Skip to content
Publicly Available Published by De Gruyter November 16, 2015

The influence of the trace element zinc on the immune system

  • Claudia Schubert , Karina Guttek , Annegret Reinhold , Kurt Grüngreiff and Dirk Reinhold EMAIL logo
From the journal LaboratoriumsMedizin

Abstract

Clinical and experimental examinations showed a close relationship between zinc as an essential trace element and the immune system. Thus, cellular and humoral components from both the innate and the adaptive immune system are affected by zinc. Human zinc deficiencies are frequently connected with disturbed immune functions. Controlled zinc substitution results in a normalization of zinc serum levels, zinc homeostasis, and the immunological parameters. As shown in in vitro experiments, low zinc concentrations stimulate functional parameters of immune cells, but high zinc concentrations are suppressive or cytotoxic for these cells. Recently, the immunosuppressive effect of zinc was demonstrated in animal models of T-cell-dependent autoimmune diseases, like experimental autoimmune encephalomyelitis. Moreover, decreased serum/plasma zinc concentrations have been detected in patients with different autoimmune diseases. Prospective studies should verify the possibility of controlled immunosuppressive zinc therapies for these diseases.

Reviewed Publication:

Sack U. Conrad K.


Introduction

The essential trace element zinc is indispensable for many biological processes in humans, animals and plants. Zinc acts as a regulator or coenzyme of more than 300 enzymes, is a component of transcription factors, acts as an antioxidant, affects the stability of biological membranes and the assembly of multi-protein complexes, such as the T-cell receptor. In addition, zinc is involved in the synthesis of DNA and RNA as well as protein synthesis, regulates the expression of hormones and hormone receptors, and is important for the metabolism of neurotransmitters, growth, sex and thyroid hormones, as well as for the storage of insulin in the pancreas. This trace element is also ascribed a crucial role in the maintenance of sensory functions and immune homeostasis [1–4].

Zinc is a regulatory element or cofactor of many enzymes, including various dehydrogenases, carboanhydrases, matrix metalloproteinases, the angiotensin-converting enzyme, alkaline phosphatase, as well as DNA and RNA polymerases. It serves catalytic, co-catalytic and enzyme-structure-supporting functions [1, 2, 5]. By participating in cellular phosphorylation and dephosphorylation processes, zinc is able to act as an intracellular signaling molecule and thus modify the action of growth factors, hormones and cytokines [6, 7]. Various research groups have also discussed a direct role of zinc, as an intracellular “second messenger” [8]. Overall, these characteristics underline the essential role of zinc in growth, as well as in the development and maintenance of biological functions.

The average total zinc content of an adult is about 2–3 g [1]. The bones and muscles contain about 85% of the total body zinc, liver and skin about 11%, with the remaining 4% being found in other organs and tissues, especially in the prostate, testes, eyes, brain, heart, and pancreas. Human serum or plasma accounts only for approximately 0.1% of the total zinc. Approximately 99% of the trace element is located in the body cells. Fifty percent of this can be detected in the cytosol, and 30%–40% in the nucleus; the remainder is membrane-bound [1, 9]. The intracellular zinc homeostasis is closely connected to the survival and development of cells, i.e. to the regulation of the cell cycle, the proliferation, differentiation and apoptosis [10, 11].

The regulation of the free and thus biologically active zinc ions in the cell plays a critical role in the development and progression of disease. This zinc homeostasis is controlled by numerous proteins. Unlike iron, which is mainly bound to heme, zinc maintains interactions with approximately 3000 human proteins [12]. Metallothionein and zinc transporters are the main components behind the maintenance of cellular zinc homeostasis. Metallothioneins are important for the resorption and storage of zinc. They, therefore, protect against metallotoxicity and oxidative stress. Zinc transporters ensure, in particular, the influx (ZIP transporters) and efflux (ZnT transporters) through cellular membranes [13]. Disorders of the zinc metabolism also affect mitochondrial function and can cause DNA damage.

To achieve a zinc homeostasis, the German Society for Nutrition, taking into account an average absorption rate of 30% as well as mandatory and optional zinc losses, has recommended a daily oral intake of 7 mg of zinc for women and 10 mg for men [14]. In particular, animal-based foods like poultry, beef, pork or dairy products are good sources of zinc and have a good bioavailability. Plant-based foods usually contain only small amounts of zinc and, due to a high content of phytate (phytic acid), can reduce zinc absorption significantly [1–3].

According to the World Health Organization (WHO), zinc deficiency is the fifth most common cause of high morbidity and mortality in developing countries [15–17]. But in industrialized countries of the West, too, it is assumed that approximately 40% of the population suffer from marginal, latent zinc deficiency [1]. In addition, in those countries, zinc deficiency is considered responsible for the pathogenesis of chronic inflammatory diseases such as diabetes mellitus, atherosclerosis, chronic liver, kidney, pancreatic and intestinal diseases, as well as rheumatic diseases.

A rapid increase in knowledge about the role of zinc in the physiological processes in various tissues and organs was achieved in the last two decades through the application of modern immunological, pharmacological, molecular-biological and physicochemical methods and procedures. Thus, close links between the trace element zinc and the immune system have been shown through clinical observations and experimental studies in the past [18–21]. Zinc, through its functional diversity, i.e. the regulation of many enzymes, transcription factors and zinc-binding proteins, influences components of both cellular and humoral immunity. The trace element affects the function and activity of cells of the innate and the adaptive immune system, such as neutrophils, granulocytes, monocytes/macrophages, NK-cells, dendritic cells as well as T- and B-lymphocytes. In addition, zinc acts on cytokine production, the activity of the complement system and antibody production [18–21].

Severe zinc deficiency in humans and zinc deficiency induced in animal experiments are associated with impaired immune cell function and consequently with secondary immunodeficiency. A number of clinical studies have shown that in those deficiency situations controlled zinc substitution results in a normalization of immunological parameters.

Depending on the cell-culture system used, low in vitro concentrations of zinc can have a stimulating effect on different functional parameters of immune cells of healthy test subjects; higher zinc concentrations, however, are suppressive or cytotoxic to these cells [2, 22, 23].

As human serum or plasma contains only approximately 0.1% of the total zinc content, the significance of an analysis of the respective zinc concentration is not optimal. Clearly diminished serum/plasma zinc concentrations have been detected in connection with a number of autoimmune diseases. Furthermore, data from animal studies suggest that in such diseases activated autoreactive T-cells might represent the target structure of a controlled zinc therapy. These aspects and issues will be discussed in more detail in the following sections.

Impact of zinc deficiency on the innate and adaptive immune system in vivo

Zinc deficiency can have alimentary (general malnutrition, phytate-rich diet or parenteral nutrition), iatrogenic (e.g. glucocorticoid therapy, contraceptives or penicillamine) or genetic (sickle cell anemia, acrodermatitis enteropathica) causes, as well as be brought on by absorption disorders (chronic inflammatory bowel disease, pancreatic insufficiency), increased zinc demand (during pregnancy, strong growth, chronic diseases, competitive sports) or increased zinc excretion (renal failure, liver cirrhosis) [1–3, 24]. Clinical symptoms of zinc deficiency can include diarrhea, increased susceptibility to infection, impaired wound healing, hair loss, dermatitis, decreased appetite, loss of taste, as well as impaired growth and reproductive capacity [2, 3].

Zinc homeostasis disorders affect both the innate and the adaptive immune system [17, 25]. Thus, zinc deficiency situations lead to decreased chemotaxis of neutrophils [26, 27]. Impaired or reduced activities and functions of these immune cells have been described in connection with zinc deficiency after parenteral nutrition and patients with Down syndrome [28–30]. In addition, it has been shown that zinc deficiency reduces the activity of NADPH oxidase of the neutrophils, resulting in a reduced formation of reactive oxygen species and reduced killing ability [31, 32]. Interestingly, the involvement of the trace element zinc in the regulation of the release of net-like extracellular structures, so-called “neutrophil extracellular traps” (NETs), has been described recently [33].

Furthermore, it has been demonstrated that zinc deficiency in vivo induces a reduced adhesion and chemotaxis of monocytes as well as a disturbance of macrophage maturation and activity [24–26]. It is interesting to note that an experimentally induced zinc deficiency in human monocytes produced both an increased elimination of bacterial pathogens by means of phagocytosis and respiratory burst and an inhibition of IL-6 and TNF-α production [34]. In contrast, studies on the monocyte/macrophage cell line HL60 have shown that zinc deficiency can induce a stimulation of the mRNA of the cytokines TNF-α, IL-1β and IL-8-mRNA [35].

Zinc deficiency also affects NK-cells. This leads to a decreased NK-cell count in peripheral blood and impaired functional activity of NK-cells. Thus, a reduced chemotaxis and decreased lysis of virus-infected cells or tumor cells have been detected [36, 37]. Prasad et al. [38] have been able to show that both tumor patients with zinc deficiency and healthy people with zinc deficiency exhibit diminished NK-cell activity due to eating habits, or in healthy people, due to experimentally-induced zinc deficiency (diet). Reduced NK-cell counts and NK-cell function have also been reported for elderly subjects with zinc deficiency [39]. A decreased number of dendritic cells (including Langerhans cells of the skin) has been detected both in patients with zinc deficiency and in an experimental murine system [40].

Deviations of zinc homeostasis, within the framework of the adaptive immune system, affect particularly strongly the formation, maturation and function of T-cells [41]. In the thymus, thymocytes (pre-T-lymphocytes) mature to T-lymphocytes, facilitated by the peptide hormone thymulin. This peptide hormone formed by thymic epithelial cells plays a key role in T-cell maturation in the thymus and requires zinc as an important structural element in order to be biologically active [42]. Consequently, zinc deficiency has a negative effect on the T-cell maturation in the thymus. As has been shown in mouse models, zinc deficiency causes thymic atrophy with an approximately 50% reduction in thymocyte counts [41, 43]. Under these deficient conditions, there is also an increased rate of apoptosis in the thymus, as has been demonstrated [44].

Furthermore, due to zinc deficiency situations in vivo, a decreased ability of mononuclear cells (MNC) from peripheral blood for mitogen-induced T-cell proliferation (lymphocyte transformation test) or cytokine production (for example, IL-2, IFNγ) has been observed. This has been confirmed for deficient conditions of the trace element after parenteral nutrition [28], in patients with liver cirrhosis [45, 46], in hemodialysis or peritoneal dialysis patients [47], in individuals with Down syndrome [30], in tumor patients and in subjects with zinc deficiency due to dietary habits (e.g. vegetarians) or after going on a diet [38].

Moreover, it has been shown that zinc also influences T-cell differentiation processes. As has been reported, children with diarrhea-caused zinc deficiency exhibit a reduced number of naive T-cells [48]. In addition, the CD4+/CD8+-T-cell ratio is reduced, in particular, by a decrease in the number of CD4+ T-cells if there is a deficit of this biometal [49]. Likewise, there is a Th1/Th2 imbalance with a reduced number of Th1-T-cells and decreased Th1 cytokine production (IL-2, IFN-γ, TNF-α), whereas the Th2 cells are affected to a minor degree [49, 50].

Apart from this, zinc deficiency causes B-cell maturation and T-cell-dependent antibody production to be reduced [25].

In the synopsis of the data one can see that zinc deficiency conditions with different causes impair immune cell functions and thus produce secondary immune deficiency (refer to Figure 1). In a series of clinical studies and animal-experiment studies, it has been shown that, in connection with zinc deficiency, a controlled zinc substitution results in a normalization of the body’s zinc homeostasis and immunological parameters described (see also “Substitution of zinc deficiency in vivo”).

Figure 1: Relationship between zinc deficiency, the dysfunction of the immune system and associated secondary immunodeficiency.
Figure 1:

Relationship between zinc deficiency, the dysfunction of the immune system and associated secondary immunodeficiency.

Influence of zinc on the function of immune cells in healthy test subjects in vitro

The influence of zinc compounds or commercial zinc preparations on the functions of the immune cells of healthy test subjects was studied intensively in different cell-culture systems in the past (especially MNC, isolated monocytes and isolated T-cells) [1, 18–21].

Preincubation of human monocytes with ZnCl2 concentrations (20–40 μM), non-toxic for these cells, reduced the lipopolysaccharide (LPS)-induced monocyte activation (adherence, formation of reactive oxygen metabolites, IL-1β mRNA expression) [51]. Incubation of MNC from peripheral blood, that is, a cell mixture of monocytes, T-cells, B-cells and NK0 cells, with ZnSO4 alone, however, led to an increased release of the cytokines IL-1β, IL-6, TNF-α and IFN-γ [52, 53]. In combination with LPS, ZnSO4 (10–100 μM) induced in MNC also increased IL-1β- and IFN-γ production [54, 55]. The mechanism for this seems to lie in the influence on the mobility of the LPS chains by means of zinc [56]. Incubation of MNC with suboptimal concentrations of LPS and zinc also stimulated the IL-1β release [57]. Interestingly, low levels of zinc have no stimulatory effect on the mitogen-stimulated cytokine release of human MNC. In contrast, when stimulated with superantigens, cytokine production was impaired by ZnSO4 [55].

Hayashi et al. [58] used Jurkat cells, a human T-cell line, to observe a zinc-dependent inhibition of IFN-γ production in the presence of elevated zinc concentrations.

Our research group has been able to document that ZnCl2, ZnO or ZnSO4, in a concentration range from 100 μM to 200 μM, impairs DNA synthesis (proliferation) and the production of different T-cell cytokines (IL-2, IL-6, IL-10) of mitogen-stimulated MNC and isolated T-lymphocytes in a dose-dependent manner [22]. Prasad et al. [59] and Sprietsma [60] first discussed an effect of zinc on the regulation of T-cell differentiation into Th1 and Th2 cells, in which, in particular, a decrease in Th1 cells occurs. Campo et al. [61] demonstrated that ZnSO4 impairs the production of the Th1 cytokine IFN-γ of human MNC, stimulated by a mixed lymphocyte culture, in a dose-dependent manner. A biphasic effect of zinc on the IFN-γ production of human activated T-cells with a maximum stimulation of 3 μM and incipient inhibition from 25 μM zinc has been reported by Aydemir et al. [62].

Mouse models have shown further that higher zinc concentrations, by affecting the IL-6/STAT3 signaling cascade, also impair the development of Th17 cells, which play a key role in the pathogenesis of autoimmune diseases. In the process, preventive zinc additions via direct STAT3 binding inhibited the maturation of naive CD4+ T-cells into Th17 cells. This led to the loss of the α-helical structure and development of STAT3 with concomitant loss of activity [63].

Our own studies on the effect of zinc hydrogen aspartate (Zink-HA, Unizink 50), a commercially available zinc preparation, on the proliferation and cytokine release of isolated human T-cells confirmed for the first time that Zink-HA inhibited the proliferation (DNA synthesis) of human anti-CD3/CD28 antibodies and mitogen-stimulated T-cells significantly in non-cytotoxic concentrations of up to 150 μM. Furthermore, Zink-HA suppressed the production of various Th1 (IL-2, IFN-γ, TNF-α), Th2 (IL5) and Th17 cytokines (IL-17, GM-CSF) in a dose-dependent manner. These results were confirmed on anti-CD3 antibody or mitogen-stimulated mouse splenocytes [64, 65].

We observed, moreover, that depending on the cell culture system, very high zinc concentrations (200–500 μM) were cytotoxic to pokeweed-mitogen (PWM)-stimulated human MNC and T-cells [22, 64, 65]. Other authors have also reported the toxic effect of high concentrations of zinc on lymphocytes and monocytes in vitro [66].

One possible explanation for the sometimes conflicting effects on MNC, monocytes and T-cells could be a concentration-dependent zinc effect on various signaling pathways of these cells and thus, inter alia, on cytokine production [20].

Consequently, stimulating and inhibiting effects can be created by different stimulations (LPS, mitogens, antibodies). The use of different zinc compounds (ZnO, ZnCl2, ZnSO4, Zink-HA) does not seem to have any influence.

In considering these results, it should be noted that the respective cell-culture systems and, in particular, the cell-culture media used have a decisive influence on the concentration of “free zinc ions”, which act on immune cells and induce the effects variously observed. Haase et al. demonstrated recently that components of cell-culture media, such as albumin or human or calf serum, significantly influenced the effect of zinc compounds in cell-culture experiments [67].

Zinc status in patients with autoimmune diseases

The quantification of zinc concentration in serum, whole blood, plasma (lithium heparinate) or 24 h urine should be carried out in approved clinical-chemistry laboratories and by flame atomic absorption spectroscopy, plasma emission spectroscopy or direct electro-thermal atomic absorption spectroscopy. Photometric methods are insufficient as a method of determination [68, 69].

Non-metal collection systems should be used for collecting blood samples. Hemolysis and the associated release of zinc from erythrocytes, for example, due to excessive congestion or aspiration during the collection of the blood sample must be avoided at all cost. Since the zinc concentration in the blood decreases after food intake, the blood should be collected on an empty stomach.

The reference range for adults is 9–18 μmol/L (serum), and 9–22 μmol/L for women and 12–26 μmol/L for men (plasma) [1, 68].

In recent years, attempts have been made by several research groups to find more suitable parameters for reliable statements about the human “zinc status” by analyzing the concentrations of zinc in cells (especially in erythrocytes, leukocytes and lymphocytes). Unfortunately, these efforts have not been successful so far. Attempts to infer from the expression of individual zinc-transporter proteins the “zinc status” of a patient have not been successful either [1, 69].

Due to the very small proportion of the total zinc content that human serum or plasma represents (only about 0.1%), the validity of an analysis of serum or plasma zinc concentrations is still a point of discussion and/or criticism. Still, serum zinc is currently considered the best biochemical indicator of the “zinc status” [70].

Thus, for the aforementioned zinc-deficiency conditions with alimentary, iatrogenic and genetic causes, it was possible to detect clearly reduced zinc serum/plasma concentrations for a number of chronic diseases, such as various autoimmune diseases [1, 25].

Autoimmune diseases affect approximately 5%–7% of the population and are, therefore, in terms of frequency, the third most common after cardiovascular diseases and cancer. In addition to a genetic predisposition, environmental variables and physical or psychological stress, other factors such as hormonal status, age and immune status appear to play a crucial role in the pathogenesis of autoimmune diseases.

The most common autoimmune diseases include autoimmune thyroid disease, rheumatoid arthritis (RA), celiac disease, type-I diabetes mellitus, systemic lupus erythematosus (SLE), autoimmune liver disease and multiple sclerosis (MS).

Various authors have observed reduced zinc concentrations in the serum or plasma of patients with RA compared with healthy subjects [71–75]. Yazar et al. [76], however, found no differences in the plasma zinc concentrations of patients with RA. Patients with celiac disease [77–80], type-I diabetes mellitus [81, 82], SLE [83, 84], autoimmune hepatitis [85, 86], primary biliary cirrhosis [87, 88] and pemphigus vulgaris [89] also exhibited decreased serum zinc concentrations.

Several authors have described decreased zinc concentrations in the plasma [90–92] or serum [93] of MS patients compared to healthy subjects. These findings are in contrast to observations made by Dore-Duffy et al. [94], who reported slightly elevated plasma zinc levels in a study of 68 MS patients compared to a healthy control group. Recently, Ghazavi et al. [95] analyzed the zinc concentrations in the serum of 60 MS patients. They also found significantly reduced zinc levels in the sera of the patients, as compared to those of a corresponding control group.

In conclusion, despite the existing problems related to the analysis/diagnosis of a zinc deficit in a variety of autoimmune diseases, there is a clear reduction of serum or plasma zinc levels. These results point to a potential regulatory role of zinc in the pathogenesis of autoimmune diseases. Immunosuppressive zinc therapy could potentially represent a novel therapeutic approach for such diseases (see also “Immunosuppressive zinc therapy in connection with autoimmune diseases in vivo”).

Substitution of zinc deficiency in vivo

Upon occurrence of clinical zinc deficiency symptoms, the serum or plasma concentrations of the trace element should be analyzed. Zinc substitution is indicated in principle for all forms of laboratory-proven zinc deficiency. This therapy should always be conducted under the control of “serum zinc” in intervals of 6–8 weeks [96].

Zinc substitution is absolutely indicated in patients with acrodermatitis enteropathica. This disease is based on a gene mutation that encodes the intestinal zinc importer ZIP 4. This leads to a severe zinc deficiency syndrome, whose immunological cardinal symptoms are characterized by thymic atrophy, quantitative and functional impairment of lymphocytes, and increased susceptibility to infection. Moynahan already wrote in 1974 [97] that zinc substitution in patients with acrodermatitis enteropathica can be a life-saving measure.

In internal medicine, chronic liver and intestinal diseases, as well as parenteral nutrition, are the main indications for zinc substitution. Accordingly, 70%–75% of patients with decompensated liver cirrhosis exhibit a more or less pronounced zinc deficiency. In various studies, an improvement of liver function, nutritional status and hepatic encephalopathy, as well as a decrease of ascites and an improvement of immunological parameters have been demonstrated in these patients following controlled zinc substitution [20, 96, 98–101].

A general dosage indication of a zinc preparation that is reliable in individual cases is hardly possible. Kästner [102] recommended a dose of ten times the regular daily intake of zinc with food (up to 10 mg per day for adults), by way of a zinc salt, for the optimal treatment of acrodermatitis enteropathica. According to Prasad [103], oral zinc doses of up to 45 mg of elemental zinc per day are considered non-toxic. Various authors have pointed out that at very high zinc doses (100–300 mg of zinc per day), disorders of the immune functions can be observed [16, 104, 105]. Our research group has found that zinc concentrations higher than 500 μmol (roughly equivalent to a daily dose of 45 mg elemental zinc) induce toxic effects on immune cells, while inhibiting DNA synthesis and cytokine production [22]. Overall, clinical and animal studies have shown that controlled zinc substitution in connection with zinc deficiency results in a normalization of the body’s zinc homeostasis and immunological parameters (see Figure 2) [25].

Figure 2: Representation of the relationship between zinc deficiency and secondary immunodeficiency with controlled zinc supplementation as well as in the case of a T-cell-mediated autoimmune disease with existing zinc deficiency and controlled immunosuppressive zinc therapy.
Figure 2:

Representation of the relationship between zinc deficiency and secondary immunodeficiency with controlled zinc supplementation as well as in the case of a T-cell-mediated autoimmune disease with existing zinc deficiency and controlled immunosuppressive zinc therapy.

Immunosuppressive zinc therapy in connection with autoimmune diseases in vivo

In recent years, the use of immunosuppressive zinc therapy has been considered as well, particularly in the context of T-cell-facilitated autoimmune diseases and transplant rejection reactions [16, 64, 65, 106]. This additional form of controlled zinc therapy is shown in the right section of Figure 2.

It must be noted at this point that so far no clinically applied, immunosuppressive high-dose zinc therapy has been established for any autoimmune disease.

The immunosuppressive effect of zinc has been confirmed via various autoimmune animal models in recent years. Tran et al. [107] and Ohkawara et al. [108] described a therapeutic effect of zinc compounds in connection with experimental colitis induced by dextran sulfate sodium (DSS).

Kitabayashi et al. [63] have shown a preventive effect of zinc in collagen-induced arthritis.

In an animal model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE), Penkowa et al. [109] and Kitabayashi et al. [63] observed a preventive effect on the severity of the disease. Our group also documented that Zink-HA (Unizink), a commercially available zinc preparation with excellent bioavailability, can reduce, preventively and therapeutically by both intraperitoneal and oral administration, the clinical symptoms of active EAE in SJL/J-mice significantly [64, 65]. In such T-cell-facilitated autoimmune diseases, thus, activated autoreactive T-cells could represent the target structure of an immunosuppressive zinc therapy. This form of therapy, too, should be conducted under close control of the “serum zinc level”.

Outlook

Overall, it can be said that zinc is an important trace element for the homeostasis of the immune system. A large number of clinical symptoms are associated with a deficiency of this biometal. Currently, the validity of an analysis of the serum or plasma zinc levels is not deemed optimal in this context. For this reason, further studies are needed to arrive at standardized statements about the “zinc status” of a person.

Furthermore, one should examine the extent by which the application spectrum of controlled zinc substitution could be expanded. In addition, future clinical studies should investigate the possible use of controlled immunosuppressive zinc therapy in connection with T-cell-facilitated autoimmune diseases, such as MS.

Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

Research funding: None declared.

Employment or leadership: None declared.

Honorarium: None declared.

Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.


Correspondence: Prof. Dr. med. Dirk Reinhold, Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany, Tel.: +0391-6715857, Fax: +0391-6715852

References

1. Rink L. Why investigate zinc? In: Rink L, editor. Zinc in human health. Amsterdam: IOS Press, 2011:3–6.Search in Google Scholar

2. Grüngreiff K, Reinhold D, editors. Zink: Bedeutung in der täglichen Praxis. 1. Auflage, Hessdorf-Klebheim: J Hartmann-Verlag, 2008:96.Search in Google Scholar

3. Grüngreiff K, editor. Zink und Leber. 7. Auflage, Freiburg: Dr. Falk Pharma GmbH, 2012:84.Search in Google Scholar

4. Prasad AS. Discovery of zinc deficiency in humans and its impact fifty years later. In: Rink L, editor. Zinc in human health. Amsterdam: IOS Press, 2011:7–28.Search in Google Scholar

5. Chasapis CT, Loutsidou AC, Spiliopoulou CA, Stefanidou ME. Zinc and human health: an update. Arch Toxicol 2012;86:521–34.10.1007/s00204-011-0775-1Search in Google Scholar

6. Beyersmann D, Haase H. Functions of zinc in signaling, proliferation and differentiation of mammalian cells. BioMetals 2001;14:331–41.10.1023/A:1012905406548Search in Google Scholar

7. Haase H, Rink L. Functional significance of zinc-related signaling pathways in immune cells. Annu Rev Nutr 2009;29:133–52.10.1146/annurev-nutr-080508-141119Search in Google Scholar

8. Yamasaki S, Sakata-Sogawa K, Hasegawa A, Suzuki T, Kabu K, Sato E, et al. Zinc is a novel intracellular second messenger. J Cell Biol 2007;177:637–45.10.1083/jcb.200702081Search in Google Scholar

9. Prasad AS. Zinc: role in immunity, oxidative stress and chronic inflammation. Curr Opin Clin Nutr Metab Care 2009;12:646–52.10.1097/MCO.0b013e3283312956Search in Google Scholar

10. Maret W. Human zinc biochemistry. In: Rink L, editor. Zinc in human health. Amsterdam: IOS Press, 2011:45–62.Search in Google Scholar

11. Huber KL, Hardy JA. Mechanism of zinc-mediated inhibition of caspase-9. Protein Sci 2012;21:1056–65.10.1002/pro.2090Search in Google Scholar

12. Maret W. Zinc and human disease. Met Ions Life Sci 2013;13:389–414.10.1007/978-94-007-7500-8_12Search in Google Scholar

13. Cousins RJ, Lichten LA. Zinc transporters. In: Rink L, editor. Zinc in human health. Amsterdam: IOS Press, 2011:136–162.Search in Google Scholar

14. Deutsche Gesellschaft für Ernährung e.V., Referenzwerte für die Nährstoffzufuhr Umschau Buchverlag, 2013.Search in Google Scholar

15. World Health report 2002 – Reducing Risks, Promoting Healthy Life. Geneva: WHO, 2002:1–250.Search in Google Scholar

16. Prasad AS. Zinc in growth and development and spectrum of human zinc deficiency. J Am Coll Nutr 1988;7:377–84.10.1080/07315724.1988.10720255Search in Google Scholar

17. Yakoob MY, Theodoratou E, Jabeen A, Imdad A, Eisele TP, Ferguson J, et al. Preventive zinc supplementation in developing countries: impact on mortality and morbidity due to diarrhea, pneumonia and malaria. BMC Public Health 2011;11:S23.10.1186/1471-2458-11-S3-S23Search in Google Scholar

18. Wellinghausen N, Kirchner H, Rink L. The immunobiology of zinc. Immunol Today 1997;18:519–21.10.1016/S0167-5699(97)01146-8Search in Google Scholar

19. Rink L, Haase H. Zinc homeostasis and immunity. Trends Immunol 2007;28:1–4.10.1016/j.it.2006.11.005Search in Google Scholar

20. Haase H, Rink L. Zinc signals and immune function. Biofactors 2014;40:27–40.10.1002/biof.1114Search in Google Scholar

21. Haase H, Rink L. Multiple impacts of zinc on immune function. Metallomics 2014;6:1175–80.10.1039/c3mt00353aSearch in Google Scholar

22. Reinhold D, Ansorge S, Grüngreiff K. Zinc regulates DNA synthesis an IL-2, IL-6, and IL-10 production of PWM-stimulated PBMC and normalizes the periphere cytokine concentration in chronic liver disease. J Trace Elem Exp Med 1997;10:19–27.10.1002/(SICI)1520-670X(1997)10:1<19::AID-JTRA3>3.0.CO;2-#Search in Google Scholar

23. Maywald M, Rink L. Zinc homeostasis and immunosenescence. J Trace Elem Med Biol 2015;29:24–30.10.1016/j.jtemb.2014.06.003Search in Google Scholar

24. Guilbert JJ. The world health report 2002 – reducing risks, promoting healthy life. Educ Health (Abingdon) 2003;16:230.10.1080/1357628031000116808Search in Google Scholar

25. Bonaventura P, Benedetti G, Albarède F, Miossec P. Zinc and its role in immunity and inflammation. Autoimmun Rev 2014;14:280–8.10.1016/j.autrev.2014.11.008Search in Google Scholar

26. Shankar AH, Prasad AS. Zinc and immune function: the biological basis of altered resistance to infection. Am J Clin Nutr 1998;68:447S–463S.10.1093/ajcn/68.2.447SSearch in Google Scholar

27. Ibs KH, Rink L. Zinc-altered immune function. J Nutr 2003;133:1452S–6S.10.1093/jn/133.5.1452SSearch in Google Scholar

28. Fan PC, Teng RJ, Chou CC, Wu TJ, Tsou Yau KI, Hsieh KH. Impaired immune function in a premature infant with zinc deficiency after total parenteral nutrition. Acta Paediatr Sin 1996;37:364–9.Search in Google Scholar

29. Licastro F, Chiricolo M, Mocchegiani E, Fabris N, Zannoti M, Beltrandi E, et al. Oral zinc supplementation in Down’s syndrome subjects decreased infections and normalized some humoral and cellular immune parameters. J Intellect Disabil Res 1994;38:149–62.10.1111/j.1365-2788.1994.tb00370.xSearch in Google Scholar

30. Stabile A, Pesaresi MA, Stabile AM, Pastore M, Sopo SM, Ricci R, et al. Immunodeficiency and plasma zinc levels in children with Down’s syndrome: a long-term follow-up of oral zinc supplementation. Clin Immunol Immunopathol 1991;58:207–16.10.1016/0090-1229(91)90137-YSearch in Google Scholar

31. 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;15:321–7.10.1002/1522-7243(200009/10)15:5<321::AID-BIO605>3.0.CO;2-OSearch in Google Scholar

32. DeCoursey TE, Morgan D, Cherny VV. The voltage dependence of NADPH oxidase reveals why phagocytes need proton channels. Nature 2003;422:531–4.10.1038/nature01523Search in Google Scholar PubMed

33. Hasan R, Rink L, Haase H. Zinc signals in neutrophil granulocytes are required for the formation of neutrophil extracellular traps. Innate Immun 2013;19:253–64.10.1177/1753425912458815Search in Google Scholar PubMed

34. 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;6:1288–95.10.1039/c4mt00051jSearch in Google Scholar PubMed

35. Bao B, Prasad AS, Beck FW, Godmere M. Zinc modulates mRNA levels of cytokines. Am J Physiol Endocrinol Metab 2003;285:E1095–102.10.1152/ajpendo.00545.2002Search in Google Scholar PubMed

36. Rajagopalan S, Winter CC, Wagtmann N, Long EO. The Ig-related killer cell inhibitory receptor binds zinc and requires zinc for recognition of HLA-C on target cells. J Immunol 1995;155:4143–6.10.4049/jimmunol.155.9.4143Search in Google Scholar

37. Rajagopalan S, Long EO. Zinc bound to the killer cell-inhibitory receptor modulates the negative signal in human NK cells. J Immunol 1998;161:1299–305.10.4049/jimmunol.161.3.1299Search in Google Scholar

38. 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;109:68–77.Search in Google Scholar

39. 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;71:590–8.10.1093/ajcn/71.2.590Search in Google Scholar PubMed

40. 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;122:722–32.10.1172/JCI58618Search in Google Scholar PubMed PubMed Central

41. Fraker PJ, King LE. Reprogramming of the immune system during zinc deficiency. Annu Rev Nutr 2004;24:277–98.10.1146/annurev.nutr.24.012003.132454Search in Google Scholar PubMed

42. Sherman AR. Zinc, copper, and iron nutriture and immunity. J Nutr 1992;122:604–9.10.1093/jn/122.suppl_3.604Search in Google Scholar PubMed

43. 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;24:494–502.10.1080/07315724.2005.10719495Search in Google Scholar PubMed

44. Fraker PJ. Roles for cell death in zinc deficiency. J Nutr 2005;135:359–62.10.1093/jn/135.3.359Search in Google Scholar PubMed

45. Badulici S, Chirulescu Z, Chirila P, Chirila M, Rosca A. Treatment with zincum metallicum CH5 in patients with liver cirrhosis. Preliminary study. Rom J Intern Med 1994;32:215–9.Search in Google Scholar

46. Vega Robledo GB, Guinzberg AL, Ramos García C, Ortiz Ortiz L. Patients with hepatic cirrhosis: altered lymphocyte response to mitogens and its relation with plasmatic zinc, albumin and transferrin. Arch Med Res 1994;25:5–9.Search in Google Scholar

47. Shu KH, Lu YS, Chen CH, Chen DC, Lee SH, Lian JD. Lymphocyte proliferation in uremic patients: correlation with zinc status. J Formos Med Assoc 1993;92:1017–20.Search in Google Scholar

48. Sheikh A, Shamsuzzaman S, Ahmad SM, Nasrin D, Nahar S, Alam MM, et al. Zinc influences innate immune responses in children with enterotoxigenic Escherichia coli-induced diarrhea. J Nutr 2010;140:1049–56.10.3945/jn.109.111492Search in Google Scholar

49. 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:E1002–7.10.1152/ajpendo.1997.272.6.E1002Search in Google Scholar

50. Prasad AS. Effects of zinc deficiency on Th1 and Th2 cytokine shifts. J Infect Dis 2000;182:S62–8.10.1086/315916Search in Google Scholar

51. Koropatnick J, Zalups RK. Effect of non-toxic mercury, zinc or cadmium pretreatment on the capacity of human monocytes to undergo lipopolysaccharide-induced activation. Br J Pharmacol 1997;120:797–806.10.1038/sj.bjp.0700975Search in Google Scholar

52. Driessen C, Hirv K, Rink L, Kirchner H. Induction of cytokines by zinc ions in human peripheral blood mononuclear cells and separated monocytes. Lymphokine Cytokine Res 1994;13:15–20.Search in Google Scholar

53. Wellinghausen N, Driessen C, Rink L. Stimulation of human peripheral blood mononuclear cells by zinc and related cations. Cytokine 1996;8:767–71.10.1006/cyto.1996.0102Search in Google Scholar

54. Wellinghausen N, Schromm AB, Seydel U, Brandenburg K, Luhm J, Kirchner H, et al. Zinc enhances lipopolysaccharide-induced monokine secretion by alteration of fluidity state of lipopolysaccharide. J Immunol 1996;157:3139–45.10.4049/jimmunol.157.7.3139Search in Google Scholar

55. Driessen C, Hirv K, Kirchner H, Rink L. Zinc regulates cytokine induction by superantigens and lipopolysaccharide. Immunology 1995;84:272–7.Search in Google Scholar

56. Wellinghausen N, Fischer A, Kirchner H, Rink L. Interaction of zinc ions with human peripheral blood mononuclear cells. Cell Immunol 1996;171:255–61.10.1006/cimm.1996.0201Search in Google Scholar

57. Driessen C, Hirv K, Kirchner H, Rink L. Divergent effects of zinc on different bacterial pathogenic agents. J Infect Dis 1995;171:486–9.10.1093/infdis/171.2.486Search in Google Scholar

58. Hayashi K, Ishizuka S, Yokoyama C, Hatae T. Attenuation of interferon-gamma mRNA expression in activated Jurkat T cells by exogenous zinc via down-regulation of the calcium-independent PKC-AP-1 signaling pathway. Life Sci 2008;83:6–11.10.1016/j.lfs.2008.04.022Search in Google Scholar

59. Prasad AS. Zinc and immunity. Mol Cell Biochem 1998;188:63–9.10.1023/A:1006868305749Search in Google Scholar

60. Sprietsma JE. Zinc-controlled Th1/Th2 switch significantly determines development of diseases. Med Hypotheses 1997;49:1–14.10.1016/S0306-9877(97)90244-9Search in Google Scholar

61. Campo CA, Wellinghausen N, Faber C, Fischer A, Rink L. Zinc inhibits the mixed lymphocyte culture. Biol Trace Elem Res 2001;79:15–22.10.1385/BTER:79:1:15Search in Google Scholar

62. Aydemir TB, Blanchard RK, Cousins RJ. Zinc supplementation of young men alters metallothionein, zinc transporter, and cytokine gene expression in leukocyte populations. Proc Natl Acad Sci USA 2006;103:1699–704.10.1073/pnas.0510407103Search in Google Scholar

63. Kitabayashi C, Fukada T, Kanamoto M, Ohashi W, Hojyo S, Atsumi T, et al. Zinck suppresses Th17 development via inhibition of STAT3 activation. Int Immunol 2010;22:375–86.10.1093/intimm/dxq017Search in Google Scholar

64. 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;25:529–39.10.1007/s10534-012-9532-zSearch in Google Scholar

65. Schubert C, Guttek K, Grüngreiff K, Thielitz A, Bühling F, Reinhold A, et al. Oral zinc aspartate treats experimental autoimmune encephalomyelitis. Biometals 2014;27:1249–62.10.1007/s10534-014-9786-8Search in Google Scholar

66. Steffensen IL, Mesna OJ, Andruchow E, Namork E, Hylland K, Andersen RA. Cytotoxicity and accumulation of Hg, Ag, Cd, Cu, Pb and Zn in human peripheral T and B lymphocytes and monocytes in vitro. Gen Pharmacol 1994;25:1621–33.10.1016/0306-3623(94)90364-6Search in Google Scholar

67. Haase H, Hebel S, Engelhardt G, Rink L. The biochemical effects of extracellular Zn(2+) and other metal ions are severely affected by their speciation in cell culture media. Metallomics 2015;7:97–106.10.1039/C4MT00206GSearch in Google Scholar

68. Thomas L. Labordiagnostik von Spurenelementen. In: Thomas L, editor. Labor und Diagnose – Indikation und Bewertung von Laborbefunden für die medizinische Diagnostik. Frankfurt/Main: Th-Books Verlagsgesellschaft mbH, 2012:545–579.Search in Google Scholar

69. Lowe NM, Fekete K, Decsi T. Methods of assessment of zinc status in humans: a systematic review. Am J Clin Nutr 2009;89:2040S–51S.10.3945/ajcn.2009.27230GSearch in Google Scholar

70. Krebs NF, Miller LV, Hambidge KM. Zinc deficiency in infants and children: a review of its complex and synergistic interactions. Paed Inter Child Health 2014;34:279–88.10.1179/2046905514Y.0000000151Search in Google Scholar

71. Önal S, Naziroglu M, Çolak M, Bulut V, Flores-Arce MF. Effects of different medical treatments on serum copper, selenium and zinc levels in patients with rheumatoid arthritis. Biol Trace Elem Res 2011;142:447–55.10.1007/s12011-010-8826-7Search in Google Scholar

72. Zoli A, Altomonte L, Caricchio R, Galossi A, Mirone L, Ruffini MP, et al. Serum zinc and copper in active rheumatoid arthritis: correlation with interleukin 1 beta and tumour necrosis factor alpha. Clin Rheumatol 1998;17:378–82.10.1007/BF01450895Search in Google Scholar

73. Milanino R, Frigo A, Bambara LM, Marrella M, Moretti U, Pasqualicchio M, et al. Copper and zinc status in rheumatoid arthritis: studies of plasma, erythrocytes, and urine, and their relationship to disease activity markers and pharmacological treatment. Clin Exp Rheumatol 1993;11:271–81.Search in Google Scholar

74. Afridi HI, Kazi TG, Kazi N, Talpur FN, Shah F, Naeemullah, et al. Evaluation of status of arsenic, cadmium, lead and zinc levels in biological samples of normal and arthritis patients of age groups (46–60) and (61–75) years. Clin Lab 2013;59:143–53.10.7754/Clin.Lab.2012.120126Search in Google Scholar

75. Afridi HI, Kazi TG, Kazi N, Shah F. Evaluation of status of zinc, copper, and iron levels in biological samples of normal and arthritis patients in age groups 46–60 and 61–75 years. Clin Lab 2012;58:705–17.10.7754/Clin.Lab.2012.120126Search in Google Scholar

76. Yazar M, Sarban S, Kocyigit A, Isikan UE. Synovial fluid and plasma selenium, copper, zinc, and iron concentrations in patients with rheumatoid arthritis and osteoarthritis. Biol Trace Elem Res 2005;106:123–32.10.1385/BTER:106:2:123Search in Google Scholar

77. Högberg L, Danielsson L, Jarleman S, Sundqvist T, Stenhammar L. Serum zinc in small children with coeliac disease. Acta Paediatr 2009;98:343–5.10.1111/j.1651-2227.2008.01085.xSearch in Google Scholar PubMed

78. Singhal N, Alam S, Sherwani R, Musarrat J. Serum zinc levels in celiac disease. Indian Pediatr 2008;45:319–21.Search in Google Scholar

79. Henker J, Gabsch HC. Serum zinc levels in children with celiac disease. Helv Paediatr Acta 1985;40:47–53.Search in Google Scholar

80. Jones PE, Peters TJ. Oral zinc supplements in non-responsive coeliac syndrome: effect on jejunal morphology, enterocyte production, and brush border disaccharidase activities. Gut 1981;22:194–8.10.1136/gut.22.3.194Search in Google Scholar PubMed PubMed Central

81. Jansen J, Rosenkranz E, Overbeck S, Warmuth S, Mocchegiani E, Giacconi R, et al. Disturbed zinc homeostasis in diabetic patients by in vitro and in vivo analysis of insulinomimetic activity of zinc. J Nutr Biochem 2012;23:1458–66.10.1016/j.jnutbio.2011.09.008Search in Google Scholar PubMed

82. Taylor CG. Zinc, the pancreas, and diabetes: insights from rodent studies and future directions. Biometals 2005;18:305–12.10.1007/s10534-005-3686-xSearch in Google Scholar PubMed

83. Yilmaz A, Sari RA, Gundogdu M, Kose N, Dag E. Trace elements and some extracellular antioxidant proteins levels in serum of patients with systemic lupus erythematosus. Clin Rheumatol 2005;24:331–5.10.1007/s10067-004-1028-ySearch in Google Scholar PubMed

84. Sahebari M, Abrishami-Moghaddam M, Moezzi A, Ghayour-Mobarhan M, Mirfeizi Z, Esmaily H, et al. Association between serum trace element concentrations and the disease activity of systemic lupus erythematosus. Lupus 2014;23:793–801.10.1177/0961203314530792Search in Google Scholar PubMed

85. Pereira TC, Saron ML, Carvalho WA, Vilela MM, Hoehr NF, Hessel G. Research on zinc blood levels and nutritional status in adolescents with autoimmune hepatitis. Arq Gastroenterol 2011;48:62–5.10.1590/S0004-28032011000100013Search in Google Scholar PubMed

86. Goode HF, Kelleher J, Walker BE. Relation between zinc status and hepatic functional reserve in patients with liver disease.Gut 1990;31:694–7.10.1136/gut.31.6.694Search in Google Scholar

87. Himoto T, Yoneyama H, Kurokochi K, Inukai M, Masugata H, Goda F, et al. Contribution of zinc deficiency to insulin resistance in patients with primary biliary cirrhosis. Biol Trace Elem Res 2011;144:133–42.10.1007/s12011-011-9049-2Search in Google Scholar

88. Sogawa K, Yamada T, Suzuki Y, Masaki T, Watanabe S, Uchida Y, et al. Elevation of ceruloplasmin activity involved in changes of hepatic metal concentration in primary biliary cirrhosis. Res Commun Chem Pathol Pharmacol 1994;84:367–70.Search in Google Scholar

89. Yazdanpanah MJ, Ghayour-Mobarhan M, Taji A, Javidi Z, Pezeshkpoor F, Tavallaie S, et al. Serum zinc and copper status in Iranian patients with pemphigus vulgaris. Int J Dermatol 2011;50:1343–6.10.1111/j.1365-4632.2011.04968.xSearch in Google Scholar

90. Wong EK Jr, Enomoto H, Leopold IH, Fleischer EB, Schoon DV, Fender D, et al. Plasma zinc levels in multiple sclerosis. Metab Pediatr Ophthalmol 1980;4:3–8.Search in Google Scholar

91. Williams CM, Lines CM, McKay EC. Iron and zinc status in multiple sclerosis patients with pressure sores. Eur J Clin Nutr 1988;42:321–8.Search in Google Scholar

92. Leopold IH. United States Patent 1981;4, 255, 419.Search in Google Scholar

93. Palm R, Hallmans G. Zinc and copper in multiple sclerosis. J Neurol Neurosurg Psychiatry 1982;45:691–8.10.1136/jnnp.45.8.691Search in Google Scholar

94. Dore-Duffy P, Catalanotto F, Donaldson JO, Ostrom KM, Testa MA. Zinc in multiple sclerosis. Ann Neurol 1983;14:450–4.10.1002/ana.410140409Search in Google Scholar

95. Ghazavi A, Kianbakht S, Ghasami K, Mosayebi G. High copper and low zinc serum levels in Iranian patients with multiple sclerosis: a case control study. Clin Lab 2012;58:161–4.Search in Google Scholar

96. Grüngreiff K, Reinhold D. Zinc and the liver. In: Rink L, editor. Zinc in human health. amsterdam: IOS Press, 2011:473–92.Search in Google Scholar

97. Moynahan EJ. Acrodermatitis enteropathica A lethal inherited human deficiency disorder. Lancet 1974;2:399–400.10.1016/S0140-6736(74)91772-3Search in Google Scholar

98. Bianchi GP, Marchesini G, Brizi M. Nutritional effects of oral zinc supplementation in cirrhosis. Nutr Res 2000;20:1079–89.10.1016/S0271-5317(00)00194-9Search in Google Scholar

99. Takuma Y, Nouso K, Makino Y, Hayashi M, Takahashi H. Clinical trial: oral zinc in hepatic encephalopathy. Aliment Pharmacol Ther 2010;32:1080–90.10.1111/j.1365-2036.2010.04448.xSearch in Google Scholar PubMed

100. Somi MH, Rezaeifar P, Ostad Rahimi A, Moshrefi B. Effects of low dose zinc supplementation on biochemical markers in non-alcoholic cirrhosis: a randomized clinical trial. Arch Iran Med 2012;15:472–6.Search in Google Scholar

101. Hayashi M, Ikezawa K, Ono A, Okabayashi S, Hayashi Y, Shimizu S, et al. Evaluation of the effects of combination therapy with branched-chain amino acid and zinc supplements on nitrogen metabolism in liver cirrhosis. Hepatol Res 2007;37:615–9.10.1111/j.1872-034X.2007.00095.xSearch in Google Scholar PubMed

102. Kästner H. Acrodermatitis enteropathica Danbolt-Closs. In: Zumkley H, editor. Spurenelemente in der inneren Medizin unter besonderer Berücksichtigung von Zink. Seeheim-Jugenheim: Innovations-Verlagsgesellschaft 1984:63–71.Search in Google Scholar

103. Prasad AS. Essentiality and toxicity of zinc. Scand J Work Environ Health 1993;19:134–6.Search in Google Scholar

104. Fosmire GJ. Zinc toxicity. Am J Clin Nutr 1990;51:225–7.10.1093/ajcn/51.2.225Search in Google Scholar PubMed

105. Sandström B, Cederblad A, Lindbald BS, Lonnerdal B. Acrodermatitis enteropathica, zinc metabolism, copper status, and immune function. Arch Pediatr Adolesc Med 1994;148:980–5.10.1001/archpedi.1994.02170090094017Search in Google Scholar PubMed

106. Wellinghausen N, Rink L. The significace of zinc for leukocyte biology. J Leukocyte Biol 1998;64:571–7.10.1002/jlb.64.5.571Search in Google Scholar PubMed

107. Tran CD, Ball JM, Sundar S, Coyle P, Howarth GS. The role of zinc and metallothionein in the dextran sulfate sodium-induced colitis mouse model. Dig Dis Sci 2007;52:2113–21.10.1007/s10620-007-9765-9Search in Google Scholar PubMed

108. Ohkawara T, Takeda H, Kato K, Miyashita K, Kato M, Iwanaga T, et al. Polaprezinc (N-(3-aminopropionyl)-L-histidinato zinc) ameliorates dextran sulfate sodium-induced colitis in mice. Scand J Gastroenterol 2005;40:1321–7.10.1080/00365520510023530Search in Google Scholar PubMed

109. Penkowa M, Hidalgo J. Treatment with metallothionein prevents demyelination and axonal damage and increases oligodendrocyte precursors and tissue repair during experimental autoimmune encephalomyelitis. J Neurosci Res 2003;72:574–86.10.1002/jnr.10615Search in Google Scholar PubMed


Article note:

Original German online version at: http://www.degruyter.com/view/j/labm.2015.39.issue-3/labmed-2015-0022/labmed-2015-0022.xml?format=INT. The German article was translated by Compuscript Ltd. and authorized by the authors.


Received: 2015-2-6
Accepted: 2015-2-10
Published Online: 2015-11-16

©2015 by De Gruyter

Downloaded on 12.6.2024 from https://www.degruyter.com/document/doi/10.1515/labmed-2015-0060/html
Scroll to top button