

    Article  CAS  PubMed  Google Scholar 

  17. Borek C. Antioxidant health effects of aged garlic extract. J Nutr. 2001;131(3s):1010s-s1015.

    Article  CAS  PubMed  Google Scholar 

  18. Shiju TM, Rajesh NG, Viswanathan P. Renoprotective effect of aged garlic extract in streptozotocin-induced diabetic rats. Indian J Pharmacol. 2013;45(1):18–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Nasr AY, Saleh HA. Aged garlic extract protects against oxidative stress and renal changes in cisplatin-treated adult male rats. Cancer Cell Int. 2014;14(1):92.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Lee TW, Bae E, Kim JH, Jang HN, Cho HS, Chang SH, et al. The aqueous extract of aged black garlic ameliorates colistin-induced acute kidney injury in rats. Ren Fail. 2019;41(1):24–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Christensen EI, Birn H. Megalin and cubilin: multifunctional endocytic receptors. Nat Rev Mol Cell Biol. 2002;3(4):256–66.

    Article  CAS  PubMed  Google Scholar 

  22. Christensen EI, Birn H, Storm T, Weyer K, Nielsen R. Endocytic receptors in the renal proximal tubule. Physiology (Bethesda). 2012;27(4):223–36.

    CAS  Google Scholar 

  23. Hori Y, Aoki N, Kuwahara S, Hosojima M, Kaseda R, Goto S, et al. Megalin blockade with cilastatin suppresses drug-induced nephrotoxicity. J Am Soc Nephrol. 2017;28(6):1783–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Cui S, Verroust PJ, Moestrup SK, Christensen EI. Megalin/gp330 mediates uptake of albumin in renal proximal tubule. Am J Physiol. 1996;271(4 Pt 2):F900–7.

    CAS  PubMed  Google Scholar 

  25. Vegt E, van Eerd JE, Eek A, Oyen WJ, Wetzels JF, de Jong M, et al. Reducing renal uptake of radiolabeled peptides using albumin fragments. J Nucl Med. 2008;49(9):1506–11.

    Article  CAS  PubMed  Google Scholar 

  26. Suzuki T, Yamaguchi H, Ogura J, Kobayashi M, Yamada T, Iseki K. Megalin contributes to kidney accumulation and nephrotoxicity of colistin. Antimicrob Agents Chemother. 2013;57(12):6319–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Salehi B, Berkay Yılmaz Y, Antika G, Boyunegmez Tumer T, Fawzi Mahomoodally M, Lobine D, et al. Insights on the use of α-lipoic acid for therapeutic purposes. Biomolecules. 2019;9(8):356.

    Article  CAS  PubMed Central  Google Scholar 

  28. Somani SM, Husain K, Whitworth C, Trammell GL, Malafa M, Rybak LP. Dose-dependent protection by lipoic acid against cisplatin-induced nephrotoxicity in rats: antioxidant defense system. Pharmacol Toxicol. 2000;86(5):234–41.

    Article  CAS  PubMed  Google Scholar 

  29. Murugavel P, Pari L. Attenuation of chloroquine-induced renal damage by alpha-lipoic acid: possible antioxidant mechanism. Ren Fail. 2004;26(5):517–24.

    Article  CAS  PubMed  Google Scholar 

  30. Kang KP, Kim DH, Jung YJ, Lee AS, Lee S, Lee SY, et al. Alpha-lipoic acid attenuates cisplatin-induced acute kidney injury in mice by suppressing renal inflammation. Nephrol Dial Transplant. 2009;24(10):3012–20.

    Article  CAS  PubMed  Google Scholar 

  31. Lee YM, Bae SY, Won NH, Pyo HJ, Kwon YJ. Alpha-lipoic acid attenuates cisplatin-induced tubulointerstitial injuries through inhibition of mitochondrial bax translocation in rats. Nephron Exp Nephrol. 2009;113(4):e104–12.

    Article  CAS  PubMed  Google Scholar 

  32. Cavdar Z, Oktan MA, Ural C, Calisir M, Kocak A, Heybeli C, et al. Renoprotective effects of alpha lipoic acid on iron overload-induced kidney injury in rats by suppressing NADPH oxidase 4 and p38 MAPK signaling. Biol Trace Elem Res. 2020;193(2):483–93.

    Article  CAS  PubMed  Google Scholar 

  33. Çakır T, Polat C, Baştürk A, Gül M, Aslaner A, Durgut H, et al. The effect of alpha lipoic acid on rat kidneys in methotrexate induced oxidative injury. Eur Rev Med Pharmacol Sci. 2015;19(11):2132–9.

    PubMed  Google Scholar 

  34. Zhang J, McCullough PA. Lipoic acid in the prevention of acute kidney injury. Nephron. 2016;134(3):133–40.

    Article  CAS  PubMed  Google Scholar 

  35. Oktan MA, Heybeli C, Ural C, Kocak A, Bilici G, Cavdar Z, et al. Alpha-lipoic acid alleviates colistin nephrotoxicity in rats. Hum Exp Toxicol. 2021;40(5):761–71.

    Article  CAS  PubMed  Google Scholar 

  36. Chang MX, Xiong F. Astaxanthin and its effects in inflammatory responses and inflammation-associated diseases: recent advances and future directions. Molecules. 2020;25(22):5342.

    Article  PubMed Central  CAS  Google Scholar 

  37. Yuan JP, Peng J, Yin K, Wang JH. Potential health-promoting effects of astaxanthin: a high-value carotenoid mostly from microalgae. Mol Nutr Food Res. 2011;55(1):150–65.

    Article  CAS  PubMed  Google Scholar 

  38. Galasso C, Orefice I, Pellone P, Cirino P, Miele R, Ianora A, et al. On the neuroprotective role of astaxanthin: new perspectives? Mar Drugs. 2018;16(8):247.

    Article  PubMed Central  CAS  Google Scholar 

  39. Davinelli S, Nielsen ME, Scapagnini G. Astaxanthin in skin health, repair, and disease: a comprehensive review. Nutrients. 2018;10(4):522.

    Article  PubMed Central  CAS  Google Scholar 

  40. Kurashige M, Okimasu E, Inoue M, Utsumi K. Inhibition of oxidative injury of biological membranes by astaxanthin. Physiol Chem Phys Med NMR. 1990;22(1):27–38.

    CAS  PubMed  Google Scholar 

  41. Naito Y, Uchiyama K, Aoi W, Hasegawa G, Nakamura N, Yoshida N, et al. Prevention of diabetic nephropathy by treatment with astaxanthin in diabetic db/db mice. BioFactors (Oxford, England). 2004;20(1):49–59.

    Article  CAS  Google Scholar 

  42. Augusti PR, Conterato GM, Somacal S, Sobieski R, Spohr PR, Torres JV, et al. Effect of astaxanthin on kidney function impairment and oxidative stress induced by mercuric chloride in rats. Food Chem Toxicol. 2008;46(1):212–9.

    Article  CAS  PubMed  Google Scholar 

  43. Ghlissi Z, Hakim A, Sila A, Mnif H, Zeghal K, Rebai T, et al. Evaluation of efficacy of natural astaxanthin and vitamin E in prevention of colistin-induced nephrotoxicity in the rat model. Environ Toxicol Pharmacol. 2014;37(3):960–6.

    Article  CAS  PubMed  Google Scholar 

  44. de Oliveira MR, Nabavi SF, Habtemariam S, Erdogan Orhan I, Daglia M, Nabavi SM. The effects of baicalein and baicalin on mitochondrial function and dynamics: a review. Pharmacol Res. 2015;100:296–308.

    Article  PubMed  CAS  Google Scholar 

  45. Liang S, Deng X, Lei L, Zheng Y, Ai J, Chen L, et al. The Comparative study of the therapeutic effects and mechanism of baicalin, baicalein, and their combination on ulcerative colitis rat. Front Pharmacol. 2019;10:1466.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Liu H, Dong Y, Gao Y, Du Z, Wang Y, Cheng P, et al. The Fascinating effects of baicalein on cancer: a review. Int J Mol Sci. 2016;17(10):1681.

    Article  PubMed Central  CAS  Google Scholar 

  47. Sahu BD, Mahesh Kumar J, Sistla R. Baicalein, a bioflavonoid, prevents cisplatin-induced acute kidney injury by up-regulating antioxidant defenses and down-regulating the MAPKs and NF-κB pathways. PLoS ONE. 2015;10(7): e0134139-e.

    Article  CAS  Google Scholar 

  48. Lai CC, Huang PH, Yang AH, Chiang SC, Tang CY, Tseng KW, et al. Baicalein, a component of Scutellaria baicalensis, attenuates kidney injury induced by myocardial ischemia and reperfusion. Planta Med. 2016;82(3):181–9.

    CAS  PubMed  Google Scholar 

  49. Dai C, Tang S, Wang Y, Velkov T, Xiao X. Baicalein acts as a nephroprotectant that ameliorates colistin-induced nephrotoxicity by activating the antioxidant defence mechanism of the kidneys and down-regulating the inflammatory response. J Antimicrob Chemother. 2017;72(9):2562–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Farkhondeh T, Samarghandian S, Bafandeh F. The cardiovascular protective effects of chrysin: a narrative review on experimental researches. Cardiovasc Hematol Agents Med Chem. 2019;17(1):17–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zheng X, Meng WD, Xu YY, Cao JG, Qing FL. Synthesis and anticancer effect of chrysin derivatives. Bioorg Med Chem Lett. 2003;13(5):881–4.

    Article  CAS  PubMed  Google Scholar 

  52. Cho H, Yun CW, Park WK, Kong JY, Kim KS, Park Y, et al. Modulation of the activity of pro-inflammatory enzymes, COX-2 and iNOS, by chrysin derivatives. Pharmacol Res. 2004;49(1):37–43.

    Article  CAS  PubMed  Google Scholar 

  53. Woodman OL, Chan E. Vascular and anti-oxidant actions of flavonols and flavones. Clin Exp Pharmacol Physiol. 2004;31(11):786–90.

    Article  CAS  PubMed  Google Scholar 

  54. Eldutar E, Kandemir FM, Kucukler S, Caglayan C. Restorative effects of Chrysin pretreatment on oxidant-antioxidant status, inflammatory cytokine production, and apoptotic and autophagic markers in acute paracetamol-induced hepatotoxicity in rats: an experimental and biochemical study. J Biochem Mol Toxicol. 2017;31(11): e21960.

    Article  CAS  Google Scholar 

  55. Mantawy EM, El-Bakly WM, Esmat A, Badr AM, El-Demerdash E. Chrysin alleviates acute doxorubicin cardiotoxicity in rats via suppression of oxidative stress, inflammation and apoptosis. Eur J Pharmacol. 2014;728:107–18.

    Article  CAS  PubMed  Google Scholar 

  56. Hanedan B, Ozkaraca M, Kirbas A, Kandemir FM, Aktas MS, Kilic K, et al. Investigation of the effects of hesperidin and chrysin on renal injury induced by colistin in rats. Biomed Pharmacother. 2018;108:1607–16.

    Article  CAS  PubMed  Google Scholar 

  57. Shayan M, Elyasi S. Cilastatin as a protective agent against drug-induced nephrotoxicity: a literature review. Expert Opin Drug Saf. 2020;19(8):999–1010.

    Article  CAS  PubMed  Google Scholar 

  58. Smiechowicz J, Barteczko B, Grotowska M, Kaiser T, Zielinski S, Kubler A. Severe acute respiratory distress syndrome complicating type A (H1N1) influenza treated with extracorporeal CO2 removal. Anestezjol Intens Ter. 2011;43(2):98–103.

    PubMed  Google Scholar 

  59. Christensen EI, Verroust PJ. Megalin and cubilin, role in proximal tubule function and during development. Pediatr Nephrol. 2002;17(12):993–9.

    Article  PubMed  Google Scholar 

  60. Vorobyeva A, Oroujeni M, Lindbo S, Hober S, Xu T, Liu Y, et al. Investigation of a pharmacological approach for reduction of renal uptake of radiolabeled ADAPT scaffold protein. Molecules. 2020;25(19):4448.

    Article  CAS  PubMed Central  Google Scholar 

  61. Hatcher H, Planalp R, Cho J, Torti FM, Torti SV. Curcumin: from ancient medicine to current clinical trials. Cell Mol Life Sci. 2008;65(11):1631–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Trujillo J, Chirino YI, Molina-Jijón E, Andérica-Romero AC, Tapia E, Pedraza-Chaverrí J. Renoprotective effect of the antioxidant curcumin: Recent findings. Redox Biol. 2013;1(1):448–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Morsy MA, Ibrahim SA, Amin EF, Kamel MY, Rifaai RA, Hassan MK. Curcumin ameliorates methotrexate-induced nephrotoxicity in rats. Adv Pharmacol Sci. 2013;2013: 387071.

    PubMed  PubMed Central  Google Scholar 

  64. Mahi-Birjand M, Yaghoubi S, Abdollahpour-Alitappeh M, Keshtkaran Z, Bagheri N, Pirouzi A, et al. Protective effects of pharmacological agents against aminoglycoside-induced nephrotoxicity: a systematic review. Expert Opin Drug Saf. 2020;19(2):167–86.

    Article  CAS  PubMed  Google Scholar 

  65. Edrees NE, Galal AAA, Abdel Monaem AR, Beheiry RR, Metwally MMM. Curcumin alleviates colistin-induced nephrotoxicity and neurotoxicity in rats via attenuation of oxidative stress, inflammation and apoptosis. Chem Biol Interact. 2018;294:56–64.

    Article  CAS  PubMed  Google Scholar 

  66. Li ZD, Luo J, Jia LH, Wang XY, Xun ZK, Liu M. Cytochrome C suppresses renal accumulation and nephrotoxicity of polymyxin B. Hum Exp Toxicol. 2019;38(2):193–200.

    Article  CAS  PubMed  Google Scholar 

  67. Erbatur ME, Sezen ŞC, Bayraktar AC, Arslan M, Kavutçu M, Aydın ME. Effects of dexmedetomidine on renal tissue after lower limb ischemia reperfusion injury in streptozotocin induced diabetic rats. Libyan J Med. 2017;12(1):1270021.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Song Y, Kim DH, Kwon TD, Han DW, Baik SH, Jung HH, et al. Effect of intraoperative dexmedetomidine on renal function after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy: a randomized, placebo-controlled trial. Int J Hyperther. 2019;36(1):1–8.

    Article  CAS  Google Scholar 

  69. Gu J, Sun P, Zhao H, Watts HR, Sanders RD, Terrando N, et al. Dexmedetomidine provides renoprotection against ischemia-reperfusion injury in mice. Critical Care (London, England). 2011;15(3):R153.

    Article  Google Scholar 

  70. Cho JS, Shim JK, Soh S, Kim MK, Kwak YL. Perioperative dexmedetomidine reduces the incidence and severity of acute kidney injury following valvular heart surgery. Kidney Int. 2016;89(3):693–700.

    Article  CAS  PubMed  Google Scholar 

  71. Talih G, Esmaoğlu A, Bayram A, Yazici C, Deniz K, Talih T. Does dexmedetomidine prevent colistin nephrotoxicity? Brazil J Anesthesiol. 2018;68(4):383–7.

    Article  Google Scholar 

  72. Vegt E, Eek A, Oyen WJG, de Jong M, Gotthardt M, Boerman OC. Albumin-derived peptides efficiently reduce renal uptake of radiolabelled peptides. Eur J Nucl Med Mol Imaging. 2010;37(2):226–34.

    Article  CAS  PubMed  Google Scholar 

  73. van Eerd JE, Vegt E, Wetzels JF, Russel FG, Masereeuw R, Corstens FH, et al. Gelatin-based plasma expander effectively reduces renal uptake of 111In-octreotide in mice and rats. J Nucl Med. 2006;47(3):528–33.

    PubMed  Google Scholar 

  74. Jones CM, Sumeray M, Heys A, Woolfson RG. Pseudo-proteinuria following gelofusine infusion. Nephrol Dial Transplant. 1999;14(4):944–5.

    Article  CAS  PubMed  Google Scholar 

  75. Sivanesan SS, Azad MAK, Schneider EK, Ahmed MU, Huang J, Wang J, et al. Gelofusine ameliorates colistin-induced nephrotoxicity. Antimicrob Agents Chemother. 2017;61(12):e00985-e1017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Rodríguez-Pérez C, García-Villanova B, Guerra-Hernández E, Verardo V. Grape Seeds proanthocyanidins: an overview of in vivo bioactivity in animal models. Nutrients. 2019;11(10):2435.

    Article  PubMed Central  CAS  Google Scholar 

  77. Yousef MI, Khalil D, Abdou HM. Neuro- and nephroprotective effect of grape seed proanthocyanidin extract against carboplatin and thalidomide through modulation of inflammation, tumor suppressor protein p53, neurotransmitters, oxidative stress and histology. Toxicol Rep. 2018;5:568–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Ding Y, Li H, Li Y, Liu D, Zhang L, Wang T, et al. Protective effects of grape seed proanthocyanidins on the kidneys of diabetic rats through the Nrf2 signalling pathway. Evid Based Complement Alternat Med. 2020;2020:5205903.

    PubMed  PubMed Central  Google Scholar 

  79. Ozkan G, Ulusoy S, Orem A, Alkanat M, Mungan S, Yulug E, et al. How does colistin-induced nephropathy develop and can it be treated? Antimicrob Agents Chemother. 2013;57(8):3463–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Catala L, Rius C, Garcia de Olalla P, Nelson JL, Alvarez J, Minguell S, et al. Pandemic A/H1N1 influenza: transmission of the first cases in Spain. Enferm Infecc Microbiol Clin. 2012;30(2):60–3.

    Article  PubMed  Google Scholar 

  81. Hajialyani M, Hosein Farzaei M, Echeverría J, Nabavi SM, Uriarte E, Sobarzo-Sánchez E. Hesperidin as a neuroprotective agent: a review of animal and clinical evidence. Molecules. 2019;24(3):648.

    Article  CAS  PubMed Central  Google Scholar 

  82. Tejada S, Pinya S, Martorell M, Capó X, Tur JA, Pons A, et al. Potential anti-inflammatory effects of hesperidin from the genus citrus. Curr Med Chem. 2018;25(37):4929–45.

    Article  CAS  PubMed  Google Scholar 

  83. Peng Pu. Protection mechanisms of hesperidin on mouse with insulin resistance. Zhongguo Zhong Yao Za Zhi. 2016;41(17):3290–5.

    Google Scholar 

  84. Chiba H, Uehara M, Wu J, Wang X, Masuyama R, Suzuki K, et al. Hesperidin, a citrus flavonoid, inhibits bone loss and decreases serum and hepatic lipids in ovariectomized mice. J Nutr. 2003;133(6):1892–7.

    Article  CAS  PubMed  Google Scholar 

  85. Liu XX, Yu DD, Chen MJ, Sun T, Li G, Huang WJ, et al. Hesperidin ameliorates lipopolysaccharide-induced acute lung injury in mice by inhibiting HMGB1 release. Int Immunopharmacol. 2015;25(2):370–6.

    Article  CAS  PubMed  Google Scholar 

  86. Sahu BD, Kuncha M, Sindhura GJ, Sistla R. Hesperidin attenuates cisplatin-induced acute renal injury by decreasing oxidative stress, inflammation and DNA damage. Phytomedicine. 2013;20(5):453–60.

    Article  CAS  PubMed  Google Scholar 

  87. Ahmad ST, Arjumand W, Nafees S, Seth A, Ali N, Rashid S, et al. Hesperidin alleviates acetaminophen induced toxicity in Wistar rats by abrogation of oxidative stress, apoptosis and inflammation. Toxicol Lett. 2012;208(2):149–61.

    Article  CAS  PubMed  Google Scholar 

  88. Wang S, He N, Xing H, Sun Y, Ding J, Liu L. Function of hesperidin alleviating inflammation and oxidative stress responses in COPD mice might be related to SIRT1/PGC-1α/NF-κB signaling axis. J Recept Signal Transduct Res. 2020;40(4):388–94.

    Article  CAS  PubMed  Google Scholar 

  89. Xu N, Zhang L, Dong J, Zhang X, Chen YG, Bao B, et al. Low-dose diet supplement of a natural flavonoid, luteolin, ameliorates diet-induced obesity and insulin resistance in mice. Mol Nutr Food Res. 2014;58(6):1258–68.

    Article  CAS  PubMed  Google Scholar 

  90. López-Lázaro M. Distribution and biological activities of the flavonoid luteolin. Mini Rev Med Chem. 2009;9(1):31–59.

    Article  PubMed  Google Scholar 

  91. Albarakati AJA, Baty RS, Aljoudi AM, Habotta OA, Elmahallawy EK, Kassab RB, et al. Luteolin protects against lead acetate-induced nephrotoxicity through antioxidant, anti-inflammatory, anti-apoptotic, and Nrf2/HO-1 signaling pathways. Mol Biol Rep. 2020;47(4):2591–603.

    Article  CAS  PubMed  Google Scholar 

  92. Xiong C, Wu Q, Fang M, Li H, Chen B, Chi T. Protective effects of luteolin on nephrotoxicity induced by long-term hyperglycaemia in rats. J Int Med Res. 2020;48(4):300060520903642.

    CAS  PubMed  Google Scholar 

  93. Hong X, Zhao X, Wang G, Zhang Z, Pei H, Liu Z. Luteolin treatment protects against renal ischemia-reperfusion injury in rats. Mediators Inflamm. 2017;2017:9783893.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Arslan BY, Arslan F, Erkalp K, Alagöl A, Sevdi MS, Yıldız G, et al. Luteolin ameliorates colistin-induced nephrotoxicity in the rat models. Ren Fail. 2016;38(10):1735–40.

    Article  CAS  PubMed  Google Scholar 

  95. Szymanska R, Pospisil P, Kruk J. Plant-derived antioxidants in disease prevention. Oxid Med Cell Longev. 2016;2016:1920208.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Bacanli M, Başaran N, Başaran AA. Lycopene: is it beneficial to human health as an antioxidant? Turk J Pharm Sci. 2017;14(3):311–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Dai C, Tang S, Deng S, Zhang S, Zhou Y, Velkov T, et al. Lycopene attenuates colistin-induced nephrotoxicity in mice via activation of the Nrf2/HO-1 pathway. Antimicrob Agents Chemother. 2015;59(1):579–85.

    Article  PubMed  CAS  Google Scholar 

  98. Bedir F, Kocaturk H, Turangezli O, Sener E, Akyuz S, Ozgeris FB, et al. The protective effect of lycopene against oxidative kidney damage associated with combined use of isoniazid and rifampicin in rats. Braz J Med Biol Res. 2021;54(8): e10660-e.

    Article  CAS  Google Scholar 

  99. Haghighipour S, Soltani R, Anjomshoa A. The protective effect of lycopene supplement against vancomycin-induced nephrotoxicity; a randomized double-blind placebo-controlled clinical trial. J Renal Inj Prev. 2020;9(4):e32-e.

    Article  CAS  Google Scholar 

  100. Raza Z, Naureen Z. Melatonin ameliorates the drug induced nephrotoxicity: molecular insights. Nefrologia. 2020;40(1):12–25.

    Article  PubMed  Google Scholar 

  101. Kostoglou-Athanassiou I. Therapeutic applications of melatonin. Ther Adv Endocrinol Metab. 2013;4(1):13–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Dudka Y, Zamorskii I, Shchudrova T, Petriuk A, Kopchuk T, Drachuk V. Nephroprotective mechanisms, therapeutic potential and perspective on melatonin use for drug-induced nephropathy. Med Sci. 2021;25(107):163–9.

    Google Scholar 

  103. Yousef JM, Chen G, Hill PA, Nation RL, Li J. Melatonin attenuates colistin-induced nephrotoxicity in rats. Antimicrob Agents Chemother. 2011;55(9):4044–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Šalamon Š, Kramar B, Marolt TP, Poljšak B, Milisav I. Medical and dietary uses of N-acetylcysteine. Antioxidants (Basel). 2019;8(5):111.

    Article  CAS  Google Scholar 

  105. Schwalfenberg GK. N-acetylcysteine: a review of clinical usefulness (an old drug with new tricks). J Nutr Metab. 2021;2021:9949453.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Sun Z, Fu Q, Cao L, Jin W, Cheng L, Li Z. Intravenous N-acetylcysteine for prevention of contrast-induced nephropathy: a meta-analysis of randomized, controlled trials. PLoS ONE. 2013;8(1):e55124-e.

    Article  CAS  Google Scholar 

  107. Huang S, You J, Wang K, Li Y, Zhang Y, Wei H, et al. N-acetylcysteine attenuates cisplatin-induced acute kidney injury by inhibiting the C5a receptor. BioMed Res Int. 2019;2019:4805853.

    PubMed  PubMed Central  Google Scholar 

  108. Chen N, Aleksa K, Woodland C, Rieder M, Koren G. N-Acetylcysteine prevents ifosfamide-induced nephrotoxicity in rats. Br J Pharmacol. 2008;153(7):1364–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Marenzi G, Assanelli E, Marana I, Lauri G, Campodonico J, Grazi M, et al. N-acetylcysteine and contrast-induced nephropathy in primary angioplasty. N Engl J Med. 2006;354(26):2773–82.

    Article  CAS  PubMed  Google Scholar 

  110. Ozyilmaz E, Ebinc FA, Derici U, Gulbahar O, Goktas G, Elmas C, et al. Could nephrotoxicity due to colistin be ameliorated with the use of N-acetylcysteine? Intensive Care Med. 2011;37(1):141–6.

    Article  CAS  PubMed  Google Scholar 

  111. Ceylan B, Ozansoy M, Kılıç Ü, Yozgat Y, Ercan Ç, Yıldız P, et al. N-acetylcysteine suppresses colistimethate sodium-induced nephrotoxicity via activation of SOD2, eNOS, and MMP3 protein expressions. Ren Fail. 2018;40(1):423–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Zhong S, Fan Y, Yan Q, Fan X, Wu B, Han Y, et al. The therapeutic effect of silymarin in the treatment of nonalcoholic fatty disease: a meta-analysis (PRISMA) of randomized control trials. Medicine. 2017;96(49):e9061-e.

    Article  CAS  Google Scholar 

  113. Karimi G, Vahabzadeh M, Lari P, Rashedinia M, Moshiri M. “Silymarin”, a promising pharmacological agent for treatment of diseases. Iran J Basic Med Sci. 2011;14(4):308–17.

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Dashti-Khavidaki S, Shahbazi F, Khalili H, Lessan-Pezeshki M. Potential renoprotective effects of silymarin against nephrotoxic drugs: a review of literature. J Pharm Pharm Sci. 2012;15(1):112–23.

    Article  PubMed  Google Scholar 

  115. Guzel S, Sahinogullari ZU, Canacankatan N, Antmen SE, Kibar D, Coskun YB. Potential renoprotective effects of silymarin against vancomycin-induced nephrotoxicity in rats. Drug Chem Toxicol. 2020;43(6):630–6.

    Article  CAS  PubMed  Google Scholar 

  116. Sedighifard Z, Roghani F, Bidram P, Harandi SA, Molavi S. Silymarin for the prevention of contrast-induced nephropathy: a placebo-controlled clinical trial. Int J Prev Med. 2016;7:23.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Hassan SMS, Youakim MF, Rizk AAE, Thomann C, Ahmad Z. Does silybin protect against toxicity induced by polymyxin E in rat kidney? Neurourol Urodyn. 2017;36(5):1278–87.

    Article  CAS  PubMed  Google Scholar 

  118. Hassan SS, Thomann C, Ettarh R, Ahmad Z. Possible protective role of silybin against polymyxin E-induced toxic effect in rat kidneys: a biochemical approach. Neurourol Urodyn. 2017;36(8):2003–10.

    Article  CAS  PubMed  Google Scholar 

  119. Dumludag B, Derici MK, Sutcuoglu O, Ogut B, Pasaoglu OT, Gonul II, et al. Role of silymarin (Silybum marianum) in the prevention of colistin-induced acute nephrotoxicity in rats. Drug Chem Toxicol. 2020. https://doi.org/10.1080/01480545.2020.1733003.

    Article  PubMed  Google Scholar 

  120. Schaffer S, Azuma J, Takahashi K, Mozaffari M. Why is taurine cytoprotective? Adv Exp Med Biol. 2003;526:307–21.

    Article  CAS  PubMed  Google Scholar 

  121. Schaffer S, Kim HW. Effects and mechanisms of taurine as a therapeutic agent. Biomol Ther (Seoul). 2018;26(3):225–41.

    Article  CAS  Google Scholar 

  122. Baliou S, Adamaki M, Ioannou P, Pappa A, Panayiotidis MI, Christodoulou I, et al. Ameliorative effect of taurine against diabetes and renal-associated disorders. Med Int. 2021;1(2):1–19.

    Article  Google Scholar 

  123. Heidari R, Behnamrad S, Khodami Z, Ommati MM, Azarpira N, Vazin A. The nephroprotective properties of taurine in colistin-treated mice is mediated through the regulation of mitochondrial function and mitigation of oxidative stress. Biomed Pharmacother. 2019;109:103–11.

    Article  CAS  PubMed  Google Scholar 

  124. Jacob RA, Sotoudeh G. Vitamin C function and status in chronic disease. Nutr Clin Care. 2002;5(2):66–74.

    Article  PubMed  Google Scholar 

  125. Grosso G, Bei R, Mistretta A, Marventano S, Calabrese G, Masuelli L, et al. Effects of vitamin C on health: a review of evidence. Front Biosci (Landmark edition). 2013;18:1017–29.

    Article  CAS  Google Scholar 

  126. Rastogi S, Gupta S, Haldar C, Chandra D. Nephroprotective effect of melatonin and L-ascorbic acid (Vitamin-C) against ampicillin- induced toxicity in Funambulus pennanti. Egypt J Basic Appl Sci. 2020;7(1):8–19.

    Google Scholar 

  127. Cho MH, Kim SN, Park HW, Chung S, Kim KS. Could vitamin E prevent contrast-induced acute kidney injury? A systematic review and meta-analysis. J Korean Med Sci. 2017;32(9):1468–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Yousef JM, Chen G, Hill PA, Nation RL, Li J. Ascorbic acid protects against the nephrotoxicity and apoptosis caused by colistin and affects its pharmacokinetics. J Antimicrob Chemother. 2012;67(2):452–9.

    Article  CAS  PubMed  Google Scholar 

  129. Sirijatuphat R, Limmahakhun S, Sirivatanauksorn V, Nation RL, Li J, Thamlikitkul V. Preliminary clinical study of the effect of ascorbic acid on colistin-associated nephrotoxicity. Antimicrob Agents Chemother. 2015;59(6):3224–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Rizvi S, Raza ST, Ahmed F, Ahmad A, Abbas S, Mahdi F. The role of vitamin e in human health and some diseases. Sultan Qaboos Univ Med J. 2014;14(2):e157–65.

    PubMed  PubMed Central  Google Scholar 

  131. Mohd Zaffarin AS, Ng SF, Ng MH, Hassan H, Alias E. Pharmacology and pharmacokinetics of vitamin E: nanoformulations to enhance bioavailability. Int J Nanomed. 2020;15:9961–74.

    Article  CAS  Google Scholar 

  132. Liu P, Feng Y, Wang Y, Zhou Y, Zhao L. Protective effect of vitamin E against acute kidney injury. Bio-Med Mater Eng. 2015;26(Suppl 1):S2133–44.

    Article  CAS  Google Scholar 

  133. Ghlissi Z, Hakim A, Mnif H, Kallel R, Zeghal K, Boudawara T, et al. Effect of vitamin E on reversibility of renal function following discontinuation of colistin in rats: histological and biochemical investigations. Saudi J Kidney Dis Transpl. 2018;29(1):10–8.

    Article  PubMed  Google Scholar 

  134. Ghlissi Z, Hakim A, Mnif H, Zeghal K, Rebai T, Boudawara T, et al. Combined use of vitamins E and C improve nephrotoxicity induced by colistin in rats. Saudi J Kidney Dis Transpl. 2018;29(3):545–53.

    Article  PubMed  Google Scholar 

  135. Bader Z, Waheed A, Hashim R. Establishment and comparison of nephrotoxicity associated with the use of various doses of conlistin in rabbits. Pak Armed Forces Med J. 2016;66(2):180–4.

    Google Scholar 

  136. Bader Z, Waheed A, Bakhtiar S, Khan IM. Alpha-tocopherol ameliorates nephrotoxicity associated with the use of colistin in rabbits. Pak J Pharm Sci. 2018;31(2):463–7.

    CAS  PubMed  Google Scholar 

  137. Moradi O, Sahraei Z, Asadi S. From bench to bedside: colistin nephrotoxicity and vitamin. Clin Microbiol Infect Dis. 2019. https://doi.org/10.15761/CMID.1000161.

    Article  Google Scholar 

Download references

Acknowledgements

The authors would like to thank Ms. A. Keivanshekouh at the Research Consultation Center (RCC) of Shiraz University of Medical Sciences for improving the use of English in the manuscript.

Funding

No funding support.

Author information

Authors and Affiliations

Authors

Contributions

EM and MM searched the literature, extracted the relevant articles, and drafted the manuscript. AV evaluated the quality of recruited studies, and critically edited the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Ehsan Mirzaei or Afsaneh Vazin.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mirjalili, M., Mirzaei, E. & Vazin, A. Pharmacological agents for the prevention of colistin-induced nephrotoxicity. Eur J Med Res 27, 64 (2022). https://doi.org/10.1186/s40001-022-00689-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40001-022-00689-w

Keywords