Skip to main content

Zinc deficiency as possible link between immunosenescence and age-related diseases

Abstract

As global life expectancy increases, research reveals a critical challenge in aging: the progressive deterioration of immune function, termed immunosenescence. This age-related immune decline is characterized by a complex dysregulation of immune responses, which leaves older adults increasingly vulnerable to infections, chronic inflammatory states, and various degenerative diseases. Without intervention, immunosenescence significantly contributes to morbidity and mortality among the elderly, intensifying healthcare burdens and diminishing quality of life on both individual and societal levels. This review explores the essential role of zinc, a trace element critical for immune health, in mitigating the impact of immunosenescence and slowing the cascade of immunological dysfunctions associated with aging. By modulating the activity of key immune cells and pathways, zinc supplementation emerges as a promising approach to strengthen immunity, reduce oxidative stress, and counteract "inflammaging," a state of chronic, low-grade inflammation that accelerates tissue damage and drives disease progression. Zinc’s involvement in cellular defense and repair mechanisms across the immune system highlights its ability to enhance immune cell functionality, resilience, and adaptability, strengthening the body's resistance to infection and its ability to manage stressors that contribute to diseases of aging. Indeed, zinc has demonstrated potential to improve immune responses, decrease inflammation, and mitigate the risk of age-related conditions including diabetes, depression, cardiovascular disease, and vision loss. Given the prevalent barriers to adequate zinc intake among older adults, including dietary limitations, decreased absorption, and interactions with medications, this review underscores the urgent need to address zinc deficiency in aging populations. Recent findings on zinc’s cellular and molecular effects on immune health present zinc supplementation as a practical, accessible intervention for supporting healthier aging and improving quality of life. By integrating zinc into targeted strategies, public health efforts may not only sustain immunity in the elderly but also extend healthy longevity, reduce healthcare costs, and potentially mitigate the incidence and impact of chronic diseases that strain healthcare systems worldwide.

Background

As the global population continues to age, the prevalence of age-related diseases such as cardiovascular disorders, neurodegenerative conditions, diabetes, and various types of cancer is expected to increase substantially. This demographic shift presents critical public health challenges, as older adults face not only the physical effects of these conditions but also an elevated vulnerability to infection and immune dysfunction [1]. A key factor in this vulnerability is the age-related decline in immune function, a phenomenon known as immunosenescence, which affects both innate and adaptive parts of the immune system. Immunosenescence results in a compromised immune response, impairing the body´s ability to defend itself against pathogens and adapt to new infections [2]. Compounding these effects is “inflammaging”, a state of chronic, low-grade inflammation that develops with age and further disrupts immune regulation. Inflammaging is associated with increased susceptibility to infections, autoimmune disorders and various age-related diseases, creating a feedback loop that accelerates the breakdown of immune health and increases morbidity and mortality risks among elderly [3, 4].

One of the essential elements in supporting immune health, particularly in the context of aging, is zinc, a trace element involved in numerous biological processes. Zinc plays a critical role in immune maintenance and regulation, as it influences DNA synthesis, cell division, and antioxidant defense mechanisms, functions vital to the integrity and effectiveness of immune responses. Zinc is necessary for the optimal function of key immune cells, including, neutrophils, macrophages, natural killer (NK) cells, and lymphocytes, all of which contribute to a robust and responsive immune system [5]. Among older adults, zinc deficiency is a common issue, arising from factors such as reduced dietary intake, decreased absorption efficiency, and altered metabolic requirements [6, 7]. This deficiency exacerbates immunosenescence and inflammaging, leading to increased inflammation, impaired immune defense, and a greater risk of chronic diseases.

The governmental German nutrition study conducted in 2008 revealed that 44% of men and 27% of women aged 65 to 80 years did not reach the recommended daily zinc intake [8]. Since then, dietary zinc intake recommendations have been revised upward. When the 2008 data are reanalyzed using the current guidelines established by the European Food Safety Authority (EFSA) [9], it is estimated that approximately 75% of individuals aged 65 to 80 years in Germany do not reach the recommended dietary zinc intake. These individuals are not definitively zinc deficient but are at greater risk for zinc deficiency or may have an inadequate zinc status, as zinc deficiency is defined by serum zinc levels below 70 µg/dL in fasting state [10]. Given this high prevalence of inadequate zinc status and likely also zinc deficiency with its adverse effect on immune function in aging populations [11], understanding and addressing zinc´s role in the immune system is critical to develop strategies that support healthier aging.

Recent research has shed light on the potential of zinc supplementation as a valuable approach to counteract immunosenescence and its associated health impacts. Zinc has been shown to modulate several crucial immune pathways, enhancing chemotaxis in neutrophils [12], promoting cytotoxic activity in NK cells [13], supporting macrophage phagocytic function [14], and preserving responsiveness of T and B cells [15, 16], which are essential components of adaptive immunity. Furthermore, zinc´s antioxidant properties contribute to its protective role against oxidative stress, a major driver of cellular aging and tissue damage in older adults [17]. Zinc also influences key gene expression pathways, including the nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-κB) and peroxisome proliferator-activated receptor (PPAR) signaling, which are critical for managing inflammatory responses and enhancing cellular resilience. By targeting these pathways, zinc can potentially reduce systemic inflammation and oxidative damage, thereby supporting immune cell health and function across both innate and adaptive immunity [18, 19].

Beyond its cellular effects, zinc´s influence has broader potential in impacting the aging process and addressing various age-related diseases. By correcting cellular dysfunction often seen with aging, zinc supplementation may help delay the onset and progression of conditions such as age-related macular degeneration [20], diabetes mellitus [21], wound-healing impairments [22], depression [23] and cardiovascular disease [24], ultimately improving health outcomes for older adults. Zinc´s ability to reduce inflammation and enhance immune response makes it a promising agent for mitigating these chronic diseases, which are prevalent in aging populations and often worsen with immune decline. Its antioxidative and anti-inflammatory properties are essential for cellular protection and repair, aiding in the maintenance of tissue integrity and function across multiple organ systems as individuals age. In AMD, zinc may help to slow down retinal degeneration and preserve vision [20, 25]. In diabetes, it plays a role in insulin regulation and blood sugar control [21]. In wound healing it supports tissue regeneration and immune defense [22]. In depression it influences neurotransmitter balance and reduces inflammation [26, 27], and in cardiovascular disease, zinc helps to reduce oxidative stress and vascular inflammation [17, 28], which are central to preserving heart health in aging populations.

Unlike certain nutrients, such as fat-soluble vitamins or iron, which the body can store for extended periods, zinc lacks a dedicated storage system [29, 30]. This means the body depends on a steady dietary supply to maintain adequate levels for vital physiological functions [31]. This is especially concerning for vulnerable populations, such as older adults, who may face challenges in maintaining adequate zinc intake, as they may experience reduced appetite, changes in taste, dietary restrictions, poor dental status and age-related digestive issues that hinder nutrient absorptions [32,33,34,35]. Medications, such as penicillamine, tetracyclines, ACE inhibitors and angiotensin-2 receptor blockers, frequently prescribed in the aged population can also interfere with zinc absorption, further compounding the risk of deficiency. Thiazide diuretics increase urinary excretion of zinc [36,37,38,39].

Limited dietary variety and a decrease in overall nutrient intake can result in lower zinc levels among elderly individuals, creating a deficiency that has significant implications for their immune health and overall well-being. Addressing zinc deficiency through dietary improvements and targeted supplementation could therefore play a critical role in maintaining immune resistance and reducing the impact of immunosenescence.

This review offers a comprehensive examination of the role of zinc in supporting immune homeostasis and countering the effect of immunosenescence. By exploring zinc´s influence on each component of the immune system, from innate responses mediated by neutrophils and macrophages to adaptive immunity regulated by T and B lymphocytes, this review underlines the importance of zinc for immune health in aging populations. Furthermore, the review delves into the clinical evidence supporting zinc supplementation as a strategy to decrease the severity of age-associated inflammatory conditions and chronic diseases, highlighting its potential to improve quality of life for older adults. By analyzing zinc´s molecular pathways, cellular interactions, and its broader impact on inflammation, this review aims to emphasize the significance of adequate zinc intake and illustrate how zinc supplementation could serve as a feasible, accessible, and effective approach to support immune health and enhance longevity among aging individuals (Fig. 1).

Fig. 1
figure 1

Overview of Zinc Status and Its Impact on Immune Function in Elderly Individuals Dysregulated immune responses and unhealthy increased reactions associated with zinc deficiency,: Restored immune balance and improved immune function achieved through zinc supplementation, (↑): increased, (↓): decreased, (↔): balanced (a) In aged and zinc deficient individuals, immune system dysregulation is characterized by increased pro-inflammatory cytokine production (IL-1, IL-6, TNF-α) by antigen-presenting cells, enhanced Th2 responses, and reduced Th1 responses. This leads to decreased IFN-γ and IL-2 production, reduced immunoglobulin (Ig) class switching, unspecific B cell activation, and decreased T regulatory cell function, promoting an inflammatory state and impaired immune balance. b Zinc supplementation restores immune homeostasis by reducing inflammation, balancing Th1/Th2 responses, and maintaining normal cytokine levels and B cell function. The presence of zinc supports APC function, enhances regulatory T cell​ activity, and promotes a balanced immune response in elderly individuals, mitigating immunosenescence. Created in https://BioRender.com

Zinc and its key proteins

Zinc is an essential trace element that plays a critical role in numerous biological processes, functioning both as a structural and catalytic cofactor. It is indispensable for the activity of more than 300 enzymes [40] and is involved in a wide range of cellular functions, including protein synthesis, DNA repair, and cell division [41]. Its cellular regulation is tightly controlled by zinc transporters, particularly the Zip (Zrt/Irt-like protein) family (Zip1-14/SLC39 A1-14), which facilitates zinc influx into the cytoplasm, and the ZnT (Zinc Transporter) family (ZnT1-10/SLC30 A1-10), which mediates zinc efflux to maintain homeostasis. Among the Zip family, ZIP4 is especially critical, as it is responsible for dietary zinc uptake in the intestine [42]. Mutations in Zip4 lead to acrodermatitis enteropathica, a rare but severe zinc deficiency disorder [43]. Zip7 is critical for intracellular zinc release from the endoplasmic reticulum into the cytosol and is essential for proper B-cell receptor signaling. A mutation in Zip7 has been identified as the cause of an autosomal recessive disorder, which is characterized by the lack of B cells and immunoglobulins, resulting in early-onset infections [44]. Other important ZIPs include Zip8, which is involved in T cell activation and IFN-γ expression [45], and Zip14, which plays a role in zinc mobilization during infection and acute-phase responses [46].

In contrast, the ZnT family primarily functions to export zinc out of the cytoplasm or into organelles. ZnT1 is the most well-known member, as it is responsible for reducing intracellular zinc levels by exporting it across the plasma membrane [42]. ZnT2 is critical for zinc secretion into breast milk [47], while ZnT3 is essential in neurons for packaging zinc into synaptic vesicles, a process vital for neurotransmission and brain function [27].

Metallothioneins (MTs), small cysteine-rich proteins, act as zinc-binding proteins to buffer and store zinc ions, protecting cells from oxidative stress and regulating zinc availability [48]. Zinc predominantly functions as a cofactor in enzymes, stabilizing protein structures through its interaction with specific amino acid residues, such as cysteine and histidine, thereby contributing to the secondary and tertiary of target proteins [49, 50]. It is essential in forming zinc finger motifs, which are critical for DNA binding and transcriptional regulation. Additionally, zinc ions can regulate enzymatic activity, DNA binding, and protein–protein interactions by inducing conformational changes in target proteins [49]. Unlike many cofactors that directly participate in redox reactions, zinc's biological role largely involves structural stabilization and catalytic activity, making it indispensable in processes such as transcription, signal transduction, immune function, and apoptosis [5] (Tables 1 and 2).

Table 1 Zinc transporters and their immunological relevance
Table 2 Zip proteins and their immunological relevance

Recommended zinc intake

Various international organizations, including the European Food Safety Authority (EFSA), the German Nutrition Society (DGE), the National Institutes of Health (NIH) and the World Health Organization (WHO), provide guidelines for zinc intake to support optimal health (Table 3).

Table 3 Comparison of zinc intake recommendations among different health organizations

EFSA provides zinc intake recommendations that account for dietary phytate levels. Phytates are naturally occurring compounds found in plant-based foods like grains, legumes, and seeds that bind to zinc in the digestive tract, reducing its absorption and bioavailability. For men, the recommended intake ranges from 9.4 mg per day with low phytate levels (300 mg/day) to 16.3 mg per day with high phytate intake (1200 mg/day). For women, the recommendations range from 7.5 mg to 12.7 mg per day across the same phytate levels [9]. Similarly, DGE suggests zinc intakes of 11 mg per day for men and 7 mg per day for women when phytate levels are low (330 mg/day). At higher phytate levels (990 mg/day), these recommendations rise to 16 mg per day for men and 10 mg per day for women [89].

WHO bases its zinc recommendations on bioavailability. In diets with high zinc bioavailability, men require 4.2 mg per day and women 3.0 mg per day. For moderate bioavailability, the recommended intake increases to 7.0 mg per day for men and 4.9 mg per day for women. Low bioavailability diets necessitate the highest zinc intakes: 14.0 mg per day for men and 9.8 mg per day for women [90]. Meanwhile, NIH recommend zinc intakes that are independent of phytate levels, suggesting 11 mg per day for men and 8 mg per day for women [91].

Excessive zinc intake, however, can lead to toxicity and significant health complications. Symptoms of acute zinc toxicity include gastrointestinal distress, such as nausea, vomiting, abdominal cramps, and diarrhea [92, 93]. Chronic high zinc consumption poses more serious risks, such as immune dysfunction and interference with copper metabolism. Zinc competes with copper for absorption in the gastrointestinal tract, and prolonged excessive zinc intake can lead to copper deficiency. This condition may result in anemia, neutropenia, and neurological symptoms such as ataxia and peripheral neuropathy. Other symptoms of zinc toxicity include headaches, fatigue, and disruptions in lipid metabolism [92,93,94].

To minimize these risks, tolerable upper intake levels (ULs) for zinc have been established by various organizations. EFSA and DGE set the UL at 25 mg per day for adults [9], while the NIH recommends a higher UL of 40 mg per day [91]. However, the upper limit of 40 mg is likely too high, as studies indicate that negative side effects can occur below this threshold. A limit of 25 mg may be a more appropriate upper boundary for zinc intake [92].

To assess whether dietary zinc intake is sufficient or excessive, the use of a food questionnaire can be particularly valuable. This tool helps to estimate zinc intake by evaluating the frequency and quantity of zinc-rich or phytate-rich foods in the diet, providing an effective method for determining whether dietary adjustments are necessary to meet individual nutritional needs [95]. Furthermore, an application accessible at www.zink-app.de enables individuals to assess their individual zinc status [96].

Sex- and gender-related differences in immunity

Immunosenescence differs significantly between sexes and genders, influenced by biological, hormonal, and environmental factors. Women generally exhibit stronger immune responses than men throughout much of their lives, a phenomenon attributed to differences in sex hormones and genetic factors [97]. Estrogen receptors are present on many immune cells, including neutrophils, macrophages, dendritic cells, natural killer cells, T lymphocytes, and B lymphocytes [98]. Estrogen, a key hormone in females, plays a critical role in enhancing immune function. It stimulates the production of immunoglobulin (Ig) G and IgM antibodies [99], whereas testosterone has the opposite effect, reducing the production of these immunoglobulins [100]. Women also have higher numbers of CD4+ T cells [101], and estrogen increases the expression of interferon (IFN)-γ, an important cytokine for immune activation [102, 103].

The phagocytic activity of neutrophils, macrophages, and other antigen-presenting cells is more pronounced in females than in males [104]. Additionally, chronic estradiol application promotes the expression of interleukin (IL)−1β, IL-6, IL-12p40, and Inducible nitric oxide synthase (iNOS) in macrophages following lipopolysaccharide (LPS) stimulation, further enhancing immune responses [105]. Women also produce significantly more IFN-α after Toll-like receptor (TLR) 7 activation compared to men [106, 107]. These factors collectively provide women with a robust capacity to combat infections and mount stronger immune responses.

The genetic advantage conferred by the presence of two X chromosomes in females further contributes to their immunological resilience. While the X chromosome does carry numerous immune-related genes, the biological implications are more nuanced than simple redundancy. To balance gene expression differences, one of the two X chromosomes in female cells undergoes X chromosome inactivation (XCI), a process that epigenetically silences most genes on the inactive X chromosome (Xi). Approximately 15%–30% of Xi-linked genes avoid this silencing, with patterns differing across cellular context, genetic background and developmental stages [108, 109]. This incomplete inactivation mosaicism results in a partially higher gene expression and provides a functional diversity that can enhance immune responses and compensate deleterious mutations. In contrast, males, with only one X chromosome, lack both the protective effects of mosaicism and the potential benefits of increased gene expression conferred by XCI escape. This may contribute to sex-based differences in immune function and disease susceptibility, while in females, the same mechanism may lead to heightened immune responses and an increased risk of autoimmunity [98, 110].

However, this sex-based advantage diminishes with age, particularly after menopause. The menopausal transition involves a significant decline in estrogen levels, which negatively impacts the immune system [111]. The protective effects of estrogen wane, leading to a reduction in immune cell populations. Postmenopausal women exhibit lower numbers of B lymphocytes and CD4+ T lymphocytes [112]. Chronic increases in inflammatory markers, including IL-1β, IL-6, and IL-10, are observed in serum during and after menopause [113,114,115]. Although IFN-γ levels initially rise during menopause, they eventually decrease to levels lower than those observed before menopause [113]. A comparison of the gene expression profiles of peripheral blood mononuclear cells (PBMCs) in elderly men and women with younger controls revealed that 48 signaling pathways are altered in females, compared to only 29 in males. This finding suggests that the aging immune system undergoes more extensive changes in women, particularly in T cell protection, which weakens, while inflammation increases [116].

This hormonal shift largely explains the narrowing of the immunological gap between postmenopausal women and men of the same age. Before menopause, men are more at risk for infections such as hepatitis, meningococcal disease, and pneumococcal disease due to their comparatively weaker immune responses. However, as women age and their immunological advantage diminishes and their risk for these infections increases to levels similar to those observed in men [117]. Hormone replacement therapy has been shown to mitigate some of these immunological alterations. For example, it increases the number of B cells [118] and decreases chronically elevated pro-inflammatory cytokines, such as IL-6, through estrogen supplementation [119].

Gender also intersects with environmental and social factors that influence aging and immune function. Societal norms and behaviors may lead to differences in lifestyle, access to healthcare, and exposure to chronic stress, all of which impact the immune system over time [120]. Furthermore, non-binary and transgender individuals may experience unique interactions between hormonal therapies, environmental factors, and aging. However, this area remains under-researched, highlighting the need for further studies.

In summary, while women generally possess stronger immune function than men due to hormonal and genetic advantages, this disparity diminishes after menopause as estrogen levels decline. Since this review focuses on the immunological changes in elderly individuals, particularly postmenopausal populations, further exploration of sex differences will not be addressed in detail.

Changes in innate immunity

Granulocytes

Granulocytes, which include neutrophils, eosinophils, and basophils, play essential roles in the innate immune response. These cells primarily perform three key functions: phagocytosis, degranulation and the production of cytokines and chemokines [121]. However, age-related changes in neutrophil function have significant implications for the immune response (Fig. 2).

Fig. 2
figure 2

Functional Decline of Neutrophils in Aging and Zinc Deficiency. : Alterations due to aging, but not described in zinc deficiency, (Black) Alterations described in both, zinc deficiency and aging, (↑): increased, (↓): decreased Created in https://BioRender.com

Neutrophil function is thought to be compromised in older adults, as indicated by increased morbidity and death caused by bacterial infections with advancing age [122]. Interestingly, while the total number of neutrophils increases during ageing [123], evidence suggests that their functional capacity declines. Changes in the expansion of polymorphonuclear precursors to granulocyte-colony stimulating factor (G-CSF) have been observed in elderly individuals [124], although responses to other stimulants like IL-3 and granulocyte–macrophage colony stimulating factor (GM-CSF) remain unchanged, ensuring sufficient neutrophilia during infection [125, 126].

The neutrophil-to-lymphocyte ratio (NLR) increases during aging. It has been recognized as a prognostic biomarker for assessing the progression of neoplastic diseases, predicting the onset of chronic degenerative conditions, and serving as a potential indirect indicator of healthy aging. Notably, the NLR typically increases in association with neoplastic diseases, reflecting systemic inflammation, while a decrease in NLR may be indicative of healthy aging [123, 127, 128]. Consistent with its role in modulating immune function, plasma zinc is inversely associated with the neutrophil-to-lymphocyte ratio [129]. This suggests that zinc supplementation in the elderly may help restore a healthier neutrophil-to-lymphocyte ratio, potentially improving immune function during aging.

The functional decline in neutrophils is associated with several factors, leading to impaired apoptosis regulation. Older neutrophils are at a higher risk of apoptosis due to a lower responsiveness to factors such as G-CSF, IL-2, or tumor necrosis factor (TNF)-α that would normally prevent them from undergoing programmed cell death [130, 131]. This phenomenon may be linked to an altered ratio in anti-apoptotic and pro-apoptotic proteins. For example, the Mcl-1/Bax ratio increases following GM-CSF stimulation in neutrophils from individuals of a younger age, whereas this ratio remains unchanged in neutrophils from older individuals [132, 133]. Zinc supplementation, of patients with down syndrome, which often goes along with zinc deficiency, could reduce the number of apoptotic cells [134, 135]. There is a significant age-related decline in oxidative burst activity, particularly in older adults. In nonagenarians, there is a higher expression of macrophage-1 antigen (MAC-1), which is involved in neutrophil adhesion and migration, compared to younger and middle-aged individuals. Interestingly, neutrophil zinc content is higher in both nonagenarians and young adults compared to middle-aged individuals, suggesting a potential age-related redistribution of zinc. Additionally nonagenarians exhibit increased superoxide dismutase (SOD) activity, which may represent a compensatory mechanism against oxidative stress [136].

Zinc influences various neutrophil functions, including migration, chemotaxis, and the oxidative burst. Zinc deficiency impairs neutrophil chemotaxis, as shown in models of crush syndrome where zinc chelation reduced neutrophil infiltration and muscle injury, indicating that adequate zinc levels are crucial for optimal neutrophil function migration [137]. Zinc deficiency has also been linked to inflammatory diseases, including acute lung injury (ALI), characterized by excessive neutrophil recruitment and hyperactivation. However, administering zinc to mice 24 h before LPS-induced ALI significantly reduced neutrophil recruitment to the lungs, inhibited their hyperactivation, and consequently decreased lung damage [138].

In zinc-treated hemodialysis patients, granulocytes exhibit enhanced responsiveness to stimuli like zymosan-activated serum, showing greater chemokinetic activity and motility [139]. Zinc also promotes chemotaxis by increasing the expression of IL-8 [12, 140]. This could be due to a Zn-induced inhibition of phosphatase activity, leading to Mitogen-activated protein kinase (MAPK) and activator protein-1 (AP-1) activation, resulting in the enhanced IL-8 expression [140]. Additionally, zinc influences GM-CSF receptor expression on immune cells differently, increasing it on neutrophils but decreasing it on monocytes [141].

Neutrophils are critical first responders to infection or injury, and their ability to migrate toward chemotactic signals is essential for effective immune defense. Zinc, along with other trace elements like copper and nickel, enhances neutrophil chemotaxis, promoting their migration to sites of infection in a manner similar to the chemotactic factor N-formylmethionyl-leucyl-phenylalanine (fMLP). Zinc and nickel, in particular, double neutrophil migration towards fMLP, demonstrating a synergistic effect in enhancing chemotaxis [142]. Another study shows that chelation of zinc with the membrane permeable chelator N,N,N′,N′-tetrakis(2-pyridinylmethyl)−1,2-ethanediamine (TPEN) reduces granulocyte migration towards fMLP and IL-8 as well as phagocytosis, oxidative burst and granule release. But the direct action of zinc as chemoattractant, as previously mentioned [142], was not observed. TPEN also inhibited the secretion of both chemotactic IL-8 and anti-inflammatory cytokine IL-1ra in response to LPS stimulation [12]. Consistent with these findings that zinc deficiency impairs neutrophil chemotaxis, it has been observed that chemotaxis is diminished in elderly individuals as well [143,144,145,146,147].

After they arrived at their destination, polymorphonuclear neutrophils (PMN) have various options for neutralizing pathogens. The main ones are: Intracellular killing through oxidative burst, phagocytosis and degranulation. Most of them are constrained during zinc deficiency [12, 142]. Given that zinc deficiency is more prevalent in the elderly, ensuring adequate zinc intake may be particularly important for maintaining neutrophil function and reducing susceptibility to infections in older adults.

While it is established that phagocytosis is significantly impaired in the context of elderly individuals [147,148,149] as well as zinc deficiency [12], the impact of zinc on oxidative burst remains controversial. Zinc can both inhibit and enhance neutrophil respiratory burst, depending on the context. For instance, zinc inhibits oxidative burst in isolated neutrophils but enhances it in whole blood, suggesting that the cellular environment may influence zinc´s effect [150]. Moreover, zinc inhibits superoxide generation in peritoneal neutrophils at concentrations as low as 10 µM [151], while other studies report enhanced superoxide release and hydrogen peroxide production at higher zinc concentrations [152]. Zinc hydroxide has been found to induce both oxidative burst and phagocytosis in rat neutrophils, further highlighting its dual role in modulating immune responses [153]. However, in this study, zinc treatment in PMNs did not improve phagocytosis or bacterial killing of Staphylococcus aureus and Staphylococcus epidermidis, but it did improve cell membrane protection, potentially mitigating damage during immune responses [154]. In the context of aging, it has become increasingly evident that oxidative burst activity declines with age [136, 148, 155, 156]. However, this phenomenon appears to be context-dependent, as oxidative burst tends to decrease in suspended cells while exhibiting an increase in adhesive cells [157].

Zinc may influence neutrophil degranulation and oxidative metabolism by activating protein kinase C (PKC) and promoting myeloperoxidase (MPO) degranulation, leading to the production of reactive oxygen species (ROS), particularly hypochlorous acid, a potent antimicrobial compound [158, 159]. More recent studies show that zinc, at low concentrations (5–12.5 µM), activates NADPH oxidase via PKC, leading to superoxide radical production. Higher zinc concentrations facilitate the conversion of superoxide to hydrogen peroxide and HOCl through MPO, enhancing microbial killing [160].

Zinc´s role in NETosis, the process by which neutrophils release extracellular traps (NETs) composed of DNA, chromatin, and granule proteins to trap pathogens, is equally complex. PKC-mediated NET formation depends on ROS production by NADP oxidase. Zinc signaling is essential for NETosis, as zinc chelation by TPEN inhibits this process without affecting PKC activity or ROS production, suggesting zinc acts downstream of ROS [161]. Zinc supplementation increases NET release, although it does not alter NET efficacy in bacterial killing [162]. However, other studies show that zinc can inhibit NET release and degranulation, possibly by reducing citrullinated histone H3 levels [163]. The impact of aging on NETosis is not well studied yet. Some studies suggest reduced NET formation in neutrophils from older individuals [164, 165], while other ones indicate an increase in NET production with age [166]. However, this heightened production may be associated with diminished functionality and efficiency, likely due to alterations in NET composition and impaired clearance mechanisms [166].

Hence, maintaining adequate zinc levels is vital for supporting neutrophil function in the elderly, as these cells play a key role in the immune defense. Zinc enhances neutrophil activities such as migration, oxidative burst, and cytokine release, which tend to decline with age (Table 4).

Table 4 Comparison of granulocyte counts and functions in aged individuals and zinc deficient state

Mast cells

Mast cells are known to be central to various pathological processes that contribute to allergic reactions. Conditions such as anaphylaxis, atopic eczema, and asthma are among the disorders where their role is particularly significant [181].

The total number of mast cells has been found to increase with age [182, 183], a pattern that bears similarities to the response observed in zinc deficiency [184]. However, with advancing age, mast cells tend to enter a state of nonspecific preactivation, while their functional capacity upon stimulation becomes diminished [183]. This decline is reflected in the reduced frequency of degranulation observed in mast cells of aged skin [185].

Zinc is a necessary component for both degranulation as well as cytokine production in mast cells. The zinc chelator TPEN has been demonstrated to inhibit cytokine production, the secretion of lipid mediators and the release of histamine in mast cells. Conversely, zinc supplementation has been shown to reverse these inhibitory effects. Other metal chelators do not exert any influence on mast cell function [186]. Furthermore, mast cells express various zinc transporters, including ZnT4, which facilitate the uptake and storage of zinc. This is particularly important in the context of inflammation, where mast cells can rapidly regain high levels of labile zinc after degranulation [187]. In addition, findings demonstrate that ZnT5 is necessary for the development of contact hypersensitivity and mast cell-mediated delayed-type allergic responses, though it is not involved in immediate passive cutaneous anaphylaxis [57]. Zinc also alters the cytokine production of mast cells. L-type calcium channels mediate zinc signaling in mast cells, influencing the production of proinflammatory cytokines such as IL-6 in response to various stimuli, including LPS and IL-33 [188], (Table 5).

Table 5 Comparison of mast cell counts and functions in aged individuals and zinc deficient state

Natural killer cells

Aging significantly affects the number, function, and phenotype of NK cells, which are critical components of the innate immune system. While no changes in NK progenitor numbers have been observed in the peripheral blood or bone marrow [192], most studies report an increase in the total number of NK cells with age [193,194,195]. However, this increase is accompanied by a significant shift in NK cell subsets. There is a notable decrease in the percentage of CD56bright cells, which are primarily responsible for cytokine secretion, including IFN-γ and TNF-α [195,196,197]. Conversely, there is an increase in CD56dim cells, which are predominantly cytotoxic [196,197,198].

Despite the increased number of cytotoxic CD56dim cells, age-related declines in NK cell cytotoxicity have been reported. These include impaired IFN-γ secretion, reduced expression of perforin and granzyme, and defective perforin release through degranulation [199,200,201]. Consistent with these findings, in vitro zinc chelation has also been shown to decrease NK cell cytotoxicity, which could be increased through zinc supplementation [202].

The ability of NK cells to recognize and kill target cells is also affected by changes in receptor expression. Aging results in reduced expression of activating receptors like NKp30 and NKp46, while the expression of inhibitory receptors like KLRG1 and killer cell immunoglobulin-like receptors (KIRs) increases [198, 201, 203]. Human leucocyte antigen (HLA)-C-specific KIRs can bind zinc, and chelating zinc reverses their inhibitory function, suggesting that zinc is essential for their activity. Zinc induces KIR-self-association, leading to the formation of multimers, a phenomenon not observed with other divalent cations [204,205,206,207]. Zinc supplementation possibly helps restoring NK cell functionality in elderly by reversing the inhibitory effects of KIRs, promoting effective target recognition, and enhancing the immune response.

Zinc also plays a pivotal role in enhancing NK cell function. For instance, peptidoglycan monomer zinc significantly improves cytotoxicity against NK-sensitive targets [208]. Studies show a positive correlation between NK cell numbers and vitamin D levels, while lifestyle factors like smoking and alcohol consumption negatively impact NK cell activity [209].

In elderly individuals, three months of zinc supplementation could temporarily boost NK cell activity [13]. Aging is associated with higher expression of metallothionein mRNA, reducing zinc bioavailability, and supplementation can restore zinc levels, enhancing NK cell cytotoxicity [210, 211].

Zinc also affects NK cell cytokine production. After stimulation with phytohemagglutinin (PHA), zinc promotes IFN-γ release and suppresses IL-10 release, shifting the immune response towards a Th1 phenotype. This cytokine shift, which enhances antiviral and antitumor immune responses, which are critical for the elderly to fight age-associated diseases, is linked to an increase in NK cell numbers, potentially explaining the enhanced IFN-γ production [212].

Zinc´s role in NK cell development is further highlighted by its effects on CD34+ progenitor cells. When cultured from both young and elderly donors, CD34+ cells from young individuals predominantly developed into CD56+CD16NK cells, while cells from elderly individuals exhibited a CD56CD16+ phenotype with lower cytotoxic activity. Zinc supplementation increased the number and cytotoxic activity of NK cells in both age groups, and notably, zinc induced the expression of the transcription factor GATA-3, which is essential for NK cell maturation [192]. These findings underline zinc´s importance for both the development and functional regulation of NK cells across the lifespan, which could support aging immune systems against infections and cancers (Fig. 3 and Table 6).

Fig. 3
figure 3

Alterations in Natural Killer Cell Function Associated with Aging and Zinc Deficiency. : Alterations due to aging, but not described in zinc deficiency, : Alterations due to zinc deficiency, but not described in aging, (Black) Alterations described in both, zinc deficiency and aging, (↑): increased, (↓): decreased Created in https://BioRender.com

Table 6 Comparison of NK cell counts and functions in aged individuals and zinc deficient state

Macrophages

Macrophages arise from monocyte precursors in the bloodstream and differentiate into tissue-specific macrophages upon entering tissues. Their primary functions include phagocytosis and regulation of inflammatory responses. However, macrophages in aged organisms exhibit several functional impairments, which contribute to a weakened immune system and increased susceptibility to inflammation and infection. Notably, both aging and zinc deficiency are associated with an increased number of macrophages [217, 218] and monocytes [167, 172, 219, 220].

As organisms age, macrophages display reduced phagocytic activity, compromising innate immune defense mechanisms. This impairment is associated with increased survival rates of pathogens, such as Streptococcus pneumoniae, and a slight rise in pro-inflammatory cytokine levels, as demonstrated in a mouse model [221]. Moreover, diminished phagocytic function in macrophages is linked to delayed wound healing [222] and impaired clearance of apoptotic cells, which contributes to chronic inflammation in aged tissues [223].

Aging also affects macrophage polarization, the process by which macrophages differentiate into either pro-inflammatory M1 or anti-inflammatory M2 phenotypes (Shapouri-Moghaddam et al. 2018). In elderly organisms, the balance between these phenotypes may shift, leading to heightened inflammation. Patients with chronic liver disease often have zinc deficiency. In a mouse model of liver fibrosis, zinc supplementation significantly suppressed fibrosis and inflammation, reduced collagen deposition, and decreased macrophage infiltration, together with inhibiting hepatic stellate cells. Zinc selectively inhibited M1 macrophage polarization and the production of M1-related inflammatory cytokines, including iNOS, monocyte chemotactic protein-1 (MCP-1), and TNF-α. Zinc´s anti-inflammatory effects were also linked to inhibition of Notch1 pathway, which regulates M1 macrophage polarization [224]. Moreover, both zinc deficiency and zinc supplementation in THP-1-derived macrophages were observed to enhance M1 polarization, while concurrently suppressing M2 polarization. The inhibitory effect of zinc supplementation on M2 polarization suggests benefits for the treatment of M2-dominated conditions, such as allergic asthma, or, more commonly observed in the aging population, cancer [225].

Mitochondrial dysfunction is another key factor in the decline of macrophage function with aging. NAD+, a cofactor critical for many enzymatic processes in macrophages, declines with age. Insufficient NAD+ synthesis leads to reduced mitochondrial respiration and phagocytosis, contributing to heightened inflammatory responses. In elderly human monocyte-derived macrophages (MDMs), increased oxygen consumption and decreased activity of mitochondrial complexes I and II have been observed, along with reduced NAD+ synthesis, which may explain the overall decline in immune function with aging [226].

Furthermore, macrophages from aged individuals exhibit impaired mitophagy, the process of recycling damaged mitochondria. This impairment leads to the accumulation of dysfunctional mitochondria, exacerbating oxidative stress and inflammation [227]. The decreased expression and signaling of TLRs, essential for recognizing pathogen-associated molecular patterns (PAMPs), further impair the ability of aging macrophages to phagocyte apoptotic cells and respond to pathogens. Reduced TLR signaling also diminishes cytokine secretion, further weakening the immune response [228].

Cytoplasmic, bioavailable zinc is essential for IL-1β production in activated human monocytes and macrophages, acting downstream of glycolysis, which is induced by the mammalian target of rapamycin complex 1 (mTORC1). Zinc enhances the phosphorylation of S6 kinase (S6 K) by mTORC1 through inhibiting protein phosphatase 2 (PP2 A), a phosphatase that dephosphorylates S6 K. The activity of PP2 A is increased in zinc deficiency [229, 230]. In patients with rheumatoid arthritis (RA), a common disease in the aged population, decreased expression of the zinc transporter Zip8 in monocytes leads to lower intracellular zinc levels. Notably, there is a positive correlation between Zip8 expression and more severe RA clinical parameters [230].

Zinc transporters, particularly Zip proteins, also play a role in macrophage polarization. In a study on hepatocellular carcinoma (HCC), Zip2 and Zip9 were upregulated in M2-polarized macrophages, while Zip9 was slightly downregulated in M1-polarized macrophages. Tumor-associated macrophages derived from human liver cancer tissues exhibited decreased transcription of Zip9 compared to adjacent, non-cancerous tissues. Dysregulated Zip expression, particularly Zip9, may influence immune responses in HCC, contributing to cancer pathogenesis and progression [231].

Zinc plays a critical role in macrophage-mediated pathogen defense. In a study on the fungal pathogen Histoplasma capsulatum, macrophages treated with the glucose analog 2-deoxy-D-glucose (2-DG) showed reduced exogenous zinc import, resulting in zinc starvation, which led to fungal death. This reduction in zinc import was likely due to impaired zinc transporter activity, emphasizing the importance of zinc in controlling infection [232]. Additionally, zinc supplementation increased the population of peritoneal macrophages in Trypanosoma cruzi infection models [233].

In chronic obstructive pulmonary disease, a disease with increased prevalence in aged individuals, zinc deficiency hinders the clearance of apoptotic epithelial cells by alveolar macrophages, contributing to persistent inflammation [234]. Zinc deficiency due to alcohol abuse similarly impairs alveolar macrophage function, reducing phagocytic activity and compromising immune response [235]. In alcohol fed rats, zinc supplementation could improve phagocytic function [236]. Moreover, zinc transporters such as Zip7 have a more direct impact on macrophage activity. In Zip7-knockdown THP-1 cells, phagocytosis is notably less efficient, through this deficiency can be reversed by zinc supplementation. Furthermore, a deficiency in Zip7 shifts macrophage activation towards an M2 phenotype, as evidenced by increased expression of the M2 marker CD206 and reduced levels of the M1 marker NOS2, along with lower production of proinflammatory cytokines such as TNF-α and IL-6. Again, zinc supplementation effectively restores these defects, underlining the role of zinc, and specifically Zip7, in supporting phagocytosis and macrophage activation [14].

In addition to supporting their function, zinc is critical for preventing macrophage apoptosis. In a genetic mouse model, the loss of zinc transporter Zip10 leads to reduced zinc levels in macrophages, triggering p53-dependent apoptosis [74]. Comparatively, aged macrophages are more sensitive to apoptotic signals as well [237].

Zinc also plays a crucial role in the differentiation of myeloid cells, but its effects can vary depending on the context. For instance, low zinc levels can block the differentiation of HL-60 cells into macrophages when induced by phorbol 12-myristate 13-acetate (PMA), an effect that can be partially reversed by adding external zinc [238]. In contrast, zinc deficiency appears to enhance the differentiation and maturation of HL-60 cells into macrophages when stimulated by 1 α, 25-dihydroxyvitamin D D(3) (1,25D(3)) [218].

Zinc´s role in modulating oxidative stress and ROS production in immune cells is context dependent. In PBMCs, including monocytes, a reduction in zinc concentration, either through the use of the chelator TPEN or by removing zinc from the culture medium, leads to a significant increase in oxidative burst and phagocytosis following infection with gram-positive S. aureus [239]. Similarly, in a study on E. coli-infected rats, zinc supplementation increased in vivo superoxide production. However, in vitro, zinc inhibited superoxide production by macrophages derived from septic rats [240]. Additionally, zinc sequestration mechanisms can enhance ROS production in infected macrophages. For instance, in Histoplasma capsulatum-infected macrophages, GM-CSF activation induces the sequestration of labile zinc through MT expression. Zinc exporters such as ZnT4 and ZnT10 are upregulated, redirecting zinc away from phagosomes and into the Golgi apparatus. This zinc redistribution enhances H+ channel function within phagosomes and triggers ROS generation, which effectively halts H. capsulatum replication and improves fungal clearance [241].

Zinc also plays a crucial role in regulating inflammatory signaling pathways in monocytes and macrophages. The activation of TLR4 in monocytes and granulocytes induces a rapid rise in intracellular zinc levels. Chelating this zinc signal with the membrane-permeable chelator TPEN effectively blocks the activation of LPS-induced signaling pathways, including the p38 MAPK, extracellular-signal regulated kinases 1/2 (ERK1/2) and NF-κB pathways, ultimately inhibiting the release of proinflammatory cytokines such as TNF-α [18]. Moreover, LPS-induced stimulation of macrophages activates the JAK-STAT1 pathway through the release of IFN-β, resulting in the expression of iNOS. In this context, free intracellular zinc ions again serve as second messengers in LPS-dependent gene expression. Chelating zinc leads to an inhibition of both LPS- and IFN-β-mediated increases in STAT1 mRNA expression in RAW 264.7 macrophages [242]. Higher zinc concentrations, however, can suppress the release of other proinflammatory cytokines, such as IL-1β and TNF-α, by inhibiting phosphodiesterase activity, thus modulating the inflammatory response [243].

Zinc is essential for the production of IFNs, especially IFN-α, which plays a crucial role in antiviral immunity. Monocytes, alongside plasmacytoid dendritic cells, are major producers of IFN-α during viral infections, contributing to its antiviral and immunomodulatory properties [244]. The age-related decline in IFN-α production [245], which increases the risk of viral infections, highlights the significance of findings by Cakman et al., who demonstrated that in vitro supplementation of blood from elderly individuals with 15 µM zinc significantly enhanced IFN-α production [167].

In summary, zinc supports aging individuals by enhancing macrophage function, improving pathogen clearance, reducing chronic inflammation and aiding wound healing. It balances macrophage polarization, supports mitochondrial health and boosts immune signaling pathways (Fig. 4 and Table 7).

Fig. 4
figure 4

Macrophage and Monocyte Dysfunction in Aging and Zinc Deficiency. : Alterations due to aging, but not described in zinc deficiency, : Alterations due to zinc deficiency, but not described in aging, (Black) Alterations described in both, zinc deficiency and aging, (↑): increased, (↓): decreased Created in https://BioRender.com

Table 7 Comparison of monocyte and macrophage counts and functions in aged individuals and zinc deficient state

Dendritic cells

Dendritic cells (DCs) play a pivotal role in the immune system by serving as professional antigen-presenting cells (APCs). They are essential for initiating and regulating immune responses. DCs capture antigens through surface receptors, process them into peptide fragments, and present these fragments as major histocompatibility complex (MHC)-peptide complexes on their surface. This process enables DCs to activate T cells, a critical step in the initiation of adaptive immune responses. Furthermore, DCs possess the ability to follow chemotactic signals, guiding them to immune-related tissues such as lymph nodes, where they interact with T cells, further underlining their crucial role in regulating immune responses [250].

DCs can be divided into two primary subtypes: plasmacytoid DCs (pDCs) and myeloid DCs (mDCs) [251]. Both subtypes have distinct roles in immune surveillance and response, with pDCs known for their ability to produce IFNs, while mDCs are more specialized in antigen presentation.

Aging has a profound impact on the function and population of DCs, particularly pDCs. In elderly individuals, there is a marked decline in the number of circulating pDCs. This decline is also associated with a decreased ability of these cells to migrate to lymph nodes, a crucial step for T cell activation [252, 253].

Elderly women exhibit a reduced number of circulating pDCs compared to younger women. Additionally, both pDCs and mDCs from older women show diminished responsiveness to TLR stimuli, including TLR7 and TLR8 activation [254]. Additionally, old DCs express lower levels of CCR7 and CCR9, resulting in decreased migratory activity, as demonstrated in studies on aged mice [255]. Despite these changes, the transendothelial migration of DCs remains unchanged compared to younger controls. Furthermore HLA-DR expression is reduced in the peripheral blood DCs of elderly subjects, which leads to diminished antigen presentation and contributes to the age-related decline in immune responsiveness [256].

Aging also affects the cytokine production of pDCs. Compared to healthy adults, pDCs from elderly individuals produce fewer type I IFNs in response to stimuli such as CpG and influenza virus [257]. Moreover, elderly pDCs are less capable of stimulating CD8+ T cells to produce perforin and granzyme, which are crucial for cytotoxic activity, and they are less efficient in promoting IFN-γ secretion by CD4+ and CD8+ T cells [258]. These functional declines are compounded by dysfunctional signaling in TLR2 and TLR4 pathways, further impairing immune responses in the elderly [259].

Aligned with the concept of inflammaging, DCs develop a more pro-inflammatory phenotype as they age [260,261,262]. In this context, zinc plays a crucial role promoting the tolerogenic phenotype in DCs and suppressing pro-inflammatory responses, thereby helping to restore a balance that is critical in aging immune systems [263, 264]. Zinc peroxide nanoparticles significantly induce a transition from immunogenic DCs to tolerogenic DCs in a mouse model of rheumatoid arthritis, thereby reducing T-cell response which drives immune progression of rheumatoid arthritis. The zinc nanoparticles reduced swelling of the ankle, improved mobility and pain reduction. However, there wase no prevention of bone destruction, but a reduced synovial inflammation [265]. Thereby zinc supplementation offers interesting potential to counteract age-related declines in DC function.

When exposed to zinc in vitro, DCs derived from bone marrow exhibited a tolerogenic profile, characterized by reduced surface MHC-II expression and increased levels of tolerogenic markers such as Programmed Death-Ligand (PD-L) 1, PD-L2 and the tryptophan degrading enzyme, Indoleamine 2,3-dioxygenase (IDO). Zn also suppressed the proinflammatory response to TLR ligands. Zn supplementation also shifted the Treg-Th17 balance towards Foxp3+ regulatory T cells (Tregs) [263, 264]. This shift can be attributed to Zn´s ability to inhibit STAT3 activation, which is crucial for Th17 differentiation. All this was shown in vivo with Histoplasma capsulatum fungal infections as well [263]. In aging populations, this balance can mitigate excessive inflammatory responses while enhancing regulatory pathways, reducing the risk of chronic inflammatory diseases.

Zinc supplementation also exerts beneficial effects in vivo. In zinc-deficient rats, zinc supplementation increased the number of DCs and mucosal cells producing secretory IgA while reducing the production of proinflammatory cytokines IL-6 and IFN-γ. This increase in DCs was observed even in rats with normal serum zinc levels [266]. Zinc deficiency also leads to a decrease in epidermal transforming growth factor (TGF)-β1, which in turn reduces the number of epidermal Langerhans cells, another important subset of DCs [267, 268]. In elderly individuals, maintaining zinc homeostasis could help preserve DC numbers and their critical roles in mucosal immunity, a key aspect of protecting against infections.

Zinc also affects DC survival and apoptosis. In mice, low concentrations of zinc stimulate ceramide formation, leading to DC apoptosis through a mechanism involving acid sphingomyelinase. However, LPS treatment was found to protect DCs from apoptosis, demonstrating the complex interplay between zinc and immune signaling [269]. Furthermore, LPS stimulation suppresses the expression of zinc importers (Zip6 and Zip10) while upregulating zinc exporters (ZnT1, ZnT4 and ZnT6), resulting in a net reduction of intracellular zinc. This decrease in intracellular zinc is associated with increased surface expression of MHC-II and CD86, key markers of DC maturation. This effect of LPS-induced zinc depletion can be replicated using zinc-chelating agents, and zinc supplementation inhibits the movement of MHC-II-positive vesicles, empathizing the importance of zinc homeostasis in DC function [270]. For aging individuals, supplementation may prevent these zinc-dependent disruptions, thereby sustaining optimal DC maturation and antigen presentation capacities.

DCs are also capable of synthesizing 1,25-dihydroxy vitamin D (1,25(OH)2D), which plays an essential role in the induction of Tregs. Animal studies suggest that high concentrations of 1,25(OH)2D are necessary to generate Tregs when supplemented, but this can lead to adverse side effects such as hypercalcemia. However, overexpression of 1α-hydroxylase in DCs enables production of 1,25(OH)2D in Treg-inducing concentrations without causing hypercalcemia. Interestingly, the addition of zinc in non-toxic concentrations significantly enhances the Treg-inducing capacity of DCs, offering a promising therapeutic strategy for immune-mediated diseases [271].

In summary, zinc deficiency worsens age-related declines in DC function, leading to reduced antigen presentation, impaired cytokine production, and weakened immune response. Effects that can possibly be improved by zinc supplementation (Table 8).

Table 8 Comparison of dendritic cell counts and functions in aged individuals and zinc deficient state

Changes in the adaptive immune system

T cells

Immunosenescence leads to significant changes in T cell populations and their effectiveness. This process involves a decrease in naïve T cells and an accumulation of memory T cells, impairing the immune system´s ability to respond to novel or fast-mutating pathogens, such as influenza virus while preserving responsiveness to previously encountered ones [283]. More specifically, in the periphery of aged people there is a rise in CD45R0+ memory T cells and a reduction in CD45RA+ naïve T cells [284].

One of the primary reasons for the decline in naïve T cells is thymic involution, a process where the thymus, responsible for producing new T cells, shrinks over time, thereby compromising the adaptive immune response [285,286,287].

Thymic epithelial cells also secrete thymulin, a hormone essential for T cell development and regulation, which depends on zinc to fold into its active form. Zinc acts as a cofactor, stabilizing thymulin's structure and ensuring its proper function in immune regulation. Without zinc, thymulin remains biologically inactive, impairing its role in T-cell differentiation and immune modulation [288]. As thymulin production decreases with age, the overall effectiveness of T cells in immune responses is further reduced [289, 290]. Thymic atrophy in the elderly is often associated with zinc deficiency, contributing to reduced activity of thymulin, lower production of naïve T cells and increased apoptosis in thymocytes [291, 292]. One reason for the increased probability of apoptosis in zinc deficient thymocytes is the higher expression of the protein p56lck [293, 294]. In a mouse model, zinc supplementation has been shown to reverse certain age-related thymic defects and enhance thymopoiesis [295, 296]. Additionally, mild zinc deficiency in humans and mice has been shown to reduce serum thymulin activity, which can be restored through zinc supplementation both in vivo and in vitro [297]. In aged mice, zinc supplementation has been observed to enhance thymulin secretion, potentially contributing to improved thymic function [290, 298].

Aging is also associated with an increase in mitochondrial content in memory CD4+ T cells, which leads to the generation of ROS and subsequently promotes the expression of proinflammatory cytokines [299]. This rise in proinflammatory cytokines is particularly evident in memory T cells, which display a senescence- associated phenotype [300,301,302].

Zinc supplementation in zinc-deficient elderly individuals, such as those in nursing homes, has been shown to improve immune function by enhancing anti-CD3/CD28 and PHA-stimulated T cell proliferation, as well as increasing the number of peripheral T cells [303].

Zinc is also critical for the activation of T cells. Following T cell receptor (TCR) activation, zinc ions enter the cell via zinc transporters such as Zip6 and Zip8, facilitating the signaling pathways necessary for T cell activation and proliferation [69, 304]. This influx of zinc enhances the production of proteins required for TCR signaling, such as lymphocyte protein tyrosine kinase (LCK) and PKC, which are essential for downstream immune responses [305, 306].

Zinc deficiency can severely impair immune responses. It leads to reduced T cell proliferation and diminished cytokine production such as IL-2 and IFN-γ [307, 308], a condition seen in aged individuals as well [309, 310], resulting in an increased susceptibility to infections.

There are also specific alterations in T cell subsets associated with aging. One notable change is the accumulation of CD8+ T cells that express senescence markers such as CD57 and the T-cell immunoreceptor with Ig and ITIM domains (TIGIT) [311]. Additionally, aged CD70+ T cells often exhibit an over-activated phenotype, characterized

by elevated levels of inhibitory receptors, including programmed cell death protein (PD-1), 2B4, and lymphocyte-activation gene 3 (LAG-3) [312]. These over-activated T cells demonstrate increased susceptibility to apoptosis and produce higher levels of cytokines. This heightened susceptibility to apoptosis is not unique to aged CD70+ T cells. T cells in general are more prone to apoptosis [313, 314]. Furthermore, zinc deficiency exacerbates apoptosis in T cell progenitors by disrupting the Bcl-2/Bax pathway, leading to elevated rates of cell death. Conversely, zinc supplementation can mitigate this effect by inhibiting caspase-3, −6, −7 and −8, thereby reducing apoptosis [315].

In addition to its impact on T cell function, zinc plays a crucial role in regulating the balance between T helper cell subsets, particularly the Th1/Th2 ratio. As individuals age, the immune response shifts towards a Th2-dominant profile, reducing the body´s ability to combat intracellular pathogens [15, 316]. Zinc supplementation can restore this balance by enhancing Th1 responses, by increasing the production of Th1 cytokines such as IFN-γ and IL-2 and promoting the expression of the transcription factor T-bet [229, 317]. Conversely, zinc deficiency shifts the immune response further towards a Th2-dominant profile, going along with the reduction of Th1 cell cytokines, such as IFN-γ, IL-2 and TNF-α. Interestingly Th2 cytokines like IL-4, IL-6 and IL-10 remained unchanged [318]. In cases of zinc deficiency, even short-term supplementation of in average 6 days can significantly improve immune function by decreasing CREMα expression and thereby increasing IL-2 production [309]. Zinc supplementation in elderly individuals has been shown to reduce the incidence of infections by increasing the numbers of CD4+ T cells and cytotoxic T lymphocytes through enhanced synthesis of IL-2 and soluble IL-2 receptor (sIL-2R) [319], (Fig. 5 and Table 9).

Fig. 5
figure 5

T Cell Dysregulation in Aging and Zinc Deficiency. : Alterations due to aging, but not described in zinc deficiency, : Alterations due to zinc deficiency, but not described in aging, (Black) Alterations described in both, zinc deficiency and aging, (↑): increased, (↓): decreased Created in https://BioRender.com

Table 9 Comparison of T cell counts and functions in aged Individuals and zinc-deficient state

Proton-pump inhibitors, such as pantoprazole, are commonly used to treat various gastrointestinal diseases in elderly patients. However, pantoprazole has been shown to reduce the expression of zinc transporter Zip8 and increase the expression of the transcription factor CREMα. This alteration results in downregulation of IFN-γ and IL-2 expression, leading to an increased risk of infections [320].

The role of zinc in modulating Tregs is another key factor in its immunoregulatory effects. Tregs are responsible for maintaining immune homeostasis by suppressing overactive immune responses, preventing autoimmune reactions, and ensuring tolerance to self-antigens [321]. The levels of Tregs in the circulating blood are diminished in older individuals [322]. During zinc deficiency, there is an increase in Treg cell numbers. However, these cells are likely nonfunctional as they express low levels of microRNS-146, which is critical for their proper function upon stimulation [323].

Zinc promotes Treg differentiation by inhibiting the activity of Sirtuin 1 (Sirt1), an enzyme that deacetylates Foxp3, a modification that leads to its ubiquitination and subsequent degradation. Foxp3 is a key transcription factor for Treg development [324]. Moreover, treatment of mixed lymphocyte cultures (MLC) with zinc and TGF-β1 triggers intracellular zinc signaling, which, in combination with increased Smad2/3 activation, enhances Foxp3 expression. This results in a mitigated allogenic immune response in MLC, suggesting that zinc can elevate Treg cells in adverse immune reactions [325]. Furthermore, an increased number of Tregs in PBMCs from atopic individuals is associated with the suppression of allergic hyperresponsiveness (Th2-mediated) and allogenic reactivity (Th1-mediated) [326].

Vitamin D3 and zinc, when administered together, more effectively induce regulatory T cells and suppress IFN-γ production in mixed lymphocyte cultures compared to treatment with either Vitamin D3 alone. Vitamin D3 enhances intracellular zinc levels by upregulating the expression of Zip13. Consequently, lower concentrations of Vitamin D3 and zinc may serve as effective treatment options, thereby minimizing the side effect associated with higher doses of Vitamin D3 [327].

The role of zinc is not universally beneficial. In the context of cancer, high dietary zinc intake has been shown to promote tumor progression by fostering a pro-tumorigenic environment mediated by T cells. Conversely, zinc deficiency or chelation of tissue zinc improves anti-tumor immunity and enhances responses to immunotherapies such as αPD-1, particularly in melanoma models [328].

Moreover, zinc supplementation has been shown to reduce the vulnerability of tumor cells to TNF-dependent lysis mediated by CD8+ T cells, whereas zinc chelation enhances this susceptibility. Additionally, the loss of the transcription factor CBFβ in tumor cells disrupts zinc homeostasis pathways, further impairing their responsiveness to TNF-dependent cytotoxicity [329].

Increased Th17 cell numbers have been observed in the elderly [322]. In both mouse models of autoimmune encephalomyelitis and human subjects, supplementation with zinc aspartate has been found to suppress the development of Th17 cells through the degradation of STAT-3, which may help alleviate the impact of Th17-driven autoimmune disorders [330, 331]. Zinc also inhibits the differentiation of Th17 cells by interfering with the IL-6/STAT3 signaling pathway and preventing the phosphorylation of IL-1 receptor-associated kinase 4 [332]. Moreover, zinc suppresses Th17 cells by inhibiting glutaminolysis, which serves as the primary energy source for these cells, thereby lowering the risk of autoimmune diseases, such as autoimmune encephalomyelitis [333].

B cells

The overall number of B cells remains relatively stable throughout most of human lifespan, with a decline only observed in advanced old age, long after the initial signs of immune system inefficacy become apparent [345, 346]. In contrast, murine studies show that while the total number of peripheral B cells in mice remains consistent as they age, the influx of new naïve B cells into peripheral tissues decreases in older mice. This decline is attributed to the slower turnover rate of splenic B cells in aged mice, allowing them to survive longer and signaling to newly generated B cells that peripheral niches are fully occupied [347, 348]. Some researchers also suggest that there is a reduced ability of naïve B cells from older mice to populate peripheral compartments [349]. The reduced influx of new B cells is also due to bone marrow cells having a reduced capacity for B cell generation in culture compared to bone marrow of younger mice [350]. Although aging does not inherently alter individual hematopoietic stem cells (HSCs), it does shift the clonal composition of the HSC pool. In aged mice, there is a decrease in lymphoid-biased HSCs and an accumulation of long lived myeloid-biased HSCs, reducing B cell production capacity [351, 352]. Additionally, the absolute number of B cell precursors in the bone marrow declines with age, particularly during adolescence [353], though B lymphopoiesis continues throughout adulthood [354]. Also, in vitro studies indicate that the responsiveness of pro-B cells to IL-7 is compromised in aged individuals [355], and IL-7 release from stromal cells in the bone marrow also declines with age [356], leading to reduced pro B-cell proliferation.

In elderly humans, there is a significant decline in CD27dull memory B cells, a subset crucial for bridging innate and adaptive immunity. Instead, highly differentiated, antigen-selected CD27bright memory cells dominate. After in vitro stimulation with CpG, B cells from older individuals produce significantly lower levels of IgM and IgA antibodies compared to younger individuals, highlighting a diminished capacity to respond to novel pathogens while maintaining responsiveness to frequently encountered antigens [357]. This is empathized by the decrease of naïve B cells and increase of memory B cells in elderlies [358]. Furthermore, there is an increase in memory B cells lacking the activation marker CD27 in peripheral blood of elderly individuals, a phenomenon also observed in chronic viral infections and autoimmune diseases such as systemic lupus erythematosus [359, 360].

There is also a decline in the IgM memory compartment with age [361], which may contribute to a weakened immune response to polysaccharide antigens, increasing susceptibility to bacterial infections like pneumococcal disease [362,363,364]. This is particularly relevant since the reduction of IgM levels in serum has been linked to decreased opsonophagocytic function [365].

In human studies examining the VH chain repertoire following pneumococcal vaccination, older adults exhibit reduced oligoclonality and a lower frequency of somatic mutations compared to younger individuals [366]. A decline in activation-induced cytidine deaminase (AID), necessary for class switch recombination (CSR) and somatic hypermutation, is observed in elderly individuals. This decrease is attributed to lower stability of E47, a transcription factor for AID, and E47 mRNA. Higher levels of microRNAs, specifically miR-155 and miR-16, in aged B cells may inhibit AID and E47 expression, reducing protein synthesis [367]. Additionally, in vitro studies have demonstrated that zinc chelation inhibits AID activity, suggesting that age-associated changes in zinc homeostasis might further exacerbate the reduction in AID functionality [368]. However, Banerjee et al. [369] demonstrated that somatic hypermutation rates are similar in both young and elderly individuals, suggesting that the increased number of mutations in Ig genes seen in older adults [370] likely results from cumulative exposure over time rather than an inherently altered mutation process.

Research on repeated influenza vaccinations in elderly individuals suggests that memory B cell generation remains intact, though serum antibody titers increase less after vaccination compared to younger controls. Despite this impairment in antibody production, protective titers were still achieved.

[371]. A reason for the reduced production of antibodies could be the notable decline in the presence of long-lived antibody-secreting plasma cells that recirculate within the bone marrow in older adults [372]. Furthermore, antibodies generated in elderly individuals are often less protective than those produced by younger ones. This is demonstrated by a decreased ability to opsonize bacteria-derived polysaccharides in vitro after vaccination [373].

Finally, aging induces a shift in baseline serum antibody specificity, with an increased prevalence of autoreactive antibodies, such as those targeting cardiolipin, DNA, and antinuclear antibodies (ANA), as well as rheumatoid factor. While this rise in autoreactivity is associated with autoimmune diseases like vasculitis, it does not necessarily result in immune pathology [374].

Zinc deficiency reduces the population of naïve B cells, which are essential for adaptive immunity [219]. This reduction is largely due to the disruption of lymphopoiesis, as zinc deficiency leads to a significant decline in pre-B cells [291]. Further research has shown that in zinc-deficient mice, while T-cell lymphopoiesis is significantly reduced, B cell development and maturation in the bone marrow are less affected [375]. However, the reduction in pre-B cells during zinc deficiency mirrors the effect observed in T cells and is associated with altered glucocorticoid metabolism and increased apoptosis, highlighting shared mechanisms of immune impairment under zinc deficiency [376].

The B cell receptor (BCR), essential for both immature and mature B cell activation and proliferation, is another area where zinc plays a pivotal role. The zinc transporter Zip10 is critical for modulating BCR signal strength [377] and preventing apoptosis in B cells, indicating zinc´s influence on humoral immunity through its regulation of BCR signaling [378].

Zinc deficiency also affects intracellular signaling pathways necessary for B cell function. Specifically, it hinders the activation of protein PKC in response to mitogens, as zinc deficiency prevents PKC´s translocation to the cell membrane. This impairment leads to a reduced proliferative response in B lymphocytes [379]. Moreover, zinc deficiency alters cytokine-mediated signaling, enhancing B cell proliferation in response to IL-6 and IL-2 but reducing it following IL-4 stimulation. This results in decreased STAT6 phosphorylation in IL-4 pathways and increased STAT3 phosphorylation in IL-6 pathways, emphasizing the role of zinc in maintaining balanced immune responses [380].

Furthermore, regulatory B cells (Bregs), identified as CD19+IL-10+ B cells, play crucial role in modulating immune responses by secreting the anti-inflammatory cytokine IL-10. By fostering an anti-inflammatory environment, Bregs help mitigate excessive pro-inflammatory responses, offering potential benefits for managing conditions such as allergies, asthma, and autoimmune diseases. This makes supporting Breg function especially important in elderly individuals, as it could help counteract the inflammatory imbalances characteristic of aging. Importantly, zinc levels significantly influence Breg production, with zinc deficiency leading to reduced Breg generation from purified B cells [381], (Fig. 6 and Table 10).

Fig. 6
figure 6

B Cell Alterations in Aging and Zinc Deficiency. : Alterations due to aging, (Black) Alternations due to both, zinc deficiency and aging, (↑): increased, (↓): decreased Created in https://BioRender.com

Table 10 Comparison of B cell counts and functions in aged individuals and zinc deficient state

In the context of vaccination, zinc supplementation has shown promising effects, particularly in elderly populations. For instance, zinc treatment improved the IgG antibody reaction to the tetanus vaccine in older adults [16]. Furthermore, in a study on pneumococcal vaccination, elderly individuals with a stronger immune response had significantly higher serum zinc levels [382]. Conversely, elderly chronic hemodialysis patients who exhibited an inadequate immune response to active diphtheria vaccination were found to have significantly lower serum zinc levels compared to those who responded effectively [383]. Zinc´s importance extends to prenatal health as well. Zinc deficiency during pregnancy has been found to impair offspring´s antibody response and T cell proliferation following hepatitis B vaccination. Additionally, maternal zinc deficiency suppresses the secretion of IFN-γ from splenocytes and reduces IFN-γ expression in both CD4+ and CD8+ T cells [384]. The timing of zinc supplementation is also critical for optimal vaccination outcomes. Research has shown that administering high doses of zinc concurrently with vaccination may result in suboptimal effects, whereas pre-vaccination zinc supplementation offers more effective enhancement of the antibody response [16, 385,386,387].

More recent studies have identified a correlation between free zinc levels and the strength of antibody responses, including the potency of neutralizing antibodies [388]. This connection is particularly significant given that antibody titers after vaccination decline more rapidly in the elderly [389], highlighting the potential role of zinc in supporting more robust and sustained immune responses in aging populations.

In studies of human B cells immortalized by Epstein-Barr virus, alterations in zinc transporter expression were observed. Specifically, there was an increase in the endoplasmic-reticulum-to-cytoplasm Zip7 mRNA and protein expression as well as phosphorylation, leading to elevated intracellular free zinc levels. These changes, however, were not limited to immortalized cells. Similar elevations were noted in vitro in activated B cells marked with CD69, suggesting that both activation and proliferation contribute to increased intracellular zinc [390]. Moreover, mutations in the Zip7 gene have been linked to a novel autosomal recessive disease characterized by agammaglobulinemia and early-onset infections, highlighting the crucial role of Zip7 in B cell development and immune function, as confirmed through CRISPR-Cas9 mutagenesis in mice [44].

Age related diseases

As highlighted earlier, immunosenescence and inflammaging contribute significantly to an increased risk of infections, cancer, and autoimmune diseases in aging individuals. For example, people over the age of 65 represented more than 68% of new cancer cases in 2019 [402], and the incidence of Clostridium difficile infections was more than threefold higher among elderly individuals compared to younger adults [403].

Additionally, the interplay between COVID-19, aging, and zinc homeostasis has garnered considerable attention, given the heightened vulnerability of older adults to severe COVID-19 outcomes. In the U.S., over 75% of deaths due to COVID-19 occurred among individuals aged 65 and older, with more than 53% of these deaths occurring in those over 75 years of age [404]. Zinc plays a pivotal role in immune function, and its deficiency has been linked to worse outcomes in COVID-19 patients. Lower serum zinc levels have been shown to correlate with more severe symptoms, such as acute respiratory distress syndrome (ARDS) and prolonged hospital stays [405, 406]. Furthermore, patients who did not receive zinc supplementation experienced longer recovery times, worse clinical presentations, and higher mortality rates compared to those who received supplemental zinc [407]. These findings underscore the critical role of zinc in modulating immune responses and mitigating the severity of COVID-19 in aging populations.

While the relationship between zinc and COVID-19 has been extensively reviewed in the literature [408], this discussion will shift focus to other age-related diseases and the broader consequences of zinc deficiency, which is not only linked to immune dysfunction but also plays a significant role in other age-associated conditions, where immunological processes are not the sole contributors. These include a range of chronic, degenerative and metabolic diseases, highlighting the indispensable role of zinc in supporting overall health and resilience during the aging process.

Age-related macular degeneration

Age-related macular degeneration (AMD) is the primary cause of blindness among older adults in industrialized countries. In 2015, AMD affected approximately 67 million individuals in Europe, with projections indicating an increase to 77 million by 2050 [409]. This acquired, degenerative condition is characterized by drusen deposits that interfere with the retinal pigment epithelium (RPE) and Bruch´s membrane, specifically affecting the macula, which is essential for central, detailed, and color vision [410]. AMD progresses in two main forms: the dry (non-vascular or early-stage) type, marked by drusen beneath the RPE, and the wet (neovascular or late-stage) type, characterized by choroidal neovascularization [411, 412]. The prevalence of AMD increases markedly with age, demonstrating a pronounced disparity between older and younger populations. Among individuals aged 64 years or younger, AMD is less common, with 9.3% exhibiting moderate signs. In contrast, this prevalence rises to 26.9% in those over 75 years, emphasizing the profound influence of aging on the progression of the condition [409].

While most AMD cases (90%) begin as the less aggressive dry type, around 10% progress to the vision-threatening wet form. However, no approved clinical therapy currently exists for dry AMD. Lifestyle changes such as avoiding smoking, following a healthy diet and taking antioxidant supplements are recommended. In contrast, wet AMD can be treated with anti- vascular endothelial growth factor (VEGF) antibodies to reduce neovascularization [413, 414]. Given that aging is the leading risk factor, alongside family history and specific genetic predispositions, understanding AMD´s development and potential interventions is vital for protecting vision in aging populations [415].

Several genes related to the complement system, including the gene of complement factor H (CFH) [416], age-related maculopathy susceptibility 2 (ARMS2) [417], and others, have been identified as risk factor for AMD. SNPs in the CFH gene can elevate the risk of developing AMD by up to six times [418]. Expanding on this, complement components C3 and C5 play a pivotal role in driving VEGF expression, a key factor in the angiogenesis characteristic of AMD [419]. Additional risk factors, such as, obesity, smoking, UV light exposure, and certain physiological traits (e.g., high blood pressure, light-colored irises, female sex), exacerbate AMD through oxidative stress mechanisms [420]. The retina possesses the highest metabolic rate of any human tissue, with the production of ROS being an inherent aspect of its normal function. Research indicates that RPE cells from individuals with AMD exhibit elevated ROS levels and an increased susceptibility to oxidative stress compared to RPE cells from non-AMD donors [421]. Zinc plays a critical protective role in maintaining retinal health, particularly in the RPE. It mitigates H2O2-induced damage, enhances SOD activity [422], and activates the antioxidant response element (ARE)-Nrf2 pathway [423], thereby protecting RPE cells against elevated ROS levels.

Zinc levels in the neural retinal decline significantly with advanced age, whereas concentrations remain stable in the RPE [424] This decline is particularly relevant given that zinc is stored in key retinal cells, including ganglion, horizontal, amacrine, and Müller cells [425, 426]. In AMD, RPE/choroid zinc levels are reduced by around 24% in humans, while zinc deposits are paradoxically high in drusen, possibly affecting CFH oligomerization [427, 428].

Due to zinc depletion in AMD-affected RPE, dietary zinc supplementation is a promising intervention to prevent disease progression. Large-scale trials, notably AREDS and AREDS2, support zinc´s efficacy in slowing AMD progression, especially in combination with antioxidants such as vitamins C and E and β-carotene [429,430,431]. Studies such as the Rotterdam Eye Study and the Blue Mountains Eye Study also observed that increased dietary zinc intake was associated with a reduced risk of AMD [20, 432, 433]. However, zinc´s effectiveness appears to vary depending on genetic background, benefiting CFH allele carriers in the Rotterdam Study [434] but also more significantly benefiting ARMS2 carriers in AREDS participants [435, 436].

Beyond AMD risk reduction, zinc´s role in the complement system is of particular interest, as zinc promotes CFH oligomerization and inactivation, potentially modulating inflammation in AMD [428]. As part of the innate immune response, complement system dysregulation is implicated in AMD pathogenesis, with zinc able to bind CFH and other complement proteins to modulate inflammatory responses [437, 438]. Another important mediator, Zip7, facilitates zinc transfer from the endoplasmic reticulum to the cytoplasm within RPE cells, a process responsive to inflammatory cytokines such as IL-1β and TNF-α. By regulating barrier dysfunction in the RPE, Zip 7 may influence AMD pathogenesis, as shown by the protective effects of inhibiting Zip7-mediated zinc movement against these cytokines [439].

Additionally, AMD´s pathogenesis involves the loss of RPE-derived VEGF signaling and defenestration of usually high permeable choriocapillaries in atrophied RPE areas, which disrupts retinal permeability [440, 441]. Zinc supplementation has shown potential in restoring these retinal fenestrations by promoting the redistribution of plasmalemmal vesicle associated protein-1 (PV-1), facilitating nutrient and waste exchange and improving retinal health in AMD [442].

In sum, zinc is a vital component in both preventive and therapeutic strategies for AMD, especially for aging populations who are at increased risk. Through its multifaceted effects, such as antioxidant properties, inflammatory modulation, genetic interactions, and RPE function support, zinc supplementation presents a low-cost, accessible option to slow AMD progression and help maintain vision quality in elderly individuals Figure 7.

Atherosclerosis

Atherosclerosis it a pathological condition characterized by the buildup of plaque, composed of fats, cholesterol, calcium and other substances, within the intima of large and medium-sized arteries- This process begins with the activation of endothelial cells, setting off a cascade that leads to vessel narrowing, activation of inflammatory pathways, and ultimately, formation of atheromatous plaques [443]. Atherosclerosis serves as the primary pathological mechanism behind most cardiovascular disease (CVD) cases, a leading global health concern and one of the top causes of disability and premature death [444, 445].

Importantly, atherosclerosis can begin early in life and often progresses silently over decades before symptoms emerge [446]. The prevalence of CVD rises sharply with age, affecting approximately 38% of individuals between 40 and 60 and reaching 83% in those over 85. Increased age is associated with conditions like elevated carotid intima-media thickness, carotid plaque, and carotid stenosis, which occur more frequently in older adults, compounding their CVD risk [447]. Given the rising prevalence of AS in elderly populations, the burden it places on personal health and healthcare systems presents a significant public health challenge requiring immediate attention.

Emerging evidence points to the critical role of zinc in modifying multiple risk factors associated with AS, particularly in older adults. Zinc impacts lipid metabolism, glucose metabolism, and blood pressure regulation, making it a promising therapeutic agent against AS (Fig. 7). Zinc-α2-glycoprotein (ZAG) is a glycoprotein with a zinc-binding site that plays a pivotal role in lipid metabolism. It promotes lipolysis through the cAMP signaling pathway [448] and enhances energy metabolism in brown adipose tissue by stimulating PPARγ and early B cell factor 2 (EBF2) [449]. Serum ZAG concentration declines progressively with age, showing an inverse correlation with fasting blood glucose and triglyceride levels, while higher body mass index (BMI) is also negatively associated with ZAG levels [450]. Furthermore, serum zinc concentrations are significantly lower in obese patients, with obesity being a major risk factor for CVD [451]. Zinc has been shown to correct lipid abnormalities in higher-fat diet models, lowering liver enzymes, including aspartate aminotransferase (AST) and alanine transferase (ALT), associated with liver stress [452]. The beneficial effects of zinc on lipid metabolism extend to improved cardiovascular outcomes. Zinc supplementation has been shown to significantly reduce total cholesterol (TC), increase high-density lipoprotein cholesterol (HDL-C) in individuals with underlying health conditions, and lower low-density lipoprotein cholesterol (LDL-C) and triglyceride levels, as demonstrated in a meta-analysis [453]. Additionally, studies assessing the TC to high-density lipoprotein cholesterol (TC/HDL-C) ratio, a recognized marker of CVD risk, indicate that zinc supplementation reduces this ratio, further highlighting its protective role in cardiovascular health [454].

Fig. 7
figure 7

Impact of Zinc Deficiency on Atherosclerosis and Cardiovascular Health Zinc deficiency contributes to dysregulated lipid metabolism, glucose metabolism, elevated blood pressure and endothelial dysfunction. Together, these effects heighten the risk of atherosclerosis and metabolic disorders in zinc-deficient individuals. (↑): Characteristic increased, (↓): Characteristic decreased Created in https://BioRender.com

Zinc also plays a protective role against atherosclerosis through its impact on glucose metabolism. Supplementation with zinc has been shown to reduce fasting glucose levels and HbA1c [455]. Additional effects of zinc on glucose metabolism will be explored in the diabetes mellitus section of this review.

Studies have demonstrated a negative association between serum zinc levels and high blood pressure [456]. Moreover, zinc supplementation has been shown to reduce systolic blood pressure, highlighting its potential therapeutic role in managing hypertension [457]. One critical organ for blood pressure regulation is the kidney, which is highly sensitive to zinc deficiency. In zinc deficient rats, kidney functions are compromised due to structural abnormalities, including reduced number of nephrons and renal fibrosis, and functional impairments, such as reduced glomerular filtration rate and proteinuria [458]. Notably, zinc plays a protective role at the cellular level, as it has been shown to significantly inhibit apoptosis in ATP-depleted renal cells [459]. Zinc-deficient rats also exhibited elevated levels of angiotensin II, along with augmented expression of angiotensin-converting enzyme 1 (ACE1) and the angiotensin receptors AT1 and AT2 at both the mRNA and protein levels. These changes enhance the activity of the renin-angiotensin system (RAAS), contributing to vasoconstriction, sodium retention, and heightened blood pressure, thereby exacerbating the risk of hypertension [460]. Further compounding the issue, zinc deficiency has been shown to upregulate the renal Na-Cl cotransporter, which increases sodium retention and further elevates blood pressure [461].

Endothelial cells (ECs) are central to the pathogenesis of atherosclerosis. Endothelial cell dysfunction, a key event, is associated with changes in hemodynamic forces. Areas of low shear stress trigger proatherogenic responses such as increased permeability, production of ROS, activation of NF-κB, and upregulation of adhesion molecules that attract leukocytes [462].

Zinc plays a critical role in maintaining endothelial health by supporting nitric oxide synthesis through the activation of endothelial nitric oxide synthase (eNOS), which is essential for vascular relaxation and anti-atherogenic effects [463]. The enzymatic activity of eNOS relies on its dimeric configuration, which is essential for its proper function. This dimeric state is stabilized by a zinc ion, playing a crucial role in maintaining the structural integrity and interaction of the two monomers [464]. The removal of zinc causes eNOS dimers to dissociate into inactive monomers in endothelial cells [465]. Hence, zinc supplementation increases intracellular NO production [466].

Moreover, Zinc deficiency enhances NF-κB activation, fueling inflammatory responses [467]. This pro-inflammatory state is further exacerbated by the reduced expression of PPARα and PPARγ in zinc-deficient endothelial cells, as these receptors are crucial inhibitors of NF-κB signaling, underlining zinc's vital role in preventing sustained inflammation [19]. Supplementation with zinc markedly reduces the binding activity of transcription factors NF-κB and AP-1, which in turn results in a significant decrease in the production of the pro-inflammatory cytokine IL-8 in endothelial cells [468]. Additionally, the hypoxia-inducible factor-1 (HIF-1) acts as a pivotal regulator during hypoxic and ischemic conditions, playing a key role in promoting cell survival and driving the production of pro-inflammatory cytokines. These actions contribute to critical processes such as angiogenesis and vascular remodeling [469]. Zinc has been shown to inhibit HIF-1α, impair its heterodimer formation and enhance its degradation via the proteasomal pathway [470].

Furthermore, a deficiency in zinc markedly impairs the barrier function of porcine endothelial cells [471], making them more prone to leukocyte infiltration and facilitating inflammatory processes [472]. Low zinc levels heighten endothelial cell vulnerability to LPS-induced apoptosis, whereas higher intracellular zinc supports defenses against apoptosis, possibly via nitric oxide pathways [473]. In addition, zinc-deficient endothelial cells exhibit increased caspase-3 activation, leading to higher rates of cell death, an effect reversible by zinc supplementation [474].

In atherosclerosis, endothelial cells produce chemokines, cytokines, and adhesion molecules that attract monocytes, leading to leukocyte migration into the vascular wall. Zinc deficiency exacerbates these processes by upregulating molecules like vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1), facilitating monocyte attachment and intensifying vascular inflammation and AS progression [475, 476].

Human studies support these findings: A randomized controlled trial demonstrated that a six-month zinc supplementation regimen in older adults increased plasma zinc levels and decreased markers of inflammation, including high-sensitivity C-reactive protein, IL-6, MCP-1, and VCAM-1. These findings suggest that addressing zinc deficiency in older populations may alleviate systemic inflammation and oxidative stress, thereby reducing the risk of atherosclerosis and improving cardiovascular outcomes [477].

Epidemiological studies have identified an inverse relationship between serum zinc levels and the risk of coronary artery disease (CAD) [478]. A meta-analysis further demonstrates that patients with heart failure have significantly lower serum zinc levels compared to healthy controls, with individuals diagnosed with idiopathic dilated cardiomyopathy exhibiting particularly reduced zinc concentrations [479]. Additionally, a significant inverse correlation has been observed between serum zinc levels and left ventricular mass, a marker of left ventricular hypertrophy [480]. Healthy individuals show higher serum zinc levels than those diagnosed with atherosclerosis or unstable angina [481, 482]. Furthermore, increased dietary zinc intake has been linked to a lower risk of coronary artery calcium progression, a known marker of subclinical atherosclerosis, particularly when the zinc is sourced from non-red meat [483]. Conversely, zinc intake from red meat sources has been positively associated with a higher risk of CVD [484]. Lastly, individuals who have suffered myocardial infarction were found to have significantly reduced zinc levels in both serum and hair samples [485].

This leads to the conclusion that zinc´s essential role in endothelial function, its modulation of inflammatory and oxidative stress pathway, and its impact on atherosclerosis progression position it as a vital nutrient for cardiovascular health in aging populations.

Depression

Depression is one of the most prevalent mental health disorders worldwide, affecting approximately 280 million people [486]. Major Depressive Disorder (MDD) specifically has an alarming presence across age groups, with a 12-month prevalence of 10.4% and a lifetime prevalence of 20.6% [487]. MDD ranks among the top five disabling disorders globally [488], leading not only to mental health challenges but also to serious physical health risks, such as cardiovascular disease, stroke, and metabolic disorders [489, 490]. For aging populations, MDD presents an even greater concern, as it significantly raises the risk of Alzheimer´s disease, dementia, frailty, and reduced health span, underlining its multifaceted burden on both mental and physical health in older adults [491,492,493]. Around 22.1% of older adults with mild dementia and 11.6% of those with moderate dementia also experience major depressive disorder [494].

The implications of MDD on mortality are profound, with affected individuals facing increased risks of death from all causes, particularly cardiovascular mortality [489, 495]. Moreover, MDD has been linked to characteristics commonly associated with aging, suggesting that it may contribute to a premature aging phenotype, particularly in older adults who are already at heightened risk [489, 495].

Treatment for depression is typically stratified by severity, with mild depression managed through psychoeducation, self-care, and psychotherapy, while moderate to severe depression often requires a combination of psychotherapy and antidepressant medications [496]. Antidepressants remain a cornerstone of treatment, demonstrating clear benefits in reducing depressive symptoms compared to placebo [497]. However, the rising prevalence of depression in the general population is coupled with an increasing number of patients who do not respond to antidepressant therapy, underlining the need to identify underlying factors to enable early detection and intervention, especially in high-risk groups [498].

Research shows that low dietary zinc intake is linked to an elevated risk of depression [23, 499, 500]. Patients with major depression frequently exhibit lower serum zinc levels than healthy controls [501, 502] and a positive correlation exists between zinc deficiency and depression severity as these studies show among female students, postmenopausal women, and hemodialysis patients [503,504,505]. Studies with human subjects have shown that dietary or supplemental zinc interventions have antidepressant-like effects and improve mood [506,507,508].

Depression has been partly linked to inflammation, as evidenced by the role of proinflammatory cytokines like IL-1β in its development [509]. Zinc deficiency promotes the production of IL-1β by increasing the accessibility of the IL-1β promoter, which possibly results in a constitutive IL-1β production even in the absence of danger signals. Zinc repletion reduces promoter accessibility, thereby limiting IL-1β production under basal conditions, although it can still be induced by stimuli (danger signals) such as LPS [510]. This mechanism may contribute to an elevated risk of depression.

Furthermore, patients with major depression show a reduced number of Tregs in their peripheral blood, a characteristic also associated with inflammatory diseases [511]. Both antidepressant therapy [512] and zinc supplementation [327] have been found to reduce IL-1β expression and increase Treg levels. Similarly, both treatments lower the production of IFN-γ, further emphasizing their shared anti-inflammatory effects [327, 513].

Beyond its role in inflammation-related mechanisms contributing to depression, zinc also influences depression through other pathways. The G protein-coupled receptor 39 (GPR39) is responsive to zinc levels, and its activation influences pathways involved in depression [26, 514]. Zinc-deficient animals have been shown to exhibit reduced GPR39 expression in brain areas such as the hippocampus and frontal cortex, similar to observations in humans with depressive symptoms who died by suicide [515]. Mice lacking GPR39 display behaviors, seen in depression and anxiety, accompanied by compromised muscle coordination. Furthermore, these mice exhibited an altered expression of the Cl transporter KCC2 in the amygdala, which influences GABAergic signal pathways [516].

The influence of zinc on depression is further underlined by its role in regulating zinc transporter proteins. Studies of stress-induced depression models have revealed lower total zinc levels, accompanied by reduced expression of ZnT1 in the prefrontal cortex and hippocampus, as well as ZnT3 in the hippocampus [517, 518].

Depression-like behaviors resulting from zinc deficiency are associated with heightened hippocampal expression of the N-methyl-D-aspartate receptor (NMDAR), and lowered α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) expression. Effects that can partially be reversed with fluoxetine treatment [519, 520].

These findings underline the importance of dietary zinc intake, particularly for aging populations who are at higher risk of both zinc deficiency and depression-related cognitive decline. Evidence from human studies suggests that dietary or supplemental zinc can have antidepressant effects, providing a promising adjunctive treatment approach for individuals who do not fully respond to conventional antidepressant therapy [506,507,508]. Recognizing zinc´s potential to support mental health, particularly in older adults, can guide treatment strategies aimed at improving both psychological and physiological outcomes in populations vulnerable to accelerated aging and mental health challenges associated with depression.

Wound healing

Elderly individuals are more susceptible to chronic wound, such as diabetic and pressure ulcers, often exacerbated by age-related declines in wound healing linked to aging immune system [521, 522]. Research highlights that essential reparative functions, including angiogenesis, fibroblast activity, and collagen deposition, are compromised in older adults. This delayed wound closure not only increases infection risks but also extends hospital stays, imposing added strain on healthcare resources [523, 524]. As wound healing is essential to tissue integrity and overall health, the care and management of chronic wounds in aging populations have become a critical public health concern, annual cost of treating chronic wounds, including various ulcers, are estimated to reach up to $96 billion in the U.S. alone [525].

The skin holds a relatively high concentration of zinc, which constitutes about 6% of the body´s total zinc content and is mainly concentrated in the skin´s epidermis. This zinc is especially significant in the wound healing process [526]. During the wound healing process, initial hemostasis is established through the aggregation of platelets and the formation of a clot, which is subsequently stabilized by the secretion and polymerization of fibrin [527]. Studies involving both rats and humans demonstrated that dietary zinc deficiency leads to impaired platelet aggregation [528, 529]. Zinc plays a crucial role in facilitating platelet aggregation by interacting with the fibrinogen receptor glycoprotein IIb/IIIa complex, enhancing the response synergistically when combined with collagen or ADP [530, 531].

In the inflammatory phase, a critical step in wound healing, zinc levels increase significantly. Rat model studies demonstrate rise of about 15% in zinc within 24 h post-injury at the wound margins, followed by an increase of up to 30% during the formation of granulation tissue and epidermal proliferation [532, 533]. This zinc signal initiates the repair process in epithelial cells [534].

Once inflammation subsides, re-epithelialization begins, typically around 2–3 days after injury, as fibroblasts infiltrate the wound site, initiating collagen and extracellular matrix (ECM) deposition to create a scaffold for tissue repair. This matrix serves a foundation supporting the migration of epithelial cells, keratinocytes, and microvascular structures [535]. The TGF-β/SMAD pathway, which zinc supports as an essential cofactor, regulates ECM deposition, aiding granulation tissue formation [325]. Additionally, zinc-dependent matrix metalloproteinases (MMPs) are involved in depredating granulation tissue to enable migration of keratinocytes, fibroblasts, and endothelial cells, promoting cell growth, migration, and angiogenesis [536]. Both zinc and calcium are essential for the efficient functioning of MMPs in vitro [537, 538].

Clinical evidence suggests that zinc deficiency can impair wound healing as low serum zinc levels correlate with poor healing outcomes, including infection and dehiscence in patients undergoing hip replacement surgeries [22]. Chronic leg ulcer patients commonly present with zinc metabolism irregularities, leading to low serum zinc levels [539,540,541]. Studies have shown that topical zinc application can enhance wound repair, as demonstrated in porcine skin wound models where zinc accelerated re-epithelization [542] and increased keratinocyte migration, contributing to epidermal re-epithelization [543]. Zinc oxide, specifically, has been effective in ulcer healing, performing comparably to enzymatic debriding agents for pressure ulcers [544,545,546] and demonstrating effectiveness in diabetic foot ulcers by promoting debridement through zinc oxide-medicated occlusive dressings, outperforming standard hydrocolloid dressings that utilize autodebridement [547]. In a study comparing topical zinc treatments with saline controls in patients with uncomplicated wounds, those treated with zinc showed significantly better healing outcomes [548].

Zinc oxide also aids in collagen degradation in neurotic wounds, which is beneficial in cases of tissue damage requiring necrotic tissue breakdown for effective healing [538]. Nutritional interventions have also shown benefits. For example, a high-calorie, high-protein oral formula fortified with zinc and other nutrients led to significant ulcer reduction in malnourished patients with chronic pressure ulcers, although zinc´s exact role within the formula remains uncertain [549]. In surgical settings, zinc infusions administered pre- and post-operatively in vascular surgery patients prevented the typical postoperative drop in serum zinc levels, resulting in a reduction in wound healing complications compared to the placebo group [550].

However, studies on systemic zinc supplementation present mixed results. For instance, a systemic review indicated that zinc sulfate did not significantly aid in treating leg ulcers unless zinc deficiency was evident [551]. In contrast, another study found general poor dietary zinc intake among patients with leg ulcers [552]. Topical zinc therapy, however showed improved healing in chronic venous leg ulcers in a meta-analysis [553].

In summary, zinc plays a multifaceted role in wound healing, making it a critical component of wound care for elderly individuals. From enhancing hemostasis and inflammation to promoting re-epithelization and ECM remodeling, zinc´s physiological functions support each phase of the wound healing process. For the aging population, which is at higher risk of delayed wound healing and associated complications, zinc-based treatments, particularly topical applications, offer promising therapeutic options, though the effectiveness of systemic supplementation remains context-dependent Figure 8.

Diabetes mellitus

Diabetes mellitus is a chronic metabolic disorder marked by elevated blood glucose levels, or hyperglycemia, and increased glycated hemoglobin, sometimes accompanied by glycosuria [554, 555]. This condition results from disruption in glucose metabolism, often stemming from insufficient insulin production by the pancreas, reduced in insulin effectiveness in body tissues (insulin resistance), or a combination of these factors, which is particularly common among older adults [556]. Prolonged hyperglycemia can lead to progressive complications, damaging vital organs such as the heart, blood vessels, eyes, kidneys, and nerves, underscoring the need for targeted management of diabetes in older populations to prevent these outcomes [556].

Diabetes ranks as the fifth leading cause of death globally, responsible for approximately 1.6 million fatalities per year [557, 558], and accounted for 11.3% of all global deaths in 2019 [559]. Type 2 diabetes, in particular, has seen a continuous increase in prevalence, driven by lifestyle shifts and population aging, especially in Westernized societies, where these factors contribute to rising obesity rates. Among those aged 70 and above, the prevalence of diabetes is notably high, affecting around 25–30% of this demographic group [560, 561].

In pancreatic β-cells, the highest concentration of zinc is in insulin secretory granules, where it plays a vital role in insulin stability and secretion. Insulin binds with zinc ions to form hexamers, which allow dense packing of insulin in storage vesicles, protecting it from degradation [50, 562]. The zinc transporter ZnT8 is essential for moving zinc into pancreatic β-cell granules, facilitating zinc-insulin hexamer formation, critical for efficient insulin release. Zinc deficiency can reduce ZnT8 expression, thereby hindering insulin secretion [563,564,565]. Single nucleotide polymorphisms (SNPs) as the rs13266634 SNP in the ZnT8 gene and is associated with an increased risk of developing type 2 diabetes [566] The risk of developing type 2 diabetes with this SNP can be lowered with zinc supplementati[on [567].

Inflammaging plays a critical role in the pathogenesis of diabetes and the destruction of pancreatic β cells as well. A key contributor to this process is the pro-inflammatory cytokine IL-1β, which is significantly elevated in aging individuals [568]. IL-1β inhibits insulin release and processing of proinsulin into insulin, impairing glucose metabolism [569]. Furthermore, IL-1β induces apoptosis in pancreatic β cells through the activation of the NF-κB pathway, exacerbating β cell loss [570]. The potential to counteract these effects has been demonstrated with IL-1 receptor antagonists, which have been shown to improve glucose tolerance and enhance insulin production [571]. Zinc supplementation offers a promising way to mitigate these effects, as it has been shown to reduce symptoms of inflammaging and lower IL-1β release. Ex vivo studies reveal that zinc supplementation can decrease IL-1β output following LPS stimulation [572, 573]. These findings suggest that zinc may help reducing the risk of diabetes mellitus in aging populations by protecting β cells from apoptosis.

The insulin signaling pathway, critical for glucose regulation, begins with insulin binding to its receptor, which activates insulin receptor substrates (IRS) and recruits phosphoinositide 3-kinase (PI3 K), which turns phosphatidylinositol-4,5-bisphosphate (PIP2) into phosphatidylinositol-3,4,5-trisphosphat (PIP3). PIP3 then activates Akt (Protein Kinase B), initiating various metabolic processes: GLUT4 translocates to the cell membrane to facilitate glucose uptake, glycogen synthase kinase 3 (GSK3) is inhibited to promote glycogen synthesis, and FOXO1 is phosphorylated, which reduces gluconeogenesis. Additionally, Akt activates ribosomal protein S6 K, enhancing protein synthesis and supporting cellular growth and metabolism. Phosphatases, such as PTEN and PTP1B, regulate this pathway by deactivating signals, thereby maintaining balance in glucose and metabolic activity [574].

Zinc is a crucial element in activating the PI3 K/Akt pathway (Fig. 8), which is vital for effective insulin signaling [575]. Research indicates that zinc independently stimulates the activation of insulin receptor β, AKT, ERK1/2 and Src homology-2 domain-containing protein tyrosine phosphatase (SHP) in myotubes, promoting glucose uptake via GLUT4 translocation in the cell membrane [575, 576]. Inhibition of the insulin receptor in C2 C12 skeletal muscle cells reduces zinc-induced Akt activation, suggesting that zinc may exert its effects through insulin receptor interaction [575], more exactly by enhancing the phosphorylation of the insulin receptor´s β-subunit [577, 578].

Fig. 8
figure 8

Impact of Zinc on Insulin Secretion, β-Cell Health, and Insulin Signaling (↑): increased, (↓): decreased, () activation, (–I): inhibition (a) Zinc transporter 8 (ZnT8) facilitates zinc storage in insulin granules within pancreatic β-cells, playing a critical role in insulin storage. Aging and zinc deficiency increase IL-1, exacerbating IL-1-mediated activation of the NF-κB pathway, which leads to increased apoptosis β-cells. b Zinc plays a modulatory role in insulin signaling pathways in target tissues such as adipocytes and myocytes. Zinc inhibits protein tyrosine phosphatase 1B (PTP1B) activity, enhancing insulin receptor signaling through IRS and PI3 K pathways. This supports Akt/PKB activation, promoting downstream effects such as increased glucose uptake via GLUT4 translocation, enhanced glycogen synthesis (via inhibition of GSK3), reduced gluconeogenesis (via FOXO1 suppression), and increased protein synthesis (via S6 K activation). Created in https://BioRender.com

Zinc is also inhibits the enzyme tyrosine phosphatase 1B (PTP1B), which would otherwise reduce the insulin signal by dephosphorylating the β-subunit of the insulin receptor [579]. Zinc also inhibits the phosphatase and tensin homolog (PTEN), which normally dephosphorylates PIP3, thereby suppressing the activation of Akt. Through PTEN inhibition, zinc promotes the further activation of Akt signaling [580, 581]. This elevated insulin signaling through zinc causes more GLUT4 transporters to move to the cell membrane, increasing glucose uptake into cells [576]. It also enhances the phosphorylation of forkhead box protein O1 (FOXO1), which suppresses gluconeogenesis [582]. Additionally, it activates S6 K, boosting protein synthesis [583], and inhibits GSK3 through phosphorylation, leading to greater glycogen synthesis [584]. GSK3 is also directly inhibited by zinc [585]. Furthermore, zinc inhibits PP2 A, which otherwise would inhibit Akt and S6 K [230, 586].

Diabetic individuals, especially older adults, often exhibit lower serum zinc levels than healthy controls. Low zinc levels are associated with higher fasting blood sugar and HbA1c levels, indicating poorer glycemic control in type 2 diabetes mellitus [587,588,589]. Zinc deficiency correlates with the severity of diabetic complications, including peripheral neuropathy. In case–control study, diabetic patients with peripheral neuropathy had lower serum zinc levels, and zinc levels were negatively correlated with HbA1c, neuropathy scores (NSC and MNSI), and nerve conduction [590]. Zinc supplementation has shown promising effects in improving neuropathic symptoms. Patients with neuropathy treated with zinc sulfate showed significant improvements in fasting and postprandial blood glucose and motor nerve conduction velocity compared to the placebo group [591, 592]. Additionally, zinc supplementation may reduce the risk of diabetic nephropathy complications [593].

Studies also indicate that zinc supplementation in type 2 diabetes mellitus patients can reduce fasting blood glucose, HbA1c, and lipid profiles, including TC and LDL-C, while increasing HDL-C, highlighting its potential as a beneficial adjunct therapy for older adults with diabetes [455, 594, 595].

Given the significant prevalence of diabetes among older adults, zinc supplementation offers an accessible strategy to improve glycemic control, mitigate complications, and enhance overall health and quality of life in this vulnerable population.

Conclusion

In conclusion, zinc is a crucial nutrient that supports immune health and reduces inflammation in older adults, providing valuable protection against age-related diseases. Zinc plays multiple roles in the immune system by strengthening the activity of various immune cells, including granulocytes, NK cells, macrophages, T cells, and B cells. As we age, immune function often weakens, leading to a higher risk of infections, chronic low-level inflammation, and reduced cellular function, challenges that zinc can help address through targeted support of these immune cells.

Zinc supports healthy cell function, which helps lower the risk of cancer and infections, reduces age-related inflammation/inflammaging, and boosts Treg activity, thereby decreasing the likelihood of autoimmune diseases. Zinc’s role in reducing oxidative stress and modulating inflammatory pathways further suggests its potential to delay or reduce the progression of specific age-related diseases, including age-related macular degeneration, diabetes, depression, atherosclerosis, and impaired wound healing. In the case of AMD, zinc is integral in combating oxidative damage in retinal cells, thereby slowing the disease’s progression and preserving vision. For diabetes, zinc’s involvement in glucose metabolism and insulin regulation can help reduce inflammatory markers associated with the disease, potentially aiding in glycemic control and protecting against diabetes-related complications. In the context of depression, zinc's anti-inflammatory and neuroprotective effects show promise in managing age-related mental health challenges by reducing inflammation that may contribute to mood disorders. Atherosclerosis, a condition driven by chronic inflammation in blood vessels, may also be mitigated by zinc’s anti-inflammatory actions, supporting cardiovascular health by reducing the buildup of plaques in arterial walls. Additionally, zinc plays a role in collagen synthesis and cellular repair, which is critical in improving wound healing, a process that often becomes compromised with age.

This review underlines the essential nature of maintaining adequate zinc levels in promoting immune health, resilience, and reducing the risk of chronic conditions in older adults. Considering its low cost, safety, and accessibility, zinc presents itself as a valuable intervention to support healthy aging and improve quality of life. By targeting both cellular health and inflammation, zinc addresses multiple facets of immunosenescence, making it a key component in comprehensive strategies to enhance immunity, delay age-related diseases, and bolster overall well-being in elderly populations.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

AID:

Activation-induced cytidine deaminase

Akt:

Protein Kinase B

ALI:

Acute lung injury

AMD:

Age-related macular degeneration

AP-1:

Activator protein-1

APC:

Antigen presenting cell

BCR:

B cell receptor

Breg:

Regulatory B cell

CFH:

Complement factor H

CVD:

Cardiovascular disease

DC:

Dendritic cell

DGE:

German Nutrition Society

ECM:

Extracellular matrix

EFSA:

European Food Safety Authority

eNOS:

Endothelial nitric oxide synthase

ERK1/2:

Extracellular-signal regulated kinases 1/2

fMLP:

N-formylmethionyl-leucyl-phenylalanine

FOXO1:

Forkhead box protein O1

G-CSF:

Granulocyte-colony stimulating factor

GM-CSF:

Granulocyte–macrophage colony stimulating factor

GPR39:

G protein-coupled receptor 39

GSK3:

Glycogen synthase kinase 3

HCC:

Hepatocellular carcinoma

HDL-C:

High-density lipoprotein cholesterol

HIF-1:

Hypoxia-inducible factor-1

HLA:

Human leucocyte antigen

HSC:

Hematopoietic stem cell

IFN:

Interferon

Ig:

Immunoglobulin

IL:

Interleukin

iNOS:

Inducible nitric oxide synthase

IRS:

Insulin receptor substrates

KIR:

Killer cell immunoglobulin-like receptor

LDL-C:

Low-density lipoprotein cholesterol

LPS:

Lipopolysaccharide

MAPK:

Mitogen-activated protein kinase

MCP-1:

Monocyte chemotactic protein-1

mDC:

Myeloid dendritic cell

MDD:

Major Depressive Disorder

MHC:

Major histocompatibility complex

MLC:

Mixed lymphocyte culture

MMP:

Matrix metalloproteinase

MPO:

Myeloperoxidase

MT:

Metallothionein

mTORC1:

Mammalian target of rapamycin complex 1

NET:

Neutrophils release extracellular trap

NF-κB:

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

NIH:

National Institutes of Health

NK cells:

Natural killer cell

NLR:

Neutrophil-to-lymphocyte ratio

PAMP:

Pathogen-associated molecular patterns

PBMC:

Peripheral blood mononuclear cell

PD-1:

Programmed Cell Death Protein 1

PD-L:

Programmed Death-Ligand

pDC:

Plasmacytoid dendritic cell

PHA:

Phytohemagglutinin

PI3 K:

Phosphoinositide 3-kinase

PIP3 :

Phosphatidylinositol-3,4,5-trisphosphat

PKC:

Protein kinase C

PMN:

Polymorphonuclear neutrophil

PP2 A:

Protein phosphatase 2

PPAR:

Peroxisome proliferator-activated receptor

PTEN:

Phosphatase and tensin homolog

PTP1B:

Tyrosine phosphatase 1B

ROS:

Reactive oxygen species

RPE:

Retinal pigment epithelium

S6 K:

S6 kinase

SNP:

Single nucleotide polymorphism

SOD:

Superoxide dismutase

TC:

Total cholesterol

TCR:

T cell receptor

TGF:

Transforming growth factor

TLR:

Toll-like receptor

TNF:

Tumor necrosis factor

TPEN:

N,N,N′,N′-Tetrakis(2-pyridinylmethyl)-1,2-ethanediamine

Tregs:

Regulatory T cell

VCAM-1:

Vascular cell adhesion molecule-1

VEGF:

Vascular endothelial growth factor

WHO:

World Health Organization

XCI:

X chromosome inactivation

Xi:

Inactive X chromosome

ZAG:

Zinc-α2-glycoprotein

ZD:

Zinc deficiency

ZnT:

Zinc transporter

References

  1. Jaul E, Barron J. Age-related diseases and clinical and public health implications for the 85 years old and over population. Front Public Health. 2017;5:335.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Aw D, Silva AB, Palmer DB. Immunosenescence: emerging challenges for an ageing population. Immunology. 2007;120(4):435–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Zheng Y, Liu Q, Goronzy JJ, Weyand CM. Immune aging - A mechanism in autoimmune disease. Semin Immunol. 2023;69:101814.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ottaviani E, et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000;908:244–54.

    Article  CAS  PubMed  Google Scholar 

  5. Wessels I, Fischer HJ, Rink L. Dietary and physiological effects of zinc on the immune system. Annu Rev Nutr. 2021;41:133–75.

    Article  CAS  PubMed  Google Scholar 

  6. August D, Janghorbani M, Young VR. Determination of zinc and copper absorption at three dietary Zn-Cu ratios by using stable isotope methods in young adult and elderly subjects. Am J Clin Nutr. 1989;50(6):1457–63.

    Article  CAS  PubMed  Google Scholar 

  7. Maares M, Haase H. A guide to human zinc absorption: general overview and recent advances of in vitro intestinal models. Nutrients. 2020;12(3):762.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Teil E. Nationale Verzehrsstudie II. Max Rubner-Institut, Editor Bundesforschungsinstitut für Ernährung und Lebensmittel. 2008.

  9. EFSA Panel on Dietetic Products N, Allergies. Scientific opinion on dietary reference values for zinc. EFSA J. 2014;12(10):3844.

  10. Haase H, Rink L. The immune system and the impact of zinc during aging. Immun Ageing. 2009;6:9.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Bailey RL, West KP Jr, Black RE. The epidemiology of global micronutrient deficiencies. Ann Nutr Metab. 2015;66 Suppl 2(Suppl. 2):22–33.

    Article  PubMed  Google Scholar 

  12. Hasan R, Rink L, Haase H. Chelation of Free Zn(2)(+) impairs chemotaxis, phagocytosis, oxidative burst, degranulation, and cytokine production by neutrophil granulocytes. Biol Trace Elem Res. 2016;171(1):79–88.

    Article  CAS  PubMed  Google Scholar 

  13. Bogden JD, Oleske JM, Lavenhar MA, Munves EM, Kemp FW, Bruening KS, et al. Effects of one year of supplementation with zinc and other micronutrients on cellular immunity in the elderly. J Am Coll Nutr. 1990;9(3):214–25.

    Article  CAS  PubMed  Google Scholar 

  14. Xie W, Xue Q, Niu L, Wong KW. Zinc transporter SLC39A7 relieves zinc deficiency to suppress alternative macrophage activation and impairment of phagocytosis. PLoS ONE. 2020;15(7):e0235776.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Uciechowski P, Kahmann L, Plumakers B, Malavolta M, Mocchegiani E, Dedoussis G, et al. TH1 and TH2 cell polarization increases with aging and is modulated by zinc supplementation. Exp Gerontol. 2008;43(5):493–8.

    Article  CAS  PubMed  Google Scholar 

  16. Duchateau J, Delepesse G, Vrijens R, Collet H. Beneficial effects of oral zinc supplementation on the immune response of old people. Am J Med. 1981;70(5):1001–4.

    Article  CAS  PubMed  Google Scholar 

  17. Hosseini R, Ferns GA, Sahebkar A, Mirshekar MA, Jalali M. Zinc supplementation is associated with a reduction in serum markers of inflammation and oxidative stress in adults: A systematic review and meta-analysis of randomized controlled trials. Cytokine. 2021;138:155396.

    Article  CAS  PubMed  Google Scholar 

  18. Haase H, Ober-Blobaum JL, Engelhardt G, Hebel S, Heit A, Heine H, et al. Zinc signals are essential for lipopolysaccharide-induced signal transduction in monocytes. J Immunol. 2008;181(9):6491–502.

    Article  CAS  PubMed  Google Scholar 

  19. Shen H, Oesterling E, Stromberg A, Toborek M, MacDonald R, Hennig B. Zinc deficiency induces vascular pro-inflammatory parameters associated with NF-kappaB and PPAR signaling. J Am Coll Nutr. 2008;27(5):577–87.

    Article  CAS  PubMed  Google Scholar 

  20. van Leeuwen R, Boekhoorn S, Vingerling JR, Witteman JC, Klaver CC, Hofman A, et al. Dietary intake of antioxidants and risk of age-related macular degeneration. JAMA. 2005;294(24):3101–7.

    Article  PubMed  Google Scholar 

  21. Farooq M. Zinc Deficiency is Associated with Poor Glycemic Control. J Coll Physicians Surg Pak. 2019;29(3):253–7.

    Article  PubMed  Google Scholar 

  22. Zorrilla P, Gomez LA, Salido JA, Silva A, Lopez-Alonso A. Low serum zinc level as a predictive factor of delayed wound healing in total hip replacement. Wound Repair Regen. 2006;14(2):119–22.

    Article  PubMed  Google Scholar 

  23. Vashum KP, McEvoy M, Milton AH, McElduff P, Hure A, Byles J, et al. Dietary zinc is associated with a lower incidence of depression: findings from two Australian cohorts. J Affect Disord. 2014;166:249–57.

    Article  CAS  PubMed  Google Scholar 

  24. Chen W, Eisenberg R, Mowrey WB, Wylie-Rosett J, Abramowitz MK, Bushinsky DA, et al. Association between dietary zinc intake and abdominal aortic calcification in US adults. Nephrol Dial Transplant. 2020;35(7):1171–8.

    Article  CAS  PubMed  Google Scholar 

  25. Newsome DA, Swartz M, Leone NC, Elston RC, Miller E. Oral zinc in macular degeneration. Arch Ophthalmol. 1988;106(2):192–8.

    Article  CAS  PubMed  Google Scholar 

  26. Tena-Campos M, Ramon E, Borroto-Escuela DO, Fuxe K, Garriga P. The zinc binding receptor GPR39 interacts with 5-HT1A and GalR1 to form dynamic heteroreceptor complexes with signaling diversity. Biochim Biophys Acta. 2015;1852(12):2585–92.

    Article  CAS  PubMed  Google Scholar 

  27. McAllister BB, Dyck RH. Zinc transporter 3 (ZnT3) and vesicular zinc in central nervous system function. Neurosci Biobehav Rev. 2017;80:329–50.

    Article  CAS  PubMed  Google Scholar 

  28. Zalewski PD, Beltrame JF, Wawer AA, Abdo AI, Murgia C. Roles for endothelial zinc homeostasis in vascular physiology and coronary artery disease. Crit Rev Food Sci Nutr. 2019;59(21):3511–25.

    Article  CAS  PubMed  Google Scholar 

  29. Orino K, Watanabe K. Molecular, physiological and clinical aspects of the iron storage protein ferritin. Vet J. 2008;178(2):191–201.

    Article  CAS  PubMed  Google Scholar 

  30. Ravisankar P, Reddy AA, Nagalakshmi B, Koushik OS, Kumar BV, Anvith PS. The comprehensive review on fat soluble vitamins. IOSR Journal of Pharmacy. 2015;5(11):12–28.

    CAS  Google Scholar 

  31. Rink L, Gabriel P. Zinc and the immune system. Proc Nutr Soc. 2000;59(4):541–52.

    Article  CAS  PubMed  Google Scholar 

  32. Rudzinska A, Piotrowicz K, Perera I, Gryglewska B, Gasowski J. Poor Appetite in Frail Older Persons-A Systematic Review. Nutrients. 2023;15(13).

  33. Volkert D, Chourdakis M, Faxen-Irving G, Fruhwald T, Landi F, Suominen MH, et al. ESPEN guidelines on nutrition in dementia. Clin Nutr. 2015;34(6):1052–73.

    Article  PubMed  Google Scholar 

  34. Carvalho TS, Lussi A. Age-related morphological, histological and functional changes in teeth. J Oral Rehabil. 2017;44(4):291–8.

    Article  CAS  PubMed  Google Scholar 

  35. Brownie S. Why are elderly individuals at risk of nutritional deficiency? Int J Nurs Pract. 2006;12(2):110–8.

    Article  PubMed  Google Scholar 

  36. Kim J, Parish AL. Polypharmacy and Medication Management in Older Adults. Nurs Clin North Am. 2017;52(3):457–68.

    Article  PubMed  Google Scholar 

  37. Braun LA, Rosenfeldt F. Pharmaco-nutrient interactions - a systematic review of zinc and antihypertensive therapy. Int J Clin Pract. 2013;67(8):717–25.

    Article  CAS  PubMed  Google Scholar 

  38. Pecoud A, Donzel P, Schelling JL. Effect of foodstuffs on the absorption of zinc sulfate. Clin Pharmacol Ther. 1975;17(4):469–74.

    Article  CAS  PubMed  Google Scholar 

  39. Lonnerdal B. Dietary factors influencing zinc absorption. J Nutr. 2000;130(5S Suppl):1378S-S1383.

    Article  CAS  PubMed  Google Scholar 

  40. Sanna A, Firinu D, Zavattari P, Valera P. Zinc Status and Autoimmunity: A Systematic Review and Meta-Analysis. Nutrients. 2018;10(1).

  41. Beyersmann D, Haase H. Functions of zinc in signaling, proliferation and differentiation of mammalian cells. Biometals. 2001;14(3–4):331–41.

    Article  CAS  PubMed  Google Scholar 

  42. Stiles LI, Ferrao K, Mehta KJ. Role of zinc in health and disease. Clin Exp Med. 2024;24(1):38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Andrews GK. Regulation and function of Zip4, the acrodermatitis enteropathica gene. Biochem Soc Trans. 2008;36(Pt 6):1242–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Anzilotti C, Swan DJ, Boisson B, Deobagkar-Lele M, Oliveira C, Chabosseau P, et al. An essential role for the Zn(2+) transporter ZIP7 in B cell development. Nat Immunol. 2019;20(3):350–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Aydemir TB, Liuzzi JP, McClellan S, Cousins RJ. Zinc transporter ZIP8 (SLC39A8) and zinc influence IFN-gamma expression in activated human T cells. J Leukoc Biol. 2009;86(2):337–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Aydemir TB, Cousins RJ. The Multiple Faces of the Metal Transporter ZIP14 (SLC39A14). J Nutr. 2018;148(2):174–84.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Golan Y, Kambe T, Assaraf YG. The role of the zinc transporter SLC30A2/ZnT2 in transient neonatal zinc deficiency. Metallomics. 2017;9(10):1352–66.

    Article  CAS  PubMed  Google Scholar 

  48. Baltaci AK, Yuce K, Mogulkoc R. Zinc Metabolism and Metallothioneins. Biol Trace Elem Res. 2018;183(1):22–31.

    Article  CAS  PubMed  Google Scholar 

  49. Matthews JM, Sunde M. Zinc Fingers-Folds for Many Occasions. IUBMB Life. 2002;54(6):351–5.

    Article  CAS  PubMed  Google Scholar 

  50. Schlichtkrull J, Årtun T, Waterman HI, Eliasson NA, Thorell B. Insulin Crystals. I. The Minimum Mole-Fraction of Metal in Insulin Crystals Prepared with Zn++, Cd++, Co++, Ni++, Cu++, Mn++, or Fe++. Acta Chemica Scandinavica. 1956;10:1455–8.

  51. Andrews GK, Wang H, Dey SK, Palmiter RD. Mouse zinc transporter 1 gene provides an essential function during early embryonic development. Genesis. 2004;40(2):74–81.

    Article  CAS  PubMed  Google Scholar 

  52. Chowanadisai W, Lönnerdal B, Kelleher SL. Identification of a mutation in SLC30A2 (ZnT-2) in women with low milk zinc concentration that results in transient neonatal zinc deficiency. J Biol Chem. 2006;281(51):39699–707.

    Article  CAS  PubMed  Google Scholar 

  53. Itsumura N, Kibihara Y, Fukue K, Miyata A, Fukushima K, Tamagawa-Mineoka R, et al. Novel mutations in SLC30A2 involved in the pathogenesis of transient neonatal zinc deficiency. Pediatr Res. 2016;80(4):586–94.

    Article  CAS  PubMed  Google Scholar 

  54. Adlard PA, Parncutt JM, Finkelstein DI, Bush AI. Cognitive loss in zinc transporter-3 knock-out mice: a phenocopy for the synaptic and memory deficits of Alzheimer’s disease? J Neurosci. 2010;30(5):1631–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Hildebrand MS, Phillips AM, Mullen SA, Adlard PA, Hardies K, Damiano JA, et al. Loss of synaptic Zn2+ transporter function increases risk of febrile seizures. Sci Rep. 2015;5:17816.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Huang L, Gitschier J. A novel gene involved in zinc transport is deficient in the lethal milk mouse. Nat Genet. 1997;17(3):292–7.

    Article  CAS  PubMed  Google Scholar 

  57. Nishida K, Hasegawa A, Nakae S, Oboki K, Saito H, Yamasaki S, et al. Zinc transporter Znt5/Slc30a5 is required for the mast cell-mediated delayed-type allergic reaction but not the immediate-type reaction. J Exp Med. 2009;206(6):1351–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Huang L, Yu YY, Kirschke CP, Gertz ER, Lloyd KK. Znt7 (Slc30a7)-deficient mice display reduced body zinc status and body fat accumulation. J Biol Chem. 2007;282(51):37053–63.

    Article  CAS  PubMed  Google Scholar 

  59. Huang L, Kirschke CP, Lay YA, Levy LB, Lamirande DE, Zhang PH. Znt7-null mice are more susceptible to diet-induced glucose intolerance and insulin resistance. J Biol Chem. 2012;287(40):33883–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wenzlau JM, Juhl K, Yu L, Moua O, Sarkar SA, Gottlieb P, et al. The cation efflux transporter ZnT8 (Slc30A8) is a major autoantigen in human type 1 diabetes. Proc Natl Acad Sci U S A. 2007;104(43):17040–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Perez Y, Shorer Z, Liani-Leibson K, Chabosseau P, Kadir R, Volodarsky M, et al. SLC30A9 mutation affecting intracellular zinc homeostasis causes a novel cerebro-renal syndrome. Brain. 2017;140(4):928–39.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Quadri M, Federico A, Zhao T, Breedveld GJ, Battisti C, Delnooz C, et al. Mutations in SLC30A10 cause parkinsonism and dystonia with hypermanganesemia, polycythemia, and chronic liver disease. Am J Hum Genet. 2012;90(3):467–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Stamelou M, Tuschl K, Chong WK, Burroughs AK, Mills PB, Bhatia KP, et al. Dystonia with brain manganese accumulation resulting from SLC30A10 mutations: a new treatable disorder. Mov Disord. 2012;27(10):1317–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Hara T, Takeda TA, Takagishi T, Fukue K, Kambe T, Fukada T. Physiological roles of zinc transporters: molecular and genetic importance in zinc homeostasis. J Physiol Sci. 2017;67(2):283–301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Dufner-Beattie J, Huang ZL, Geiser J, Xu W, Andrews GK. Mouse ZIP1 and ZIP3 genes together are essential for adaptation to dietary zinc deficiency during pregnancy. Genesis. 2006;44(5):239–51.

    Article  CAS  PubMed  Google Scholar 

  66. Peters JL, Dufner-Beattie J, Xu W, Geiser J, Lahner B, Salt DE, et al. Targeting of the mouse Slc39a2 (Zip2) gene reveals highly cell-specific patterns of expression, and unique functions in zinc, iron, and calcium homeostasis. Genesis. 2007;45(6):339–52.

    Article  CAS  PubMed  Google Scholar 

  67. Dufner-Beattie J, Huang ZL, Geiser J, Xu W, Andrews GK. Generation and characterization of mice lacking the zinc uptake transporter ZIP3. Mol Cell Biol. 2005;25(13):5607–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Guo H, Jin X, Zhu T, Wang T, Tong P, Tian L, et al. SLC39A5 mutations interfering with the BMP/TGF-β pathway in non-syndromic high myopia. J Med Genet. 2014;51(8):518–25.

    Article  CAS  PubMed  Google Scholar 

  69. Yu M, Lee WW, Tomar D, Pryshchep S, Czesnikiewicz-Guzik M, Lamar DL, et al. Regulation of T cell receptor signaling by activation-induced zinc influx. J Exp Med. 2011;208(4):775–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Taylor KM, Muraina IA, Brethour D, Schmitt-Ulms G, Nimmanon T, Ziliotto S, et al. Zinc transporter ZIP10 forms a heteromer with ZIP6 which regulates embryonic development and cell migration. Biochem J. 2016;473(16):2531–44.

    Article  CAS  PubMed  Google Scholar 

  71. Li D, Achkar JP, Haritunians T, Jacobs JP, Hui KY, D’Amato M, et al. A Pleiotropic Missense Variant in SLC39A8 Is Associated With Crohn’s Disease and Human Gut Microbiome Composition. Gastroenterology. 2016;151(4):724–32.

    Article  CAS  PubMed  Google Scholar 

  72. Thomas P, Pang Y, Dong J, Berg AH. Identification and characterization of membrane androgen receptors in the ZIP9 zinc transporter subfamily: II. Role of human ZIP9 in testosterone-induced prostate and breast cancer cell apoptosis. Endocrinology. 2014;155(11):4250–65.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Shintaro H, Tomohiro M, Toshiyuki F. B-cell receptor strength and zinc signaling: Unraveling the role of zinc transporter ZIP10 in humoral immunity. Receptors & Clinical Investigation. 2015.

  74. Gao H, Zhao L, Wang H, Xie E, Wang X, Wu Q, et al. Metal transporter Slc39a10 regulates susceptibility to inflammatory stimuli by controlling macrophage survival. Proc Natl Acad Sci U S A. 2017;114(49):12940–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Martin AB, Aydemir TB, Guthrie GJ, Samuelson DA, Chang SM, Cousins RJ. Gastric and colonic zinc transporter ZIP11 (Slc39a11) in mice responds to dietary zinc and exhibits nuclear localization. J Nutr. 2013;143(12):1882–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Kang X, Chen R, Zhang J, Li G, Dai PG, Chen C, et al. Expression Profile Analysis of Zinc Transporters (ZIP4, ZIP9, ZIP11, ZnT9) in Gliomas and their Correlation with IDH1 Mutation Status. Asian Pac J Cancer Prev. 2015;16(8):3355–60.

    Article  PubMed  Google Scholar 

  77. Kelleher SL, Velasquez V, Croxford TP, McCormick NH, Lopez V, MacDavid J. Mapping the zinc-transporting system in mammary cells: molecular analysis reveals a phenotype-dependent zinc-transporting network during lactation. J Cell Physiol. 2012;227(4):1761–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Yu Y, Wu A, Zhang Z, Yan G, Zhang F, Zhang L, et al. Characterization of the GufA subfamily member SLC39A11/Zip11 as a zinc transporter. J Nutr Biochem. 2013;24(10):1697–708.

    Article  CAS  PubMed  Google Scholar 

  79. Davis DN, Strong MD, Chambers E, Hart MD, Bettaieb A, Clarke SL, et al. A role for zinc transporter gene SLC39A12 in the nervous system and beyond. Gene. 2021;799:145824.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Zhu X, Yu C, Wu W, Shi L, Jiang C, Wang L, et al. Zinc transporter ZIP12 maintains zinc homeostasis and protects spermatogonia from oxidative stress during spermatogenesis. Reprod Biol Endocrinol. 2022;20(1):17.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Ye C, Lian G, Wang T, Chen A, Chen W, Gong J, et al. The zinc transporter ZIP12 regulates monocrotaline-induced proliferation and migration of pulmonary arterial smooth muscle cells via the AKT/ERK signaling pathways. BMC Pulm Med. 2022;22(1):111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Fukada T, Civic N, Furuichi T, Shimoda S, Mishima K, Higashiyama H, et al. The zinc transporter SLC39A13/ZIP13 is required for connective tissue development; its involvement in BMP/TGF-beta signaling pathways. PLoS One. 2008;3(11):e3642.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Giunta C, Elçioglu NH, Albrecht B, Eich G, Chambaz C, Janecke AR, et al. Spondylocheiro dysplastic form of the Ehlers-Danlos syndrome–an autosomal-recessive entity caused by mutations in the zinc transporter gene SLC39A13. Am J Hum Genet. 2008;82(6):1290–305.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Tuschl K, Meyer E, Valdivia LE, Zhao N, Dadswell C, Abdul-Sada A, et al. Mutations in SLC39A14 disrupt manganese homeostasis and cause childhood-onset parkinsonism-dystonia. Nat Commun. 2016;7:11601.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Hojyo S, Fukada T, Shimoda S, Ohashi W, Bin BH, Koseki H, et al. The zinc transporter SLC39A14/ZIP14 controls G-protein coupled receptor-mediated signaling required for systemic growth. PLoS One. 2011;6(3):e18059.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Jimenez-Rondan FR, Ruggiero CH, Riva A, Yu F, Stafford LS, Cross TR, et al. Deletion of metal transporter Zip14 reduces major histocompatibility complex II expression in murine small intestinal epithelial cells. Proc Natl Acad Sci U S A. 2025;122(1):e2422321121.

    Article  CAS  PubMed  Google Scholar 

  87. Wessels I, Cousins RJ. Zinc dyshomeostasis during polymicrobial sepsis in mice involves zinc transporter Zip14 and can be overcome by zinc supplementation. Am J Physiol Gastrointest Liver Physiol. 2015;309(9):G768–78.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Aydemir TB, Chang SM, Guthrie GJ, Maki AB, Ryu MS, Karabiyik A, et al. Zinc transporter ZIP14 functions in hepatic zinc, iron and glucose homeostasis during the innate immune response (endotoxemia). PLoS ONE. 2012;7(10):e48679.

    Article  CAS  PubMed  Google Scholar 

  89. Zink: Deutsche Gesellschaft für Ernährung e. V.; [Available from: https://www.dge.de/wissenschaft/referenzwerte/zink/.

  90. Organization WH. Vitamin and mineral requirements in human nutrition: World Health Organization; 2004.

  91. Zinc - Fact Sheet for Health Professionals: National Institutes of Health; [Available from: https://ods.od.nih.gov/factsheets/Zinc-HealthProfessional/.

  92. Schoofs H, Schmit J, Rink L. Zinc Toxicity: Understanding the Limits. Molecules. 2024;29(13).

  93. Plum LM, Rink L, Haase H. The Essential Toxin: Impact of Zinc on Human Health. Int J Environ Res Public Health. 2010;7(4):1342–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Wahab A, Mushtaq K, Borak SG, Bellam N. Zinc-induced copper deficiency, sideroblastic anemia, and neutropenia: A perplexing facet of zinc excess. Clin Case Rep. 2020;8(9):1666–71.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Trame S, Wessels I, Haase H, Rink L. A short 18 items food frequency questionnaire biochemically validated to estimate zinc status in humans. J Trace Elem Med Biol. 2018;49:285–95.

    Article  CAS  PubMed  Google Scholar 

  96. Trame S, Brüggemann A, Rink L. Calculating zinc uptake by Zinc-APP. J Trace Elem Med Biol. 2023;77:127132.

    Article  PubMed  Google Scholar 

  97. Giefing-Kröll C, Berger P, Lepperdinger G, Grubeck-Loebenstein B. How sex and age affect immune responses, susceptibility to infections, and response to vaccination. Aging Cell. 2015;14(3):309–21.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Fish EN. The X-files in immunity: sex-based differences predispose immune responses. Nat Rev Immunol. 2008;8(9):737–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Kanda N, Tamaki K. Estrogen enhances immunoglobulin production by human PBMCs. J Allergy Clin Immunol. 1999;103(2):282–8.

    Article  CAS  PubMed  Google Scholar 

  100. Kanda N, Tsuchida T, Tamaki K. Testosterone inhibits immunoglobulin production by human peripheral blood mononuclear cells. Clin Exp Immunol. 1996;106(2):410–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Amadori A, Zamarchi R, De Silvestro G, Forza G, Cavatton G, Danieli GA, et al. Genetic control of the CD4/CD8 T-cell ratio in humans. Nat Med. 1995;1(12):1279–83.

    Article  CAS  PubMed  Google Scholar 

  102. Gilmore W, Weiner LP, Correale J. Effect of estradiol on cytokine secretion by proteolipid protein-specific T cell clones isolated from multiple sclerosis patients and normal control subjects. J Immunol (Baltimore, Md: 1950). 1997;158(1):446–51.

    Article  CAS  Google Scholar 

  103. Fox H, Bond B, Parslow T. Estrogen regulates the IFN-gamma promoter. J Immunol (Baltimore Md: 1950). 1991;146(12):4362–7.

    Article  CAS  Google Scholar 

  104. Bouman A, Heineman MJ, Faas MM. Sex hormones and the immune response in humans. Hum Reprod Update. 2005;11(4):411–23.

    Article  CAS  PubMed  Google Scholar 

  105. Calippe B, Douin-Echinard V, Laffargue M, Laurell H, Rana-Poussine V, Pipy B, et al. Chronic estradiol administration in vivo promotes the proinflammatory response of macrophages to TLR4 activation: involvement of the phosphatidylinositol 3-kinase pathway. J Immunol. 2008;180(12):7980–8.

    Article  CAS  PubMed  Google Scholar 

  106. Berghöfer B, Frommer T, Haley G, Fink L, Bein G, Hackstein H. TLR7 ligands induce higher IFN-α production in females. J Immunol. 2006;177(4):2088–96.

    Article  PubMed  Google Scholar 

  107. Griesbeck M, Ziegler S, Laffont S, Smith N, Chauveau L, Tomezsko P, et al. Sex differences in plasmacytoid dendritic cell levels of IRF5 drive higher IFN-α production in women. J Immunol. 2015;195(11):5327–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Carrel L, Willard HF. X-inactivation profile reveals extensive variability in X-linked gene expression in females. Nature. 2005;434(7031):400–4.

    Article  CAS  PubMed  Google Scholar 

  109. Balaton BP, Brown CJ. Escape Artists of the X Chromosome. Trends Genet. 2016;32(6):348–59.

    Article  CAS  PubMed  Google Scholar 

  110. Sun Z, Fan J, Wang Y. X-Chromosome Inactivation and Related Diseases. Genet Res (Camb). 2022;2022:1391807.

    Article  PubMed  Google Scholar 

  111. Buckler H. The menopause transition: endocrine changes and clinical symptoms. British Menopause Society Journal. 2005;11(2):61–5.

    Article  Google Scholar 

  112. Giglio T, Imro MA, Filaci G, Scudeletti M, Puppo F, De Cecco L, et al. Immune cell circulating subsets are affected by gonadal function. Life Sci. 1994;54(18):1305–12.

    Article  CAS  PubMed  Google Scholar 

  113. Deguchi K, Kamada M, Irahara M, Maegawa M, Yamamoto S, Ohmoto Y, et al. Postmenopausal changes in production of type 1 and type 2 cytokines and the effects of hormone replacement therapy. Menopause. 2001;8(4):266–73.

    Article  CAS  PubMed  Google Scholar 

  114. Vural P, Akgul C, Canbaz M. Effects of hormone replacement therapy on plasma pro-inflammatory and anti-inflammatory cytokines and some bone turnover markers in postmenopausal women. Pharmacol Res. 2006;54(4):298–302.

    Article  CAS  PubMed  Google Scholar 

  115. Yasui T, Maegawa M, Tomita J, Miyatani Y, Yamada M, Uemura H, et al. Changes in serum cytokine concentrations during the menopausal transition. Maturitas. 2007;56(4):396–403.

    Article  CAS  PubMed  Google Scholar 

  116. Marttila S, Jylhävä J, Nevalainen T, Nykter M, Jylhä M, Hervonen A, et al. Transcriptional analysis reveals gender-specific changes in the aging of the human immune system. PLoS One. 2013;8(6):e66229.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Giefing-Kröll C, Berger P, Lepperdinger G, Grubeck-Loebenstein B. How sex and age affect immune responses, susceptibility to infections, and response to vaccination. Aging Cell. 2015;14(3):309–21.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Kamada M, Irahara M, Maegawa M, Yasui T, Yamano S, Yamada M, et al. B cell subsets in postmenopausal women and the effect of hormone replacement therapy. Maturitas. 2001;37(3):173–9.

    Article  CAS  PubMed  Google Scholar 

  119. Saucedo R, Rico G, Basurto L, Ochoa R, Zárate A. Transdermal estradiol in menopausal women depresses interleukin-6 without affecting other markers of immune response. Gynecol Obstet Invest. 2002;53(2):114–7.

    Article  CAS  PubMed  Google Scholar 

  120. Calabrò A, Accardi G, Aiello A, Caruso C, Candore G. Sex and gender affect immune aging. Front Aging. 2023;4:1272118.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Amulic B, Cazalet C, Hayes GL, Metzler KD, Zychlinsky A. Neutrophil function: from mechanisms to disease. Annu Rev Immunol. 2012;30(1):459–89.

    Article  CAS  PubMed  Google Scholar 

  122. Laupland KB, Church DL, Mucenski M, Sutherland LR, Davies HD. Population-based study of the epidemiology of and the risk factors for invasive Staphylococcus aureus infections. J Infect Dis. 2003;187(9):1452–9.

    Article  PubMed  Google Scholar 

  123. Pellegrino R, Paganelli R, Di Iorio A, Bandinelli S, Moretti A, Iolascon G, et al. Neutrophil, lymphocyte count, and neutrophil to lymphocyte ratio predict multimorbidity and mortality-results from the Baltimore Longitudinal Study on Aging follow-up study. Geroscience. 2024;46(3):3047–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Uciechowski P, Rink L. Neutrophil Granulocyte Functions in the Elderly. 2009.

  125. Chatta GS, Andrews RG, Rodger E, Schrag M, Hammond WP, Dale DC. Hematopoietic progenitors and aging: alterations in granulocytic precursors and responsiveness to recombinant human G-CSF, GM-CSF, and IL-3. J Gerontol. 1993;48(5):M207–12.

    Article  CAS  PubMed  Google Scholar 

  126. Fortin CF, Larbi A, Dupuis G, Lesur O, Fulop T Jr. GM-CSF activates the Jak/STAT pathway to rescue polymorphonuclear neutrophils from spontaneous apoptosis in young but not elderly individuals. Biogerontology. 2007;8(2):173–87.

    Article  CAS  PubMed  Google Scholar 

  127. Grilz E, Posch F, Konigsbrugge O, Schwarzinger I, Lang IM, Marosi C, et al. Association of Platelet-to-Lymphocyte Ratio and Neutrophil-to-Lymphocyte Ratio with the Risk of Thromboembolism and Mortality in Patients with Cancer. Thromb Haemost. 2018;118(11):1875–84.

    Article  PubMed  Google Scholar 

  128. Shah N, Parikh V, Patel N, Patel N, Badheka A, Deshmukh A, et al. Neutrophil lymphocyte ratio significantly improves the Framingham risk score in prediction of coronary heart disease mortality: insights from the National Health and Nutrition Examination Survey-III. Int J Cardiol. 2014;171(3):390–7.

    Article  PubMed  Google Scholar 

  129. Rodic S, McCudden C, van Walraven C. relationship between plasma zinc and red blood cell zinc levels in hospitalized patients. J Appl Lab Med. 2022;7(6):1412–23.

    Article  PubMed  Google Scholar 

  130. Fulop T Jr, Fouquet C, Allaire P, Perrin N, Lacombe G, Stankova J, et al. Changes in apoptosis of human polymorphonuclear granulocytes with aging. Mech Ageing Dev. 1997;96(1–3):15–34.

    Article  CAS  PubMed  Google Scholar 

  131. Tortorella C, Simone O, Piazzolla G, Stella I, Cappiello V, Antonaci S. Role of phosphoinositide 3-kinase and extracellular signal-regulated kinase pathways in granulocyte macrophage-colony-stimulating factor failure to delay fas-induced neutrophil apoptosis in elderly humans. J Gerontol A Biol Sci Med Sci. 2006;61(11):1111–8.

    Article  PubMed  Google Scholar 

  132. Schroder AK, von der Ohe M, Kolling U, Altstaedt J, Uciechowski P, Fleischer D, et al. Polymorphonuclear leucocytes selectively produce anti-inflammatory interleukin-1 receptor antagonist and chemokines, but fail to produce pro-inflammatory mediators. Immunology. 2006;119(3):317–27.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Fulop T, Larbi A, Douziech N, Fortin C, Guerard KP, Lesur O, et al. Signal transduction and functional changes in neutrophils with aging. Aging Cell. 2004;3(4):217–26.

    Article  CAS  PubMed  Google Scholar 

  134. Antonucci A, Di Baldassarre A, Di Giacomo F, Stuppia L, Palka G. Detection of apoptosis in peripheral blood cells of 31 subjects affected by Down syndrome before and after zinc therapy. Ultrastruct Pathol. 1997;21(5):449–52.

    Article  CAS  PubMed  Google Scholar 

  135. Oishi K, Machida K. Inhibition of neutrophil apoptosis by antioxidants in culture medium. Scand J Immunol. 1997;45(1):21–7.

    Article  CAS  PubMed  Google Scholar 

  136. Moroni F, Di Paolo ML, Rigo A, Cipriano C, Giacconi R, Recchioni R, et al. Interrelationship among neutrophil efficiency, inflammation, antioxidant activity and zinc pool in very old age. Biogerontology. 2005;6(4):271–81.

    Article  CAS  PubMed  Google Scholar 

  137. Haruta Y, Kobayakawa K, Saiwai H, Hata K, Tamaru T, Iura H, et al. Zinc chelator treatment in crush syndrome model mice attenuates ischemia-reperfusion-induced muscle injury due to suppressing of neutrophil infiltration. Sci Rep. 2022;12(1):15580.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Wessels I, Pupke JT, von Trotha KT, Gombert A, Himmelsbach A, Fischer HJ, et al. Zinc supplementation ameliorates lung injury by reducing neutrophil recruitment and activity. Thorax. 2020;75(3):253–61.

    Article  PubMed  Google Scholar 

  139. Briggs WA, Pedersen MM, Mahajan SK, Sillix DH, Prasad AS, McDonald FD. Lymphocyte and granulocyte function in zinc-treated and zinc-deficient hemodialysis patients. Kidney Int. 1982;21(6):827–32.

    Article  CAS  PubMed  Google Scholar 

  140. Kim YM, Reed W, Wu W, Bromberg PA, Graves LM, Samet JM. Zn2+-induced IL-8 expression involves AP-1, JNK, and ERK activities in human airway epithelial cells. Am J Physiol Lung Cell Mol Physiol. 2006;290(5):L1028–35.

    Article  CAS  PubMed  Google Scholar 

  141. van der Laan M, Buttgenbach A, Wolf J, Rink L, Wessels I. The Role of Zinc in GM-CSF-induced signaling in human polymorphonuclear leukocytes. Mol Nutr Food Res. 2022;66(14):e2101106.

    Article  PubMed  Google Scholar 

  142. Hujanen ES, Seppa ST, Virtanen K. Polymorphonuclear leukocyte chemotaxis induced by zinc, copper and nickel in vitro. Biochim Biophys Acta. 1995;1245(2):145–52.

    Article  PubMed  Google Scholar 

  143. Muniain M, Rodrigues C, Pozuelo F, Romero A, Mata R, Perez-Venegas J, et al. Chemotaxis in the elderly: Modifications after the intake of ascorbic acid and levamisole. Age. 1988;11(3):88–92.

    Article  CAS  Google Scholar 

  144. Niwa Y, Kasama T, Miyachi Y, Kanoh T. Neutrophil chemotaxis, phagocytosis and parameters of reactive oxygen species in human aging: cross-sectional and longitudinal studies. Life Sci. 1989;44(22):1655–64.

    Article  CAS  PubMed  Google Scholar 

  145. Sapey E, Greenwood H, Walton G, Mann E, Love A, Aaronson N, et al. Phosphoinositide 3-kinase inhibition restores neutrophil accuracy in the elderly: toward targeted treatments for immunosenescence. Blood. 2014;123(2):239–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Sapey E, Patel JM, Greenwood HL, Walton GM, Hazeldine J, Sadhra C, et al. Pulmonary Infections in the Elderly Lead to Impaired Neutrophil Targeting, Which Is Improved by Simvastatin. Am J Respir Crit Care Med. 2017;196(10):1325–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Wenisch C, Patruta S, Daxbock F, Krause R, Horl W. Effect of age on human neutrophil function. J Leukoc Biol. 2000;67(1):40–5.

    Article  CAS  PubMed  Google Scholar 

  148. Sauce D, Dong Y, Campillo-Gimenez L, Casulli S, Bayard C, Autran B, et al. Reduced Oxidative Burst by Primed Neutrophils in the Elderly Individuals Is Associated With Increased Levels of the CD16bright/CD62Ldim Immunosuppressive Subset. J Gerontol A Biol Sci Med Sci. 2017;72(2):163–72.

    Article  CAS  PubMed  Google Scholar 

  149. Butcher SK, Chahal H, Nayak L, Sinclair A, Henriquez NV, Sapey E, et al. Senescence in innate immune responses: reduced neutrophil phagocytic capacity and CD16 expression in elderly humans. J Leukoc Biol. 2001;70(6):881–6.

    Article  CAS  PubMed  Google Scholar 

  150. Beswick PH, Brannen PC, Hurles SS. The effects of smoking and zinc on the oxidative reactions of human neutrophils. J Clin Lab Immunol. 1986;21(2):71–5.

    CAS  PubMed  Google Scholar 

  151. Yatsuyanagi J, Muraoka S, Tsutsui R, Ogiso T. Inhibition of superoxide generation and of increase in intracellular Ca2+ concentration by zinc in rat neutrophils stimulated with zymosan. Chem Pharm Bull (Tokyo). 1990;38(6):1656–9.

    Article  CAS  PubMed  Google Scholar 

  152. Takeyama Y, Ogino K, Segawa H, Kobayashi H, Uda T, Houbara T. Effects of zinc on production of active oxygen species by rat neutrophils. Pharmacol Toxicol. 1995;76(1):50–5.

    Article  CAS  PubMed  Google Scholar 

  153. Ogino K, Izumi Y, Segawa H, Takeyama Y, Ishiyama H, Houbara T, et al. Zinc hydroxide induced respiratory burst in rat neutrophils. Eur J Pharmacol. 1994;270(1):73–8.

    CAS  PubMed  Google Scholar 

  154. Sunzel B, Holm S, Reuterving CO, Soderberg T, Hallmans G, Hanstrom L. The effect of zinc on bacterial phagocytosis, killing and cytoprotection in human polymorphonuclear leucocytes. APMIS. 1995;103(9):635–44.

    Article  CAS  PubMed  Google Scholar 

  155. Braga PC, Sala MT, Dal Sasso M, Mancini L, Sandrini MC, Annoni G. Influence of age on oxidative bursts (chemiluminescence) of polymorphonuclear neutrophil leukocytes. Gerontology. 1998;44(4):192–7.

    Article  CAS  PubMed  Google Scholar 

  156. Vitlic A, Khanfer R, Lord JM, Carroll D, Phillips AC. Bereavement reduces neutrophil oxidative burst only in older adults: role of the HPA axis and immunesenescence. Immun Ageing. 2014;11(1):13.

    Article  PubMed  PubMed Central  Google Scholar 

  157. Tortorella C, Ottolenghi A, Pugliese P, Jirillo E, Antonaci S. Relationship between respiratory burst and adhesiveness capacity in elderly polymorphonuclear cells. Mech Ageing Dev. 1993;69(1–2):53–63.

    Article  CAS  PubMed  Google Scholar 

  158. Lindahl M, Leanderson P, Tagesson C. Novel aspect on metal fume fever: zinc stimulates oxygen radical formation in human neutrophils. Hum Exp Toxicol. 1998;17(2):105–10.

    Article  CAS  PubMed  Google Scholar 

  159. Hasegawa H, Suzuki K, Suzuki K, Nakaji S, Sugawara K. Effects of zinc on the reactive oxygen species generating capacity of human neutrophils and on the serum opsonic activity in vitro. Luminescence. 2000;15(5):321–7.

    Article  CAS  PubMed  Google Scholar 

  160. Freitas M, Porto G, Lima JL, Fernandes E. Zinc activates neutrophils’ oxidative burst. Biometals. 2010;23(1):31–41.

    Article  CAS  PubMed  Google Scholar 

  161. Hasan R, Rink L, Haase H. Zinc signals in neutrophil granulocytes are required for the formation of neutrophil extracellular traps. Innate Immun. 2013;19(3):253–64.

    Article  PubMed  Google Scholar 

  162. Ganatra HA, Varisco BM, Harmon K, Lahni P, Opoka A, Wong HR. Zinc supplementation leads to immune modulation and improved survival in a juvenile model of murine sepsis. Innate Immun. 2017;23(1):67–76.

    Article  CAS  PubMed  Google Scholar 

  163. Kuzmicka W, Manda-Handzlik A, Cieloch A, Mroczek A, Demkow U, Wachowska M, et al. Zinc Supplementation Modulates NETs Release and Neutrophils' Degranulation. Nutrients. 2020;13(1).

  164. Tseng CW, Kyme PA, Arruda A, Ramanujan VK, Tawackoli W, Liu GY. Innate immune dysfunctions in aged mice facilitate the systemic dissemination of methicillin-resistant S. aureus. PLoS One. 2012;7(7):e41454.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Hazeldine J, Harris P, Chapple IL, Grant M, Greenwood H, Livesey A, et al. Impaired neutrophil extracellular trap formation: a novel defect in the innate immune system of aged individuals. Aging Cell. 2014;13(4):690–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Sabbatini M, Bona E, Novello G, Migliario M, Reno F. Aging hampers neutrophil extracellular traps (NETs) efficacy. Aging Clin Exp Res. 2022;34(10):2345–53.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Cakman I, Kirchner H, Rink L. Zinc supplementation reconstitutes the production of interferon-alpha by leukocytes from elderly persons. J Interferon Cytokine Res. 1997;17(8):469–72.

    Article  CAS  PubMed  Google Scholar 

  168. Starr JM, Deary IJ. Sex differences in blood cell counts in the Lothian Birth Cohort 1921 between 79 and 87 years. Maturitas. 2011;69(4):373–6.

    Article  PubMed  Google Scholar 

  169. Tsukamoto K, Machida K. Effects of life events and stress on neutrophil functions in elderly men. Immun Ageing. 2012;9(1):13.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Wessels I, Jansen J, Rink L, Uciechowski P. Immunosenescence of polymorphonuclear neutrophils. ScientificWorldJournal. 2010;10(1):145–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Pinna K, Kelley DS, Taylor PC, King JC. Research Communication: Immune Functions Are Maintained in Healthy Men with Low Zinc Intake. J Nutr. 2002;132(7):2033–6.

    Article  CAS  PubMed  Google Scholar 

  172. Someya Y, Tanihata J, Sato S, Kawano F, Shirato K, Sugiyama M, et al. Zinc-deficiency induced changes in the distribution of rat white blood cells. J Nutr Sci Vitaminol (Tokyo). 2009;55(2):162–9.

    Article  CAS  PubMed  Google Scholar 

  173. Sakakibara Y, Sato S, Kawashima Y, Someya Y, Shirato K, Tachiyashiki K, et al. Different recovery responses from dietary zinc-deficiency in the distribution of rat granulocytes. J Nutr Sci Vitaminol (Tokyo). 2011;57(2):197–201.

    Article  CAS  PubMed  Google Scholar 

  174. Poli V, Pui-Yan Ma V, Di Gioia M, Broggi A, Benamar M, Chen Q, et al. Zinc-dependent histone deacetylases drive neutrophil extracellular trap formation and potentiate local and systemic inflammation. iScience. 2021;24(11):103256.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Song C, Vandewoude M, Stevens W, De Clerck L, Van der Planken M, Whelan A, et al. Alterations in immune functions during normal aging and Alzheimer’s disease. Psychiatry Res. 1999;85(1):71–80.

    Article  CAS  PubMed  Google Scholar 

  176. van Beek AA, Fransen F, Meijer B, de Vos P, Knol EF, Savelkoul HFJ. Aged mice display altered numbers and phenotype of basophils, and bone marrow-derived basophil activation, with a limited role for aging-associated microbiota. Immun Ageing. 2018;15:32.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Scott ME, Koski KG. Zinc deficiency impairs immune responses against parasitic nematode infections at intestinal and systemic sites. J Nutr. 2000;130(5S Suppl):1412S-S1420.

    Article  CAS  PubMed  Google Scholar 

  178. Richter M, Bonneau R, Girard MA, Beaulieu C, Larivee P. Zinc status modulates bronchopulmonary eosinophil infiltration in a murine model of allergic inflammation. Chest. 2003;123(3 Suppl):446S.

    Article  PubMed  Google Scholar 

  179. Petrov VV, Vasil’eva OV, Kornilova NK, Gunin AG. Age-related changes in mast cells and eosinophils of human dermis. Ontogenez. 2013;44(3):179–85.

    CAS  PubMed  Google Scholar 

  180. Suzuki M, Ramezanpour M, Cooksley C, Lee TJ, Jeong B, Kao S, et al. Zinc-depletion associates with tissue eosinophilia and collagen depletion in chronic rhinosinusitis. Rhinology. 2020;58(5):451–9.

    CAS  PubMed  Google Scholar 

  181. Theoharides TC, Alysandratos KD, Angelidou A, Delivanis DA, Sismanopoulos N, Zhang B, et al. Mast cells and inflammation. Biochim Biophys Acta. 2012;1822(1):21–33.

    Article  CAS  PubMed  Google Scholar 

  182. Abdel Hafez SMN. Age related changes in the dermal mast cells and the associated changes in the dermal collagen and cells: A histological and electron microscopy study. Acta Histochem. 2019;121(5):619–27.

    Article  CAS  PubMed  Google Scholar 

  183. Kutukova NA, Nazarov PG, Kudryavtseva GV, Shishkin VI. Mast cells and aging. Adv Gerontol. 2017;7(1):68–75.

    Article  Google Scholar 

  184. Belanger LF. The influence of zinc-deprivation on the mast cell population of the bone marrow and other tissues. J Nutr. 1978;108(8):1315–21.

    Article  CAS  PubMed  Google Scholar 

  185. Pilkington SM, Barron MJ, Watson REB, Griffiths CEM, Bulfone-Paus S. Aged human skin accumulates mast cells with altered functionality that localize to macrophages and vasoactive intestinal peptide-positive nerve fibres. Br J Dermatol. 2019;180(4):849–58.

    Article  CAS  PubMed  Google Scholar 

  186. Kabu K, Yamasaki S, Kamimura D, Ito Y, Hasegawa A, Sato E, et al. Zinc is required for Fc epsilon RI-mediated mast cell activation. J Immunol. 2006;177(2):1296–305.

    Article  CAS  PubMed  Google Scholar 

  187. Ho LH, Ruffin RE, Murgia C, Li L, Krilis SA, Zalewski PD. Labile zinc and zinc transporter ZnT4 in mast cell granules: role in regulation of caspase activation and NF-kappaB translocation. J Immunol. 2004;172(12):7750–60.

    Article  CAS  PubMed  Google Scholar 

  188. Uchida R, Xiang H, Arai H, Kitamura H, Nishida K. L-Type Calcium Channel-Mediated Zinc Wave Is Involved in the Regulation of IL-6 by Stimulating Non-IgE with LPS and IL-33 in Mast Cells and Dendritic Cells. Biol Pharm Bull. 2019;42(1):87–93.

    Article  CAS  PubMed  Google Scholar 

  189. Kutukova NA, Nazarov PG, Kudryavtseva GV, Shishkin VI. Mast cells and aging. Adv Gerontol. 2016;29(4):586–93.

    CAS  PubMed  Google Scholar 

  190. Chatterjee V, Gashev AA. Aging-associated shifts in functional status of mast cells located by adult and aged mesenteric lymphatic vessels. Am J Physiol Heart Circ Physiol. 2012;303(6):H693-702.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Stoncius LV, Ashrafi SH, Meyer J. Ultrastructure of mast cells in the hyperplastic buccal mucosa of the zinc-deficient rat. J Oral Pathol. 1985;14(5):375–82.

    Article  CAS  PubMed  Google Scholar 

  192. Muzzioli M, Stecconi R, Moresi R, Provinciali M. Zinc improves the development of human CD34+ cell progenitors towards NK cells and increases the expression of GATA-3 transcription factor in young and old ages. Biogerontology. 2009;10(5):593–604.

    Article  CAS  PubMed  Google Scholar 

  193. Ligthart GJ, van Vlokhoven PC, Schuit HR, Hijmans W. The expanded null cell compartment in ageing: increase in the number of natural killer cells and changes in T-cell and NK-cell subsets in human blood. Immunology. 1986;59(3):353–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Mariani E, Monaco MC, Cattini L, Sinoppi M, Facchini A. Distribution and lytic activity of NK cell subsets in the elderly. Mech Ageing Dev. 1994;76(2–3):177–87.

    Article  CAS  PubMed  Google Scholar 

  195. Gounder SS, Abdullah BJJ, Radzuanb N, Zain F, Sait NBM, Chua C, et al. Effect of Aging on NK Cell Population and Their Proliferation at Ex Vivo Culture Condition. Anal Cell Pathol (Amst). 2018;2018:7871814.

    PubMed  Google Scholar 

  196. Borrego F, Alonso MC, Galiani MD, Carracedo J, Ramirez R, Ostos B, et al. NK phenotypic markers and IL2 response in NK cells from elderly people. Exp Gerontol. 1999;34(2):253–65.

    Article  CAS  PubMed  Google Scholar 

  197. Michel T, Poli A, Cuapio A, Briquemont B, Iserentant G, Ollert M, et al. Human CD56bright NK Cells: An Update. J Immunol. 2016;196(7):2923–31.

    Article  CAS  PubMed  Google Scholar 

  198. Muller-Durovic B, Lanna A, Covre LP, Mills RS, Henson SM, Akbar AN. Killer Cell Lectin-like Receptor G1 Inhibits NK Cell Function through Activation of Adenosine 5’-Monophosphate-Activated Protein Kinase. J Immunol. 2016;197(7):2891–9.

    Article  PubMed  Google Scholar 

  199. Judge SJ, Murphy WJ, Canter RJ. Characterizing the Dysfunctional NK Cell: Assessing the Clinical Relevance of Exhaustion, Anergy, and Senescence. Front Cell Infect Microbiol. 2020;10:49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Rukavina D, Laskarin G, Rubesa G, Strbo N, Bedenicki I, Manestar D, et al. Age-related decline of perforin expression in human cytotoxic T lymphocytes and natural killer cells. Blood. 1998;92(7):2410–20.

    Article  CAS  PubMed  Google Scholar 

  201. Hazeldine J, Hampson P, Lord JM. Reduced release and binding of perforin at the immunological synapse underlies the age-related decline in natural killer cell cytotoxicity. Aging Cell. 2012;11(5):751–9.

    Article  CAS  PubMed  Google Scholar 

  202. Rolles B, Maywald M, Rink L. Influence of zinc deficiency and supplementation on NK cell cytotoxicity. Journal of Functional Foods. 2018;48:322–8.

    Article  CAS  Google Scholar 

  203. Almeida-Oliveira A, Smith-Carvalho M, Porto LC, Cardoso-Oliveira J, Ribeiro Ados S, Falcao RR, et al. Age-related changes in natural killer cell receptors from childhood through old age. Hum Immunol. 2011;72(4):319–29.

    Article  CAS  PubMed  Google Scholar 

  204. Sumati R, Sumati R, Christine W, Christine W, Christine CW, Nicolai W, et al. The Ig-related killer cell inhibitory receptor binds zinc and requires zinc for recognition of HLA-C on target cells. Journal of Immunology. 1995.

  205. Rajagopalan S, Long EO. Zinc bound to the killer cell-inhibitory receptor modulates the negative signal in human NK cells. J Immunol. 1998;161(3):1299–305.

    Article  CAS  PubMed  Google Scholar 

  206. Vales-Gomez M, Erskine RA, Deacon MP, Strominger JL, Reyburn HT. The role of zinc in the binding of killer cell Ig-like receptors to class I MHC proteins. Proc Natl Acad Sci U S A. 2001;98(4):1734–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Kumar S, Rajagopalan S, Sarkar P, Dorward DW, Peterson ME, Liao HS, et al. Zinc-Induced Polymerization of Killer-Cell Ig-like Receptor into Filaments Promotes Its Inhibitory Function at Cytotoxic Immunological Synapses. Mol Cell. 2016;62(1):21–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Mrakovcic-Sutic I, Radosevic-Stasic B, Simin M, Muhvic D, Rukavina D. Augmentation of NKT and NK cell-mediated cytotoxicity by peptidoglycan monomer linked with zinc. Mediators Inflamm. 2002;11(2):129–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Mariani E, Ravaglia G, Forti P, Meneghetti A, Tarozzi A, Maioli F, et al. Vitamin D, thyroid hormones and muscle mass influence natural killer (NK) innate immunity in healthy nonagenarians and centenarians. Clin Exp Immunol. 1999;116(1):19–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Mocchegiani E, Muzzioli M, Giacconi R, Cipriano C, Gasparini N, Franceschi C, et al. Metallothioneins/PARP-1/IL-6 interplay on natural killer cell activity in elderly: parallelism with nonagenarians and old infected humans. Effect of zinc supply. Mech Ageing Dev. 2003;124(4):459–68.

    Article  CAS  PubMed  Google Scholar 

  211. Mocchegiani E, Giacconi R, Cipriano C, Malavolta M. NK and NKT cells in aging and longevity: role of zinc and metallothioneins. J Clin Immunol. 2009;29(4):416–25.

    Article  CAS  PubMed  Google Scholar 

  212. Metz CH, Schroder AK, Overbeck S, Kahmann L, Plumakers B, Rink L. T-helper type 1 cytokine release is enhanced by in vitro zinc supplementation due to increased natural killer cells. Nutrition. 2007;23(2):157–63.

    Article  CAS  PubMed  Google Scholar 

  213. Solana R, Alonso MC, Pena J. Natural killer cells in healthy aging. Exp Gerontol. 1999;34(3):435–43.

    Article  CAS  PubMed  Google Scholar 

  214. Facchini A, Mariani E, Mariani AR, Papa S, Vitale M, Manzoli FA. Increased number of circulating Leu 11+ (CD 16) large granular lymphocytes and decreased NK activity during human ageing. Clin Exp Immunol. 1987;68(2):340–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  215. Sansoni P, Cossarizza A, Brianti V, Fagnoni F, Snelli G, Monti D, et al. Lymphocyte subsets and natural killer cell activity in healthy old people and centenarians [see comments]. 1993.

  216. Ravaglia G, Forti P, Maioli F, Bastagli L, Facchini A, Mariani E, et al. Effect of micronutrient status on natural killer cell immune function in healthy free-living subjects aged >/=90 y. Am J Clin Nutr. 2000;71(2):590–8.

    Article  CAS  PubMed  Google Scholar 

  217. Wong WT. Microglial aging in the healthy CNS: phenotypes, drivers, and rejuvenation. Front Cell Neurosci. 2013;7:22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Dubben S, Honscheid A, Winkler K, Rink L, Haase H. Cellular zinc homeostasis is a regulator in monocyte differentiation of HL-60 cells by 1 alpha,25-dihydroxyvitamin D3. J Leukoc Biol. 2010;87(5):833–44.

    Article  CAS  PubMed  Google Scholar 

  219. King LE, Fraker PJ. Zinc deficiency in mice alters myelopoiesis and hematopoiesis. J Nutr. 2002;132(11):3301–7.

    Article  CAS  PubMed  Google Scholar 

  220. Puissant-Lubrano B, Apoil PA, Guedj K, Congy-Jolivet N, Roubinet F, Guyonnet S, et al. Distinct effect of age, sex, and CMV seropositivity on dendritic cells and monocytes in human blood. Immunol Cell Biol. 2018;96(1):114–20.

    Article  CAS  PubMed  Google Scholar 

  221. Inomata M, Xu S, Chandra P, Meydani SN, Takemura G, Philips JA, et al. Macrophage LC3-associated phagocytosis is an immune defense against Streptococcus pneumoniae that diminishes with host aging. Proc Natl Acad Sci U S A. 2020;117(52):33561–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Linehan E, Dombrowski Y, Snoddy R, Fallon PG, Kissenpfennig A, Fitzgerald DC. Aging impairs peritoneal but not bone marrow-derived macrophage phagocytosis. Aging Cell. 2014;13(4):699–708.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Rymut N, Heinz J, Sadhu S, Hosseini Z, Riley CO, Marinello M, et al. Resolvin D1 promotes efferocytosis in aging by limiting senescent cell-induced MerTK cleavage. FASEB J. 2020;34(1):597–609.

    Article  CAS  PubMed  Google Scholar 

  224. Xie C, Wan L, Li C, Feng Y, Kang YJ. Selective suppression of M1 macrophages is involved in zinc inhibition of liver fibrosis in mice. J Nutr Biochem. 2021;97:108802.

    Article  CAS  PubMed  Google Scholar 

  225. Dierichs L, Kloubert V, Rink L. Cellular zinc homeostasis modulates polarization of THP-1-derived macrophages. Eur J Nutr. 2018;57(6):2161–9.

    Article  CAS  PubMed  Google Scholar 

  226. Minhas PS, Liu L, Moon PK, Joshi AU, Dove C, Mhatre S, et al. Macrophage de novo NAD(+) synthesis specifies immune function in aging and inflammation. Nat Immunol. 2019;20(1):50–63.

    Article  CAS  PubMed  Google Scholar 

  227. Zhong W, Rao Z, Xu J, Sun Y, Hu H, Wang P, et al. Defective mitophagy in aged macrophages promotes mitochondrial DNA cytosolic leakage to activate STING signaling during liver sterile inflammation. Aging Cell. 2022;21(6):e13622.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Connors J, Taramangalam B, Cusimano G, Bell MR, Matt SM, Runner K, et al. Aging alters antiviral signaling pathways resulting in functional impairment in innate immunity in response to pattern recognition receptor agonists. Geroscience. 2022;44(5):2555–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Kloubert V, Wessels I, Wolf J, Blaabjerg K, Janssens V, Hapala J, et al. Zinc deficiency leads to reduced interleukin-2 production by active gene silencing due to enhanced CREMalpha expression in T cells. Clin Nutr. 2021;40(5):3263–78.

    Article  CAS  PubMed  Google Scholar 

  230. Kim B, Kim HY, Yoon BR, Yeo J, In Jung J, Yu KS, et al. Cytoplasmic zinc promotes IL-1beta production by monocytes and macrophages through mTORC1-induced glycolysis in rheumatoid arthritis. Sci Signal. 2022;15(716):eabi7400.

    Article  CAS  PubMed  Google Scholar 

  231. Gou Y, Yang D, Tian T, Zhu X, Zhang R, Ren J, et al. The Transcription of ZIP9 is associated with the macrophage polarization and the pathogenesis of hepatocellular carcinoma. Front Immunol. 2022;13:725595.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Rossi DC, Figueroa JAL, Buesing WR, Candor K, Blancett LT, Evans HM, et al. A metabolic inhibitor arms macrophages to kill intracellular fungal pathogens by manipulating zinc homeostasis. J Clin Invest. 2021;131(16).

  233. Brazao V, Caetano LC, Del Vecchio Filipin M, Paula Alonso Toldo M, Caetano LN, do Prado JC, Jr. Zinc supplementation increases resistance to experimental infection by Trypanosoma cruzi. Vet Parasitol. 2008;154(1-2):32-7.

  234. Hamon R, Homan CC, Tran HB, Mukaro VR, Lester SE, Roscioli E, et al. Zinc and zinc transporters in macrophages and their roles in efferocytosis in COPD. PLoS One. 2014;9(10):e110056.

    Article  PubMed  PubMed Central  Google Scholar 

  235. Mehta AJ, Yeligar SM, Elon L, Brown LA, Guidot DM. Alcoholism causes alveolar macrophage zinc deficiency and immune dysfunction. Am J Respir Crit Care Med. 2013;188(6):716–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Mehta AJ, Joshi PC, Fan X, Brown LA, Ritzenthaler JD, Roman J, et al. Zinc supplementation restores PU.1 and Nrf2 nuclear binding in alveolar macrophages and improves redox balance and bacterial clearance in the lungs of alcohol-fed rats. Alcohol Clin Exp Res. 2011;35(8):1519–28.

    CAS  PubMed  PubMed Central  Google Scholar 

  237. Becker L, Nguyen L, Gill J, Kulkarni S, Pasricha PJ, Habtezion A. Age-dependent shift in macrophage polarisation causes inflammation-mediated degeneration of enteric nervous system. Gut. 2018;67(5):827–36.

    Article  CAS  PubMed  Google Scholar 

  238. Glesne D, Vogt S, Maser J, Legnini D, Huberman E. Regulatory properties and cellular redistribution of zinc during macrophage differentiation of human leukemia cells. J Struct Biol. 2006;155(1):2–11.

    Article  CAS  PubMed  Google Scholar 

  239. Mayer LS, Uciechowski P, Meyer S, Schwerdtle T, Rink L, Haase H. Differential impact of zinc deficiency on phagocytosis, oxidative burst, and production of pro-inflammatory cytokines by human monocytes. Metallomics. 2014;6(7):1288–95.

    Article  CAS  PubMed  Google Scholar 

  240. Srinivas U, Jeppsson B, Braconier JH. Superoxide production of peritoneal macrophages in experimental gram-negative sepsis; influence of in vitro and in vivo supplements of zinc. APMIS. 1989;97(8):682–8.

    Article  CAS  PubMed  Google Scholar 

  241. Subramanian Vignesh K, Landero Figueroa JA, Porollo A, Caruso JA, Deepe GS Jr. Granulocyte macrophage-colony stimulating factor induced Zn sequestration enhances macrophage superoxide and limits intracellular pathogen survival. Immunity. 2013;39(4):697–710.

    Article  CAS  PubMed  Google Scholar 

  242. Reiber C, Brieger A, Engelhardt G, Hebel S, Rink L, Haase H. Zinc chelation decreases IFN-beta-induced STAT1 upregulation and iNOS expression in RAW 264.7 macrophages. J Trace Elem Med Biol. 2017;44:76–82.

    Article  CAS  PubMed  Google Scholar 

  243. von Bulow V, Rink L, Haase H. Zinc-mediated inhibition of cyclic nucleotide phosphodiesterase activity and expression suppresses TNF-alpha and IL-1 beta production in monocytes by elevation of guanosine 3’,5’-cyclic monophosphate. J Immunol. 2005;175(7):4697–705.

    Article  Google Scholar 

  244. De Maeyer RPH, Chambers ES. The impact of ageing on monocytes and macrophages. Immunol Lett. 2021;230:1–10.

    Article  PubMed  Google Scholar 

  245. Katschinski DM, Neustock P, Kluter H, Kirchner H. Influence of various factors on interferon-alpha production in cultures of human leukocytes. J Interferon Res. 1994;14(3):105–10.

    Article  CAS  PubMed  Google Scholar 

  246. Kido T, Ishiwata K, Suka M, Yanagisawa H. Inflammatory response under zinc deficiency is exacerbated by dysfunction of the T helper type 2 lymphocyte–M2 macrophage pathway. Immunology. 2019;156(4):356–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  247. Jacinto TA, Meireles GS, Dias AT, Aires R, Porto ML, Gava AL, et al. Increased ROS production and DNA damage in monocytes are biomarkers of aging and atherosclerosis. Biol Res. 2018;51(1):33.

    Article  PubMed  PubMed Central  Google Scholar 

  248. Ghassemifar MR, Olsson MG, Agren MS, Franzen LE. Impaired function of postoperative macrophages from zinc-deficient rats decreases collagen contraction. Brief report APMIS. 1995;103(5):395–400.

    Article  CAS  PubMed  Google Scholar 

  249. Wirth JJ, Fraker PJ, Kierszenbaum F. Zinc requirement for macrophage function: effect of zinc deficiency on uptake and killing of a protozoan parasite. Immunology. 1989;68(1):114–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  250. Cabeza-Cabrerizo M, Cardoso A, Minutti CM. Pereira da Costa M, Reis e Sousa C. Dendritic Cells Revisited Annu Rev Immunol. 2021;39(1):131–66.

    Article  CAS  PubMed  Google Scholar 

  251. Balan S, Saxena M, Bhardwaj N. Dendritic cell subsets and locations. Int Rev Cell Mol Biol. 2019;348:1–68.

    Article  CAS  PubMed  Google Scholar 

  252. Canaday DH, Amponsah NA, Jones L, Tisch DJ, Hornick TR, Ramachandra L. Influenza-induced production of interferon-alpha is defective in geriatric individuals. J Clin Immunol. 2010;30(3):373–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  253. Wong C, Goldstein DR. Impact of aging on antigen presentation cell function of dendritic cells. Curr Opin Immunol. 2013;25(4):535–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  254. Splunter MV, Perdijk O, Fick-Brinkhof H, Floris-Vollenbroek EG, Meijer B, Brugman S, et al. Plasmacytoid dendritic cell and myeloid dendritic cell function in ageing: A comparison between elderly and young adult women. PLoS One. 2019;14(12):e0225825.

    Article  PubMed  PubMed Central  Google Scholar 

  255. Bashir H, Singh S, Singh RP, Agrewala JN, Kumar R. Age-mediated gut microbiota dysbiosis promotes the loss of dendritic cells tolerance. Aging Cell. 2023;22(6):e13838.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Pietschmann P, Hahn P, Kudlacek S, Thomas R, Peterlik M. Surface markers and transendothelial migration of dendritic cells from elderly subjects. Exp Gerontol. 2000;35(2):213–24.

    Article  CAS  PubMed  Google Scholar 

  257. Sridharan A, Esposo M, Kaushal K, Tay J, Osann K, Agrawal S, et al. Age-associated impaired plasmacytoid dendritic cell functions lead to decreased CD4 and CD8 T cell immunity. Age (Dordr). 2011;33(3):363–76.

    Article  CAS  PubMed  Google Scholar 

  258. Agrawal A, Agrawal S, Cao JN, Su H, Osann K, Gupta S. Altered innate immune functioning of dendritic cells in elderly humans: a role of phosphoinositide 3-kinase-signaling pathway. J Immunol. 2007;178(11):6912–22.

    Article  CAS  PubMed  Google Scholar 

  259. Bailey KL, Smith LM, Heires AJ, Katafiasz DM, Romberger DJ, LeVan TD. Aging leads to dysfunctional innate immune responses to TLR2 and TLR4 agonists. Aging Clin Exp Res. 2019;31(9):1185–93.

    Article  PubMed  Google Scholar 

  260. Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L, et al. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. J Immunol. 2010;184(5):2518–27.

    Article  CAS  PubMed  Google Scholar 

  261. Prakash S, Agrawal S, Vahed H, Ngyuen M, BenMohamed L, Gupta S, et al. Dendritic cells from aged subjects contribute to chronic airway inflammation by activating bronchial epithelial cells under steady state. Mucosal Immunol. 2014;7(6):1386–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. Agrawal A, Tay J, Ton S, Agrawal S, Gupta S. Increased reactivity of dendritic cells from aged subjects to self-antigen, the human DNA. J Immunol. 2009;182(2):1138–45.

    Article  CAS  PubMed  Google Scholar 

  263. George MM, Subramanian Vignesh K, Landero Figueroa JA, Caruso JA, Deepe GS Jr. Zinc Induces Dendritic Cell Tolerogenic Phenotype and Skews Regulatory T Cell-Th17 Balance. J Immunol. 2016;197(5):1864–76.

    Article  CAS  PubMed  Google Scholar 

  264. Rosenkranz E, Maywald M, Hilgers RD, Brieger A, Clarner T, Kipp M, et al. Induction of regulatory T cells in Th1-/Th17-driven experimental autoimmune encephalomyelitis by zinc administration. J Nutr Biochem. 2016;29:116–23.

    Article  CAS  PubMed  Google Scholar 

  265. Qiao H, Mei J, Yuan K, Zhang K, Zhou F, Tang T, et al. Immune-regulating strategy against rheumatoid arthritis by inducing tolerogenic dendritic cells with modified zinc peroxide nanoparticles. J Nanobiotechnology. 2022;20(1):323.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  266. Ranuh RG, Athiyyah AF, Darma A, Candra R, Setyoningrum RA, Endaryanto A, et al. ZINC SUPPLEMENTATION ALTERED BRONCHUS MUCOSAL IMMUNE

  267. STATUS EXPRESSED BY IFN-γ,. IL-6, DENDRITIC CELLS AND sIgA. Carpathian Journal of Food Science and Technology. 2019;11(5):67–72.

    Google Scholar 

  268. Ogawa Y, Kawamura T, Shimada S. Zinc and skin biology. Arch Biochem Biophys. 2016;611:113–9.

    Article  CAS  PubMed  Google Scholar 

  269. Kawamura T, Ogawa Y, Nakamura Y, Nakamizo S, Ohta Y, Nakano H, et al. Severe dermatitis with loss of epidermal Langerhans cells in human and mouse zinc deficiency. J Clin Invest. 2012;122(2):722–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Shumilina E, Xuan NT, Schmid E, Bhavsar SK, Szteyn K, Gu S, et al. Zinc induced apoptotic death of mouse dendritic cells. Apoptosis. 2010;15(10):1177–86.

    Article  CAS  PubMed  Google Scholar 

  271. Kitamura H, Morikawa H, Kamon H, Iguchi M, Hojyo S, Fukada T, et al. Toll-like receptor-mediated regulation of zinc homeostasis influences dendritic cell function. Nat Immunol. 2006;7(9):971–7.

    Article  CAS  PubMed  Google Scholar 

  272. Shaikh NA, Zhang XB, Abdalla MI, Baylink DJ, Tang X. Enhancing Human Treg Cell Induction through Engineered Dendritic Cells and Zinc Supplementation. Crit Rev Immunol. 2024;44(3):37–52.

    Article  PubMed  PubMed Central  Google Scholar 

  273. Steger MM, Maczek C, Grubeck-Loebenstein B. Morphologically and functionally intact dendritic cells can be derived from the peripheral blood of aged individuals. Clin Exp Immunol. 1996;105(3):544–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. John E, Laskow TC, Buchser WJ, Pitt BR, Basse PH, Butterfield LH, et al. Zinc in innate and adaptive tumor immunity. J Transl Med. 2010;8(1):118.

    Article  PubMed  PubMed Central  Google Scholar 

  275. Schulz AR, Malzer JN, Domingo C, Jurchott K, Grutzkau A, Babel N, et al. Low Thymic Activity and Dendritic Cell Numbers Are Associated with the Immune Response to Primary Viral Infection in Elderly Humans. J Immunol. 2015;195(10):4699–711.

    Article  CAS  PubMed  Google Scholar 

  276. Jing Y, Shaheen E, Drake RR, Chen N, Gravenstein S, Deng Y. Aging is associated with a numerical and functional decline in plasmacytoid dendritic cells, whereas myeloid dendritic cells are relatively unaltered in human peripheral blood. Hum Immunol. 2009;70(10):777–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  277. Orsini G, Legitimo A, Failli A, Massei F, Biver P, Consolini R. Enumeration of human peripheral blood dendritic cells throughout the life. Int Immunol. 2012;24(6):347–56.

    Article  CAS  PubMed  Google Scholar 

  278. Dvergsten CL, Johnson LA, Sandstead HH. Alterations in the postnatal development of the cerebellar cortex due to zinc deficiency. III. Impaired dendritic differentiation of basket and stellate cells. Brain Res. 1984;318(1):21–6.

    Article  CAS  PubMed  Google Scholar 

  279. Xu YP, Qi RQ, Chen W, Shi Y, Cui ZZ, Gao XH, et al. Aging affects epidermal Langerhans cell development and function and alters their miRNA gene expression profile. Aging (Albany NY). 2012;4(11):742–54.

    Article  CAS  PubMed  Google Scholar 

  280. Chambers ES, Vukmanovic-Stejic M. Skin barrier immunity and ageing. Immunology. 2020;160(2):116–25.

    Article  CAS  PubMed  Google Scholar 

  281. Hasegawa T, Feng Z, Yan Z, Ngo KH, Hosoi J, Demehri S. Reduction in human epidermal langerhans cells with age is associated with decline in CXCL14-Mediated Recruitment of CD14(+) Monocytes. J Invest Dermatol. 2020;140(7):1327–34.

    Article  CAS  PubMed  Google Scholar 

  282. Nakamura H, Sekiguchi A, Ogawa Y, Kawamura T, Akai R, Iwawaki T, et al. Zinc deficiency exacerbates pressure ulcers by increasing oxidative stress and ATP in the skin. J Dermatol Sci. 2019;95(2):62–9.

    Article  CAS  PubMed  Google Scholar 

  283. Sekiguchi A, Ishikawa O, Motegi SI. Case of zinc deficiency-induced dermatomyositis-like dermatitis: Association between absence of CD1a-positive Langerhans cells and development of dermatitis. J Dermatol. 2020;47(8):e286–8.

    Article  PubMed  Google Scholar 

  284. Talbot HK. Influenza in older adults. Infect Dis Clin North Am. 2017;31(4):757–66.

    Article  PubMed  Google Scholar 

  285. Cakman I, Rohwer J, Schutz RM, Kirchner H, Rink L. Dysregulation between TH1 and TH2 T cell subpopulations in the elderly. Mech Ageing Dev. 1996;87(3):197–209.

    Article  CAS  PubMed  Google Scholar 

  286. Chinn IK, Blackburn CC, Manley NR, Sempowski GD. Changes in primary lymphoid organs with aging. Semin Immunol. 2012;24(5):309–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  287. Palmer DB. The effect of age on thymic function. Front Immunol. 2013;4:316.

    Article  PubMed  PubMed Central  Google Scholar 

  288. Min H, Montecino-Rodriguez E, Dorshkind K. Reduction in the developmental potential of intrathymic T cell progenitors with age. J Immunol. 2004;173(1):245–50.

    Article  CAS  PubMed  Google Scholar 

  289. Dardenne M, Pleau JM. Interactions between zinc and thymulin. Met Based Drugs. 1994;1(2–3):233–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  290. Consolini R, Legitimo A, Calleri A, Milani M. Distribution of age-related thymulin titres in normal subjects through the course of life. Clin Exp Immunol. 2000;121(3):444–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  291. Mocchegiani E, Fabris N. Age-related thymus involution: zinc reverses in vitro the thymulin secretion defect. Int J Immunopharmacol. 1995;17(9):745–9.

    Article  CAS  PubMed  Google Scholar 

  292. King LE, Osati-Ashtiani F, Fraker PJ. Apoptosis plays a distinct role in the loss of precursor lymphocytes during zinc deficiency in mice. J Nutr. 2002;132(5):974–9.

    Article  CAS  PubMed  Google Scholar 

  293. Dardenne M, Pleau JM, Nabarra B, Lefrancier P, Derrien M, Choay J, et al. Contribution of zinc and other metals to the biological activity of the serum thymic factor. Proc Natl Acad Sci U S A. 1982;79(17):5370–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  294. Di Somma MM, Nuti S, Telford JL, Baldari CT. p56lck plays a key role in transducing apoptotic signals in T cells. FEBS Lett. 1995;363(1–2):101–4.

    Article  PubMed  Google Scholar 

  295. Taylor CG, Giesbrecht JA. Dietary zinc deficiency and expression of T lymphocyte signal transduction proteins. Can J Physiol Pharmacol. 2000;78(10):823–8.

    Article  CAS  PubMed  Google Scholar 

  296. Wong CP, Song Y, Elias VD, Magnusson KR, Ho E. Zinc supplementation increases zinc status and thymopoiesis in aged mice. J Nutr. 2009;139(7):1393–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  297. Fraker PJ, King LE. Reprogramming of the immune system during zinc deficiency. Annu Rev Nutr. 2004;24:277–98.

    Article  CAS  PubMed  Google Scholar 

  298. Prasad AS, Meftah S, Abdallah J, Kaplan J, Brewer GJ, Bach JF, et al. Serum thymulin in human zinc deficiency. J Clin Invest. 1988;82(4):1202–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  299. DiSilvestro RA, Dardenne M, Joseph E. Comparison of Thymulin Activity with Other Measures of Marginal Zinc Deficiency. Biol Trace Elem Res. 2021;199(2):585–7.

    Article  PubMed  Google Scholar 

  300. Chen Y, Ye Y, Krauss PL, Lowe P, Pfeiffenberger M, Damerau A, et al. Age-related increase of mitochondrial content in human memory CD4+ T cells contributes to ROS-mediated increased expression of proinflammatory cytokines. Front Immunol. 2022;13:911050.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  301. Henson SM, Lanna A, Riddell NE, Franzese O, Macaulay R, Griffiths SJ, et al. p38 signaling inhibits mTORC1-independent autophagy in senescent human CD8(+) T cells. J Clin Invest. 2014;124(9):4004–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  302. Akbar AN, Henson SM, Lanna A. Senescence of T Lymphocytes: Implications for Enhancing Human Immunity. Trends Immunol. 2016;37(12):866–76.

    Article  CAS  PubMed  Google Scholar 

  303. Ye J, Huang X, Hsueh EC, Zhang Q, Ma C, Zhang Y, et al. Human regulatory T cells induce T-lymphocyte senescence. Blood. 2012;120(10):2021–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  304. Barnett JB, Dao MC, Hamer DH, Kandel R, Brandeis G, Wu D, et al. Effect of zinc supplementation on serum zinc concentration and T cell proliferation in nursing home elderly: a randomized, double-blind, placebo-controlled trial. Am J Clin Nutr. 2016;103(3):942–51.

    Article  CAS  PubMed  Google Scholar 

  305. Liu MJ, Bao S, Galvez-Peralta M, Pyle CJ, Rudawsky AC, Pavlovicz RE, et al. ZIP8 regulates host defense through zinc-mediated inhibition of NF-kappaB. Cell Rep. 2013;3(2):386–400.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  306. Korichneva I, Hoyos B, Chua R, Levi E, Hammerling U. Zinc release from protein kinase C as the common event during activation by lipid second messenger or reactive oxygen. J Biol Chem. 2002;277(46):44327–31.

    Article  CAS  PubMed  Google Scholar 

  307. Pernelle JJ, Creuzet C, Loeb J, Gacon G. Phosphorylation of the lymphoid cell kinase p56lck is stimulated by micromolar concentrations of Zn2+. FEBS Lett. 1991;281(1–2):278–82.

    Article  CAS  PubMed  Google Scholar 

  308. Daaboul D, Rosenkranz E, Uciechowski P, Rink L. Repletion of zinc in zinc-deficient cells strongly up-regulates IL-1beta-induced IL-2 production in T-cells. Metallomics. 2012;4(10):1088–97.

    Article  CAS  PubMed  Google Scholar 

  309. Wong CP, Magnusson KR, Sharpton TJ, Ho E. Effects of zinc status on age-related T cell dysfunction and chronic inflammation. Biometals. 2021;34(2):291–301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  310. Baarz BR, Laurentius T, Wolf J, Wessels I, Bollheimer LC, Rink L. Short-term zinc supplementation of zinc-deficient seniors counteracts CREMalpha - mediated IL-2 suppression. Immun Ageing. 2022;19(1):40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  311. Nagel JE, Chopra RK, Chrest FJ, McCoy MT, Schneider EL, Holbrook NJ, et al. Decreased proliferation, interleukin 2 synthesis, and interleukin 2 receptor expression are accompanied by decreased mRNA expression in phytohemagglutinin-stimulated cells from elderly donors. J Clin Invest. 1988;81(4):1096–102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  312. Pieren DKJ, Smits NAM, Postel RJ, Kandiah V, de Wit J, van Beek J, et al. Co-Expression of TIGIT and Helios Marks Immunosenescent CD8(+) T Cells During Aging. Front Immunol. 2022;13:833531.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  313. Wang D, Du J, Song Y, Wang B, Song R, Hao Y, et al. CD70 contributes to age-associated T cell defects and overwhelming inflammatory responses. Aging (Albany NY). 2020;12(12):12032–50.

    Article  CAS  PubMed  Google Scholar 

  314. Phelouzat MA, Laforge T, Arbogast A, Quadri RA, Boutet S, Proust JJ. Susceptibility to apoptosis of T lymphocytes from elderly humans is associated with increased in vivo expression of functional Fas receptors. Mech Ageing Dev. 1997;96(1–3):35–46.

    Article  CAS  PubMed  Google Scholar 

  315. Heigele A, Joas S, Regensburger K, Kirchhoff F. Increased susceptibility of CD4+ T cells from elderly individuals to HIV-1 infection and apoptosis is associated with reduced CD4 and enhanced CXCR4 and FAS surface expression levels. Retrovirology. 2015;12(1):86.

    Article  PubMed  PubMed Central  Google Scholar 

  316. Honscheid A, Rink L, Haase H. T-lymphocytes: a target for stimulatory and inhibitory effects of zinc ions. Endocr Metab Immune Disord Drug Targets. 2009;9(2):132–44.

    Article  PubMed  Google Scholar 

  317. Hawkley LC, Cacioppo JT. Stress and the aging immune system. Brain Behav Immun. 2004;18(2):114–9.

    Article  CAS  PubMed  Google Scholar 

  318. Bao B, Prasad AS, Beck FW, Bao GW, Singh T, Ali S, et al. Intracellular free zinc up-regulates IFN-gamma and T-bet essential for Th1 differentiation in Con-A stimulated HUT-78 cells. Biochem Biophys Res Commun. 2011;407(4):703–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  319. Prasad AS. Effects of zinc deficiency on Th1 and Th2 cytokine shifts. J Infect Dis. 2000;182(Suppl 1):S62–8.

    Article  CAS  PubMed  Google Scholar 

  320. Fortes C, Forastiere F, Agabiti N, Fano V, Pacifici R, Virgili F, et al. The effect of zinc and vitamin A supplementation on immune response in an older population. J Am Geriatr Soc. 1998;46(1):19–26.

    Article  CAS  PubMed  Google Scholar 

  321. Liu W, Jakobs J, Rink L. Proton-Pump Inhibitors Suppress T Cell Response by Shifting Intracellular Zinc Distribution. Int J Mol Sci. 2023;24(2).

  322. Rocamora-Reverte L, Melzer FL, Wurzner R, Weinberger B. The Complex role of regulatory T cells in immunity and aging. Front Immunol. 2020;11:616949.

    Article  CAS  PubMed  Google Scholar 

  323. Schmitt V, Rink L, Uciechowski P. The Th17/Treg balance is disturbed during aging. Exp Gerontol. 2013;48(12):1379–86.

    Article  CAS  PubMed  Google Scholar 

  324. Kulik L, Maywald M, Kloubert V, Wessels I, Rink L. Zinc deficiency drives Th17 polarization and promotes loss of Treg cell function. J Nutr Biochem. 2019;63:11–8.

    Article  CAS  PubMed  Google Scholar 

  325. Rosenkranz E, Metz CH, Maywald M, Hilgers RD, Wessels I, Senff T, et al. Zinc supplementation induces regulatory T cells by inhibition of Sirt-1 deacetylase in mixed lymphocyte cultures. Mol Nutr Food Res. 2016;60(3):661–71.

    Article  CAS  PubMed  Google Scholar 

  326. Maywald M, Meurer SK, Weiskirchen R, Rink L. Zinc supplementation augments TGF-beta1-dependent regulatory T cell induction. Mol Nutr Food Res. 2017;61(3).

  327. Rosenkranz E, Hilgers RD, Uciechowski P, Petersen A, Plumakers B, Rink L. Zinc enhances the number of regulatory T cells in allergen-stimulated cells from atopic subjects. Eur J Nutr. 2017;56(2):557–67.

    Article  CAS  PubMed  Google Scholar 

  328. Schmitt AK, Puppa MA, Wessels I, Rink L. Vitamin D3 and zinc synergistically induce regulatory T cells and suppress interferon-gamma production in mixed lymphocyte culture. J Nutr Biochem. 2022;102:108942.

    Article  CAS  PubMed  Google Scholar 

  329. Awasthi A, Narayan S, Dalal R, Rizvi ZA. Zinc dampens anti-tumour immunity by promoting Foxp3+ regulatory T cells. 2023.

  330. Lelliott EJ, Naddaf J, Ganio K, Michie J, Wang S, Liu L, et al. Intracellular zinc protects tumours from T cell-mediated cytotoxicity. Cell Death Differ. 2024.

  331. Stoye D, Schubert C, Goihl A, Guttek K, Reinhold A, Brocke S, et al. Zinc aspartate suppresses T cell activation in vitro and relapsing experimental autoimmune encephalomyelitis in SJL/J mice. Biometals. 2012;25(3):529–39.

    Article  CAS  PubMed  Google Scholar 

  332. Kitabayashi C, Fukada T, Kanamoto M, Ohashi W, Hojyo S, Atsumi T, et al. Zinc suppresses Th17 development via inhibition of STAT3 activation. Int Immunol. 2010;22(5):375–86.

    Article  CAS  PubMed  Google Scholar 

  333. Lee H, Kim B, Choi YH, Hwang Y, Kim DH, Cho S, et al. Inhibition of interleukin-1beta-mediated interleukin-1 receptor-associated kinase 4 phosphorylation by zinc leads to repression of memory T helper type 17 response in humans. Immunology. 2015;146(4):645–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  334. Guzel N, Rink L, Fischer HJ. Zinc modulates glutamine metabolism in T Cells. Mol Nutr Food Res. 2023;67(20):e2300155.

    Article  PubMed  Google Scholar 

  335. Golden MH, Jackson AA, Golden BE. Effect of zinc on thymus of recently malnourished children. Lancet. 1977;2(8047):1057–9.

    Article  CAS  PubMed  Google Scholar 

  336. Nodera M, Yanagisawa H, Wada O. Increased apoptosis in a variety of tissues of zinc-deficient rats. Life Sci. 2001;69(14):1639–49.

    Article  CAS  PubMed  Google Scholar 

  337. Ferrando-Martinez S, Ruiz-Mateos E, Hernandez A, Gutierrez E, Rodriguez-Mendez Mdel M, Ordonez A, et al. Age-related deregulation of naive T cell homeostasis in elderly humans. Age (Dordr). 2011;33(2):197–207.

    Article  CAS  PubMed  Google Scholar 

  338. Pfister G, Weiskopf D, Lazuardi L, Kovaiou RD, Cioca DP, Keller M, et al. Naive T cells in the elderly: are they still there? Ann N Y Acad Sci. 2006;1067(1):152–7.

    Article  CAS  PubMed  Google Scholar 

  339. Derhovanessian E, Maier AB, Beck R, Jahn G, Hahnel K, Slagboom PE, et al. Hallmark features of immunosenescence are absent in familial longevity. J Immunol. 2010;185(8):4618–24.

    Article  CAS  PubMed  Google Scholar 

  340. Li M, Yao D, Zeng X, Kasakovski D, Zhang Y, Chen S, et al. Age related human T cell subset evolution and senescence. Immun Ageing. 2019;16:24.

    Article  PubMed  PubMed Central  Google Scholar 

  341. Prasad AS, Beck FW, Grabowski SM, Kaplan J, Mathog RH. Zinc deficiency: changes in cytokine production and T-cell subpopulations in patients with head and neck cancer and in noncancer subjects. Proc Assoc Am Physicians. 1997;109(1):68–77.

    CAS  PubMed  Google Scholar 

  342. Le Page A, Dupuis G, Larbi A, Witkowski JM, Fulop T. Signal transduction changes in CD4(+) and CD8(+) T cell subpopulations with aging. Exp Gerontol. 2018;105:128–39.

    Article  PubMed  Google Scholar 

  343. Elyahu Y, Hekselman I, Eizenberg-Magar I, Berner O, Strominger I, Schiller M, et al. Aging promotes reorganization of the CD4 T cell landscape toward extreme regulatory and effector phenotypes. Sci Adv. 2019;5(8):eaaw8330.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  344. Beck FW, Prasad AS, Kaplan J, Fitzgerald JT, Brewer GJ. Changes in cytokine production and T cell subpopulations in experimentally induced zinc-deficient humans. Am J Physiol. 1997;272(6 Pt 1):E1002–7.

    CAS  PubMed  Google Scholar 

  345. Prasad AS. Zinc: Mechanisms of Host Defense1. J Nutr. 2007;137(5):1345–9.

    Article  CAS  PubMed  Google Scholar 

  346. Tavares SM, Junior Wde L, Lopes ESMR. Normal lymphocyte immunophenotype in an elderly population. Rev Bras Hematol Hemoter. 2014;36(3):180–3.

    Article  PubMed  PubMed Central  Google Scholar 

  347. Ferguson FG, Wikby A, Maxson P, Olsson J, Johansson B. Immune parameters in a longitudinal study of a very old population of Swedish people: a comparison between survivors and nonsurvivors. J Gerontol A Biol Sci Med Sci. 1995;50(6):B378–82.

    Article  CAS  PubMed  Google Scholar 

  348. Kline GH, Hayden TA, Klinman NR. B cell maintenance in aged mice reflects both increased B cell longevity and decreased B cell generation. J Immunol. 1999;162(6):3342–9.

    Article  CAS  PubMed  Google Scholar 

  349. Wols HA, Johnson KM, Ippolito JA, Birjandi SZ, Su Y, Le PT, et al. Migration of immature and mature B cells in the aged microenvironment. Immunology. 2010;129(2):278–90.

    Article  PubMed  Google Scholar 

  350. Johnson KM, Owen K, Witte PL. Aging and developmental transitions in the B cell lineage. Int Immunol. 2002;14(11):1313–23.

    Article  CAS  PubMed  Google Scholar 

  351. Zharhary D. Age-related changes in the capability of the bone marrow to generate B cells. J Immunol. 1988;141(6):1863–9.

    Article  CAS  PubMed  Google Scholar 

  352. Cho RH, Sieburg HB, Muller-Sieburg CE. A new mechanism for the aging of hematopoietic stem cells: aging changes the clonal composition of the stem cell compartment but not individual stem cells. Blood. 2008;111(12):5553–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  353. Guerrettaz LM, Johnson SA, Cambier JC. Acquired hematopoietic stem cell defects determine B-cell repertoire changes associated with aging. Proc Natl Acad Sci U S A. 2008;105(33):11898–902.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  354. McKenna RW, Washington LT, Aquino DB, Picker LJ, Kroft SH. Immunophenotypic analysis of hematogones (B-lymphocyte precursors) in 662 consecutive bone marrow specimens by 4-color flow cytometry. Blood. 2001;98(8):2498–507.

    Article  CAS  PubMed  Google Scholar 

  355. Rossi MI, Yokota T, Medina KL, Garrett KP, Comp PC, Schipul AH Jr, et al. B lymphopoiesis is active throughout human life, but there are developmental age-related changes. Blood. 2003;101(2):576–84.

    Article  CAS  PubMed  Google Scholar 

  356. Stephan RP, Lill-Elghanian DA, Witte PL. Development of B cells in aged mice: decline in the ability of pro-B cells to respond to IL-7 but not to other growth factors. J Immunol. 1997;158(4):1598–609.

    Article  CAS  PubMed  Google Scholar 

  357. Stephan RP, Reilly CR, Witte PL. Impaired ability of bone marrow stromal cells to support B-lymphopoiesis with age. Blood. 1998;91(1):75–88.

    Article  CAS  PubMed  Google Scholar 

  358. Ciocca M, Zaffina S, Fernandez Salinas A, Bocci C, Palomba P, Conti MG, et al. Evolution of human memory B cells from childhood to old age. Front Immunol. 2021;12:690534.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  359. Colonna-Romano G, Buffa S, Bulati M, Candore G, Lio D, Pellicano M, et al. B cells compartment in centenarian offspring and old people. Curr Pharm Des. 2010;16(6):604–8.

    Article  CAS  PubMed  Google Scholar 

  360. Wei C, Anolik J, Cappione A, Zheng B, Pugh-Bernard A, Brooks J, et al. A new population of cells lacking expression of CD27 represents a notable component of the B cell memory compartment in systemic lupus erythematosus. J Immunol. 2007;178(10):6624–33.

    Article  CAS  PubMed  Google Scholar 

  361. Moir S, Ho J, Malaspina A, Wang W, DiPoto AC, O’Shea MA, et al. Evidence for HIV-associated B cell exhaustion in a dysfunctional memory B cell compartment in HIV-infected viremic individuals. J Exp Med. 2008;205(8):1797–805.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  362. Martin V, Wu YC, Kipling D, Dunn-Walters DK. Age-related aspects of human IgM(+) B cell heterogeneity. Ann N Y Acad Sci. 2015;1362(1):153–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  363. Marrie TJ. Community-acquired pneumonia in the elderly. Clin Infect Dis. 2000;31(4):1066–78.

    Article  CAS  PubMed  Google Scholar 

  364. Shi Y, Yamazaki T, Okubo Y, Uehara Y, Sugane K, Agematsu K. Regulation of aged humoral immune defense against pneumococcal bacteria by IgM memory B cell. J Immunol. 2005;175(5):3262–7.

    Article  CAS  PubMed  Google Scholar 

  365. Moberley SA, Holden J, Tatham DP, Andrews RM. Vaccines for preventing pneumococcal infection in adults. Cochrane Database Syst Rev. 2008;1:CD000422.

    Google Scholar 

  366. Park S, Nahm MH. Older adults have a low capacity to opsonize pneumococci due to low IgM antibody response to pneumococcal vaccinations. Infect Immun. 2011;79(1):314–20.

    Article  CAS  PubMed  Google Scholar 

  367. Kolibab K, Smithson SL, Rabquer B, Khuder S, Westerink MA. Immune response to pneumococcal polysaccharides 4 and 14 in elderly and young adults: analysis of the variable heavy chain repertoire. Infect Immun. 2005;73(11):7465–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  368. Frasca D, Diaz A, Romero M, Ferracci F, Blomberg BB. MicroRNAs miR-155 and miR-16 Decrease AID and E47 in B Cells from Elderly Individuals. J Immunol. 2015;195(5):2134–40.

    Article  CAS  PubMed  Google Scholar 

  369. Muramatsu M, Sankaranand VS, Anant S, Sugai M, Kinoshita K, Davidson NO, et al. Specific expression of activation-induced cytidine deaminase (AID), a novel member of the RNA-editing deaminase family in germinal center B cells. J Biol Chem. 1999;274(26):18470–6.

    Article  CAS  PubMed  Google Scholar 

  370. Banerjee M, Mehr R, Belelovsky A, Spencer J, Dunn-Walters DK. Age- and tissue-specific differences in human germinal center B cell selection revealed by analysis of IgVH gene hypermutation and lineage trees. Eur J Immunol. 2002;32(7):1947–57.

    Article  CAS  PubMed  Google Scholar 

  371. Chong Y, Ikematsu H, Yamaji K, Nishimura M, Kashiwagi S, Hayashi J. Age-related accumulation of Ig V(H) gene somatic mutations in peripheral B cells from aged humans. Clin Exp Immunol. 2003;133(1):59–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  372. Frasca D, Diaz A, Romero M, Blomberg BB. The generation of memory B cells is maintained, but the antibody response is not, in the elderly after repeated influenza immunizations. Vaccine. 2016;34(25):2834–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  373. Manz RA, Thiel A, Radbruch A. Lifetime of plasma cells in the bone marrow. Nature. 1997;388(6638):133–4.

    Article  CAS  PubMed  Google Scholar 

  374. Schenkein JG, Park S, Nahm MH. Pneumococcal vaccination in older adults induces antibodies with low opsonic capacity and reduced antibody potency. Vaccine. 2008;26(43):5521–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  375. Andersen-Ranberg K, M HO-M, Wiik A, Jeune B, Hegedus L. High prevalence of autoantibodies among Danish centenarians. Clin Exp Immunol. 2004;138(1):158-63.

  376. King LE, Frentzel JW, Mann JJ, Fraker PJ. Chronic zinc deficiency in mice disrupted T cell lymphopoiesis and erythropoiesis while B cell lymphopoiesis and myelopoiesis were maintained. J Am Coll Nutr. 2005;24(6):494–502.

    Article  CAS  PubMed  Google Scholar 

  377. Fraker PJ, Osati-Ashtiani F, Wagner MA, King LE. Possible roles for glucocorticoids and apoptosis in the suppression of lymphopoiesis during zinc deficiency: a review. J Am Coll Nutr. 1995;14(1):11–7.

    Article  CAS  PubMed  Google Scholar 

  378. Hojyo S, Miyai T, Fujishiro H, Kawamura M, Yasuda T, Hijikata A, et al. Zinc transporter SLC39A10/ZIP10 controls humoral immunity by modulating B-cell receptor signal strength. Proc Natl Acad Sci U S A. 2014;111(32):11786–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  379. Miyai T, Hojyo S, Ikawa T, Kawamura M, Irie T, Ogura H, et al. Zinc transporter SLC39A10/ZIP10 facilitates antiapoptotic signaling during early B-cell development. Proc Natl Acad Sci U S A. 2014;111(32):11780–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  380. Klecha AJ, Salgueiro J, Wald M, Boccio J, Zubillaga M, Leonardi NM, et al. In vivo iron and zinc deficiency diminished T- and B-selective mitogen stimulation of murine lymphoid cells through protein kinase C-mediated mechanism. Biol Trace Elem Res. 2005;104(2):173–83.

    Article  CAS  PubMed  Google Scholar 

  381. Gruber K, Maywald M, Rosenkranz E, Haase H, Plumakers B, Rink L. Zinc deficiency adversely influences interleukin-4 and interleukin-6 signaling. J Biol Regul Homeost Agents. 2013;27(3):661–71.

    CAS  PubMed  Google Scholar 

  382. Puppa MA, Bennstein SB, Fischer HJ, Rink L. Zinc deficiency impairs the development of human regulatory B cells from purified B cells. J Trace Elem Med Biol. 2024;86:127556.

    Article  CAS  PubMed  Google Scholar 

  383. Hamza SA, Mousa SM, Taha SE, Adel LA, Samaha HE, Hussein DA. Immune response of 23-valent pneumococcal polysaccharide vaccinated elderly and its relation to frailty indices, nutritional status, and serum zinc levels. Geriatr Gerontol Int. 2012;12(2):223–9.

    Article  PubMed  Google Scholar 

  384. Kreft B, Fischer A, Krüger S, Sack K, Kirchner H, Rink L. The impaired immune response to diphtheria vaccinationin elderly chronic hemodialysis patients is related to zinc deficiency. Biogerontology. 2000;1:61–6.

    Article  CAS  PubMed  Google Scholar 

  385. Zhao N, Wang X, Zhang Y, Gu Q, Huang F, Zheng W, et al. Gestational zinc deficiency impairs humoral and cellular immune responses to hepatitis B vaccination in offspring mice. PLoS ONE. 2013;8(9):e73461.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  386. Albert MJ, Qadri F, Wahed MA, Ahmed T, Rahman AS, Ahmed F, et al. Supplementation with zinc, but not vitamin A, improves seroconversion to vibriocidal antibody in children given an oral cholera vaccine. J Infect Dis. 2003;187(6):909–13.

    Article  CAS  PubMed  Google Scholar 

  387. Sundaram ME, Meydani SN, Vandermause M, Shay DK, Coleman LA. Vitamin E, vitamin A, and zinc status are not related to serologic response to influenza vaccine in older adults: an observational prospective cohort study. Nutr Res. 2014;34(2):149–54.

    Article  CAS  PubMed  Google Scholar 

  388. Braga CB, Santos IK, Palmeira P, Peria FM, Ribeiro SM, Martinez EZ, et al. Effect of Zinc Supplementation on Serological Response to Vaccination Against Streptococcus Pneumoniae in Patients Undergoing Chemotherapy for Colorectal Cancer. Nutr Cancer. 2015;67(6):926–32.

    Article  CAS  PubMed  Google Scholar 

  389. Chillon TS, Maares M, Demircan K, Hackler J, Sun Q, Heller RA, et al. Serum Free Zinc is associated with vaccination response to SARS-CoV-2. Front Immunol. 2022;13:906551.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  390. Skowronski DM, Tweed SA, De Serres G. Rapid decline of influenza vaccine-induced antibody in the elderly: is it real, or is it relevant? J Infect Dis. 2008;197(4):490–502.

    Article  PubMed  Google Scholar 

  391. Ollig J, Kloubert V, Taylor KM, Rink L. B cell activation and proliferation increase intracellular zinc levels. J Nutr Biochem. 2019;64:72–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  392. Frasca D, Diaz A, Romero M, Landin AM, Blomberg BB. Age effects on B cells and humoral immunity in humans. Ageing Res Rev. 2011;10(3):330–5.

    Article  CAS  PubMed  Google Scholar 

  393. Ibs KH, Rink L. Zinc-altered immune function. J Nutr. 2003;133(5 Suppl 1):1452S-S1456.

    Article  CAS  PubMed  Google Scholar 

  394. Frasca D, Blomberg BB. Aging induces B cell defects and decreased antibody responses to influenza infection and vaccination. Immun Ageing. 2020;17(1):37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  395. Ratliff M, Alter S, Frasca D, Blomberg BB, Riley RL. In senescence, age-associated B cells secrete TNFalpha and inhibit survival of B-cell precursors. Aging Cell. 2013;12(2):303–11.

    Article  CAS  PubMed  Google Scholar 

  396. de Mol J, Kuiper J, Tsiantoulas D, Foks AC. The dynamics of B cell aging in health and disease. Front Immunol. 2021;12:733566.

    Article  PubMed  PubMed Central  Google Scholar 

  397. Frasca D, Landin AM, Lechner SC, Ryan JG, Schwartz R, Riley RL, et al. Aging down-regulates the transcription factor E2A, activation-induced cytidine deaminase, and Ig class switch in human B cells. J Immunol. 2008;180(8):5283–90.

    Article  CAS  PubMed  Google Scholar 

  398. Conticello SG, Langlois MA, Yang Z, Neuberger MS. DNA Deamination in Immunity: AID in the Context of Its APOBEC Relatives. Advances in Immunology. 94: Academic Press; 2007. p. 37–73.

  399. Howard WA, Gibson KL, Dunn-Walters DK. Antibody quality in old age. Rejuvenation Res. 2006;9(1):117–25.

    Article  CAS  PubMed  Google Scholar 

  400. Rink L, Seyfarth M. Characteristics of immunologic test values in the elderly. Z Gerontol Geriatr. 1997;30(3):220–5.

    CAS  PubMed  Google Scholar 

  401. ListÌ F, Candore G, Modica MA, Russo M, Lorenzo GD, Esposito-Pellitteri M, et al. A Study of Serum Immunoglobulin Levels in Elderly Persons That Provides New Insights into B Cell Immunosenescence. Ann N Y Acad Sci. 2006;1089(1):487–95.

    Article  PubMed  Google Scholar 

  402. Frasca D, Diaz A, Romero M, Mendez NV, Landin AM, Blomberg BB. Effects of age on H1N1-specific serum IgG1 and IgG3 levels evaluated during the 2011–2012 influenza vaccine season. Immun Ageing. 2013;10(1):14.

    Article  PubMed  PubMed Central  Google Scholar 

  403. Xiang D, Hu S, Mai T, Zhang X, Zhang L, Wang S, et al. Worldwide cancer statistics of adults over 75 years old in 2019: a systematic analysis of the global burden of disease study 2019. BMC Public Health. 2022;22(1):1979.

    Article  PubMed  PubMed Central  Google Scholar 

  404. Pechal A, Lin K, Allen S, Reveles K. National age group trends in Clostridium difficile infection incidence and health outcomes in United States Community Hospitals. BMC Infect Dis. 2016;16(1):682.

    Article  PubMed  PubMed Central  Google Scholar 

  405. : Center for Disease Control and Prevention; [Available from: https://data.cdc.gov/NCHS/Provisional-COVID-19-Deaths-by-Sex-and-Age/9bhg-hcku/about_data.

  406. Jothimani D, Kailasam E, Danielraj S, Nallathambi B, Ramachandran H, Sekar P, et al. COVID-19: Poor outcomes in patients with zinc deficiency. Int J Infect Dis. 2020;100:343–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  407. Gonçalves TJM, Gonçalves S, Guarnieri A, Risegato RC, Guimarães MP, de Freitas DC, et al. Association Between Low Zinc Levels and Severity of Acute Respiratory Distress Syndrome by New Coronavirus SARS-CoV-2. Nutr Clin Pract. 2021;36(1):186–91.

    Article  PubMed  Google Scholar 

  408. Vogel-González M, Talló-Parra M, Herrera-Fernández V, Pérez-Vilaró G, Chillón M, Nogués X, et al. Low Zinc Levels at Admission Associates with Poor Clinical Outcomes in SARS-CoV-2 Infection. Nutrients. 2021;13(2).

  409. Wessels I, Rolles B, Slusarenko AJ, Rink L. Zinc deficiency as a possible risk factor for increased susceptibility and severe progression of Corona Virus Disease 19. Br J Nutr. 2022;127(2):214–32.

    Article  CAS  PubMed  Google Scholar 

  410. Li JQ, Welchowski T, Schmid M, Mauschitz MM, Holz FG, Finger RP. Prevalence and incidence of age-related macular degeneration in Europe: a systematic review and meta-analysis. Br J Ophthalmol. 2020;104(8):1077–84.

    Article  PubMed  Google Scholar 

  411. Al-Zamil WM, Yassin SA. Recent developments in age-related macular degeneration: a review. Clin Interv Aging. 2017;12:1313–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  412. Seah I, Zhao X, Lin Q, Liu Z, Su SZZ, Yuen YS, et al. Use of biomaterials for sustained delivery of anti-VEGF to treat retinal diseases. Eye (Lond). 2020;34(8):1341–56.

    Article  PubMed  Google Scholar 

  413. Karkhaneh R, Faghihi H, Riazi-Esfahani H, Abrishami M, Bazvand F, Ebrahimiadib N, et al. Evaluating the Efficacy and Safety of Aflibercept Biosimilar (P041) Compared with Originator Product in Patients with Neovascular Age-Related Macular Degeneration. Ophthalmol Retina. 2024;8(8):744–53.

    Article  PubMed  Google Scholar 

  414. Comparison of Age-related Macular Degeneration Treatments Trials Research G, Maguire MG, Martin DF, Ying GS, Jaffe GJ, Daniel E, et al. Five-Year Outcomes with Anti-Vascular Endothelial Growth Factor Treatment of Neovascular Age-Related Macular Degeneration: The Comparison of Age-Related Macular Degeneration Treatments Trials. Ophthalmology. 2016;123(8):1751-61.

  415. Gil-Martinez M, Santos-Ramos P, Fernandez-Rodriguez M, Abraldes MJ, Rodriguez-Cid MJ, Santiago-Varela M, et al. Pharmacological Advances in the Treatment of Age-related Macular Degeneration. Curr Med Chem. 2020;27(4):583–98.

    Article  CAS  PubMed  Google Scholar 

  416. Age-Related Eye Disease Study Research G. Risk factors associated with age-related macular degeneration. A case-control study in the age-related eye disease study: Age-Related Eye Disease Study Report Number 3. Ophthalmology. 2000;107(12):2224–32.

  417. Hageman GS, Anderson DH, Johnson LV, Hancox LS, Taiber AJ, Hardisty LI, et al. A common haplotype in the complement regulatory gene factor H (HF1/CFH) predisposes individuals to age-related macular degeneration. Proc Natl Acad Sci U S A. 2005;102(20):7227–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  418. Katta S, Kaur I, Chakrabarti S. The molecular genetic basis of age-related macular degeneration: an overview. J Genet. 2009;88(4):425–49.

    Article  CAS  PubMed  Google Scholar 

  419. Thakkinstian A, Han P, McEvoy M, Smith W, Hoh J, Magnusson K, et al. Systematic review and meta-analysis of the association between complement factor H Y402H polymorphisms and age-related macular degeneration. Hum Mol Genet. 2006;15(18):2784–90.

    Article  CAS  PubMed  Google Scholar 

  420. Nozaki M, Raisler BJ, Sakurai E, Sarma JV, Barnum SR, Lambris JD, et al. Drusen complement components C3a and C5a promote choroidal neovascularization. Proc Natl Acad Sci U S A. 2006;103(7):2328–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  421. Davoudi S, Sobrin L. Epidemiology and Risk Factors in Age-Related Macular Degeneration (AMD). In: Albert DM, Miller JW, Azar DT, Young LH, editors. Albert and Jakobiec’s Principles and Practice of Ophthalmology. Cham: Springer International Publishing; 2022. p. 3833–55.

    Chapter  Google Scholar 

  422. Golestaneh N, Chu Y, Xiao YY, Stoleru GL, Theos AC. Dysfunctional autophagy in RPE, a contributing factor in age-related macular degeneration. Cell Death Dis. 2017;8(1):e2537.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  423. Tate DJ Jr, Miceli MV, Newsome DA. Zinc protects against oxidative damage in cultured human retinal pigment epithelial cells. Free Radic Biol Med. 1999;26(5–6):704–13.

    Article  CAS  PubMed  Google Scholar 

  424. Ha KN, Chen Y, Cai J, Sternberg P Jr. Increased glutathione synthesis through an ARE-Nrf2-dependent pathway by zinc in the RPE: implication for protection against oxidative stress. Invest Ophthalmol Vis Sci. 2006;47(6):2709–15.

    Article  PubMed  Google Scholar 

  425. Wills NK, Ramanujam VM, Kalariya N, Lewis JR, van Kuijk FJ. Copper and zinc distribution in the human retina: relationship to cadmium accumulation, age, and gender. Exp Eye Res. 2008;87(2):80–8.

    Article  CAS  PubMed  Google Scholar 

  426. Akagi T, Kaneda M, Ishii K, Hashikawa T. Differential subcellular localization of zinc in the rat retina. J Histochem Cytochem. 2001;49(1):87–96.

    Article  CAS  PubMed  Google Scholar 

  427. Kaneda M, Ishii K, Akagi T, Tatsukawa T, Hashikawa T. Endogenous zinc can be a modulator of glycinergic signaling pathway in the rat retina. J Mol Histol. 2005;36(3):179–85.

    Article  CAS  PubMed  Google Scholar 

  428. Lengyel I, Flinn JM, Peto T, Linkous DH, Cano K, Bird AC, et al. High concentration of zinc in sub-retinal pigment epithelial deposits. Exp Eye Res. 2007;84(4):772–80.

    Article  CAS  PubMed  Google Scholar 

  429. Nan R, Gor J, Lengyel I, Perkins SJ. Uncontrolled zinc- and copper-induced oligomerisation of the human complement regulator factor H and its possible implications for function and disease. J Mol Biol. 2008;384(5):1341–52.

    Article  CAS  PubMed  Google Scholar 

  430. Chew EY, Clemons TE, Agron E, Sperduto RD, Sangiovanni JP, Kurinij N, et al. Long-term effects of vitamins C and E, beta-carotene, and zinc on age-related macular degeneration: AREDS report no. 35. Ophthalmology. 2013;120(8):1604–11 e4.

  431. Newsome DA. A randomized, prospective, placebo-controlled clinical trial of a novel zinc-monocysteine compound in age-related macular degeneration. Curr Eye Res. 2008;33(7):591–8.

    Article  CAS  PubMed  Google Scholar 

  432. Wong P, Markey M, Rapp CM, Darrow RM, Ziesel A, Organisciak DT. Enhancing the efficacy of AREDS antioxidants in light-induced retinal degeneration. Mol Vis. 2017;23:718–39.

    CAS  PubMed  PubMed Central  Google Scholar 

  433. Gopinath B, Liew G, Russell J, Cosatto V, Burlutsky G, Mitchell P. Intake of key micronutrients and food groups in patients with late-stage age-related macular degeneration compared with age-sex-matched controls. Br J Ophthalmol. 2017;101(8):1027–31.

    Article  PubMed  Google Scholar 

  434. Tan JS, Wang JJ, Flood V, Rochtchina E, Smith W, Mitchell P. Dietary antioxidants and the long-term incidence of age-related macular degeneration: the Blue Mountains Eye Study. Ophthalmology. 2008;115(2):334–41.

    Article  PubMed  Google Scholar 

  435. Ho L, van Leeuwen R, Witteman JC, van Duijn CM, Uitterlinden AG, Hofman A, et al. Reducing the genetic risk of age-related macular degeneration with dietary antioxidants, zinc, and omega-3 fatty acids: the Rotterdam study. Arch Ophthalmol. 2011;129(6):758–66.

    Article  CAS  PubMed  Google Scholar 

  436. Awh CC, Hawken S, Zanke BW. Treatment response to antioxidants and zinc based on CFH and ARMS2 genetic risk allele number in the age-related eye disease study. Ophthalmology. 2015;122(1):162–9.

    Article  PubMed  Google Scholar 

  437. Awh CC, Lane AM, Hawken S, Zanke B, Kim IK. CFH and ARMS2 genetic polymorphisms predict response to antioxidants and zinc in patients with age-related macular degeneration. Ophthalmology. 2013;120(11):2317–23.

    Article  PubMed  Google Scholar 

  438. Blom AM, Kask L, Ramesh B, Hillarp A. Effects of zinc on factor I cofactor activity of C4b-binding protein and factor H. Arch Biochem Biophys. 2003;418(2):108–18.

    Article  CAS  PubMed  Google Scholar 

  439. Evans BD, Amiraian K. Effect of zinc on lysis of sheep erythrocytes by guinea pig C4-deficient serum. Mol Immunol. 1987;24(12):1345–50.

    Article  CAS  PubMed  Google Scholar 

  440. Xu Y, Twarog M, Li N, Banks A, Schustak J, Bao Y, et al. Zinc transport from the endoplasmic reticulum to the cytoplasm via Zip7 is necessary for barrier dysfunction mediated by inflammatory signaling in RPE cells. PLoS ONE. 2022;17(7): e0271656.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  441. McLeod DS, Grebe R, Bhutto I, Merges C, Baba T, Lutty GA. Relationship between RPE and choriocapillaris in age-related macular degeneration. Invest Ophthalmol Vis Sci. 2009;50(10):4982–91.

    Article  PubMed  Google Scholar 

  442. Saint-Geniez M, Kurihara T, Sekiyama E, Maldonado AE, D’Amore PA. An essential role for RPE-derived soluble VEGF in the maintenance of the choriocapillaris. Proc Natl Acad Sci U S A. 2009;106(44):18751–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  443. Cunningham F, Cahyadi S, Lengyel I. A Potential New Role for Zinc in Age-Related Macular Degeneration through Regulation of Endothelial Fenestration. Int J Mol Sci. 2021;22(21).

  444. Jebari-Benslaiman S, Galicia-Garcia U, Larrea-Sebal A, Olaetxea JR, Alloza I, Vandenbroeck K, et al. Pathophysiology of Atherosclerosis. Int J Mol Sci. 2022;23(6):3346.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  445. Collaborators GMD. Global, regional, and national burden of 12 mental disorders in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. The Lancet Psychiatry. 2022;9(2):137–50.

    Article  Google Scholar 

  446. Beaglehole R, Yach D. Globalisation and the prevention and control of non-communicable disease: the neglected chronic diseases of adults. Lancet. 2003;362(9387):903–8.

    Article  CAS  PubMed  Google Scholar 

  447. Hong YM. Atherosclerotic cardiovascular disease beginning in childhood. Korean Circ J. 2010;40(1):1–9.

    Article  PubMed  PubMed Central  Google Scholar 

  448. Song P, Fang Z, Wang H, Cai Y, Rahimi K, Zhu Y, et al. Global and regional prevalence, burden, and risk factors for carotid atherosclerosis: a systematic review, meta-analysis, and modelling study. Lancet Glob Health. 2020;8(5):e721–9.

    Article  PubMed  Google Scholar 

  449. Tisdale MJ. Zinc-alpha2-glycoprotein in cachexia and obesity. Curr Opin Support Palliat Care. 2009;3(4):288–93.

    Article  PubMed  Google Scholar 

  450. Elattar S, Dimri M, Satyanarayana A. The tumor secretory factor ZAG promotes white adipose tissue browning and energy wasting. FASEB J. 2018;32(9):4727–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  451. Yang CJ, Han RY, Wang SS. Correlation of serum zinc alpha 2 glycoprotein with blood lipid and reproductive hormone levels in men. Zhonghua Nan Ke Xue. 2017;23(11):997–1001.

    PubMed  Google Scholar 

  452. Suliburska J, Cofta S, Gajewska E, Kalmus G, Sobieska M, Samborski W, et al. The evaluation of selected serum mineral concentrations and their association with insulin resistance in obese adolescents. Eur Rev Med Pharmacol Sci. 2013;17(17):2396–400.

    CAS  PubMed  Google Scholar 

  453. Xu C, Huang Z, Liu L, Luo C, Lu G, Li Q, et al. Zinc Regulates Lipid Metabolism and MMPs Expression in Lipid Disturbance Rabbits. Biol Trace Elem Res. 2015;168(2):411–20.

    Article  CAS  PubMed  Google Scholar 

  454. Ranasinghe P, Wathurapatha WS, Ishara MH, Jayawardana R, Galappatthy P, Katulanda P, et al. Effects of Zinc supplementation on serum lipids: a systematic review and meta-analysis. Nutr Metab (Lond). 2015;12:26.

    Article  PubMed  Google Scholar 

  455. Gunasekara P, Hettiarachchi M, Liyanage C, Lekamwasam S. Effects of zinc and multimineral vitamin supplementation on glycemic and lipid control in adult diabetes. Diabetes Metab Syndr Obes. 2011;4:53–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  456. Afkhami-Ardekani M, Karimi M, Mohammadi SM, Nourani F. Effect of zinc sulfate supplementation on lipid and glucose in type 2 diabetic patients. Pak J Nutr. 2008;7(4):550–3.

    Article  CAS  Google Scholar 

  457. Bergomi M, Rovesti S, Vinceti M, Vivoli R, Caselgrandi E, Vivoli G. Zinc and copper status and blood pressure. J Trace Elem Med Biol. 1997;11(3):166–9.

    Article  CAS  PubMed  Google Scholar 

  458. Mousavi SM, Mofrad MD, do Nascimento IJB, Milajerdi A, Mokhtari T, Esmaillzadeh A. The effect of zinc supplementation on blood pressure: a systematic review and dose-response meta-analysis of randomized-controlled trials. Eur J Nutr. 2020;59(5):1815–27.

  459. Tomat AL, Inserra F, Veiras L, Vallone MC, Balaszczuk AM, Costa MA, et al. Moderate zinc restriction during fetal and postnatal growth of rats: effects on adult arterial blood pressure and kidney. Am J Physiol Regul Integr Comp Physiol. 2008;295(2):R543–9.

    Article  CAS  PubMed  Google Scholar 

  460. Wei Q, Wang J, Wang MH, Yu F, Dong Z. Inhibition of apoptosis by Zn2+ in renal tubular cells following ATP depletion. Am J Physiol Renal Physiol. 2004;287(3):F492-500.

    Article  CAS  PubMed  Google Scholar 

  461. Gobetto MN, Mendes Garrido Abregu F, Caniffi C, Veiras L, Elesgaray R, Gironacci M, et al. Fetal and postnatal zinc restriction: sex differences in the renal renin-angiotensin system of newborn and adult Wistar rats. J Nutr Biochem. 2020;81:108385.

  462. Williams CR, Mistry M, Cheriyan AM, Williams JM, Naraine MK, Ellis CL, et al. Zinc deficiency induces hypertension by promoting renal Na(+) reabsorption. Am J Physiol Renal Physiol. 2019;316(4):F646–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  463. Davies PF, Civelek M, Fang Y, Fleming I. The atherosusceptible endothelium: endothelial phenotypes in complex haemodynamic shear stress regions in vivo. Cardiovasc Res. 2013;99(2):315–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  464. Tran N, Garcia T, Aniqa M, Ali S, Ally A, Nauli SM. Endothelial Nitric Oxide Synthase (eNOS) and the Cardiovascular System: in Physiology and in Disease States. Am J Biomed Sci Res. 2022;15(2):153–77.

    CAS  PubMed  PubMed Central  Google Scholar 

  465. Chreifi G, Li H, McInnes CR, Gibson CL, Suckling CJ, Poulos TL. Communication between the zinc and tetrahydrobiopterin binding sites in nitric oxide synthase. Biochemistry. 2014;53(25):4216–23.

    Article  CAS  PubMed  Google Scholar 

  466. Zou MH, Shi C, Cohen RA. Oxidation of the zinc-thiolate complex and uncoupling of endothelial nitric oxide synthase by peroxynitrite. J Clin Invest. 2002;109(6):817–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  467. Zhuang X, Pang X, Zhang W, Wu W, Zhao J, Yang H, et al. Effects of zinc and manganese on advanced glycation end products (AGEs) formation and AGEs-mediated endothelial cell dysfunction. Life Sci. 2012;90(3–4):131–9.

    Article  CAS  PubMed  Google Scholar 

  468. Hennig B, Meerarani P, Toborek M, McClain CJ. Antioxidant-like properties of zinc in activated endothelial cells. J Am Coll Nutr. 1999;18(2):152–8.

    Article  CAS  PubMed  Google Scholar 

  469. Connell P, Young VM, Toborek M, Cohen DA, Barve S, McClain CJ, et al. Zinc attenuates tumor necrosis factor-mediated activation of transcription factors in endothelial cells. J Am Coll Nutr. 1997;16(5):411–7.

    Article  CAS  PubMed  Google Scholar 

  470. Palazon A, Goldrath AW, Nizet V, Johnson RS. HIF transcription factors, inflammation, and immunity. Immunity. 2014;41(4):518–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  471. Nardinocchi L, Pantisano V, Puca R, Porru M, Aiello A, Grasselli A, et al. Zinc downregulates HIF-1alpha and inhibits its activity in tumor cells in vitro and in vivo. PLoS One. 2010;5(12):e15048.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  472. Hennig B, Wang Y, Ramasamy S, McClain CJ. Zinc deficiency alters barrier function of cultured porcine endothelial cells. J Nutr. 1992;122(6):1242–7.

    Article  CAS  PubMed  Google Scholar 

  473. Pang B, Dong G, Pang T, Sun X, Liu X, Nie Y, et al. Emerging insights into the pathogenesis and therapeutic strategies for vascular endothelial injury-associated diseases: focus on mitochondrial dysfunction. Angiogenesis. 2024;27(4):623–39.

    Article  PubMed  PubMed Central  Google Scholar 

  474. Thambiayya K, Wasserloos K, Kagan VE, Stoyanovsky D, Pitt BR. A critical role for increased labile zinc in reducing sensitivity of cultured sheep pulmonary artery endothelial cells to LPS-induced apoptosis. Am J Physiol Lung Cell Mol Physiol. 2012;302(12):L1287–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  475. Meerarani P, Ramadass P, Toborek M, Bauer HC, Bauer H, Hennig B. Zinc protects against apoptosis of endothelial cells induced by linoleic acid and tumor necrosis factor alpha. Am J Clin Nutr. 2000;71(1):81–7.

    Article  CAS  PubMed  Google Scholar 

  476. Kuvibidila SR, Sandoval M, Lao J, Velez M, Yu L, Ode D, et al. Plasma zinc levels inversely correlate with vascular cell adhesion molecule-1 concentration in children with sickle cell disease. J Natl Med Assoc. 2006;98(8):1263–72.

    PubMed  PubMed Central  Google Scholar 

  477. Cho YE, Choi JE, Alam MJ, Lee MH, Sohn HY, Beattie JH, et al. Zinc deficiency decreased cell viability both in endothelial EA. hy926 cells and mouse aortic culture ex vivo and its implication for anti-atherosclerosis. Nutr Res Pract. 2008;2(2):74–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  478. Bao B, Prasad AS, Beck FW, Fitzgerald JT, Snell D, Bao GW, et al. Zinc decreases C-reactive protein, lipid peroxidation, and inflammatory cytokines in elderly subjects: a potential implication of zinc as an atheroprotective agent. Am J Clin Nutr. 2010;91(6):1634–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  479. Singh RB, Gupta UC, Mittal N, Niaz MA, Ghosh S, Rastogi V. Epidemiologic study of trace elements and magnesium on risk of coronary artery disease in rural and urban Indian populations. J Am Coll Nutr. 1997;16(1):62–7.

    Article  CAS  PubMed  Google Scholar 

  480. Yu X, Huang L, Zhao J, Wang Z, Yao W, Wu X, et al. The Relationship between Serum Zinc Level and Heart Failure: A Meta-Analysis. Biomed Res Int. 2018;2018:2739014.

    PubMed  PubMed Central  Google Scholar 

  481. Huang L, Teng T, Bian B, Yao W, Yu X, Wang Z, et al. Zinc Levels in Left Ventricular Hypertrophy. Biol Trace Elem Res. 2017;176(1):48–55.

    Article  CAS  PubMed  Google Scholar 

  482. Alissa EM, Bahjri SM, Ahmed WH, Al-Ama N, Ferns GA. Trace element status in Saudi patients with established atherosclerosis. J Trace Elem Med Biol. 2006;20(2):105–14.

    Article  CAS  PubMed  Google Scholar 

  483. Qazmooz HA, Smesam HN, Mousa RF, Al-Hakeim HK, Maes M. Trace element, immune and opioid biomarkers of unstable angina, increased atherogenicity and insulin resistance: Results of machine learning. J Trace Elem Med Biol. 2021;64:126703.

    Article  CAS  PubMed  Google Scholar 

  484. Gao JW, Zhang SL, Hao QY, Huang FF, Liu ZY, Zhang HF, et al. Association of dietary zinc intake with coronary artery calcium progression: the Multi-Ethnic Study of Atherosclerosis (MESA). Eur J Nutr. 2021;60(5):2759–67.

    Article  CAS  PubMed  Google Scholar 

  485. de Oliveira Otto MC, Alonso A, Lee DH, Delclos GL, Bertoni AG, Jiang R, et al. Dietary intakes of zinc and heme iron from red meat, but not from other sources, are associated with greater risk of metabolic syndrome and cardiovascular disease. J Nutr. 2012;142(3):526–33.

    Article  PubMed  PubMed Central  Google Scholar 

  486. Liu B, Cai ZQ, Zhou YM. Deficient zinc levels and myocardial infarction : association between deficient zinc levels and myocardial infarction: a meta-analysis. Biol Trace Elem Res. 2015;165(1):41–50.

    Article  CAS  PubMed  Google Scholar 

  487. Depressive disorder (depression): World Health Organisation; 2023 [Available from: https://www.who.int/en/news-room/fact-sheets/detail/depression.

  488. Hasin DS, Sarvet AL, Meyers JL, Saha TD, Ruan WJ, Stohl M, et al. Epidemiology of Adult DSM-5 Major Depressive Disorder and Its Specifiers in the United States. JAMA Psychiat. 2018;75(4):336–46.

    Article  Google Scholar 

  489. Whiteford HA, Degenhardt L, Rehm J, Baxter AJ, Ferrari AJ, Erskine HE, et al. Global burden of disease attributable to mental and substance use disorders: findings from the Global Burden of Disease Study 2010. Lancet. 2013;382(9904):1575–86.

    Article  PubMed  Google Scholar 

  490. Leung YW, Flora DB, Gravely S, Irvine J, Carney RM, Grace SL. The impact of premorbid and postmorbid depression onset on mortality and cardiac morbidity among patients with coronary heart disease: meta-analysis. Psychosom Med. 2012;74(8):786–801.

    Article  PubMed  PubMed Central  Google Scholar 

  491. Richmond-Rakerd LS, D’Souza S, Milne BJ, Caspi A, Moffitt TE. Longitudinal associations of mental disorders with physical diseases and mortality among 2.3 Million New Zealand Citizens. JAMA Netw Open. 2021;4(1):e2033448.

    Article  PubMed  PubMed Central  Google Scholar 

  492. Diniz BS, Butters MA, Albert SM, Dew MA, Reynolds CF 3rd. Late-life depression and risk of vascular dementia and Alzheimer’s disease: systematic review and meta-analysis of community-based cohort studies. Br J Psychiatry. 2013;202(5):329–35.

    Article  PubMed  PubMed Central  Google Scholar 

  493. Richmond-Rakerd LS, D’Souza S, Milne BJ, Caspi A, Moffitt TE. Longitudinal associations of mental disorders with dementia: 30-year analysis of 1.7 Million New Zealand Citizens. JAMA Psychiatry. 2022;79(4):333–40.

    Article  PubMed  PubMed Central  Google Scholar 

  494. Soysal P, Veronese N, Thompson T, Kahl KG, Fernandes BS, Prina AM, et al. Relationship between depression and frailty in older adults: A systematic review and meta-analysis. Ageing Res Rev. 2017;36:78–87.

    Article  PubMed  Google Scholar 

  495. Asmer MS, Kirkham J, Newton H, Ismail Z, Elbayoumi H, Leung RH, et al. Meta-Analysis of the Prevalence of Major Depressive Disorder Among Older Adults With Dementia. J Clin Psychiatry. 2018;79(5).

  496. Diniz BS, Reynolds CF 3rd, Butters MA, Dew MA, Firmo JO, Lima-Costa MF, et al. The effect of gender, age, and symptom severity in late-life depression on the risk of all-cause mortality: the Bambui Cohort Study of Aging. Depress Anxiety. 2014;31(9):787–95.

    Article  PubMed  Google Scholar 

  497. Kennedy SH, Lam RW, McIntyre RS, Tourjman SV, Bhat V, Blier P, et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 Clinical Guidelines for the Management of Adults with Major Depressive Disorder: Section 3. Pharmacological Treatments Can J Psychiatry. 2016;61(9):540–60.

    Article  PubMed  Google Scholar 

  498. Cipriani A, Furukawa TA, Salanti G, Chaimani A, Atkinson LZ, Ogawa Y, et al. comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: a systematic review and network meta-analysis. Focus (Am Psychiatr Publ). 2018;16(4):420–9.

    PubMed  PubMed Central  Google Scholar 

  499. Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, Warden D, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry. 2006;163(11):1905–17.

    Article  PubMed  Google Scholar 

  500. Anbari-Nogyni Z, Bidaki R, Madadizadeh F, Sangsefidi ZS, Fallahzadeh H, Karimi-Nazari E, et al. Relationship of zinc status with depression and anxiety among elderly population. Clin Nutr ESPEN. 2020;37:233–9.

    Article  PubMed  Google Scholar 

  501. Li Z, Wang W, Xin X, Song X, Zhang D. Association of total zinc, iron, copper and selenium intakes with depression in the US adults. J Affect Disord. 2018;228:68–74.

    Article  CAS  PubMed  Google Scholar 

  502. Al-Fartusie FS, Al-Bairmani HK, Al-Garawi ZS, Yousif AH. Evaluation of Some Trace Elements and Vitamins in Major Depressive Disorder Patients: a Case-Control Study. Biol Trace Elem Res. 2019;189(2):412–9.

    Article  CAS  PubMed  Google Scholar 

  503. Islam MR, Islam MR, Shalahuddin Qusar MMA, Islam MS, Kabir MH, Mustafizur Rahman GKM, et al. Alterations of serum macro-minerals and trace elements are associated with major depressive disorder: a case-control study. BMC Psychiatry. 2018;18(1):94.

    Article  PubMed  PubMed Central  Google Scholar 

  504. Amani R, Saeidi S, Nazari Z, Nematpour S. Correlation between dietary zinc intakes and its serum levels with depression scales in young female students. Biol Trace Elem Res. 2010;137(2):150–8.

    Article  CAS  PubMed  Google Scholar 

  505. Stanislawska M, Szkup-Jablonska M, Jurczak A, Wieder-Huszla S, Samochowiec A, Jasiewicz A, et al. The severity of depressive symptoms vs. serum Mg and Zn levels in postmenopausal women. Biol Trace Elem Res. 2014;157(1):30–5.

    Article  CAS  PubMed  Google Scholar 

  506. Roozbeh J, Sharifian M, Ghanizadeh A, Sahraian A, Sagheb MM, Shabani S, et al. Association of zinc deficiency and depression in the patients with end-stage renal disease on hemodialysis. J Ren Nutr. 2011;21(2):184–7.

    Article  CAS  PubMed  Google Scholar 

  507. Nowak G, Siwek M, Dudek D, Zieba A, Pilc A. Effect of zinc supplementation on antidepressant therapy in unipolar depression: a preliminary placebo-controlled study. Pol J Pharmacol. 2003;55(6):1143–7.

    CAS  PubMed  Google Scholar 

  508. Sawada T, Yokoi K. Effect of zinc supplementation on mood states in young women: a pilot study. Eur J Clin Nutr. 2010;64(3):331–3.

    Article  CAS  PubMed  Google Scholar 

  509. Nguyen PH, Grajeda R, Melgar P, Marcinkevage J, DiGirolamo AM, Flores R, et al. Micronutrient supplementation may reduce symptoms of depression in Guatemalan women. Arch Latinoam Nutr. 2009;59(3):278–86.

    CAS  PubMed  Google Scholar 

  510. Konsman JP, Veeneman J, Combe C, Poole S, Luheshi GN, Dantzer R. Central nervous action of interleukin-1 mediates activation of limbic structures and behavioural depression in response to peripheral administration of bacterial lipopolysaccharide. Eur J Neurosci. 2008;28(12):2499–510.

    Article  CAS  PubMed  Google Scholar 

  511. Wessels I, Haase H, Engelhardt G, Rink L, Uciechowski P. Zinc deficiency induces production of the proinflammatory cytokines IL-1beta and TNFalpha in promyeloid cells via epigenetic and redox-dependent mechanisms. J Nutr Biochem. 2013;24(1):289–97.

    Article  CAS  PubMed  Google Scholar 

  512. Li Y, Xiao B, Qiu W, Yang L, Hu B, Tian X, et al. Altered expression of CD4(+)CD25(+) regulatory T cells and its 5-HT(1a) receptor in patients with major depression disorder. J Affect Disord. 2010;124(1–2):68–75.

    Article  CAS  PubMed  Google Scholar 

  513. Himmerich H, Milenovic S, Fulda S, Plumakers B, Sheldrick AJ, Michel TM, et al. Regulatory T cells increased while IL-1beta decreased during antidepressant therapy. J Psychiatr Res. 2010;44(15):1052–7.

    Article  PubMed  Google Scholar 

  514. Himmerich H, Fulda S, Sheldrick AJ, Plumakers B, Rink L. IFN-gamma reduction by tricyclic antidepressants. Int J Psychiatry Med. 2010;40(4):413–24.

    Article  PubMed  Google Scholar 

  515. Mo F, Tang Y, Du P, Shen Z, Yang J, Cai M, et al. GPR39 protects against corticosterone-induced neuronal injury in hippocampal cells through the CREB-BDNF signaling pathway. J Affect Disord. 2020;272:474–84.

    Article  CAS  PubMed  Google Scholar 

  516. Mlyniec K, Doboszewska U, Szewczyk B, Sowa-Kucma M, Misztak P, Piekoszewski W, et al. The involvement of the GPR39-Zn(2+)-sensing receptor in the pathophysiology of depression. Studies in rodent models and suicide victims. Neuropharmacology. 2014;79:290–7.

    Article  CAS  PubMed  Google Scholar 

  517. Sagi R, Chakraborty M, Bogdanovic M, Asraf H, Sekler I, Kofman O, et al. Loss of the zinc receptor ZnR/GPR39 in mice enhances anxiety-related behavior and motor deficits, and modulates KCC2 expression in the amygdala. Behav Brain Funct. 2024;20(1):31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  518. Suh SW, Won SJ, Hamby AM, Yoo BH, Fan Y, Sheline CT, et al. Decreased brain zinc availability reduces hippocampal neurogenesis in mice and rats. J Cereb Blood Flow Metab. 2009;29(9):1579–88.

    Article  CAS  PubMed  Google Scholar 

  519. Rafalo-Ulinska A, Pochwat B, Misztak P, Bugno R, Kryczyk-Poprawa A, Opoka W, et al. Zinc Deficiency Blunts the Effectiveness of Antidepressants in the Olfactory Bulbectomy Model of Depression in Rats. Nutrients. 2022;14(13).

  520. Doboszewska U, Szewczyk B, Sowa-Kucma M, Mlyniec K, Rafalo A, Ostachowicz B, et al. Antidepressant activity of fluoxetine in the zinc deficiency model in rats involves the NMDA receptor complex. Behav Brain Res. 2015;287:323–30.

    Article  CAS  PubMed  Google Scholar 

  521. Doboszewska U, Sowa-Kucma M, Mlyniec K, Pochwat B, Holuj M, Ostachowicz B, et al. Zinc deficiency in rats is associated with up-regulation of hippocampal NMDA receptor. Prog Neuropsychopharmacol Biol Psychiatry. 2015;56:254–63.

    Article  CAS  PubMed  Google Scholar 

  522. Thanapaul R, Shvedova M, Shin GH, Roh DS. An Insight into Aging, Senescence, and Their Impacts on Wound Healing. Adv Geriatr Med Res. 2021;3(3).

  523. Wang Z, Shi C. Cellular senescence is a promising target for chronic wounds: a comprehensive review. Burns Trauma. 2020;8:tkaa021.

    Article  PubMed  PubMed Central  Google Scholar 

  524. Gould L, Abadir P, Brem H, Carter M, Conner-Kerr T, Davidson J, et al. Chronic wound repair and healing in older adults: current status and future research. Wound Repair Regen. 2015;23(1):1–13.

    Article  PubMed  PubMed Central  Google Scholar 

  525. Markiewicz-Gospodarek A, Koziol M, Tobiasz M, Baj J, Radzikowska-Buchner E, Przekora A. Burn Wound Healing: Clinical Complications, Medical Care, Treatment, and Dressing Types: The Current State of Knowledge for Clinical Practice. Int J Environ Res Public Health. 2022;19(3).

  526. Nussbaum SR, Carter MJ, Fife CE, DaVanzo J, Haught R, Nusgart M, et al. An economic evaluation of the impact, cost, and medicare policy implications of chronic nonhealing wounds. Value Health. 2018;21(1):27–32.

    Article  PubMed  Google Scholar 

  527. Gupta M, Mahajan VK, Mehta KS, Chauhan PS. Zinc therapy in dermatology: a review. Dermatol Res Pract. 2014;2014:709152.

    Article  PubMed  PubMed Central  Google Scholar 

  528. Nurden AT, Nurden P, Sanchez M, Andia I, Anitua E. Platelets and wound healing. Front Biosci. 2008;13(9):3532–48.

    PubMed  Google Scholar 

  529. Gordon PR, Woodruff CW, Anderson HL, O’Dell BL. Effect of acute zinc deprivation on plasma zinc and platelet aggregation in adult males. Am J Clin Nutr. 1982;35(1):113–9.

    Article  CAS  PubMed  Google Scholar 

  530. Emery MP, Browning JD, O’Dell BL. Impaired hemostasis and platelet function in rats fed low zinc diets based on egg white protein. J Nutr. 1990;120(9):1062–7.

    Article  CAS  PubMed  Google Scholar 

  531. Heyns Adu P, Eldor A, Yarom R, Marx G. Zinc-induced platelet aggregation is mediated by the fibrinogen receptor and is not accompanied by release or by thromboxane synthesis. Blood. 1985;66(1):213–9.

    Article  PubMed  Google Scholar 

  532. Marx G, Krugliak J, Shaklai M. Nutritional zinc increases platelet reactivity. Am J Hematol. 1991;38(3):161–5.

    Article  CAS  PubMed  Google Scholar 

  533. Lansdown AB, Sampson B, Rowe A. Sequential changes in trace metal, metallothionein and calmodulin concentrations in healing skin wounds. J Anat. 1999;195 ( Pt 3)(Pt 3):375–86.

  534. Richards MP, Cousins RJ. Metallothionein and its relationship to the metabolism of dietary zinc in rats. J Nutr. 1976;106(11):1591–9.

    Article  CAS  PubMed  Google Scholar 

  535. Sharir H, Zinger A, Nevo A, Sekler I, Hershfinkel M. Zinc released from injured cells is acting via the Zn2+-sensing receptor, ZnR, to trigger signaling leading to epithelial repair. J Biol Chem. 2010;285(34):26097–106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  536. Pastar I, Stojadinovic O, Yin NC, Ramirez H, Nusbaum AG, Sawaya A, et al. Epithelialization in wound healing: a comprehensive review. Adv Wound Care (New Rochelle). 2014;3(7):445–64.

    Article  PubMed  Google Scholar 

  537. Martins VL, Caley M, O’Toole EA. Matrix metalloproteinases and epidermal wound repair. Cell Tissue Res. 2013;351(2):255–68.

    Article  CAS  PubMed  Google Scholar 

  538. Tezvergil-Mutluay A, Agee KA, Hoshika T, Carrilho M, Breschi L, Tjaderhane L, et al. The requirement of zinc and calcium ions for functional MMP activity in demineralized dentin matrices. Dent Mater. 2010;26(11):1059–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  539. Agren MS. Zinc oxide increases degradation of collagen in necrotic wound tissue. Br J Dermatol. 1993;129(2):221.

    Article  CAS  PubMed  Google Scholar 

  540. Raffoul W, Far MS, Cayeux MC, Berger MM. Nutritional status and food intake in nine patients with chronic low-limb ulcers and pressure ulcers: importance of oral supplements. Nutrition. 2006;22(1):82–8.

    Article  CAS  PubMed  Google Scholar 

  541. Rojas AI, Phillips TJ. Patients with chronic leg ulcers show diminished levels of vitamins A and E, carotenes, and zinc. Dermatol Surg. 1999;25(8):601–4.

    Article  CAS  PubMed  Google Scholar 

  542. Hawkins T, Marks JM, Plummer VM, Greaves MW. Whole body monitoring and other studies of zinc-65 metabolism in patients with dermatological diseases. Clin Exp Dermatol. 1976;1(3):243–52.

    Article  CAS  PubMed  Google Scholar 

  543. Agren MS, Chvapil M, Franzen L. Enhancement of re-epithelialization with topical zinc oxide in porcine partial-thickness wounds. J Surg Res. 1991;50(2):101–5.

    Article  CAS  PubMed  Google Scholar 

  544. Tenaud I, Leroy S, Chebassier N, Dreno B. Zinc, copper and manganese enhanced keratinocyte migration through a functional modulation of keratinocyte integrins. Exp Dermatol. 2000;9(6):407–16.

    Article  CAS  PubMed  Google Scholar 

  545. Kietzmann. Improvement and retardation of wound healing: effects of pharmacological agents in laboratory animal studies. Vet Dermatol. 1999;10(2):83-8.

  546. Stromberg HE, Agren MS. Topical zinc oxide treatment improves arterial and venous leg ulcers. Br J Dermatol. 1984;111(4):461–8.

    Article  CAS  PubMed  Google Scholar 

  547. Agren MS, Stromberg HE. Topical treatment of pressure ulcers. A randomized comparative trial of Varidase and zinc oxide. Scand J Plast Reconstr Surg. 1985;19(1):97-100.

  548. Apelqvist J, Larsson J, Stenstrom A. Topical treatment of necrotic foot ulcers in diabetic patients: a comparative trial of DuoDerm and MeZinc. Br J Dermatol. 1990;123(6):787–92.

    Article  CAS  PubMed  Google Scholar 

  549. Attia EA, Belal DM, El Samahy MH, El Hamamsy MH. A pilot trial using topical regular crystalline insulin vs. aqueous zinc solution for uncomplicated cutaneous wound healing: Impact on quality of life. Wound Repair Regen. 2014;22(1):52-7.

  550. Cereda E, Gini A, Pedrolli C, Vanotti A. Disease-specific, versus standard, nutritional support for the treatment of pressure ulcers in institutionalized older adults: a randomized controlled trial. J Am Geriatr Soc. 2009;57(8):1395–402.

    Article  PubMed  Google Scholar 

  551. Faure H, Peyrin JC, Richard MJ, Favier A. Parenteral supplementation with zinc in surgical patients corrects postoperative serum-zinc drop. Biol Trace Elem Res. 1991;30(1):37–45.

    Article  CAS  PubMed  Google Scholar 

  552. Wilkinson EA, Hawke CI. Does oral zinc aid the healing of chronic leg ulcers? A systematic literature review. Arch Dermatol. 1998;134(12):1556–60.

    Article  CAS  PubMed  Google Scholar 

  553. Wissing U, Unosson M, Lennernas MA, Ek AC. Nutritional intake and physical activity in leg ulcer patients. J Adv Nurs. 1997;25(3):571–8.

    Article  CAS  PubMed  Google Scholar 

  554. O’Connor S, Murphy S. Chronic venous leg ulcers: is topical zinc the answer? A review of the literature. Adv Skin Wound Care. 2014;27(1):35–44 (quiz 5-6).

    Article  PubMed  Google Scholar 

  555. Hu Y, Liu W, Chen Y, Zhang M, Wang L, Zhou H, et al. Combined use of fasting plasma glucose and glycated hemoglobin A1c in the screening of diabetes and impaired glucose tolerance. Acta Diabetol. 2010;47(3):231–6.

    Article  CAS  PubMed  Google Scholar 

  556. d’Emden MC, Shaw JE, Jones GR, Cheung NW. Guidance concerning the use of glycated haemoglobin (HbA1c) for the diagnosis of diabetes mellitus. Med J Aust. 2015;203(2):89–90.

    Article  PubMed  Google Scholar 

  557. Fowler MJ. Microvascular and macrovascular complications of diabetes. Clinical diabetes. 2008;26(2):77–82.

    Article  Google Scholar 

  558. Murray CJ, Abbafati C, Abbas KM, Abbasi M, Abbasi-Kangevari M, Abd-Allah F, et al. Five insights from the global burden of disease study 2019. The Lancet. 2020;396(10258):1135–59.

    Article  Google Scholar 

  559. Global report on diabetes: World Health Organization; 2016.

  560. Saeedi P, Salpea P, Karuranga S, Petersohn I, Malanda B, Gregg EW, et al. Mortality attributable to diabetes in 20–79 years old adults, 2019 estimates: Results from the International Diabetes Federation Diabetes Atlas, 9(th) edition. Diabetes Res Clin Pract. 2020;162:108086.

    Article  PubMed  Google Scholar 

  561. Domjan BA, Ferencz V, Tanczer T, Szili-Janicsek Z, Barkai L, Hidvegi T, et al. Large increase in the prevalence of self-reported diabetes based on a nationally representative survey in Hungary. Prim Care Diabetes. 2017;11(2):107–11.

    Article  PubMed  Google Scholar 

  562. Koenen M, Hill MA, Cohen P, Sowers JR. Obesity, adipose tissue and vascular dysfunction. Circ Res. 2021;128(7):951–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  563. Dunn MF. Zinc-ligand interactions modulate assembly and stability of the insulin hexamer – a review. Biometals. 2005;18(4):295–303.

    Article  CAS  PubMed  Google Scholar 

  564. Wijesekara N, Dai FF, Hardy AB, Giglou PR, Bhattacharjee A, Koshkin V, et al. Beta cell-specific Znt8 deletion in mice causes marked defects in insulin processing, crystallisation and secretion. Diabetologia. 2010;53(8):1656–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  565. Lefebvre B, Vandewalle B, Balavoine AS, Queniat G, Moerman E, Vantyghem MC, et al. Regulation and functional effects of ZNT8 in human pancreatic islets. J Endocrinol. 2012;214(2):225–32.

    Article  CAS  PubMed  Google Scholar 

  566. Davidson HW, Wenzlau JM, O’Brien RM. Zinc transporter 8 (ZnT8) and beta cell function. Trends Endocrinol Metab. 2014;25(8):415–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  567. Salem SD, Saif-Ali R, Ismail IS, Al-Hamodi Z, Muniandy S. Contribution of SLC30A8 variants to the risk of type 2 diabetes in a multi-ethnic population: a case control study. BMC Endocr Disord. 2014;14:2.

    Article  PubMed  PubMed Central  Google Scholar 

  568. Drake I, Hindy G, Ericson U, Orho-Melander M. A prospective study of dietary and supplemental zinc intake and risk of type 2 diabetes depending on genetic variation in SLC30A8. Genes Nutr. 2017;12:30.

    Article  PubMed  PubMed Central  Google Scholar 

  569. Paganelli R, Scala E, Quinti I, Ansotegui IJ. Humoral immunity in aging. Aging (Milano). 1994;6(3):143–50.

    CAS  PubMed  Google Scholar 

  570. Mandrup-Poulsen T. The role of interleukin-1 in the pathogenesis of IDDM. Diabetologia. 1996;39(9):1005–29.

    Article  CAS  PubMed  Google Scholar 

  571. Cnop M, Welsh N, Jonas JC, Jorns A, Lenzen S, Eizirik DL. Mechanisms of pancreatic beta-cell death in type 1 and type 2 diabetes: many differences, few similarities. Diabetes. 2005;54 Suppl 2(suppl_2):S97-107.

  572. Sauter NS, Schulthess FT, Galasso R, Castellani LW, Maedler K. The antiinflammatory cytokine interleukin-1 receptor antagonist protects from high-fat diet-induced hyperglycemia. Endocrinology. 2008;149(5):2208–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  573. Prasad AS, Bao B, Beck FW, Kucuk O, Sarkar FH. Antioxidant effect of zinc in humans. Free Radic Biol Med. 2004;37(8):1182–90.

    Article  CAS  PubMed  Google Scholar 

  574. Kahmann L, Uciechowski P, Warmuth S, Plumakers B, Gressner AM, Malavolta M, et al. Zinc supplementation in the elderly reduces spontaneous inflammatory cytokine release and restores T cell functions. Rejuvenation Res. 2008;11(1):227–37.

    Article  CAS  PubMed  Google Scholar 

  575. Sakaguchi M. The role of insulin signaling with FOXO and FOXK transcription factors. Endocr J. 2024;71(10):939–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  576. Norouzi S, Adulcikas J, Sohal SS, Myers S. Zinc stimulates glucose oxidation and glycemic control by modulating the insulin signaling pathway in human and mouse skeletal muscle cell lines. PLoS One. 2018;13(1):e0191727.

    Article  PubMed  PubMed Central  Google Scholar 

  577. Wu Y, Lu H, Yang H, Li C, Sang Q, Liu X, et al. Zinc stimulates glucose consumption by modulating the insulin signaling pathway in L6 myotubes: essential roles of Akt-GLUT4, GSK3beta and mTOR-S6K1. J Nutr Biochem. 2016;34:126–35.

    Article  PubMed  Google Scholar 

  578. Nakayama A, Hiromura M, Adachi Y, Sakurai H. Molecular mechanism of antidiabetic zinc-allixin complexes: regulations of glucose utilization and lipid metabolism. J Biol Inorg Chem. 2008;13(5):675–84.

    Article  CAS  PubMed  Google Scholar 

  579. Basuki W, Hiromura M, Sakurai H. Insulinomimetic Zn complex (Zn(opt)2) enhances insulin signaling pathway in 3T3-L1 adipocytes. J Inorg Biochem. 2007;101(4):692–9.

    Article  CAS  PubMed  Google Scholar 

  580. Wilson M, Hogstrand C, Maret W. Picomolar concentrations of free zinc(II) ions regulate receptor protein-tyrosine phosphatase beta activity. J Biol Chem. 2012;287(12):9322–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  581. Wu W, Wang X, Zhang W, Reed W, Samet JM, Whang YE, et al. Zinc-induced PTEN protein degradation through the proteasome pathway in human airway epithelial cells. J Biol Chem. 2003;278(30):28258–63.

    Article  CAS  PubMed  Google Scholar 

  582. Plum LM, Brieger A, Engelhardt G, Hebel S, Nessel A, Arlt M, et al. PTEN-inhibition by zinc ions augments interleukin-2-mediated Akt phosphorylation. Metallomics. 2014;6(7):1277–87.

    Article  CAS  PubMed  Google Scholar 

  583. Walter PL, Kampkotter A, Eckers A, Barthel A, Schmoll D, Sies H, et al. Modulation of FoxO signaling in human hepatoma cells by exposure to copper or zinc ions. Arch Biochem Biophys. 2006;454(2):107–13.

    Article  CAS  PubMed  Google Scholar 

  584. Zhang Y, Zhang Y, Yu Y. Global Phosphoproteomic Analysis of Insulin/Akt/mTORC1/S6K Signaling in Rat Hepatocytes. J Proteome Res. 2017;16(8):2825–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  585. Tang X, Shay NF. Zinc has an insulin-like effect on glucose transport mediated by phosphoinositol-3-kinase and Akt in 3T3-L1 fibroblasts and adipocytes. J Nutr. 2001;131(5):1414–20.

    Article  CAS  PubMed  Google Scholar 

  586. Ilouz R, Kaidanovich O, Gurwitz D, Eldar-Finkelman H. Inhibition of glycogen synthase kinase-3beta by bivalent zinc ions: insight into the insulin-mimetic action of zinc. Biochem Biophys Res Commun. 2002;295(1):102–6.

    Article  CAS  PubMed  Google Scholar 

  587. Ugi S, Imamura T, Maegawa H, Egawa K, Yoshizaki T, Shi K, et al. Protein phosphatase 2A negatively regulates insulin’s metabolic signaling pathway by inhibiting Akt (protein kinase B) activity in 3T3-L1 adipocytes. Mol Cell Biol. 2004;24(19):8778–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  588. Al-Maroof RA, Al-Sharbatti SS. Serum zinc levels in diabetic patients and effect of zinc supplementation on glycemic control of type 2 diabetics. Saudi Med J. 2006;27(3):344–50.

    PubMed  Google Scholar 

  589. Williams NR, Rajput-Williams J, West JA, Nigdikar SV, Foote JW, Howard AN. Plasma, granulocyte and mononuclear cell copper and zinc in patients with diabetes mellitus. Analyst. 1995;120(3):887–90.

    Article  CAS  PubMed  Google Scholar 

  590. Saharia GK, Goswami RK. Evaluation of serum zinc status and glycated hemoglobin of type 2 diabetes mellitus patients in a tertiary care hospital of assam. J Lab Physicians. 2013;5(1):30–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  591. Hussein M, Fathy W, Hassan A, Elkareem RA, Marzouk S, Kamal YS. Zinc deficiency correlates with severity of diabetic polyneuropathy. Brain Behav. 2021;11(10):e2349.

    Article  PubMed  PubMed Central  Google Scholar 

  592. Hayee MA, Mohammad QD, Haque A. Diabetic neuropathy and zinc therapy. Bangladesh Med Res Counc Bull. 2005;31(2):62–7.

    CAS  PubMed  Google Scholar 

  593. Gupta R, Garg VK, Mathur DK, Goyal RK. Oral zinc therapy in diabetic neuropathy. J Assoc Physicians India. 1998;46(11):939–42.

    CAS  PubMed  Google Scholar 

  594. Khan MI, Siddique KU, Ashfaq F, Ali W, Reddy HD, Mishra A. Effect of high-dose zinc supplementation with oral hypoglycemic agents on glycemic control and inflammation in type-2 diabetic nephropathy patients. J Nat Sci Biol Med. 2013;4(2):336–40.

    Article  PubMed  PubMed Central  Google Scholar 

  595. Farvid MS, Siassi F, Jalali M, Hosseini M, Saadat N. The impact of vitamin and/or mineral supplementation on lipid profiles in type 2 diabetes. Diabetes Res Clin Pract. 2004;65(1):21–8.

    Article  CAS  PubMed  Google Scholar 

  596. Kadhim HM, Ismail SH, Hussein KI, Bakir IH, Sahib AS, Khalaf BH, et al. Effects of melatonin and zinc on lipid profile and renal function in type 2 diabetic patients poorly controlled with metformin. J Pineal Res. 2006;41(2):189–93.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge the use of large language models, for improving the written English of the manuscript. These tools were not employed in the research or data interpretation of this work. All figures were made using BioRender.com

Funding

Open Access funding enabled and organized by Projekt DEAL. This research did not receive any external funding.

Author information

Authors and Affiliations

Authors

Contributions

M.T.S. wrote the main text and prepared the figures. L.R. concepted the review and edited the text, content and figures. All authors reviewed the final manuscript.

Corresponding author

Correspondence to Lothar Rink.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schulz, M.T., Rink, L. Zinc deficiency as possible link between immunosenescence and age-related diseases. Immun Ageing 22, 19 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12979-025-00511-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12979-025-00511-1

Keywords