Next Article in Journal
Molecular Characterization and Antibiotic Susceptibility of Non-PCV13 Pneumococcal Serotypes among Vaccinated Children in Cape Coast, Ghana
Next Article in Special Issue
Gut Microbiota and Metabolome Changes in Three Pulmonary Hypertension Rat Models
Previous Article in Journal
A Promising Use of Trimethyl Chitosan for Removing Microcystis aeruginosa in Water Treatment Processes
Previous Article in Special Issue
Gut Microbiota Profiles in Children and Adolescents with Psychiatric Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Protective Effects of Nutraceutical Components in Methotrexate-Induced Toxicity Models—An Overview

by
Gheorghe-Eduard Marin
1,
Maria-Adriana Neag
2,*,
Codrin-Constantin Burlacu
1,* and
Anca-Dana Buzoianu
2
1
Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
2
Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
*
Authors to whom correspondence should be addressed.
Microorganisms 2022, 10(10), 2053; https://doi.org/10.3390/microorganisms10102053
Submission received: 28 September 2022 / Revised: 11 October 2022 / Accepted: 13 October 2022 / Published: 18 October 2022
(This article belongs to the Special Issue Gut Microbiota in Disease)

Abstract

:
There are multiple concerns associated with methotrexate (MTX), widely recognized for anti-neoplastic and anti-inflammatory effects in life-threatening disease conditions, i.e., acute lymphoblastic leukemia, non-Hodgkin’s lymphoma, psoriasis, and rheumatoid arthritis, due to long-term side effects and associated toxicity, which limits its valuable potential. MTX acts as an inhibitor of dihydrofolate reductase, leading to suppression of purine and pyrimidine synthesis in high metabolic and turnover cells, targeting cancer and dysregulated immune cells. Due to low discrimination between neoplastic cells and naturally high turnover cells, MTX is prone to inhibiting the division of all fast-dividing cells, causing toxicity in multiple organs. Nutraceutical compounds are plant-based or food-derived compounds, used for their preventive and therapeutic role, ascertained in multiple organ dysfunctions, including cardiovascular disease, ischemic stroke, cancer, and neurodegenerative diseases. Gut microbiota and microbiota-derived metabolites take part in multiple physiological processes, their dysregulation being involved in disease pathogenesis. Modulation of gut microbiota by using nutraceutical compounds represents a promising therapeutic direction to restore intestinal dysfunction associated with MTX treatment. In this review, we address the main organ dysfunctions induced by MTX treatment, and modulations of them by using nutraceutical compounds. Moreover, we revealed the protective mechanisms of nutraceuticals in MTX-induced intestinal dysfunctions by modulation of gut microbiota.

1. Introduction

Antifolate drugs represent an old class of pharmaceuticals which interfere with folate metabolism and have been used to treat proliferative disorders, e.g., acute lymphocytic leukemia, breast cancer, and parasitic, and microbial diseases [1,2,3].
Methotrexate (MTX), an antimetabolite of folic acid, which acts as an inhibitor of dihydrofolate reductase (DHFR), exhibited anti-neoplastic and immunomodulatory effects in the area of malignant and non-malignant diseases [4]. Though the mechanisms behind its anti-inflammatory effects against psoriasis, rheumatoid arthritis (RA) and anti-cancer effects upon blood malignancies and other neoplasia are quite understood, the precise therapeutic effect of MTX is not clarified [5].
Although MTX therapy attracted attention in preclinical and clinical settings of numerous inflammatory and cancer disorders, its associated adverse effects and the toxicity on multiple organs related to MTX remain an important concern and a cause of drug withdrawal [6,7].
Nutraceutical-based therapy, first described by DeFelice in 1989, refers to food-based compounds which exhibited multifaced roles in preventing and treating life-threatening diseases [8,9]. Nutraceutical compounds are represented by a wide range of nutrients and phytochemicals, such as dietary bioactive peptides and lipids, fat soluble vitamins, amino-acids, micronutrients [10]. Some of the most common nutraceuticals, such as resveratrol [11], curcumin [12], coenzyme Q10 (ubiquinone), α-lipoic acid [13], β-carotene [14], quercetin [15] have been shown to alleviate oxidative stress, and apoptosis-induced cellular injury, targeting pro-inflammatory signaling pathways, by acting on nuclear and mitochondrial targets [16,17].
Growing evidence suggested the protective effects of nutraceuticals in multiple in vitro and vivo disease models, including ischemia–reperfusion injury [18], cardiovascular diseases [19], ischemic stroke [20], cancer [21], neurodegenerative diseases [22], and inflammatory bowel diseases [23].
Within MTX therapy, preclinical and clinical studies reported long-term side effects and toxicity effects on multiple organs and a promising therapeutic strategy aimed at restoring the toxicity of MTX is currently needed. The negative effects of MTX treatment are reflected on multiple organs, including hepatic fibrosis, acute lung injury, dysregulation of gut microbiota and nephrotoxicity [24,25,26,27]. There are a wide range of short- and long-term side effects, ranging from nausea, drowsiness, liver enzymes elevation, to renal insufficiency, hepatic fibrosis/cirrhosis, pulmonary fibrosis and life-threatening blood disorders, i.e., pancytopenia and aplastic anemia [5].
MTX toxicity and MTX side effects restrict its clinical uses to moderate and severe forms of disease or drug-resistant diseases [28]. Thus, finding promising natural components aimed at ameliorating toxicity and side effects of MTX represents a future direction in MTX therapy. In MTX acute nephrotoxicity, the management is focused on fast intravenous fluid hydration, urine alkalinization, and rescue of folic acid [29].
Multiple preclinical studies explored the anti-inflammatory and immunomodulatory effects of several Nutraceuticals in liver and kidney injury, acute lung injury and gut microbiota dysregulation induced by treatment with MTX [30,31,32]. An in-depth overview of the mechanism of action of nutraceuticals in experimental MTX toxicity models could provide further directions in MTX therapy. This review aims to overview the main nutraceuticals involved in the beneficial effects against MTX-induced organ injuries and to propose promising nutraceuticals as adjuvant therapy in patients treated with MTX.

2. MTX as a Disease-Modifying Agent

DHFR is a crucial enzyme involved in the reductive process of dihydrofolic acid to tetrahydrofolic acid, an essential oxido-reductive process required for the de novo synthesis of purines, pyrimidines, and certain amino acids [33]. These chemical components are essential for cell proliferation and cell growth [33].
MTX is an antimetabolite of folic acid, which was first approved by FDA for treating only life-threatening malignancies, treatment-resistant or treatment non-respondent forms of psoriasis and RA, and also those forms with severe course of disease [4]. The mechanism of action underlying anti-neoplastic and immunomodulatory effects of MTX is based on inhibition of DHFR, thus promoting suppression of purine and pyrimidine synthesis in cell proliferation [34,35].
MTX binds to the same active site of DHFR, with a 1000-fold increase in the affinity of DHFR over that of dihydrofolic acid [36]. Binding of the MTX to its substrate, DHFR, acts in a NADPH-dependent manner [36,37]. This dependency could also play a critical role for the selective toxicity of MTX within malignant cells, malignant cells revealing a higher NADPH/NADP ratio compared to the intact and viable cells [36,38].
However, the anti-inflammatory mechanism of MTX does not proceed by inhibiting DHFR enzyme and in-depth mechanistic data of anti-inflammatory features are still needed. Mounting experimental studies proposed several mechanisms of action of MTX treatment, including counteracting reactive oxygen species (ROS) production [39,40,41], inhibition of pyrimidine pathway enzymes [42], release of adenosine [43,44], and regulation of cytokine production [45,46].
Multiple ways of administration, i.e., orally, intravenously, intramuscularly, or intrathecally allow a good bioavailability and an effective mechanism of action of MTX in multiple pathological contexts [47].
After oral administration, it is actively absorbed within proximal jejunum, the enteric transport being mediated by proton-coupled folate transporter (PCFT/SLC46A1) [5]. Even in the absorption phase, a small percentage of 5% of MTX is metabolized to 4-amino-4deoxy-N10-methylpterrroic acid, an inactive metabolite of MTX [5].
Within the cell membrane, the cellular uptake and efflux are mediated by specific transporters, including reduced folate carrier (RFC1), the proton-coupled folate transporter (PCFT), and ATP-binding cassette proteins (ABCC) [5]. The bioavailability ranges from 30% to 90% [5,48], varying widely among different subtypes of patients and it decreases with increasing dose, suggesting the saturation of the active transporters with MTX [49,50].
The distribution of MTX to the body’s tissues mainly depends on the reduced folate carrier 1 (RFC1), involved in the transport of reduced folates such as 5-methyl THF [51]. Once distributed to cells and tissues, MTX is rapidly converted to MTX polyglutamates (MTX-PG) by folylpolyglutamate synthase, which binds six glutamate residues to MTX, therefore sustaining its intracellular retention and increasing enzyme binding affinity [52]. Some MTX is hydrolyzed to 7-hydroxymethotrexate by aldehyde oxidase in the liver [53].
Renal excretion constitutes the primary elimination route, mainly in the intact form (more than 80%) and 3% as the 7-hidroylated form [54]. MTX is recycled by enterohepatic circulation, about 8% and 26% of plasmatic MTX being excreted in the bile [52,55,56]. The clinical use of MTX ranges from neoplastic diseases, such as acute lymphoblastic leukemia, acute promyelocytic leukemia, non-Hodgkin’s lymphoma, to epidermoid cancers of the head and neck, early-stage breast cancer, osteosarcoma and several types of gestational trophoblastic neoplasia [57,58].
In addition to anti-cancer effects, MTX poses anti-inflammatory and immunomodulatory effects. Inflammatory diseases, such as inflammatory bowel diseases, vasculitis, systemic lupus erythematosus, multiple sclerosis, transplantation surgeries could benefit from MTX therapy [59,60]. As a disease-modifying agent, MTX is used for treating RA, juvenile idiopathic arthritis (JIA), and psoriasis [57,58,61].
Despite its widespread clinical use and usefulness, MTX comes with pitfalls, including short- and long-term side effects such as hepatotoxicity, nephrotoxicity, and leukopenia, which can predispose patients to severe infections [57,61]. An overview of the main pharmacological features of MTX as a therapeutic agent in non-malignant and malignant diseases is provided in Table 1.

3. Gut Microbiota-Related Changes following Treatment with MTX

3.1. Involvement of Gut Microbiota in Health and Disease

Microbiota is part of our human microecosystem, which poses multiple regulatory roles in our body in both health and pathological conditions [63]. Widely distributed within the organism, microbial species colonize multiple organs and cavities, i.e., oral cavity, gut, lung, skin, vagina, etc. [64]. Microbiota consists of a large abundance and density of microorganisms, including bacteria, fungi, viruses, protozoa, and archaeal, which live in symbiotic or parasitic relationships [64,65].
With the greatest diversity and abundance, the gut microbiota harbors an extensive community of over 100 trillion microbial cells, with a 150-fold increase in the gut-regulatory genes compared to human genome [66,67]. The two most dominant intestinal phyla include Firmicutes and Bacteroidetes, with Lactobacillus, Faecalibacterium, Clostridium, Enterococcus accounting for dominant genera of Firmicutes and Bacteroides, and Prevotella, reaching the most of Bacteroidetes genera [68].
Gut microbiota and its derived metabolites exert multiple essential roles in the body, from immune modulation, to metabolic, digestive functions and biosynthesis of active compounds [63,69]. Compositional changes in gut bacterial species have been reported in large spectrum of disease conditions, i.e., cardiovascular dysfunctions [70], stroke [71], neurodegenerative and cognitive disorders [72,73], cancer-related disease and autoimmune disorders [74,75]. Aging, diet, smoking and patient-associated comorbidities, i.e., diabetes, obesity are influencing factors with a decisive impact on gut bacterial profile [76,77,78,79].

3.2. The Role and Protective Effects Exhibited by Microbiota

Gut microbiota interacts in a bidirectional manner with multiple organ systems, influencing each other; systemic changes contribute to intestinal dysbiosis, and also intestinal microbiota dysregulation is involved in disease pathogenesis and organ dysfunction [80]. Evolving research studies evaluated the relationship between dysregulation of gut microbiota and dysfunctions of other organ systems, in relation with disease pathogenesis and therapeutic insights. Referring to this interaction, they are classified as the gut–liver axis [81], the gut–brain axis [82], and the gut–liver–brain axis [83].
Gut microbiota exerts immunomodulatory and anti-inflammatory functions and interact in a bidirectional manner with multiple organ systems through its key mediators, microbiota-derived metabolites, specifically short-chain fatty acids, (SCFAs) [84]. SCFAs, consisting of acetate, propionate, and butyrate are carboxylic acids, formed by chains of 2–6 carbon atoms, which are produced by anaerobic bacterial fermentation of complex dietary carbohydrates within intestinal lumen [85]. The source of biosynthesis is represented by dietary fibers such as plant cell wall-derived polysaccharide, soluble oligosaccharide, and also endogenous molecules, such as mucin [85,86]. Different microbial taxa are responsible for SCFAs synthesis: acetate formation is mostly mediated by enteral bacteria, such as Prevotella spp., Bifidobacterium spp., Bacteroides spp., Clostridium spp., Ruminococcus spp., and Streptococcus spp. [87]; propionate formation is regulated by few bacterial genera, Salmonella enterica serovar Typhimurium and R. inulinivorans [88,89]; and dominant species responsible for butyrate formation are Coprococcus, F. prausnitzii, E. hallii, E. rectale, Ruminococcus bromii [90,91].
After synthesis in the intestinal lumen, the cellular uptake of SCFAs proceeds via specific transporters expressed on epithelial cells of the small intestine and colon, in a manner dependent on Na+/H or Cl/HCO3 co-transporters [86]. The highest SCFAs concentration is reached in the cecum, followed by descending colon and ileum, in relation to biodiversity and composition of microbial species [92]. After entering into intestinal epithelial cells, SCFAs exert regulatory roles by interacting with specific receptors, G protein-coupled receptors (GPCR) and or histone deacetylases (HDACs) [93,94,95]. At the nuclear level, propionate and butyrate induce transcriptional regulation and post-translational modification of histones, by targeting lysine and histone deacetylase (K/HDAC) [86].
SCFAs could interact with different immune cells, contributing to innate and adaptive immune homeostasis [84,93,94,95]. SCFAs, acetate or propionate stimulate bone marrow hematopoiesis, along with activation of Th cells differentiation and increasing expression of specific chemoattractant molecules on immune cells [96,97]. Local or systemic immune responses are mediated by SCFAs, butyrate inhibiting pro-inflammatory pathways, such as NF-κB, thus decreasing pro-inflammatory cytokines, i.e., TNF-α, IL-6, IL-12 and activating anti-inflammatory cytokines, i.e., IL-10 [92].
Changes in the abundance and diversity of gut microflora species, known as “dysbiosis”, are reported in multiple intestinal inflammatory dysfunctions, providing high value as theragnostic tools in disease settings [98]. Evolving research studies depicted metabolomic and microbiome profiling data of serum and fecal patient samples, giving insights into bacterial-based biomarkers as promising predictors of therapeutic response and clinical outcome [99,100].

3.3. The Role of Microbiota for Promoting an Intact Epithelial Cell Barrier

Intestinal stem cell niches (ISCN) are at the basis of epithelial barrier renewal and maintaining of intestinal barrier integrity [101]. By sustaining proliferative and differentiation processes in a dynamic manner, ISCNs give rise to specialized epithelial cells: enterocytes, enteroendocrine cells, Paneth cells, microfold (M) cells and goblet cells [102]. Epithelial cell surface expresses specific receptors, namely pattern recognition receptors (PRRs), which bind to microbial ligands expressed on enteric commensal bacteria to maintain intestinal epithelial homeostasis against pathogen bacteria species and other intestinal insults [103,103]. Therefore, bacterial molecules and its associated metabolites are recognized by several PRRs, consisting of Toll-like receptors (TLRs), which are expressed on epithelial and immune cells, aimed at protecting and immunomodulating intestinal surface barrier [103,103]. Specific conserved motifs in epithelial cell receptors, including TLR/MyD88 and Nucleotide oligomerization domain (NOD)-like receptors (NLRs), exhibited protective roles against invasion of gut microbiota species of epithelial cells, by synthesis of antimicrobial factors, such as defensins and cathelicidins [103,104,105]. Inflammatory responses triggered by commensal bacteria are prevented by sequestration of microflora by mucosal epithelial cells, preventing the “deleterious” activation of TLRs by beneficial microflora [106,107]. In a model of chemical colitis of pathogen-free mice, Rakoff-Nahoum et al. found that activation of TLR2 and TLR4 by enteric commensal bacteria is necessary for protection upon mucosal injuries and mice-related mortality [103]. In addition to protective effects of epithelial cell barrier, PRRs by interacting with microbial components promoted pathways involved in cellular proliferation [105].
The breakdown of intestinal barrier integrity is involved in multiple intestinal dysfunctions, such as irritable bowel syndrome, metabolic syndrome, inflammatory bowel diseases, and necrotizing enterocolitis, and obesity [108,109,110]. The functional state of intestinal barrier function is a hallmark of gut homeostasis, depending on multiple influencing factors, (e.g., cellular, biochemical, immunological, and bacterial) [105]. Commensal gut bacterial species sustain both maintenance and/or restoring processes of epithelial gut barrier, by promoting cellular processes of differentiation, proliferation and migration and survival [111,112]. SCFAs elicited several modulatory roles to maintain an intact intestinal epithelial barrier at different levels.
Butyrate constitutes the main energy source of colonocytes, contributing to their structural integrity [86]. SCFAs by interacting with GPCRs, (e.g., butyrate-GPR109a, acetate, propionate, butyrate—GPR43, GPR41) induce gene expression regulation and signaling transduction, promoting epithelial cell differentiation, apoptosis, and proliferation [113]. SCFAs sustain epithelial barrier integrity, by reducing epithelial permeability (e.g., modulation of HIF, STAT3 signaling pathways) [86], regulating tight junction (TJ) proteins [105], and mucus layer thickness (e.g., targeting MUC expression) [114] and promoting antimicrobial peptides synthesis [115].
The distinctive features of the intestinal surface, such as the “intact” barrier and, at the same time, being selective and dynamically permeable, are conferred by intercellular connections, consisting of TJs, adherens junctions, the desmosomes, and the gap junctions [116,117]. Gut microbiota bacterial species modulate expression of several intercellular connections, therefore maintaining intestinal epithelial barrier homeostasis. In Caco-2 and HT-29 cells cultured on human milk oligosaccharides, B. infantis and B. bifidum regulated TJs proteins such as ZO-1 and occludin expression, whereas L. rhamnosus and Faecalibacterium prausnitzii modulated Occludin and E-cadherin expression to alleviate impaired gut barrier function in a mouse model [118,119].

3.4. The Influence of MTX on Gut Microbiota

The first experimental report describing the response of small-intestine epithelial cell under MTX was mentioned by Taminiau et al., in 1980 [120]. The authors reported that, under an intravenous dose of 30 mg/kg for 24 and 48 h, MTX exhibited suppressive effects on cellular mitoses in crypts, decreased intestinal villi, and also inactivated thymidine kinase activity, an essential enzyme within epithelial cells from intestinal crypts [120]. In a metabolomic and microbiome profiling study conducted in male Sprague Dawley rats treated with MTX, authors depicted changes in fecal samples metabolites and compositional bacterial changes at different time-points after MTX treatment [26]. Changes in the metabolomic profile of fecal samples, specifically 2,4-diamino-N(10)-methylpteroic acid (DAMPA) concentration in faces have been positively correlated with abundance of Prevotellaceae, Anaeroplasmataceae, Lactobacillaceae and Ruminococcaceae, and negatively correlated with bacterial species of Deferribacteraceae and Coriobacteriaceae [26]. Moreover, urine sample of rats exhibited methionine sulfoximine metabolites, which was associated with Ruminococcaceae species at 48 h. Bacterial abundance of Prevotellaceae and Anaeroplasmataceae was associated with two metabolites under MTX treatment, fecal glutamate and urine 5-hydroxyindole acetic acid at 48 h. Up to 24 metabolic compounds, i.e., dipeptides, tripeptide, organic acids, have been shown to be altered in fecal samples of rats upon MTX treatment in a dose-dependent manner [26]. Gut microbiota of rats exhibited changes in bacterial species after MTX treatment, with an enrichment of the abundance of Firmicutes over the Bacteroidetes, when treating rats with low doses, with an reverse trend at high doses [26]. Changes of gut microbial signature of rats under MTX treatment were driven by an increase in Peptostreptococcaceae and Porphyromonadaceae and a decrease in the relative abundance of Ruminococcaceae, Erysipelotrichaceae [26]. Alteration in gut microbial composition has been revealed in MTX-induced liver injury, which was restored after 40 mg/kg of Magnesium Isoglycyrrhizinate (MgIG) supplementation [32]. Under a long course of 30 days of MTX treatment, gut microbiota in mice exhibited an increase in bacterial proliferation of Muribaculaceae, and also a decrease in Lactobacillus abundance [32]. Within colon permeability, MTX treatment induces alteration in expression of TJs, i.e., ZO-1 and Claudin-1 and cell adhesion protein, E-cadherin, suggesting the impact of dysregulated gut microflora on the expression of intercellular connections [32]. MgIG could reverse the dysregulated expression levels of these proteins induced by MTX treatment, mitigating epithelial changes in leaky gut [32].
In addition to the alteration of gut microbiota induced by MTX, bacterial changes in gut microflora occurred during RA, for which MTX is indicated, suggesting the influence of disease progression on gut microbiota and further influencing MTX efficacy. In 212 fecal samples of RA patients, the bacterial proliferation of Haemophilus spp. was decreased, being negatively correlated with serum autoantibodies levels, whereas RA patients exhibited an enrichment of Lactobacillus salivarius [121]. By DNA sequencing of stool samples of 29 children with JIA, Öman et al. depicted distinct signature of gut microbiota composition upon MTX treatment, with an increase in Subdoligranulum and under-representation of Rikenellaceae, Veillonellaceae, Bacteroidales_S24-7_group [122]. Moreover, the SCFAs levels differ in MTX group compared with Etanercept group, with an increase level of iso-butyrate in fecal samples of 15 JIA patients treated with MTX [122]. Multiple experimental studies examined the potential of pre/probiotics, vitamins, plant-based extracts to alleviate structural changes in the intestine architecture upon difference disease models [123]. Changes in the bacterial profile of gut microbiota upon MTX treatment suggest the role of adjuvant therapies in restoring the intestinal bacterial balance, nutraceutical compounds being promising candidates.
Several bacterial changes in gut microbiota of animal models or patients treated with MTX are viewed in Table 2.

4. Nutraceuticals Use to Counteract MTX Toxicity in Experimental Models

Given the plethora of side effects, both acute and chronic, the management of MTX toxicity needs to incorporate strategies aimed to prevent the side effects associated with long-term use and adjusting treatment plans for acute toxicity [5,47]. The schematic bellow (Figure 1) represents an overview of the main molecular and cellular mechanisms involved in MTX multi-organ toxicity according to experimental models.
Preventing the toxicity associated with long-term use could be achieved by using the lowest effective dose, monitoring the blood concentration of MTX by using commercially available immunoassays [125], and co-treatment of MTX with natural compounds which can prevent buildup of MTX, slow down or prevent side effects.
Although the therapeutic effect is obtained by administering the effective dose therapy [47,125], dose individualization considering both the severity of disease and the MTX metabolic rate could prevent toxicity events [47,61].
Therapeutic drug monitoring (TDM) has become a staple for many therapies that utilize drugs with a narrow therapeutic index such as MTX. TDM can be used for two reasons in MTX treatment: to ensure that MTX concentration is high enough to be therapeutically effective, and to minimize dose-dependent toxic events [47,126,127]. MTX monitoring can be performed in a clinical setting using commercially available immunoassay kits, thus minimizing the difficulty and time requirement of such operation [125].
Several combined therapeutic strategies aimed at preventing both acute toxicity and side effects associated with long-term use have been recently proposed. One such example is the use of folic acid or folinic acid (5-formyl derivative of tetrahydrofolic acid, Leucovorin) supplements in order to prevents folate depletion for healthy tissue, and thus prevent hematologic side effects [127,128], but also allow the use of higher doses of MTX in order to achieve a better therapeutic result without significantly increasing the chance for adverse events [125,127]. Another example is the use of urine alkalinization compounds, in order to increase the excretion of MTX and prevent nephrotoxicity [127,128]. Moreover, urine alkalinization can be used for episodes of acute renal toxicity that result in acute kidney insufficiency (AKI) together with glucarpidase (a recombinant bacterial carboxypeptidase G2). Creating a pH of 7.5 will reduce the amount of MTX and its 7-hydroxilated form crystalizing, further improving renal excretion, while glucarpidase will cleave MTX into DAMPA and glutamate, two non-toxic metabolites, thus rapidly lowering MTX plasma concentration in patients with AKI [127,128,129].
Nutraceuticals are defined as plant or food-based compounds generally sold in medicinal forms, for both preventive and therapeutic roles [8,9]. Additionally, the beneficial roles in physiological or disease conditions for providing protection against chronic disease have been demonstrated in multiple preclinical and clinical studies [130]. Under this definition several items are covered, such as vitamins, minerals, herbs and other botanicals extracts, to amino acids, prebiotics, probiotics, and other dietary substances [131]. A considerable amount of the current research into nutraceuticals focuses on plant-based therapeutics, through the use of concentrates and purified extracts, due to the large variety of active compounds found in plants [131,132].
Emerging preclinical and clinical studies ascertained the role of nutraceuticals in multiple disease contexts, including cardiovascular diseases, diabetes, cancer, allergies, and visual disorders [133,134,135,136,137,138,139,140]. Through these studies nutraceuticals make themselves out as a viable and simple method not only of treatment, but also of prevention of certain diseases.
Multiple chemotherapeutical agents frequently used in cancer therapy proved to have multi organ long-term side effects and toxicity that hinders their therapeutic efficacy and the life expectancy of oncologic patients [125,141,142,143,144,145,146,147,148]. These pitfalls of chemotherapeutic agents prompt the need for therapeutic strategies aimed at reducing the toxicity of cancer drugs. Plant-based therapies have been established as a protective measure in cancer therapy toxicity models [149,150,151].
MTX is part of those chemotherapeutical agents to have multi organ long-term side effects, with studies showing significant hepatotoxicity, nephrotoxicity, and potential pulmonary fibrosis as some of the most dangerous adverse effect after employing MTX [127,128], and list them as a common cause of therapy withdrawal [47,57,61,128]. Thus, MTX could benefit from nutraceutical co-treatment in order to reduce or prevent certain adverse events.

4.1. Hepatotoxicity

The underling molecular mechanism behind MTX-induced hepatotoxicity is not yet completely understood; however, considerable experimental and clinical evidence proposed oxidative stress as a contributing factor through the increased generation of ROS, alongside with decreased antioxidant defense systems [152,153,154].
Prevention of MTX-induced hepatotoxicity using nutraceuticals has been extensively studied, consisting in a wide variety of nutraceutical categories, including vitamins, nutrients and dietary supplements, with varying levels of effectiveness [155,156,157].
In an experimental mouse model of MTX toxicity, pre-treatment with epicatechin at doses of 25, 50 and 100 mg/kg have been shown to have protective effects. The beneficial effects elicited by epicatechin compound might be explained by improving the antioxidant defense system, posing anti-inflammatory effects, and alleviating histopathological changes [30]. In another experimental model, treatment with thymoquinone at a dose of 10 mg/kg/day for 10 days has been shown to have protective effects in rats treated with MTX, by regulating antioxidant, anti-nitrosative, anti-inflammatory, and antiapoptotic mechanisms [158].
An amount of 50 and 100 mg/kg/day of ferulic acid has been shown to have protective effects in MTX-induced mice model of oxidative stress injury [159].
Rhein treatment in rats proved to be another protective measure against MTX-induced hepatotoxicity, as reported by Bu et al. [160]. The underling mechanism of this effect was linked to the upregulation of several signaling pathways, such as nuclear factor (erythroid-derived 2) like 2 factor (nrf2), B-cell lymphoma-2 (Bcl-2), heme oxygenase 1 (HO-1), and glutamate-cysteine ligase catalytic subunit (GCLC) [160].
Gossypin, an anti-tumoral and anti-inflammatory nutraceutical, was examined by Mohamed et al. in a rat model of MTX toxicity. The anti-inflammatory mechanism responsible for the reduction in inflammatory cell infiltration of the liver proceeds by inhibiting the TGF-β/NFκ-B signaling pathway [161].
Berberine is a plant-based nutraceutical that has been extensively tested in multiple disease models for its therapeutic effects. Using a MTX-induced hepatotoxicity model in rat model Mehrzadi et al. [162] analyzed the protective effects of berberine pre-treatment, using 100 mg/kg doses for 10 days. Berberine extract proved to have hepatoprotective effects by activating antioxidant defense enzymes such as GSH and GPx [162].
Resveratrol’s effectiveness as a protective agent in MTX hepatotoxicity was analyzed and confirmed in two different animal studies. Tunali-Akbay et al. [163] reported a neutrophil-dependent antioxidant mechanism through which resveratrol enacts its hepatoprotective effects, while Dalaklioglu et al. [152] reported the inhibition of lipid peroxidation through scavenging of superoxide and hydroxyl radicals by resveratrol.
Similar to resveratrol, gallic acid inhibits lipid peroxidation through the scavenging of ROS [164]. Jafaripour et al. [165] reported that inhibition of lipid peroxidation was behind the action of another nutraceutical, rosmarinic acid, which was able to mitigate the oxidative stress induced by MTX.
The effectiveness of two plant extracts widely used in traditional medicine, turmeric and Ginkgo biloba extract, was evaluated in two separate studies using rat model. Experimental data of Ginkgo biloba extract revealed a dose-dependent protective effect of the nutraceutical [166]. The beneficial effect might be explained through the modulation of the innate antioxidative mechanisms such as glutathione and glutathione S-transferase [166]. Moghadam et al. [167] reported similar findings while evaluating the effectives of turmeric extracts, with the added benefit of scavenging of ROS, and thus reducing lipid peroxidation.
Ahmad et al. [168] described the hepatoprotective effect manifested by sinapic acid through the regulation of the nrf2/heme oxygenase 1 (HO-1) and NF-κB signaling pathways. By modulating both oxidative enzymes, such as malondialdehyde, nitric oxide and catalase, and antioxidative systems, i.e., glutathione peroxidase, glutathione reductase activity, naringin was able to provide significant protection against MTX-induced oxidative stress and preserve the histological structure of rat hepatic tissue [169].
Rutin is a glycoside flavanol found in several plants, including citrus plants [170], which was previously demonstrated to exert a hepatoprotective effect against other chemotherapeutical agents [171,172] Erdogan et al. [173] reported that rutin, by elevating tissue superoxide dismutase and plasma glutathione peroxidase, showed similar hepatoprotective effects against MTX toxicity.
Propolis and propolis extracts have been examined as nutraceutics as well. Çetin et al. [174] reported the beneficial effects of propolis in preventing MTX hepatic injury in rats. Chrysin, a flavonoid extracted from honey and propolis, has been reported to alleviate oxidative stress and apoptosis induced by MTX in rats [175].
Two dietary supplements with promising effects have been tested for their supposed protective effects, indole-3-carbinol and alfa lipolic acid. Indole-3-carbinol, by upregulating antioxidant defense systems, alleviates MTX-induced hepatic injury [176]. On the other hand, alfa lipolic acid was able to prevent MTX-induced hepatotoxicity through the scavenging of ROS [157]
Vitamin supplements have been tested as well, with various results. Akbulut et al. [156] demonstrated the limited protective effect of ascorbic acid compared to other hepatoprotective agents in MTX-induced toxicity. In rat model of MTX-induced liver injury therapeutic delivery of β-carotene, an important source of vitamin A in the human diet, with antioxidant properties, led to decreased hepatic MDA activity and increased hepatic SOD, CAT and GSH peroxidase activities under conditions of [153]. Amirfakhrian et al. [177] showed the protective effect of vitamin E on liver architecture assessed by 99mTc-phytate functional imaging. Ismail et al. [178] report that while thiamine pyrophosphate exhibited protective effects against MTX hepatotoxicity, thiamine alone was ineffective. The beneficial effect was attributed to a reduction in MTX-induced NADP inhibition.
Dietary deficiencies were also reported to influence the severity of MTX hepatotoxicity [155]. A choline deficient diet resulted in an increase in the extent of fatty infiltration in rats treated with MTX as compared to normal rats treated with MTX, outlining the importance of choline in preventing MTX-induced fatty liver injury.

4.2. Nephrotoxicity

The most commonly described mechanism of MTX nephrotoxicity is the precipitation of MTX and its metabolites in the renal tubules [127]. Other mechanisms have also been proposed to play a concomitant role, such as constriction of the afferent capillary and direct effects on the mesangial or tubular epithelial cells [179,180].
Because the primary elimination route of MTX is renal excretion, with a high likelihood of nephrotoxicity, preventing those adverse events has been attempted through the use of various nutraceutical compounds.
Aladaileh et al. [181] have described the mechanism behind formononetin nephroprotective properties. Through the upregulation of nrf2/HO-1, formononetin successfully prevented MTX-induced renal injuries in a rat model. A similar molecular mechanism was found by Hassanein et al. [182] when studying the effectiveness of berberine as a nephroprotective agent. Rosmarinic acid, a polyphenolic nutraceutical compound, was reported to regulate the same pathway as berberine and formononetin, having a similar effect in rat models [165].
Paeonol and paeoniflorin are two aromatic compounds found in plants from the genus Paeonia spp. [183,184,185]. Morsy et al. [186] reported how paeonol administration increased the expression of the renal efflux transporter P-glycoprotein, which accelerates MTX elimination, limiting the nephrotoxic effect of the drug. The protective effect of paeoniflorin-6′-O-benzene sulfonate against MTX toxicity proceeds by targeting expression apoptotic proteins, such as Bax, cleaved-caspase-3, and cleaved-caspase-8 [187].
In an experimental model of MTX nephrotoxicity in rats, Elsawy et al. [188] observed the protective effects of naringin at doses of 20–40 and 80 mg/kg.
In another experimental model, Oguz et al. [189] evaluated the effectiveness of lycopene alone and in combination with melatonin. Both therapeutic regimes provided significant reduction in TNF-α, interleukin 1-beta (IL-1β) and ceruloplasmin levels. Further histopathological evaluation of renal tissue revealed a superior effect for the combined regime, with both schemes showing a significant protective effect against MTX-induced histological changes.
Inhibition of lipid peroxidation and increase in antioxidative status of cells occurred as a result of treatment with caffeic acid phenethyl ester at a dose of 10 mmol/kg in MTX-induced oxidative stress in rat kidney [190].
The effectiveness of gallic acid as a nephroprotector was assessed in two different animal studies. Asci et al. [191] and Olayinka et al. [164] have reported that gallic acid, through the reduction in oxidative stress, was effective at preventing renal injury induced by MTX.
Curcumin, primarily found in turmeric powder, is reported to have antioxidant properties, thus posing nephroprotective effects in rat models for MTX toxicity [192].
Quercetin, in a 15 mg/kg/day for 5 days, has been shown to lessen the degenerative changes and reduce apoptosis in rat kidney upon MTX treatment. Additionally, quercetin increased superoxide dismutase, glutathione peroxidase, and catalase levels, effectively reducing oxidative stress induced by MTX [193].
Several plant extracts have been tested for nephroprotective effects against MTX toxicity. Sherif et al. [194] have tested Ginkgo Biloba extracts, finding that it decreased renal TGF-β mRNA and MALAT1 expression, and regulated PI3K/Akt/mTOR signaling. Nigella sativa oil increased glutathione levels and prevented histological changes of renal tissue in a prolonged exposure of animal model [195]. Pre-treatment and co-treatment of MTX with garlic extracts in a rat model proved to be effective at preventing renal injury by increasing renal antioxidant enzyme activity [196]. Abouelela et al. [197] report the use of Ceiba pentandra extract to prevent MTX-induced renal toxicity in rats. The effects might be explained by improved renal antioxidant capacity and reduced MTX-induced oxidative stress. Hydro-Alcoholic extract of raspberry fruits has been shown to protect against MTX nephrotoxicity by upregulating the expression of aquaporin 1 in a dose-dependent matter [198]. Polyherbal combinations have been teste as well, with Sharma et al. [199] reporting on the use of Roots of B. diffusa and R. emodi, flowers of N. nucifera and stem bark of C. nurvala concomitantly. Histopathological analysis showed an alleviation of rat renal injury caused by MTX, when rats were treated with the polyherbal mixture, an effect explained by ROS scavenging and by improved renal antioxidant capacity.

4.3. Gastrointestinal Toxicity

Studies have shown that ROS production plays a key role in the mechanism behind gastrointestinal mucositis and enteritis caused by MTX treatment [200,201]. By having a low discrimination ability between tumor cells and fast-dividing cells and inhibiting DNA and RNA synthesis of fast-dividing cells, MTX counteracts epithelial intestine cells with a high turnover [120,202]. This results in the inhibition of division, a decrease in cell population, and ultimately leads to architectural and functional changes of intestinal epithelium.
Turmeric extracts, and mainly curcumin, have been reported to have various protective effects against MTX-induced toxicity. Song et al. [203] reported protective effects of curcumin against MTX aggression against intestinal mucosa. The beneficial effect might be explained by activation of mitogen-activated protein kinase phosphatase-1 and antioxidative mechanisms of superoxide dismutase and repression of NF-κB [203].
The effect of glutamine supplementation in MTX-induced enteritis has been studied on multiple animal models. in rat models, glutamine reduced intestinal injury, improved nutritional status, decreased bacterial translocation by preserving intestinal mucosa integrity, and improved survival rate [204]. However, in a cat model, glutamine supplementation was unable to preserve intestinal function [205].
Gastroprotective effects of several plant extracts have been tested against multiple animal models of gastrointestinal toxicity caused by MTX treatment. Shi et al. [206] explored gastroprotective effect of steamed root of Rehmannia glutinosa Libosch in intestinal mucositis model of MTX in a rat model. The extract mitigates MTX intestinal injury by alleviating oxidative stress and inflammatory responses [206]. Paullinia cupana, by increasing antioxidant systems and inhibiting IL-1β, has been shown to preserve intestinal integrity against MTX toxicity [207]. Wang et al. [208] have tested the effectiveness of glycyrrhizin acid, constituent of Glycyrrhiza glabra root, against MTX-induced enteritis. By suppressing the NF-κB and MAPK signaling pathways, glycyrrhizin acid showed significant protective effects against enteritis caused by MTX treatment in rats. Albiflorin is a glycoside isolated from the same plant as paeoniflorin, which has anti-inflammatory properties. Zhang et al. [209] reported immunomodulatory and anti-inflammatory features of albiflorin by inhibiting NF-κB/NLRP3 pathway, and also significantly reducing oxidative stress in MTX-induced enteritis.
Fatty acids and fatty acid derivates have been experimentally investigated as potential protective agents against oral mucositis, gastric mucositis and loss of intestinal integrity. Alfa lipolic acid, a naturally occurring caprylic acid derivative, has been tested as a preventive agent against oral mucositis and oxidative stress induced by MTX in rats [210]. Through the increase in glutathione and superoxide dismutase, and inhibition of apoptosis, alfa lipolic acid attenuated MTX-evoked alterations of the intestinal wall [210]. da Silva Ferreira et al. [211] reinforced the potential prophylactic benefits of butyrate, through the use of a novel model using 3D intestinal organoids derived from mouse ileum. The anti-inflammatory and anti-apoptotic properties of omega 3 fatty acids was reported to limit the intestinal damage in rats treated with MTX [212]. The molecular mechanism relies on downregulation of NF-κB, ciclooxigenase-2 and TNF-α.
Yilmaz et al. [213] reported the beneficial effect of daily use of vitamin supplement injections to ameliorate intestinal inflammation and protect rats against MTX-induced mucositis. Vitamin C and B2 efficacy has been tested by da Silva Ferreira et al. [214] using an in vitro bacterial growth model, and an in vivo rat model. Both vitamins were able to enhance the growth of gut bacteria, leading to enrichment of Blautia coccoides and Roseburia intestinalis. However, despite the fact that vitamin C ameliorated clinical symptoms of mucositis, neither of the vitamins was able to modulate the course of MTX-induced mucositis, as assessed by citrulline plasma levels [214].
Salecan is a non-toxic water-soluble β-glucan exhibited dose-dependent effects against intestinal mucositis in rats treated with MTX [215]. Salecan treatment inhibited oxidative stress through the effective scavenging of ROS, therefore maintaining mucosal architecture and integrity. Similarly, sodium alginate, a salt of alginic acid found in algae, was reported to protect intestinal architecture, and even mitigate hematologic side effects of MTX treatment in rats [216].
While probiotics have not been extensively explored as preventive tools for MTX-induced multi-organ toxicity, they might have useful properties in preventing intestinal damage. In an MTX toxicity model in rats, cow’s milk yogurt fermented with Lactobacillus johnsonii and sheep’s milk yogurt fermented with a combination of Lactobacillus bulgaricus and Streptococcus thermophilus were able to improve small intestinal barrier function and prevent MTX damage to the small intestine [217].

4.4. Pulmonary Toxicity

While the mechanism behind MTX pulmonary toxicity is not well understood, it is likely to be an idiosyncratic reaction and not linked to folate antagonism, as it appears in both high-dose and low-dose treatment schemes [25,218,219]. The presence of mononuclear cell infiltration and inflammatory granulomas in lung injury of animal models upon MTX treatment suggests that MTX pneumonitis represents a hypersensitivity reaction [213,218,219,220]. Pulmonary protective nutraceuticals have not been studied extensively, with few compounds being tested in experimental setting.
Polydatin, also known as picedin, is a major resveratrol derivative found in grape juice. Polydatin’s antioxidant and anti-inflammatory properties increased cellular antioxidant status and reduced inflammation and fibrosis in rats [31]. In rat models of pulmonary oxidative damage, the effects of lutein and alfa lipoic acid elicits antioxidative and anti-inflammatory features in lung tissue, by increasing glutathione levels, and decreasing proinflammatory cytokines [221,222].
A synopsis of the main natural components used to counteract multi-organ injury induced by MTX toxicity is provided in Table 3.
While cost-wise and therapeutically effective, nutraceutical therapy for MTX-induced toxicity in humans has not been fully explored in terms of high-dose-induced adverse effects and dose accumulation of multiple nutraceuticals simultaneously administered.
Most studies have tested the effect of the nutraceutical alone on the health of animal models, with no ill effect detected. However, there are not enough studies that explored the effect of nutraceuticals on the pharmacological profile of MTX or other common medications [10]. While most nutraceuticals come from sources that humans have been regularly consuming with no adverse effects, it is unclear if those same compound administered in higher doses are as safe [131]. Thus, before any clinical trials can begin, further testing, with longer periods of nutraceutical administration needs to be performed, in order to assess the effects of long-term administration of nutraceuticals.

5. Conclusions and Future Perspectives

MTX is, and will continue to be clinically relevant for years to come due to a wide sera of therapeutic applications, from chemotherapeutical agents for malignant hemopathies, osteosarcoma, breast cancer, etc. [47,57,58], to anti-inflammatory medication for vasculitis, systemic lupus erythematosus, multiple sclerosis, transplantation surgeries [59,60], and further as a disease-modifying agent for RA, JIA, and psoriasis [57,58,61]. However, MTX therapy comes with pitfalls, making it less than perfect. The severe side effects associated with MTX use can occur in both low- and high-dose regiments, often leading to temporary or permanent interruption of MTX medication [47,57,61,128], which limits its clinical potential in patients with urgent need of it [47,61,127,128].
Clinicians could benefit from adjuvant compounds which are designed to reduce the severity and frequency of adverse effects and can be used as a MTX co-treatments. Emerging nutraceutical compounds demonstrated translational relevance as adjuvant therapy into experimental models of MTX toxicity posing several advantages that make them good candidates for such a task [133,134,135,136,137,138,139,140]. With a wide variety of natural compounds that do not pose a health risk for patients, are easy to administer, and relatively inexpensive, clinicians could effectively prevent therapeutic limitations and debilitating side effects of MTX treatment, by using nutraceuticals. Therefore, to take these experimental data from bench to bedside, further clinical trials to explore the clinical potential of such compounds in patients treated with MTX are needed.

Author Contributions

Conceptualization, G.-E.M., M.-A.N., C.-C.B. and A.-D.B.; writing—original draft preparation, G.-E.M., M.-A.N., C.-C.B. and A.-D.B.; writing—review and editing, G.-E.M., M.-A.N. and C.-C.B., supervision M.-A.N. and A.-D.B.; project administration, G.-E.M. funding acquisition, G.-E.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by “Iuliu Haţieganu” university of medicine and pharmacy, grant number (35.177/17.12.2021).

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Fleisher, M. Antifolate Analogs: Mechanism of Action, Analytical Methodology, and Clinical Efficacy. Ther. Drug Monit. 1993, 15, 521–526. [Google Scholar] [CrossRef]
  2. Costi, M.P.; Ferrari, S. Update on Antifolate Drugs Targets. Curr. Drug Targets 2001, 2, 135–166. [Google Scholar] [CrossRef]
  3. Gonen, N.; Assaraf, Y.G. Antifolates in Cancer Therapy: Structure, Activity and Mechanisms of Drug Resistance. Drug Resist. Updat. 2012, 15, 183–210. [Google Scholar] [CrossRef]
  4. Bannwarth, B.; Labat, L.; Moride, Y.; Schaeverbeke, T. Methotrexate in Rheumatoid Arthritis. An Update. Drugs 1994, 47, 25–50. [Google Scholar] [CrossRef]
  5. Bedoui, Y.; Guillot, X.; Sélambarom, J.; Guiraud, P.; Giry, C.; Jaffar-Bandjee, M.C.; Ralandison, S.; Gasque, P. Methotrexate an Old Drug with New Tricks. Int. J. Mol. Sci. 2019, 20, 5023. [Google Scholar] [CrossRef] [Green Version]
  6. Prey, S.; Paul, C. Effect of Folic or Folinic Acid Supplementation on Methotrexate-Associated Safety and Efficacy in Inflammatory Disease: A Systematic Review. Br. J. Dermatol. 2009, 160, 622–628. [Google Scholar] [CrossRef]
  7. Schnabel, A.; Gross, W.L. Low-Dose Methotrexate in Rheumatic Diseases—Efficacy, Side Effects, and Risk Factors for Side Effects. Semin. Arthritis Rheum. 1994, 23, 310–327. [Google Scholar] [CrossRef]
  8. Kalra, E.K. Nutraceutical--Definition and Introduction. AAPS PharmSci. 2003, 5, E25. [Google Scholar] [CrossRef] [Green Version]
  9. Brower, V. Nutraceuticals: Poised for a Healthy Slice of the Healthcare Market? Nat. Biotechnol. 1998, 16, 728–731. [Google Scholar] [CrossRef]
  10. Larussa, T.; Imeneo, M.; Luzza, F. Potential Role of Nutraceutical Compounds in Inflammatory Bowel Disease. World J. Gastroenterol. 2017, 23, 2483–2492. [Google Scholar] [CrossRef]
  11. Kumar, A.; Sharma, S.S. NF-KappaB Inhibitory Action of Resveratrol: A Probable Mechanism of Neuroprotection in Experimental Diabetic Neuropathy. Biochem. Biophys. Res. Commun. 2010, 394, 360–365. [Google Scholar] [CrossRef] [PubMed]
  12. Soto-Urquieta, M.G.; López-Briones, S.; Pérez-Vázquez, V.; Saavedra-Molina, A.; González-Hernández, G.A.; Ramírez-Emiliano, J. Curcumin Restores Mitochondrial Functions and Decreases Lipid Peroxidation in Liver and Kidneys of Diabetic Db/Db Mice. Biol. Res. 2014, 47, 74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Dadhania, V.P.; Tripathi, D.N.; Vikram, A.; Ramarao, P.; Jena, G.B. Intervention of Alpha-Lipoic Acid Ameliorates Methotrexate-Induced Oxidative Stress and Genotoxicity: A Study in Rat Intestine. Chem. Biol. Interact. 2010, 183, 85–97. [Google Scholar] [CrossRef]
  14. Lesmana, R.; Yusuf, I.F.; Goenawan, H.; Achadiyani, A.; Khairani, A.F.; Fatimah, S.N.; Supratman, U. Low Dose of β-Carotene Regulates Inflammation, Reduces Caspase Signaling, and Correlates with Autophagy Activation in Cardiomyoblast Cell Lines. Med. Sci. Monit. Basic Res. 2020, 26, e928648-1–e928648-11. [Google Scholar] [CrossRef]
  15. Ohnishi, E.; Bannai, H. Quercetin Potentiates TNF-Induced Antiviral Activity. Antivir. Res. 1993, 22, 327–331. [Google Scholar] [CrossRef]
  16. Singh, S.; Kola, P.; Kaur, D.; Singla, G.; Mishra, V.; Panesar, P.S.; Mallikarjunan, K.; Krishania, M. Therapeutic Potential of Nutraceuticals and Dietary Supplements in the Prevention of Viral Diseases: A Review. Front. Nutr. 2021, 8, 640. [Google Scholar] [CrossRef]
  17. Naoi, M.; Wu, Y.; Shamoto-Nagai, M.; Maruyama, W. Mitochondria in Neuroprotection by Phytochemicals: Bioactive Polyphenols Modulate Mitochondrial Apoptosis System, Function and Structure. Int. J. Mol. Sci. 2019, 20, 2451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Lapi, D.; Stornaiuolo, M.; Sabatino, L.; Sommella, E.; Tenore, G.; Daglia, M.; Scuri, R.; Di Maro, M.; Colantuoni, A.; Novellino, E. The Pomace Extract Taurisolo Protects Rat Brain From Ischemia-Reperfusion Injury. Front. Cell. Neurosci. 2020, 14, 3. [Google Scholar] [CrossRef]
  19. Tognola, C.; Alessandro, M.; Milani, M.; Cartella, I.; Tavecchia, G.; Grasso, E.; Sun, J.; Giannattasio, C. Nutraceuticals in Chronic Coronary Syndromes: Preclinical Data and Translational Experiences. High Blood Press. Cardiovasc. Prev. 2021, 28, 13–25. [Google Scholar] [CrossRef]
  20. Chelluboina, B.; Vemuganti, R. Therapeutic Potential of Nutraceuticals to Protect Brain after Stroke. Neurochem. Int. 2021, 142, 104908. [Google Scholar] [CrossRef]
  21. Maiuolo, J.; Gliozzi, M.; Carresi, C.; Musolino, V.; Oppedisano, F.; Scarano, F.; Nucera, S.; Scicchitano, M.; Bosco, F.; Macri, R.; et al. Nutraceuticals and Cancer: Potential for Natural Polyphenols. Nutrients 2021, 13, 3834. [Google Scholar] [CrossRef] [PubMed]
  22. Dadhania, V.P.; Trivedi, P.P.; Vikram, A.; Tripathi, D.N. Nutraceuticals against Neurodegeneration: A Mechanistic Insight. Curr. Neuropharmacol. 2016, 14, 627–640. [Google Scholar] [CrossRef] [PubMed]
  23. Uranga, J.A.; López-Miranda, V.; Lombó, F.; Abalo, R. Food, Nutrients and Nutraceuticals Affecting the Course of Inflammatory Bowel Disease. Pharmacol. Rep. 2016, 68, 816–826. [Google Scholar] [CrossRef] [PubMed]
  24. Cheng, H.S.; Rademaker, M. Monitoring Methotrexate-Induced Liver Fibrosis in Patients with Psoriasis: Utility of Transient Elastography. Psoriasis 2018, 8, 21–29. [Google Scholar] [CrossRef] [Green Version]
  25. Kim, Y.-J.; Song, M.; Ryu, J.-C. Mechanisms Underlying Methotrexate-Induced Pulmonary Toxicity. Expert Opin. Drug Saf. 2009, 8, 451–458. [Google Scholar] [CrossRef]
  26. Letertre, M.P.M.; Munjoma, N.; Wolfer, K.; Pechlivanis, A.; McDonald, J.A.K.; Hardwick, R.N.; Cherrington, N.J.; Coen, M.; Nicholson, J.K.; Hoyles, L.; et al. A Two-Way Interaction between Methotrexate and the Gut Microbiota of Male Sprague–Dawley Rats. J. Proteome Res. 2020, 19, 3326–3339. [Google Scholar] [CrossRef]
  27. Ramalanjaona, B.; Hevroni, G.; Cham, S.; Page, C.; Salifu, M.O.; McFarlane, S.I. Nephrotoxicity Associated with Low-Dose Methotrexate and Outpatient Parenteral Microbial Therapy: A Case Report, Review of the Literature and Pathophysiologic Insights. Am. J. Med. Case Rep. 2020, 8, 400–404. [Google Scholar] [CrossRef]
  28. Psoriasis—ClinicalKey. Available online: https://www.clinicalkey.com/#!/content/book/3-s2.0-B9780702062759000088 (accessed on 20 September 2022).
  29. Feinsilber, D.; Leoni, R.J.; Siripala, D.; Leuck, J.; Mears, K.A. Evaluation, Identification, and Management of Acute Methotrexate Toxicity in High-Dose Methotrexate Administration in Hematologic Malignancies. Cureus 2018, 10, e2040. [Google Scholar] [CrossRef] [Green Version]
  30. Azadnasab, R.; Kalantar, H.; Khorsandi, L.; Kalantari, H.; Khodayar, M.J. Epicatechin Ameliorative Effects on Methotrexate-Induced Hepatotoxicity in Mice. Hum. Exp. Toxicol. 2021, 40, S603–S610. [Google Scholar] [CrossRef]
  31. Polydatin Combats Methotrexate-Induced Pulmonary Fibrosis in Rats: Involvement of Biochemical and Histopathological Assessment—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/35174937/ (accessed on 19 September 2022).
  32. Xia, Y.; Shi, H.; Qian, C.; Han, H.; Lu, K.; Tao, R.; Gu, R.; Zhao, Y.; Wei, Z.; Lu, Y. Modulation of Gut Microbiota by Magnesium Isoglycyrrhizinate Mediates Enhancement of Intestinal Barrier Function and Amelioration of Methotrexate-Induced Liver Injury. Front. Immunol. 2022, 13, 874878. [Google Scholar] [CrossRef]
  33. Schnell, J.R.; Dyson, H.J.; Wright, P.E. Structure, Dynamics, and Catalytic Function of Dihydrofolate Reductase. Annu. Rev. Biophys. Biomol. Struct. 2004, 33, 119–140. [Google Scholar] [CrossRef] [Green Version]
  34. Farber, S.; Diamond, L.K.; Mercer, R.D.; Sylvester, R.F.; Wolff, J.A. Temporary Remissions in Acute Leukemia in Children Produced by Folic Acid Antagonist, 4-Aminopteroyl-Glutamic Acid (Aminopterin). N. Engl. J. Med. 1948, 238, 787–793. [Google Scholar] [CrossRef]
  35. Seeger, D.R.; Cosulich, D.B.; Smith, J.M.; Hultquist, M.E. Analogs of Pteroylglutamic Acid. III. 4-Amino Derivatives. Available online: https://pubs.acs.org/doi/pdf/10.1021/ja01173a061 (accessed on 13 September 2022).
  36. Avendaño, C.; Menéndez, J.C. Antimetabolites. In Medicinal Chemistry of Anticancer Drugs; Elsevier: Amsterdam, The Netherlands, 2008; pp. 9–52. ISBN 978-0-444-52824-7. [Google Scholar]
  37. Blakley, R.L.; Cocco, L. Role of Isomerization of Initial Complexes in the Binding of Inhibitors to Dihydrofolate Reductase. Biochemistry 1985, 24, 4772–4777. [Google Scholar] [CrossRef]
  38. Pereira da Silva, C.M.; Pedroso de Lima, M.C. Application of Flow Microcalorimetry to the Study of Dehydrofolate Reductase Activities in Crude Tissue Homogenates. J. Therm. Anal. 1992, 38, 821–833. [Google Scholar] [CrossRef]
  39. Phillips, D.C.; Woollard, K.J.; Griffiths, H.R. The Anti-Inflammatory Actions of Methotrexate Are Critically Dependent upon the Production of Reactive Oxygen Species. Br. J. Pharmacol. 2003, 138, 501–511. [Google Scholar] [CrossRef] [Green Version]
  40. Herman, S.; Zurgil, N.; Deutsch, M. Low Dose Methotrexate Induces Apoptosis with Reactive Oxygen Species Involvement in T Lymphocytic Cell Lines to a Greater Extent than in Monocytic Lines. Inflamm. Res. 2005, 54, 273–280. [Google Scholar] [CrossRef]
  41. Huang, C.-C.; Hsu, P.-C.; Hung, Y.-C.; Liao, Y.-F.; Liu, C.-C.; Hour, C.-T.; Kao, M.-C.; Tsay, G.J.; Hung, H.-C.; Liu, G.Y. Ornithine Decarboxylase Prevents Methotrexate-Induced Apoptosis by Reducing Intracellular Reactive Oxygen Species Production. Apoptosis 2005, 10, 895–907. [Google Scholar] [CrossRef]
  42. Quéméneur, L.; Gerland, L.-M.; Flacher, M.; Ffrench, M.; Revillard, J.-P.; Genestier, L. Differential Control of Cell Cycle, Proliferation, and Survival of Primary T Lymphocytes by Purine and Pyrimidine Nucleotides. J. Immunol. 2003, 170, 4986–4995. [Google Scholar] [CrossRef] [Green Version]
  43. Cronstein, B.N. Low-Dose Methotrexate: A Mainstay in the Treatment of Rheumatoid Arthritis. Pharmacol. Rev. 2005, 57, 163–172. [Google Scholar] [CrossRef] [Green Version]
  44. Montesinos, M.C.; Desai, A.; Delano, D.; Chen, J.-F.; Fink, J.S.; Jacobson, M.A.; Cronstein, B.N. Adenosine A2A or A3 Receptors Are Required for Inhibition of Inflammation by Methotrexate and Its Analog MX-68. Arthritis Rheum 2003, 48, 240–247. [Google Scholar] [CrossRef]
  45. Gerards, A.H.; de Lathouder, S.; de Groot, E.R.; Dijkmans, B.A.C.; Aarden, L.A. Inhibition of Cytokine Production by Methotrexate. Studies in Healthy Volunteers and Patients with Rheumatoid Arthritis. Rheumatology 2003, 42, 1189–1196. [Google Scholar] [CrossRef] [Green Version]
  46. de Lathouder, S.; Gerards, A.H.; Dijkmans, B.A.C.; Aarden, L.A. Two Inhibitors of DNA-Synthesis Lead to Inhibition of Cytokine Production via a Different Mechanism. Nucleosides Nucleotides Nucleic Acids 2004, 23, 1089–1100. [Google Scholar] [CrossRef] [PubMed]
  47. Routledge, P.A.; Hutchings, A.D. Therapeutic Drug Monitoring (TDM). In The Immunoassay Handbook; Elsevier: Amsterdam, The Netherlands, 2013; pp. 945–962. ISBN 978-0-08-097037-0. [Google Scholar]
  48. van Roon, E.N.; van de Laar, M.A.F.J. Methotrexate Bioavailability. Clin. Exp. Rheumatol. 2010, 28, S27–S32. [Google Scholar]
  49. Hoekstra, M.; Haagsma, C.; Neef, C.; Proost, J.; Knuif, A.; van de Laar, M. Bioavailability of Higher Dose Methotrexate Comparing Oral and Subcutaneous Administration in Patients with Rheumatoid Arthritis. J. Rheumatol. 2004, 31, 645–648. [Google Scholar]
  50. Schiff, M.H.; Jaffe, J.S.; Freundlich, B. Head-to-Head, Randomised, Crossover Study of Oral versus Subcutaneous Methotrexate in Patients with Rheumatoid Arthritis: Drug-Exposure Limitations of Oral Methotrexate at Doses ≥15 Mg May Be Overcome with Subcutaneous Administration. Ann. Rheum. Dis. 2014, 73, 1549–1551. [Google Scholar] [CrossRef] [Green Version]
  51. Dixon, K.H.; Lanpher, B.C.; Chiu, J.; Kelley, K.; Cowan, K.H. A Novel CDNA Restores Reduced Folate Carrier Activity and Methotrexate Sensitivity to Transport Deficient Cells. J. Biol. Chem. 1994, 269, 17–20. [Google Scholar] [CrossRef]
  52. Inoue, K.; Yuasa, H. Molecular Basis for Pharmacokinetics and Pharmacodynamics of Methotrexate in Rheumatoid Arthritis Therapy. Drug Metab. Pharmacokinet. 2014, 29, 12–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Cannella, A.C.; O’Dell, J.R. Chapter 61—Traditional DMARDs: Methotrexate, Leflunomide, Sulfasalazine, Hydroxychloroquine, and Combination Therapies. In Kelley and Firestein’s Textbook of Rheumatology, 10th ed.; Firestein, G.S., Budd, R.C., Gabriel, S.E., McInnes, I.B., O’Dell, J.R., Eds.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 958–982.e7. ISBN 978-0-323-31696-5. [Google Scholar]
  54. Seideman, P.; Beck, O.; Eksborg, S.; Wennberg, M. The Pharmacokinetics of Methotrexate and Its 7-Hydroxy Metabolite in Patients with Rheumatoid Arthritis. Br. J. Clin. Pharmacol. 1993, 35, 409–412. [Google Scholar] [CrossRef] [Green Version]
  55. Nuernberg, B.; Koehnke, R.; Solsky, M.; Hoffman, J.; Furst, D.E. Biliary Elimination of Low-Dose Methotrexate in Humans. Arthritis Rheum. 1990, 33, 898–902. [Google Scholar] [CrossRef]
  56. Grim, J.; Chládek, J.; Martínková, J. Pharmacokinetics and Pharmacodynamics of Methotrexate in Non-Neoplastic Diseases. Clin. Pharm. 2003, 42, 139–151. [Google Scholar] [CrossRef] [PubMed]
  57. METHOTREXATE TABLETS, USP. 27.
  58. World Health Organization. World Health Organization Model List of Essential Medicines: 22nd List (2021); World Health Organization: Geneva, Switzerland, 2021. [Google Scholar]
  59. Chan, E.S.L.; Cronstein, B.N. Mechanisms of Action of Methotrexate. Bull. NYU Hosp. Jt. Dis. 2013, 71, S5–S8. [Google Scholar]
  60. Chan, E.S.L.; Cronstein, B.N. Methotrexate—How Does It Really Work? Nat. Rev. Rheumatol. 2010, 6, 175–178. [Google Scholar] [CrossRef] [PubMed]
  61. Methotrexate Monograph for Professionals—Drugs.com. Available online: https://web.archive.org/web/20161008130258/https://www.drugs.com/monograph/methotrexate.html (accessed on 10 September 2022).
  62. Methotrexate Side Effects in Detail—Drugs.com. Available online: https://web.archive.org/web/20160614022351/http://www.drugs.com/sfx/methotrexate-side-effects.html (accessed on 10 September 2022).
  63. Shreiner, A.B.; Kao, J.Y.; Young, V.B. The Gut Microbiome in Health and in Disease. Curr. Opin. Gastroenterol. 2015, 31, 69–75. [Google Scholar] [CrossRef] [PubMed]
  64. Hou, K.; Wu, Z.-X.; Chen, X.-Y.; Wang, J.-Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in Health and Diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar] [CrossRef]
  65. Gill, S.R.; Pop, M.; DeBoy, R.T.; Eckburg, P.B.; Turnbaugh, P.J.; Samuel, B.S.; Gordon, J.I.; Relman, D.A.; Fraser-Liggett, C.M.; Nelson, K.E. Metagenomic Analysis of the Human Distal Gut Microbiome. Science 2006, 312, 1355–1359. [Google Scholar] [CrossRef] [Green Version]
  66. Guinane, C.M.; Cotter, P.D. Role of the Gut Microbiota in Health and Chronic Gastrointestinal Disease: Understanding a Hidden Metabolic Organ. Therap. Adv. Gastroenterol. 2013, 6, 295–308. [Google Scholar] [CrossRef] [Green Version]
  67. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A Human Gut Microbial Gene Catalogue Established by Metagenomic Sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef] [Green Version]
  68. Senghor, B.; Sokhna, C.; Ruimy, R.; Lagier, J.-C. Gut Microbiota Diversity According to Dietary Habits and Geographical Provenance. Hum. Microbiome J. 2018, 7–8, 1–9. [Google Scholar] [CrossRef]
  69. Gianchecchi, E.; Fierabracci, A. Recent Advances on Microbiota Involvement in the Pathogenesis of Autoimmunity. Int. J. Mol. Sci. 2019, 20, 283. [Google Scholar] [CrossRef] [Green Version]
  70. Zhou, W.; Cheng, Y.; Zhu, P.; Nasser, M.I.; Zhang, X.; Zhao, M. Implication of Gut Microbiota in Cardiovascular Diseases. Oxidative Med. Cell. Longev. 2020, 2020, 5394096. [Google Scholar] [CrossRef]
  71. Battaglini, D.; Pimentel-Coelho, P.M.; Robba, C.; dos Santos, C.C.; Cruz, F.F.; Pelosi, P.; Rocco, P.R.M. Gut Microbiota in Acute Ischemic Stroke: From Pathophysiology to Therapeutic Implications. Front. Neurol. 2020, 11, 598. [Google Scholar] [CrossRef]
  72. Zhang, H.; Chen, Y.; Wang, Z.; Xie, G.; Liu, M.; Yuan, B.; Chai, H.; Wang, W.; Cheng, P. Implications of Gut Microbiota in Neurodegenerative Diseases. Front. Neurol. 2022, 13, 785644. [Google Scholar] [CrossRef]
  73. Meyer, K.; Lulla, A.; Debroy, K.; Shikany, J.M.; Yaffe, K.; Meirelles, O.; Launer, L.J. Association of the Gut Microbiota With Cognitive Function in Midlife. JAMA Netw. Open 2022, 5, e2143941. [Google Scholar] [CrossRef] [PubMed]
  74. Akbar, N.; Khan, N.A.; Muhammad, J.S.; Siddiqui, R. The Role of Gut Microbiome in Cancer Genesis and Cancer Prevention. Health Sci. Rev. 2022, 2, 100010. [Google Scholar] [CrossRef]
  75. Xu, H.; Liu, M.; Cao, J.; Li, X.; Fan, D.; Xia, Y.; Lu, X.; Li, J.; Ju, D.; Zhao, H. The Dynamic Interplay between the Gut Microbiota and Autoimmune Diseases. J. Immunol. Res. 2019, 2019, 7546047. [Google Scholar] [CrossRef] [Green Version]
  76. Salazar, N.; Arboleya, S.; Fernández-Navarro, T.; de los Reyes-Gavilán, C.G.; Gonzalez, S.; Gueimonde, M. Age-Associated Changes in Gut Microbiota and Dietary Components Related with the Immune System in Adulthood and Old Age: A Cross-Sectional Study. Nutrients 2019, 11, 1765. [Google Scholar] [CrossRef] [Green Version]
  77. Sanchez-Morate, E.; Gimeno-Mallench, L.; Stromsnes, K.; Sanz-Ros, J.; Román-Domínguez, A.; Parejo-Pedrajas, S.; Inglés, M.; Olaso, G.; Gambini, J.; Mas-Bargues, C. Relationship between Diet, Microbiota, and Healthy Aging. Biomedicines 2020, 8, 287. [Google Scholar] [CrossRef]
  78. Musso, G.; Gambino, R.; Cassader, M. Obesity, Diabetes, and Gut Microbiota. Diabetes Care 2010, 33, 2277–2284. [Google Scholar] [CrossRef] [Green Version]
  79. Teuwen, L.-A.; Geldhof, V.; Pasut, A.; Carmeliet, P. COVID-19: The Vasculature Unleashed. Nat. Rev. Immunol. 2020, 20, 389–391. [Google Scholar] [CrossRef] [PubMed]
  80. Neag, M.A.; Mitre, A.O.; Catinean, A.; Buzoianu, A.D. Overview of the Microbiota in the Gut-Liver Axis in Viral B and C Hepatitis. World J. Gastroenterol. 2021, 27, 7446–7461. [Google Scholar] [CrossRef] [PubMed]
  81. Albillos, A.; de Gottardi, A.; Rescigno, M. The Gut-Liver Axis in Liver Disease: Pathophysiological Basis for Therapy. J. Hepatol. 2020, 72, 558–577. [Google Scholar] [CrossRef] [Green Version]
  82. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The Gut-Brain Axis: Interactions between Enteric Microbiota, Central and Enteric Nervous Systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar] [PubMed]
  83. Ding, J.-H.; Jin, Z.; Yang, X.-X.; Lou, J.; Shan, W.-X.; Hu, Y.-X.; Du, Q.; Liao, Q.-S.; Xie, R.; Xu, J.-Y. Role of Gut Microbiota via the Gut-Liver-Brain Axis in Digestive Diseases. World J. Gastroenterol. 2020, 26, 6141–6162. [Google Scholar] [CrossRef] [PubMed]
  84. Honda, K.; Littman, D.R. The Microbiota in Adaptive Immune Homeostasis and Disease. Nature 2016, 535, 75–84. [Google Scholar] [CrossRef] [PubMed]
  85. Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The Role of Short-Chain Fatty Acids in Microbiota-Gut-Brain Communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef]
  86. Martin-Gallausiaux, C.; Marinelli, L.; Blottière, H.M.; Larraufie, P.; Lapaque, N. SCFA: Mechanisms and Functional Importance in the Gut. Proc. Nutr. Soc. 2021, 80, 37–49. [Google Scholar] [CrossRef]
  87. Rey, F.E.; Faith, J.J.; Bain, J.; Muehlbauer, M.J.; Stevens, R.D.; Newgard, C.B.; Gordon, J.I. Dissecting the in Vivo Metabolic Potential of Two Human Gut Acetogens. J. Biol. Chem. 2010, 285, 22082–22090. [Google Scholar] [CrossRef] [Green Version]
  88. Bobik, T.A.; Havemann, G.D.; Busch, R.J.; Williams, D.S.; Aldrich, H.C. The Propanediol Utilization (Pdu) Operon of Salmonella Enterica Serovar Typhimurium LT2 Includes Genes Necessary for Formation of Polyhedral Organelles Involved in Coenzyme B(12)-Dependent 1, 2-Propanediol Degradation. J. Bacteriol. 1999, 181, 5967–5975. [Google Scholar] [CrossRef] [Green Version]
  89. Scott, K.P.; Martin, J.C.; Campbell, G.; Mayer, C.-D.; Flint, H.J. Whole-Genome Transcription Profiling Reveals Genes up-Regulated by Growth on Fucose in the Human Gut Bacterium “Roseburia inulinivorans”. J. Bacteriol. 2006, 188, 4340–4349. [Google Scholar] [CrossRef] [Green Version]
  90. Flint, H.J.; Duncan, S.H.; Scott, K.P.; Louis, P. Links between Diet, Gut Microbiota Composition and Gut Metabolism. Proc. Nutr. Soc. 2015, 74, 13–22. [Google Scholar] [CrossRef] [Green Version]
  91. Louis, P.; Young, P.; Holtrop, G.; Flint, H.J. Diversity of Human Colonic Butyrate-Producing Bacteria Revealed by Analysis of the Butyryl-CoA:Acetate CoA-Transferase Gene. Environ. Microbiol. 2010, 12, 304–314. [Google Scholar] [CrossRef] [PubMed]
  92. Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef] [Green Version]
  93. Kau, A.L.; Ahern, P.P.; Griffin, N.W.; Goodman, A.L.; Gordon, J.I. Human Nutrition, the Gut Microbiome and the Immune System. Nature 2011, 474, 327–336. [Google Scholar] [CrossRef] [PubMed]
  94. Zhao, L.; Zhang, F.; Ding, X.; Wu, G.; Lam, Y.Y.; Wang, X.; Fu, H.; Xue, X.; Lu, C.; Ma, J.; et al. Gut Bacteria Selectively Promoted by Dietary Fibers Alleviate Type 2 Diabetes. Science 2018, 359, 1151–1156. [Google Scholar] [CrossRef] [Green Version]
  95. Husted, A.S.; Trauelsen, M.; Rudenko, O.; Hjorth, S.A.; Schwartz, T.W. GPCR-Mediated Signaling of Metabolites. Cell Metab. 2017, 25, 777–796. [Google Scholar] [CrossRef] [Green Version]
  96. Landsman, L.; Jung, S. Lung Macrophages Serve as Obligatory Intermediate between Blood Monocytes and Alveolar Macrophages. J. Immunol. 2007, 179, 3488–3494. [Google Scholar] [CrossRef] [Green Version]
  97. Landsman, L.; Varol, C.; Jung, S. Distinct Differentiation Potential of Blood Monocyte Subsets in the Lung. J. Immunol. 2007, 178, 2000–2007. [Google Scholar] [CrossRef] [Green Version]
  98. Antonio, N.; Andrea, T.; Claudio, T.; Beatrice, P.; Pamela, C.; Chiara, M.; de’ Gian, L.A.; Francesco, D.M.; Tiziana, M. Digestive Disorders and Intestinal Microbiota. Acta Biomed. 2018, 89, 47–51. [Google Scholar] [CrossRef]
  99. Rueca, M.; Fontana, A.; Bartolini, B.; Piselli, P.; Mazzarelli, A.; Copetti, M.; Binda, E.; Perri, F.; Gruber, C.E.M.; Nicastri, E.; et al. Investigation of Nasal/Oropharyngeal Microbial Community of COVID-19 Patients by 16S RDNA Sequencing. Int. J. Environ. Res. Public Health 2021, 18, 2174. [Google Scholar] [CrossRef]
  100. Hajjo, R.; Sabbah, D.A.; Al Bawab, A.Q. Unlocking the Potential of the Human Microbiome for Identifying Disease Diagnostic Biomarkers. Diagnostics 2022, 12, 1742. [Google Scholar] [CrossRef]
  101. Peck, B.C.E.; Shanahan, M.T.; Singh, A.P.; Sethupathy, P. Gut Microbial Influences on the Mammalian Intestinal Stem Cell Niche. Stem Cells Int. 2017, 2017, 5604727. [Google Scholar] [CrossRef] [Green Version]
  102. Umar, S. Intestinal Stem Cells. Curr. Gastroenterol. Rep. 2010, 12, 340–348. [Google Scholar] [CrossRef]
  103. Rakoff-Nahoum, S.; Paglino, J.; Eslami-Varzaneh, F.; Edberg, S.; Medzhitov, R. Recognition of Commensal Microflora by Toll-like Receptors Is Required for Intestinal Homeostasis. Cell 2004, 118, 229–241. [Google Scholar] [CrossRef]
  104. Vaishnava, S.; Yamamoto, M.; Severson, K.M.; Ruhn, K.A.; Yu, X.; Koren, O.; Ley, R.; Wakeland, E.K.; Hooper, L.V. The Antibacterial Lectin RegIIIgamma Promotes the Spatial Segregation of Microbiota and Host in the Intestine. Science 2011, 334, 255–258. [Google Scholar] [CrossRef] [Green Version]
  105. Alam, A.; Neish, A. Role of Gut Microbiota in Intestinal Wound Healing and Barrier Function. Tissue Barriers 2018, 6, 1539595. [Google Scholar] [CrossRef]
  106. Gewirtz, A.T.; Navas, T.A.; Lyons, S.; Godowski, P.J.; Madara, J.L. Cutting Edge: Bacterial Flagellin Activates Basolaterally Expressed TLR5 to Induce Epithelial Proinflammatory Gene Expression. J. Immunol. 2001, 167, 1882–1885. [Google Scholar] [CrossRef] [Green Version]
  107. Sansonetti, P. Host-Pathogen Interactions: The Seduction of Molecular Cross Talk. Gut 2002, 50 (Suppl. 3), III2–III8. [Google Scholar] [CrossRef]
  108. Morgan, X.C.; Tickle, T.L.; Sokol, H.; Gevers, D.; Devaney, K.L.; Ward, D.V.; Reyes, J.A.; Shah, S.A.; LeLeiko, N.; Snapper, S.B.; et al. Dysfunction of the Intestinal Microbiome in Inflammatory Bowel Disease and Treatment. Genome Biol. 2012, 13, R79. [Google Scholar] [CrossRef]
  109. Hall, A.B.; Tolonen, A.C.; Xavier, R.J. Human Genetic Variation and the Gut Microbiome in Disease. Nat. Rev. Genet. 2017, 18, 690–699. [Google Scholar] [CrossRef] [PubMed]
  110. Farkas, A.E.; Nusrat, A. Pharmacological Targeting of the Inflamed Intestinal Barrier. Curr. Pharm. Des. 2016, 22, 5400–5414. [Google Scholar] [CrossRef] [PubMed]
  111. Ismail, A.S.; Hooper, L.V. Epithelial Cells and Their Neighbors. IV. Bacterial Contributions to Intestinal Epithelial Barrier Integrity. Am. J. Physiol. Gastrointest. Liver Physiol. 2005, 289, G779–G784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Smith, K.; McCoy, K.D.; Macpherson, A.J. Use of Axenic Animals in Studying the Adaptation of Mammals to Their Commensal Intestinal Microbiota. Semin. Immunol. 2007, 19, 59–69. [Google Scholar] [CrossRef] [PubMed]
  113. Fellows, R.; Denizot, J.; Stellato, C.; Cuomo, A.; Jain, P.; Stoyanova, E.; Balázsi, S.; Hajnády, Z.; Liebert, A.; Kazakevych, J.; et al. Microbiota Derived Short Chain Fatty Acids Promote Histone Crotonylation in the Colon through Histone Deacetylases. Nat. Commun. 2018, 9, 105. [Google Scholar] [CrossRef]
  114. Gaudier, E.; Jarry, A.; Blottière, H.M.; de Coppet, P.; Buisine, M.P.; Aubert, J.P.; Laboisse, C.; Cherbut, C.; Hoebler, C. Butyrate Specifically Modulates MUC Gene Expression in Intestinal Epithelial Goblet Cells Deprived of Glucose. Am. J. Physiol. Gastrointest Liver Physiol. 2004, 287, G1168–G1174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Raqib, R.; Sarker, P.; Bergman, P.; Ara, G.; Lindh, M.; Sack, D.A.; Nasirul Islam, K.M.; Gudmundsson, G.H.; Andersson, J.; Agerberth, B. Improved Outcome in Shigellosis Associated with Butyrate Induction of an Endogenous Peptide Antibiotic. Proc. Natl. Acad. Sci. USA 2006, 103, 9178–9183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Laukoetter, M.G.; Bruewer, M.; Nusrat, A. Regulation of the Intestinal Epithelial Barrier by the Apical Junctional Complex. Curr. Opin Gastroenterol. 2006, 22, 85–89. [Google Scholar] [CrossRef] [PubMed]
  117. Buckley, A.; Turner, J.R. Cell Biology of Tight Junction Barrier Regulation and Mucosal Disease. Cold Spring Harb. Perspect Biol. 2018, 10, a029314. [Google Scholar] [CrossRef]
  118. Laval, L.; Martin, R.; Natividad, J.N.; Chain, F.; Miquel, S.; Desclée de Maredsous, C.; Capronnier, S.; Sokol, H.; Verdu, E.F.; van Hylckama Vlieg, J.E.T.; et al. Lactobacillus Rhamnosus CNCM I-3690 and the Commensal Bacterium Faecalibacterium Prausnitzii A2-165 Exhibit Similar Protective Effects to Induced Barrier Hyper-Permeability in Mice. Gut Microbes 2015, 6, 1–9. [Google Scholar] [CrossRef] [Green Version]
  119. Chichlowski, M.; De Lartigue, G.; German, J.B.; Raybould, H.E.; Mills, D.A. Bifidobacteria Isolated from Infants and Cultured on Human Milk Oligosaccharides Affect Intestinal Epithelial Function. J. Pediatr. Gastroenterol. Nutr. 2012, 55, 321–327. [Google Scholar] [CrossRef] [Green Version]
  120. Taminiau, J.A.; Gall, D.G.; Hamilton, J.R. Response of the Rat Small-Intestine Epithelium to Methotrexate. Gut 1980, 21, 486–492. [Google Scholar] [CrossRef] [Green Version]
  121. Zhang, X.; Zhang, D.; Jia, H.; Feng, Q.; Wang, D.; Liang, D.; Wu, X.; Li, J.; Tang, L.; Li, Y.; et al. The Oral and Gut Microbiomes Are Perturbed in Rheumatoid Arthritis and Partly Normalized after Treatment. Nat. Med. 2015, 21, 895–905. [Google Scholar] [CrossRef] [PubMed]
  122. Öman, A.; Dicksved, J.; Engstrand, L.; Berntson, L. Fecal Microbiota in Children with Juvenile Idiopathic Arthritis Treated with Methotrexate or Etanercept. Pediatr. Rheumatol. Online J. 2021, 19, 55. [Google Scholar] [CrossRef]
  123. Anand, S.; Mande, S.S. Diet, Microbiota and Gut-Lung Connection. Front. Microbiol. 2018, 9, 2147. [Google Scholar] [CrossRef] [Green Version]
  124. Picchianti-Diamanti, A.; Panebianco, C.; Salemi, S.; Sorgi, M.L.; Di Rosa, R.; Tropea, A.; Sgrulletti, M.; Salerno, G.; Terracciano, F.; D’Amelio, R.; et al. Analysis of Gut Microbiota in Rheumatoid Arthritis Patients: Disease-Related Dysbiosis and Modifications Induced by Etanercept. Int. J. Mol. Sci. 2018, 19, 2938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Hempel, G. Dose and Therapy Individualisation in Cancer Chemotherapy. In Handbook of Analytical Separations; Elsevier: Amsterdam, The Netherlands, 2004; Volume 5, pp. 169–194. ISBN 978-0-444-50972-7. [Google Scholar]
  126. Bonello, M.; Ronco, C. CHAPTER 69—Acute Renal Failure in Oncological Disorders and Tumor Lysis Syndrome. In Critical Care Nephrology, 2nd ed.; Ronco, C., Bellomo, R., Kellum, J.A., Eds.; W.B. Saunders: Philadelphia, PA, USA, 2009; pp. 379–384. ISBN 978-1-4160-4252-5. [Google Scholar]
  127. Howard, S.C.; McCormick, J.; Pui, C.-H.; Buddington, R.K.; Harvey, R.D. Preventing and Managing Toxicities of High-Dose Methotrexate. Oncologist 2016, 21, 1471–1482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Methotrexate. In Meyler’s Side Effects of Drugs; Elsevier: Amsterdam, The Netherlands, 2016; pp. 886–911. ISBN 978-0-444-53716-4.
  129. Green, J.M. Glucarpidase to Combat Toxic Levels of Methotrexate in Patients. Ther. Clin. Risk Manag. 2012, 8, 403–413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Canada, H. ARCHIVED—Policy Paper—Nutraceuticals/Functional Foods and Health Claims on Foods. Available online: https://www.canada.ca/en/health-canada/services/food-nutrition/food-labelling/health-claims/nutraceuticals-functional-foods-health-claims-foods-policy-paper.html (accessed on 13 September 2022).
  131. Gupta, S.; Chauhan, D.; Mehla, K.; Sood, P.; Nair, A. An Overview of Nutraceuticals: Current Scenario. J. Basic Clin. Pharm. 2010, 1, 55–62. [Google Scholar] [PubMed]
  132. Maddi, V.; Aragade, P.; Digge, V.; Nitalikar, M. Short Review Importance of Nutraceuticals in Health Management. Pharmacogn. Rev. 2007, 1, 377–379. [Google Scholar]
  133. Hollman, P.C.; Feskens, E.J.; Katan, M.B. Tea Flavonols in Cardiovascular Disease and Cancer Epidemiology. Proc. Soc. Exp. Biol. Med. 1999, 220, 198–202. [Google Scholar] [CrossRef] [Green Version]
  134. German, J.B.; Walzem, R.L. The Health Benefits of Wine. Annu. Rev. Nutr. 2000, 20, 561–593. [Google Scholar] [CrossRef]
  135. Thomas, B.; Ghebremeskel, K.; Lowy, C.; Crawford, M.; Offley-Shore, B. Nutrient Intake of Women with and without Gestational Diabetes with a Specific Focus on Fatty Acids. Nutrition 2006, 22, 230–236. [Google Scholar] [CrossRef] [PubMed]
  136. Coleman, M.D.; Eason, R.C.; Bailey, C.J. The Therapeutic Use of Lipoic Acid in Diabetes: A Current Perspective. Environ. Toxicol. Pharmacol. 2001, 10, 167–172. [Google Scholar] [CrossRef]
  137. Aggarwal, B.B.; Kumar, A.; Bharti, A.C. Anticancer Potential of Curcumin: Preclinical and Clinical Studies. Anticance. Res. 2003, 23, 363–398. [Google Scholar]
  138. ACS: Coriolus Versicolor. Available online: https://web.archive.org/web/20100625052416/http://www.cancer.org/docroot/ETO/content/ETO_5_3X_Coriolous_Versicolor.asp (accessed on 13 September 2022).
  139. Chidambara Murthy, K.N.; Vanitha, A.; Rajesha, J.; Mahadeva Swamy, M.; Sowmya, P.R.; Ravishankar, G.A. In Vivo Antioxidant Activity of Carotenoids from Dunaliella Salina—A Green Microalga. Life Sci. 2005, 76, 1381–1390. [Google Scholar] [CrossRef] [PubMed]
  140. Liu, R.; Wang, T.; Zhang, B.; Qin, L.; Wu, C.; Li, Q.; Ma, L. Lutein and Zeaxanthin Supplementation and Association with Visual Function in Age-Related Macular Degeneration. Investig. Ophthalmol. Vis. Sci. 2014, 56, 252–258. [Google Scholar] [CrossRef]
  141. Altmann, K.-H. Cancer Chemotherapy: Basic Science to the Clinic. By Rachel Airley. ChemMedChem 2009, 4, 2123–2124. [Google Scholar] [CrossRef]
  142. Shaikh, A.Y.; Shih, J.A. Chemotherapy-Induced Cardiotoxicity. Curr. Heart Fail Rep. 2012, 9, 117–127. [Google Scholar] [CrossRef]
  143. Thatishetty, A.V.; Agresti, N.; O’Brien, C.B. Chemotherapy-Induced Hepatotoxicity. Clin. Liver Dis. 2013, 17, 671–686. [Google Scholar] [CrossRef]
  144. Beijers, A.J.M.; Jongen, J.L.M.; Vreugdenhil, G. Chemotherapy-Induced Neurotoxicity: The Value of Neuroprotective Strategies. Neth. J. Med. 2012, 70, 18–25. [Google Scholar]
  145. Nerve Problems (Peripheral Neuropathy) and Cancer Treatment—Side Effects—NCI. Available online: https://www.cancer.gov/about-cancer/treatment/side-effects/nerve-problems (accessed on 13 September 2022).
  146. Davila, M.L. Neutropenic Enterocolitis. Curr. Opin. Gastroenterol. 2006, 22, 44–47. [Google Scholar] [CrossRef]
  147. Keidan, R.D.; Fanning, J.; Gatenby, R.A.; Weese, J.L. Recurrent Typhlitis. A Disease Resulting from Aggressive Chemotherapy. Dis. Colon Rectum 1989, 32, 206–209. [Google Scholar] [CrossRef] [PubMed]
  148. de Jonge, M.J.A.; Verweij, J. Renal Toxicities of Chemotherapy. Semin. Oncol. 2006, 33, 68–73. [Google Scholar] [CrossRef] [PubMed]
  149. Cocetta, V.; Tinazzi, M.; Giacomini, I.; Rosato, B.; Ragazzi, E.; Berretta, M.; Montopoli, M. Clinical Evidence of Interaction between Nutraceutical Supplementation and Platinum-Based Chemotherapy. Available online: https://www.ingentaconnect.com/content/ben/cmc/pre-prints/content-35638272 (accessed on 14 September 2022).
  150. Binu, P.; Gifty, K.; Vineetha, R.C.; Abhilash, S.; Arathi, P.; Nair, R.H. Eugenol, a Plant-Derived Phenolic Nutraceutical, Protects Thiol (SH) Group in Myocardium from ROS-Mediated Oxidation under Chemotherapeutic Stress Induced by Arsenic Trioxide—A in Vivo Model Study. Drug Chem. Toxicol. 2018, 41, 352–357. [Google Scholar] [CrossRef] [PubMed]
  151. Chiancone, F.; Carrino, M.; Fedelini, M.; Fabiano, M.; Persico, F.; Meccariello, C.; Fedelini, P. The Role of Protopine Associated with Nuciferine in Controlling Adverse Events during Hyperthermic Intravesical Chemotherapy Instillations. A Nutraceutical Approach to Control Adverse Event during Intravesical Instillations. Arch. Ital. Urol. Androl. 2020, 92, 177–181. [Google Scholar] [CrossRef] [PubMed]
  152. Dalaklioglu, S.; Genc, G.E.; Aksoy, N.H.; Akcit, F.; Gumuslu, S. Resveratrol Ameliorates Methotrexate-Induced Hepatotoxicity in Rats via Inhibition of Lipid Peroxidation. Hum. Exp. Toxicol. 2013, 32, 662–671. [Google Scholar] [CrossRef]
  153. Vardi, N.; Parlakpinar, H.; Cetin, A.; Erdogan, A.; Cetin Ozturk, I. Protective Effect of Beta-Carotene on Methotrexate-Induced Oxidative Liver Damage. Toxicol. Pathol. 2010, 38, 592–597. [Google Scholar] [CrossRef]
  154. Hadi, N.R.; Al-Amran, F.G.; Swadi, A. Metformin Ameliorates Methotrexate-Induced Hepatotoxicity. J. Pharmacol. Pharmacother. 2012, 3, 248–253. [Google Scholar] [CrossRef] [Green Version]
  155. Pomfret, E.A.; daCosta, K.-A.; Zeisel, S.H. Effects of Choline Deficiency and Methotrexate Treatment upon Rat Liver. J. Nutr. Biochem. 1990, 1, 533–541. [Google Scholar] [CrossRef]
  156. Akbulut, S.; Elbe, H.; Eris, C.; Dogan, Z.; Toprak, G.; Otan, E.; Erdemli, E.; Turkoz, Y. Cytoprotective Effects of Amifostine, Ascorbic Acid and N-Acetylcysteine against Methotrexate-Induced Hepatotoxicity in Rats. World J. Gastroenterol. 2014, 20, 10158–10165. [Google Scholar] [CrossRef]
  157. Çakır, T.; Baştürk, A.; Polat, C.; Aslaner, A.; Durgut, H.; Şehirli, A.Ö.; Gül, M.; Öğünç, A.V.; Gül, S.; Sabuncuoglu, M.Z.; et al. Does Alfa Lipoic Acid Prevent Liver from Methotrexate Induced Oxidative Injury in Rats? Acta Cir. Bras. 2015, 30, 247–252. [Google Scholar] [CrossRef] [Green Version]
  158. El-Sheikh, A.A.K.; Morsy, M.A.; Abdalla, A.M.; Hamouda, A.H.; Alhaider, I.A. Mechanisms of Thymoquinone Hepatorenal Protection in Methotrexate-Induced Toxicity in Rats. Mediat. Inflamm. 2015, 2015, e859383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Roghani, M.; Kalantari, H.; Khodayar, M.J.; Khorsandi, L.; Kalantar, M.; Goudarzi, M.; Kalantar, H. Alleviation of Liver Dysfunction, Oxidative Stress and Inflammation Underlies the Protective Effect of Ferulic Acid in Methotrexate-Induced Hepatotoxicity. Drug Des. Devel. Ther. 2020, 14, 1933–1941. [Google Scholar] [CrossRef] [PubMed]
  160. Bu, T.; Wang, C.; Meng, Q.; Huo, X.; Sun, H.; Sun, P.; Zheng, S.; Ma, X.; Liu, Z.; Liu, K. Hepatoprotective Effect of Rhein against Methotrexate-Induced Liver Toxicity. Eur. J. Pharmacol. 2018, 834, 266–273. [Google Scholar] [CrossRef]
  161. Mohamed, M.; El Sheikh, A.K.; Mohammed, H.H. Modulation of Liver P-Glycoprotien Expression May Contribute to Gossypin Protection against Methotrexate-Induced Hepatotoxicity. Indian J. Pharmacol. 2021, 53, 25–30. [Google Scholar] [CrossRef] [PubMed]
  162. Mehrzadi, S.; Fatemi, I.; Esmaeilizadeh, M.; Ghaznavi, H.; Kalantar, H.; Goudarzi, M. Hepatoprotective Effect of Berberine against Methotrexate Induced Liver Toxicity in Rats. Biomed. Pharmacother. 2018, 97, 233–239. [Google Scholar] [CrossRef] [PubMed]
  163. Tunalı-Akbay, T.; Sehirli, O.; Ercan, F.; Sener, G. Resveratrol Protects Against Methotrexate-Induced Hepatic Injury in Rats. J. Pharm. Pharm. Sci. 2010, 13, 303–310. [Google Scholar] [CrossRef]
  164. Olayinka, E.T.; Ore, A.; Adeyemo, O.A.; Ola, O.S. Ameliorative Effect of Gallic Acid on Methotrexate-Induced Hepatotoxicity and Nephrotoxicity in Rat. J. Xenobiotics 2016, 6, 14–18. [Google Scholar] [CrossRef] [Green Version]
  165. Jafaripour, L.; Naserzadeh, R.; Alizamani, E.; Javad Mashhadi, S.M.; Moghadam, E.R.; Nouryazdan, N.; Ahmadvand, H. Effects of Rosmarinic Acid on Methotrexate-Induced Nephrotoxicity and Hepatotoxicity in Wistar Rats. Indian J. Nephrol. 2021, 31, 218–224. [Google Scholar] [CrossRef]
  166. Al Kury, L.T.; Dayyan, F.; Ali Shah, F.; Malik, Z.; Khalil, A.A.K.; Alattar, A.; Alshaman, R.; Ali, A.; Khan, Z. Ginkgo Biloba Extract Protects against Methotrexate-Induced Hepatotoxicity: A Computational and Pharmacological Approach. Molecules 2020, 25, 2540. [Google Scholar] [CrossRef]
  167. Moghadam, A.R.; Tutunchi, S.; Namvaran-Abbas-Abad, A.; Yazdi, M.; Bonyadi, F.; Mohajeri, D.; Mazani, M.; Marzban, H.; Łos, M.J.; Ghavami, S. Pre-Administration of Turmeric Prevents Methotrexate-Induced Liver Toxicity and Oxidative Stress. BMC Complement. Altern Med. 2015, 15, 246. [Google Scholar] [CrossRef] [Green Version]
  168. Ahmad, A.; Alkharfy, K.M.; Bin Jardan, Y.A.; Shahid, M.; Ansari, M.A.; Alqahtani, S.; Jan, B.L.; Al-Jenoobi, F.I.; Raish, M. Sinapic Acid Mitigates Methotrexate-Induced Hepatic Injuries in Rats through Modulation of Nrf-2/HO-1 Signaling. Environ. Toxicol. 2021, 36, 1261–1268. [Google Scholar] [CrossRef] [PubMed]
  169. Elsawy, H.; Algefare, A.I.; Alfwuaires, M.; Khalil, M.; Elmenshawy, O.M.; Sedky, A.; Abdel-Moneim, A.M. Naringin Alleviates Methotrexate-Induced Liver Injury in Male Albino Rats and Enhances Its Antitumor Efficacy in HepG2 Cells. Biosci. Rep. 2020, 40, BSR20193686. [Google Scholar] [CrossRef] [PubMed]
  170. Wang, Y.-C.; Chuang, Y.-C.; Hsu, H.-W. The Flavonoid, Carotenoid and Pectin Content in Peels of Citrus Cultivated in Taiwan. Food Chem. 2008, 106, 277–284. [Google Scholar] [CrossRef]
  171. Nafees, S.; Rashid, S.; Ali, N.; Hasan, S.K.; Sultana, S. Rutin Ameliorates Cyclophosphamide Induced Oxidative Stress and Inflammation in Wistar Rats: Role of NFκB/MAPK Pathway. Chem. Biol. Interact. 2015, 231, 98–107. [Google Scholar] [CrossRef] [PubMed]
  172. Schwingel, T.E.; Klein, C.P.; Nicoletti, N.F.; Dora, C.L.; Hadrich, G.; Bica, C.G.; Lopes, T.G.; da Silva, V.D.; Morrone, F.B. Effects of the Compounds Resveratrol, Rutin, Quercetin, and Quercetin Nanoemulsion on Oxaliplatin-Induced Hepatotoxicity and Neurotoxicity in Mice. Naunyn. Schmiedebergs Arch. Pharmacol. 2014, 387, 837–848. [Google Scholar] [CrossRef] [PubMed]
  173. Erdogan, E.; Ilgaz, Y.; Gurgor, P.N.; Oztas, Y.; Topal, T.; Oztas, E. Rutin Ameliorates Methotrexate Induced Hepatic Injury in Rats. Acta Cir. Bras. 2015, 30, 778–784. [Google Scholar] [CrossRef]
  174. Çetin, A.; Kaynar, L.; Eser, B.; Karadağ, C.; Sarayman, B.; Öztürk, A.; Koçyiğit, İ.; Hacıoğlu, S.K.; Çiçek, B.; Silici, S. Beneficial Effects of Propolis on Methotrexate-Induced Liver Injury in Rats. Acta Oncol. Turc. 2011, 44, 18–23. [Google Scholar]
  175. Ali, N.; Rashid, S.; Nafees, S.; Hasan, S.K.; Sultana, S. Beneficial Effects of Chrysin against Methotrexate-Induced Hepatotoxicity via Attenuation of Oxidative Stress and Apoptosis. Mol. Cell Biochem. 2014, 385, 215–223. [Google Scholar] [CrossRef]
  176. Hasan, H.; Ismail, H.; El-Orfali, Y.; Khawaja, G. Therapeutic Benefits of Indole-3-Carbinol in Adjuvant-Induced Arthritis and Its Protective Effect against Methotrexate Induced-Hepatic Toxicity. BMC Complement. Altern Med. 2018, 18, 337. [Google Scholar] [CrossRef]
  177. Amirfakhrian, H.; Abedi, S.M.; Sadeghi, H.; Azizi, S.; Hosseinimehr, S.J. The Use of 99mTc-Phytate for Assessment the Protective Effect of Vitamin E against Hepatotoxicity Induced by Methotrexat in Rat. Nucl. Med. Rev. 2018, 21, 8–13. [Google Scholar] [CrossRef] [Green Version]
  178. Demiryilmaz, I.; Sener, E.; Cetin, N.; Altuner, D.; Suleyman, B.; Albayrak, F.; Akcay, F.; Suleyman, H. Biochemically and Histopathologically Comparative Review of Thiamine’s and Thiamine Pyrophosphate’s Oxidative Stress Effects Generated with Methotrexate in Rat Liver. Med. Sci. Monit. 2012, 18, BR475–BR481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Widemann, B.C.; Adamson, P.C. Understanding and Managing Methotrexate Nephrotoxicity. Oncologist 2006, 11, 694–703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Kitamura, M.; Kitamura, S.; Fujioka, M.; Kamijo, R.; Sato, S.; Sawayama, Y.; Uramatsu, T.; Obata, Y.; Mochizuki, Y.; Nishikido, M.; et al. Methotrexate-Induced Acute Kidney Injury in Patients with Hematological Malignancies: Three Case Reports with Literature Review. Ren. Replace. Ther. 2018, 4, 39. [Google Scholar] [CrossRef]
  181. Aladaileh, S.H.; Hussein, O.E.; Abukhalil, M.H.; Saghir, S.A.M.; Bin-Jumah, M.; Alfwuaires, M.A.; Germoush, M.O.; Almaiman, A.A.; Mahmoud, A.M. Formononetin Upregulates Nrf2/HO-1 Signaling and Prevents Oxidative Stress, Inflammation, and Kidney Injury in Methotrexate-Induced Rats. Antioxidants 2019, 8, 430. [Google Scholar] [CrossRef] [Green Version]
  182. Hassanein, E.H.M.; Shalkami, A.-G.S.; Khalaf, M.M.; Mohamed, W.R.; Hemeida, R.A.M. The Impact of Keap1/Nrf2, P38MAPK/NF-ΚB and Bax/Bcl2/Caspase-3 Signaling Pathways in the Protective Effects of Berberine against Methotrexate-Induced Nephrotoxicity. Biomed. Pharmacother. 2019, 109, 47–56. [Google Scholar] [CrossRef]
  183. Paeonol: A Bio-Antimutagen Isolated from a Crude Drug, Moutan Cortex. Available online: https://www.jstage.jst.go.jp/article/bbb1961/51/5/51_5_1441/_article (accessed on 17 September 2022).
  184. Zhang, L.; Li, D.-C.; Liu, L.-F. Paeonol: Pharmacological Effects and Mechanisms of Action. Int. Immunopharmacol. 2019, 72, 413–421. [Google Scholar] [CrossRef]
  185. Yan, D.; Saito, K.; Ohmi, Y.; Fujie, N.; Ohtsuka, K. Paeoniflorin, a Novel Heat Shock Protein-Inducing Compound. Cell Stress Chaperones 2004, 9, 378–389. [Google Scholar] [CrossRef]
  186. Morsy, M.A.; El-Sheikh, A.A.K.; Abdel-Hafez, S.M.N.; Kandeel, M.; Abdel-Gaber, S.A. Paeonol Protects Against Methotrexate-Induced Nephrotoxicity via Upregulation of P-Gp Expression and Inhibition of TLR4/NF-ΚB Pathway. Front. Pharmacol. 2022, 13, 774387. [Google Scholar] [CrossRef]
  187. Wei, X.; Wu, Y.; Tang, H.; Wang, B.; Wang, Y.; Sun, W.; Asenso, J.; Xiao, F.; Wang, C. CP-25 Ameliorates Methotrexate Induced Nephrotoxicity via Improving Renal Apoptosis and Methotrexate Excretion. J. Pharmacol. Sci. 2021, 146, 21–28. [Google Scholar] [CrossRef]
  188. Elsawy, H.; Alzahrani, A.M.; Alfwuaires, M.; Abdel-Moneim, A.M.; Khalil, M. Nephroprotective Effect of Naringin in Methotrexate Induced Renal Toxicity in Male Rats. Biomed. Pharmacother. 2021, 143, 112180. [Google Scholar] [CrossRef]
  189. Oguz, E.; Kocarslan, S.; Tabur, S.; Sezen, H.; Yilmaz, Z.; Aksoy, N. Effects of Lycopene Alone or Combined with Melatonin on Methotrexate-Induced Nephrotoxicity in Rats. Asian Pac. J. Cancer Prev. 2015, 16, 6061–6066. [Google Scholar] [CrossRef] [Green Version]
  190. Öktem, F.; Yilmaz, H.R.; Ozguner, F.; Olgar, S.; Ayata, A.; Uzar, E.; Uz, E. Methotrexate-Induced Renal Oxidative Stress in Rats: The Role of a Novel Antioxidant Caffeic Acid Phenethyl Ester. Toxicol. Ind. Health 2006, 22, 241–247. [Google Scholar] [CrossRef]
  191. Asci, H.; Ozmen, O.; Ellidag, H.Y.; Aydin, B.; Bas, E.; Yilmaz, N. The Impact of Gallic Acid on the Methotrexate-Induced Kidney Damage in Rats. J. Food Drug Anal. 2017, 25, 890–897. [Google Scholar] [CrossRef]
  192. Morsy, M.A.; Ibrahim, S.A.; Amin, E.F.; Kamel, M.Y.; Rifaai, R.A.; Hassan, M.K. Curcumin Ameliorates Methotrexate-Induced Nephrotoxicity in Rats. Adv. Pharmacol. Sci. 2013, 2013, 387071. [Google Scholar] [CrossRef] [Green Version]
  193. Erboga, M.; Aktas, C.; Erboga, Z.F.; Donmez, Y.B.; Gurel, A. Quercetin Ameliorates Methotrexate-Induced Renal Damage, Apoptosis and Oxidative Stress in Rats. Ren. Fail. 2015, 37, 1492–1497. [Google Scholar] [CrossRef] [Green Version]
  194. Sherif, I.O.; Al-Shaalan, N.H.; Sabry, D. Ginkgo Biloba Extract Alleviates Methotrexate-Induced Renal Injury: New Impact on PI3K/Akt/MTOR Signaling and MALAT1 Expression. Biomolecules 2019, 9, 691. [Google Scholar] [CrossRef] [Green Version]
  195. Ahmed, J.H.; Abdulmajeed, I.M. Effect of Nigella Sativa (Black Seeds) against Methotrexate-Induced Nephrotoxicity in Mice. J. Intercult Ethnopharmacol. 2017, 6, 9–13. [Google Scholar] [CrossRef]
  196. Ahmed, W.; Zaki, A.; Nabil, T. Prevention of Methotrexate-Induced Nephrotoxicity by Concomitant Administration of Garlic Aqueous Extract in Rat. Turk. J. Med. Sci. 2015, 45, 507–516. [Google Scholar] [CrossRef]
  197. Abouelela, M.E.; Orabi, M.A.A.; Abdelhamid, R.A.; Abdelkader, M.S.; Madkor, H.R.; Darwish, F.M.M.; Hatano, T.; Elsadek, B.E.M. Ethyl Acetate Extract of Ceiba pentandra (L.) Gaertn. Reduces Methotrexate-Induced Renal Damage in Rats via Antioxidant, Anti-Inflammatory, and Antiapoptotic Actions. J. Tradit Complement. Med. 2020, 10, 478–486. [Google Scholar] [CrossRef]
  198. Khoshnoud, S.; Mohseni Kouchesfahani, H.; Nabiuni, M. Evaluation of The Protective Effect of Hydro-Alcoholic Extract of Raspberry Fruit on Aquaporin1 Expression in Rats Kidney Treated by Methotrexate. Cell J. 2017, 19, 306–313. [Google Scholar] [CrossRef]
  199. Sharma, S.; Baboota, S.; Amin, S.; Mir, S.R. Ameliorative Effect of a Standardized Polyherbal Combination in Methotrexate-Induced Nephrotoxicity in the Rat. Pharm. Biol. 2020, 58, 184–199. [Google Scholar] [CrossRef] [Green Version]
  200. Miyazono, Y.; Gao, F.; Horie, T. Oxidative Stress Contributes to Methotrexate-Induced Small Intestinal Toxicity in Rats. Scand. J. Gastroenterol. 2004, 39, 1119–1127. [Google Scholar] [CrossRef]
  201. Gao, F.; Horie, T. A Synthetic Analog of Prostaglandin E1 Prevents the Production of Reactive Oxygen Species in the Intestinal Mucosa of Methotrexate-Treated Rats. Life Sci. 2002, 71, 1091–1099. [Google Scholar] [CrossRef]
  202. Altmann, G.G. Changes in the Mucosa of the Small Intestine Following Methotrexate Administration or Abdominal X-Irradiation. Am. J. Anat. 1974, 140, 263–279. [Google Scholar] [CrossRef]
  203. Song, W.-B.; Wang, Y.-Y.; Meng, F.-S.; Zhang, Q.-H.; Zeng, J.-Y.; Xiao, L.-P.; Yu, X.-P.; Peng, D.; Su, L.; Xiao, B.; et al. Curcumin Protects Intestinal Mucosal Barrier Function of Rat Enteritis via Activation of MKP-1 and Attenuation of P38 and NF-ΚB Activation. PLoS ONE 2010, 5, e12969. [Google Scholar] [CrossRef]
  204. Fox, A.D.; Kripke, S.A.; De Paula, J.; Berman, J.M.; Settle, R.G.; Rombeau, J.L. Effect of a Glutamine-Supplemented Enteral Diet on Methotrexate-Induced Enterocolitis. JPEN J. Parenter Enter. Nutr. 1988, 12, 325–331. [Google Scholar] [CrossRef]
  205. Harsha, W.T.F.; Kalandarova, E.; McNutt, P.; Irwin, R.; Noel, J. Nutritional Supplementation with Transforming Growth Factor-Beta, Glutamine, and Short Chain Fatty Acids Minimizes Methotrexate-Induced Injury. J. Pediatr. Gastroenterol. Nutr. 2006, 42, 53–58. [Google Scholar] [CrossRef]
  206. Marks, S.L.; Cook, A.K.; Reader, R.; Kass, P.H.; Théon, A.P.; Greve, C.; Rogers, Q.R. Effects of Glutamine Supplementation of an Amino Acid-Based Purified Diet on Intestinal Mucosal Integrity in Cats with Methotrexate-Induced Enteritis. Am. J. Vet. Res. 1999, 60, 755–763. [Google Scholar]
  207. Shi, C.-J.; Wen, X.-S.; Gao, H.-F.; Liu, Z.-H.; Xu, X.-K.; Li, L.-F.; Shen, T.; Xian, C.J. Steamed Root of Rehmannia Glutinosa Libosch (Plantaginaceae) Alleviates Methotrexate-Induced Intestinal Mucositis in Rats. J. Ethnopharmacol. 2016, 183, 143–150. [Google Scholar] [CrossRef]
  208. Aldhahrani, A. Protective Effects of Guarana (Paullinia Cupana) against Methotrexate-Induced Intestinal Damage in Mice. Food Sci. Nutr. 2021, 9, 3397–3404. [Google Scholar] [CrossRef]
  209. Zhang, H.; Wang, J.; Lang, W.; Liu, H.; Zhang, Z.; Wu, T.; Li, H.; Bai, L.; Shi, Q. Albiflorin Ameliorates Inflammation and Oxidative Stress by Regulating the NF-ΚB/NLRP3 Pathway in Methotrexate-Induced Enteritis. Int. Immunopharmacol. 2022, 109, 108824. [Google Scholar] [CrossRef]
  210. Ahmed, A.A.M.; Selim, M.A.A.; El-Sayed, N.M. α-Lipoic Acid Ameliorates Oral Mucositis and Oxidative Stress Induced by Methotrexate in Rats. Histological and Immunohistochemical Study. Life Sci. 2017, 171, 51–59. [Google Scholar] [CrossRef]
  211. da Silva Ferreira, A.R.; van der Aa, S.A.J.; Wehkamp, T.; Wardill, H.R.; Ten Klooster, J.P.; Garssen, J.; Harthoorn, L.F.; Hartog, A.; Harmsen, H.J.M.; Tissing, W.J.E.; et al. Development of a Self-Limiting Model of Methotrexate-Induced Mucositis Reinforces Butyrate as a Potential Therapy. Sci. Rep. 2021, 11, 22911. [Google Scholar] [CrossRef]
  212. Koppelmann, T.; Pollak, Y.; Ben-Shahar, Y.; Gorelik, G.; Sukhotnik, I. The Mechanisms of the Anti-Inflammatory and Anti-Apoptotic Effects of Omega-3 Polyunsaturated Fatty Acids during Methotrexate-Induced Intestinal Damage in Cell Line and in a Rat Model. Nutrients 2021, 13, 888. [Google Scholar] [CrossRef]
  213. Yilmaz, E.; Azizoglu, Z.B.; Aslan, K.; Erdem, S.; Haliloglu, Y.; Suna, P.A.; Yay, A.H.; Deniz, K.; Tasdemir, A.; Per, S.; et al. Therapeutic Effects of Vitamin D and IL-22 on Methotrexate-Induced Mucositis in Mice. Anticancer Drugs 2022, 33, 11–18. [Google Scholar] [CrossRef]
  214. da Silva Ferreira, A.R.; Wardill, H.R.; Havinga, R.; Tissing, W.J.E.; Harmsen, H.J.M. Prophylactic Treatment with Vitamins C and B2 for Methotrexate-Induced Gastrointestinal Mucositis. Biomolecules 2020, 11, 34. [Google Scholar] [CrossRef]
  215. Gao, Y.; Sun, Q.; Yang, X.; Lu, W.; Zhao, Y.; Ge, W.; Yang, Y.; Xu, X.; Zhang, J. Orally Administered Salecan Ameliorates Methotrexate-Induced Intestinal Mucositis in Mice. Cancer Chemother. Pharmacol. 2019, 84, 105–116. [Google Scholar] [CrossRef]
  216. Yamamoto, A.; Itoh, T.; Nasu, R.; Kajiwara, E.; Nishida, R. Sodium Alginate Inhibits Methotrexate-Induced Gastrointestinal Mucositis in Rats. Biol. Pharm. Bull. 2013, 36, 1528–1534. [Google Scholar] [CrossRef]
  217. Southcott, E.; Tooley, K.L.; Howarth, G.S.; Davidson, G.P.; Butler, R.N. Yoghurts Containing Probiotics Reduce Disruption of the Small Intestinal Barrier in Methotrexate-Treated Rats. Dig. Dis. Sci. 2008, 53, 1837–1841. [Google Scholar] [CrossRef]
  218. Cannon, G.W. Methotrexate Pulmonary Toxicity. Rheum Dis. Clin. N. Am. 1997, 23, 917–937. [Google Scholar] [CrossRef]
  219. Lateef, O.; Shakoor, N.; Balk, R.A. Methotrexate Pulmonary Toxicity. Expert Opin. Drug Saf. 2005, 4, 723–730. [Google Scholar] [CrossRef] [PubMed]
  220. Dawson, J.K.; Clewes, A.R.; Hendry, J. Pulmonary Effects of Low-Dose Methotrexate Therapy. Clin. Pulm. Med. 2004, 11, 307–317. [Google Scholar] [CrossRef]
  221. Mammadov, R.; Suleyman, B.; Akturan, S.; Cimen, F.K.; Kurt, N.; Suleyman, Z.; Malkoc, İ. Effect of Lutein on Methotrexate-Induced Oxidative Lung Damage in Rats: A Biochemical and Histopathological Assessment. Korean J. Intern. Med. 2019, 34, 1279–1286. [Google Scholar] [CrossRef] [PubMed]
  222. Arpag, H.; Gül, M.; Aydemir, Y.; Atilla, N.; Yiğitcan, B.; Cakir, T.; Polat, C.; Þehirli, Ö.; Sayan, M. Protective Effects of Alpha-Lipoic Acid on Methotrexate-Induced Oxidative Lung Injury in Rats. J. Investig. Surg 2018, 31, 107–113. [Google Scholar] [CrossRef]
Figure 1. The main mechanisms of MTX toxicity in experimental disease models of organ injury. Several organ dysfunctions have been reported following MTX treatment: MTX-induced liver toxicity represented by fibrosis and cirrhosis, MTX-associated lung dysfunction consisted of pneumonitis and fibrosis, kidney injury related to MTX treatment, including acute kidney injury and tubulo-nephritis, intestinal changes upon MTX including mucositis, enteritis, and dysbiosis. Abbreviations: AKI, acute kidney injury; GSH, glutathione; ROS, reactive oxygen species.
Figure 1. The main mechanisms of MTX toxicity in experimental disease models of organ injury. Several organ dysfunctions have been reported following MTX treatment: MTX-induced liver toxicity represented by fibrosis and cirrhosis, MTX-associated lung dysfunction consisted of pneumonitis and fibrosis, kidney injury related to MTX treatment, including acute kidney injury and tubulo-nephritis, intestinal changes upon MTX including mucositis, enteritis, and dysbiosis. Abbreviations: AKI, acute kidney injury; GSH, glutathione; ROS, reactive oxygen species.
Microorganisms 10 02053 g001
Table 1. Synopsis of MTX as disease-modifying agent in multiple disease conditions.
Table 1. Synopsis of MTX as disease-modifying agent in multiple disease conditions.
DiseaseEnd-Organ EffectMolecular MechanismEfficient Dose IndicatedMost Common Adverse Effects
Acute lymphoblastic leukemiaNeoplastic cells, abnormally fast-dividing cellsDHFR inhibition, disruption of de novo nucleotide biosynthesis and DNA replication, resulting in cell deathInduction 3.3 mg/m2
Maintenance 30 mg/m2/week
-
Liver enzymes elevation
-
Hepatic fibrosis/cirrhosis
-
GI bleed
-
Diarrhea
-
Ulcerative stomatitis
-
Leukopenia
-
Anemia
-
Aplastic anemia
-
Pancytopenia
-
Pneumonia
-
Pulmonary fibrosis
-
Renal insufficiency
-
Hematuria
-
Toxic epidermal necrolysis
-
Nausea
-
Drowsiness
Acute promyelocytic leukemia
Meningeal leukemia12 mg/m2/every 2–5 days until the cell count of the CSF returns to normal
Burkitt’s lymphoma and other non-Hodgkin’s lymphomas10–25 mg/day
4–8 days
Mycosis Fungoides2.5–10 mg/day orally or 50 mg/week i.m.
Epidermoid cancers of the head and neck30–40 mg/m2/week i.v
Early-stage breast cancer
Squamous cell carcinoma
Small cell carcinoma
Osteosarcoma12 g/m2 i.v.
Chorioadenoma destruens15–30 mg/day
5 day course
Hydatidiform mole
Severe psoriasisT cells
B cells
Adenosine accumulation, inhibition of T-cell activation, downregulation of B cellsSingle dose 10–25 mg/week
Rheumatoid arthritisSingle dose 7.5 mg/week
Polyarticular course juvenile rheumatoid arthritisSingle dose 10 mg/m2/week
References [47,57,58,61,62].
Table 2. Gut microbiota-related changes following treatment with MTX and the associated epithelial changes.
Table 2. Gut microbiota-related changes following treatment with MTX and the associated epithelial changes.
Gut Microbiota Changes in Subjects Exposed to MTXIntestinal Epithelial ChangesSamplesTechniqueReferences
Peptostreptococcaceae and Porphyromonadaceae
Ruminococcaceae, Erysipelotrichaceae
NR22 Male Sprague Dawley rats 7 to 8 weeks oldFecal DNA extraction and sequencing[26]
Lactobacillus
Muribaculaceae
↓ ZO-1, claudin-1, and E-cadherin 8-week-old male mice (6 per group)Fecal DNA extraction and sequencing[32]
H. filiformis and Bacteroides sp.
P. intermedia
NR21 RA patients at pre and post-MTX + tripterygium glycosidesMetagenomic shotgun sequencing[121]
↑ Subdoligranulum
Rikenellaceae, Veillonellaceae, Bacteroidales_S24-7_group, Alistipes Prevotellaceae_NK3B31_group
NRFecal samples from 29 children with
JIA treated with MTX
DNA extraction, amplification, and sequencing[122]
EnterobaterialesNR11 patients with RA receiving MTXDNA extraction and metagenomic sequencing[124]
Abbreviations: JIA, juvenile idiopathic arthritis; MTX, methotrexate; NR, not reported; RA, rheumatoid arthritis; ZO-1, zonula occludens-1.
Table 3. End-organ effects of nutraceuticals used in experimental models of MTX-induced toxicity.
Table 3. End-organ effects of nutraceuticals used in experimental models of MTX-induced toxicity.
NameModelClassSourceTherapeutic EffectsRef.
CholineIn vivo liver toxicity rat modelVitamin/nutrientMultiple sources in meat and plants↑ PCho, GroPCho, and betaine [155]
Gossypin/gossypentinIn vivo liver toixicity rat modelFlavonoid/plant extractHibiscus sabdariffa↓BAX, TGF-β, caspase 3, and NF-κB
↓hepatic fibrosis
[161]
Epicatechin/CatechinIn vivo liver toxicity mice modelFlavonoid/plant extractMimosa catechu↓ IL-1β, TNF-α, and NO
↓ MDA
GSH level and activity level of catalase, SOD, and GPx ↑
[30]
Thiamine and thiamine pyrophosphateIn vivo liver toixicity rat modelVitaminWhole grains, legumes, and some meats and fishThiamine no protective effects reported
TPP effects on: MDA and MPO ↓
GSH and SOD ↑
[178]
ThymoquinoneIn vivo liver toixicity rat modelPlant extractNigella sativa↓TNF-α, NF-κB COX-2 expressions
↓MDA
↑glutathione and catalase
[158]
Ferulic acidIn vivo liver toxicity mice modelPlant extractFerula communis↓MDA, IL-6, and TNF-α
↓accumulation of inflammatory cells
↓nuclear pyknosis
↑GSH, CAT, TAC
[159]
Rhein/cassic acidIn vivo liver toxicity rat model
In vitro normal human hepatocyte (L02 cells) model
Plant extractRheum undulatum, Rheum palmatum,
Cassia reticulata
↑cell survival rate
↓apoptosis
↑Nrf2, Bcl-2, HO-1 and GCLC
↓Bax
↓NF-κB, TNF-α and caspase-3
[160]
BerberineIn vivo liver toixicity rat modelPlant extractBerberis vulgaris, Berberis aristata, Mahonia aquifolium, Hydrastis canadensis, Xanthorhiza simplicissima, Phellodendron amurense ↓MDA, PC, NO levels and MPO activity
↑GSH level, SOD, GPx and CAT activities
[162]
ResveratrolIn vivo liver toixicity rat modelPlant extract Skin of grapes, blueberries, raspberries, mulberries, and peanuts↓MDA levels, MPO and TF activities and collagen contents
↑GSH
↓TNF-α
↓TBARS, CAT, and GST
[152,163]
Ginko bilobaIn silico bio computational model
In vivo liver toxicity rat model
Plant extractGinko biloba tree↓caspase-3, JNK and TNF-α
↓apoptosis
↑GSH and GST
in silico: drug-receptor interactions stabilized by a low energy value and with a good number of hydrogen bonds
[166]
Abbreviations: BAX, BCL2-associated X protein; Bcl-2, B-cell lymphoma-2; caspase-3, cysteine aspartic acid-specific protease 3; CAT, catalase; COX2: cyclooxygenase-2; GCLC, glutamate-cysteine ligase catalytic subunit; GPx, glutathione peroxidase; GroPCho, glycerophosphocholine; GSH, gluthatione; GST, glutathione S-transferase; HO-1, heme oxygenase 1, IL-1β, interleukin 1 beta; IL-6, interleukin-6; JNK, c-Jun N-terminal kinases; MDA, malondialdehyde; MPO, mieloperoxidase; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NO, nitric oxide; Nrf2, erythroid 2-related factor 2; PCho, phosphocholine; SOD, superoxide dismutase, TAC, total antioxidant capacity; TBARS, thiobarbituric acid reactive substances; TF, tissue factor; TNF-α, tumor necrosis factor-alpha; TNF-β, tumor necrosis factor-beta; TPP, thiamine pyrophosphate.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Marin, G.-E.; Neag, M.-A.; Burlacu, C.-C.; Buzoianu, A.-D. The Protective Effects of Nutraceutical Components in Methotrexate-Induced Toxicity Models—An Overview. Microorganisms 2022, 10, 2053. https://doi.org/10.3390/microorganisms10102053

AMA Style

Marin G-E, Neag M-A, Burlacu C-C, Buzoianu A-D. The Protective Effects of Nutraceutical Components in Methotrexate-Induced Toxicity Models—An Overview. Microorganisms. 2022; 10(10):2053. https://doi.org/10.3390/microorganisms10102053

Chicago/Turabian Style

Marin, Gheorghe-Eduard, Maria-Adriana Neag, Codrin-Constantin Burlacu, and Anca-Dana Buzoianu. 2022. "The Protective Effects of Nutraceutical Components in Methotrexate-Induced Toxicity Models—An Overview" Microorganisms 10, no. 10: 2053. https://doi.org/10.3390/microorganisms10102053

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop