Please cite this paper as :

Ghosh, P., Dey, T. and Bandyopadhyay, D. 2022. The pleiotropic role of melatonin against chromium-induced cardiovascular infirmities: a mechanistic insight. Melatonin Research. 5, 3 (Sep. 2022), 209-253. DOI:https://doi.org/https://doi.org/10.32794/mr112500130.


Review 

The pleiotropic role of melatonin against chromium-induced cardiovascular infirmities: a mechanistic insight

Priyanka Ghosh1, Tiyasa Dey1, Debasish Bandyopadhyay1* 

1Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, 92, APC Road, Kolkata-700009, India

*Correspondence: debasish63@gmail.comTel: +91 9433072066

Running title: Melatonin protects chromium induced myocardial damages

Received: May 6, 2022; Accepted: August 30, 2022


ABSTRACT

     Currently, cardiovascular diseases are still the number one killer in the world. These include hypertension, coronary heart disease, ischemic heart disease, myocardial infarction, congestive heart failure, cardiac arrhythmias, etc. One of the risk factors for cardiovascular diseases is environmental heavy metal pollution which makes the victims more vulnerable to sudden cardiac death. Chromium (Cr) is one of the metals. Cr(VI) is the most hazardous one among its variants.  It is readily across the plasma membrane to cause oxidative damage to intracellular molecules including LDL, proteins, and DNA; therefore, promotes endothelial dysfunction and Ca2+ overload in the heart. As to its molecular mechanism, Cr(VI) downregulates the expressions of SIRTUINS, FOXOs, PGC-1α, and AMPK and upregulates the P53, Akt, and NF-κβ, causing alteration in metabolic pathways, inhibiting mitochondrial biogenesis, inducing autophagy and apoptosis. In addition, Cr(VI) alters the expressions of Th1 cytokines (IL-1β, IL-2, IL-12, TNF-α, and IFN-γ) as well as Th2 cytokines (IL-4, IL-5, and IL-10) to induce myocardial inflammation. Melatonin, a potent antioxidant, and an efficient metal chelator can neutralize almost all the alterations caused by Cr(VI). Thus, melatonin can be a selected molecule to protect against Cr(VI)-induced cardiovascular toxicity. This review highlights the etiology of Cr(VI) associated heart diseases and the potentiality of melatonin to prevent Cr(VI)-mediated cardiac oxidative stress, apoptosis and inflammation. 

Keywords: Chromium, oxidative stress, reactive oxygen species (ROS), apoptosis, inflammation, cardiovascular diseases, myocardial ischemia, antioxidant, melatonin.

 _____________________________________________________________________________________________________________________________________________________________________________________


1. INTRODUCTION 

     Environmental pollutants are destroying our ecosystem at an expedited rate and become a worldwide threat to human health. Heavy metals are the topmost concerning because of their widely distribution made by human activity. These metals are cardiac toxic and can cause cardiovascular disease (CVD), coronary artery disease (CAD), and stroke (1). Several complications such as thrombosis, arrhythmias, hypertension, fibrinolysis, and ischemic heart disease (IHD) are associated with the increased mortality and morbidity from CVD (2). The association between heavy metals and CVD is well defined. Buhari et al. (3) have claimed that the formation of free radicals by heavy metals increased serum lipid levels and oxidation of low-density lipoprotein cholesterol (LDL-C) which results in atherosclerosis (3). In addition, the oxidative stress caused by heavy metals hampers several cellular events, including growth, proliferation, differentiation, damage-repairing and apoptosis, leading to irresistible tissue injuries (4). Moreover, the disruption of the antioxidant defence system further promotes generation of reactive oxygen species (ROS) from mitochondria. The excessive increase of ROS in mitochondria leads to a catastrophic cycle of mitochondrial DNA (mtDNA) damage that ultimately causes heart failure (5).

     Although heavy metals are toxic to cells, among them chromium (Cr) is a two-faced metal since it exerts both beneficial and detrimental effects (6). Cr is a naturally occurring compound found extensively in the earth’s crust; however, anthropogenic processes such as mining, textile, steel, and chrome plating pollute the air, water, and food. Cr has several oxidation states ranging from -2 to +6. In comparison to different oxidation states, trivalent (CrIII) and hexavalent (CrVI) are the most stable and dominant forms in nature (7). Chromium (III) is a biologically active form of Cr and is widely used as a therapeutic or pharmacological substance that improves glucose tolerance, insulin sensitivity and fat metabolism (8–10). Current studies have cast the doubt on the nutritive action of Cr(III) because it may also bind to  DNA to induce genotoxicity and mutation (11, 12). Cr(VI) promotes ROS generation [such as hydroxyl radical (HO.), superoxide anion (O2.-), hydrogen peroxide (H2O2), alters intracellular metabolisms, damages DNA, and disrupts cellular integrity (13–15). In addition, recent studies have reported that Cr(VI) aggravates heart dysfunction (16), myocardial infarction (MI) (17), and other cardiovascular risks (18).

     Based on the data of the World Health Organization (WHO-2019), CVDs are the leading cause of death globally. An estimated 17.9 million people died from CVDs, and 85% were due to heart attack (19). This evokes the demand for suitable medicines and therapeutic approaches which can reduce death rate of CVDs. In the path of searching, we found that melatonin as a potent antioxidant was conventionally used for the treatment of heart diseases (20, 21). Melatonin is found in both animal and plant tissues (22, 23). It reduces oxidative stress, lipid peroxidation (LPO), inflammation, necrosis, apoptosis, and mitochondrial permeability transition pore opening (24). The cardio-protective mechanisms of melatonin involve both direct and indirect pathways including antihypertensive, antilipidemic, antiadrenergic, and immunomodulatory activities (23). Evidence showed that melatonin reduced hypertension, hypertensive cardiomyopathy (25, 26), coronary heart disease (CHD) (27), myocardial infarction (28), congestive heart failure (CHF) (29, 30), cardiac arrhythmias (31) as well as sudden cardiac arrest (32). This review is focused on the cardiac toxicity of Cr(VI) as well as the protective effects of melatonin on the cardiovascular damages caused by  Cr(VI).

 

2. POSITIVE AND NEGATIVE EFFECTS OF TWO MAJOR CHROMIUM COMPOUNDS ON CARDIOVASCULAR SYSTEM  

     The cardiovascular system, also known as the circulatory system, is susceptible to oxidative damage. Imbalanced redox status triggers several cellular and molecular damaging pathways that ultimately lead to the progression of hypertension, atherosclerosis, and other vascular complications.

     Both Cr(VI), and Cr(III) are used vigorously in industrial processes. However,  significant differences are present between them including oxidation states, occurrence, chemical/biological properties, and toxicity as well (33). Cr(III) is used as a nutritional supplement and it is 10 to 100 times less toxic than Cr(VI) due to its poor absorption (about 1%) from the gut (34) as well as its impermeability through the plasma membrane (35). The exposure to Cr(III) is rare from environment since Cr(III) is rapidly oxidized into Cr(VI) in sludge, soil as well as in groundwater under aerobic conditions (36–38). While Cr(VI) can be exposed through water, food, and the occupational area. The United States Environmental Protection Agency (USEPA) has categorized Cr(VI) under the top 20 hazardous contaminants (39), and International Agency for Research on Cancer (IARC) has declared Cr(VI) as a human carcinogen (40). 

2.1. Beneficial effects of Cr(III) on cardiovascular system.

     Studies of the past few decades have focused on the regulatory roles of Cr(III) on fat, protein, and glucose metabolism (9, 41) as well as the maintenance of levels and activities of insulin (34, 42). It promotes glucose oxidation, lipogenesis, and transportation of D-galactose across the membrane mediated by insulin  (34). Cr(III) is an essential micronutrient popularly known to form a biologically active organic complex called the ‘glucose tolerance factor’ (GTF) (43). In mammals, Cr(III) deficiency causes impaired glucose tolerance, high insulin, free fatty acids, low respiratory quotient, and abnormal nitrogen metabolism (44). Cr(III) is thought to be beneficial for cardiac health because it can modulate the levels of cholesterol and specific lipoproteins (45). These activities may be mediated by insulin since insulin has a lipogenic activity (46) to lowering triglyceride synthesis in the liver and enhancing the number of LDL receptors to minimize the level of LDL (47). In addition, Cr(III) is incapable of oxidizing LDL particles (48). Several studies have reported that Cr(III) reduces plaque formation in the area of aortic intima by simultaneously lowering the total cholesterol and increasing the HDL fractions in the blood (49–52). Glucose intolerance and high level of insulin promote the onset of diabetes. Diabetes mellitus is the primary reason for large vessel disease and CAD because it reduces  prostacyclin production and promotes hyperlipidemia, and platelet hyper-aggregation (53, 54). Thus, Cr(III) deficiency results in type 2 diabetes mellitus and cardiovascular disorder (55, 56). Toxic effects of Cr (III) mainly occur only after its conversion to  Cr(VI)  (7, 57) which will be discussed below. 

2.2. Toxicity of Cr(VI) on heart.

     The environmental contamination of Cr(VI) is a greater risk for human health and it can cause various disorders including liver disease (58, 59), chronic lung diseases (60), skin ulcers (61), allergic contact dermatitis (62, 63), kidney disease (64), immunological disorder (65), etc. The cardiovascular system is highly susceptible to any oxidant. Therefore. the most common disorder caused by Cr(VI) is CVD. 

2.2.1. Cr(VI) metabolism.

     Cr(VI) is a strong oxidizing agent and highly soluble in water (66). By reacting with molecular oxygen (O2), it can form two different predominant species in different pH conditions, including chromate (CrO42-) (basic) and dichromate (CrO72-) (acidic) (7). After entering the body, Cr(VI) is rapidly absorbed in the gastrointestinal tract (GI tract) and then taken up by the cells of different tissues (67). In the form of the tetrahedral divalent, (CrO42-), it can cross the plasma membrane via phosphate (68) and sulfate anionic carrier (69). In the intracellular environment, Cr(VI) is gradually reduced into its stable intermediate Cr(III) by utilizing both non-enzymatic reductants such as glutathione (GSH), ascorbate (Asc), and thioredoxin (70, 71), and enzymes including mitochondrial electron transport complex enzymes, microsomal cytochrome P450/nicotinamide-adenine dinucleotide phosphate (oxidized) (NADPH)-cytochrome P450 reductase (72, 73), glutathione reductase (GR) and nicotinamide-adenine dinucleotide phosphate (reduced) (NADP+) oxidoreductase (13, 74). Likewise, extracellular conversion of Cr(VI) to Cr(III) has been reported in saliva, gastric juice (33), and intestinal bacteria (75). Small portion of Cr(III) can enter the intracellular environment through phagocytosis (76) while majority of Cr(III) is incapable of crossing the membrane barrier; therefore, it is trapped in the place where it is converted by Cr(VI) (33). 

2.2.2. Effects of Cr(VI)-induced oxidative stress on CVD.

     CVD is the outcome of complex heterogeneous pathophysiology where oxidative stress has been defined as a common etiology (77). In the intracellular environment, mitochondria and other organelles, including peroxisome and endoplasmic reticulum are considered as the main sources of ROS formation which contributes to ischemia-reperfusion injury (78). Apart from that, several enzymes such as xanthine oxidase, lipoxygenases, cyclooxygenases, nitric oxide synthases, and cytochrome P450 also generate  ROS (79). In cardiomyocytes, a transmembrane enzyme, NADPH oxidase (NOX family enzyme), is attributed to produce significant amount of ROS and reactive nitrogen species (RNS) which are associated with heart failure (80, 81). NOX4, which is the predominant isoform of NADPH oxidase expressed primarily in mitochondria of cardiomyocytes, stimulates O2.- production and promotes apoptosis and mitochondrial dysfunction through mitochondrial swelling, cytochrome C release, decreasing mitochondrial DNA and aconitase activity (82–84).

     Cr(VI) is classified as a redox-active metal because it directly undergoes redox cycling to produce a large amount of ROS (85). It facilitates HO., formation by the Fenton and Haber-Weiss reactions (33, 74). ROS attacks the cellular lipid and protein resulting in membrane injury (86–88). Likewise, intracellular and extracellular accumulated Crs modulates the morphological structures of cell surface by disrupting the lipid-protein networks of membranes, leading to loss of cellular integrity (76) and the leakage of cellular content such as intracellular enzymes as well as the incorporation of the viral component into the cardiac sarcolemma and ultimately leads to cardiac myopathies (89). Cr(VI) aggravates LDL oxidation by binding with the tryptophan residue of LDL particles to generate tryptophan radicals in the intracellular lysosomal compartment leading to LDL aggregation. These aggregated LDL particles precipitated and retained in the subcellular compartment and tissue, resulting in lysosomal dysfunction, cell damage, vascular injury, and atherogenic processes, which confers the progression of atherosclerosis (48). As a result, LDL is repeatedly oxidized, and about 90% of lipoproteins are re-entering the circulation by passing through the vascular wall, contributing to the systematic lipo-peroxide load (90). The study exhibits that NADPH-dependent flavoenzymes have chromate-reductase activity by binding with an adjacent cysteine residue (cys46 and 41) of glutathione reductase and also cross-linking with two peptide fragments (tyrosine 67 and asparagine 52) of cytochrome-c to involve in chromate reduction and finally binds with Cr(III) (73). Therefore, it may bind with Cr(VI) and further stimulate the expression of NADPH oxidase (91). 

2.2.3. Impacts of Cr(VI) on endothelial dysfunction and cardiovascular diseases.

     Endothelium dysfunction is a potential cause of CVD. The endothelium is a highly active monolayer that maintains vascular tone, cell proliferation and angiogenesis, coagulation, cell adhesion, and anti-inflammatory properties by releasing endothelium-derived contracting factor (EDCF) and endothelium-derived relaxing factor (EDRF) (92, 93). Cardiovascular complications are mainly associated with the imbalance of EDRF that results in heart failure, diabetes, atherosclerosis, and hypertension (94). Alterations in the EDRF lower the activity of endothelial nitric oxide synthase (eNOS) that generates nitric oxide (NO), a vasoactive compound (95, 96). NO contributes to regulation of non-enzymatic lipid oxidation, production of inflammatory and vasoactive eicosanoids, modulation of macrophage migration, LDL oxidation, suppression of vascular lesions and free radical detoxification (97). NO is also a vascular relaxation molecule, while under oxidative stress, NO rapidly reacts with O2.- to form peroxynitrite (ONOO-) at rapid rate (98). A reduction in the bioavailability of NO results in vasoconstriction, thereby increasing the risk of hypertension (99). On the other hand, ONOO- causes LPO formation with a massive amount of H2O2 production. This process hampers the energy metabolism by inhibiting the mitochondrial electron transport chain (ETC) as well as Kreb's cycle enzyme aconitase. The outcome is the development of atherosclerosis (100–103). Interestingly, hypercholesterolemia also impairs endothelial-dependent vasodilatation in large conductance vessels, micro vessels, and peripheral circulation (104–106). Components of oxidized LDL such as lys phosphatidylcholine accumulate in the vascular wall and stimulate the production of protein kinase C and O2.- which can cause atherosclerotic lesions (107, 108). Kojda (96) reports that hypercholesterolemia and CAD together are associated with the imbalance of vascular production of NO and which in turn generates other toxic radicals, including H2O2 and ONOO- further promoting the progression of other CVDs (Figure 1.).

Being a redox-active transition metal, Cr(VI) increases the production of  which reacts with NO rapidly to produce ONOO- to enhance the endothelial cell expression. This escalates the intracellular adhesion molecule (ICAM) and adhesive properties of the endothelium (109), leading to endothelium dysfunction and tissue injury. Like other enzymatic and non-enzymatic reductive actions, NOS, by one-step reduction, can reduce Cr(VI) to Cr (V). Cr(V) is further reduced by O2.- to produce the cytotoxic effect (110). On the contrary, Kushwaha et al. (111) claim that Cr(VI) reduces endogenous NO production by inhibiting nitric oxide synthase activity and stimulates ROS production through downregulating the ascorbate-glutathione cycle and glutathione biosynthesis in plant tissue. Although there are no such comprehensible pieces of evidence found on Cr(VI) mediated endothelial dysfunction of heart tissue; thus, essentially further studies need to be done in the future for a clear understanding. 

2.2.4. Effects of Cr(VI) induced calcium (Ca2+) overload on cardiovascular system.

Although ROS can act as an intracellular secondary messenger, excessive ROS induces hypertension due to the vasoconstriction caused by intracellular Ca2+ overload (112). In turn, Ca2+ overload further increases the ROS generation to form a vicious cycle (113). Ca2+ overload also occurs under several other conditions including (1) the increased concentration of sodium in the intracellular environment (114, 115), (2) the elevated membrane permeability and impaired protein function with the accumulation of long-chain fatty acids in the cardiac membrane (116, 117), (3) lack of adenosine triphosphate (ATP) in the cellular environment during ischemia (118), (4) the ischemia-reperfusion associated adrenergic stimulation (119). Intracellular Ca2+ overload causes cellular injury either by interfering with the Ca2+ handling proteins or damaging the membrane lipids (120). ONOOalso causes pronounced injury to the cardiomyocytes by impairing plasma membrane and contractile protein through unconstrained calcium influx (121).

     Cr(VI) mediated excessive ROS generation can cause mitochondrial and other organelle membrane damage, resulting in cellular Ca2+ overload due to Ca2+ influx from the calcium stores (113). Cr(VI) also increases the Ca2+ by inhibiting the activities of Ca2+- magnesium (Mg2+) -ATPase, and sodium (Na+)- potassium (K+) -ATPase. Ca2+- Mg2+ ATPase is present in the plasma membrane, which plays a crucial role in calcium transportation to maintain intracellular calcium homeostasis (122, 123). Whereas Na+-K+-ATPase is essential for the uptake of K+, that is essential to maintaining the osmotic pressure inside and outside of the cell and regulating the cell volume (124). Calmodulin (CaM) is a calcium-binding protein; calcium-mediated cellular functions are synchronized by binding with CaM (125). The surge in Ca2+ concentration simultaneously increases the CaM consumption and reduces the expression of CaM mRNA and protein as well as the Ca2+-ATPase activity. Reduced expression of CaM fails to maintain intracellular calcium homeostasis. Eventually, Ca2+ overload causes the release of apoptotic factors from mitochondria leading to DNA damage,  apoptosis, and necrosis (126, 127) (Figure 1.).

 

Figure 1.jpg


Fig, 1.  A mode of Cr-induced oxidative stress, Ca2+ overload, and endothelial dysfunction.

     (Cr)- Chromium, (Ca)- Calcium, (CaM)- Calmodulin, (H2O2)- Hydrogen Peroxide, (HO.)- hydroxyl radical, (- Superoxide Radical, (NO)- Nitric Oxide, (ONOO.)- Peroxynitrite, (ROS)- Reactive Oxygen Species, (Mg)- Magnesium, (K)- Potassium, (Ox- LDL)- Oxidized Low-Density Lipoprotein, (ATP)- Adenosine- Tri- Phosphate.


3. ROLES OF Cr(VI) ON SIGNAL TRANSDUCTION PATHWAYS INVOLVED IN CARDIAC DISEASES

     In cells, various signals are involved in the expressions of genes that play crucial roles in ROS defense, cellular metabolism, mitochondrial biogenesis, anti-inflammation, and apoptosis. These include but not limited to the silent information regulators of transcription-1 (SIRT 1), forkhead box transcription factor (FOXO), AKT, and peroxisome proliferator-activated receptor-gamma (PPAR-ϒ) coactivator 1-alpha (PGC-1α) (59). The details will be discussed below. 

3.1. Effects of Cr(VI) on expression of SIRTUINS, and FOXOs in the progression of cardiovascular diseases.

     Among the mitochondrial biogenesis regulators, SIRTs are nicotinamide adenine dinucleotide (NAD+) dependent histone deacetylases that serve different functions by located in separate subcellular compartments including cytoplasm (SIRT 1, 2), mitochondria (SIRT 3, 4 and 5), and nucleus (SIRT 1, 2, 6 and 7) (128). Activities of them are highly dependent on NAD+ (oxidized) and NADH (reduced). SIRTs are primarily involved in various physiological processes such as energy metabolism, stress resistance, mitochondrial function, DNA damage repair, apoptosis, and inflammation (129, 130). Cr(VI) inhibits the expression of SIRT 1, resulting in the initiation of apoptosis (131). Furthermore, Cr(VI) aggravates cellular apoptosis in rat hepatocytes by inhibiting the deacetylation of SIRT 1, as reported by Yang et al. (132). Suppression of SIRT 1 by Cr (VI) may also hamper the energy metabolism, reduce stress defense mechanism, and induce inflammation. Thus, it inhibits the activities of rate-limiting enzymes of the tricarboxylic acid cycle (TCA cycle) [isocitrate dehydrogenase (ICDH), succinate dehydrogenase (SDH), and pyruvate dehydrogenase (PDH)], enzymes involved in oxidative phosphorylation (NADH dehydrogenase) and enzymes of gluconeogenic pathway (133).

     All SIRTs are equally essential, but SIRT 1, 2 and 3 are well-known cardiac isoforms that protect the heart from oxidative damage by upregulating the FOXO4, FOXO1, and FOXO3a respectively (134). Increased expression of FOXOs enhances the intracellular antioxidant capacity by modulating Mn-SOD activity (135–137). It has been reported that hindrance of deacetylation of SIRT 1 after the Cr(VI) exposure causes inhibition of FOXO1 expression in hepatic tissue resulting in decreasing activity of Mn-SOD, leading to lower stress resistance and increased tissue damage (132). However, SIRT 1 and SIRT 2 also enhance the protein expression of other antioxidant enzymes, including catalase (CAT), glutathione peroxidase (GPx), and thioredoxin (Trx), to protect against oxidative injuries (129, 138, 139). The Cr(VI)  diminished the antioxidant enzyme activities [including SOD, glutathione-S-transferase (GST), CAT, GPx, GR, and thioredoxin reductase (TR) due to increased ROS generation (140, 141) or SIRT 1 inhibition. But at low concentration, Cr(VI) tends to increase the activity of both SOD1 and SOD2, respectively (59). SIRT 2 and SIRT 3 boost the production of NADPH which in turn increases the conversion of oxidized glutathione (GSSG) to GSH to mitigate the oxidative stress. For example, SIRT 2 deacetylates glucose-6-phosphate dehydrogenase (G6PDH) to produce NADPH (142). There is a complex mechanism involved in Cr(VI) redox status and SIRT 2 and 3 inhibition. Cr(VI) can be reduced to its intermediates by consuming GSH (71), NADPH (143), and NADPH-linked enzymes, including glucose-6-phosphate (G6P) (72), to reduce their availability in the intracellular environment. Therefore, inhibition of SIRT 2 and SIRT 3 by Cr(VI) also indirectly reduces the production of both enzymatic and non-enzymatic antioxidants, resulting in the diminution of stress resistance and increased cellular damage. Similar to  SIRT 1, SIRT 3 also activates TCA cycle enzymes, including succinate dehydrogenase (SDH) (144) and isocitrate dehydrogenase 2 (ICDH2) (145), and glutamate dehydrogenase (146). SIRT 3, therefore, plays a vital role in the deacetylation of mitochondrial proteins to maintain metabolic homeostasis (147). Mitochondrial function is crucial for cancer cell proliferation and survival (148). SIRT 3 regulates oxidative phosphorylation (OXPHOS) via deacetylating complexes I and III to maintain standard ROS generation (149). Thus, the inhibition of SIRT 3 activity by Cr(VI) alters the mitochondrial membrane potential (MMP) and ATP production (150). 

3.2. Modulatory effects of Cr(VI) on the expression of AMPK and PGC-1α in the progression of cardiovascular diseases.

     On the other hand, adenosine monophosphate-activated protein kinase (AMPK) is found primarily on lysosomes and also distributed in the golgi apparatus, endoplasmic reticulum, and mitochondria (151). Activation of AMPK depends upon the cellular AMP/ATP ratio. Hence, defects in energy production and enhanced energy consumption activate AMPK (152). During energy stress, AMPK regulates mitochondrial gene expression and enhances the gene expression related to glucose metabolism (153), mitochondrial respiration (154) and lipid synthesis (155). Cr(VI) inhibits the formation of neutrophil extracellular traps (NETs) via AMPK signaling, which primarily controls the extracellular killing mechanism that can cause neutrophil toxicity and apoptosis. Cr(VI)-induced NET inhibition is further associated with the down-regulation of protein expression of myeloperoxidase (MPO)/Histone-3 (H3) and the decreased NET is accompanied by the reduced DNA levels in neutrophils both intracellular and extracellularly (156). However, Cr(VI) also inhibits the activation of the AMPK-mediated Nrf2 pathway to induce excessive ROS generation (156). A pathway of AMPK/FOXO3a is a crucial regulator of cellular metabolism. SIRT 6 protects the cardiomyocytes via activating the AMPK/FOXO3a  pathway (157). This pathway is activated during metabolic stress, which primarily induces autophagy to preserve cell survival by retaining the energy, but under persistent stress, it provokes autophagic cell death (158). It has been reported that Cr(VI)-induced autophagy via suppressing the AMPK/FOXO3a associated mTOR signaling in rat hepatocytes (123). mTOR is important to regulate cell growth and cellular homeostasis and control cell proliferation by modulating the expression of transcription factor FOXO3a (159). SIRT 1 and AMPK are two primary metabolic sensors that act as the gatekeepers and regulators of mitochondrial biogenesis and respiration by activating PGC-1α (160). Transcription of many genes related to fatty acid metabolism (161) and the mitochondrial metabolism are under the regulation of PGC-1α network (162). PGC-1α activation is primarily dependent on the expressions of SIRT1 and AMPK, which upregulates its activity through phosphorylation (163), acetylation (164,165), and methylation (166). Cr(VI) is known to deactivate the pathway of SIRT1/PGC-1α/Nrf2 (131), which may promote ROS production, reduce activities of antioxidant enzymes, inhibit mitochondrial biogenesis, and cause metabolic disturbances.

3.3. Insights of Cr(VI) on P53 signaling pathway in the progression of cardiac diseases.

     Being a tumor suppressor, P53 is also a vital regulator of the cell cycle and senescence (167, 168). Activation of p53 decreases intracellular ROS by increasing antioxidant levels (169). Acetylation of p53 results in excessive ROS generation, oxidative DNA damage and upregulation of growth-suppressive genes (170). Therefore, mutation of P53 are highly associated with metastatic spread and activation of oncogenes (171). Cr(VI) induced oxidative stress promotes the acetylation and activation of P53, which in turn increases the expression of apoptotic protein Bcl2 Associated X (Bax) (13, 172) and alters the activity of anti-apoptotic proteins such as B Cell Lymphoma-2 (Bcl-2) and B Cell Lymphoma extra-large (Bcl-XL) (173). It has been reported that Cr(VI)-induced P53 activation uses hydroxyl radical as a messenger (174) which was generated by Fenton or Haber-Weiss reaction (33). This p53 activation also activates apoptotic machinery by releasing cytochrome c into the cytosol (175). The accumulation of cytochrome c in the cytosol activates caspase-3 (176) which helps in sustaining cellular homeostasis by regulating cell death and inflammation. Accordingly, caspases are broadly classified into two groups that are important for apoptosis (caspase 3, 6, 7, 8, and 9 in mammals) and inflammation (caspase 1, 4, 5, 12 in humans) (177). Activating apoptotic caspase cascade by releasing caspase-3 to the intracellular environment results in cardiomyocyte damage, leading to heart failure (178, 179). Thus, activating apoptotic signaling via P53 and releasing caspase3 by  Cr(VI) causes cardiomyocyte injury (174, 180). On the other hand, both SIRT1 and SIRT7 stimulate the deacetylation of P53, thereby reducing cellular senescence and apoptotic cell deaths (181). Thus, inhibition of SIRT1 to increase the acetylation of P53 and P53-upregulated modulator of apoptosis (PUMA) aggravates cellular apoptosis. Inhibition of SIRT1 also upregulates expression of Bax and cleavage of caspase-3, whereas it downregulates the Bcl-2 and Bcl-XL expression (182).

3.4. Consequences of Cr(VI) mediated Akt and NF-κβ signaling pathway in the progression of cardiovascular diseases.

     In mammals, cellular stress activates various apoptotic pathways in mitochondria. SIRT3 inhibits mitochondrial ROS production and suppresses the ROS-induced Ras-P13K-Akt pathway (137). Cr(VI) can stimulate renin-angiotensin system (Ras)  to produce  O2.- by using molecular oxygen and NADPH oxidase and this is responsible for further signal transduction from Ras to nucleus leading to activation of various apoptotic signals (183). Via activation of Ras, Cr(VI) causes endoplasmic reticulum (ER) stress,  disrupted redox state and carcinogenesis  (184). Protein kinase B (PKB), also known as Akt, is recognized for its anti-apoptotic activity (185). It promotes cell survival by regulating transcription factor nuclear factor kappa β (NF-κβ) and transcription of pro-survival genes (186) and inhibits pro-apoptotic family members of Bcl2, including BAD and BAX (187). But under stressful conditions, Ras depended PI3K activation is associated with pro-survival signaling due to the involvement of Akt. Activated PI3K or Akt revokes apoptosis, but the dominant-negative Akt [(BAX and Bcl2 Associated Agonist of Cell Death (BAD)] can enhances the apoptosis by inhibiting the activation of anti-apoptotic proteins Bcl2 and Bcl-XL (188, 189). Apart from that, another protein called insulin-like growth factor (IGF-1) also controls the P13K/Akt pathway to induce cardiac hypertrophy, which causes the progression of heart failure (190). Chronic exposure to Cr(VI) causes activation of PI3K/Akt depended signaling cascades, i.e., PI3K/AKT/GSK-3β/β-catenin and PI3K/AKT/mTOR to promote carcinogenesis (191). On the contrary, Zhang et al(192, 193) reported that long-term but low-dose exposure to Cr(VI) could inhibit the PI3K/Akt pathway, which can cause premature senescence to L-02 hepatocytes. Cr(VI) increases the phosphorylation of Akt to stimulate the Akt-dependent phosphorylation of the ERK/AMPK pathway, in which ROS initiates mitochondrial-depended apoptosis (194). Dysfunction of mitochondrial biogenesis caused by Cr(VI) also aggravated apoptosis by downregulating the Akt/STAT3 pathway (195). Members of the signal transducer and activator of transcription (STAT) protein family, especially STAT3, are closely associated with cell survival, tumor appearance, and development (196). STAT3 can inhibit tumor cell apoptosis both directly and indirectly by enhancing the expression of Bcl-2 and Bcl-XL, other anti-apoptotic proteins, and interleukins (ILs) such as IL-6 IL-10, IL-11, etc. (197). Despite that, it is also essential for regulating the activity of mitochondrial respiratory chain enzymes, thus, maintaining the production of ATP and reducing the autophagy of cardiomyocytes (198). However, it has also been reported that Cr(VI) can induce autophagy in L-02 hepatocytes, resulting in autophagosome accumulation. Inhibition of the Akt/mTOR pathway is the primary reason behind the activation of autophagy, as described by the authors. Cr(VI) downregulates the protein expression of the critical mediators, including PI3K, AKT, and mTOR, and decreases the phosphorylation of Akt (199). During hypoxia, excessive carbon dioxide retention leads to autophagy of brain neurons due to phosphorylation of PI3K and inhibition of the Akt/mTOR pathway (200). Liang et al. (199) claimed that “Cr(VI)-induced autophagy protects L-02 hepatocytes from apoptosis through the ROS/Akt/mTOR pathway”.

     Conversely, NF-κβ is a versatile protein that simultaneously controls both the inflammatory response and apoptosis. NF-κβ generally regulates the transcription of anti-apoptotic protein, promoting the proliferation and tumor growth (201). The elevated ROS generated by Cr(VI) reduction upregulates the activity of NF-κβ (202) that finally resulting in the development of carcinogenesis (203). Other than excessive ROS production, the accumulation of intracellular Ca2+ induced by Cr(VI) also exacerbates the activity of NF-κβ,  thereby leading to premature senescence through the ROS/Ca2+/ NF-κβ pathway (204). AP-1 is a dimeric transcription factor consisting of different protein family members such as JUN, FOS, ATF, and MAF. Its activity primarily depends on transcription, post-transitional modification, and interaction with other proteins. AP-1 can act as both oncogenic and anti-oncogenic because it regulates the closely related genes to cell proliferation, differentiation, apoptosis, angiogenesis, and tumor invasion (205). After Cr(VI) exposure, activations of NF-κβ and AP-1  trigger the expression of genes responsible for inflammation and apoptosis (206). Thus, Cr(VI) can alter all the apoptosis and autophagy signalling pathways to cause cell death, resulting in carcinogenesis and heart failure. 

3.5. Effects of Cr(VI) induced ER stress signaling pathway on the progression of cardiovascular diseases.

     The endoplasmic reticulum (ER) is the vital cellular organelle where protein folding occurs. Stressful conditions jeopardize the internal environment of the ER and also impair protein maturation resulting in the assembling of unfolded protein and starting a characteristic stress response known as unfolded protein response (UPR) (207). Although, disturbances in the ER also lead to oxidative stress, disruption of calcium homeostasis, and aggregation of unfolded protein can cause overexpression of both normal and abnormal folded protein (208). Evidence shows that Cr(VI) induced ER stress leads to autophagy and apoptosis (209). ER stress-mediated signal transduction pathway minimizes the accumulation of unfolded proteins by enhancing ER native chaperones, inhibiting protein translation, and accelerating the degradation of misfolded proteins through initiating the UPR. Prolonged ER stress accelerates apoptosis and causes cell death via both mitochondria-dependent and independent pathways (208, 210). Studies have also indicated that Cr(VI) can cause mitochondrial damage by persuading ER stress (211) and prolonged UPR provokes apoptosis (207). All these alterations mentioned on chapter 3 are illustrated in Figure 2.

Figure 2.jpg

Fig. 2. Effects of Cr(VI)-induced oxidative stress on cellular signal transduction pathways.

     (AMPK)- Adenosine Monophosphate-Activated Protein Kinase, (AP-1)– Activator Protein-1, (BAX)- Bcl2 Associated X, (Bcl-2)- B Cell Lymphoma-2, (Bcl-XL)- B Cell Lymphoma Extra Large, (CAT)- Catalase, (ERK)- Extracellular Signal Regulated Kinase, (FOXO)- Forkhead Box Transcription Factor, (GPx)- Glutathione Peroxidase, (GR)- Glutathione Reductase, (GST)- Glutathione-S-Transferase, (NF-κβ)- Nuclear Factor-Kappa β, (PGC-1α)- Peroxisome Proliferator-Activated Receptor-Gamma (PPAR-ϒ) Coactivator 1-Alpha, (PI3K)- Phosphatidylinositol-3-Phosphate, (PKB)- Protein Kinase B, (PUMA)- P53-Upregulated Modulator Of Apoptosis, (RAS)- Renin-Angiotensin System, (SIRT)- Silent Information Regulators Of Transcription, (SOD)- Superoxide Dismutase, (STAT)- Signal Transducer And Activator Of Transcription, (TRx)- Thioredoxin.

 

4. EFFECTS OF Cr(VI) INDUCED INFLAMMATION ON PREDISPOSITION OF CARDIOVASCULAR DISEASES

     The immune system triggers inflammation in response to environmental stimuli such as pathogens, irradiation, and other toxic compounds. In response to tissue injury, the body triggers various signaling cascades at the cellular level to initiate the repairing and healing processes. Pro-inflammatory and anti-inflammatory mechanisms are primarily dependent on stressors (212). For example, everything related to cardiovascular complications, from endothelial cell dysfunction, plaque formation, progression, and rupture to architectural instability, depends on the cellular inflammatory response driven by the cytokines and interleukins (213). 

4.1. Effects of interleukins and tumor necrosis factor α (TNFα) in Cr(VI) mediated inflammation on the cardiac tissues.

     Cr(VI) exposure results in a wide range of immunological responses depending on the dose, time, routes of exposure, and valency and type of chromium compounds (214). Tissue inflammation primarily leads to the release of pro-inflammatory cytokines from monocytes/macrophages, vascular endothelial cells, fibroblasts, etc. Pro-inflammatory cytokines trigger the inflammation reaction by up-regulating the expression of C-reactive protein (CRP), IL-6, TNFα, IL-1β, and NF-ԟB (215, 216), and interferon-gamma (IFN-ƴ) (217). Evidence demonstrates that Cr(VI) can significantly increase inflammatory cytokines, including IL-6, TNF-α, and IFN-ƴ (218). However,  a negative association was found between the Cr(VI)  and CRP levels in blood (219,220). A 2-hour Cr(VI) exposure can significantly increase chemokine IL-8 homolog and cytokine IL-6 (60). In addition, Cr(VI) enhances the mRNA expression of toll like receptor 2/4 (TLR2/4), which subsequently elevates the ROS levels to trigger the inflammation process by increasing the TNFα and IL-1β mRNA levels in a concentration-dependent manner in hepatocytes (59). Cr(VI) dose-dependently inhibits certain interleukins including  IL-2, IL-4, and IL-10 by  48%, 57%, and 35% at the concentration of 2μM and 65%, 81%, and 51% at 5μM, respectively (221). 

4.2. Impacts of T and B lymphocytes in Cr(VI) mediated inflammation on the cardiac tissues.

     Interleukins are classified as heterogeneous cytokines involved in activating B and T lymphocytes and macrophages. T cell is the major component of host anti-tumor immunity (222). T cells require activation and clonal expansion of both CD4+ and CD8+ and trigger the immune response to destroy tumor cells (222). Cr(VI) inhibits the proliferation and activation of CD4+ and CD8+ T cells and significantly decreases their viability at the tumor site, resulting in impairment of cytolytic function for the tumors (221). A subsequent increase in CD4+ (helper T cell) and CD8+ (toxic T cell) T lymphocytes have also been found in response to Cr(VI) induced lung injury in the early exposure (60). CD4+ tumor T (Th) cells primarily participate in tumor immunology and are divided into subgroups according to their immunological functions, such as Th1 and Th2 cytokines. Th1 cytokines are generally related to tumor-suppressive activity, including IL-1β, IL-2, IL-12, TNF-α, and IFN-γ, whereas, Th2 cytokines such as IL-4, IL-5, and IL-10 are associated with tumor growth and metastasis (223). Cr(VI) can alter the immune response and may induce a hypersensitivity reaction by stimulating or suppressing the expression of T and B lymphocytes, macrophages, and cytokine production (214, 224). 

4.3. Influences of neutrophils and macrophages in Cr(VI) mediated inflammation on the cardiac tissues.

     Cr(VI) causes a 2-fold increase of macrophages by replacing the neutrophils content at the site of tissue injury (60). Neutrophils and macrophages are involved in phagocytosis to engulf the foreign particles and amplify the inflammatory response through cytokine production (225, 226). In addition, Cr(VI) inhibits the formation of neutrophil extracellular traps (NET), which is responsible for innate immune response, and its release is considered to be a vital part of the extracellular killing mechanism that promotes neutrophil apoptosis (156). Therefore, lower doses of Cr(VI) stimulate the process of phagocytosis and increase the humoral immune response. In contrast, higher doses of Cr(VI) depress the phagocytic activity of alveolar macrophages and the humoral immune response (227). 

4.4. Effects of Cr(VI) induced  IFN- ƴ  and inflammation on the myocardial tissues.

     IFN-ƴ is a cytokine primarily secreted from the activated T cells that can activate macrophages, stimulate antigen production, and mediate antiviral and antibacterial activity (228). A positive correlation between the Cr(VI) content of the blood and IFN-ƴ production has been reported (229). IFN-ƴ upregulates the expression of other Th1 cytokines such as TNFα and IL-12, reduce macrophage polarization, inflammation (230) and the activity of Th2 cytokines of IL-4 and IL-5, to reduce atherosclerotic lesions (231). The Cr(VI) induced IFN-ƴ release may activate Th1 cytokines and macrophages to promote microbicidal activity and upregulate the class II MHC (major histocompatibility complex) to maintain the host defense system primarily (229). On the contrary, other studies have reported that the enhanced IFN-ƴ causes severe cardiovascular complications. IFN-ƴ inhibits the production of smooth muscle cells (SMCs) in collagen that weaken the fibrous structure of plaque and make it vulnerable to rupture, which is responsible for developing a thrombotic complication (232). SMCs protect an integral part of the plaque by forming a thick fibrotic layer to stabilize it, preventing the plaque rupture  (233). 

4.5. Effects of cyclooxygenase-2 (COX2) in Cr(VI) mediated inflammation on the cardiac tissues.

     COX2, also known as prostaglandin H synthase 2 (PGH2), plays a crucial role in arachidonic acid metabolism (234). It is expressed in several cell types, including monocytes/macrophages, vascular endothelial cells, and colorectal cancer cells, in response to inflammatory cytokines and growth factors (235, 236). COX-2 derivatives such as PGE2 and prostacyclin maintain blood pressure, endothelial thromboresistance, pain, and inflammation. Inhibition of COX-2 in the vascular endothelial, SMC, and cardiomyocytes cause a decrease in PGI2 and PGE2 activities associated with the reduction of NO and endothelial dysfunction (235). This can ultimately lead to increased arterial blood pressure, risk of atherothrombosis, rupture of coronary plaque, myocardial infarction, arrhythmias, and stroke (237).

     Data shows that Cr(VI) induces autophagy by activating ER stress through enhancing COX-2 activity (238). Previous studies indicate that the initiation of ER stress triggers autophagy (239). Autophagy is a dynamic process involved with autophagosome and lysosomal degradation (240) through initiating the catabolic process to break down the damaged organelles and large protein aggregates (241). The controversy is also present on the actions of COX-2 and Cr(VI) in heart tissue. COX-2 may have a protective effect on the heart since its inhibition also results in inflammation of the heart tissue. In contrast, heavy metals, especially Cr(VI), cause overexpression of COX-2, which usually increases the inflammation in tissues and cell lines. It is still the argument whether  COX-2 is a perpetrator or protector (242). Therefore, whether COX-2 expression in heart tissue after Cr(VI) exposure is beneficial or detrimental remains to be clarified. Judging the data from other heavy metals, such as cadmium,  the increased activity of COX2 will enhances the NADPH oxidase activity and reduces the bio-availability of NO. these events can cause endothelial dysfunction and the establishment of atherosclerosis along with hypertension (243).

4.6. Effects of NF-ԟB in Cr(VI) mediated inflammation on the cardiac tissues.

     The transcriptional factor NF-ԟB plays a pivotal role in maintaining inflammatory response by modulation of innate and adaptive immunity. NF-ԟB regulates the expression of different pro-inflammatory genes and participates in inflammasome regulation. All these actions of  NF-ԟB are made by upregulation of cytokines and chemokines that critically control activation, differentiation, and survival of innate immune cells and inflammatory T cells (244). Cr(VI) markedly increases the activity of NF-ԟB in rat hepatic tissue, which initiates inflammatory cascades by releasing a wide variety of pro-inflammatory cytokines (245). NF-ԟB appears to be cardioprotective during hypoxia and reperfusion injury. However, long-term activation of NF-ԟB promotes heart failure via ER stress-mediated cell death and chronic inflammation by enhancing the expression of specific cytokines including  TNF-α, IL-1, and IL-6 (246). The actions of Cr(VI) induced inflammation on predisposition of cardiovascular diseases are summarized in the Figure 3.

Figure 3.jpg

Fig. 3. Actions of Cr(VI)-induced inflammation on the chronic and acute stressed cardiomyocytes.

     (COX-2)- Cyclooxygenase-2, (IFN-Ƴ)- Interferon Gamma, (NF-κβ)- Nuclear Factor-Kappa β, (TLR)- Toll-Like Receptor, (TNF-α)- Tumor Necrosis Factor Α, (ER stress)- Endoplasmic Reticulum Stress, (IL)- Interleukin, (MHC)- Major Histocompatibility Complex.

 

5. EFFECTS OF MELATONIN ON Cr(VI) CAUSED CVD

     As discussed above, Cr(VI) is toxic to the heart by targeting various intracellular and extracellular molecules that are highly associated with cardiac health. Therefore, to minimize the death rate of Cr(VI) induced cardiac diseases, the introduction of melatonin should be necessary judging from its cardioprotective property. To identify an antioxidant with no or less pro-oxidant activity and side effects is challenging. Melatonin seems to be one of the unique antioxidants among others (including GSH, vitamin C, Vitamin- E, etc.). The protective effects of melatonin against Cr(VI) toxicities with its distinctive and versatile properties in heart are discussed below. 

5.1. Evidence of melatonin as a cardioprotective molecule.

     Melatonin (N-acetyl-5-methoxytryptamine) is a neuroendocrine hormone synthesized from the pineal gland to maintain circadian rhythm, but its original function serves as a potent antioxidant. Other functions of melatonin are acquired during evaluation (247). It is present in animals, plants, and bacteria (248). Melatonin exhibits antioxidant, anti-inflammatory, and anti-cancer activity. It can directly scavenge ROS and RNS and also can stimulate the gene expression of antioxidant enzymes and inhibit the actions of pro-oxidant enzymes(249). Being a nutraceutical substance (250), melatonin chelates metals convincingly up to 95% and reduces metal-catalyzed molecular damage (251). Melatonin synthetic enzymes are found in almost all organs and tissues, including the brain, retina, lens, cochlea, Harderian gland, airway epithelium, skin, gastrointestinal tract, liver, kidney, thyroid, pancreas, thymus, spleen, immune system cells, carotid body, reproductive tract, and endothelial cells. Melatonin also presents in all biological fluids, including saliva, cerebrospinal fluid, bile, synovial fluid, amniotic fluid, and breast milk (252). 

     The protective actions of melatonin on CVDs including myocardial ischemia-reperfusion injury (MIRI), myocardial hypoxia-reoxygenation injury, pulmonary hypertension, hypertension, atherosclerosis, valvular heart diseases, and other CVDs have been documented (253). In a clinical study, melatonin treatment reduced platelet aggregation, pulsatility index in the internal carotid artery, and catecholamine level with the improved systolic and diastolic blood pressure and the low melatonin levels in the blood are associated with the predisposition of CCD, arterial hypertension, and congestive heart failure (254). The epidemiological evidence showed that the sudden incidents of cardiac events such as MI, sudden cardiac death, and cardiac arrhythmias generally occur in the early morning when the circulating levels of melatonin are low in the bloodstream (27). Additionally, melatonin is able to reduce blood pressure through the following mechanisms: 1) stimulating the hypothalamus; 2) lowering the blood pressure through its antioxidant activity, 3) minimizing the level of catecholamines and  4) dilating the smooth muscle in the aorta wall (255). 

5.2. Effects of melatonin’s receptor and non-receptor mediated activities on cardiovascular diseases.

     Melatonin acts through both the receptor- and non-receptor-mediated pathways. In the cardiovascular system (cardiomyocytes, left ventricle, and coronary arteries), melatonin can act on its membrane receptors (MT1 and MT2) (23). The vascular melatonin receptors are functionally linked to either vasocontractory or vasodilatory effects of melatonin (255). The MT1 receptor induces arterial vasoconstriction. Whereas activation of MT2 receptor mediates vasodilation (256). The receptor-independent actions of melatonin are attributed to its antioxidant and mitochondrial-protecting effects (257). Intracoronary-melatonin simultaneously amplifies the coronary blood flow to maintain normal cardiac functions through MT1 and MT2, β-adrenoreceptors, and NO release (258). It is reported that MT1 rather than MT2 efficiently ameliorate MI/R injury and improve cardiac dysfunction, which was accompanied by attenuation of oxidative stress, ER stress, and mitochondrial dysfunction (259). The activity of MT2 receptors is found to be altered in the sections of isolated coronary arteries, aorta, and left ventricular specimens in dilated and ischemic cardiomyopathy patients compared to the healthy heart donor (260). Mechanistically, melatonin stimulates the MT2/Notch1/Hes1/RORα signaling pathway to protect the primary cardiomyocytes against hypoxia/reoxygenation injury (259).

     Melatonin plays an important role in regulating blood pressure (BP) and reducing hypertension. Frank et. al. (261) has reported that bedtime melatonin intake lowers the systolic and diastolic BP by about 6- and 4-mm hg without affecting the heart rate in patient with essential hypertension. An increase in systolic and diastolic BP in day-night proportions is around 15 and 25 %, respectively when melatonin level is low. Melatonin supplementation reduces the nighttime blood pressure level significantly in nocturnal hypertensive patients (systolic blood pressure from 136+/-9 to 130+/-10 mm Hg and diastolic BP from 72+/-11 to 69+/-9 mm Hg) (262). The mechanism of action of melatonin to regulate BP involves both receptor and non-receptor mediated pathways (263, 264) including: a. Both MT1 and MT2 receptors are present in vascular smooth muscle cells as well as endothelial cells; activation of these receptors causes a decrease in cyclic AMP and phospatidylino-inositol-4,5-biphosphate hydrolysis resulting in vasodilation and vasoconstriction (265). b. Activation of the MT2 receptor on endothelial cells subsequently enhances the level of NO as well as lowers the cytosolic Ca2+ level leading to vasodilation (25,265). c. Melatonin ameliorates the sympathetic responses to reduce orthostatic stress (266). d. Interacting with CaM, it inhibits the Ca2+ channel and stimulates the Ca2+ pumping to protect the cardiomyocytes (265). e. Melatonin enhances coronary blood flow as well as cardiac contractility through NO release and β-adrenoreceptors (258). f. It modulates cardiac autonomic activity by maintaining circadian pacemaker (267).

5.3. Effects of melatonin on Cr(VI)-induced oxidative cardiovascular diseases.

     Unlike other classical antioxidants, melatonin does not undergo redox cycling; thus, it does not participate in  pro-oxidant activity (268). Melatonin can directly interact with a large number of ROS, including peroxyl radical (ROO.) (269), hypochlorous acid (HOCl) (270),  HO(271), H2Oand O2.- (272). Thus, melatonin can effectively inhibit the Cr(VI) mediated H2O2, O2.-, HOproduction either through scavenging activity or by chelating the metal (273). The cardiac membrane structures are prone to oxidative damage which is associated with various heart diseases (274), therefore, protecting the membrane structure is becoming the priority to reduce the death associated heart diseases. Melatonin is an amphiphilic molecule and it can easily cross the subcellular morphophysiological barrier and located between the polar and non-polar head group of membrane phospholipid to protect the membrane against both lipophilic and hydrophilic radicals emerging from the aqueous environment (275). Melatonin shields the cell membrane structures by directly detoxifying the lipo-peroxyl radicals (LOO.) (276) or indirectly by inhibiting the gene expression of lipo-oxygenase (277). Melatonin also blocks the site of membrane lipid to counteract LPO and prevents the rigidity of phospholipid bilayer (278, 279). Thus, it can maintain cellular integrity and attenuates the Cr(VI) induced membrane damage. Melatonin also modulates the expression of two dominant pro-oxidant enzymes of the heart, such as NOX2 and NOX4 (280). Melatonin pretreatment alleviates ROS overproduction and preserves cell viability by downregulating the Cr(VI)-induced NOX4 via activating the MT1 receptor (281).

     In addition, melatonin possesses anti-atherogenic activity. Melatonin inhibits LDL oxidation more efficiently than that of vitamin E (282). A study reported a limited protective effect of melatonin on LPO of LDL (283) while other study reported that melatonin reduced the susceptibility of LDL particles to oxidative modifications (284). Melatonin has the ability to suppress the vaso-spastic effect of ox-LDL (oxidized LDL, responsible for the development of atherosclerosis) since it is capable of neutralizing the HO. produced from the fractions of lipid (255). Accordingly, a high level of LDL was found in persons with low levels of blood melatonin indicating elevated levels of ox-LDL impaired melatonin synthesis at night (285). Chronic melatonin administration decreases total serum cholesterol and LDL while increasing the level of HDL in diet-induced hypercholesterolemic rats (286). It has been found that melatonin can suppress the formation of cholesterol and LDL receptor activity by 38% and 42%, respectively, in human mononuclear leukocytes (287). Therefore, it can assume that melatonin is effective in protecting against Cr(VI) induced oxidative cardiac damage. 

5.4. Effects of melatonin on Cr(VI) induced endothelial dysfunction.

     Cardiovascular diseases are highly associated with the influence of inflammatory cytokinesox-LDL, in damaged endothelium. Endothelial dysfunction is primarily caused the adhesion of molecules and the activation of leukocytes (288). Leukocytes adhere to the endothelium via adhesion molecules, including ICAM and vascular cell adhesion molecule (VCAM), leading to a cascade of events toward to endothelial dysfunction and damage (289). Melatonin ameliorates the ox-LDL-induced endothelial dysfunction by decreasing the ER stress and increasing the sustainability of ER homeostasis and mitochondrial function by inhibiting the JNK/Mff pathway. Furthermore, melatonin also reduces the aggregation of ICAM, VCAM, and leukocytes within the endothelial wall, thereby, protecting the endothelial cells against ox-LDL-induced endothelial dysfunction (290). Besides the antioxidant activity, melatonin and its metabolites N1-acetyl-5-methoxykynuramine (AMK) inhibit both inducible and neuronal NO synthases (291). Thus, it reduces NO formation, which diminishes NO interaction with O2.-, inhibiting the formation of ONOO-. Moreover, melatonin and AMK readily neutralize the ONOOand its carbon adducts including carbonate radicals and NO2 (291). On the contrary, melatonin increases NO bioavailability and protects the heart against endothelial damage, vasoconstriction, platelet aggregation, and leukocyte infiltration (292). Thus, melatonin may be able to reverse the Cr(VI) induced endothelial dysfunction. 

5.5. Effects of melatonin on Cr(VI) associated Ca2+ overload in myocardial tissues.

     Intracellular Ca2+overload is associated with hypertension and other cardiovascular complication in humans. Melatonin is a potent ROS scavenger; it curtails ROS production to reduce the Ca2+ cytosolic output (293). Melatonin abrogates the Ca2+ deposition in the vascular SMCs via activating mitochondrial fusion and mitophagy through the AMPK/OPA1 signaling pathway (294), protecting the mitochondrial membrane and diminishing the Ca2+ leakage. An increase in the Ca2+ overload induced by Cr(VI) causes inhibition of Ca2+ regulating ATPase signaling. Melatonin prevents the suppression of Na+-K+-ATPase, Ca2+- Mg2+ ATPase (295), thereby, maintaining the intracellular calcium homeostasis and osmotic pressure inside and outside the cell.

 

6. EFFECTS OF MELATONIN ON Cr(VI) PERSUED SIGNAL TRANSDUCTION PATHWAYS IN CVD

     Alterations in the intracellular redox status contribute to low melatonin levels in the intracellular environment and apoptosis (296). Melatonin alleviates several pathways involving apoptosis. SIRT 1 is one of them, overexpression and activation of SIRT 1 alleviate metabolic and cardiovascular complications. Melatonin treatment increases the activity of SIRT 1 and the expression of SIRT 2 and 3 in cells and tissues (297, 298). An increase in the SIRT 3 activity by melatonin subsequently increases the FOXO3a activity and transcription of SOD2 and CAT (299). Banerjee et al. (273) showed that the reduced activity of SOD, CAT, GPx, and GR caused by Cr (VI) was recovered by melatonin treatment at a dose-dependent manner. This may be attributed to the upregulated expression of proteins of SIRT1 and FOXOs by melatonin.  

     SIRT 1 also regulates cellular metabolic signaling through the acetylation and activation of the coactivator PGC-1α, which instigates mitochondrial transcription factors and promotes both mitochondrial biogenesis and the anti-oxidative capacity (300). An association between AMPK and PGC-1α also exists to regulate energy metabolism, fatty acid oxidation, and mitochondrial biogenesis (301). Therefore, dysfunction of this pathway results in metabolic disorders. Cr(VI) suppresses this SIRT1/PGC-1α pathway while this suppression is significantly attenuated by melatonin via promotion of  SIRT1/PGC-1α/Nrf2 and AMPK/Nrf2 pathway (131, 180).

     P53 is a tumor suppressor, but its overexpression leads to activation of an oncogene that finally results in cancernogenesis. P53 is just a component of an extensive network which regulates the carcinogenesis  (302). An excessive ROS generation in cardiomyocytes caused by Cr(VI) activates the P53 pathway resulting in mitochondrial damage and release of cytochrome c in the cytosol. Cytochrome c upregulates Bax and downregulates Bcl-2 and Bcl-XL protein expressions leading to cleavage of caspase-3 and apoptosis. Melatonin, as an antioxidant, activates the AMPK/Nrf2 pathway to inhibit ROS generation and protects the cardiomyocytes from oxidative stress and apoptosis (180).

     The Ras mainly maintains vascular homeostasis, blood volume, and electrolyte composition (303). But Ras also modulates melatonin synthesis by establishing the circadian rhythm. On other hand,  melatonin can suppress Ras activity by deactivating the Ras blockades (304). Currently it is found that melatonin can suppress the overexpression of Ras (305). Thus, melatonin may ameliorate the Cr(VI) mediated overexpression of Ras to protect the cell from apoptosis and terminate the carcinogenic signals. Furthermore, melatonin suppresses the proliferation, migration, and invasion of gall bladder cancer cells via arresting the PI3K/Akt/mTOR signaling pathway by inhibiting the phosphorylation of PI3K/Akt and mTOR in a time-dependent manner (306). Chen et al. (307) demonstrated that melatonin inhibits AKT phosphorylation through the MT1 receptor, preventing PI3K activation in glioblastoma cancer stem cells. Cr(VI) also downregulates the pathway of PI3K/Akt to induce autophagy. On the contrary, via MT1  melatonin promotes phosphorylation of Akt that activates the PI3K signaling to reduce autophagy of primary astrocytes (308, 309). Whether Cr(VI) promotes apoptosis or autophagy may be depended on the time, and routes of exposure. Thus, it can presume that melatonin can be a protective molecule against Cr(VI)-induced PI3K/Akt mediated apoptosis and autophagy.

     Formation of unfolded protein and initiation of UPR are caused by oxidative imbalance in the ER. Melatonin as a potent antioxidant protects cell from apoptosis through modulating the ER stress (207). This is evidenced by melatonin’s supplementation suppressing ER stress by inhibiting ROS generation as well as AMPK activation (310). Melatonin also increases the protein folding ability of ER and decreases the expression and accumulation of ER stress-associated unfolded and toxic proteins (311–313). Thus, melatonin is able to reduce ER stress and protects the cells from autophagy and apoptosis induced by Cr(VI) mediated ER stress.

 

7. EFFECTS OF MELATONIN ON Cr(VI)-INDUCED INFLAMMATION IN CVD

     Inflammation is a sequence of responses of the tissues and organs to injury or pathogen invasions. Cr(VI) induces inflammation by triggering diverse inflammatory responses driven by different cytokines (213). Melatonin is considered as a buffering molecule for the inflammation or the immune system. It acts as a stimulant under both basal or immunosuppressive conditions as well as an anti-inflammatory compound in the presence of intensified immune responses, such as acute inflammation (314). The release of cytokines, including TNF-α, IL-1β, IL-6, IL-12, and IFN-γ, and infiltrated macrophages in the tissues cause microvascular dysfunction and organ failure (315). Melatonin blocks the overproduction of pro-inflammatory cytokines, especially TNF-α (316), IL-1β, IL-6 and IL-8 (317, 318) and increases IL-10 levels (319). In addition, melatonin modulates the expression of TLR4-induced inflammatory gene expressions to control the activities of other pro and anti-inflammatory cytokines (320).

     Melatonin enhances immune response in aged individuals by boosting the immunosenescence suppressed T lymphocytes, especially the CD4+ lymphocyte. It mediates the activation, proliferation, and differentiation of helper T cells time-dependently (321). Melatonin increases cytokine levels within a short period, leading to the gradual recovery of cytokine expression. Moreover, T/B cell activation in pinealectomized mice was compromised indicating melatonin’s critical role in regulation of immune balance by modulating the activation of T and B lymphocytes (322). Presumably, melatonin can alleviate the Cr(VI)-mediated altered immune response.

     The pineal gland is also an essential immune target. IFN-γ was found to increase melatonin production from rat pineal glands in an in vitro-culture (323). Melatonin can enhance the B cell proliferation and the Th1 cytokines (IL-2 and IFN-γ) production (324, 325). A surge has been found in human cytokine production in the early morning, indicating that the IFN-γ/IL-10 is positively correlated with plasma melatonin (326).

     Likewise, other activities of melatonin also contribute to the protection against COX-2 induce chronic and intermittent hypoxia associated myocardial injury (253). Melatonin inhibits transcriptional activation of both COX2 and iNOS by suppressing the acetylation and binding of P52, thus it may hinder the initiation of the inflammation induced by Cr(VI) (327). COX-2-derived prostanoids are associated with vasoconstriction and vasodilation. Melatonin alleviates pulmonary vasoconstriction by enhancing the expression of vasodilator prostaglandins and reducing the expression of vasoconstrictor prostaglandins in pulmonary hypertensive neonates (328). In addition, melatonin persuades prostacyclin synthase expression, enhances the prostacyclin receptor's protein expression, and decreases the activity of COX-2 without changing the expression of the prostanoid vasoconstrictor (thromboxane) pathway (329).

     The upregulation of NF-κβ expression induced by Cr(VI) promotes ROS generation and increases the activity of IL-1β with myocardial inflammation which, if uncontrolled, will cause heart tissue damage. Melatonin inhibits the NF-κβ activity by suppressing ROS production through the AMPK/Nrf2 pathway (180). In addition, melatonin also inhibits NF-κβ transcriptional activation and  NF-κβ aggravated pro-inflammatory cytokines production to reduce inflammation (330). The potential mechanisms of melatonin on the Cr(VI) induced CVD are summarized in Figure 4.

 

8. CONCLUSION

     Cr(VI) induces oxidative stress and resultant inflammation, Ca2+ overload, endothelial dysfunction, and apoptosis in cardiovascular system. Cr(VI) is a classified group I carcinogen and causes carcinogenesis by inhibiting the expressions of different proteins. Melatonin an indolamine synthesized in mitochondria is a potent antioxidant and a metal chelator. It can also be classified as a nutraceutical substance since it is widely present in food staff including eggs, milk, fish, nuts, cherries, vegetables and wine, an integral part of our daily diet. Melatonin exerts its protective effects on Cr(VI) induced oxidative stress, either by chelating Cr(VI) or by scavenging the ROS and RNS; therefore, it normalizes the NO metabolism and reduces the Ca2+ overload to protect the cardiac endothelium. Melatonin also maintains the functions of SIRTUINS, FOXOs, PGC-1α, and AMPK to preserve nutrients and energy metabolism and mitochondrial biogenesis while suppressing the overreactions of Akt and P53. Melatonin also buffers the immune system and protects the cardiac tissue from Cr(VI)-induced inflammation by inhibiting activities of pro-inflammatory cytokines and enhancing the production of the anti-inflammatory cytokines. The protective mechanisms of melatonin against Cr(VI)-induced damages are summarized in Figure 4. Thus, we hypothesize that melatonin can serve as a therapeutic as well as nutraceutical molecule to reduce Cr(VI)-induced CVDs clinically.

Figure 4.jpg

 Fig. 4. The potential protective mechanisms of melatonin on Cr(VI)-induced endothelial dysfunction, Ca2+ overload, inflammation and apoptosis mediated CVD.

     (AMPK)- Adenosine Monophosphate-Activated Protein Kinase, (BAX)- Bcl2 Associated X, (Bcl-2)- B Cell Lymphoma-2, (Bcl-XL)- B Cell Lymphoma Extra Large, (FOXO)- Forkhead Box Transcription Factor, (NF-κβ)- Nuclear Factor-Kappa β, (PGC-1α)- Peroxisome Proliferator-Activated Receptor-Gamma (PPAR-ϒ) Coactivator 1-Alpha, (PI3K)- Phosphatidylinositol-3-Phosphate, (SIRT)- Silent Information Regulators Of Transcription, (COX-2)- Cyclooxygenase-2, (IFN-Ƴ)- Interferon Gamma, (NF-κβ)- Nuclear Factor-Kappa β, (ER stress)- Endoplasmic Reticulum Stress, (IL)- Interleukin, Nrf-2- Nuclear Factor Erythroid-2 Related Factor-2, (NO)- Nitric Oxide, (ONOO.)- Peroxynitrite, (ROS)- Reactive Oxygen Species, (Mg)- Magnesium, (K)- Potassium, (LDL)- Low-Density Lipoprotein, ICAM- Intercellular Adhesion Molecule, VCAM- Vascular Cell Adhesion Molecule

 

ACKNOWLEDGEMENTS

     Priyanka Ghosh thankfully acknowledges the receipt of the UGC Junior Research Fellowship (ref no.1571/PWD (NET-NOV2017), Govt. of India. Tiyasa Dey gratefully acknowledges the receipt of the UGC Senior Research Fellowship (ref no.1418/ (NET-JUNE 2015), Govt. of India. DB is supported by departmental BI grant of the University of Calcutta. DB also gratefully acknowledges the support he received from the DST PURSE Program awarded to the University of Calcutta.

 

AUTHORSHIP

     Dr. DB contributed to the conception and critical revision of the manuscript and approved it. PG prepared figures and drafted the manuscript. TD edited the manuscript.

 

CONFLICT OF INTEREST

     The authors declare no conflict of interest.

 

ABBREVIATIONS

 O2.- - Superoxide Anion

AMP- Adenosine Mono-Phosphate

AMPK- Adenosine Monophosphate-Activated Protein Kinase

AP-1 – Activator Protein-1

ATP- Adenosine Tri-Phosphate

BAD- Bcl2 Associated Agonist of Cell Death

BAX- Bcl2 Associated X

Bcl-2- B Cell Lymphoma-2

Bcl-XL- B Cell Lymphoma Extra Large

CAD - Coronary Artery Disease

CAT- Catalase

CHD - Coronary Heart Disease

CHF- Congestive Heart Failure

COX-2- Cyclooxygenase-2

Cr - Chromium

CRP- C-Reactive Protein

CVD - Cardiovascular Disease

EDCF - Endothelium Derived Contracting Factor

EDRF - Endothelium Derived Relaxing Factor

ERK- Extracellular Signal Regulated Kinase

FOXO- Forkhead Box Transcription Factor

G6PDH- Glucose-6-Phosphate Dehydrogenase

GPx- Glutathione Peroxidase

GSH- Reduced Glutathione

GTF- Glucose Tolerance Factor

GTP- Guanosine Triphosphate

H2O2 - Hydrogen Peroxide

HDL- High-Density Lipoprotein

HO. - Hydroxyl Radical

HOCl- Hypochlorous Acid

ICAM- intercellular adhesion molecule

IFN-Ƴ- Interferon Gamma

IGF-1- Insulin-Like Growth Factor

IHD - Ischemic Heart Diseases

LDL-C - Low-Density Lipoprotein Cholesterol

LOO– Lipo-peroxyl Radical

LPO - Lipid Peroxidation

MHC- Major Histocompatibility Complex

MI - Myocardial Infarction

Mt DNA - Mitochondrial DNA

mTOR- Mammalian Target Of Rapamycin

NAD+- Nicotinamide Adenine Dinucleotide (Oxidized)

NADH- Nicotinamide Adenine Dinucleotide (Reduced)

NADPH - Nicotinamide Adenine Dinucleotide Phosphate

NET- Neutrophil Extracellular Traps

NF-κβ- Nuclear Factor-Kappa β

Nrf-2- Nuclear Factor Erythroid-2 Related Factor-2

ONOO- - Peroxynitrite

ox-LDL- Oxidized LDL

PGC-1α- Peroxisome Proliferator-Activated Receptor-Gamma (PPAR-ϒ) Coactivator 1-Alpha

PGH2- Prostaglandin H Synthase 2

PI3K- Phosphatidylinositol-3-Phosphate

PKB- Protein Kinase B

PPAR-ϒ - Peroxisome Proliferator-Activated Receptor-Gamma (PPAR-ϒ)

PUMA- P53-Upregulated Modulator of Apoptosis

RAS- Renin Angiotensin System

RNS - Reactive Nitrogen Species

ROO.Peroxyl Radical

ROS – Reactive Oxygen Species

SIRT- Silent Information Regulators of Transcription

SMC- Smooth Muscle Cell

STAT- Signal Transducer and Activator Of Transcription

Th1 And 2- Type 1 And 2 T Helper Cells

TLR- Toll-Like Receptor

TNF-α- Tumor Necrosis Factor Α

Trx- Thioredoxin

VCAM- Vascular Cell Adhesion Molecule

WHO - World Health Organization

 

REFERENCES

 

  1. Chowdhury R, Ramond A, O’Keeffe LM, Shahzad S, Kunutsor SK, Muka T, Gregson J, Willeit P, Warnakula S, Khan H, Chowdhury S, Gobin R, Franco OH, Di Angelantonio E (2018) Environmental toxic metal contaminants and risk of cardiovascular disease: Systematic review and meta-analysis. BMJ Open 362 (k3310): 14–16. DOI: 10.1136/bmj.k3310.

  2. Tanwar V, Katapadi A, Adelstein JM, Grimmer JA, Wold LE (2018) Cardiac pathophysiology in response to environmental stress: a current review. Curr. Opin. Physiol. 1: 198–205. DOI: 10.1016/j.cophys.2017.11.005.

  3. Buhari O, Dayyab FM, Igbinoba O, Atanda A, Medhane F, Faillace RT (2020) The association between heavy metal and serum cholesterol levels in the US population: National Health and Nutrition Examination Survey 2009–2012. Hum. Exp. Toxicol. 39 (3): 355–364. DOI: 10.1177/0960327119889654.

  4. Balali-Mood M, Naseri K, Tahergorabi Z, Khazdair MR, Sadeghi M (2021) Toxic Mechanisms of Five Heavy Metals: Mercury, Lead, Chromium, Cadmium, and Arsenic. Front. Pharmacol. 12 (12): 1–19. DOI: 10.3389/fphar.2021.643972.

  5. Tsutsui H, Kinugawa S, Matsushima S (2011) Oxidative stress and heart failure. Am. J. Physiol. - Hear. Circ. Physiol. 301 (6): 2181–2190. DOI: 10.1152/ajpheart.00554.2011.

  6. Tchounwou PB, Yedjou CG, Patlolla AK, Sutton DJ (2012) Heavy Metal Toxicity and the Environment. Mol. Clin. Environ. Toxicol. 101: 133–164. DOI: 10.1007/978-3-7643-8340-4.

  7. Ghosh P, Dey T, Chattopadhyay A, Bandyopadhyay D (2021) An insight into the ameliorative effects of melatonin against chromium induced oxidative stress and DNA damage: a review. Melatonin Res. 4 (3): 377–407. DOI: 10.32794/MR112500101.

  8. Ali A, Ma Y, Reynolds J, Wise JP, Inzucchi SE, Katz DL (2011) Chromiun effects on glucose tolerance and insulin sensitivity in persons at risk for diabetes mellitus. Endocr. Pract. 17 (1): 16–25. DOI: 10.4158/EP10131.OR.

  9. Sahin K, Onderci M, Tuzcu M, Ustundag B, Cikim G, Ozercan IH, Sriramoju V, Juturu V, Komorowski JR (2007) Effect of chromium on carbohydrate and lipid metabolism in a rat model of type 2 diabetes mellitus: the fat-fed, streptozotocin-treated rat. Metabolism 56 (9): 1233–1240. DOI: 10.1016/j.metabol.2007.04.021.

  10. Hua Y, Clark S RJ and NS (2012) Molecular Mechanisms of Chromium in Alleviating Insulin Resistance. J. Nutr. Biochem. 23 (4): 313–319. DOI: 10.1016/j.jnutbio.2011.11.001.Molecular.

  11. Sandra S. Wise JPW (2012) Chromium and genomic stability. Mutat. Res. 733 (1–2): 78–82. DOI: 10.1016/j.mrfmmm.2011.12.002.

  12. Qi W, Reiter RJ, Tan DX, Garcia JJ, Manchester LC, Karbownik M, Calvo JR (2000) Chromium(III)-induced 8-hydroxydeoxyguanosine in DNA and its reduction by antioxidants: Comparative effects of melatonin, ascorbate, and vitamin E. Environ. Health Perspect. 108 (5): 399–403. DOI: 10.1289/ehp.00108399.

  13. Bagchi D, Bagchi M, Stohs SJ (2001) Chromium (VI)-induced oxidative stress, apoptotic cell death and modulation of p53 tumor suppressor gene. Mol. Cell. Biochem. 222 (1–2): 149–158. DOI: 10.1023/A:1017958028256.

  14. Bagchi D,  Balmoori J, Bagchi M, Ye X, Williams CB, Stohs SJ (2000) Role of P53 tumor suppressor gene in the toxicity of TCDD, endrin, naphthalene, and chromium (VI) in liver and brain tissues of mice. Free Radic. Biol. Med. 28 (6): 895–903. DOI: 10.1016/s0891-5849(00)00173-8.

  15. Fang Z, Zhao M, Zhen H, Chen L, Shi P, Huang Z (2014) Genotoxicity of tri- and hexavalent chromium compounds in vivo and their modes of action on DNA damage in vitro. PLoS One 9 (8): 1–9. DOI: 10.1371/journal.pone.0103194.

  16. ang D, Yang Q, Fu N, Li S, Han B, Liu Y, Tang Y, Guo X, Lv Z, Zhang Z (2021) Hexavalent chromium induced heart dysfunction via Sesn2-mediated impairment of mitochondrial function and energy supply. Chemosphere 264 (128547): 1–10. DOI: 10.1016/j.chemosphere.2020.128547.

  17. Abraham AS, Sonnenblick M, Eni M, Shemesh O, Batt AP (1980) Serum chromium in patients with recent and old myocardial infarction. Am. Heart J. 99 (5): 604–606. DOI: 10.1016/0002-8703(80)90734-6.

  18. Gutiérrez-Bedmar M, Martínez-González MÁ, Muñoz-Bravo C, Ruiz-Canela M, Mariscal A, Salas-Salvadó J et al. (2017) Chromium exposure and risk of cardiovascular disease in high cardiovascular risk subjects: Nested case-control study in the prevention with mediterranean diet (PREDIMED) study. Circ. J. 81 (8): 1183–1190. DOI: 10.1253/circj.CJ-17-0032.

  19. Benjamin EJ, Muntner P, Alonso A, Bittencourt MS, Callaway CW, Carson AP et al. (2019) Heart disease and stroke statistics-2019 update: A report from the american heart association. Circulation 139 (10): e56–e528. DOI: 10.1161/CIR.0000000000000659.

  20. Sharman EH, Bondy SC (2016) Melatonin: A safe nutraceutical and clinical agent. Nutraceuticals: efficacy, safety and toxicity. Elsevier Inc.; 2016. 501–509 p. DOI: 10.1016/B978-0-12-802147-7.00036-X.

  21. Domínguez-Rodríguez A, Abreu-González P, Báez-Ferrer N, Reiter RJ, Avanzas P, Hernández-Vaquero D (2021) Melatonin and cardioprotection in humans: A systematic review and meta-Analysis of randomized controlled trials. Front. Cardiovasc. Med. 12 (8): 1–6. DOI: 10.3389/fcvm.2021.635083.

  22. Tan DX, Hardeland R, Manchester LC, Korkmaz A, Ma S, Rosales-Corral S, Reiter RJ (2012) Functional roles of melatonin in plants, and perspectives in nutritional and agricultural science. J. Exp. Bot. 63 (2): 577–597. DOI: 10.1093/jxb/err256.

  23. Jiki Z, Lecour S, Nduhirabandi F (2018) Cardiovascular benefits of dietary melatonin: A myth or a reality? Front. Physiol. 9 (528): 1–17. DOI: 10.3389/fphys.2018.00528.

  24. Yang Y, Sun Y, Yi W, Li Y, Fan C, Xin Z, Jiang S, Di S, Qu Y, Reiter RJ, Yi D (2014) A review of melatonin as a suitable antioxidant against myocardial ischemia-reperfusion injury and clinical heart diseases. J. Pineal Res. 57 (4): 357–366. DOI: 10.1111/jpi.12175.

  25. Koziróg M, Poliwczak AR, Duchnowicz P, Koter-Michalak M, Sikora J, Broncel M (2011) Melatonin treatment improves blood pressure, lipid profile, and parameters of oxidative stress in patients with metabolic syndrome. J. Pineal Res. 50 (3): 261–266. DOI: 10.1111/j.1600-079X.2010.00835.x.

  26. Dominguez-Rodriguez A, Abreu-Gonzalez P, Reiter RJ (2014) The potential usefulness of serum melatonin level to predict heart failure in patients with hypertensive cardiomyopathy. Int. J. Cardiol. 174 (2): 415–417. DOI: 10.1016/j.ijcard.2014.04.044.

  27. Altun A, Yaprak M, Aktoz M, Vardar A, Betul UA, Ozbay G (2002) Impaired nocturnal synthesis of melatonin in patients with cardiac syndrome X. Neurosci. Lett. 327 (2): 143–145. DOI: 10.1016/S0304-3940(02)00368-3.

  28. Domínguez-Rodríguez A, Abreu-González P, García MJ, Sanchez J, Marrero F, De Armas-Trujillo D (2002) Decreased nocturnal melatonin levels during acute myocardial infarction. J. Pineal Res. 33 (4): 248–252. DOI: 10.1034/j.1600-079X.2002.02938.x.

  29. Girotti L, Lago M, Ianovsky O, Elizari M V., Dini A, Pérez Lloret S, Albornoz LE, Cardinali DP (2003) Low Urinary 6-Sulfatoxymelatonin Levels in Patients with Severe Congestive Heart Failure. Endocrine 22 (3): 245–248. DOI: 10.1385/ENDO:22:3:245.

  30. Dzida G, Prystupa A, Lachowska-Kotowska P, Kardas T, Kamieński P, Kimak E, Hałabiś M, Kiciński P (2013) Alteration in diurnal and nocturnal melatonin serum level in patients with chronic heart failure. Ann. Agric. Environ. Med. 20 (4): 745–748.

  31. Siegel D, Black DM, Seeley DG, Hulley SB (1992) Circadian Variation in Ventricular arrhythmias in hypertensive men. Am. J. Cardiol. 69 (4): 344–347. DOI: 10.1016/0002-9149(92)90231-m.

  32. Savopoulos C, Ziakas A, Hatzitolios A, Delivoria C, Kounanis A, Mylonas S, Tsougas M, Psaroulis D (2006) Circadian rhythm in sudden cardiac death: A retrospective study of 2,665 cases. Angiology 57 (2): 197–204. DOI: 10.1177/000331970605700210.

  33. Sun H, Brocato J, Costa M (2015) Oral chromium exposure and toxicity. Curr. Env. Heal. Rep. 2 (3): 295–303. DOI: 10.1007/s40572-015-0054-z.

  34. Simonoff M (1984) Chromium deficiency and cardiovascular risk. Cardiovasc. Res. 18 (10): 591–596. DOI: 10.1093/CVR/18.10.591.

  35. Nickens KP, Patierno SR, Ceryak S (2010) Chromium genotoxicity: A double-edged sword. Chem. Biol. Interact. 188 (2): 276–288. DOI: 10.1016/j.cbi.2010.04.018.

  36. Oliveira H (2012) Chromium as an environmental pollutant: Insights on induced plant toxicity. J. Bot. 2012 : 2090–2120. DOI: 10.1155/2012/375843.

  37. Liang J, Huang X, Yan J, Li Y, Zhao Z, Liu Y, Ye J, Wei Y (2021) A review of the formation of Cr(VI) via Cr(III) oxidation in soils and groundwater. Sci. Total Environ. 774 (145762): 1–13. DOI: 10.1016/J.SCITOTENV.2021.145762.

  38. Apte AD, Tare V, Bose P (2006) Extent of oxidation of Cr(III) to Cr(VI) under various conditions pertaining to natural environment. J. Hazard. Mater. 128 (2–3): 164–174. DOI: 10.1016/J.JHAZMAT.2005.07.057.

  39. Wilbur S, Abadin H, Fay M, Yu D, Tencza B, Ingerman L, Klotzbach J, James S (2012) Toxicological Profile for Chromium. Agency for Toxic Substances and Disease Registry (US) (September): 1-9. www.atsdr.cdc.gov.

  40. International Agency for Research on Cancer 2012. (2012) Arsenic, Metals, Fibres, and Dusts. Vol. 1989, IARC Monographs: Arsenic, Metals, Fibres, and Dusts in a Review of Human Carcinogens. 2012. 147–168 p.

  41. Wayne W. Campbell , Lyndon J.O Joseph, Richard A. Anderson, Stephen L. Davey JH and WJE (2002) Effects of resistive training and chromium picolinate on body composition and skeletal muscle size in older women. Int. J. Sport Nutr. Exerc. Metab. 12 (2): 125–135. DOI: 10.1123/ijsnem.12.2.125.

  42. Anderson RA (1998) Chromium, glucose intolerance and diabetes. J. Am. Coll. Nutr. 17 (6): 548–555. DOI: 10.1080/07315724.1998.10718802.

  43. Schwarz K, Mertz W (1959) Chromium(III) and the glucose tolerance factor. Arch. Biochem. Biophys. 85 (1): 292–295. DOI: 10.1016/0003-9861(59)90479-5.

  44. Jeejeebhoy KN, Chu RC, Marliss EB, Greenberg GR, Bruce-Robertson A (1977) Chromium deficiency, glucose intolerance, and neuropathy reversed by chromium supplementation, in a patient receiving long-term total parenteral nutrition. Am. J. Clin. Nutr. 30 (4): 531–538. DOI: 10.1093/AJCN/30.4.531.

  45. Ngala RA, Awe MA, Nsiah P (2018) The effects of plasma chromium on lipid profile, glucose metabolism and cardiovascular risk in type 2 diabetes mellitus. A case - control study. PLoS One 13 (7): 1–11. DOI: 10.1371/journal.pone.0197977.

  46. Scherer T, O’Hare J, Diggs-Andrews K, Schweiger M, Cheng B, Lindtner C, Zielinski E, Vempati P, Su K, Dighe S, Milsom T, Puchowicz M, Scheja L, Zechner R, Fisher SJ, Previs SF, Buettner C (2011) Brain insulin controls adipose tissue lipolysis and lipogenesis. Cell Metab. 13 (2): 183–194. DOI: 10.1016/j.cmet.2011.01.008.

  47. Duvillard L, Florentin E, Lizard G, Petit JM, Galland F, Monier S, Gambert P, Vergès B (2003) Cell surface expression of LDL receptor is decreased in type 2 diabetic patients and is normalized by insulin therapy. Diabetes Care 26 (5): 1540–1544. DOI: 10.2337/DIACARE.26.5.1540.

  48. Lapenna D, Ciofani G (2020) Chromium and human low-density lipoprotein oxidation. J. Trace Elem. Med. Biol. 59 (126411): 1–29. DOI: 10.1016/J.JTEMB.2019.126411.

  49. Abraham AS, Sonnenblick M, Eini M, Shemesh O, Batt AP (1980) The effect of chromium on established atherosclerotic plaques in rabbits. Am. J. Clin. Nutr. 33 (11): 2294–2298. DOI: 10.1093/AJCN/33.11.2294.

  50. Uusitupa MI, Kumpulainen JT, Voutilainen E, Hersio K, Sarlund H, Pyörälä KP, Koivistoinen PE, Lehto JT (1983) Effect of Inorganic chromium supplementation on glucose tolerance, insulin response, and serum lipids in noninsulin-dependent diabetics. Am. J. Clin. Nutr. 38 (3): 404–410. DOI: 10.1093/ajcn/38.3.404.

  51. Krzysik M, Grajeta H, Prescha A, Weber R (2011) Effect of cellulose, pectin and chromium(III) on lipid and carbohydrate metabolism in rats. J. Trace Elem. Med. Biol. 25 (2): 97–102. DOI: 10.1016/J.JTEMB.2011.01.003.

  52. Swaroop A, Bagchi M, Preuss HG, Zafra-Stone S, Ahmad T, Bagchi D 2019. (2019) Benefits of chromium(III) complexes in animal and human health. In: The Nutritional Biochemistry of Chromium (III). Elsevier B.V.; 2019. p. 251–278. DOI: 10.1016/b978-0-444-64121-2.00008-8.

  53. Barrett-Connor E (2003) Diabetes and heart disease. Diabetes Care 26 (10): 2947–2958. DOI: 10.2337/DIACARE.26.10.2947.

  54. Haffner SM (2009) Coronary heart disease in patients with diabetes. N. Engl. J. Med. 342 (14): 1040–1042. DOI: 10.1056/NEJM200004063421408.

  55. Wang ZQ, Qin J, Martin J, Zhang XH, Sereda O, Anderson RA, Pinsonat P, Cefalu WT (2007) Phenotype of subjects with type 2 diabetes mellitus may determine clinical response to chromium supplementation. Metabolism 56 (12): 1652–1655. DOI: 10.1016/j.metabol.2007.07.007.

  56. Pattar GR, Tackett L, Liu P, Elmendorf JS (2006) Chromium picolinate positively influences the glucose transporter system via affecting cholesterol homeostasis in adipocytes cultured under hyperglycemic diabetic conditions. Mutat. Res. - Genet. Toxicol. Environ. Mutagen. 610 (1–2): 93–100. DOI: 10.1016/j.mrgentox.2006.06.018.

  57. Raja NS, Nair BU (2008) Chromium(III) complexes inhibit transcription factors binding to DNA and associated gene expression. Toxicology 251 (1–3): 61–65. DOI: 10.1016/j.tox.2008.07.052.

  58. Das J, Sarkar A, Sil PC (2015) Hexavalent chromium induces apoptosis in human liver (HepG2) cells via redox imbalance. Toxicol. Reports 2: 600–608. DOI: 10.1016/J.TOXREP.2015.03.013.

  59. Zhong X, da Silveira e Sá R de C, Zhong C (2017) Mitochondrial biogenesis in response to chromium (VI) toxicity in human liver cells. Int. J. Mol. Sci. 18 (9): 1–17. DOI: 10.3390/IJMS18091877.

  60. Beaver LM, Stemmy EJ, Schwartz AM, Damsker JM, Constant SL, Ceryak SM, Patierno SR (2009) Lung inflammation, injury, and proliferative response after repetitive particulate hexavalent chromium exposure. Environ. Health Perspect. 117 (12): 1896–1902. DOI: 10.1289/ehp.0900715.

  61. Fleeger AK, Deng JF (2011) A case study of chromium VI-induced skin ulcerations during a porcelain enamel curing operation. Appl. Occup. Environ. Hyg. 5 (6): 1589–1605. DOI: 10.1080/1047322X.1990.10389658.

  62. Hessel EVS, Staal YCM, Piersma AH, den Braver-Sewradj SP, Ezendam J (2021) Occupational exposure to hexavalent chromium. Part I. Hazard assessment of non-cancer health effects. Regul. Toxicol. Pharmacol. 126 (105048): 1–18. DOI: 10.1016/j.yrtph.2021.105048.

  63. Shelnutt SR, Goad P, Belsito D V. (2007) Dermatological toxicity of hexavalent chromium. Crit. Rev. Toxicol. 37 (5): 375–387.

  64. Wedeen RP, Qian L (1991) Chromium-Induced Kidney Disease. Environ. Health Perspect. 92 : 71–74. DOI: 10.2307/3431139.

  65. Shrivastava R, Upreti RK, Seth PK, Chaturvedi UC (2002) Effects of chromium on the immune system. FEMS Immunol. Med. Microbiol. 34 (1): 1–7. DOI: 10.1111/J.1574-695X.2002.TB00596.X.

  66. Katz SA (1991) The analytical biochemistry of chromium. Environ. Health Perspect. 92 : 13–16. DOI: 10.1289/ehp.919213.

  67. Costa M (1997) Toxicity and carcinogenicity of Cr(VI) in animal models and humans. Crit. Rev. Toxicol. 27 (5): 431–442. DOI: 10.3109/10408449709078442.

  68. Chiu A, Shi XL, Lee WKP, Hill R, Wakeman TP, Katz A, Xu B, Dalal NS, Robertson JD, Chen C, Chiu N, Donehower L (2010) Review of chromium (VI) apoptosis, cell-cycle-arrest, and carcinogenesis. J. Environ. Sci. Heal. - Part C Environ. Carcinog. Ecotoxicol. Rev. 28 (3): 188–230. DOI: 10.1080/10590501.2010.504980.

  69. Jennette KW (1979) Chromate metabolism in liver microsomes. Biol. Trace Elem. Res. 1 (1): 55–62. DOI: 10.1016/0006-291x(78)90931-2.

  70. O’Brien T, Xu J, Patierno SR (2001) Effects of glutathione on chromium-induced DNA crosslinking and DNA polymerase arrest. Mol. Cell. Biochem. 222 (1–2): 173–182. DOI: 10.1023/A:1017918330073.

  71. Chen QY, Murphy A, Sun H CM (2019) Molecular and epigenetic mechanisms of Cr(VI)-induced carcinogenesis. Toxicol. Appl. Pharmacol. 377 (114636): 1–22. DOI: 10.1016/j.taap.2019.114636.

  72. Aiyar J, De Flora S, Wetterhahn KE (1992) Reduction of chromium(vi) to chromium(v) by rat liver cytosolic and microsomal fractions: Is dt-diaphorase involved? Carcinogenesis 13 (7): 1159–1166. DOI: 10.1093/carcin/13.7.1159.

  73. Connett PH, Wetterhahn KE (1983) Metabolism of the carcinogen chromate by cellular constituents. Inorg. Elem. Biochem. 54 : 93–124. DOI: 10.1007/bfb0111319.

  74. Shi X, Chiu A, Chen CT, Halliwell B, Castranova V, Vallyathan V (1999) Reduction of chromium (VI) and its relationship to carcinogenesis. J. Toxicol. Environ. Heal. - Part B Crit. Rev. 2 (1): 87–104. DOI: 10.1080/109374099281241.

  75. Maret W (2019) Chromium supplementation in human health, metabolic syndrome, and diabetes. Met. Ions Life Sci. 19 : 231–251. DOI: 10.1515/9783110527872-015.

  76. Wang Y, Su H, Gu Y, Song X, Zhao J (2017) Carcinogenicity of chromium and chemoprevention: A brief update. Onco. Targets Ther. 10: 4065–4079. DOI: 10.2147/OTT.S139262.

  77. Senoner T, Dichtl W (2019) Oxidative stress in cardiovascular diseases: Still a therapeutic target? Nutrients 11 (9): 1–25. DOI: 10.3390/nu11092090.

  78. Janero DR, Hreniuk D, Sharif HM (1993) Hydrogen peroxide-induced oxidative stress to the mammalian heart-muscle cell (cardiomyocyte): nonperoxidative purine and pyrimidine nucleotide depletion. J. Cell. Physiol. 155 (3): 494–504. DOI: 10.1002/JCP.1041550308.

  79. Holmström KM, Finkel T (2014) Cellular mechanisms and physiological consequences of redox-dependent signalling. Nat. Rev. Mol. Cell Biol. 15 (6): 411–421. DOI: 10.1038/nrm3801.

  80. Li JM, Gall NP, Grieve DJ, Chen M, Shah AM (2002) Activation of NADPH oxidase during progression of cardiac hypertrophy to failure. Hypertension 40 (4): 477–484. DOI: 10.1161/01.HYP.0000032031.30374.32.

  81. Heymes C, Bendall JK, Ratajczak P, Cave AC, Samuel JL, Hasenfuss G, Shah AM (2003) Increased myocardial NADPH oxidase activity in human heart failure. J. Am. Coll. Cardiol. 41 (12): 2164–2171. DOI: 10.1016/S0735-1097(03)00471-6.

  82. Kuroda J, Sadoshima J (2010) NADPH oxidase and cardiac failure. J. Cardiovasc. Transl. Res. 3 (4): 314–320. DOI: 10.1007/s12265-010-9184-8.

  83. Kuroda J, Ago T, Matsushima S, Zhai P, Schneider MD, Sadoshima J (2010) NADPH oxidase 4 (Nox4) is a major source of oxidative stress in the failing heart. Proc. Natl. Acad. Sci. U. S. A. 107 (35): 15565–15570. DOI: 10.1073/pnas.1002178107.

  84. Ago T, Kuroda J, Pain J, Fu C, Li H, Sadoshima J (2010) Upregulation of Nox4 by hypertrophic stimuli promotes apoptosis and mitochondrial dysfunction in cardiac myocytes. Circ. Res. 106 (7): 1253–1264. DOI: 10.1161/CIRCRESAHA.109.213116.

  85. Stohs SJ, Bagchi D (1995) Oxidative mechanisms in the toxicity of metal ions. Free Radic. Biol. Med. 18 (2): 321–336. DOI: 10.1016/0891-5849(94)00159-H.

  86. Kanner J, German JB, Kinsella JE (1987) Initiation of lipid peroxidation in biological systems. C R C Crit. Rev. Food Sci. Nutr. 25 (4): 317–364. DOI: 10.1080/10408398709527457.

  87. Halliwell B, Chirico S (1993) Lipid peroxidation: its mechanism, measurement, and significance. Am. J. Clin. Nutr. 57 (5): 715–725. DOI: 10.1093/ajcn/57.5.715S.

  88. Upadhyay RK, Panda SK (2010) Influence of chromium salts on increased lipid peroxidation and differential pattern in antioxidant metabolism in Pistia stratiotes L. Brazilian Arch. Biol. Technol. 53 (5): 1137–1144. DOI: 10.1590/S1516-89132010000500008.

  89. Katz AM, Freston JW, Messineo FC, Herbette LG (1985) Membrane damage and the pathogenesis of cardiomyopathies. J. Mol. Cell. Cardiol. 17 (SUPPL. 2): 11–20. DOI: 10.1016/0022-2828(85)90004-5.

  90. Schwenke DC, Carew TE (1989) Initiation of atherosclerotic lesions in cholesterol-fed rabbits. II. Selective retention of LDL vs. selective increases in LDL permeability in susceptible sites of arteries. Arteriosclerosis 9 (6): 908–918. DOI: 10.1161/01.ATV.9.6.908.

  91. Wang X, Son YO, Chang Q, Sun L, Hitron JA, Budhraja A, Zhang Z, Ke Z, Chen F, Luo J, Shi X (2011) NADPH oxidase activation is required in reactive oxygen species generation and cell transformation induced by hexavalent chromium. Toxicol. Sci. 123 (2): 399–410. DOI: 10.1093/toxsci/kfr180.

  92. Rajendran P, Rengarajan T, Thangavel J, Nishigaki Y, Sakthisekaran D, Sethi G, Nishigaki I (2013) The vascular endothelium and human diseases. Int. J. Biol. Sci. 9 (10): 1057–1069. DOI: 10.7150/ijbs.7502.

  93. Harrison DG (1997) Cellular and molecular mechanisms of endothelial cell dysfunction. J. Clin. Invest. 100 (9): 2153–2157. DOI: 10.1172/JCI119751.

  94. Salvador M, Higgs A (1992) The l-arginine-nitric oxide pathway. N. Engl. J. Med. 397 (27): 2002–2012. DOI: 10.1056/NEJM199312303292706.

  95. Forstermann U, Münzel T (2006) Endothelial nitric oxide synthase in vascular disease: From marvel to menace. Circulation 113 (13): 1708–1714. DOI: 10.1161/CIRCULATIONAHA.105.602532.

  96. Kojda G, Harrison D (1999) Interactions between NO and reactive oxygen species: Pathophysiological importance in atherosclerosis, hypertension, diabetes and heart failure. Cardiovasc. Res. 43 (3): 562–571. DOI: 10.1016/S0008-6363(99)00169-8.

  97. Bloodsworth A, Donnell VBO, Freeman B  (2000) Nitric oxide regulation of free radical– and enzyme-mediated lipid and lipoprotein oxidation. Arterioscler. Thromb. 20 (7): 1707–1715. DOI: 10.1161/01.atv.20.7.1707.

  98. Thomson L, Trujillo M, Telleri R, Radi R (1995) Kinetics of cytochrome c2+ oxidation by peroxynitrite implications for superoxide measurements in nitric oxide-producing biological systems. Arch. Biochem. Biophys. 319 (2): 491–497. DOI: 10.1006/abbi.1995.1321.

  99. Touyz RM (2004) Reactive oxygen species, vascular oxidative stress, and redox signaling in hypertension: What is the clinical significance? Hypertension 44 (3): 248–252. DOI: 10.1161/01.HYP.0000138070.47616.9d.

  100. Beckman JS, Beckman TW, Chen J, Marshall PA, Freeman BA (1990) Apparent hydroxyl radical production by peroxynitrite: Implications for endothelial injury from nitric oxide and superoxide. Proc. Natl. Acad. Sci. U. S. A. 87 (4): 1620–1624. DOI: 10.1073/pnas.87.4.1620.

  101. White CR, Brock TA, Chang LY, Crapo J, Briscoe P, Ku D, Bradley WA, Gianturco SH, Gore J, Freeman BA, Tarpey MM (1994) Superoxide and peroxynitrite in atherosclerosis. Proc. Natl. Acad. Sci. U. S. A. 91 (3): 1044–1048. DOI: 10.1073/pnas.91.3.1044.

  102. Radi R, Rodriguez M, Castro L, Telleri R (1994) Inhibition of mitochondrial electron transport by peroxynitrite. Arch. Biochem. Biophys. 308 (1): 89–95. DOI: 10.1006/abbi.1994.1013.

  103. Castro L, Rodriguez M, Radi R (1994) Aconitase is readily inactivated by peroxynitrite, but not by its precursor, nitric oxide. J. Biol. Chem. 269 (47): 29409–29415. DOI: 10.1016/s0021-9258(18)43894-x.

  104. Sellke FW, Armstrong ML, Harrison DG (1990) Endothelium-dependment vascular relaxation is abnormal in the coronary microcirculation of atherosclerotic primates. Circulation 81 (5): 1586–1593. DOI: 10.1161/01.CIR.81.5.1586.

  105. Zeiher AM, Drexler H, Wollschlager H, Just H (1991) Endothelial dysfunction of the coronary microvasculature is associated with impaired coronary blood flow regulation in patients with early atherosclerosis. Circulation 84 (5): 1984–1992. DOI: 10.1161/01.CIR.84.5.1984.

  106. Creager MA, Cooke JP, Mendelsohn ME, Gallagher SJ, Coleman SM, Loscalzo J, Dzau VJ (1990) Impaired vasodilation of forearm resistance vessels in hypercholesterolemic humans. J. Clin. Invest. 86 (1): 228–234. DOI: 10.1172/JCI114688.

  107. Oishi K, Raynor RL, Charp PA, Kuo JF (1988) Regulation of protein kinase C by lysophospholipids: Potential role in signal transduction. J. Biol. Chem. 263 (14): 6865–6871. DOI: 10.1016/s0021-9258(18)68724-1.

  108. Ohara Y, Peterson TE, Zheng B, Kuo JF, Harrison DG (1994) Lysophosphatidylcholine increases vascular superoxide anion production via protein kinase C activation. Arterioscler. Thromb. Vasc. Biol. 14 (6): 1007–1013. DOI: 10.1161/01.ATV.14.6.1007.

  109. Pritchard J, Ackerman A, Kalyanaraman B (2000) Chromium (VI) increases endothelial cell expression of ICAM-1 and decreases nitric oxide activity. J. Environ. Pathol. Toxicol. Oncol. 19 (3): 251–260.

  110. Porter R, Jáchymová M, Martásek P, Kalyanaraman B, Vásquez-Vivar J (2005) Reductive activation of Cr(VI) by nitric oxide synthase. Chem. Res. Toxicol. 18 (5): 834–843. DOI: 10.1021/TX049778E.

  111. Kushwaha BK, Ali HM, Siddiqui MH, Singh VP (2020) Nitric oxide-mediated regulation of sub-cellular chromium distribution, ascorbate-glutathione cycle and glutathione biosynthesis in tomato roots under chromium (VI) toxicity. J. Biotechnol. 318 (2): 68–77. DOI: 10.1016/J.JBIOTEC.2020.05.006.

  112. Brito R, Castillo G, González J, Valls N, Rodrigo R (2015) Oxidative stress in hypertension: Mechanisms and therapeutic opportunities. Exp. Clin. Endocrinol. Diabetes 123 (6): 325–335. DOI: 10.1055/s-0035-1548765.

  113. Görlach A, Bertram K, Hudecova S, Krizanova O (2015) Calcium and ROS: A mutual interplay. Redox Biol. 6 : 260–271. DOI: 10.1016/j.redox.2015.08.010.

  114.  Fiolet J, Baartscheer A, Schumacher C, Coronel R, Terwelle H (1984) The change of the free energy of ATP hydrolysis during global ischemia and anoxia in the rat heartIts possible role in the regulation of transsarcolemmal sodium and potassium gradients. J. Mol. Cell. Cardiol. 16 (11): 1023–1036. DOI: 10.1016/S0022-2828(84)80015-2.

  115. Vanechteld C, Kirkels J, Eijgelshoven M, Vandermeer P, Ruigrok T (1991) Intracellular sodium during ischemia and calcium-free perfusion: A 23Na NMR study. J. Mol. Cell. Cardiol. 23 (3): 297–307. DOI: 10.1016/0022-2828(91)90066-U.

  116. Philipson KD, Ward R (1985) Effects of fatty acids on Na+-Ca2+ exchange and Ca2+ permeability of cardiac sarcolemmal vesicles. J. Biol. Chem. 260 (17): 9666–9671. DOI: 10.1016/s0021-9258(17)39290-6.

  117. Dhalla NS, Elimban V, Rupp H (1992) Paradoxical role of lipid metabolism in heart function and dysfunction. Mol. Cell. Biochem. 116 (1–2): 3–9. DOI: 10.1007/BF01270562.

  118. Tani M (1990) Mechanisms of Ca2+ overload in reperfused ischemic myocardium. Annu. Rev. Physiol. 52 (1): 543–559. DOI: 10.1146/annurev.physiol.52.1.543.

  119. Mukherjee A, Wong TM, Buja LM, Lefkowitz RJ, Willerson JT (1979) Beta adrenergic and muscarinic cholinergic receptors in canine myocardium: Effects of ischemia. J. Clin. Invest. 64 (5): 1423–1428. DOI: 10.1172/JCI109600.

  120. Dhalla NS, Temsah RM, Netticadan T (2000) Role of oxidative stress in cardiovascular diseases. J. Hypertens. 18 (6): 655–673. DOI: 10.1097/00004872-200018060-00002.

  121. Ishida H, Ichimori K, Hirota Y, Fukahori M, Nakazawa H (1996) Peroxynitrite-induced cardiac myocyte injury. Free Radic. Biol. Med. 20 (3): 343–350. DOI: 10.1016/0891-5849(96)02060-6.

  122. Guerini D, Coletto L, Carafoli E (2005) Exporting calcium from cells. Cell Calcium. 38 (3–4): 281–289. DOI: 10.1016/j.ceca.2005.06.032.

  123. Xie Y, Xiao F, Luo L, Zhong C (2014) Activation of autophagy protects against ROS-mediated mitochondria-dependent apoptosis in L-02 hepatocytes induced by Cr(VI). Cell. Physiol. Biochem. 33 (3): 705–716. DOI: 10.1159/000358646.

  124. Kaplan JH (2003) Biochemistry of Na,K-ATPase. Annu. Rev. Biochem. 71: 511–535. DOI: 10.1146/ANNUREV.BIOCHEM.71.102201.141218.

  125. Cheung WY (1980) Calmodulin plays a pivotal role in cellular regulation. Science  207 (4426): 19–27. DOI: 10.1126/SCIENCE.6243188.

  126. Zhang X, Wang Y, Chen M, Zeng M (2021) Hexavalent chromium-induced apoptosis in Hep3B cells is accompanied by calcium overload, mitochondrial damage, and AIF translocation. Ecotoxicol. Environ. Saf. 208 (111391): 1–10. DOI: 10.1016/j.ecoenv.2020.111391.

  127. Liang Q, Zhang Y, Zeng M, Guan L, Xiao Y, Xiao F (2018) The role of IP3R-SOCCs in Cr(vi)-induced cytosolic Ca 2+ overload and apoptosis in L-02 hepatocytes. Toxicol. Res. (Camb). 7 (3): 521–528. DOI: 10.1039/C8TX00029H.

  128. Finkel T, Deng CX, Mostoslavsky R (2009) Recent progress in the biology and physiology of sirtuins. Nature 460 (7255): 587–591. DOI: 10.1038/nature08197.

  129. Matsushima S, Sadoshima J (2015) The role of sirtuins in cardiac disease. Am. J. Physiol. - Hear. Circ. Physiol. 309 (9): H1375–H1389. DOI: 10.1152/AJPHEART.00053.2015.

  130. Xu S, Bai P, Jin ZG (2016) Sirtuins in cardiovascular health and diseases. Trends Endocrinol. Metab. 27 (10): 677–678. DOI: 10.1016/j.tem.2016.07.004.

  131. Han B, Li S, Lv Y, Yang D, Li J, Yang Q, Wu P, Lv Z, Zhang Z (2019) Dietary melatonin attenuates chromium-induced lung injury via activating the Sirt1/Pgc-1α/Nrf2 pathway. Food Funct. 10 (9): 5555–5565. DOI: 10.1039/C9FO01152H.

  132. Yang Q, Han B, Li S, Wang X, Wu P, Liu Y, Li J, Han B, Deng N, Zhang Z (2022) The link between deacetylation and hepatotoxicity induced by exposure to hexavalent chromium. J. Adv. Res. 35 : 129–140. DOI: 10.1016/j.jare.2021.04.002.

  133. Shil K, Pal S (2018) Metabolic adaptability in hexavalent chromium-treated renal tissue: an in vivo study. Clin. Kidney J. 11 (2): 222–229. DOI: 10.1093/ckj/sfx069.

  134. Tanno M, Kuno A, Horio Y, Miura T (2012) Emerging beneficial roles of sirtuins in heart failure. Basic Res. Cardiol. 107 (4): 1–14. DOI: 10.1007/s00395-012-0273-5.

  135. Dioum EM, Chen R, Alexander MS, Zhang Q, Hogg RT, Gerard RD, Garcia JA (2009) Regulation of hypoxia-inducible factor 2alpha signaling by the stress-responsive deacetylase sirtuin 1. Science (80-. ). 324 (5932): 1289–1293. DOI: 10.1126/SCIENCE.1169956.

  136. Daitoku H, Hatta M, Matsuzaki H, Aratani S, Ohshima T, Miyagishi M, Nakajima T, Fukamizu A (2004) Silent information regulator 2 potentiates Foxo1-mediated transcription through its deacetylase activity. Proc. Natl. Acad. Sci. U. S. A. 101 (27): 10042–10047. DOI: 10.1073/PNAS.0400593101.

  137. Sundaresan NR, Gupta M, Kim G, Rajamohan SB, Isbatan A, Gupta MP (2009) Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent antioxidant defense mechanisms in mice. J. Clin. Invest. 119 (9): 2758–2771. DOI: 10.1172/JCI39162.

  138. Alcendor RR, Gao S, Zhai P, Zablocki D, Holle E, Yu X, Tian B, Wagner T, Vatner SF, Sadoshima J (2007) Sirt1 regulates aging and resistance to oxidative stress in the heart. Circ. Res. 100 (10): 1512–1521. DOI: 10.1161/01.RES.0000267723.65696.4A.

  139. Hsu CP, Zhai P, Yamamoto T, Maejima Y, Matsushima S, Hariharan N, Shao D, Takagi H, Oka S, Sadoshima J (2010) Silent information regulator 1 protects the heart from ischemia/reperfusion. Circulation 122 (21): 2170–2182. DOI: 10.1161/CIRCULATIONAHA.110.958033.

  140. Ahmad MK, Syma S, Mahmood R (2011) Cr(VI) induces lipid peroxidation, protein oxidation and alters the activities of antioxidant enzymes in human erythrocytes. Biol. Trace Elem. Res. 144 (1–3): 426–435. DOI: 10.1007/s12011-011-9119-5.

  141. Kalahasthi RB, Rao RHR, Murthy RBK, Kumar MK (2006) Effect of chromium(VI) on the status of plasma lipid peroxidation and erythrocyte antioxidant enzymes in chromium plating workers. Chem. Biol. Interact. 164 (3): 192–199. DOI: 10.1016/j.cbi.2006.09.012.

  142. Wang YP, Zhou LS, Zhao YZ, Wang SW, Chen LL, Liu LX, Ling ZQ, Hu FJ, Sun YP, Zhang JY, Yang C, Yang Y, Xiong Y, Guan KL, Ye D (2014) Regulation of G6PD acetylation by SIRT2 and KAT9 modulates NADPH homeostasis and cell survival during oxidative stress. EMBO J. 33 (12): 1304–1320. DOI: 10.1002/EMBJ.201387224.

  143.  Cupo DY, Wetterhahn KE (1985) Modification of chromium(VI)-induced DNA damage by glutathione and cytochromes P-450 in chicken embryo hepatocytes. Proc. Natl. Acad. Sci. U. S. A. 82 (20): 6755–6759. DOI: 10.1073/pnas.82.20.6755.

  144. Finley LWS, Haas W, Desquiret-Dumas V, Wallace DC, Procaccio V, Gygi SP, Haigis MC (2011) Succinate dehydrogenase is a direct target of sirtuin 3 deacetylase activity. PLoS One 6 (8): 1–6. DOI: 10.1371/journal.pone.0023295.

  145. Someya S, Yu W, Hallows WC, Xu J, Vann JM, Leeuwenburgh C, Tanokura M, Denu JM, Prolla TA (2010) Sirt3 mediates reduction of oxidative damage and prevention of age-related hearing loss under caloric restriction. Cell 143 (5): 802–812. DOI: 10.1016/j.cell.2010.10.002.

  146. Lombard DB, Alt FW, Cheng H-L, Bunkenborg J, Streeper RS, Mostoslavsky R et al. (2007) Mammalian Sir2 homolog SIRT3 regulates global mitochondrial lysine acetylation. Mol. Cell. Biol. 27 (24): 8807–8814. DOI: 10.1128/MCB.01636-07.

  147. Giralt A, Villarroya F (2012) SIRT3, a pivotal actor in mitochondrial functions: metabolism, cell death and aging. Biochem. J. 444 (1): 1–10. DOI: 10.1042/BJ20120030.

  148. Finley LWS, Haigis MC (2012) Metabolic regulation by SIRT3: implications for tumorigenesis. Trends Mol. Med. 18 (9): 516–523. DOI: 10.1016/j.molmed.2012.05.004.

  149. Ahn B-H, Kim H-S, Song S, Lee IH, Liu J, Vassilopoulos A, Deng C-X, Finkel T (2008) A role for the mitochondrial deacetylase Sirt3 in regulating energy homeostasis. Proc. Natl. Acad. Sci. U.S.A. 105 (38): 14447–14452. DOI: 10.1073/pnas.0803790105.

  150. Clementino M, Kim D, Zhang Z (2019) Constitutive activation of NAD-dependent sirtuin 3 plays an important role in tumorigenesis of chromium(VI)-transformed cells. Toxicol. Sci. 169 (1): 224–234. DOI: 10.1093/toxsci/kfz032.

  151. Chauhan AS, Zhuang L, Gan B (2020) Spatial control of AMPK signaling at subcellular compartments. Crit. Rev. Biochem. Mol. Biol. 55 (1): 17–32. DOI: 10.1080/10409238.2020.1727840.

  152. Cantó C, Auwerx J (2009) PGC-1alpha, SIRT1 and AMPK, an energy sensing network that controls energy expenditure. Curr. Opin. Lipidol. 20 (2): 98–105. DOI: 10.1097/MOL.0B013E328328D0A4.

  153. Ojuka EO, Nolte LA, Holloszy JO (2000) Increased expression of GLUT-4 and hexokinase in rat epitrochlearis muscles exposed to AICAR in vitro. J. Appl. Physiol. 88 (3): 1072–1075. DOI: 10.1152/JAPPL.2000.88.3.1072.

  154. Winder WW, Holmes BF, Rubink DS, Jensen EB, Chen M, Holloszy JO (2000) Activation of AMP-activated protein kinase increases mitochondrial enzymes in skeletal muscle. J. Appl. Physiol. 88 (6): 2219–2226. DOI: 10.1152/JAPPL.2000.88.6.2219.

  155. Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre J, Doebber T, Fujii N, Musi N, Hirshman MF, Goodyear LJ, Moller DE (2001) Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Invest. 108 (8): 1167–1174. DOI: 10.1172/JCI13505.

  156. Ma Y, Li S, Ye S, Tang S, Hu D, Wei L, Xiao F (2021) Hexavalent chromium inhibits the formation of neutrophil extracellular traps and promotes the apoptosis of neutrophils via AMPK signaling pathway. Ecotoxicol. Environ. Saf. 223 (112614): 1–12. DOI: 10.1016/j.ecoenv.2021.112614.

  157. Wang X-X, Wang X-L, Tong M, Gan L, Chen H, Wu S, Chen J-X, Li R-L, Wu Y, Zhang H, Zhu Y, Li Y, He J, Wang M, Jiang W (2016) SIRT6 protects cardiomyocytes against ischemia/reperfusion injury by augmenting FoxO3α-dependent antioxidant defense mechanisms. Basic Res. Cardiol. 111 (2): 1–19. DOI: 10.1007/s00395-016-0531-z.

  158. Chiacchiera A F, Simone C (2010) The AMPK-FoxO3A axis as a target for cancer treatment. Cell Cycle 9 (6): 1091–1096. DOI: 10.4161/CC.9.6.11035.

  159. Mori S, Nada S, Kimura H, Tajima S, Takahashi Y, Kitamura A, Oneyama C, Okada M (2014) The mTOR pathway controls cell proliferation by regulating the FoxO3a transcription factor via SGK1 kinase. PLoS One 9 (2): 1–12. DOI: 10.1371/journal.pone.0088891.

  160. Cantó C, Auwerx J (2009) PGC-1α, SIRT1 and AMPK, an energy sensing network that controls energy expenditure. Curr. Opin. Lipidol. 20 (2): 98–105. DOI: 10.1097/MOL.0b013e328328d0a4.

  161. Liang H, Ward WF (2006) PGC-1α: a key regulator of energy metabolism. Adv. Physiol. Educ. 30 (4): 145–151. DOI: 10.1152/advan.00052.2006.

  162. Onishi Y, Ueha T, Kawamoto T, Hara H, Toda M, Harada R, Minoda M, Kurosaka M, Akisue T (2015) Regulation of mitochondrial proliferation by PGC-1α induces cellular apoptosis in musculoskeletal malignancies. Sci. Rep. 4 (3916): 1–8. DOI: 10.1038/srep03916.

  163. Jäer S, Handschin C, St-Pierre J, Spiegelman BM (2007) AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha. Proc. Natl. Acad. Sci. U. S. A. 104 (29): 12017–12022. DOI: 10.1073/PNAS.0705070104.

  164. Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P (2005) Nutrient control of glucose homeostasis through a complex of PGC-1α and SIRT1. Nature 434 (7029): 113–118. DOI: 10.1038/NATURE03354.

  165. Nemoto S, Fergusson MM, Finkel T (2005) SIRT1 functionally interacts with the metabolic regulator and transcriptional coactivator PGC-1α. J. Biol. Chem. 280 (16): 16456–16460. DOI: 10.1074/jbc.M501485200.

  166. Teyssier C, Ma H, Emter R, Kralli A, Stallcup MR (2005) Activation of nuclear receptor coactivator PGC-1α by arginine methylation. Genes Dev. 19 (12): 1466–1472. DOI: 10.1101/GAD.1295005.

  167. Furukawa A, Tada-Oikawa S, Kawanishi S, Oikawa S (2007) H2O2 accelerates cellular senescence by accumulation of acetylated p53 via decrease in the function of SIRT1 by NAD+ depletion. Cell. Physiol. Biochem. 20 (1–4): 045–054. DOI: 10.1159/000104152.

  168. Wang Y, Meng A, Zhou D (2004) Inhibition of phosphatidylinostol 3-kinase uncouples H2O2-induced senescent phenotype and cell cycle arrest in normal human diploid fibroblasts. Exp. Cell Res. 298 (1): 188–196. DOI: 10.1016/j.yexcr.2004.04.012.

  169. Sablina AA, Budanov A V., Ilyinskaya G V., Agapova LS, Kravchenko JE, Chumakov PM (2005) The antioxidant function of the p53 tumor suppressor. Nat. Med. 11 (12): 1306–1313. DOI: 10.1038/nm1320.

  170. Luo J, Li M, Tang Y, Laszkowska M, Roeder RG, Gu W (2004) Acetylation of p53 augments its site-specific DNA binding both in vitro and in vivo. Proc. Natl. Acad. Sci. 101 (8): 2259–2264. DOI: 10.1073/pnas.0308762101.

  171. Davidoff AM, Herndon JE, Glover NS, Kerns BJ, Pence JC, Lglehart JD MJ (1991) Relation between p53 overexpression and established prognostic factors in breast cancer. Surgery. 110 (2): 259–264.

  172. Ramadan MA, Shawkey AE, Rabeh MA, Abdellatif AO (2019) Expression of P53, BAX, and BCL-2 in human malignant melanoma and squamous cell carcinoma cells after tea tree oil treatment in vitro. Cytotechnology 71 (1): 461–473. DOI: 10.1007/S10616-018-0287-4.

  173. Gahl RF, Dwivedi P, Tjandra N (2016) Bcl-2 proteins bid and bax form a network to permeabilize the mitochondria at the onset of apoptosis. Cell Death Dis. 7 (10): e2424–e2424. DOI: 10.1038/cddis.2016.320.

  174. Li H, Shi J, Gao H, Yang X, Fu Y, Peng Y, Xia Y, Zhou D (2021) Hexavalent chromium causes apoptosis and autophagy by inducing mitochondrial dysfunction and oxidative stress in broiler cardiomyocytes. Biol. Trace Elem. Res. 200 (6): 1–10. DOI: 10.1007/S12011-021-02877-X/FIGURES/6.

  175. Schuler M, Bossy-Wetzel E, Goldstein JC, Fitzgerald P, Green DR (2000) p53 induces apoptosis by caspase activation through mitochondrial cytochrome c release. J. Biol. Chem. 275 (10): 7337–7342. DOI: 10.1074/JBC.275.10.7337.

  176. Narula J, Pandey P, Arbustini E, Haider N, Narula N, Kolodgie FD, Dal Bello B, Semigran MJ, Bielsa-Masdeu A, Dec GW, Israels S, Ballester M, Virmani R, Saxena S, Kharbanda S (1999) Apoptosis in heart failure: Release of cytochrome c from mitochondria and activation of caspase-3 in human cardiomyopathy. Proc. Natl. Acad. Sci. U. S. A. 96 (14): 8144–8149. DOI: 10.1073/PNAS.96.14.8144.

  177. McIlwain DR, Berger T, Mak TW (2013) Caspase functions in cell death and disease. Cold Spring Harb. Perspect. Biol. 5 (4): 1–28. DOI: 10.1101/CSHPERSPECT.A008656.

  178. Moorjani N, Ahmad M, Catarino P, Brittin R, Trabzuni D, Al-Mohanna F, Narula N, Narula J, Westaby S (2006) Activation of apoptotic caspase cascade during the ansition to pressure overload-induced heart failure. J. Am. Coll. Cardiol. 48 (7): 1451–1458. DOI: 10.1016/j.jacc.2006.05.065.

  179. Yang B, Ye D, Wang Y (2013) Caspase-3 as a therapeutic target for heart failure. Expert Opin. Ther. Targets 17 (3): 255–263. DOI: 10.1517/14728222.2013.745513.

  180. Li J, Zheng X, Ma X, Xu X, Du Y, Lv Q, Li X, Wu Y, Sun H, Yu L, Zhang Z (2019) Melatonin protects against chromium(VI)-induced cardiac injury via activating the AMPK/Nrf2 pathway. J. Inorg. Biochem. 197 (110698): 1–9. DOI: 10.1016/j.jinorgbio.2019.110698.

  181. Vakhrusheva O, Smolka C, Gajawada P, Kostin S, Boettger T, Kubin T, Braun T, Bober E (2008) Sirt7 increases stress resistance of cardiomyocytes and prevents apoptosis and inflammatory cardiomyopathy in mice. Circ. Res. 102 (6): 703–710. DOI: 10.1161/CIRCRESAHA.107.164558.

  182. Sivakumar KK, Stanley JA, Behlen JC, Wuri L, Dutta S, Wu J, Arosh JA, Banu SK (2022) Inhibition of Sirtuin-1 hyperacetylates p53 and abrogates Sirtuin-1-p53 interaction in Cr(VI)-induced apoptosis in the ovary. Reprod. Toxicol. 109 : 121–134. DOI: 10.1016/J.REPROTOX.2022.03.007.

  183. Wang S, Leonard SS, Ye J, Gao N, Wang L, Shi X (2004) Role of reactive oxygen species and Cr(VI) in Ras-mediated signal transduction. Mol. Cell. Biochem. 255 (1–2): 119–127. DOI: 10.1023/B:MCBI.0000007268.42733.53.

  184. Ganapathy S, Li P, Lafontant J, Xiong R, Yu T, Zhang G, Chen C (2017) Chromium IV exposure, via Src/Ras signaling, promotes cell transformation. Mol. Carcinog. 56 (7): 1808–1815. DOI: 10.1002/MC.22639.

  185. Castellano E, Downward J (2011) RAS interaction with PI3K: More than just another effector pathway. Genes Cancer 2 (3): 261–274. DOI: 10.1177/1947601911408079.

  186. Duronio V (2008) The life of a cell: apoptosis regulation by the PI3K/PKB pathway. Biochem. J. 415 (3): 333–344. DOI: 10.1042/BJ20081056.

  187. Cantley LC (2002) The phosphoinositide 3-Kinase pathway. Science 296 (5573): 1655–1657. DOI: 10.1126/science.296.5573.1655.

  188. Vivanco I, Sawyers CL (2002) The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat. Rev. Cancer 2 (7): 489–501. DOI: 10.1038/NRC839.

  189. Cox AD, Der CJ (2003) The dark side of Ras: regulation of apoptosis. Oncogene 22 (56): 8999–9006. DOI: 10.1038/SJ.ONC.1207111.

  190. Sundaresan NR, Vasudevan P, Zhong L, Kim G, Samant S, Parekh V, Pillai VB, Ravindra P V., Gupta M, Jeevanandam V, Cunningham JM, Deng C-X, Lombard DB, Mostoslavsky R, Gupta MP (2012) The sirtuin SIRT6 blocks IGF-Akt signaling and development of cardiac hypertrophy by targeting c-Jun. Nat. Med. 18 (11): 1643–1650. DOI: 10.1038/nm.2961.

  191. Son YO, Pratheeshkumar P, Wang L, Wang X, Fan J, Kim DH, Lee JY, Zhang Z, Lee JC, Shi X (2013) Reactive oxygen species mediate Cr(VI)-induced carcinogenesis through PI3K/AKT-dependent activation of GSK-3β/β-catenin signaling. Toxicol. Appl. Pharmacol. 271 (2): 239–248. DOI: 10.1016/J.TAAP.2013.04.036.

  192. Zhang Y, Zhang Y, Xiao Y, Zhong C, Xiao F (2019) Expression of clusterin suppresses Cr(VI)-induced premature senescence through activation of PI3K/AKT pathway. Ecotoxicol. Environ. Saf. 183 (109465): 1–10. DOI: 10.1016/J.ECOENV.2019.109465.

  193. Zhang Y, Xiao F, Liu X, Liu K, Zhou X, Zhong C (2017) Cr(VI) induces cytotoxicity in vitro through activation of ROS-mediated endoplasmic reticulum stress and mitochondrial dysfunction via the PI3K/Akt signaling pathway. Toxicol. Vitr. 41 : 232–244. DOI: 10.1016/j.tiv.2017.03.003.

  194. Fu S-C, Liu J-M, Lee K-I, Tang F-C, Fang K-M, Yang C-Y, Su C-C, Chen H-H, Hsu R-J, Chen Y-W (2020) Cr(VI) induces ROS-mediated mitochondrial-dependent apoptosis in neuronal cells via the activation of Akt/ERK/AMPK signaling pathway. Toxicol. Vitr. 65 (104795): 104795. DOI: 10.1016/j.tiv.2020.104795.

  195. Liang N, Li S, Liang Y, Ma Y, Tang S, Ye S, Xiao F (2021) Clusterin inhibits Cr(VI)-induced apoptosis via enhancing mitochondrial biogenesis through AKT-associated STAT3 activation in L02 hepatocytes. Ecotoxicol. Environ. Saf. 221 (112447): 1–11. DOI: 10.1016/j.ecoenv.2021.112447.

  196. Bowman T, Garcia R, Turkson J, Jove R (2000) STATs in oncogenesis. Oncogene 19 (21): 2474–2488. DOI: 10.1038/sj.onc.1203527.

  197. Bromberg J, Darnell JE (2000) The role of STATs in transcriptional control and their impact on cellular function. Oncogene  19 (21): 2468–2473. DOI: 10.1038/sj.onc.1203476.

  198. Elschami M, Scherr M, Philippens B, Gerardy-Schahn R (2013) Reduction of STAT3 expression induces mitochondrial dysfunction and autophagy in cardiac HL-1 cells. Eur. J. Cell Biol. 92 (1): 21–29. DOI: 10.1016/j.ejcb.2012.09.002.

  199. Liang Q, Xiao Y, Liu K, Zhong C, Zeng M, Xiao F (2018) Cr(VI)-induced autophagy protects l-02 hepatocytes from apoptosis through the ROS-AKT-mTOR pathway. Cell. Physiol. Biochem. 51 (4): 1863–1878. DOI: 10.1159/000495713.

  200. Chen A, Xiong L-J, Tong Y, Mao M (2013) Neuroprotective effect of brain-derived neurotrophic factor mediated by autophagy through the PI3K/Akt/mTOR pathway. Mol. Med. Rep. 8 (4): 1011–1016. DOI: 10.3892/mmr.2013.1628.

  201. Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M (2006) Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem. Biol. Interact. 160 (1): 1–40. DOI: 10.1016/j.cbi.2005.12.009.

  202. Zhao L, Song Y, Pu J, Guo J, Wang Y, Chen Z, Chen T, Gu Y, Jia G (2014) Effects of repeated Cr(VI) intratracheal instillation on club (Clara) cells and activation of nuclear factor-kappa B pathway via oxidative stress. Toxicol. Lett. 231 (1): 72–81. DOI: 10.1016/j.toxlet.2014.09.011.

  203. Wang L, Qiu J-G, He J, Liu W-J, Ge X, Zhou F-M, Huang Y-X, Jiang B-H, Liu L-Z (2019) Suppression of miR-143 contributes to overexpression of IL-6, HIF-1α and NF-κB p65 in Cr(VI)-induced human exposure and tumor growth. Toxicol. Appl. Pharmacol. 378 (114603): 1–7. DOI: 10.1016/j.taap.2019.114603.

  204. Zhang Y, Yang G, Huang S, Yang X, Yuan F, Song Y, Liu S, Yu X (2022) Regulation of Cr(VI)-induced premature senescence in l02 hepatocytes by ROS-Ca2+-NF-κB signaling. Oxid. Med. Cell. Longev. 2022 (7295224): 1–16. DOI: 10.1155/2022/7295224.

  205. Eferl R, Wagner EF (2003) AP-1: A double-edged sword in tumorigenesis. Nat. Rev. Cancer 3 (11): 859–868. DOI: 10.1038/nrc1209.

  206. Chen F, Ding M, Lu Y, Leonard SS, Vallyathan V, Castranova V, Shi X (2000) Participation of MAP kinase p38 and IkappaB kinase in chromium (VI)-induced NF-kappaB and AP-1 activation. J. Environ. Pathol. Toxicol. Oncol. 19 (3): 231–238.

  207. Fernández A, Ordóñez R, Reiter RJ, González‐Gallego J, Mauriz JL (2015) Melatonin and endoplasmic reticulum stress: relation to autophagy and apoptosis. J. Pineal Res. 59 (3): 292–307. DOI: 10.1111/jpi.12264.

  208. Minamino T, Kitakaze M (2010) ER stress in cardiovascular disease. J. Mol. Cell. Cardiol. 48 (6): 1105–1110. DOI: 10.1016/J.YJMCC.2009.10.026.

  209. Ge H, Li Z, Jiang L, Li Q, Geng C, Yao X, Shi X, Liu Y, Cao J (2019) Cr (VI) induces crosstalk between apoptosis and autophagy through endoplasmic reticulum stress in A549 cells. Chem. Biol. Interact. 298 : 35–42. DOI: 10.1016/j.cbi.2018.10.024.

  210. Liu MQ, Chen Z, Chen LX (2016) Endoplasmic reticulum stress: a novel mechanism and therapeutic target for cardiovascular diseases. Acta Pharmacol. Sin. 37 (4): 425–443. DOI: 10.1038/APS.2015.145.

  211. Liang Q, Zhang Y, Huang M, Xiao Y, Xiao F (2019) Role of mitochondrial damage in Cr(VI)‑induced endoplasmic reticulum stress in L‑02 hepatocytes. Mol. Med. Rep. 19 (2): 1256–1265. DOI: 10.3892/MMR.2018.9704.

  212. Liu YZ, Wang YX, Jiang CL (2017) Inflammation: The common pathway of stress-related diseases. Front. Hum. Neurosci. 11 (316): 1–11. DOI: 10.3389/FNHUM.2017.00316/BIBTEX.

  213. Alfaddagh A, Martin SS, Leucker TM, Michos ED, Blaha MJ, Lowenstein CJ, Jones SR, Toth PP (2020) Inflammation and cardiovascular disease: From mechanisms to therapeutics. Am. J. Prev. Cardiol. 4 (100130): 1–19. DOI: 10.1016/j.ajpc.2020.100130.

  214. Shrivastava R, Upreti RK, Seth PK, Chaturvedi UC (2002) Effects of chromium on the immune system. FEMS Immunol. Med. Microbiol. 34 (1): 1–7. DOI: 10.1016/S0928-8244(02)00345-0.

  215. Miller AH, Maletic V, Raison CL (2009) Inflammation and Its Discontents: The role of cytokines in the pathophysiology of major depression. Biol. Psychiatry 65 (9): 732–741. DOI: 10.1016/j.biopsych.2008.11.029.

  216. Zhang JM, An J (2007) Cytokines, inflammation and pain. Int. Anesthesiol. Clin. 45 (2): 27–37. DOI: 10.1097/AIA.0B013E318034194E.

  217. Lee SH, Kwon JY, Kim SY, Jung KA, Cho M La (2017) Interferon-gamma regulates inflammatory cell death by targeting necroptosis in experimental autoimmune arthritis. Sci. Rep. 7 (1): 1–9. DOI: 10.1038/s41598-017-09767-0.

  218. Wang Y, Wang L, Wang X, Cheng G, Xing Y, Zhang M, Zhang P, Liu J (2022) Inflammatory injury and mitophagy in the cock heart induced by the oral administration of hexavalent chromium. Biol. Trace Elem. Res. 200 (3): 1312–1320. DOI: 10.1007/s12011-021-02715-0.

  219. Hu G, Wang T, Liu J, Chen Z, Zhong L, Yu S, Zhao Z, Zhai M, Jia G (2017) Serum protein expression profiling and bioinformatics analysis in workers occupationally exposed to chromium (VI). Toxicol. Lett. 277: 76–83. DOI: 10.1016/J.TOXLET.2017.05.026.

  220. Hu G, Long C, Hu L, Zhang Y, Hong S, Zhang Q, Zheng P, Su Z, Xu J, Wang L, Gao X, Zhu X, Yuan F, Wang T, Yu S, Jia G (2022) Blood chromium exposure, immune inflammation and genetic damage: Exploring associations and mediation effects in chromate exposed population. J. Hazard. Mater. 425 (127769): 1–9. DOI: 10.1016/j.jhazmat.2021.127769.

  221. Dai L, Xu W, Li H, Frank JA, He C, Zhang Z, Chen G (2017) Effects of hexavalent chromium on mouse splenic T lymphocytes. Toxicol. Vitr. 45 (1): 166–171. DOI: 10.1016/j.tiv.2017.09.006.

  222. Hadrup S, Donia M, Thor Straten P (2012) Effector CD4 and CD8 T cells and their role in the tumor microenvironment. Cancer Microenviron. 6 (2): 123–133. DOI: 10.1007/S12307-012-0127-6.

  223. Lee HL, Jang JW, Lee SW, Yoo SH, Kwon JH, Nam SW, Bae SH, Choi JY, Han NI, Yoon SK (2019) Inflammatory cytokines and change of Th1/Th2 balance as prognostic indicators for hepatocellular carcinoma in patients treated with transarterial chemoembolization. Sci. Rep. 9 (1): 1–8. DOI: 10.1038/s41598-019-40078-8.

  224. Burastero SE, Paolucci C, Breda D, Ponti J, Munaro B, Sabbioni E (2006) Chromium (VI)-induced immunotoxicity and intracellular accumulation in human primary dendritic cells. Int. J. Immunopathol. Pharmacol. 19 (3): 581–591. DOI: 10.1177/039463200601900314.

  225. Eming SA, Krieg T, Davidson JM (2007) Inflammation in wound repair: Molecular and cellular mechanisms. J. Invest. Dermatol. 127 (3): 514–525. DOI: 10.1038/sj.jid.5700701.

  226. Freeman CM, Curtis JL, Chensue SW (2007) CC chemokine receptor 5 and CXC chemokine receptor 6 expression by lung CD8+ cells correlates with chronic obstructive pulmonary disease severity. Am. J. Pathol. 171 (3): 767–776. DOI: 10.2353/ajpath.2007.061177.

  227. Glaser U, Hochrainer D, Klppel H, Kuhnen H (1985) Low level chromium (VI) inhalation effects on alveolar macrophages and immune functions in Wistar rats. Arch. Toxicol. 57 (4): 250–256. DOI: 10.1007/BF00324787.

  228. Tau G, Rothman P (1999) Biologic functions of the IFN‐γ receptors. Allergy 54 (12): 1233–1251. DOI: 10.1034/J.1398-9995.1999.00099.X.

  229. Katiyar S, Awasthi SK, Srivastava JK (2009) Effect of chromium on the level of IL-12 and IFN-gamma in occupationally exposed workers. Sci. Total Environ. 407 (6): 1868–1874. DOI: 10.1016/j.scitotenv.2008.11.057.

  230. Tabas I, Lichtman AH (2017) Monocyte-macrophages and T cells in atherosclerosis. Immunity 47 (4): 621–634. DOI: 10.1016/j.immuni.2017.09.008.

  231. Teixeira LK, Fonseca BPF, Barboza BA, Viola JPB (2005) The role of interferon-gamma on immune and allergic responses. Mem. Inst. Oswaldo Cruz. 100 (suppl 1): 137–144. DOI: 10.1590/S0074-02762005000900024.

  232. Ovchinnikova O, Robertson A-KL, Wågsäter D, Folco EJ, Hyry M, Myllyharju J, Eriksson P, Libby P, Hansson GK (2009) T-cell activation leads to reduced collagen maturation in atherosclerotic plaques of Apoe−/− mice. Am. J. Pathol. 174 (2): 693–700. DOI: 10.2353/ajpath.2009.080561.

  233. Bennett MR, Sinha S, Owens GK (2016) Vascular smooth muscle cells in atherosclerosis. Circ. Res. 118 (4): 692–702. DOI: 10.1161/CIRCRESAHA.115.306361.

  234. Michael Garavito R, Malkowski MG, DeWitt DL (2002) The structures of prostaglandin endoperoxide H synthases-1 and -2. Prostaglandins Other Lipid Mediat. 6869 : 129–152. DOI: 10.1016/S0090-6980(02)00026-6.

  235. Patrono C (2016) Cardiovascular effects of cyclooxygenase-2 inhibitors: a mechanistic and clinical perspective. Br. J. Clin. Pharmacol. 82 (4): 957–964. DOI: 10.1111/bcp.13048.

  236. Smith WL, Langenbach R (2001) Why there are two cyclooxygenase isozymes. J. Clin. Invest. 107 (12): 1491–1495. DOI: 10.1172/JCI13271.

  237. Grosser T (2005) Biological basis for the cardiovascular consequences of COX-2 inhibition: therapeutic challenges and opportunities. J. Clin. Invest. 116 (1): 4–15. DOI: 10.1172/JCI27291.

  238. Chen P, Geng N, Zhou D, Zhu Y, Xu Y, Liu K, Liu Y, Liu J (2019) The regulatory role of COX-2 in the interaction between Cr(VI)-induced endoplasmic reticulum stress and autophagy in DF-1 cells. Ecotoxicol. Environ. Saf. 170: 112–119. DOI: 10.1016/J.ECOENV.2018.11.120.

  239. Klionsky DJ, Abdel-Aziz AK, Abdelfatah S, Abdellatif M, Abdoli A, Abel S et al. (2021) Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 17 (1): 1–382.

  240. Yorimitsu T, Nair U, Yang Z, Klionsky DJ (2006) Endoplasmic reticulum stress triggers autophagy. J. Biol. Chem. 281 (40): 30299–30304. DOI: 10.1074/JBC.M607007200.

  241. Murrow L, Debnath J (2013) Autophagy as a stress-response and quality-control mechanism: Implications for cell injury and human disease. Annu. Rev. Pathol. 8 : 105–137. DOI: 10.1146/ANNUREV-PATHOL-020712-163918.

  242. Luo C, He M, Bohlin L (2005) Is COX-2 a perpetrator or a protector? Selective COX-2 inhibitors remain controversial. Acta Pharmacol. Sin. 26 (8): 926–933. DOI: 10.1111/j.1745-7254.2005.00150.x.

  243. Angeli JK, Cruz Pereira CA, de Oliveira Faria T, Stefanon I, Padilha AS, Vassallo DV (2013) Cadmium exposure induces vascular injury due to endothelial oxidative stress: the role of local angiotensin II and COX-2. Free Radic. Biol. Med. 65 : 838–848. DOI: 10.1016/j.freeradbiomed.2013.08.167.

  244. Liu T, Zhang L, Joo D, Sun S-C (2017) NF-κB signaling in inflammation. Signal Transduct. Target Ther. 2 (17023): 1–9. DOI: 10.1038/sigtrans.2017.23.

  245. Anandasadagopan SK, Sundaramoorthy C, Pandurangan AK, Nagarajan V, Srinivasan K, Ganapasam S (2017) S-Allyl cysteine alleviates inflammation by modulating the expression of NF-ΰB during chromium (VI)-induced hepatotoxicity in rats. Hum. Exp. Toxicol. 36 (11): 1186–1200. DOI: 10.1177/0960327116680275.

  246. Gordon JW, Shaw JA, Kirshenbaum LA (2011) Multiple facets of NF-κB in the heart. Circ. Res. 108 (9): 1122–1132. DOI: 10.1161/CIRCRESAHA.110.226928.

  247. Zhao D, Yu Y, Shen Y, Liu Q, Zhao Z, Sharma R, Reiter RJ (2019) Melatonin synthesis and function: Evolutionary history in animals and plants. Front. Endocrinol. (Lausanne). 10 (249): 1–16. DOI: 10.3389/FENDO.2019.00249/BIBTEX.

  248. Reiter RJ, Tan DX, Zhou Z, Cruz MHC, Fuentes-Broto L, Galano A (2015) Phytomelatonin: Assisting plants to survive and thrive. Molecules 20 (4): 7396–7437. DOI: 10.3390/molecules20047396.

  249. Reiter RJ, Mayo JC, Tan D-X, Sainz RM, Alatorre-Jimenez M, Qin L (2016) Melatonin as an antioxidant: under promises but over delivers. J. Pineal Res. 61 (3): 253–278. DOI: 10.1111/jpi.12360.

  250. Sharman EH, Bondy SC (2016) Melatonin: A safe nutraceutical and clinical agent. Nutraceuticals: efficacy, safety and toxicity. Elsevier; 2016. 501–509 p. DOI: 10.1016/B978-0-12-802147-7.00036-X.

  251. Romero A, Ramos E, De Los Ríos C, Egea J, Del Pino J, Reiter RJ (2014) A review of metal-catalyzed molecular damage: Protection by melatonin. J. Pineal Res. 56 (4): 343–370. DOI: 10.1111/jpi.12132.

  252. Acuña-Castroviejo D, Escames G, Venegas C, Díaz-Casado ME, Lima-Cabello E, López LC, Rosales-Corral S, Tan D-X, Reiter RJ (2014) Extrapineal melatonin: sources, regulation, and potential functions. Cell. Mol. Life Sci. 71 (16): 2997–3025. DOI: 10.1007/s00018-014-1579-2.

  253. Sun H, Gusdon AM, Qu S (2016) Effects of melatonin on cardiovascular diseases. Curr. Opin. Lipidol. 27 (4): 408–413. DOI: 10.1097/MOL.0000000000000314.

  254. Pandi-Perumal SR, BaHammam AS, Ojike NI, Akinseye OA, Kendzerska T, Buttoo K, Dhandapany PS, Brown GM, Cardinali DP (2016) Melatonin and human cardiovascular disease: J. Cardiovasc. Pharmacol. Ther. 22 (2): 122–132. DOI: 10.1177/1074248416660622.

  255. Sewerynek E (2002) Melatonin and the cardiovascular system. Neuroendocrinol. Lett. 4 (suppl 1): 79–83.

  256. Liu J, Clough SJ, Hutchinson AJ, Adamah-Biassi EB, Popovska-Gorevski M, Dubocovich ML (2016) MT 1 and MT 2 melatonin receptors: A therapeutic perspective. Annu. Rev. Pharmacol. Toxicol. 56 (1): 361–383. DOI: 10.1146/annurev-pharmtox-010814-124742.

  257. Hardeland R, Cardinali DP, Srinivasan V, Spence DW, Brown GM, Pandi-Perumal SR (2011) Melatonin—A pleiotropic, orchestrating regulator molecule. Prog. Neurobiol. 93 (3): 350–384. DOI: 10.1016/j.pneurobio.2010.12.004.

  258. Grossini E, Molinari C, Uberti F, Mary DASG, Vacca G, Caimmi PP (2011) Intracoronary melatonin increases coronary blood flow and cardiac function through β-adrenoreceptors, MT1/MT2 receptors, and nitric oxide in anesthetized pigs. J. Pineal Res. 51 (2): 246–257. DOI: 10.1111/J.1600-079X.2011.00886.X.

  259. Han D, Wang Y, Chen J, Zhang J, Yu P, Zhang R, Li S, Tao B, Wang Y, Qiu Y, Xu M, Gao E, Cao F (2019) Activation of melatonin receptor 2 but not melatonin receptor 1 mediates melatonin-conferred cardioprotection against myocardial ischemia/reperfusion injury. J. Pineal Res. 67 (1): e12571. DOI: 10.1111/JPI.12571.

  260. Ekmekcioglu C, Thalhammer T, Humpeler S, Mehrabi MR, Glogar HD, Hölzenbein T, Markovic O, Leibetseder VJ, Strauss-Blasche G, Marktl W (2003) The melatonin receptor subtype MT2 is present in the human cardiovascular system. J. Pineal Res. 35 (1): 40–44. DOI: 10.1034/J.1600-079X.2003.00051.X.

  261. Scheer FAJL, Van Montfrans GA, Van Someren EJW, Mairuhu G, Buijs RM (2004) Daily nighttime melatonin reduces blood pressure in male patients with essential hypertension. Hypertension 43 (2): 192–197. DOI: 10.1161/01.HYP.0000113293.15186.3B.

  262. Grossman E, Laudon M, Yalcin R, Zengil H, Peleg E, Sharabi Y, Kamari Y, Shen-Orr Z, Zisapel N (2006) Melatonin reduces night blood pressure in patients with nocturnal hypertension. Am. J. Med. 119 (10): 898–902. DOI: 10.1016/J.AMJMED.2006.02.002.

  263. Katsi V, Karagiorgi I, Makris T, Papavasileiou M, Androulakis AE, Tsioufis C, Tousoulis D, Stefanadis C, Kallikazaros IE (2012) The role of melatonin in hypertension: A brief review. Cardiovasc. Endocrinol. 1 (1): 13–18. DOI: 10.1097/XCE.0B013E3283565783.

  264. Simko F, Pechanova O (2009) Potential roles of melatonin and chronotherapy among the new trends in hypertension treatment. J. Pineal Res. 47 (2): 127–133. DOI: 10.1111/J.1600-079X.2009.00697.X.

  265. Paulis L, Šimko F (2007) Blood pressure modulation and cardiovascular protection by melatonin: potential mechanisms behind. Physiol. Res. 56 (6): 671–684. DOI: 10.33549/PHYSIOLRES.931236.

  266. Ray CA (2003) Melatonin attenuates the sympathetic nerve responses to orthostatic stress in humans. J. Physiol. 551 (Pt 3): 1043–1048. DOI: 10.1113/JPHYSIOL.2003.043182.

  267. Nishiyama K, Yasue H, Moriyama Y, Tsunoda R, Ogawa H, Yoshimura M, Kugiyama K (2001) Acute effects of melatonin administration on cardiovascular autonomic regulation in healthy men. Am. Heart J. 141 (5): 1–5. DOI: 10.1067/MHJ.2001.114368.

  268. Chan TY, Tang PL (1996) Characterization of the antioxidant effects of melatonin and related indoleamines in vitro. J. Pineal Res. 20 (4): 187–191. DOI: 10.1111/j.1600-079X.1996.tb00257.x.

  269. Mayo JC, Tan DX, Sainz RM, Lopez-Burillo S, Reiter RJ (2003) Oxidative damage to catalase induced by peroxyl radicals: Functional protection by melatonin and other antioxidants. Free Radic. Res. 37 (5): 543–553. DOI: 10.1080/1071576031000083206.

  270. Marshall KA, Reiter RJ, Poeggeler B, Aruoma OI, Halliwell B (1996) Evaluation of the antioxidant activity of melatonin in vitro. Free Radic. Biol. Med. 21 (3): 307–315. DOI: 10.1016/0891-5849(96)00046-9.

  271. Stasica P, Paneth P, Rosiak JM (2000) Hydroxyl radical reaction with melatonin molecule: A computational study. J. Pineal Res. 29 (2): 125–127. DOI: 10.1034/j.1600-079X.2000.290209.x.

  272. Zang LY, Cosma G, Gardner H, Vallyathan V (1998) Scavenging of reactive oxygen species by melatonin. Biochim. Biophys. Acta - Gen. Subj. 1425 (3): 469–477. DOI: 10.1016/S0304-4165(98)00099-3.

  273. Banerjee S, Joshi N, Mukherjee R, Singh PK, Baxi D, Ramachandran A V. (2017) Melatonin protects against chromium (VI) induced hepatic oxidative stress and toxicity: Duration dependent study with realistic dosage. Interdiscip. Toxicol. 10 (1): 20–29. DOI: 10.1515/intox-2017-0003.

  274. Lee SH, Kim DH, Kuzmanov U, Gramolini AO (2021) Membrane proteomic profiling of the heart: Past, present, and future. Am. J. Physiol. - Hear. Circ. Physiol. 320 (1): H417–H423. DOI: 10.1152/AJPHEART.00659.2020/ASSET/IMAGES/LARGE/AJ-AHRT200097F003.JPEG.

  275. Ceraulo L, Ferrugia M, Tesoriere L, Segreto S, Livrea MA, Turco Liveri V (1999) Interactions of melatonin with membrane models: Portioning of melatonin in AOT and lecithin reversed micelles. J. Pineal Res. 26 (2): 108–112. DOI: 10.1111/j.1600-079X.1999.tb00570.x.

  276. Livrea MA, Tesoriere L, Arpa D, Morreale M (1997) Reaction of melatonin with lipoperoxyl radicals in phospholipid bilayers. Free Radic. Biol. Med. 23 (5): 706–711. DOI: 10.1016/S0891-5849(97)00018-X.

  277. Reiter RJ, Acuña-Castroviejo D, Tan DX, Burkhardt S (2001) Free radical-mediated molecular damage: Mechanisms for the protective actions of melatonin in the central nervous system. Ann. N. Y. Acad. Sci. 939: 200–215. DOI: 10.1111/j.1749-6632.2001.tb03627.x.

  278. Daniels WMU, Van Rensburg SJ, Van Zyl JM, Taljaard JJF (1998) Melatonin prevents β-amyloid-induced lipid peroxidation. J. Pineal Res. 24 (2): 78–82. DOI: 10.1111/j.1600-079X.1998.tb00370.x.

  279. García JJ, Reiter RJ, Guerrero JM, Escames G, Yu BP, Oh CS, Muñoz-Hoyos A (1997) Melatonin prevents changes in microsomal membrane fluidity during induced lipid peroxidation. FEBS Lett. 408 (3): 297–300. DOI: 10.1016/S0014-5793(97)00447-X.

  280. Aryafar T, Amini P, Rezapoor S, Shabeeb DD, Eleojo Musa A, Najafi M, Shirazi AI (2021) Modulation of radiation-induced NADPH oxidases in rat’s heart tissues by melatonin. J. Biomed. Phys. Eng. 11 (4): 465–472. DOI: 10.31661/JBPE.V0I0.1094.

  281. Lv Y, Li T, Yang M, Su L, Zhu Z, Zhao S, Zeng W, Zheng Y (2021) Melatonin attenuates chromium (VI)-induced spermatogonial stem cell/progenitor mitophagy by restoration of METTL3-mediated RNA N6-methyladenosine modification. Front. Cell Dev. Biol. 9 (684398): 1–19. DOI: 10.3389/FCELL.2021.684398.

  282. Seeger H, Mueck AO, Lippert TH (1997) Effect of melatonin and metabolites on copper-mediated oxidation of low density lipoprotein. Br. J. Clin. Pharmacol. 44 (3): 283–284. DOI: 10.1046/J.1365-2125.1997.00648.X.

  283. Abuja PM, Liebmann P, Hayn M, Schauenstein K, Esterbauer H (1997) Antioxidant role of melatonin in lipid peroxidation of human LDL. FEBS Lett. 413 (2): 289–293. DOI: 10.1016/S0014-5793(97)00918-6.

  284. Kelly MR, Loo G (1997) Melatonin inhibits oxidative modification of human low-density lipoprotein. J. Pineal Res. 22 (4): 203–209. DOI: 10.1111/J.1600-079X.1997.TB00323.X.

  285. Dominguez-Rodriguez A, Abreu-Gonzalez P, Garcia-Gonzalez M, Ferrer-Hita J, Vargas M, Reiter RJ (2005) Elevated levels of oxidized low-density lipoprotein and impaired nocturnal synthesis of melatonin in patients with myocardial infarction. Atherosclerosis 180 (1): 101–105. DOI: 10.1016/j.atherosclerosis.2004.11.003.

  286. Hoyos M, Guerrero JM, Perez-Cano R, Olivan J, Fabiani F, Garcia-Pergañeda A, Osuna C (2002) Serum cholesterol and lipid peroxidation are decreased by melatonin in diet-induced hypercholesterolemic rats. J. Pineal Res. 28 (3): 150–155. DOI: 10.1034/j.1600-079X.2001.280304.x.

  287. Mullerwieland D, Behnke B, Koopmann K, Krone W (1994) Melatonin inhibits LDL receptor activity and cholesterol-synthesis in freshly isolated human mononuclear leukocytes. Biochem. Biophys. Res. Commun. 203 (1): 416–421. DOI: 10.1006/bbrc.1994.2198.

  288. Haghjooyjavanmard S, Nematbakhsh M, Monajemi A, Soleimani M (2008) von Willebrand factor, C-reactive protein, nitric oxide, and vascular endothelial growth factor in a dietary reversal model of hypercholesterolemia in rabbit. Biomed. Pap. Med. Fac. Univ. Palacky. Olomouc. Czech. Repub. 152 (1): 91–95. DOI: 10.5507/BP.2008.014.

  289. Javanmard SH, Heshmat-Ghahdarijani K, Mirmohammad-Sadeghi M, Sonbolestan SA, Ziayi A (2016) The effect of melatonin on endothelial dysfunction in patient undergoing coronary artery bypass grafting surgery. Adv. Biomed. Res. 5 (1): 174–179. DOI: 10.4103/2277-9175.194801.

  290. Li P, Xie C, Zhong J, Guo Z, Guo K, Tu Q (2021) Melatonin attenuates ox-LDL-induced endothelial dysfunction by reducing ER stress and inhibiting JNK/Mff signaling. Oxid. Med. Cell. Longev. 2021 (5589612): 1–10. DOI: 10.1155/2021/5589612.

  291. Hardeland R (2021) Melatonin, its metabolites and their interference with reactive nitrogen compounds. Molecules 26 (4105): 1–28. DOI: 10.3390/molecules26134105.

  292. Fabrizio RL, Gaia F, Eleonora F, Claudia R, Stefania C, Claudio L, Rita R (2013) Vascular endothelial cells and dysfunctions: role of melatonin. Front. Biosci. (Elite Ed). 5 (1): 119–129. DOI: 10.2741/E601.

  293. Suwanjang W, Abramov AY, Charngkaew K, Govitrapong P, Chetsawang B (2016) Melatonin prevents cytosolic calcium overload, mitochondrial damage and cell death due to toxically high doses of dexamethasone-induced oxidative stress in human neuroblastoma SH-SY5Y cells. Neurochem. Int. 97: 34–41. DOI: 10.1016/J.NEUINT.2016.05.003.

  294. Chen WR, Zhou YJ, Yang JQ, Liu F, Wu XP, Sha Y (2020) Melatonin attenuates calcium deposition from vascular smooth muscle cells by activating mitochondrial fusion and mitophagy via an AMPK/OPA1 signaling pathway. Oxid. Med. Cell. Longev. 2020 (5298483): 1–23. DOI: 10.1155/2020/5298483.

  295. Chen LD, Kumar P, Reiter RJ, Tan DX, Manchester LC, Chambers JP, Poeggeler B, Saarela S (1994) Melatonin prevents the suppression of cardiac Ca(2+)-stimulated ATPase activity induced by alloxan. Am. J. Physiol. 267 (1): E57-62. DOI: 10.1152/AJPENDO.1994.267.1.E57.

  296. Rodriguez C, Martín V, Herrera F, García-Santos G, Rodriguez-Blanco J, Casado-Zapico S, Sánchez-Sánchez A, Suárez S, Puente-Moncada N, Anítua M, Antolín I (2013) Mechanisms involved in the pro-apoptotic effect of melatonin in cancer cells. Int. J. Mol. Sci. 14 (4): 6597–6613. DOI: 10.3390/ijms14046597.

  297. Hardeland R (2018) Melatonin and inflammation—Story of a double‐edged blade. Wiley Online Libr. 65 (4): 1–23. DOI: 10.1111/jpi.12525.

  298. Mayo JC, Sainz RM, González Menéndez P, Cepas V, Tan D-X, Reiter RJ (2017) Melatonin and sirtuins: A “not-so unexpected” relationship. J. Pineal Res. 62 (2): 1–17. DOI: 10.1111/jpi.12391.

  299. Rangarajan P, Karthikeyan A, Lu J, Ling E-A, Dheen ST (2015) Sirtuin 3 regulates Foxo3a-mediated antioxidant pathway in microglia. Neuroscience 311 : 398–414. DOI: 10.1016/j.neuroscience.2015.10.048.

  300. Favero G, Franco C, Stacchiotti A, Rodella LF, Rezzani R (2020) Sirtuin1 role in the melatonin protective effects against obesity-related heart injury. Front. Physiol. 11 (103): 1–11. DOI: 10.3389/fphys.2020.00103.

  301. Zhang C-L, Feng H, Li L, Wang J-Y, Wu D, Hao Y-T, Wang Z, Zhang Y, Wu L-L (2017) Globular CTRP3 promotes mitochondrial biogenesis in cardiomyocytes through AMPK/PGC-1α pathway. Biochim. Biophys. Acta - Gen. Subj. 1861 (1): 3085–3094. DOI: 10.1016/j.bbagen.2016.10.022.

  302. National Center for Biotechnology Information (US). Genes and Disease. (1998) The p53 tumor suppressor protein. https://www.ncbi.nlm.nih.gov/books/NBK22268/.

  303. Peach MJ (1977) Renin-angiotensin system: biochemistry and mechanisms of action. Physiol. Rev. 57 (2): 313–370. DOI: 10.1152/PHYSREV.1977.57.2.313.

  304. Campos LA, Cipolla-Neto J, Amaral FG, Michelini LC, Bader M, Baltatu OC (2013) The angiotensin-melatonin axis. Int. J. Hypertens. 2013 (521783): 1–7. DOI: 10.1155/2013/521783.

  305. Ohashi N, Ishigaki S, Isobe S (2019) The pivotal role of melatonin in ameliorating chronic kidney disease by suppression of the renin–angiotensin system in the kidney. Hypertens. Res. 42 (6): 761–768. DOI: 10.1038/s41440-018-0186-2.

  306. Chen K, Zhu P, Chen W, Luo K, Shi XJ, Zhai W (2021) Melatonin inhibits proliferation, migration, and invasion by inducing ROS-mediated apoptosis via suppression of the PI3K/Akt/mTOR signaling pathway in gallbladder cancer cells. Aging (Albany. NY). 13 (18): 22502–22515. DOI: 10.18632/AGING.203561.

  307. Chen X, Hao A, Li X, Du Z, Li H, Wang H, Yang H, Fang Z (2016) Melatonin inhibits tumorigenicity of glioblastoma stem-like cells via the AKT-EZH2-STAT3 signaling axis. J. Pineal Res. 61 (2): 208–217. DOI: 10.1111/jpi.12341.

  308. Kong PJ, Byun JS, Lim SY, Lee JJ, Hong SJ, Kwon KJ, Kim SS (2008) Melatonin induces Akt phosphorylation through melatonin receptor- and PI3K-dependent pathways in primary astrocytes. Korean J. Physiol. Pharmacol. 12 (2): 37–41. DOI: 10.4196/KJPP.2008.12.2.37.

  309. Kilic U, Caglayan AB, Beker MC, Gunal MY, Caglayan B, Yalcin E, Kelestemur T, Gundogdu RZ, Yulug B, Yılmaz B, Kerman BE, Kilic E (2017) Particular phosphorylation of PI3K/Akt on Thr308 via PDK-1 and PTEN mediates melatonin’s neuroprotective activity after focal cerebral ischemia in mice. Redox Biol. 12 : 657–665. DOI: 10.1016/j.redox.2017.04.006.

  310. Fan C, Feng J, Tang C, Zhang Z, Feng Y, Duan W, Zhai M, Yan Z, Zhu L, Feng L, Zhu H, Luo E (2020) Melatonin suppresses ER stress-dependent proapoptotic effects via AMPK in bone mesenchymal stem cells during mitochondrial oxidative damage. Stem Cell Res. Ther. 11 (1): 1–22. DOI: 10.1186/S13287-020-01948-5/FIGURES/9.

  311. Chen Y, Zhao Q, Sun Y, Jin Y, Zhang J, Wu J (2018) Melatonin induces anti-inflammatory effects viaendoplasmic reticulum stress in RAW264.7 macrophages. Mol. Med. Rep. 17 (4): 6122–6129. DOI: 10.3892/MMR.2018.8613/HTML.

  312. Ozgur R, Uzilday B, Turkan I, Sekmen AH (2017) The Effects of melatonin on transcriptional profile of unfolded protein response genes under endoplasmic reticulum stress in arabidopsis thaliana. Plant Mol. Biol. Report. 35 (2): 188–202. DOI: 10.1007/s11105-016-1015-x.

  313. Aouichat S, Navarro-Alarcon M, Alarcón-Guijo P, Salagre D, Ncir M, Zourgui L, Agil A (2021) Melatonin improves endoplasmic reticulum stress-mediated IRE1α pathway in Zücker diabetic fatty rat. Pharmaceuticals 14 (232): 1–14. DOI: 10.3390/PH14030232.

  314. Carrillo-Vico A, Lardone P, Álvarez-Sánchez N, Rodríguez-Rodríguez A, Guerrero J (2013) Melatonin: Buffering the immune system. Int. J. Mol. Sci. 14 (4): 8638–8683. DOI: 10.3390/ijms14048638.

  315. Annane PD, Bellissant PE, Cavaillon JM (2005) Septic shock. Lancet 365 (9453): 63–78. DOI: 10.1016/S0140-6736(04)17667-8.

  316. Haddadi GH, Fardid R (2015) Oral administration of melatonin modulates the expression of tumor necrosis factor-α (TNF-α) gene in irradiated rat cervical spinal cord. Reports Pract. Oncol. Radiother. 20 (2): 123–127. DOI: 10.1016/j.rpor.2014.11.003.

  317. Yu GM, Kubota H, Okita M, Maeda T (2017) The anti-inflammatory and antioxidant effects of melatonin on LPS-stimulated bovine mammary epithelial cells. PLoS One 12 (5): 1–17. DOI: 10.1371/JOURNAL.PONE.0178525.

  318. Mannino G, Caradonna F, Cruciata I, Lauria A, Perrone A, Gentile C (2019) Melatonin reduces inflammatory response in human intestinal epithelial cells stimulated by interleukin-1β. J. Pineal Res. 67 (3): 1–28. DOI: 10.1111/JPI.12598.

  319. Lowes DA, Webster NR, Murphy MP, Galley HF (2013) Antioxidants that protect mitochondria reduce interleukin-6 and oxidative stress, improve mitochondrial function, and reduce biochemical markers of organ dysfunction in a rat model of acute sepsis. Br. J. Anaesth. 110 (3): 472–480. DOI: 10.1093/bja/aes577.

  320. Xia MZ, Liang YL, Wang H, Chen X, Huang YY, Zhang ZH, Chen YH, Zhang C, Zhao M, Xu DX, Song LH (2012) Melatonin modulates TLR4-mediated inflammatory genes through MyD88- and TRIF-dependent signaling pathways in lipopolysaccharide-stimulated RAW264.7 cells. J. Pineal Res. 53 (4): 325–334. DOI: 10.1111/J.1600-079X.2012.01002.X.

  321. Yoo YM, Jang SK, Kim GH, Park JY, Joo SS (2016) Pharmacological advantages of melatonin in immunosenescence by improving activity of T lymphocytes. J. Biomed. Res. 30 (4): 314–321. DOI: 10.7555/JBR.30.2016K0010.

  322. Luo J, Zhang Z, Sun H, Song J, Chen X, Huang J, Lin X, Zhou R (2020) Effect of melatonin on T/B cell activation and immune regulation in pinealectomy mice. Life Sci. 242 (117191): 1–11. DOI: 10.1016/j.lfs.2019.117191.

  323. Withyachumnarnkul B, Nonaka KO, Santana C, Attia AM, Reiter RJ (1990) Interferon-gamma modulates melatonin production in rat pineal glands in organ culture. J. Interferon Res. 10 (4): 403–411. DOI: 10.1089/JIR.1990.10.403.

  324. Maurino SG, Gonzalez-Haba MG, Calvo JR RM (1997) Melatonin enhances IL-2, IL-6, and IFN-γ production by human circulating CD4+ cells: A possible nuclear receptor-mediated mechanism involving T helper type 1 lymphocytes and monocytes. J. Immunol. 159 (2): 574–581.

  325. Inserra P, Zhang Z, Ardestani SK, Araghi-Niknam M, Liang B, Jiang S, Shaw D, Molitor M, Elliott K, Watson RR (1998) Modulation of cytokine production by dehydroepiandrosterone (DHEA) plus melatonin (MLT) supplementation of old mice. Exp. Biol. Med. 218 (1): 76–82. DOI: 10.3181/00379727-218-44270.

  326. Petrovsky N HL (1997) Diurnal rhythmicity of human cytokine production: A dynamic disequilibrium in T helper cell type 1/T helper cell type 2 balance? J. Immunol. 158 (11): 5163–5168.

  327. Deng WG, Tang ST, Tseng HP, Wu KK (2006) Melatonin suppresses macrophage cyclooxygenase-2 and inducible nitric oxide synthase expression by inhibiting p52 acetylation and binding. Blood 108 (2): 518–524. DOI: 10.1182/BLOOD-2005-09-3691.

  328. Figueroa EG, Gonzaléz-Candia A, Villanueva CA, Ebensperger G, Reyes R V., Llanos AJ, Herrera EA (2021) Beneficial effects of melatonin on prostanoids pathways in pulmonary hypertensive neonates. Vascul. Pharmacol. 138 (106853): 1–11. DOI: 10.1016/j.vph.2021.106853.

  329. Aguilar SA, Arias P V., Canquil I, Ebensperger G, Llanos AJ, Reyes R V., González-Candia A, Herrera EA (2019) Melatonin modulates the expression of pulmonary prostanoids. Rev. Med. Chil. 147 (3): 281–288. DOI: 10.4067/S0034-98872019000300281.

  330. García JA, Volt H, Venegas C, Doerrier C, Escames G, López LC, Acuña-Castroviejo D (2015) Disruption of the NF-κB/NLRP3 connection by melatonin requires retinoid-related orphan receptor-α and blocks the septic response in mice. FASEB J. 29 (9): 3863–3875. DOI: 10.1096/FJ.15-273656.

 

CCBY.png

This work is licensed under a Creative Commons Attribution 4.0 International License