You are on page 1of 8

Archives of Biochemistry and Biophysics 611 (2016) 58e65

Contents lists available at ScienceDirect

Archives of Biochemistry and Biophysics


journal homepage: www.elsevier.com/locate/yabbi

Zinc and immunity: An essential interrelation


Maria Maares, Hajo Haase*
Department of Food Chemistry and Toxicology, Berlin Institute of Technology, Gustav-Meyer-Allee 25, D-13355 Berlin, Germany

a r t i c l e i n f o a b s t r a c t

Article history: The significance of the essential trace element zinc for immune function has been known for several
Received 1 February 2016 decades. Zinc deficiency affects immune cells, resulting in altered host defense, increased risk of
Received in revised form inflammation, and even death. The micronutrient zinc is important for maintenance and development of
16 March 2016
immune cells of both the innate and adaptive immune system. A disrupted zinc homeostasis affects these
Accepted 23 March 2016
cells, leading to impaired formation, activation, and maturation of lymphocytes, disturbed intercellular
Available online 26 March 2016
communication via cytokines, and weakened innate host defense via phagocytosis and oxidative burst.
This review outlines the connection between zinc and immunity by giving a survey on the major roles of
Keywords:
Zinc
zinc in immune cell function, and their potential consequences in vivo.
Immune system © 2016 Elsevier Inc. All rights reserved.
Cytokines
Innate immunity
Adaptive immunity

1. Introduction results in a plateau of plasma zinc, with most studies averaging


around a value of 14 mM [20].
Zinc plays an important role in the immune system, affecting A disrupted zinc homeostasis causes impaired immune func-
both innate and adaptive immune cells. This is highlighted by the tion, leading to compromised host defense and an increased risk of
effects of zinc deficiency, including thymic atrophy, lymphopenia, excessive inflammation. Deficiency and supplementation of this
impaired cellular and antibody-mediated immune responses, and essential micronutrient are important factors influencing the im-
even death [1,2]. Since the relationship between zinc deficiency and mune system. Thus, the significance of zinc for selected innate and
immune dysfunction was discovered about 50 years ago, it has been adaptive immune cells is discussed below.
widely investigated [3]. The role of zinc in immune function, its
effects on immune cells, and the underlying molecular mechanisms
have been described [4,5], including its importance as a signaling 2. Innate immune system
molecule [6e8]. Because zinc deficiency is closely linked to insuf-
ficient dietary intake or impaired resorption, especially people from Upon recognition of characteristic molecular patterns of path-
developing countries, elderly, vegetarians/vegans, and patients ogens, the innate immune system initiates an immediate immune
with some chronic diseases, such as renal insufficiency and chronic response, mainly maintained by neutrophil granulocytes, mono-
diarrhea, are affected [9e12]. Zinc supplementation can either cytes, and macrophages. They directly attack pathogens, com-
restore or even improve immune function [13e16] and has been plemented by natural killer cells, which recognize and kill virus-
used as a therapeutic treatment in chronic gastrointestinal disor- infected and tumor cells [21,22]. Additionally, innate immune
ders, renal diseases, and genetic predispositions, such as sickle cell cells release various cytokines and chemokines, soluble proteins
anemia and the zinc malabsorption syndrome acrodermatitis that are recognized by corresponding receptors on innate and
enteropathica [11]. Physiological plasma zinc levels vary around adaptive immune cells. These signals enable intercellular commu-
12e16 mM [17,18], providing an important pool for immune cells nication, which coordinates the immune response [23,24]. During
[19]. In healthy individuals, substantial dietary zinc restriction the acute phase of an infection, one systemic response that is co-
significantly decreases plasma levels, whereas elevated zinc intake ordinated by cytokines leads to decreased concentrations of several
essential trace elements in the plasma, including zinc. This process
is known as nutritional immunity. Not only leukocytes, but also
* Corresponding author. pathogens critically depend on zinc, and withdrawal of nutrients
E-mail address: Haase@TU-Berlin.de (H. Haase). aims to hinder pathogenic growth [25,26].

http://dx.doi.org/10.1016/j.abb.2016.03.022
0003-9861/© 2016 Elsevier Inc. All rights reserved.
M. Maares, H. Haase / Archives of Biochemistry and Biophysics 611 (2016) 58e65 59

Abbreviations NK natural killer cell


NKT natural killer T-cell
APC antigen presenting cell PBMC peripheral blood mononuclear cells
BCR B-cell receptor PDE cyclic nucleotide phosphodiesterases
cAMP cyclic adenosine monophosphate PKA protein kinase A
Con A Concanavalin A PKC protein kinase C
CD cluster of differentiation PMA phorbol-12-myristate 13-acetate
DC dendritic cells PMN polymorphonuclear leukocytes
EAE experimental autoimmune encephalomyelitis TH T helper cells
cGMP cyclic guanosine monophosphate T-bet T-box transcription factor
IL interleukin TCR T-cell receptor
IL-2Ra IL-2 receptor alpha chain TLR Toll-like receptor
IL-12Rb2 IL-12 receptor beta 2 subunit TNF-a tumor necrosis factor-a
IFN interferon TRIF toll/IL-1R domain-containing adapter inducing IFN-b
IRF3 IFN regulatory factor 3 TPEN N, N, N0 , N0 -tetrakis-[2-pyridylmethyl]-
KIR killer inhibitory receptors ethylenediamine
LPS lipopolysaccharide Treg regulatory T-cell
MHC major histocompatibility complex ZnT zinc transporter
MyD88 myeloid differentiation primary response gene 88 ZIP Zrt/Irt-like protein
NF-kB nuclear factor-kB

2.1. Polymorphonuclear leucocytes traps (NETs). These NETs are composed of DNA, chromatin, and
granule proteins and released by PMNs to capture and kill extra-
After invasion of pathogens or inflammation, poly- cellular pathogens. Zinc signals are involved in the formation of
morphonuclear leukocytes (PMN) (also referred to as granulocytes) NETs. Compared to untreated control cells, decreased NETosis was
migrate via adhesion and chemotaxis into the infected tissue as a observed in PMN in vitro after zinc chelation by the chelator N, N,
response to signals derived from the source of infection [27,28]. The N0 , N0 -tetrakis-[2-pyridylmethyl]-ethylenediamine (TPEN), because
most abundant PMN are neutrophil granulocytes, alongside with the ROS-dependent signal transduction cascade that leads to
eosinophils and basophiles. They provide the first line of host de- NETosis requires zinc signals [31]. Taken together, sufficient supply
fense by phagocytosis, NETosis, degranulation and secretion of of zinc enables regular function of neutrophil granulocytes,
several cytokines [27,29,30]. In addition to phagocytosis, oxidative whereas either zinc deprivation or excess impair their activity.
burst represents an important defense mechanism of activated
neutrophils. After internalization, granulocytes kill the pathogens
by generating reactive oxygen species (ROS), which is mainly 2.2. Monocytes and macrophages
mediated by high NADPH oxidase activity [21,31]. Physiological zinc
concentrations were reported to be essential for ROS generation in Monocytes represent another important cell type of innate
human leukocytes, whereas extremely high zinc concentrations immunity. They migrate into the infected tissue shortly after the
(10 mM) resulted in a strong decrease [32]. Moreover, zinc defi- PMN, where they differentiate into macrophages. Alongside with
ciency was shown to increase the amount of ROS in vitro after in- PMN, they mediate host defense via phagocytosis and oxidative
duction with phorbol-12-myristate-13-acetate (PMA) [33]. Zinc is a burst. In addition, they are antigen presenting cells (APCs) and
known inhibitor of the NADPH oxidase [34], suggesting that zinc secrete pro-inflammatory cytokines to regulate the immune
deficiency might lead to enhanced NADPH oxidase activity, thereby response [21,41]. During in vitro zinc deficiency, phagocytosis and
elevating ROS production. Yet, zinc has a multitude of other pro- cytotoxicity of monocytes increase and oxidative burst is enhanced
antioxidant functions [35,36], which may also be contributing to [21,42]. Additionally, increased maturation into macrophages was
elevated ROS levels during a shortage of zinc. observed after zinc depletion, suggesting that low zinc status pro-
During zinc deficiency, impaired function of PMN in vivo was motes differentiation of monocytes [42] (Fig. 1). Conversely, acti-
reported, resulting in decreased activity, phagocytosis (Fig. 1) and vation of these cells by the adaptive immune system is impaired
altered host defense [37,38]. Neutrophil granulocytes secrete during zinc deficiency. This is due to decreased T helper type 1
interleukin (IL)-8, a chemokine that attracts additional gran- (TH1)-cell-mediated secretion of interferon (IFN)-g [43], a TH1
ulocytes and acts as a chemoattractant for T-cells. Additionally, they cytokine of paramount importance for activation of APCs [44].
release the anti-inflammatory IL-1 receptor antagonist (IL-1ra), Moreover, zinc deficiency reduces the generation of IL-12 by
which highlights their role in regulation and resolution of the in- monocytes and macrophages [36,45]. IL-12 is an important regu-
flammatory response. Lipopolysaccharides (LPS) from the cell wall lator of TH1-cell differentiation [46], making both IL-12 and IFN-g
of gram-negative bacteria induce the release of IL-8 and IL-1ra [39]. critical cytokines for phagocytic activation by T-cells [36]. Thus,
This is impaired by zinc deprivation, indicating the importance of host defense via phagocytosis and oxidative burst is maintained
zinc for granulocyte function. Moreover, chemotaxis was observed during zinc deficiency, but ROS generation in monocytes and
to be impaired by zinc deficiency and induced by high zinc con- macrophages seems to be less dependent on adaptive immunity
centrations (500 mM) in rat granulocytes in vitro, suggesting that and cellular communication.
zinc itself acts as a chemoattractant [40]. However, others could not Several reports focus on zinc-induced release of pro-
reproduce this direct action of zinc in human leukocytes [39]. inflammatory cytokines in vitro (IL-1, IL-6 and tumor necrosis fac-
NETosis is the formation and release of neutrophil extracellular tor (TNF)-a) [4,21,47]. In monocytes, zinc influences the secretion of
pro-inflammatory cytokines in a concentration-depended manner
60 M. Maares, H. Haase / Archives of Biochemistry and Biophysics 611 (2016) 58e65

Fig. 1. Innate and adaptive immunity during zinc deficiency. Overview of innate and adaptive immune cells and their interactions during zinc deficiency. For reasons of clarity only
selected cells and their mediated operations are shown. Immune cell numbers are, in most cases, decreased during zinc deficiency. Immediate immune defense against pathogens is
still maintained, because phagocytosis and oxidative burst are not declined in macrophages and monocytes, unless zinc deficiency becomes too severe. Cytokine release is
particularly impaired leading to altered intercellular communication and regulation.

e both enhancing and inhibiting cytokine release [4]. Low zinc occurs during zinc deficiency [55]. Such a shift from adaptive to
concentrations increase LPS-induced secretion of pro- predominantly innate immunity and immediate host defense
inflammatory cytokines, whereas higher concentrations nega- would be accompanied by a decreased requirement for communi-
tively affect cytokine production in monocytes in vitro. However, cation between innate and adaptive immune cells by cytokines.
the specific amount of zinc, which influences cytokine release, Instead, monocytes and macrophages focus on phagocytosis and
depends on the cell model and experimental conditions. In human oxidative burst during zinc deficiency.
peripheral blood mononuclear cells (PBMC), incubation with zinc On the molecular level, zinc regulates secretion of pro-
concentrations of 100 mM in LPS-free medium led to stimulation of inflammatory cytokines by monocytes through modulating tran-
mRNA expression and release of IL-6, IL-1b, and TNF-a, suggesting scription of their genes via activation or inhibition of the nuclear
that high zinc concentrations stimulate monokine release in vitro factor-kB (NF-kB) [4,24]. The underlying molecular pathway is
[48e50]. Moreover, when incubated simultaneously under “non- based on Toll-like receptor (TLR)-4 signaling and its modulation by
stimulatory concentrations” (<100 mM zinc) zinc potentiated the zinc signals [7] (Fig. 2). TLR-4 is activated by conserved molecular
LPS effect, leading to increased secretion of cytokines [17,51]. In the structures associated with pathogens, such as LPS [7,56], initiating
human cell line Mono Mac 1, 25 mM zinc was sufficient to down- two different signaling pathways, either depending on the myeloid
regulate mRNA levels of TNF-a and IL-1b after incubation with LPS, differentiation primary response gene 88 (MyD88) or on Toll/IL-1R
whereas 1 mM zinc induced cytokine release [4,52]. However, in this domain-containing adapter inducing interferon-b (TRIF). The
study zinc-incubation was conducted in the presence of pyrithione, MyD88-pathway leads to production of cytokines via NF-kB and
an ionophore that facilitates zinc absorption [52]. Findings of was shown to be activated by fast zinc signals, which occur in a few
cytokine release in zinc-deficient states are inconsistent and vary seconds or minutes after triggering of the receptor [6]. In contrast,
depending on cell line, experimental condition and duration of zinc TRIF-dependent signals are inhibited by zinc, resulting in decreased
depletion. In HL-60 cells, a promyelocytic cell line, zinc deficiency IFN-b-production by reduced activation of IFN regulatory factor 3
augmented PMA-stimulated release of IL-1b, TNF-a, and IL-8 (IRF3) [56]. Furthermore, zinc also affects TLR-4-mediated cytokine
[33,47]. This was due to downregulation of A20, a protein which release via cyclic nucleotide signals. Cyclic nucleotide phosphodi-
inhibits cytokine production on the molecular level [47,53]. esterases (PDEs) degrade the second messengers cyclic adenosine
Furthermore, in vivo studies with mice showed that zinc depriva- monophosphate (cAMP) and cyclic guanosine monophosphate
tion by TPEN increases levels of IL-1b and IL-6 after LPS-injection (cGMP) and depend on low zinc concentrations for their activity.
[54]. In contrast, cytokine production is decreased by mild zinc Conversely, higher zinc concentrations inhibit PDE activity, result-
deficiency in human PBMC [21], whereas phagocytosis and oxida- ing in elevated cGMP, which cross-activates protein kinase A (PKA).
tive burst are not affected, reinforcing the assumption of a shift Through inhibitory phosphorylation of Raf1, this finally leads to
away from intercellular communication via cytokines, concen- impairment of NF-kB activation. Yet another type of NF-kB-regu-
trating the cellular activity on direct anti-pathogenic functions [21]. lation by zinc was found in HL-60 cells, in which the zinc finger
It has been postulated that a reprogramming of the immune system protein A20 is upregulated by zinc. Increased A20 then suppresses
M. Maares, H. Haase / Archives of Biochemistry and Biophysics 611 (2016) 58e65 61

Fig. 2. Role of TLR-4 and zinc in antigen presenting cells. The LPS-induced MyD88-pathway leads to pro-inflammatory cytokine release and, together with increased levels of TRIF-
dependent IFN-b, NF-kB-activation triggers expression of the inducible nitric oxide synthase (iNOS). Upregulation of zinc exporters (ZnT) and downregulation of the zinc importer
ZIP6 via TRIF-dependent signaling result in decreased intracellular free zinc, subsequently leading to increased transport of major histocompatibility complex (MHC) class II
molecules to the plasma membrane and cell maturation. Broken lines implicate a series of different steps that are not depicted in detail for reasons of clarity.

TLR-induced activation of NF-kB leading to downregulated cyto- plasma membrane of NK-cells, which detect the expression of MHC
kines [34,57]. In fact, in comparison to zinc sufficient HL-60 cells, I molecules on the surface of the target cell and inhibit cytotoxic
zinc deficiency led to decreased A20 mRNA levels, relieving its mechanisms, are modulated by zinc. Zinc is required for multi-
inhibitory effect on NF-kB, followed by augmented cytokine release merization of KIR [65] and its interaction with MHC I molecules,
[53]. Hence, zinc signals are required for NF-kB activation and while stimulation of the killing process is not zinc-dependent [66].
subsequent cytokine production. In the presence of critically Thus, during zinc deficiency these inhibitory signals and the proper
decreased or elevated intracellular zinc concentrations, these NF- interaction with MHC I on target cells could be impaired and lead to
kB-activating pathways are inhibited, resulting in impaired cyto- unspecific killing. However, the latter might be only of limited
kine release. relevance in light of the abovementioned functional impairment of
NK-cell cytotoxic activities during zinc deficiency.
2.3. Natural killer cells Besides their cytotoxic defense mechanisms, NK-cells also
regulate the immune response and facilitate the activation of im-
Natural killer (NK-) cells are lymphocytes that are essential for mune cells by secreting cytokines, which was shown to be
maintaining the innate immune response and host defense against enhanced after zinc supplementation [61]. One of these cytokines is
viral infections and tumors. They recognize and kill infected and IFN-g, which activates dendritic cells, promotes their maturation
transformed cells through cytotoxic mechanisms. The expression of and initiates immune response by T helper cells (TH)-cells and their
major histocompatibility complex (MHC) class I molecules on the differentiation, which is discussed in detail below [61].
cell surface is often downregulated in tumors and virally infected
cells, hiding them from cytotoxic T-cells, which recognize their 2.4. Dendritic cells
target cells by antigens presented on MHC I. In contrast, NK-cells
can detect these cells via missing antigen presentation [22,58]. Other important leukocytes in innate immune defense are
Similar to the other innate immune cells, NK-cell activity and dendritic cells (DC), which are often described as a linker between
function is zinc-dependent. Impaired NK-cell function was reported innate and adaptive immune system. They are essential for the
in zinc-deficient humans [38,59] and after in vitro treatment with a innate immune system, secreting cytokines (IL-2, IL-6, IL-10; IL-12,
zinc chelator [38]. This was mainly due to decreased stimulation by TNF-a [67e69]), and activating NK- and NKT-cells [58,70]. Addi-
decreased T-cell-secreted IL-2 [60]. So far, only effects of extracel- tionally, they provide information about dimension and nature of
lular added zinc have been reported - not specifying its intracellular infection and communicate with T-cells via antigen presentation
role in NK-cell activity. [71]. Intracellular zinc concentrations play a critical role in DC
Zinc supplementation significantly increased NK-cell numbers function and activity. LPS stimulation of murine dendritic cells led
in whole blood cultures [61] and NK-cell activity in vivo [62], to downregulation of the zinc importer Zrt/Irt-like protein 6 (ZIP6)
whereas zinc depletion with TPEN did not lead to a decrease in the and upregulation of zinc exporters via TLR-4, resulting in decreased
number of NK-cells [61]. NK-cells, as all other leukocytes, are free zinc and increased cell maturation [72]. In fact, reduction of
generated from pluripotent hematopoietic stem cells in the bone intracellular zinc was found to be crucial for surface expression of
marrow that express the cluster of differentiation protein (CD) 34þ MHC II molecules [72], which present antigens to CD4þT-cells,
on their surface [63]. Differentiation of a CD34þ common he- hereby activating them during an infection. In immature DC, MHC II
matopoietic progenitor cell toward NK-cells has been observed to is located in lysosomes and endosomes [73]. Transport of MHC II
be augmented by moderate extracellular zinc concentrations [64], molecules from these compartments to the plasma membrane is
implying an involvement of zinc in NK-cell formation and inhibited by zinc and a reduction of cellular zinc is required to
differentiation. enable antigen presentation and hereby communication with
On the functional level, killer inhibitory receptors (KIR) on the adaptive immunity [72] (Fig. 2).
62 M. Maares, H. Haase / Archives of Biochemistry and Biophysics 611 (2016) 58e65

3. Adaptive immune system affected, leading to a net shift of the TH1/TH2 ratio toward TH2
[43,90]. Thus, zinc influences the TH1/TH2 balance, resulting in
The adaptive immune system is based on T- and B-lymphocytes, altered immune function due to decreased TH1 cytokine secretion
which express specific receptors on their surface to detect antigens [77].
- T-cell receptor (TCR) and B-cell receptor (BCR) [74,75]. Antigens Several studies investigated the correlation of TH1 cytokine
are defined as structures, which are recognized by these receptors release and zinc concentration. Studies with the T-cell line Hut-78
or antibodies (¼ soluble BCR) and are mainly proteins and peptides, analyzed cytokine release after incubation with extremely low
next to carbohydrates, lipids or any synthetic structure. Their spe- (1 mM), moderate (15 mM) and high zinc concentrations
cific recognition by these receptors results in a selective expansion (50e100 mM) [5,94]. While cells with zinc deficiency (incubated
in numbers and activation of the T- and B-cell clones carrying a with 1 mM zinc) showed significant decrease in IL-2, TNF-a, IFN-g,
matching receptor, leading to the participation of these highly and the IL-2 receptor alpha chain (IL-2Ra) gene expression [5], high
antigen-specific cells in the immune response. Some of these cells zinc supplementation led to mild mRNA-downregulation and
persist as memory cells even after the end of an infection. Hence, decreased production of these cytokines compared to incubation
subsequent contacts with the same antigen lead to a faster and with 15 mM zinc [94]. Zinc affects mRNA levels of cytokines via NF-
more efficient response of the adaptive immune system. kB inhibition or induction, and, accordingly, NF-kB DNA-binding
Altered zinc homeostasis influences lymphocyte activity, func- activity in Hut-78 cells was also decreased in the same
tion, and development by directly affecting their formation (lym- concentration-dependent manner [94]. Furthermore, TNF-a is
phopoiesis) and cytokine secretion (Fig. 1) or indirectly due to known to stimulate NF-kB [24], thus downregulation of TNF-a
impaired stimulation by innate immune cells [2,76]. might also affect NF-kB activity [94]. In addition to the molecular
mechanism of cytokine expression via the protein kinase C (PKC)/
3.1. T-cells Raf1/NF-kB pathway [4,52], it has been reported that intracellular
zinc levels modulate TH1 cell differentiation via IFN-g and T-box
T-cells express CD4þ or CD8þ co-receptors on their surface, transcription factor (T-bet) upregulation in Hut-78 cells after
defining their function. These co-receptors lead to enhanced Concanavalin A (Con A)-induced TCR-stimulation [45]. More spe-
interaction with MHC molecules on the surface of target cells cifically, increase of intracellular free zinc after Con A stimulation in
(CD4þ/MHC II; CD8þ/MHC I) and regulate the specific presentation cells treated with 5 mM zinc is suggested to enhance IFN-g, T-bet,
of antigens to the TCR. Depending on their co-receptor, T-cells are and IL-12 receptor beta 2 subunit (IL-12Rb2) via activated PKC-q.
divided in cytotoxic CD8þ T-cells and CD4þ T helper (TH)-cells Because IFN-g, T-bet, and IL-12Rb2 are important for differentiation
[77,78]. TH-cells promote functions of other immune cells, whereas of naïve CD4þT-cells toward TH1 [44], upregulation of these cyto-
cytotoxic T-cells recognize and eliminate virally infected and tumor kines emphasizes that zinc promotes TH1 differentiation [45].
cells [6]. Regulatory T-cells (Tregs) play a special role in controlling
The thymus is a lymphoid organ, where maturation of T-cells adaptive and innate immunity by suppressing immune cells
takes place. Several reports describe the occurrence of thymic at- [16,95]. Several studies report unwanted T-cell-mediated immune
rophy due to zinc deficiency in human children [79,80] and animals responses and their prevention by zinc treatment in vivo and
in vivo [81], reporting a serious size reduction of the thymus, which in vitro [16,96e98]. Moderate zinc supplementation was suggested
is caused by increased glucocorticoid levels and enhanced as means to control allergen specific immune reactions by regula-
apoptosis of pre T-cells. Subsequently, the amount of mature T-cells tion of the TH1/TH2 ratio and Treg activity [43,95]. Zinc was observed
significantly declines, leading to reduced activity of CD8þ cytotoxic to modulate the allergic immune response by increasing the
T-cells and CD4þ TH-cells [76,82,83]. number of regulatory T-cells, limited allergen-induced immune cell
This crucial decrease in T-cells during zinc deficiency is not only proliferation, and development of the immune response toward
caused by enhanced apoptosis of pre T-cells, but also by impaired TH1 by decreased TH2 cytokine IL-10 and enhanced IFN-g in human
thymulin activity [1,84]. Thymulin is a thymic peptide hormone PBMC after incubation with timothy grass allergen and 50 mM zinc
that requires zinc for its activity and is crucial for T-cell differenti- [16]. Conversely, in a mixed lymphocyte culture the addition of
ation and function [84,85]. Additionally, it induces IL-2 cytokine 50 mM zinc induced Tregs but decreased IFN-g, suggesting a pre-
secretion and CD8þ cell proliferation [86,87]. In humans with mild vention of allograft rejection e rejection of transplanted tissue from
zinc deficiency, thymulin activity and number of T-cells decrease the same species [99]. These immunomodulatory effects of zinc by
significantly [1]. Similarly, thymulin activity was diminished in regulation of cytokine signaling in T-cells were also reported in vivo
zinc-deficient mice without thymic atrophy and could be restored [98]. Here, allogeneic reactions were suppressed after supple-
by zinc supplementation [84]. mentation with high zinc doses in vitro and in vivo (80 mg/day;
Decreased proliferation and differentiation affect TH-cell func- 1wk) [96,100].
tion and cytokine production. There are several types of TH-cells TH17 are another subpopulation of TH-cells, which induces the
and their balance is of crucial importance for an effective immune recruitment of neutrophil granulocytes and regulates cell-mediated
response. TH1- and TH2-cells antagonize each other, directing the immune response against extracellular pathogens [101]. Cytokines
immune response either toward cellular immunity (TH1), especially released by TH17-cells promote inflammation and are connected
activation of macrophages, or toward humoral immunity (TH2), with autoimmune diseases [102]. Beneficial effects of zinc supple-
mediated by B-cell antibody production [88]. TH1 cytokines (IL-2, mentation were observed in investigations on zinc and TH17-
IFN-g, and TNF-a) are reduced by zinc deficiency [43,89,90]. The mediated autoimmune diseases. In mice with experimental auto-
TH1 cytokine IFN-g mainly stimulates TH1 development, macro- immune encephalomyelitis (EAE), an experimental model for
phages, and DCs and induces MHC molecule expression [44,45]. multiple sclerosis, moderate zinc doses led to declined TH17 cell
Besides, IFN-g attenuates TH2 [91]. IL-2 is an important cytokine for numbers and diminished EAE severity [97,103]. This was accom-
T-cell proliferation, activation, and differentiation and additionally panied by an augmented number of inducible Tregs in the central
represses TH2 [89,92]. Because IL-2 stimulates NK-cells and mono- nervous system [103].
cytes, impaired IL-2 secretion alters their activity and function as Treatment of individuals with pharmacological zinc supple-
well [47,93]. Production of TH1 cytokines is decreased in zinc- mentation has a profound effect on the immune system. Similarly
deficient states, whereas TH2 cytokines (IL-4, IL-6, IL-10) are not to the other immune cells, supplementation of zinc-deficient
M. Maares, H. Haase / Archives of Biochemistry and Biophysics 611 (2016) 58e65 63

individuals can reverse the negative effects of zinc deficiency on T- beta, and NF-kappa B, J. Immunol. 179 (6) (2007) 4180e4186.
[5] A.S. Prasad, et al., Zinc activates NF-kappaB in HUT-78 cells, J. Lab. Clin. Med.
cells [41,104]. Treatment of individuals with normal zinc levels was
138 (4) (2001) 250e256.
shown to modulate the immune system dependent on the sup- [6] H. Haase, L. Rink, Zinc signals and immune function, Biofactors 40 (1) (2014)
plemented amount of zinc. Moderate zinc concentrations improve 27e40.
immune function and extenuate unwanted T-cell-mediated im- [7] H. Haase, et al., Zinc signals are essential for lipopolysaccharide-induced
signal transduction in monocytes, J. Immunol. 181 (9) (2008) 6491e6502.
mune response [97,103], whereas high zinc doses inhibit immune [8] T. Miyai, et al., Zinc transporter SLC39A10/ZIP10 facilitates antiapoptotic
function [17,100]. signaling during early B-cell development, Proc. Natl. Acad. Sci. U. S. A. 111
(32) (2014) 11780e11785.
[9] H. Haase, E. Mocchegiani, L. Rink, Correlation between zinc status and im-
3.2. B-cells mune function in the elderly, Biogerontology 7 (2006) 421e428.
[10] D. Ford, Intestinal and placental zinc transport pathways, Proc. Nutr. Soc. 63
(1) (2004) 21e29.
B-cells play an essential role in the humoral immune response
[11] P.J. Fraker, et al., The dynamic link between the integrity of the immune
by producing antibodies [74]. Because lymphopoiesis is impaired system and zinc status, J. Nutr. 130 (5S Suppl) (2000) 1399se1406s.
during zinc deficiency, pre-B-cell numbers decrease during zinc [12] R.A. Wapnir, Zinc deficiency, malnutrition and the gastrointestinal tract,
deprivation [83] (Fig. 1). Interestingly, an animal study with zinc- J. Nutr. 130 (2000) 13885e13925.
[13] M. Bonomini, et al., Effects of zinc supplementation in chronic haemodialysis
deficient mice showed a strong decrease in lymphopoiesis for T- patients, Nephrol. Dial. Transpl. 8 (10) (1993) 1166e1168.
cells, whereas B-cells and their maturation in the bone marrow [14] W. Maret, H.H. Sandstead, Zinc requirements and the risks and benefits of
were less affected [2]. Comparable to T-cells, the decreased pre B- zinc supplementation, J. Trace Elem. Med. Biol. 20 (1) (2006) 3e18.
[15] S. Maggini, S. Beveridge, M. Suter, A Combination of high-dose vitamin C plus
cell number in zinc-deficient states is connected to glucocorticoid zinc for the common cold, J. Int. Med. Res. 40 (1) (2012) 28e42.
metabolism and increased apoptosis [105]. A recent report by [16] E. Rosenkranz, et al., Zinc enhances the number of regulatory T cells in
Hojyo et al. has provided new insights into the role of zinc in B-cell allergen-stimulated cells from atopic subjects, Eur. J. Nutr. (2015), http://
dx.doi.org/10.1007/s00394-015-1100-1.
development. The antigen receptor BCR, expressed on the surface [17] C. Driessen, et al., Zinc regulates cytokine induction by superantigens and
of immature and mature B-cells, controls the activation status of B- lipopolysaccharide, Immunology 84 (2) (1995) 272e277.
cells and regulates their proliferation [106]. The zinc transporter [18] N. Wellinghausen, H. Kirchner, L. Rink, The immunobiology of zinc, Immunol.
Today 18 (1997) 519e521.
ZIP10 regulates B-cell function and has an important role in hu- [19] S. Overbeck, L. Rink, H. Haase, Modulating the immune response by oral zinc
moral immunity by modulating BCR signals [106] and preventing supplementation: a single approach for multiple diseases, Arch. Immunol.
apoptosis in B-cells [8]. Ther. Exp. Warsz. 56 (1) (2008) 15e30.
[20] R.S. Gibson, et al., Indicators of zinc status at the population level: a review of
In zinc-deficient states, a reduced immune response to vacci-
the evidence, Br. J. Nutr. 99 (Suppl 3) (2008) S14eS23.
nation in vivo was reported several times [107e109], suggesting a [21] L.S. Mayer, et al., Differential impact of zinc deficiency on phagocytosis,
possible enhancement of antibody formation by zinc supplemen- oxidative burst, and production of pro-inflammatory cytokines by human
tation [110]. Multiple studies investigated the impact of zinc sup- monocytes, Metallomics 6 (7) (2014) 1288e1295.
[22] E. Vivier, et al., Innate or adaptive immunity? The example of natural killer
plementation on vaccination leading to different results, mainly cells, Science 331 (6013) (2011) 44e49.
due to heterogeneity of experimental design (as reviewed in [23] F. Belardelli, M. Ferrantini, Cytokines as a link between innate and adaptive
Ref. [19]). In general, simultaneous supplementation of high antitumor immunity, Trends Immunol. 23 (4) (2002) 201e208.
[24] U. Siebenlist, G. Franzoso, K. Brown, Structure, regulation and function of NF-
amounts of zinc at the time of vaccination resulted in poorer out- kappa B, Annu. Rev. Cell Biol. 10 (1994) 405e455.
comes, whereas zinc treatment in advance of vaccination seems to [25] E. Rosenkranz, A. Prasad, L. Rink, Immunobiology and Hematology of Zinc,
be a far more promising way to improve the antibody response 2011.
[26] P.G. Sohnle, et al., Zinc-reversible antimicrobial activity of recombinant cal-
[109e112]. protectin (migration inhibitory factor-related proteins 8 and 14), J. Infect.
Dis. 182 (4) (2000) 1272e1275.
[27] A.K. Schro €der, et al., Polymorphonuclear leucocytes selectively produce anti-
4. Conclusion
inflammatory interleukin-1 receptor antagonist and chemokines, but fail to
produce pro-inflammatory mediators, Immunology 119 (3) (2006) 317e327.
The role of zinc in the immune system has been investigated in [28] B. Amulic, et al., Neutrophil function: from mechanisms to disease, Annu.
depth. As discussed above, altered zinc homeostasis critically in- Rev. Immunol. 30 (2012) 459e489.
[29] A.R. Lloyd, J.J. Oppenheim, Poly's lament: the neglected role of the poly-
fluences innate and adaptive immunity, and therefore host defense morphonuclear neutrophil in the afferent limb of the immune response,
and the immune response in general. Immunol. Today 13 (5) (1992) 169e172.
Zinc supplementation can reverse the negative effects of zinc [30] W.L. Lee, R.E. Harrison, S. Grinstein, Phagocytosis by neutrophils, Microbes
Infect. 5 (14) (2003) 1299e1306.
deficiency, including impaired immune cell development, [31] R. Hasan, L. Rink, H. Haase, Zinc signals in neutrophil granulocytes are
compromised T-cell-mediated immune response, decreased required for the formation of neutrophil extracellular traps, Innate Immun.
oxidative burst and many more. The potential benefits of zinc 19 (3) (2012) 253e264.
[32] H. Hasegawa, et al., Effects of zinc on the reactive oxygen species generating
supplementation go from anti-inflammatory and immunomodu- capacity of human neutrophils and on the serum opsonic activity in vitro,
latory effects, to prevention of allergies and autoimmunity, or even Luminescence 15 (5) (2000) 321e327.
suppression of allogenic reactions. A more detailed understanding [33] I. Wessels, et al., Zinc deficiency induces production of the proinflammatory
cytokines IL-1beta and TNFalpha in promyeloid cells via epigenetic and
of the molecular effects of zinc and their controlled manipulation
redox-dependent mechanisms, J. Nutr. Biochem. 24 (1) (2013) 289e297.
might prove to be a useful future tool for targeted modulation of the [34] A.S. Prasad, et al., Antioxidant effect of zinc in humans, Free Radic. Biol. Med.
immune system with very limited side effects. 37 (8) (2004) 1182e1190.
[35] W. Maret, Zinc coordination environments in proteins as redox sensors and
signal transducers, Antioxid. Redox Signal 8 (9e10) (2006) 1419e1441.
References [36] A.S. Prasad, Zinc is an antioxidant and anti-inflammatory agent: its role in
human health, Front. Nutr. (2014) 1.
[1] A.S. Prasad, et al., Serum thymulin in human zinc deficiency, J. Clin. Invest. 82 [37] C.L. Keen, M.E. Gershwin, Zinc deficiency and immune function, Annu. Rev.
(4) (1988) 1202e1210. Nutr. 10 (1990) 415e431.
[2] L.E. King, et al., Chronic zinc deficiency in mice disrupted T cell lympho- [38] J.I. Allen, et al., Alterations in human natural killer cell activity and monocyte
poiesis and erythropoiesis while B Cell lymphopoiesis and myelopoiesis cytotoxicity induced by zinc deficiency, J. Lab. Clin. Med. 102 (4) (1983)
were maintained, J. Am. Coll. Nutr. 24 (6) (2005) 494e502. 577e589.
[3] J.A. Halsted, A.S. Prasad, Syndrome of iron deficiency anemia, hep- [39] R. Hasan, L. Rink, H. Haase, Chelation of free Zn Impairs chemotaxis,
atosplenomegaly, hypogonadism, dwarfism and geophagia, Trans. Am. Clin. phagocytosis, oxidative burst, degranulation, and cytokine production by
Climatol. Assoc. 72 (1961) 130e149. neutrophil granulocytes, Biol. Trace Elem. Res. (2015), http://dx.doi.org/
[4] V. von Bulow, et al., Zinc-dependent suppression of TNF-alpha production is 10.1007/s12011-015-0515-0.
mediated by protein kinase A-induced inhibition of Raf-1, I kappa B kinase [40] E.S. Hujanen, S.T. Seppa, K. Virtanen, Polymorphonuclear leukocyte
64 M. Maares, H. Haase / Archives of Biochemistry and Biophysics 611 (2016) 58e65

chemotaxis induced by zinc, copper and nickel in vitro, Biochim. Biophys. transport from lysosomes to the plasma membrane, Nature 418 (6901)
Acta 1245 (2) (1995) 145e152. (2002) 988e994.
[41] A.S. Prasad, Zinc: role in immunity, oxidative stress and chronic inflamma- [74] R.R. Hardy, K. Hayakawa, B cell development pathways, Annu. Rev. Immunol.
tion, Curr. Opin. Clin. Nutr. Metab. Care 12 (6) (2009) 646e652. 19 (2001) 595e621.
[42] S. Dubben, et al., Cellular zinc homeostasis is a regulator in monocyte dif- [75] D. Cantrell, T cell antigen receptor signal transduction pathways, Annu. Rev.
ferentiation of HL-60 cells by 1 alpha,25-dihydroxyvitamin D3, J. Leukoc. Immunol. 14 (1) (1996) 259e274.
Biol. 87 (5) (2010) 833e844. [76] P.J. Fraker, L.E. King, A distinct role for apoptosis in the changes in lym-
[43] A.S. Prasad, Effects of zinc deficiency on Th1 and Th2 cytokine shifts, J. Infect. phopoiesis and myelopoiesis created by deficiencies in zinc, Faseb J. 15 (14)
Dis. 182 (Suppl 1) (2000) S62eS68. (2001) 2572e2578.
[44] D. Agnello, et al., Cytokines and transcription factors that regulate T helper [77] T.R. Mosmann, R.L. Coffman, TH1 and TH2 cells: different patterns of
cell differentiation: new players and new insights, J. Clin. Immunol. 23 (3) lymphokine secretion lead to different functional properties, Annu. Rev.
(2003) 147e161. Immunol. 7 (1989) 145e173.
[45] B. Bao, et al., Intracellular free zinc up-regulates IFN-gamma and T-bet [78] A.S. Prasad, Zinc: mechanisms of host defense, J. Nutr. 137 (5) (2007)
essential for Th1 differentiation in Con-A stimulated HUT-78 cells, Biochem. 1345e1349.
Biophys. Res. Commun. 407 (4) (2011) 703e707. [79] J.M. Oleske, et al., Zinc therapy of depressed cellular immunity in acro-
[46] C.L. Langrish, et al., IL-12 and IL-23: master regulators of innate and adaptive dermatitis enteropathica. Its correction, Am. J. Dis. Child. 133 (9) (1979)
immunity, Immunol. Rev. 202 (2004) 96e105. 915e918.
[47] B. Bao, et al., Zinc modulates mRNA levels of cytokines, Am. J. Physiol. [80] M.N. Golden, A. Jackson, B. Golden, Effect of zinc on thymus of recently
Endocrinol. Metab. 285 (2003) E1095eE1102. malnourished children, Lancet 310 (8047) (1977) 1057e1059.
[48] N. Wellinghausen, C. Driessen, L. Rink, Stimulation of human peripheral [81] P.S. Dowd, J. Kelleher, P.J. Guillou, T-lymphocyte subsets and interleukin-2
blood mononuclear cells by zinc and related cations, Cytokine 8 (10) (1996) production in zinc-deficient rats, Br. J. Nutr. 55 (1) (1986) 59e69.
767e771. [82] P. DePasquale-Jardieu, P. Fraker, Further characterization of the role of
[49] N. Wellinghausen, M. Martin, L. Rink, Zinc inhibits interleukin-1-dependent corticosterone in the loss of humoral immunity in zinc-deficient A/J mice as
T cell stimulation, Eur. J. Immunol. 27 (10) (1997) 2529e2535. determined by adrenalectomy, J. Immunol. 124 (1980) 2650e2655.
[50] C. Driessen, et al., Induction of cytokines by zinc ions in human peripheral [83] L.E. King, F. Osati-Ashtiani, P.J. Fraker, Apoptosis plays a distinct role in the
blood mononuclear cells and separated monocytes, Lymphokine Cytokine loss of precursor lymphocytes during zinc deficiency in mice, J. Nutr. 132 (5)
Res. 13 (1) (1994) 15e20. (2002) 974e979.
[51] C. Driessen, et al., Divergent effects of zinc on different bacterial pathogenic [84] M. Dardenne, et al., In vivo and in vitro studies of thymulin in marginally
agents, J. Infect. Dis. 171 (2) (1995) 486e489. zinc-deficient mice, Eur. J. Immunol. 14 (1984) 454e458.
[52] V. von Bulow, L. Rink, H. Haase, Zinc-mediated inhibition of cyclic nucleotide [85] J.J. Haddad, Thymulin and zinc (Zn2þ)-mediated inhibition of endotoxin-
phosphodiesterase activity and expression suppresses TNF-alpha and IL-1 induced production of proinflammatory cytokines and NF-kappaB nuclear
beta production in monocytes by elevation of guanosine 3',5'-cyclic mono- translocation and activation in the alveolar epithelium: unraveling the mo-
phosphate, J. Immunol. 175 (7) (2005) 4697e4705. lecular immunomodulatory, anti-inflammatory effect of thymulin/Zn2þ
[53] A.S. Prasad, et al., Zinc-suppressed inflammatory cytokines by induction of in vitro, Mol. Immunol. 47 (2e3) (2009) 205e214.
A20-mediated inhibition of nuclear factor-kappaB, Nutrition 27 (7e8) (2010) [86] B. Safieh-Garabedian, et al., Thymulin modulates cytokine release by pe-
816e823. ripheral blood mononuclear cells: a comparison between healthy volunteers
[54] H. Summersgill, et al., Zinc depletion regulates the processing and secretion and patients with systemic lupus erythematosus, Int. Arch. Allergy Immunol.
of IL-1j[beta], Cell Death Dis. 5 (1) (2014). 101 (2) (1993) 126e131.
[55] P.J. Fraker, L.E. King, Reprogramming of the immune system during zinc [87] M. Dardenne, Zinc and immune function, Eur. J. Clin. Nutr. 56 (Suppl 3)
deficiency, Annu. Rev. Nutr. 24 (2004) 277e298. (2002) S20eS23.
[56] A. Brieger, L. Rink, H. Haase, Differential regulation of TLR-dependent MyD88 [88] A.L. Rothermel, K.M. Gilbert, W.O. Weigle, Differential abilities of Th1 and
and TRIF signaling pathways by free zinc ions, J. Immunol. 191 (4) (2013) Th2 to induce polyclonal B cell proliferation, Cell Immunol. 135 (1) (1991)
1808e1817. 1e15.
[57] B. Bao, et al., Zinc decreases C-reactive protein, lipid peroxidation, and in- [89] K. Gruber, et al., Zinc deficiency adversely influences interleukin-4 and
flammatory cytokines in elderly subjects: a potential implication of zinc as interleukin-6 signaling, J. Biol. Regul. Homeost. Agents 27 (3) (2013)
an atheroprotective agent, Am. J. Clin. Nutr. 91 (6) (2010) 1634e1641. 661e671.
[58] W.M. Yokoyama, B.F.M. Plougastel, Immune functions encoded by the nat- [90] F.W. Beck, et al., Changes in cytokine production and T cell subpopulations in
ural killer gene complex, Nat. Rev. Immunol. 3 (4) (2003) 304e316. experimentally induced zinc-deficient humans, Am. J. Physiol. 272 (6 Pt 1)
[59] E. Tapazoglou, et al., Decreased natural killer cell activity in patients with (1997) E1002eE1007.
zinc deficiency with sickle cell disease, J. Lab. Clin. Med. 105 (1985) 19e22. [91] B. Elser, et al., IFN-gamma represses IL-4 expression via IRF-1 and IRF-2,
[60] A. Prasad, Effects of zinc deficiency on Th1 and Th2 cytokine shifts, J. Infect. Immunity 17 (6) (2002) 703e712.
Dis. 2182 (Suppl 1) (2000) 62e68. [92] A.S. Prasad, Zinc in human health: effect of zinc on immune cells, Mol. Med.
[61] C.H. Metz, et al., T-helper type 1 cytokine release is enhanced by in vitro zinc 14 (5e6) (2008) 353e357.
supplementation due to increased natural killer cells, Nutrition 23 (2) (2006) [93] I. Espinoza-Delgado, et al., Interleukin-2 and human monocyte activation,
157e163. J. Leukoc. Biol. 57 (1) (1995) 13e19.
[62] E. Mariani, et al., Effect of zinc supplementation on plasma IL-6 and MCP-1 [94] B. Bao, et al., Toxic effect of zinc on NF-kappaB, IL-2, IL-2 receptor alpha, and
production and NK cell function in healthy elderly: interactive influence TNF-alpha in HUT-78 (Th(0)) cells, Toxicol. Lett. 166 (3) (2006) 222e228.
of þ647 MT1a and -174 IL-6 polymorphic alleles, Exp. Gerontol. 43 (5) [95] C.M. Hawrylowicz, A. O'Garra, Potential role of interleukin-10-secreting
(2008) 462e471. regulatory T cells in allergy and asthma, Nat. Rev. Immunol. 5 (4) (2005)
[63] I.T. Kao, et al., Generation of natural killer cells from serum-free, expanded 271e283.
human umbilical cord blood CD34þ cells, Stem Cells Dev. 16 (6) (2007) [96] C. Faber, et al., Zinc in pharmacological doses suppresses allogeneic reaction
1043e1051. without affecting the antigenic response, Bone Marrow Transpl. 33 (12)
[64] M. Muzzioli, et al., Zinc improves the development of human CD34þ cell (2004) 1241e1246.
progenitors towards Natural Killer cells and induces the expression of GATA- [97] C. Kitabayashi, et al., Zinc suppresses Th17 development via inhibition of
3 transcription factor, Int. J. Biochem. Cell Biol. 39 (5) (2007) 955e965. STAT3 activation, Int. Immunol. 22 (5) (2010) 375e386.
[65] M. Vales-Gomez, et al., The role of zinc in the binding of killer cell Ig-like [98] A. Varin, et al., In vitro and in vivo effects of zinc on cytokine signalling in
receptors to class I MHC proteins, Immunology 96 (2000) 1734e1739. human T cells, Exp. Gerontol. 43 (5) (2008) 472e482.
[66] S. Rajagopalan, et al., The Ig-related killer cell inhibitory receptor binds zinc [99] E. Rosenkranz, et al., Zinc Supplementation Induces Regulatory T Cells by
and requires zinc for recognition of HLA-C on target cells, J. Immunol. 155 (9) Inhibition of Sirt-1 Deacetylase in Mixed Lymphocyte Cultures, 2016.
(1995) 4143e4146. [100] C.A. Campo, et al., Zinc inhibits the mixed lymphocyte culture, Biol. Trace
[67] F. Granucci, et al., Inducible IL-2 production by dendritic cells revealed by Elem. Res. 79 (1) (2001) 15e22.
global gene expression analysis, Nat. Immunol. 2 (9) (2001) 882e888. [101] W. Ouyang, J.K. Kolls, Y. Zheng, The biological functions of T helper 17 cell
[68] A. Langenkamp, et al., Kinetics of dendritic cell activation: impact on priming effector cytokines in inflammation, Immunity 28 (4) (2008) 454e467.
of TH1, TH2 and nonpolarized T cells, Nat. Immunol. 1 (4) (2001) 311e316. [102] B. Afzali, et al., The role of T helper 17 (Th17) and regulatory T cells (Treg) in
[69] S.E. Macatonia, et al., Dendritic cells produce IL-12 and direct the develop- human organ transplantation and autoimmune disease, Clin. Exp. Immunol.
ment of Th1 cells from naive CD4þ T cells, J. Immunol. 154 (10) (1995) 148 (1) (2007) 32e46.
5071e5079. [103] E. Rosenkranz, et al., Induction of regulatory T cells in Th1-/Th17-driven
[70] D. Sarhan, et al., Dendritic cell regulation of NK-cell responses involves experimental autoimmune encephalomyelitis by zinc administration, J. Nutr.
lymphotoxin-alpha, IL-12, and TGF-beta, Eur. J. Immunol. 45 (6) (2015) Biochem. 29 (2016) 116e123.
1783e1793. [104] S. Bhatnagar, et al., Zinc as adjunct treatment in infants aged between 7 and
[71] I. Mellman, R.M. Steinman, Dendritic cells: specialized and regulated antigen 120 days with probable serious bacterial infection: a randomised, double-
processing machines, Cell 106 (3) (2001) 255e258. blind, placebo-controlled trial, Lancet 379 (9831) (2012) 2072e2078.
[72] H. Kitamura, et al., Toll-like receptor-mediated regulation of zinc homeo- [105] P.J. Fraker, et al., Possible roles for glucocorticoids and apoptosis in the
stasis influences dendritic cell function, Nat. Immunol. 7 (9) (2006) 971e977. suppression of lymphopoiesis during zinc deficiency: a review, J. Am. Coll.
[73] A. Chow, et al., Dendritic cell maturation triggers retrograde MHC class II Nutr. 14 (1) (1995) 11e17.
M. Maares, H. Haase / Archives of Biochemistry and Biophysics 611 (2016) 58e65 65

[106] S. Hojyo, et al., Zinc transporter SLC39A10/ZIP10 controls humoral immunity immune response of old people, Am. J. Med. 70 (5) (1981) 1001e1004.
by modulating B-cell receptor signal strength, Proc. Natl. Acad. Sci. U. S. A. [110] M.J. Albert, et al., Supplementation with zinc, but not vitamin A, improves
111 (32) (2014) 11786e11791. seroconversion to vibriocidal antibody in children given an oral cholera
[107] T.A. Strand, et al., Effects of zinc deficiency and pneumococcal surface protein vaccine, J. Infect. Dis. 187 (6) (2003) 909e913.
a immunization on zinc status and the risk of severe infection in mice, Infect. [111] C.B. Braga, et al., Effect of zinc supplementation on serological response to
Immun. 71 (4) (2003) 2009e2013. vaccination against streptococcus pneumoniae in patients undergoing
[108] N. Zhao, et al., Gestational zinc deficiency impairs humoral and cellular chemotherapy for colorectal cancer, Nutr. Cancer 67 (6) (2015) 926e932.
immune responses to hepatitis B vaccination in offspring mice, PLoS One 8 [112] M.E. Sundaram, et al., Vitamin E, vitamin A, and zinc status are not related to
(9) (2013) e73461. serologic response to influenza vaccine in older adults: an observational
[109] J. Duchateau, et al., Beneficial effects of oral zinc supplementation on the prospective cohort study, Nutr. Res. 34 (2) (2014) 149e154.

You might also like