Skip to main content
Health Science Reports logoLink to Health Science Reports
. 2025 Sep 22;8(9):e71279. doi: 10.1002/hsr2.71279

Emerging Roles of Metallothioneins in Human Pathophysiology: A Review

Ousman Mohammed 1,2, Abdisa Tufa 1, Solomon Tebeje Gizaw 1,
PMCID: PMC12453973  PMID: 40994777

ABSTRACT

Background

Metallothioneins (MTs) are small molecular‐weight proteins that either bind or release essential transition metals, depending on a specific cell situation. There are 4 major isoforms of MTs (1–4), of which MT1/2 is found in every cell in the body.

Aim

To provide a comprehensive discussion on the role of metallothionein in human pathophysiology.

Methods

This review was conducted using a variety of search engines, including Google Scholar, PubMed Central, Scopus, Web of Science, and others.

Results

The antioxidant property of MT is enhanced based on the availability of zinc. MTs reduce Pro‐inflammatory cytokine production and shift the Th17/Treg balance, ultimately alleviating inflammatory diseases. In neurodegenerative diseases, MTs interact with protein aggregates and modulate their formation and toxicity. The MT's molecular processes in neurodegenerative illnesses include metal ion control, antioxidant activity, and anti‐inflammatory actions. In cancer, MT acts as a tumor suppressor in the early stages and promotes tumor progression in the later stages. Only a few MT isoform‐targeting approaches have been approved for use as therapeutic drugs.

Conclusion

Metallothioneins play vital roles in controlling oxidative stress, immunity, metal homeostasis and protein aggregate for many diseases. Their dual roles as defenders in inflammation and neurodegeneration, and as suppressors and facilitators in cancer, highlight the complex nature of their biological role.

Keywords: cancer, inflammatory bowel diseases, metallothionein, multiple sclerosis, neurodegenerative diseases, rheumatoid arthritis


Abbreviations

AD

alzheimer's disease

AKt

protein kinase B

APT

adenosine triphosphate

AREs

antioxidant response elements

CNS

central nervous system

DSS

dextran sulfate sodium

GREs

glucocorticoid response elements

HCC

hepatocellular cancer

IBDs

inflammatory bowel diseases

IL

interleukins

KO

knockout

MFT1

metal response element‐binding transcription factor 1

MMP

matrix metalloproteinase

MREs

metal response elements

MS

multiple sclerosis

MTs

metallothioneins

NF‐κB

nuclear factor‐kappa B

NSCLC

non‐small cell lung cancer

PD

parkinson's disease

RA

rheumatoid arthritis

ROS

reactive oxygen species

SCC

squamous cell carcinoma

STAT

signal transducer and activator of transcription

TFs

transcription factors

TNF‐a

tumor necrosis factor‐a

VEGF

vascular endothelial growth factor

1. Background

Metallothioneins (MTs) are tiny proteins with a molecular weight of 6–7 kDa that either bind or release biological transition metals, depending on a specific cell situation. In 1957, Vallee and Margoshe discovered MTs as proteins that bind to cadmium in the renal cortex of horses [1]. Initial research concentrated on protein structure, but it quickly expanded to other functional and biochemical characteristics [2]. The human metallothionein (MT) gene cluster on chromosome 16q13 consists of several functional genes (MT1A‐MT1X, MT2A, MT3, MT4) and a number of pseudogenes, corresponding to a well‐conserved evolutionary structure which may correlate with coordinated gene regulation. Variation in the regulatory regions as well as in the coding region of the MT genes affects expression levels, the binding of metals, and is implicated in diseases like cancer and metal toxicity. Single nucleotide polymorphisms (SNPs) in the MT2A gene, notably rs28366003, rs10636, and rs1610216, are linked to disrupted metal homeostasis and increased risk of cancer, cardiovascular diseases, and type 2 diabetes, serving as potential biomarkers of metal toxicity and disease susceptibility [3, 4, 5]. MT plays a crucial role in transporting metals like copper and cadmium, regulating their metabolism, and protecting cells from the toxicity of unbound cadmium [3].

Metallothioneins can bind seven divalent metal ions, and this binding stabilizes their three‐dimensional structure. The MT is shaped like a finger, and it connects with the body when the metal level rises by activating these fingers. If the body has low levels of metal, the opposite occurs [1, 2, 3, 4]. MTs have long been known to regulate metal levels, preventing stress and heavy metal toxicity; however, emerging evidence shows their immunomodulatory effects influencing signal transduction pathways [6, 7]. MTs play key roles in metal detoxification, oxidative damage defense, DNA repair, cell proliferation, differentiation, neuroprotection, and apoptosis, with relevance to inflammatory diseases, neurological disorders, carcinogenesis, and chemotherapeutic development [6, 7, 8, 9].

Metallothionein provides zinc to activate transcription factors, crucial for the homeostasis of zinc‐dependent proteins which is essential for cell function and metabolism [10]. Zinc fingers, composed of cysteine, bind DNA bases and play roles in DNA recognition, transcription, RNA packing, protein folding, and apoptosis, regulating tissue development and neoplastic transformation [11, 12]. Because zinc availability is critical for a wide range of cellular activities, it is expected that MT dysregulation is linked to both normal and pathological conditions [13]. Studies show MT isoforms play a key immunoregulatory role in cancer [14], neurological disorders [11], and inflammation [10], making them a crucial immunotherapy target [10, 14]. Recent studies have explored the pathophysiological functions of metallothionein, but no comprehensive review has examined its significant role in various illnesses. As a powerful antioxidant and anti‐inflammatory, metallothionein can play an important role in a wide range of disease processes. A thorough understanding of metallothionein's function in various disease conditions is required for its full potential use. Therefore, this review aims to comprehensively discuss MT's role in human pathophysiology.

1.1. Cellular Localization and Biochemical Characteristics of Metallothioneins

Metallothionein is primarily synthesized in the liver and kidney and intestine and is found in high concentrations in fast‐dividing and transforming cells. Studies suggest that MT plays a key role in cell differentiation and proliferation [15, 16]. MT1 and MT2 are primarily cytoplasmic in non‐pathological tissues but are also detected in the nucleus, lysosomes, and the intermembrane space of liver mitochondria [15]. Their location varies during the cell cycle, with higher cytoplasmic expression in G0/G1, nuclear presence in S/G2, and cytoplasmic expression post‐G2/M transition [17, 18]. MTs translocate to the nucleus during liver regeneration post‐hepatectomy, likely reflecting their role in supplying zinc to transcription factors and enzymes for cell division [10, 19, 20].

MT translocation to the nucleus during the G1 to S phase transition likely protects DNA from free radicals and supplies zinc to enzymes involved in DNA replication, transcription, and RNA processing [21, 22]. MTs can diffuse into the nucleus via the nuclear pore complex due to their small size, with translocation requiring the small GTPase and other cytosolic molecules besides importins [23]. Additionally, reports exist regarding the import of MTs through the nuclear perinuclear cytoskeleton‐associated MT1 and MT2 mRNA. Effective protein shuttling at the G1 to S phase transition relies on MT1 and MT2 mRNA being anchored to the perinuclear cytoskeleton via eEF1 binding at nucleotides 21–36 and 66–76, enabling localized protein production [23, 24, 25].

Mammalian metallothioneins (MTs) are small proteins composed of 61 to 68 amino acids, with a high cysteine content and minimal histidine or aromatic amino acids [2, 4, 26]. MT isoforms are categorized based on factors like molecular weight, metal binding, and amino acid sequence [27]. MT1 and MT2 are expressed in all body cells, while MT3, primarily found in the CNS, plays a role in brain development and apoptosis and is found in normal skin and malignant skin lesions as well as breast and urinary bladder cancers [16, 28, 29]. MT4, located in the stratified epithelium, is involved in pH regulation and protection against skin injuries as well as contributing to taste and texture characterization on the tongue [16, 28, 29].

1.2. Metallothionein and Zinc

Metallothioneins play a crucial role in zinc homeostasis through the oxidation and reduction of cysteine sulfur ligands, which facilitate zinc release. Zinc and cadmium binding is essential to maintain the stability of MT; this ensures intracellular zinc availability [30, 31]. Zinc‐bound MT sources zinc from cellular pools, influenced by diet and transporter function, and can donate or extract zinc for metalloproteins [32]. Apo‐MT prevents DNA binding of zinc‐finger transcription factors like Spl and TFIIIA, while Zn‐MT restores activity [9, 13]. MTs regulate apoptosis, cell division, and gene expression by supplying zinc to proteins and transcription factors. During oxidative stress, they scavenge free radicals, enhancing zinc release and forming MT‐disulfide. This disulfide form is unstable and converts back to the thiol form in a more reduced environment, aided by selenium and elevated GSH/GSSG levels [32, 33]. Consequently, MT is essential to avert glutathione depletion. MT's superior antioxidant capacity over GSH is not just due to its higher thiol content, but also to its unique cysteine‐cluster structure, which enables efficient free radical scavenging and redox cycling [33].

1.3. Metallothionein Expression and Regulation

Despite not having a classical signal peptide MT can be released under stress through nonclassical pathways adding to the extracellular pool found in tissue spaces milk urine and serum. Using receptors like megalin (LRP‐2) and CD91 (LRP‐1) this extracellular MT can alter inflammatory and immune responses. Furthermore MTs ability to both respond to and induce the expression of Pro‐inflammatory cytokines (IL‐1 IL‐6 and TNF‐α) emphasizes a reciprocal regulatory role in cytokine signaling and inflammatory pathophysiology [34]. In response to tissue injury, infection, inflammation, and neoplastic illness, MT expression can be altered by physiological mediators through response elements in the promoter [35]. Cytosolic MT levels can increase up to 100 times after inflammation, sequestering zinc. MT induction occurs within 2–4 h and depends on zinc availability [35, 36, 37]. MT gene expression is regulated by elements like the TATA box, GC box, glucocorticoid response elements (GREs), antioxidant response elements (ARE), and metal response elements (MREs), which interact with TFIID, AP1, and AP2 and are crucial for metal induction [8, 32, 37, 38]. Zinc activates MTF1 by displacing its inhibitor, allowing it to bind to MREs in the MT promoter, promoting MT transcription. MTF1 translocates to the nucleus after zinc activation, and its activity is regulated by phosphorylation/dephosphorylation, with protein phosphatase 2 A (PP2A) controlling MT production [39, 40, 41, 42, 43]. Zn is the only metal that activates metal response element‐binding transcription factor 1 (MTF1) [44]. Other heavy metals, like cadmium and lead, can promote MT transcription via MRE. MT gene expression is regulated by the TATA box, response elements like ARE and MRE, and factors like Sp1, TPA, USF, Ap1, Ap2, and MTF‐1 (Figure 1) [7, 35, 37].

Figure 1.

Figure 1

A summary of the regulation and function of the metallothionein (MT) gene [35]. Many response elements in the MT promoter up‐regulate transcription. These are some examples: 1) following zinc occupancy, MTF‐1 activates MRE at the promoter region, which is determined by zinc from dietary supply; 2) glucocorticoid response elements (GRE); 3) elements that are triggered by cytokine signaling through STAT proteins; and 4) antioxidant response elements (ARE), also known as electrophile response elements, which are triggered by redox conditions. Methylation also suppresses expression in certain tumor cells; 5) Release of Zn from Zn7MT to zinc‐dependent apoproteins, indicating MT's role in metal homeostasis.

2. The Role of Metallothioneins in Diabetes

Metallothioneins, notably MT1 and MT2A, exert opposing and context‐dependent effects on pancreatic beta‐cell function and diabetes. In the transgenic mice overexpressing the human MT2A gene under the insulin promoter (MT2A‐Tg) models, MTs have been shown to possess a protective antioxidant activity during STZ‐induced hyperglycemia and islet viability, which protects against oxidative and nitrosative damage [45, 46, 47]. These protective effects lead to better islet graft survival and glycemic control in transplantation animals. Similarly, exogenous administration of recombinant MTs fused to cell‐penetrating peptides, such as Tat‐MT1A, provides resistance to glucolipotoxicity, hypoxia, and cytokine‐induced oxidative stress in beta cells in vitro and in vivo [48]. However, contradicting evidence from nonobese diabetic (NOD) mice suggests that MT2A overexpression can speed up diabetes onset, particularly in males, despite decreasing cytokine‐induced ROS. This harmful effect is connected to more beta‐cell death, weaker insulin signaling (because of less AKT/FOXO1 phosphorylation), and lower levels of PDX1, suggesting that MTs might change key survival and function processes without relying on their antioxidant properties [49, 50].

2.1. The Role of Metallothioneins in Oxidative Stress and Inflammatory Diseases

Metallothioneins (MTs) neutralize free radicals through redox‐dependent mechanisms, with thiolate ligands in cysteine residues contributing to antioxidant activity via oxidative stress‐induced zinc mobilization [2, 4]. Rising oxidative stress promotes MT gene expression, which allows for ROS scavenging [8, 34, 35, 36], lysosomal stabilization, and decreased apoptosis [47]. MTs surpass glutathione at lowering ROS and preventing lipid peroxidation [51, 52]. Inflammation, infection, and stress all activate MTs, which behave similarly to acute‐phase proteins. Lipopolysaccharide (LPS) enhances MT1, particularly in the liver, whereas MT‐null animals exhibit immunological dysfunction [2, 9, 10]. MTs control immunological responses by modulating zinc‐dependent proteins such as p53, NF‐kB, and Sp1 [37, 38]. LPS stimulates MT1 production via IL6RE and glucocorticoid synergy near the MT1 promoter [53, 54], and IL‐6‐induced STAT phosphorylation mediates MT's anti‐inflammatory actions [55, 56]. MT‐KO mice show greater tissue injury and cytokine levels [54, 57, 58, 59]. MTs reduce inflammation by disrupting NO formation and facilitating zinc withdrawal from microbes, supporting antibacterial function [22, 60]. Extracellular MTs act as chemokine‐like danger signals despite lacking classic structure due to high cysteine content [61, 62].

2.2. The Role of Metallothionein in Rheumatoid Arthritis Pathogenesis

Rheumatoid arthritis (RA) is an inflammatory disease that commonly affects tissues and joints [63]. MT has functional effects on immune cells and may play a role in preventing RA as a stress response protein that sequesters toxins. Studies have revealed that MT1 expression is highly elevated in RA and is directly linked to disease progression. Similarly, when MT1 was administered locally, it significantly decreased pathogenic signs and synovial inflammation in rheumatoid mice [64, 65]. An imbalance between Th17 and Treg cells leads to RA development, with Th17 causing joint destruction and Tregs preserving immunological tolerance [66, 67]. MT1 reduces RA inflammation by lowering Pro‐inflammatory cytokines and shifting CD4 + T cell development to Treg cells while decreasing Th17 cells [66]. This shift occurs through STAT3 pathway suppression, which suppresses STAT3‐mediated RORγt activation and Th17‐related cytokines [54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66]. MT1 also controls Th1 responses in autoimmune and arthritic diseases [54].

Evidence from MT1 and MT2 deficient and transgenic mice suggests that in autoimmune arthritis, MT1 may be crucial for controlling the Th1 response [68]. In addition, in mice with developed arthritis, daily injections of MT1 and MT2 proteins significantly reduced disease occurrence and severity [66]. Furthermore, MT directly inhibits the expression of RA‐related inflammatory cytokines throughout the pathogenesis of RA. Pro‐inflammatory cytokines, such as IL‐6, IL‐1, IFN‐γ, IL‐17, and TNF‐α, which are key mediators in RA progression, were significantly suppressed in the peripheral blood mononuclear cells of RA patients after treatment with recombinant MT1, while this phenomenon was not observed in the control group [65, 69]. Evidence from these studies suggests that MT1 may have potential therapeutic value for this disease [54].

2.3. Metallothionein and Inflammatory Bowel Diseases

Inflammatory bowel disorders (IBDs) have a complex pathophysiology involving immune system disorders, environmental factors, gut microbiome changes, and local immune abnormalities [70, 71]. IBD patients exhibit increased pro‐inflammatory cytokine release, oxidative stress, and altered apoptosis [56]. MTs contribute to IBD inflammation, with animal studies showing significantly higher MT1 and MT2 expression in inflamed colitis tissue. MT1 and MT2 deficiency worsens DSS‐induced colitis, while MTs help prevent mucosal inflammation by reducing pro‐inflammatory cytokine production [56, 72]. In MT1/2 (‐/‐) KO mice, colitis induction caused greater myeloperoxidase and cytokine production than in wild‐type mice, indicating MTs' protective role in reducing inflammation [68].

IBD patients have weakened antioxidant barriers and severe local ROS activity, correlating with disease activity [73, 74]. MT upregulation may defend against ROS damage [56, 75], yet local MT expression in IBD epithelium is lower than in healthy individuals [76]. However, some studies found MTs non‐protective, with MT‐null mice showing reduced disease markers post‐DSS treatment [77, 78, 79]. Further studies reported no significant differences in mucosal MT expression among IBD patients [80, 81] nor genetic variations predisposing to IBD [82]. MTs reduce disease severity by inducing anti‐inflammatory cytokines like TGF‐β and IL‐10. In all animal models, MTs act as barriers to local inflammation [83, 84]. MTs help reduce IBD severity by reducing inflammation, modulating oxidative stress, immune responses, apoptosis, and zinc homeostasis through antioxidant, anti‐inflammatory, and Antiapoptotic functions [56, 77, 78, 79, 83].

2.4. Metallothionein in Multiple Sclerosis Pathogenesis

Multiple sclerosis (MS) is an inflammatory demyelinating condition of the central nervous system (CNS) with complex pathophysiology [85]. Oxidative stress promotes demyelination and neuronal degeneration in MS, and MT plays a key role as an antioxidant. MT1 and MT2 expression is elevated in MS brain lesions, particularly in macrophages, microglia, and reactive astrocytes, with higher expression in inactive lesions, suggesting a role in alleviating the disease [86, 87]. MT maintains zinc homeostasis, and zinc deficiency disrupts immune cell function, leading to lower metallothionein levels, reduced innate defense, and T‐cell‐mediated autoimmunity [9, 10, 54, 88]. Zinc deficiency also affects Th17 downregulation and the balance of T cells in MS [66, 68].

MT1/2 levels are elevated in MS and experimental autoimmune encephalomyelitis (EAE) [89, 90]. MT1 and MT2 knockout mice are more susceptible to disease [91], and inflammatory cytokines likely control MT production in MS lesions [92, 93, 94]. In EAE, MT1 and MT2 deficiency increases T cell and macrophage infiltration in the CNS and promotes pro‐inflammatory cytokines like TNF‐a, IL‐6, and IL‐1b. MT therapy can reduce EAE progression, alleviate tissue loss, and reduce CNS inflammation. MT2 treatment reduces symptoms and inflammation in EAE mice, while MT1 has antiapoptotic and antioxidant effects [93, 94, 95].

MT2 primarily promotes disease remission in EAE, while MT1 has minimal impact. MT1 influences serum cytokine patterns, and MT2 boosts Th2 responses and preserves myelin in the spinal cord, with less WBC infiltration in MT2‐treated mice [92]. MT2's stronger Zn‐thionein character maintains Zn (II), while MT1's lower Zn (II) properties allow for Cu (I) binding [91, 96].

2.5. Metallothioneins in Neurodegenerative Diseases: Neuroprotection and Metal Homeostasis

A high oxygen consumption by the central nervous system (CNS) makes it highly vulnerable to oxidative stress [97]. By scavenging reactive oxygen species and binding toxic metal ions, MTs are essential for cellular defense against oxidative stress and heavy metal toxicity [97, 98, 99]. Their potential as both neuroprotective agents and indicators of metal‐induced neurotoxicity is highlighted by their overexpression in the central nervous system, especially when exposed to heavy metals like Pb, Cd, and Hg over an extended period of time. The antioxidant effects of MT play an important role in tackling the clumping of altered proteins and their toxicity [96, 97, 98, 99].

Reactive oxygen species play a central role in AD due to limited brain regeneration. MT1/2 are upregulated near amyloid plaques, especially in the hippocampus and cortex, suggesting involvement in AD pathogenesis [97, 100, 101, 102, 103]. MT1 suppresses Aβ‐induced microglial activation and neurotoxicity [104], while disrupted zinc homeostasis and elevated Zn and Fe levels in affected brain regions contribute to pathology [7, 105]. MT2A inhibits Aβ aggregation and reduces inflammation and neurotoxicity [7, 101, 102, 103]. MT3, though protective, is downregulated in AD; it blocks Aβ1‐40 toxicity and prevents harmful aggregates [7, 97, 106, 107, 108]. MT3 also scavenges Aβ via transthyretin binding [97, 98], and Zn7MT3 neutralizes redox‐active copper, preventing ROS and neuronal damage [109, 110, 111, 112]. Both Zn7MT3 and Zn7MT2A prevent Cu‐Aβ1‐40‐induced neurotoxicity [113]. MTs inhibit apoptosis by blocking caspase‐3 and DNA fragmentation [6, 7, 99, 100, 101]. The protective effect of human Zn7MT3 against copper‐mediated toxicity in neurodegenerative diseases is summarized (Figure 2).

Figure 2.

Figure 2

Metal‐swap reactions and Cu(II) redox‐silencing by Zn7MT3 in neurodegenerative diseases [100]. The metal switch between Zn7MT3 and the disease‐specific amyloidogenic Cu(II)‐Aβ1‐40 peptide, Cu(II)–α‐Syn, and Cu(II)‐PrP proteins eliminates ROS generation and the cellular toxicity that goes with it. Cu(II) is reduced by the thiolates as it binds to the N‐terminal domain, creating the Cu(I)4Zn4MT‐3 species as well as the non‐redox‐active Zn(II)‐Aβ1‐40, Zn(II)‐‐Syn, and Zn(II)‐PrP. The N‐terminal β‐domain of Cu(I)4Zn4MT‐3 contains an air‐stable Cu(I)4‐thiolate cluster and two disulfide residues.

Metallothioneins also play a role in the etiology of Parkinson's disease (PD), providing neuroprotective benefits via metal chelation, antioxidative action, and regulation of microglial activation [111]. PD is characterized by the gradual loss of dopamine neurons that provide signals to the striatum [114]. In PD, dopaminergic neurons can produce excessive dopamine, leading to neurotoxicity. Intrinsic MT may protect against dopamine quinone‐induced neurotoxicity in hemiparkinsonian animals lacking MT1 and MT2 receptors [115]. In an animal model of PD, MT2 treatment exhibited a neuroprotective effect that mitigates rotenone‐induced neurodegeneration [116]. Levodopa increased MT3 mRNA expression in the undamaged striatum, suggesting a link between MT3 regulation and progressive degeneration in PD, highlighting the role of metallothioneins in dopaminergic dysfunction [117, 118, 119].

In laboratory experiments investigating the interaction of Zn7MT‐3 with copper (II)‐bound Syn and prion proteins, it was discovered that Zn7MT‐3 acts protectively by sequestering copper [120]. The protein's thiol groups simultaneously reduce Cu(II) while binding to the N‐terminal region, leading to the formation of the Cu(I)₄Zn₄MT‐3 complex and non‐redox‐active Zn(II)‐Aβ1‐40, Zn(II)‐α‐Syn, and Zn(II)‐PrP species [97]. Human MT1A, transduced into mitochondria, restored tyrosine hydroxylase expression, improved mitochondrial function, reduced ROS, and alleviated dopaminergic neuron degeneration in PD, suggesting a potential treatment option [121].

Studies have also shown that MT induction aids in the production of coenzyme Q10 in the brains of transgenic and KO mice in PD models. This is accomplished by activating lipoamide dehydrogenase, which improves coenzyme Q10's antioxidant capacity [122]. Ubiquinone reduces pro‐inflammatory cytokines and boosts the production of ubiquinol but decreases synuclein nitration [122]. Likewise, investigations in vitro as well as in vivo revealed that MT gene overexpression decreased the amount of ROS produced, caspase‐3 activation, and lipid peroxidation caused by lipid peroxidating agents [123, 124]. The therapeutic applications of MTs and MT‐derived peptides for reducing metal‐induced misfolded protein aggregation and inflammation make them a possible therapeutic target in neurodegenerative illnesses like PD [111, 120].

3. Roles of Metallothionein in Carcinogenesis

Metallothioneins may promote tumor growth through mitogenic actions and apoptosis suppression. MT overexpression has been associated with resistance to anticancer medicines and radiotherapy [19, 20, 32]. MTs are essential to tumor cell viability because they combat metal toxicity, oxidative stress, and proliferation [3, 21]. In some situations, MT expression is related to more aggressive and higher‐grade tumors, whereas in others, it's related to more differentiated, lower‐grade tumors [16]. Reduced MT expression has been seen in various cancer types, including liver [16, 25, 125], colon [126], and prostate [14]. Besides, the expression of MT decreases gradually throughout the change from normal colorectal tissue to adenomatous polyps and cancer [127]. In contrast, abnormal MT overexpression has been discovered in malignancies, including breast cancer and oral squamous cell carcinoma [13, 16, 32, 128, 129].

Overexpression of MT has a role in medication resistance because nuclear expression of MT protects DNA in cancer cells from the damaging effects of therapies [16, 17, 20]. Indeed, MT knockout mice were found to have a greater rate of induced carcinogenesis [16]. There is proof that MT1/2 both offer protection against chemical carcinogens. After 14 weeks of 7,12‐dimethylbenz[a]anthracene (DMBA) therapy, MT‐/‐ mice developed cutaneous tumors, but MT + /+ mice did not. These MTs were discovered to be endogenic protection factors against DMBA‐induced skin malignancies [16, 130]. In cancer, MT has a dual function, suppressing tumor growth in the early stages and promoting tumor progression in the later stages [131, 132, 133].

3.1. Metallothioneins and Tumour Progression

Tumors with elevated MT expression may have enhanced the breakdown of extracellular matrix components and facilitated tumor cell invasion [129]. The possible antiapoptotic action of MT due to its regulatory action on p53 or NF‐κB activity by donating or withdrawing zinc ions may be connected to a less favorable prognosis in individuals with increased MT expression [134, 135, 136, 137]. MTs remove zinc from P53, favoring an aggregation‐prone state that leads to p53 instability, inactivation, and loss of function. As a result, uncontrolled cell growth occurs. Overexpression of MTs had a strong inhibitory effect on P53 transcriptional activity, which results from the metal chelation function of MTs [138, 139]. MTs interact with NF‐κB and alter its antiapoptotic actions, most likely because zinc is required for NF‐κB's DNA‐binding function [135]. MT2A also influences the activation of the NF‐κB pathway and thereby contributes to tumor aggression in gastric and colorectal cancers [140, 141]. By increasing NF‐κB activity, MT1M downregulation can also aid in hepatocellular cancer development [142].

Persistent apo‐MT expression in tumor cells contributes to faster growth and survival of these cells by inducing inactive p53 [137]. Metal‐free MT interacts with p53 in tumor cells, preventing DNA binding and affecting transcription and apoptosis, suggesting an oncogenic role [143, 144]. MT1 and MT2 null cells have been documented in vitro and in vivo to be more susceptible to apoptotic stimuli and to have roughly 3–4‐fold greater basal levels of normally functioning p53 than counter‐MT‐containing cells [143]. Besides, following cisplatin treatment of cells from MT1 and MT2 gene KO mice, the amounts of p53 and the death effector protein Bax were much higher than in normal cells. Thus, MT deficiency promotes apoptosis susceptibility [145, 146].

Other researchers, however, have discovered no link between MT1 and 2 protein concentrations and p53 expression in cancerous tumors [147]. MT1 and 2 induction was greater in well‐functioning p53‐containing cells [148]. Apo‐MT removes Zn from p53, inactivating it, while Zn‐bound MT1 and 2 supports p53 function, highlighting the role of MT form, timing, and dosage in cancer. Elevated MT1 and MT2 levels reduce apoptosis in various cancers [147, 149]. MT1 and MT2 has been established to prevent apoptosis by increasing bcl‐2 and c‐myc, which are antiapoptotic oncogenes, while inhibiting proapoptotic tumor suppressor proteins such as caspase‐1 and‐3 and cytochrome c leakage [150]. The fact that Zn also impacts caspase‐3 activity may explain the link between MT1 and MT2 levels and caspase‐3 [23, 134].

MT1E promoter methylation is likely in breast cancer; a process of gene inactivation leads to decreased MT1E expression [150]. Moreover, cancer development has been correlated with MT1G suppression in esophageal, liver, thyroid, and prostate cancers. Promoter hypermethylation has been connected to MT1G silencing [151, 152]. The downregulation of MT1G in hepatocellular malignancy was likewise caused by an allelic loss on chromosomes 16q12.1–q23.1 [153, 154]. MT1G stabilizes p53 by inhibiting MDM2 and donating zinc, enhancing its activity. MT1G mutation induces Bax‐mediated cell death and p21‐driven arrest, potentially preventing HCC tumors [154, 155, 156, 157, 158].

CpG island hypermethylation promotes gastric cancer by inactivating MT3, MT1H, and MT1G [138]. The dephosphorylation of the C/EBP transcription factor in liver cancer suppresses MT1 and MT2A genes through the PI3K/AKT pathway. In thyroid cancer, MT1G has anticancer effects by reducing cell invasion and proliferation and promoting cell cycle termination and cell death. This is achieved by suppressing Akt and Rb phosphorylation or altering the PI3K/AKT and Rb/E2F signaling pathways [159, 160]. Zinc ions promoted MT isoform synthesis in breast cancer cells while also marginally increasing VEGF expression [161]. MT1/2‐deficient mice that produce IL‐6 demonstrated diminished IL‐6‐induced angiogenesis, showing that MT1/2 plays an angiogenic role through angiogenic molecule regulation [162]. MTs activate gelatinase (MMP), which is required for ECM breakdown and metastasis [163]. MMP‐9, which is connected to neovascularization and ECM remodeling, interacts with MTs and promotes tumor growth [164, 165].

3.2. Metallothionein 1 Modulated Signaling Pathways in Cancer

The inhibitory impact of MT1G on tumors in cancer depends on the PI3K/Akt pathway. Reduced Akt activation by MT1G enhances Mdm2 overexpression and suppresses cell adhesion, crucial in thyroid carcinogenesis [166]. Additionally, genes related to the Wnt/β‐catenin pathway were enriched in HCC patients with reduced MT1H expression. MT1H overexpression in HepG2 and Hep3B cells downregulates Wnt/β‐catenin target genes by lowering β‐catenin nuclear translocation, suppressing the pathway, and reducing tumorigenicity [167].

MT1 may have dual roles in NF‐kB activation. MT loss decreases the nuclear p65 subunit in fibroblast cell lines without affecting NF‐kB transcriptional function, increasing apoptosis susceptibility. Restoring MT1 expression elevates p65 levels, NF‐kB activity, and apoptosis resistance, making MT1 essential for NF‐kB activation [57, 168]. Similarly, MT overexpression enhances NF‐kB DNA binding and gene activation in breast tumor cell lines. However, MT1 gene transfection into MT‐KO cells inhibits TNF‐a‐induced NF‐kB‐dependent gene expression by preventing IkBa degradation [169]. MT1M expression also blocks TNF‐induced IkB degradation and NF‐kB transactivation in HCC, suggesting MT1 deficiency promotes carcinogenesis by increasing NF‐kB activity [140]. Zinc and zinc ionophores reduce NF‐kB DNA binding in HeLa cells, indicating MT1's role in NF‐kB signaling via zinc sequestration (Figure 3) [170].

Figure 3.

Figure 3

Signaling pathways that MT1 controls [57]. MT1 inhibits STAT1/3 phosphorylation, impeding the development of Tr1 cells that produce IL‐10. MT1 also suppresses Th17 cell proliferation by decreasing STAT3 phosphorylation. The elevation of p50/P65 activity by MT1 can positively affect the NF‐kB signaling pathway, which promotes the motility of tumor cells, tumor cell invesion, and cell death resistance. MT1 can also inhibit carcinogenesis by negatively regulating TNF‐a‐induced IkBa degradation. These fundamental activities are facilitated by MT1‐mediated suppression of free Zn2 + . By inhibiting Akt phosphorylation, MT1 inhibits the PI3K/Akt pathway. MT1 inhibits the Wnt/b‐catenin pathway by decreasing b‐catenin nuclear translocation.

3.3. Metallothionein Isoforms in Various Types of Cancer

MTs have a function in the onset of cancer and its course, but their expression is not ubiquitous in all human malignancies [14, 16, 25, 125, 126, 127, 128, 129]. So far, the underlying molecular processes that underlie the upregulation of MT expression in certain cancers and downregulation in others are not yet understood. Changes in isoform expression patterns could be one such explanation. Therefore, it was discovered that overexpression of particular MT isoforms affects the proliferation of low‐MT‐expressing cancer cells [5]. MT3 isoforms and all MT1 sub‐isoforms except MT1B are found in breast cancer. MT2A is upregulated in renal tumors, while MT1A and MT1G are downregulated. MT3 is present in renal carcinoma and normal kidney tubules but absent in normal bladder tissue, making it a potential bladder cancer biomarker. Bladder cancer also shows high MT1X mRNA expression [26, 171]. While the MT1X isoform was downregulated in cases of advanced prostate cancer, the MT1A, E, X, and MT2A isoforms were all prevalent in healthy prostate tissue. Additionally, MT1 and MT2 isoforms have been linked to the proliferative activity of human malignancies in the breast, colon, and prostate [26].

A study found that MT1X overexpression in HCC cells slows the cell cycle, enhances apoptosis, inhibits tumor development, promotes lung metastasis, and stops cell division through NF‐kB signaling [158]. In gastric adenocarcinoma, elevated levels of MT1X were observed in cancer‐associated fibroblasts, which subsequently enhanced angiogenesis [172]. MT1M is expressed in a range of typical hepatic tissues, with hepatic tissues having the greatest expression. MT1M expression, however, was significantly reduced in HCC specimens. According to a methylation profile investigation, most of the HCCs that have been investigated have methylated MT1M promoters [173]. MT expression in normal lung tissue appears to protect against potentially carcinogenic causes [174]. However, high‐MT lung epithelial cells are valuable for the subsequent activation of oncogenesis. Thus, it appears that MTs protect lung cells from harmful substances until a key event occurs, at which point they contribute to tumor formation [158, 175]. MTs' expression in tumors influences metastasis, cell adhesion, invasion, and migration. Understanding these changes can help identify treatment goals and inhibit tumor development [49, 176].

Although MT isoforms and sub‐isoforms (Table 1) have been proposed as diagnostic and prognostic biomarkers, there are limitations in their clinical utility because MT proteins can be induced by virtually any exogenous stressor (such as heavy metals, oxidative stress, inflammation, or exposure to drugs) thereby precluding disease specific interpretations. For example, increased MT levels could simply be a mark of cellular stress more generally, not a specific disease such as cancer. As suggested by previous studies, optimizing the isoform‐specificance detection method and combination with other onco‐markers is necessary to enhance detection sensitivity and specificity for diagnosis in clinical practice [3, 171].

Table 1.

Summary of Metallothionein Isoforms in Cancer.

MT isoform Function/role Regulation Cancer‐related effects References
MT1 (general) Zinc homeostasis, Antiapoptotic, redox regulation Induced by metals, stress, NF‐κB, methylation status Promotes tumor survival, inhibits p53, modulates NF‐κB and PI3K/Akt signaling, enhances angiogenesis [129, 134, 135, 136, 138, 139, 140, 141, 162, 164]
MT2A Similar to MT1; regulates NF‐κB Upregulated in renal tumors; PI3K/AKT involvement Promotes invasion, survival; activates NF‐κB in gastric and colorectal cancers [140, 141, 165]
MT3 Brain‐specific; growth‐inhibitory in normal tissue Inactivated by promoter methylation in gastric cancer Silencing contributes to cancer progression [138, 171]
MT1E Antiapoptotic, zinc donor Promoter methylation in breast cancer → inactivation Downregulation contributes to tumor progression [150]
MT1G Stabilizes p53, inhibits MDM2, enhances p21 and Bax Silenced via hypermethylation and chromosomal loss Tumor suppressor in liver, thyroid, prostate, and esophageal cancers [151, 152, 153, 154, 155, 156, 157, 158]
MT1H Regulates Wnt/β‐catenin, suppresses proliferation Downregulated in HCC; promoter methylation Tumor suppressor; its loss enhances β‐catenin activity [159, 167]
MT1M Regulates NF‐κB, suppresses tumor growth Downregulated in HCC; promoter methylation Inhibits TNF‐α‐induced NF‐κB activation, acts as tumor suppressor [140, 142, 173]
MT1X Modulates NF‐κB, apoptosis, and cell cycle Overexpressed in HCC and gastric cancer fibroblasts Suppresses tumor growth; enhances angiogenesis in fibroblasts [158, 172]

3.4. Metallothioneins and Their Roles in Chemoresistance

Tumor MT expression is linked to both innate and acquired resistance to antitumor drugs. MTs contribute to chemoresistance by deactivating and chelating drugs, protecting biomolecules from cytotoxicity, and potentially driving multidrug resistance (MDR) [5, 32, 177]. Drug or metabolite sequestration by MT may restrict the antineoplastic medications' ability to interact directly with their intracellular targets, lowering the effectiveness of their actions. As a result, MT expression in malignant cells shields them while increasing tumor growth rate, resulting in shorter patient survival [5, 178]. Moreover, the antiapoptotic effects of MTs and their transcription factor (de)activation and ROS scavenging are favorable for the survival and growth of cancer cells, besides their protection against the immune system of the host [140, 141, 142, 143, 144]. Thus, MTs are double agents; they have significant implications for physiological mechanisms as well as cancer, providing resistance to antineoplastic drugs [5, 158, 179, 180, 181]. For instance, MT1G overexpression leads to sorafenib chemoresistance in HCC by suppressing non‐apoptotic regulated cell death, ferroptosis, which participates in glutathione depletion, lipid peroxidation, and iron metabolism [181]. MT‐3 promotes cisplatin resistance in neuroblastoma cells, revealing its role in tumor‐specific chemoresistance mechanisms [177]. It follows that the possibility of using tumor cell expression of MT to tailor treatment strategies is not surprising [158, 182].

3.5. Metallothioneins as Cancer Therapeutic Targets

MTs have beneficial effects for alleviating inflammatory and neurodegenerative diseases. However, MTs are double agents for cancer progression [5, 53, 158]. Because of their nucleophilicity, MTs have to shield cells from the lethal consequences of electrophilic antitumor medications. Depending on the isoforms of MTs, therapeutic options for targeting them include DNA methylation, siRNA, antisense mRNA, and microRNA. Therefore, MT gene KO is becoming more and more popular as a therapeutic objective for malignant tumors. One common method for permanently reducing gene expression is RNA interference [158, 183, 184]. To target MT2A in ovarian cancer, researchers created pRNA and siRNA chimeras. The pRNA/siRNA complex may limit MT2A expression, reducing cell proliferation. Thus, siRNA‐mediated silencing of the MT2A gene limits breast and ovarian tumor cell proliferation [185]. Conversely, MT1G overexpression in gastric cancer played tumor suppressive roles. It suppresses rapid proliferation, metastasis, and invasion by promoting feroptosis and autophagy, thereby inhibiting gastric cancer cell function and providing novel mechanisms and therapeutic alternatives [186]. To date, only a few MT isoform‐targeting approaches are approved for use as therapeutic drugs [5, 187]. More study is required to develop and refine these technologies, as well as to determine the clinical safety of different MT‐modulating approaches. Targeting specific MT isoforms in a range of malignancies suggests a bright future for MT biological applications in cancer treatment [5, 158].

4. Concluding Remarks

The antioxidant property of MT is enhanced based on zinc's accessibility. MTs reduce pro‐inflammatory cytokine production and shift the Th17/Treg balance, ultimately alleviating inflammatory diseases. In neurodegenerative diseases, MTs interact with protein aggregates and modulate their formation and toxicity. The MT's molecular mechanisms in neurodegenerative diseases involve its regulation of metal ion levels, antioxidant properties, and anti‐inflammatory effects. MT plays two roles in cancer, suppressing tumors in the early stages while promoting tumor progression in the later stages. Only a few MT isoform‐targeting approaches have been approved for use as therapeutic drugs. More study is required to develop and refine these technologies and assess the level of clinical safety of different MT‐modulating approaches.

5. Perspectives

In the future, the focus should be put on the generation of isoform specific metallothionein targeting compounds, based on the observation that metallothioneins have diverse functions in various pathophysiologies. Developments in high‐throughput screening and structural biology may speed discovery of safe and effective MT‐directed therapeutics. Studying MT functions in conditions other than those currently investigated may uncover new modes of action and therapeutic time windows, especially in neuroinflammatory and neurodegenerative diseases. Combining MT modulation with established therapies might improve disease course by focusing attention on the importance of a collaborative strategy in translating MT biology to the clinics.

Author Contributions

Ousman Mohammed: conceptualization, writing – original draft. Abdisa Tufa: resources. Solomon Tebeje Gizaw: investigation, writing – review and editing, writing – original draft, supervision, resources, project administration.

Ethics Statement

The authors have nothing to report.

Consent

The authors have nothing to report.

Conflicts of Interest

The authors declare no conflicts of interest.

Transparency Statement

The lead author Ousman Mohammed affirms that this manuscript is an honest, accurate, and transparent account of the study being reported; that no important aspects of the study have been omitted; and that any discrepancies from the study as planned (and, if relevant, registered) have been explained.

Mohammed O., Tufa A., and Gizaw S. T., “Emerging Roles of Metallothioneins in Human Pathophysiology: A review,” Health Science Reports 8 (2025). 10.1002/hsr2.71279.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

References

  • 1. Margoshes M. and Vallee B. L., “A Cadmium Protein From Equine Kidney Cortex,” Journal of the American Chemical Society 79 (1957): 4813–4814. [Google Scholar]
  • 2. Bhardwaj H., Singh C., Kumar B. S., and Singh D., “Role of Metallothionein to Moderate Heavy Metals Toxicity in Animals: A Review,” Cancer 8, no. 9 (2021): 638–648. [Google Scholar]
  • 3. Sekovanić A., Jurasović J., and Piasek M., “Metallothionein 2A Gene Polymorphisms in Relation to Diseases and Trace Element Levels in Humans,” Archives of Industrial Hygiene and Toxicology 71, no. 1 (2020): 27–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Kayaaltı Z. and Söylemezoğlu T., “The Polymorphism of Core Promoter Region on Metallothionein 2A‐metal Binding Protein in Turkish Population,” Molecular Biology Reports 37 (2010): 185–190. [DOI] [PubMed] [Google Scholar]
  • 5. Zulijani A., Dekanić A., Ćabov T., and Jakovac H., “Metallothioneins and Megalin Expression Profiling in Premalignant and Malignant Oral Squamous Epithelial Lesions,” Cancers 13, no. 18 (2021): 4530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Marikar F. and Zi‐Chun H., “Metal‐Binding Protein: Metallothionein,” International Journal of Medical Biochemistry 6, no. 1 (2023): 57–62. [Google Scholar]
  • 7. Rodrigo M. A., Jimemez A. M., Haddad Y., et al., “Metallothionein Isoforms as Double Agents—Their Roles in Carcinogenesis, Cancer Progression and Chemoresistance,” Drug Resistance Updates 52 (2020): 100691. [DOI] [PubMed] [Google Scholar]
  • 8. Yang R., Roshani D., Gao B., Li P., and Shang N., “Metallothionein: A Comprehensive Review of Its Classification, Structure, Biological Functions, and Applications,” Antioxidants 13, no. 7 (2024): 825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Juárez‐Rebollar D., Rios C., Nava‐Ruíz C., and Méndez‐Armenta M., “Metallothionein in Brain Disorders,” Oxidative Medicine and Cellular Longevity 2017 (2017): 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Subramanian Vignesh K. and Deepe G. Jr., “Metallothioneins: Emerging Modulators in Immunity and Infection,” International Journal of Molecular Sciences 18, no. 10 (2017): 2197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Wani A. L., Parveen N., Ansari M. O., Ahmad M. F., Jameel S., and Shadab G. G. H. A., “Zinc: An Element of Extensive Medical Importance,” Current Medicine Research and Practice 7, no. 3 (2017): 90–98. [Google Scholar]
  • 12. Baltaci A. K., Yuce K., and Mogulkoc R., “Zinc Metabolism and Metallothioneins,” Biological Trace Element Research 183 (2018): 22–31. [DOI] [PubMed] [Google Scholar]
  • 13. Li M., Liu Y., Wei Y., et al., “Zinc‐Finger Protein YY1 Suppresses Tumor Growth of Human Nasopharyngeal Carcinoma by Inactivating C‐Myc–Mediated microRNA‐141 Transcription,” Journal of Biological Chemistry 294, no. 15 (2019): 6172–6187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Atrián‐Blasco E., Santoro A., Pountney D. L., Meloni G., Hureau C., and Faller P., “Chemistry of Mammalian Metallothioneins and Their Interaction With Amyloidogenic Peptides and Proteins,” Chemical Society Reviews 46, no. 24 (2017): 7683–7693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Ye B., Maret W., and Vallee B. L., “Zinc Metallothionein Imported Into Liver Mitochondria Modulates Respiration,” Proceedings of the National Academy of Sciences 98, no. 5 (2001): 2317–2322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Ma Y., Yin Z., Dai H., et al., “Increased Metallothionein‐1 Associated With Gout Activity and Tophi,” Immunological Investigations 52, no. 3 (2023): 319–331. [DOI] [PubMed] [Google Scholar]
  • 17. Nordberg M. and Nordberg G. F., “Metallothionein and Cadmium Toxicology—Historical Review and Commentary,” Biomolecules 12, no. 3 (2022): 360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Rodrigo M. A., Jimemez A. M., Haddad Y., et al., “Metallothionein Isoforms as Double Agents—Their Roles in Carcinogenesis, Cancer Progression and Chemoresistance,” Drug Resistance Updates 52 (2020): 100691. [DOI] [PubMed] [Google Scholar]
  • 19. Cherian M. G. and Apostolova M. D., “Nuclear Localization of Metallothionein During Cell Proliferation and Differentiation,” Cellular and Molecular Biology (Noisy‐le‐Grand, France) 46 (2000): 347–356. [PubMed] [Google Scholar]
  • 20. Smith P. J., Wiltshire M., Furon E., Beattie J. H., and Errington R. J., “Impact of Overexpression of Metallothionein‐1 on Cell Cycle Progression and Zinc Toxicity,” American Journal of Physiology‐Cell Physiology 295, no. 5 (2008): C1399–C1408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Tapiero H. and Tew K. D., “Trace Elements in Human Physiology and Pathology: Zinc and Metallothioneins,” Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie 57, no. 9 (2003): 399–411. [DOI] [PubMed] [Google Scholar]
  • 22. Dziegiel P., Pula B., Kobierzycki C., Stasiolek M., and Podhorska‐Okolow M., Metallothioneins in normal and cancer cells (Springer, 2016). 218, 1–117. [DOI] [PubMed] [Google Scholar]
  • 23. Mickleburgh I., Chabanon H., Nury D., et al., “Elongation Factor 1α Binds to the Region of the Metallothionein‐1 mRNA Implicated in Perinuclear Localization—Importance of an Internal Stem–Loop,” RNA 12, no. 7 (2006): 1397–1407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Coyle P., Philcox J. C., Carey L. C., and Rofe A. M., “Metallothionein: The Multipurpose Protein,” Cellular and Molecular Life Sciences (CMLS) 59 (2002): 627–647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Mahim A. and Petering D. H., “Zinc Trafficking to Apo‐Zn‐Proteins 2. Cellular Interplay of Proteome, Metallothionein, and Glutathione,” Metallomics 14, no. 11 (2022): mfac081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Soda N., Sharan A., Gupta B. K., Singla‐Pareek S. L., and Pareek A., “Evidence for Nuclear Interaction of a Cytoskeleton Protein (OsIFL) With Metallothionein and Its Role in Salinity Stress Tolerance,” Scientific Reports 6, no. 1 (2016): 34762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Pula B., Tazbierski T., Zamirska A., et al., “Metallothionein 3 Expression in Normal Skin and Malignant Skin Lesions,” Pathology Oncology Research: POR 21 (2015): 187–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Liang G. Y., Lu S. X., Xu G., Liu X. D., Li J., and Zhang D. S., “Expression of Metallothionein and Nrf2 Pathway Genes in Lung Cancer and Cancer‐Surrounding Tissues,” World Journal of Surgical Oncology 11 (2013): 199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Singh A. K., Pomorski A., Wu S., Peris‐Díaz M. D., Czepczyńska‐Krężel H., and Krężel A., “The Connection of α‐and β‐Domains in Mammalian Metallothionein‐2 Differentiates Zn (Ii) Binding Affinities, Affects Folding, and Determines Zinc Buffering Properties,” Metallomics 15, no. 6 (2023): mfad029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Koh J. Y. and Lee S. J., “Metallothionein‐3 as a Multifunctional Player in the Control of Cellular Processes and Diseases,” Molecular Brain 13 (2020): 116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Shamsi T. N. and Fatima S., “Metallothionein: Classification, Biochemical Features and Clinical Applications,” Journal of Proteins & Proteomics 5, no. 1 (2014): 1. [Google Scholar]
  • 32. Aziz J., Rahman M. T., and Vaithilingam R. D., “Dysregulation of Metallothionein and Zinc Aggravates Periodontal Diseases,” Journal of Trace Elements in Medicine and Biology 66 (2021): 126754. [DOI] [PubMed] [Google Scholar]
  • 33. Domán A., Dóka É., Garai D., et al., “Interactions of Reactive Sulfur Species With Metalloproteins,” Redox Biology 60 (2023): 102617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Klaassen C. D., Liu J., and Diwan B. A., “Metallothionein Protection of Cadmium Toxicity,” Toxicology and Applied Pharmacology 238, no. 3 (2009): 215–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Davis S. R. and Cousins R. J., “Metallothionein Expression in Animals: A Physiological Perspective on Function,” Journal of Nutrition 130, no. 5 (2000): 1085–1088. [DOI] [PubMed] [Google Scholar]
  • 36. Li F. J., Fu S., Ye H., et al., “Metallothionein Alleviates Glutathione Depletion–Induced Oxidative Cardiomyopathy Through CISD1‐Dependent Regulation of Ferroptosis in Murine Hearts,” American Journal of Pathology 194, no. 6 (2024): 912–926. [DOI] [PubMed] [Google Scholar]
  • 37. Verhoef J. N. C., Allen A. L., Harding J. C. S., and Al‐Dissi A. N., “Metallothionein Expression in Horses With Chronic Liver Disease and Its Correlation With Ki‐67 Immunoreactivity,” Veterinary Pathology 55, no. 5 (2018): 703–710. [DOI] [PubMed] [Google Scholar]
  • 38. Perveen H., Chattopadhyay S., Maity M., Dash M., and Islam S. S., “Involvement of Proinflammatory Cytokines and Metallothionein in the Repairing of Arsenic‐Mediated Uterine Tissue Damage by Curcumin,” Journal of Basic and Clinical Physiology and Pharmacology 30, no. 4 (2019): 20170179. [DOI] [PubMed] [Google Scholar]
  • 39. De S. K., McMaster M. T., and Andrews G. K., “Endotoxin Induction of Murine Metallothionein Gene Expression,” Journal of Biological Chemistry 265, no. 25 (1990): 15267–15274. [PubMed] [Google Scholar]
  • 40. Sekovanić A., Jurasović J., and Piasek M., “Metallothionein 2A Gene Polymorphisms in Relation to Diseases and Trace Element Levels in Humans,” Archives of Industrial Hygiene and Toxicology 71, no. 1 (2020): 27–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Lee W., Haslinger A., Karin M., and Tjian R., “Activation of Transcription by Two Factors That Bind Promoter and Enhancer Sequences of the Human Metallothionein Gene and SV40,” Nature 325, no. 6102 (1987): 368–372. [DOI] [PubMed] [Google Scholar]
  • 42. Palmiter R. D., “Regulation of Metallothionein Genes by Heavy Metals Appears to be Mediated by a Zinc‐Sensitive Inhibitor That Interacts With a Constitutively Active Transcription Factor, MTF‐1,” Proceedings of the National Academy of Sciences 91, no. 4 (1994): 1219–1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Krizkova S., Kepinska M., Emri G., et al., “Microarray Analysis of Metallothioneins in Human Diseases—A Review,” Journal of Pharmaceutical and Biomedical Analysis 117 (2016): 464–473. [DOI] [PubMed] [Google Scholar]
  • 44. Haq F., “Signaling Events for Metallothionein Induction,” Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis 533, no. 1–2 (2003): 211–226. [DOI] [PubMed] [Google Scholar]
  • 45. Chen H., Carlson E. C., Pellet L., Moritz J. T., and Epstein P. N., “Overexpression of Metallothionein in Pancreatic β‐Cells Reduces Streptozotocin‐Induced DNA Damage and Diabetes,” Diabetes 50 (2001): 2040–2046. [DOI] [PubMed] [Google Scholar]
  • 46. Chen S., Han J., and Liu Y., “Dual Opposing Roles of Metallothionein Overexpression in C57BL/6J Mouse Pancreatic Β‐Cells,” PLoS One 10 (2015): e0137583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Bensellam M., Laybutt D. R., and Jonas J. C., “Emerging Roles of Metallothioneins in Beta Cell Pathophysiology: Beyond and above Metal Homeostasis and Antioxidant Response,” Biology 10, no. 3 (2021): 176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Kim M. J., Hwang Y. H., Kim Y. H., and Lee D. Y., “Immunomodulation of Cell‐Penetrating Tat‐Metallothionein for Successful Outcome of Xenotransplanted Pancreatic Islet,” Journal of Drug Targeting 25 (2016): 350–359. [DOI] [PubMed] [Google Scholar]
  • 49. Li X., Chen H., and Epstein P. N., “Metallothionein and Catalase Sensitize to Diabetes in Nonobese Diabetic Mice,” Diabetes 55 (2006): 1592–1604. [DOI] [PubMed] [Google Scholar]
  • 50. Chou F. C. and Sytwu H. K., “Overexpression of Thioredoxin in Islets Transduced by a Lentiviral Vector Prolongs Graft Survival in Autoimmune Diabetic Nod Mice,” Journal of Biomedical Science 16 (2009): 71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Babula P., Masarik M., Adam V., et al., “Mammalian Metallothioneins: Properties and Functions,” Metallomics 4, no. 8 (2012): 739–750. [DOI] [PubMed] [Google Scholar]
  • 52. Bakiu R., Pacchini S., Piva E., et al., “Metallothionein Expression as a Physiological Response Against Metal Toxicity in the Striped Rockcod Trematomus hansoni ,” International Journal of Molecular Sciences 23, no. 21 (2022): 12799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Baird S. K., Kurz T., and Brunk U. T., “Metallothionein Protects Against Oxidative Stress‐Induced Lysosomal Destabilization,” Biochemical Journal 394, no. 1 (2006): 275–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Miura T., Muraoka S., and Ogiso T., “Antioxidant Activity of Metallothionein Compared With Reduced Glutathione,” Life Sciences 60, no. 21 (1997): 301–309. [DOI] [PubMed] [Google Scholar]
  • 55. Samuel M. S., Datta S., Khandge R. S., and Selvarajan E., “A State of the Art Review on Characterization of Heavy Metal Binding Metallothioneins Proteins and Their Widespread Applications,” Science of the Total Environment 775 (2021): 145829. [Google Scholar]
  • 56. Lin X., Jagadapillai R., Cai J., Cai L., Shao G., and Gozal E., “Metallothionein Induction Attenuates the Progression of Lung Injury in Mice Exposed to Long‐Term Intermittent Hypoxia,” Inflammation Research 69 (2020): 15–26. [DOI] [PubMed] [Google Scholar]
  • 57. Dai H., Wang L., Li L., Huang Z., and Ye L., “Metallothionein 1: A New Spotlight on Inflammatory Diseases,” Frontiers in Immunology 12 (2021): 739918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Inoue K., Takano H., Shimada A., et al., “Role of Metallothionein in Coagulatory Disturbance and Systemic Inflammation Induced by Lipopolysaccharide in Mice,” The FASEB Journal 20, no. 3 (2006): 533–535. [DOI] [PubMed] [Google Scholar]
  • 59. Socha‐Banasiak A., Sputa‐Grzegrzółka P., Grzegrzółka J., et al., “Metallothioneins in Inflammatory Bowel Diseases: Importance in Pathogenesis and Potential Therapy Target,” Canadian Journal of Gastroenterology and Hepatology 2021 (2021): 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Yang J., Liu L., Li M., Huang X., Yang H., and Li K., “Naringenin Inhibits Pro‑Inflammatory Cytokine Production in Macrophages Through Inducing MT1G to Suppress the Activation of NF‑Κb,” Molecular Immunology 137 (2021): 155–162. [DOI] [PubMed] [Google Scholar]
  • 61. Read S. A., Parnell G., Booth D., Douglas M. W., George J., and Ahlenstiel G., “The Antiviral Role of Zinc and Metallothioneins in Hepatitis C Infection,” Journal of Viral Hepatitis 25, no. 5 (2018): 491–501. [DOI] [PubMed] [Google Scholar]
  • 62. Zangger K., Öz G., Haslinger E., Kunert O., and Armitage I. M., “Nitric Oxide Selectively Releases Metals From the N‐Terminal Domain of Metallothioneins: Potential Role at Inflammatory Sites,” The FASEB Journal 15, no. 7 (2001): 1303–1305. [DOI] [PubMed] [Google Scholar]
  • 63. Yin X., Knecht D. A., and Lynes M. A., “Metallothionein Mediates Leukocyte Chemotaxis,” BMC Immunology 6 (2005): 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Laukens D., Waeytens A., De Bleser P., Cuvelier C., and De Vos M., “Human Metallothionein Expression Under Normal and Pathological Conditions: Mechanisms of Gene Regulation Based on in Silico Promoter Analysis,” Critical Reviews™ in Eukaryotic Gene Expression 19, no. 4 (2009): 301–317. [DOI] [PubMed] [Google Scholar]
  • 65. Herrera‐Quintana L., Vázquez‐Lorente H., Gamarra‐Morales Y., Molina‐López J., and Planells E., “Evolution of Status of Trace Elements and Metallothioneins in Patients With COVID‐19: Relationship With Clinical, Biochemical, and Inflammatory Parameters,” Metabolites 13, no. 8 (2023): 931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. McInnes I. B. and Schett G., “Cytokines in the Pathogenesis of Rheumatoid Arthritis,” Nature Reviews Immunology 7, no. 6 (2007): 429–442. [DOI] [PubMed] [Google Scholar]
  • 67. Aziz J., Vaithilingam R. D., Radzi Z., and Rahman M. T., “Inflammatory Responses in Periodontitis With or Without Rheumatoid Arthritis Alter Salivary Metallothionein and Zinc,” Biological Trace Element Research 201, no. 7 (2023): 3162–3174. [DOI] [PubMed] [Google Scholar]
  • 68. Xia Y., Yang Q., Li Q., et al., “Metallothionein‐1 Mitigates the Advancement of Osteoarthritis by Regulating Th17/Treg Balance,” Cellular Immunology 405–406 (2024): 104877. [DOI] [PubMed] [Google Scholar]
  • 69. Sun J., Li L., Li L., et al., “Metallothionein‐1 Suppresses Rheumatoid Arthritis Pathogenesis by Shifting the Th17/Treg Balance,” European Journal of Immunology 48, no. 9 (2018): 1550–1562. [DOI] [PubMed] [Google Scholar]
  • 70. Jakovac H., Tota M., Grebic D., et al., “Metallothionein I+ II Expression as an Early Sign of Chronic Relapsing Experimental Autoimmune Encephalomyelitis in Rats,” Current Aging Science 6, no. 1 (2013): 37–44. [DOI] [PubMed] [Google Scholar]
  • 71. Penkowa M., Espejo C., Martínez‐Cáceres E. M., Montalban X., and Hidalgo J., “Increased Demyelination and Axonal Damage in Metallothionein I+ II‐Deficient Mice During Experimental Autoimmune Encephalomyelitis,” Cellular and Molecular Life Sciences (CMLS) 60 (2003): 185–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Ananthakrishnan A. N., “Epidemiology and Risk Factors for IBD,” Nature Reviews Gastroenterology & Hepatology 12, no. 4 (2015): 205–217. [DOI] [PubMed] [Google Scholar]
  • 73. De Souza H. S. and Fiocchi C., “Immunopathogenesis of IBD: Current State of the Art,” Nature Reviews Gastroenterology & Hepatology 13, no. 1 (2016): 13–27. [DOI] [PubMed] [Google Scholar]
  • 74. Tsuji T., Naito Y., Takagi T., et al., “Role of Metallothionein in Murine Experimental Colitis,” International Journal of Molecular Medicine 31, no. 5 (2013): 1037–1046. [DOI] [PubMed] [Google Scholar]
  • 75. Tian T., Wang Z., and Zhang J., “Pathomechanisms of Oxidative Stress in Inflammatory Bowel Disease and Potential Antioxidant Therapies,” Oxidative Medicine and Cellular Longevity 2017 (2017): 4535194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Yuksel M., Ates I., Kaplan M., et al., “Is Oxidative Stress Associated With Activation and Pathogenesis of Inflammatory Bowel Disease,” Journal of Medical Biochemistry 36, no. 4 (2017): 341–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Navarro‐sempere A., Martínez‐Peinado P., Rodrigues A. S., et al., “Metallothionein Expression in the Central Nervous System in Response to Chronic Heavy Metal Exposure: Possible Neuroprotective Mechanism,” Environmental Geochemistry and Health 45, no. 11 (2023): 8257–8269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Brüwer M., Schmid K. W., Metz K. A., Krieglstein C. F., Senninger N., and Schürmann G., “Increased Expression of Metallothionein in Inflammatory Bowel Disease,” Inflammation Research 50 (2001): 289–293. [DOI] [PubMed] [Google Scholar]
  • 79. Tran C. D., Ball J. M., Sundar S., Coyle P., and Howarth G. S., “The Role of Zinc and Metallothionein in the Dextran Sulfate Sodium‐Induced Colitis Mouse Model,” Digestive Diseases and Sciences 52 (2007): 2113–2121. [DOI] [PubMed] [Google Scholar]
  • 80. Oz H. S., Chen T., de Villiers W. J., and McClain C. J., “Metallothionein Overexpression Does Not Protect Against Inflammatory Bowel Disease in a Murine Colitis Model,” Medical Science Monitor 11, no. 3 (2004): BR69–BR73. [PubMed] [Google Scholar]
  • 81. Devisscher L., Hindryckx P., Lynes M. A., et al., “Role of Metallothioneins as Danger Signals in the Pathogenesis of Colitis,” Journal of Pathology 233, no. 1 (2014): 89–100. [DOI] [PubMed] [Google Scholar]
  • 82. Dooley T. P., Curto E. V., Reddy S. P., et al., “Regulation of Gene Expression in Inflammatory Bowel Disease and Correlation With IBD Drugs. Screening by DNA Microarrays,” Inflammatory Bowel Diseases 10, no. 1 (2004): 1–14. [DOI] [PubMed] [Google Scholar]
  • 83. Loachim E., Michael M., Katsanos C., Demou A., and Tsianos E. V., “The Immunohistochemical Expression of Metallothionein in Inflammatory Bowel Disease. Correlation With HLA‐DR Antigen Expression, Lymphocyte Subpopulations and Proliferation‐Associated Indices,” Histology and Histopathology 18 (2003): 75–82. [DOI] [PubMed] [Google Scholar]
  • 84. Morgan A. R., Fraser A. G., and Ferguson L. R., “Metallothionein Genes: No Association With Crohn's Disease in a New Zealand Population,” Journal of Negative Results in Biomedicine 11, no. 1 (2012): 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Stoop J. N., Harry R. A., von Delwig A., Isaacs J. D., Robinson J. H., and Hilkens C. M., “Therapeutic Effect of Tolerogenic Dendritic Cells in Established Collagen‐Induced Arthritis is Associated With a Reduction in Th17 Responses,” Arthritis and Rheumatism 62, no. 12 (2010): 3656–3665. [DOI] [PubMed] [Google Scholar]
  • 86. Lindeque J. Z., Levanets O., Louw R., and Van Der Westhuizen F. H., “The Involvement of Metallothioneins in Mitochondrial Function and Disease,” Current Protein & Peptide Science 11, no. 4 (2010): 292–309. [DOI] [PubMed] [Google Scholar]
  • 87. Smythe C., “Suppression of Arthritis by Metallothionein,” Arthritis Research & Therapy 4 (2002): 78052. [Google Scholar]
  • 88. Hemmer B., Kerschensteiner M., and Korn T., “Role of the Innate and Adaptive Immune Responses in the Course of Multiple Sclerosis,” The Lancet Neurology 14, no. 4 (2015): 406–419. [DOI] [PubMed] [Google Scholar]
  • 89. Nakamura S., Shimazawa M., and Hara H., “Physiological Roles of Metallothioneins in Central Nervous System Diseases,” Biological and Pharmaceutical Bulletin 41, no. 7 (2018): 1006–1013. [DOI] [PubMed] [Google Scholar]
  • 90. Keçecioğlu C., Sarıkaya C., Aydın A., Charehsaz M., and Efendi H., “Investigation of the Relationship Between Heavy Metals (Cadmium, Arsenic, and Lead) and Metallothionein in Multiple Sclerosis,” Cureus 16, no. 8 (2024): e66754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Waller R., Woodroofe M. N., Wharton S. B., et al., “Gene Expression Profiling of the Astrocyte Transcriptome in Multiple Sclerosis Normal Appearing White Matter Reveals a Neuroprotective Role,” Journal of Neuroimmunology 299 (2016): 139–146. [DOI] [PubMed] [Google Scholar]
  • 92. Jakovac H., Grebić D., Tota M., et al., “Time‐Course Expression of Metallothioneins and Tissue Metals in Chronic Relapsing Form of Experimental Autoimmune Encephalomyelitis,” Histology and Histopathology 26, no. 2 (2011): 233–245. [DOI] [PubMed] [Google Scholar]
  • 93. Jakovac H., Grubić Kezele T., Šućurović S., Mulac‐Jeričević B., and Radošević‐Stašić B., “Osteopontin–Metallothionein I/II Interactions in Experimental Autoimmunune Encephalomyelitis,” Neuroscience 350 (2017): 133–145. [DOI] [PubMed] [Google Scholar]
  • 94. Penkowa M., Espejo C., Martínez‐Cáceres E. M., Poulsen C. B., Montalban X., and Hidalgo J., “Altered Inflammatory Response and Increased Neurodegeneration in Metallothionein I+ II Deficient Mice During Experimental Autoimmune Encephalomyelitis,” Journal of Neuroimmunology 119, no. 2 (2001): 248–260. [DOI] [PubMed] [Google Scholar]
  • 95. Comes G., Fernandez‐Gayol O., Molinero A., et al., “Mouse Metallothionein‐1 and Metallothionein‐2 Are Not Biologically Interchangeable in an Animal Model of Multiple Sclerosis, EAE,” Metallomics 11, no. 2 (2019): 327–337. [DOI] [PubMed] [Google Scholar]
  • 96. Grubić‐Kezele T., Blagojević Zagorac G., Jakovac H., Domitrović R., Milin C., and Radošević‐Stašić B., “Hepatic Expression of Metallothionein I/II, Glycoprotein 96, IL‐6, and TGF‐β in Rat Strains With Different Susceptibilities to Experimental Autoimmune Encephalomyelitis,” Clinical & Developmental Immunology 2013 (2013): 750406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Morgan A. R., Fraser A. G., and Ferguson L. R., “Metallothionein Genes: No Association With Crohn's Disease in a New Zealand Population,” Journal of Negative Results in Biomedicine 11, no. 1 (2012): 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Waeytens A., De Vos M., and Laukens D., “Evidence for a Potential Role of Metallothioneins in Inflammatory Bowel Diseases,” Mediators of Inflammation 2009 (2009): 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Dostie K. E., Thees A. V., and Lynes M. A., “Metallothionein: A Novel Therapeutic Target for Treatment of Inflammatory Bowel Disease,” Current Pharmaceutical Design 24, no. 27 (2018): 3155–3161. [DOI] [PubMed] [Google Scholar]
  • 100. Vašák M. and Meloni G., “Mammalian metallothionein‐3: New Functional and Structural Insights,” International Journal of Molecular Sciences 18, no. 6 (2017): 1117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Zaręba N. and Kepinska M., “The Function of Transthyretin Complexes With Metallothionein in Alzheimer's Disease,” International Journal of Molecular Sciences 21, no. 23 (2020): 9003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Knopman D. S., Amieva H., Petersen R. C., et al., “Alzheimer Disease,” Nature Reviews Disease Primers 7, no. 1 (2021): 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Breijyeh Z. and Karaman R., “Comprehensive Review on Alzheimer's Disease: Causes and Treatment,” Molecules 25, no. 24 (2020): 5789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Jiang Z., Shen B., and Xiang J., “Metal‐Dependent Interactions of Metallothionein‐3 β‐domain With Amyloid‐β Peptide and Related Physiological Implications,” Journal of Inorganic Biochemistry 196 (2019): 110693. [DOI] [PubMed] [Google Scholar]
  • 105. Kim J. H., Nam Y. P., Jeon S. M., Han H. S., and Suk K., “Amyloid Neurotoxicity is Attenuated by Metallothionein: Dual Mechanisms at Work,” Journal of Neurochemistry 121, no. 5 (2012): 751–762. [DOI] [PubMed] [Google Scholar]
  • 106. Chung R. S., Adlard P. A., Dittmann J., Vickers J. C., Chuah M. I., and West A. K., “Neuron‐Glia Communication: Metallothionein Expression is Specifically up‐Regulated by Astrocytes in Response to Neuronal Injury,” Journal of Neurochemistry 88, no. 2 (2004): 454–461. [DOI] [PubMed] [Google Scholar]
  • 107. Valko M., Leibfritz D., Moncol J., Cronin M. T., Mazur M., and Telser J., “Free Radicals and Antioxidants in Normal Physiological Functions and Human Disease,” The International Journal of Biochemistry & Cell Biology 39, no. 1 (2007): 44–84. [DOI] [PubMed] [Google Scholar]
  • 108. Li Y., Jiao Q., Xu H., et al., “Biometal Dyshomeostasis and Toxic Metal Accumulations in the Development of Alzheimer's Disease,” Frontiers in Molecular Neuroscience 10 (2017): 339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Manso Y., Carrasco J., Comes G., et al., “Characterization of the Role of Metallothionein‐3 in an Animal Model of Alzheimer's Disease,” Cellular and Molecular Life Sciences 69 (2012): 3683–3700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Cheignon C., Tomas M., Bonnefont‐Rousselot D., Faller P., Hureau C., and Collin F., “Oxidative Stress and the Amyloid Beta Peptide in Alzheimer's Disease,” Redox Biology 14 (2018): 450–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Yu W. H., Lukiw W. J., Bergeron C., Niznik H. B., and Fraser P. E., “Metallothionein III is Reduced in Alzheimer's Disease,” Brain Research 894, no. 1 (2001): 37–45. [DOI] [PubMed] [Google Scholar]
  • 112. Irie Y. and Keung W. M., “Metallothionein‐III Antagonizes the Neurotoxic and Neurotrophic Effects of Amyloid β Peptides,” Biochemical and Biophysical Research Communications 282, no. 2 (2001): 416–420. [DOI] [PubMed] [Google Scholar]
  • 113. Liu K., Liu X., Wen L., et al., “Blocking Metallothionein‐2 Expression by Copper‐Doped Carbon Dots Induces Cellular Antioxidant System Collapse for Antitumor Therapy,” Nano Letters 24, no. 34 (2024): 10699–10709. [DOI] [PubMed] [Google Scholar]
  • 114. Miyazaki I. and Asanuma M., “Multifunctional Metallothioneins as a Target for Neuroprotection in Parkinson's Disease,” Antioxidants 12, no. 4 (2023): 894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Meloni G., Sonois V., Delaine T., et al., “Metal Swap Between Zn7‐Metallothionein‐3 and Amyloid‐β–Cu Protects Against Amyloid‐β Toxicity,” Nature Chemical Biology 4, no. 6 (2008): 366–372. [DOI] [PubMed] [Google Scholar]
  • 116. Chung R. S., Howells C., Eaton E. D., et al., “The Native Copper‐and Zinc‐Binding Protein Metallothionein Blocks Copper‐Mediated Aβ Aggregation and Toxicity in Rat Cortical Neurons,” PLoS One 5, no. 8 (2010): e12030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Jankovic J. and Tan E. K., “Parkinson's Disease: Etiopathogenesis and Treatment,” Journal of Neurology, Neurosurgery & Psychiatry 91, no. 8 (2020): 795–808. [DOI] [PubMed] [Google Scholar]
  • 118. Nies Y. H., Lim W. L., Abd Karim N., Yahaya M. F., and Teoh S. L., “Metallothionein II Treatment Mitigates Rotenone‐Induced Neurodegeneration in Zebrafish Models of Parkinson's Disease,” Frontiers in Pharmacology 16 (2025): 1478013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Miyazaki I., Asanuma M., Hozumi H., Miyoshi K., and Sogawa N., “Protective Effects of Metallothionein Against Dopamine Quinone‐Induced Dopaminergic Neurotoxicity,” FEBS Letters 581, no. 25 (2007): 5003–5008. [DOI] [PubMed] [Google Scholar]
  • 120. Miyazaki I., Sogawa C. A., Asanuma M., et al., “Expression of Metallothionein‐III mRNA and Its Regulation by Levodopa in the Basal Ganglia of Hemi‐Parkinsonian Rats,” Neuroscience Letters 293, no. 1 (2000): 65–68. [DOI] [PubMed] [Google Scholar]
  • 121. Chung R. S., Penkowa M., Dittmann J., et al., “Redefining the Role of Metallothionein Within the Injured Brain,” Journal of Biological Chemistry 283, no. 22 (2008): 15349–15358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Li Y., Yang C., Wang S., et al., “Copper and Iron Ions Accelerate the Prion‐Like Propagation of α‐synuclein: A Vicious Cycle in Parkinson's Disease,” International Journal of Biological Macromolecules 163 (2020): 562–573. [DOI] [PubMed] [Google Scholar]
  • 123. Meloni G., Crameri A., Fritz G., et al., “The Catalytic Redox Activity of Prion Protein–CuII is Controlled by Metal Exchange With the ZnII–Thiolate Clusters of Zn7Metallothionein‐3,” ChemBioChem 13, no. 9 (2012): 1261–1265. [DOI] [PubMed] [Google Scholar]
  • 124. Kang Y. C., Son M., Kang S., et al., “Cell‐Penetrating Artificial Mitochondria‐Targeting Peptide‐Conjugated Metallothionein 1A Alleviates Mitochondrial Damage in Parkinson's Disease Models,” Experimental & Molecular Medicine 50, no. 8 (2018): 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Manoharan S., Guillemin G. J., Abiramasundari R. S., Essa M. M., Akbar M., and Akbar M. D., “The Role of Reactive Oxygen Species in the Pathogenesis of Alzheimer's Disease, Parkinson's Disease, and Huntington's Disease: A Mini Review,” Oxidative Medicine and Cellular Longevity 16 (2016): 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Sharma S. and Ebadi M., “Metallothioneins as Early and Sensitive Biomarkers of Redox Signaling in Neurodegenerative Disorders,” IIOAB Journal 2, no. 6 (2011): 98–106. [Google Scholar]
  • 127. Sharma S., Moon C. S., Khogali A., et al., “Biomarkers in Parkinson's Disease (Recent Update),” Neurochemistry International 63, no. 3 (2013): 201–229. [DOI] [PubMed] [Google Scholar]
  • 128. Tamai Y., Iwasa M., Eguchi A., et al., “Serum Copper, Zinc and Metallothionein Serve as Potential Biomarkers for Hepatocellular Carcinoma,” PLoS One 15, no. 8 (2020): e0237370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Yan D. W., Fan J. W., Yu Z., et al., “Downregulation of Metallothionein 1F, a Putative Oncosuppressor, by Loss of Heterozygosity in Colon Cancer Tissue,” Biochimica et Biophysica Acta (BBA)‐Molecular Basis of Disease 1822, no. 6 (2012): 918–926. [DOI] [PubMed] [Google Scholar]
  • 130. Liu X., Quan J., Shen Z., et al., “Metallothionein 2A (MT2A) Controls Cell Proliferation and Liver Metastasis by Controlling the MST1/LATS2/YAP1 Signaling Pathway in Colorectal Cancer,” Cancer Cell International 22, no. 1 (2022): 205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Kamal H. M., EL Sayed W. M., Ibrahem A. H., EL Sokary M. A., and Behiry E. G., “Study of metallothionein‐2A mRNA Relative Expression and Oxidant Status in Females With Breast Cancer,” Meta Gene 24 (2020): 100678. [Google Scholar]
  • 132. Sinduja P., Ramani P., Gheena S., and Ramasubramanian A., “Expression of Metallothionein in Oral Squamous Cell Carcinoma: A Systematic Review,” Journal of Oral and Maxillofacial Pathology 24, no. 1 (2020): 143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Zhang Y., Tian Y., Zhang H., Xu B., and Chen H., “Potential Pathways of Zinc Deficiency‐Promoted Tumorigenesis,” Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie 133 (2021): 110983. [DOI] [PubMed] [Google Scholar]
  • 134. Lim D., Jocelyn K. M. X., Yip G. W. C., and Bay B. H., “Silencing the Metallothionein‐2A Gene Inhibits Cell Cycle Progression From G1‐to S‐Phase Involving ATM and cdc25A Signaling in Breast Cancer Cells,” Cancer Letters 276, no. 1 (2009): 109–117. [DOI] [PubMed] [Google Scholar]
  • 135. Guan C., Zou X., Shi W., et al., “Metallothionein 1B Attenuates Inflammation and Hepatic Steatosis in MASH by Inhibiting the AKT/PI3K Pathway,” Journal of Lipid Research 66, no. 1 (2025): 100701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Bezerra G. M. R., Pontes F. S. C., Conte Neto N., et al., “Nuclear Metallothionein in Oral Squamous Cell Carcinoma: Clinicopathological Parameters and Patient Survival,” Brazilian Oral Research 32 (2018): e105. [DOI] [PubMed] [Google Scholar]
  • 137. Eriten B. and Goze F., “Comparison of the Effects of C‐KIT, MMP‐2, Ki‐67, Bcl‐2 and Metallothionein Expression on Prognostic Factors in Skin and Non‐Skin Malignant Melanomas,” Medicine Science | International Medical Journal 11, no. 3 (2022): 953. [Google Scholar]
  • 138. Wahyudi L. D., Yu S. H., and Cho M. K., “The Effect of Curcumin on the Cadmium‐Induced Mitochondrial Apoptosis Pathway by Metallothionein 2A Regulation,” Life Sciences 310 (2022): 121076. [DOI] [PubMed] [Google Scholar]
  • 139. Li K., Zhang Z., Mei Y., et al., “Metallothionein‐1G Suppresses Pancreatic Cancer Cell Stemness by Limiting Activin A Secretion via NF‐κB Inhibition,” Theranostics 11, no. 7 (2021): 3196–3212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Ha J. H., Prela O., Carpizo D. R., and Loh S. N., “p53 and Zinc: A Malleable Relationship,” Frontiers in Molecular Biosciences 9 (2022): 895887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Ostrakhovitch E. A., Olsson P. E., Jiang S., and Cherian M. G., “Interaction of Metallothionein With Tumor Suppressor p53 Protein,” FEBS Letters 580, no. 5 (2006): 1235–1238. [DOI] [PubMed] [Google Scholar]
  • 142. Pan Y., Huang J., Xing R., et al., “Metallothionein 2A Inhibits NF‐κB Pathway Activation and Predicts Clinical Outcome Segregated With TNM Stage in Gastric Cancer Patients Following Radical Resection,” Journal of Translational Medicine 11, no. 1 (2013): 173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Marikar F. M. M. T., Jin G., Sheng W., Ma D., and Hua Z., “Metallothionein 2A an Interactive Protein Linking Phosphorylated FADD to NF‐κB Pathway Leads to Colorectal Cancer Formation,” Chinese Clinical Oncology 5, no. 6 (2016): 76. [DOI] [PubMed] [Google Scholar]
  • 144. Mao J., Yu H., Wang C., et al., “Metallothionein MT1M is a Tumor Suppressor of Human Hepatocellular Carcinomas,” Carcinogenesis 33, no. 12 (2012): 2568–2577. [DOI] [PubMed] [Google Scholar]
  • 145. Takahashi S., “Molecular Functions of Metallothionein and Its Role in Hematological Malignancies,” Journal of Hematology & Oncology 5, no. 1 (2012): 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Cardoso S. V., Silveira‐Júnior J. B., De Carvalho Machado V., De‐Paula A. M., Loyola A. M., and De Aguiar M. C., “Expression of Metallothionein and p53 Antigens Are Correlated in Oral Squamous Cell Carcinoma,” Anticancer Research 29, no. 4 (2009): 1189–1193. [PubMed] [Google Scholar]
  • 147. Cho Y. H., Lee S. H., Lee S. J., Kim H. N., and Koh J. Y., “A Role of metallothionein‐3 in Radiation‐Induced Autophagy in Glioma Cells,” Scientific Reports 10, no. 1 (2020): 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Voels B., Wang L., Sens D. A., Garrett S. H., Zhang K., and Somji S., “The Unique C‐And N‐Terminal Sequences of Metallothionein Isoform 3 Mediate Growth Inhibition and Vectorial Active Transport in MCF‐7 Cells,” BMC Cancer 17 (2017): 369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Pastuszewski W., Dziegiel P., Krecicki T., et al., “Prognostic Significance of Metallothionein, p53 Protein and Ki‐67 Antigen Expression in Laryngeal Cancer,” Anticancer Research 27, no. 1A (2007): 335–342. [PubMed] [Google Scholar]
  • 150. Fan L. Z. and Cherian M. G., “Potential Role of p53 on Metallothionein Induction in Human Epithelial Breast Cancer Cells,” British Journal of Cancer 87, no. 9 (2002): 1019–1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Dutsch‐Wicherek M., “The Possible Biological Role of Metallothionein in Apoptosis,” Frontiers in Bioscience 13, no. 9 (2008): 4029–4038. [DOI] [PubMed] [Google Scholar]
  • 152. Pedersen M. Ø., Larsen A., Stoltenberg M., and Penkowa M., “The Role of Metallothionein in Oncogenesis and Cancer Prognosis,” Progress in Histochemistry and Cytochemistry 44, no. 1 (2009): 29–64. [DOI] [PubMed] [Google Scholar]
  • 153. Shimoda R., “Metallothionein is a Potential Negative Regulator of Apoptosis,” Toxicological Sciences 73, no. 2 (2003): 294–300. [DOI] [PubMed] [Google Scholar]
  • 154. Shen Y., Wang H., and Pan C., “Effect of Serum Tumor Necrosis Factor Receptor P55 and Metallothionein 1E on Clinical Outcome of Estrogen Receptor Positive Breast Cancer Patients Undergoing Radical Mastectomy,” Journal of Clinical Surgery 32, no. 5 (2024): 490–493. [Google Scholar]
  • 155. Kanda M., Nomoto S., Okamura Y., et al., “Detection of Metallothionein 1G as a Methylated Tumor Suppressor Gene in Human Hepatocellular Carcinoma Using a Novel Method of Double Combination Array Analysis,” International Journal of Oncology 35, no. 3 (2009): 477–483. [DOI] [PubMed] [Google Scholar]
  • 156. Sakamoto L. H. T., De Camargo B., Cajaiba M., Soares F. A., and Vettore A. L., “MT1G Hypermethylation: A Potential Prognostic Marker for Hepatoblastoma,” Pediatric Research 67, no. 4 (2010): 387–393. [DOI] [PubMed] [Google Scholar]
  • 157. Tao Y. F., Xu L. X., Lu J., et al., “Metallothionein III (MT3) is a Putative Tumor Suppressor Gene That is Frequently Inactivated in Pediatric Acute Myeloid Leukemia by Promoter Hypermethylation,” Journal of Translational Medicine 12, no. 1 (2014): 182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Wang Y., Wang G., Tan X., et al., “MT1G Serves as a Tumor Suppressor in Hepatocellular Carcinoma by Interacting With P53,” Oncogenesis 8, no. 12 (2019): 67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Wojtczak B., Pula B., Gomulkiewicz A., et al., “Metallothionein Isoform Expression in Benign and Malignant Thyroid Lesions,” Anticancer Research 37, no. 9 (2017): 5179–5185. [DOI] [PubMed] [Google Scholar]
  • 160. Fu J., Lv H., Guan H., et al., “Metallothionein 1G Functions as a Tumor Suppressor in Thyroid Cancer Through Modulating the PI3K/Akt Signaling Pathway,” BMC Cancer 13, no. 1 (2013): 462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Wierzowiecka B., Gomulkiewicz A., Cwynar‐Zajac L., et al., “Expression of Metallothionein and Vascular Endothelial Growth Factor Isoforms in Breast Cancer Cells,” In Vivo (Athens, Greece) 30, no. 3 (2016): 271–278. [PubMed] [Google Scholar]
  • 162. Si M. and Lang J., “The Roles of Metallothioneins in Carcinogenesis,” Journal of Hematology & Oncology 11, no. 1 (2018): 1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Zitka O., Kukacka J., Krizkov S., et al., “Matrix Metalloproteinases,” Current Medicinal Chemistry 17, no. 31 (2010): 3751–3768. [DOI] [PubMed] [Google Scholar]
  • 164. Farina A. and Mackay A., “Gelatinase B/MMP‐9 in Tumour Pathogenesis and Progression,” Cancers 6, no. 1 (2014): 240–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Zalewska M., Trefon J., and Milnerowicz H., “The Role of Metallothionein Interactions With Other Proteins,” Proteomics 14, no. 11 (2014): 1343–1356. [DOI] [PubMed] [Google Scholar]
  • 166. Zhao Y., Zhao L., Mao T., and Zhong L., “Assessment of Risk Based on Variant Pathways and Establishment of an Artificial Neural Network Model of Thyroid Cancer,” BMC Medical Genetics 20, no. 1 (2019): 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Zheng Y., Jiang L., Hu Y., et al., “Metallothionein 1H (MT1H) Functions as a Tumor Suppressor in Hepatocellular Carcinoma Through Regulating Wnt/β‐catenin Signaling Pathway,” BMC Cancer 17 (2017): 161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Butcher H. L., Kennette W. A., Collins O., Zalups R. K., and Koropatnick J., “Metallothionein Mediates the Level and Activity of Nuclear Factor κB in Murine Fibroblasts,” The Journal of Pharmacology and Experimental Therapeutics 310, no. 2 (2004): 589–598. [DOI] [PubMed] [Google Scholar]
  • 169. Kimura T. and Itoh N., “Function of Metallothionein in Gene Expression and Signal Transduction: Newly Found Protective Role of Metallothionein,” Journal of Health Science 54, no. 3 (2008): 251–260. [Google Scholar]
  • 170. Kim C. H., Kim J. H., Lee J., and Ahn Y. S., “Zinc‐Induced NF‐κB Inhibition Can Be Modulated by Changes in the Intracellular Metallothionein Level,” Toxicology and Applied Pharmacology 190, no. 2 (2003): 189–196. [DOI] [PubMed] [Google Scholar]
  • 171. Cherian M., “Metallothioneins in Human Tumors and Potential Roles in Carcinogenesis,” Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis 533, no. 1–2 (2003): 201–209. [DOI] [PubMed] [Google Scholar]
  • 172. Jin X., Tian Y., Zhu H., Sun Y., and Zhang Z., “Computer‐Aided Analysis Reveals Metallothionein‐Positive Cancer‐Associated Fibroblasts Promote Angiogenesis in Gastric Adenocarcinoma,” Discover Oncology 15, no. 1 (2024): 751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Krizkova S., Kepinska M., Emri G., et al., “An Insight Into the Complex Roles of Metallothioneins in Malignant Diseases With Emphasis on (Sub)Isoforms/Isoforms and Epigenetics Phenomena,” Pharmacology & Therapeutics 183 (2018): 90–117. [DOI] [PubMed] [Google Scholar]
  • 174. Person R. J., Tokar E. J., Xu Y., Orihuela R., Ngalame N. N. O., and Waalkes M. P., “Chronic Cadmium Exposure In Vitro Induces Cancer Cell Characteristics in Human Lung Cells,” Toxicology and Applied Pharmacology 273, no. 2 (2013): 281–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Dziegiel P., Pula B., Kobierzycki C., et al., “The Role of Metallothioneins in Carcinogenesis,” Metallothioneins in Normal and Cancer Cells 1 (2016): 29–63. [DOI] [PubMed] [Google Scholar]
  • 176. Huang M., Lin Y., Wang C., et al., “New Insights Into Antiangiogenic Therapy Resistance in Cancer: Mechanisms and Therapeutic Aspects,” Drug Resistance Updates 64 (2022): 100849. [DOI] [PubMed] [Google Scholar]
  • 177. Rodrigo M. A. M., Michalkova H., Strmiska V., et al., “Metallothionein‐3 Promotes Cisplatin Chemoresistance Remodelling in Neuroblastoma,” Scientific Reports 11, no. 1 (2021): 5496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Masiulionytė B., Valiulytė I., Tamašauskas A., and Skiriutė D., “Metallothionein Genes Are Highly Expressed in Malignant Astrocytomas and Associated With Patient Survival,” Scientific Reports 9, no. 1 (2019): 5406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Wong D. L. and Stillman M. J., “Capturing Platinum in Cisplatin: Kinetic Reactions With Recombinant Human Apo‐Metallothionein 1A,” Metallomics 10, no. 5 (2018): 713–721. [DOI] [PubMed] [Google Scholar]
  • 180. Tariba B., Živković T., Krasnići N., et al., “Serum Metallothionein in Patients With Testicular Cancer,” Cancer Chemotherapy and Pharmacology 75 (2015): 813–820. [DOI] [PubMed] [Google Scholar]
  • 181. Rodrigo M. A. M., Michalkova H., Jimenez A. M. J., et al., “Metallothionein‐3 is a Multifunctional Driver That Modulates the Development of Sorafenib‐Resistant Phenotype in Hepatocellular Carcinoma Cells,” Biomarker Research 12, no. 1 (2024): 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Dziegiel P., Pula B., Kobierzycki C., et al., “The Role of Metallothioneins in Carcinogenesis,” Metallothioneins in Normal and Cancer Cells 1 (2016): 29–63. [DOI] [PubMed] [Google Scholar]
  • 183. Guo S., Huang F., and Guo P., “Construction of Folate‐Conjugated pRNA of Bacteriophage phi29 DNA Packaging Motor for Delivery of Chimeric siRNA to Nasopharyngeal Carcinoma Cells,” Gene Therapy 13, no. 10 (2006): 814–820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Kang H., Ga Y. J., Kim S. H., et al., “Small Interfering RNA (siRNA)‐Based Therapeutic Applications Against Viruses: Principles, Potential, and Challenges,” Journal of Biomedical Science 30, no. 1 (2023): 88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Tarapore P., Shu Y., Guo P., and Ho S. M., “Application of phi29 Motor pRNA for Targeted Therapeutic Delivery of siRNA Silencing Metallothionein‐IIA and Survivin in Ovarian Cancers,” Molecular Therapy 19, no. 2 (2011): 386–394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Meng K., Song J., Qi F., Li J., Fang Z., and Song L., “MT1G Promotes Iron Autophagy and Inhibits the Function of Gastric Cancer Cell Lines by Intervening in GPX4/SQSTM1,” Scientific Reports 14, no. 1 (2024): 28539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Wang X. L., Schnoor M., and Yin L. M., “Metallothionein‐2: An Emerging Target in Inflammatory Diseases and Cancers,” Pharmacology & Therapeutics 244 (2023): 108374. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.


Articles from Health Science Reports are provided here courtesy of Wiley

RESOURCES