Hostname: page-component-8448b6f56d-t5pn6 Total loading time: 0 Render date: 2024-04-18T05:27:51.396Z Has data issue: false hasContentIssue false

Zinc deficiency as a possible risk factor for increased susceptibility and severe progression of Corona Virus Disease 19

Published online by Cambridge University Press:  01 March 2021

Inga Wessels
Affiliation:
Institute of Immunology, Faculty of Medicine, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074Aachen, Germany
Benjamin Rolles
Affiliation:
Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074Aachen, Germany
Alan J. Slusarenko
Affiliation:
Department of Plant Physiology, RWTH Aachen University, Worringer Weg 1, 52074Aachen, Germany
Lothar Rink*
Affiliation:
Institute of Immunology, Faculty of Medicine, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074Aachen, Germany
*
*Corresponding author: Dr Lothar Rink, email LRink@ukaachen.de
Rights & Permissions [Opens in a new window]

Abstract

The importance of Zn for human health becomes obvious during Zn deficiency. Even mild insufficiencies of Zn cause alterations in haematopoiesis and immune functions, resulting in a proinflammatory phenotype and a disturbed redox metabolism. Although immune system malfunction has the most obvious effect, the functions of several tissue cell types are disturbed if Zn supply is limiting. Adhesion molecules and tight junction proteins decrease, while cell death increases, generating barrier dysfunction and possibly organ failure. Taken together, Zn deficiency both weakens the resistance of the human body towards pathogens and at the same time increases the danger of an overactive immune response that may cause tissue damage. The case numbers of Corona Virus Disease 19 (COVID-19) are still increasing, which is causing enormous problems for health systems and economies. There is an urgent need to reduce both the number of severe cases and the resulting deaths. While therapeutic options are still under investigation, and first vaccines have been approved, cost-effective ways to reduce the likelihood of or even prevent infection, and the transition from mild symptoms to more serious detrimental disease, are highly desirable. Nutritional supplementation might be an effective option to achieve these aims. In this review, we discuss known Zn deficiency effects in the context of an infection with Severe Acute Respiratory Syndrome-Coronavirus-2 and its currently known pathogenic mechanisms and elaborate on how severe pre-existing Zn deficiency may pre-dispose patients to a severe progression of COVID-19. First published clinical data on the association of Zn homoeostasis with COVID-19 and registered studies in progress are listed.

Type
Full Papers
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited
Copyright
© The Author(s), 2021. Published by Cambridge University Press on behalf of The Nutrition Society

In March 2020, the WHO declared the Corona Virus Disease 19 (COVID-19) to be a pandemic(1). Infections with Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) can be asymptomatic (40 % of the cases) or cause a mild illness (40 %), but in about 15 % of the cases, severe disease develops, characterised by clinical signs of pneumonia (fever, cough and dyspnoea) plus one of the following: respiratory rate > 30 breaths/min; severe respiratory distress or SpO2 < 90 % on room air as defined by the WHO. Patients with acute respiratory distress syndrome (ARDS), sepsis or septic shock are categorised as critically ill which is the case in about 5 % of the cases(2). Amongst the co-morbidities, resulting in severe COVID-19 progression, inappropriate nutrition is increasingly attracting attention(Reference Bencivenga, Rengo and Varricchi3). According to the WHO, 1·9 billion adults are overweight or obese, while 462 million are underweight(4), underlining the relevance of taking inappropriate nutrition into account when discussing prevention and treatment of COVID-19. It is important to mention that Zn deficiency is frequently observed in undernutrition as well as in obesity, although the underlying mechanisms are different(Reference Rios-Lugo, Madrigal-Arellano and Gaytán-Hernández5).

In a previous article, we drew attention to the strong overlap of risk groups for severe progression of COVID-19 with the groups where Zn deficiency is frequently diagnosed(Reference Wessels, Rolles and Rink6). The effects of Zn supplementation were described and discussed(Reference Wessels, Rolles and Rink6,Reference Skalny, Rink and Ajsuvakova7) . In this article, we would like to discuss how pre-existing Zn deficiency might increase the susceptibility to COVID-19 infections as well as pre-dispose individuals for severe progression of disease as summarised in Fig. 1. Despite the many improvements in Zn research, we still lack a valid biomarker to reliably assess the Zn status of an individual(Reference Sandstead and Freeland-Graves8,Reference Lowe, Fekete and Decsi9) . Serum or plasma Zn levels are often used but are not completely reliable. Thus, serum levels below 642·5 μg/l are taken as an indication of Zn deficiency, but only partially reflect intracellular concentrations and the Zn status of an individual. Therefore, clear clinical signs of Zn deficiency can be observed even if serum Zn levels are above this critical value or in the normal range. Circadian variations of serum Zn levels were observed, and serum Zn also depends upon recent food intake and the degree of hydration/dehydration of an individual(Reference Heller, Sun and Hackler10,Reference Guillard, Piriou and Gombert11) . Early effects of Zn deficiency are often general and include functional changes that can be associated with various diseases. For this reason, mild Zn deficiency can be ‘hidden’(Reference Sandstead and Freeland-Graves8). Functional deficiencies in Zn-dependent immunological processes have been shown in human subjects and mice without any significantly different serum or plasma Zn levels compared with controls(Reference King, Frentzel and Mann12Reference Beck, Prasad and Kaplan14). Currently, Zn deficiency is mostly defined by using a combination of clinical symptoms, calculating Zn and phytate intake from food and measuring immunological changes(Reference Trame, Wessels and Haase15,Reference Prasad16) . For growing infants (<2 years) and children (<5 years), the ‘height-for-age ratio’ should be determined as an additional parameter(Reference King, Brown and Gibson17). Moreover, it has been suggested that serum and plasma Zn values need to be adjusted for situations where inflammation is present(Reference McDonald, Suchdev and Krebs18,Reference Likoswe, Phiri and Broadley19) . For these reasons, Zn deficiency is often investigated using animal models of severe and induced Zn deficiency and well-defined low-Zn diets. Alternatively, Zn deficiency can reliably be modelled in cell cultures with either Zn-depleted media or by using Zn-specific chelators. Whether the latter rather models severe or mild Zn deficiency in the context of the whole organism is hard to predict. In this review, we describe data derived from clearly Zn-deficient humans and mice. Individuals with subclinical Zn deficiency might be less severely affected, but the effects are probably still not negligible(Reference Sandstead20). The consequences of Zn deficiency are manifold(Reference Prasad16,Reference Prasad21Reference Hambidge and Krebs24) , and only effects that are relevant regarding the susceptibility and progression of infectious diseases such as COVID-19 (Fig. 1) are included here. In regard to innate immunity, the article will focus on the effects of Zn on the integrity of the epithelial cell barriers, on neutrophil and macrophage maturation and functions, and in regard to adaptive immunity, we focus on lymphocyte maturation and differentiation, and cytokine and antibody production. Known effects of Zn deficiency on the vascular system and the association of those effects with diseases affecting the heart, kidney, central nervous system and intestine are described in relation to COVID-19.

Fig. 1 Summary of complications that can be expected in patients with pre-existing zinc deficiency, when challenged by Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2). A patient with no co-morbidities and a balanced zinc homoeostasis will most likely develop no or mild symptoms or complications if infected with SARS-CoV-2 because immune cell numbers and functions are balanced, as are the other parameters listed in the Figure. However, zinc deficiency alone will result in the alterations indicated in the Figure. Preconditions resulting from zinc deficiency may result in the development of severe symptoms, critical illness and even death if the patient becomes infected with SARS-CoV-2. ARDS, acute respiratory distress syndrome; CNS, central nervous system; IFN, interferon; MMP, matrix metalloproteinase; TH, T helper cell; Treg, regulatory T cell; ZA, zinc adequate; ZD: zinc deficient.

In addition to nutritional causes (undernutrition, malnutrition, veganism, geophagy, a phytate-rich diet, low-Zn parenteral nutrition), conditioned Zn deficiency has been observed in association with many diseases and inflammatory reactions(Reference McDonald, Suchdev and Krebs18,Reference Wessels and Rink25) . Attention was drawn to Zn deficiency in the 1960s due to a traditional soil-eating diet (geophagy) in a group in Iran leading to a severe Zn deficiency associated with dwarfism. The group revealed that a severely disturbed immune response, was more susceptible to infection, suffered from lethargy, and none survived beyond the age of 25 years(Reference Prasad16). Untreated severe Zn deficiency, such as that seen during acrodermatitis enteropathica, has a high mortality rate often because of the inefficient clearance of infections(Reference Barnes and Moynahan26). The subjects with acrodermatitis enteropathica, and the above-described group in Iran, suffered from severe Zn deficiency. However, studies in mice and human subjects have shown that detrimental effects are seen not only in severe Zn deficiency but that also a slight to moderate Zn deficiency can result in alterations of haematopoiesis and defects in the functions of immune cells(Reference King, Frentzel and Mann12Reference Beck, Prasad and Kaplan14,Reference Kahmann, Uciechowski and Warmuth27) , which thus increases the susceptibility to infection. It is important to recall that the immune system is affected negatively by Zn deficiency before any other symptoms become obvious and before serum Zn levels drop below 642·5 μg/l(Reference Roohani, Hurrell and Kelishadi28). Besides being essential for a robustly functioning immune system, Zn is also important for DNA synthesis, cell proliferation, cell differentiation, apoptosis, protein structure, protein–protein interactions and signal transduction as a second messenger for all kinds of cells. In the nervous system, Zn serves as an individual neurotransmitter that is secreted into the synaptic cleft(Reference Beyersmann and Haase29Reference Tóth33). Zn deficiency can manifest itself in a variety of ways; amongst others, there are increased frequencies of pneumonia and diarrhoea, an altered sense of smell and taste, cytopenia, poor wound healing, hair thinning, eczema, reduced fertility, increased fatigue, sicca syndrome and nail dysplasia(Reference Ackland and Michalczyk22). Zn deficiency is a significant public health problem, and high numbers of deaths worldwide, especially in children, are associated with severe Zn deficiency(Reference Sandstead and Freeland-Graves8,Reference Hambidge and Krebs24,Reference Caulfield, Onis and Blössner34,Reference Fischer Walker, Ezzati and Black35) .

The magnitude of the effects of a pre-existing Zn deficiency, and the significance of mild compared with severe Zn deficiency, remains to be clearly defined and clarified in relation to COVID-19. A series of studies have been registered to analyse retrospectively the serum Zn levels of patients (online Supplementary Table S1), and the first published data in this regard are starting to appear. Data from further registered studies, investigating prophylactic Zn supplementation to decrease the susceptibility for infections and severe disease, especially in medical and military personnel, are also underway (online Supplementary Table S1). In the absence of experimental data, we extrapolate the information from the existing literature, in anticipation of the data from clinical studies, which should soon be available (online Supplementary Table S1).

Zinc deficiency alters haematopoiesis and disturbs the balance of innate and adaptive immune cells largely to the detriment of cells from the lymphoid lineage

Severe infections with SARS-CoV-2 can cause major hematopoietic changes. Most prominently, a decrease in lymphocytes has been noted, especially affecting the T cells. In COVID-19 patients with severe symptoms, the reduction in number and the functional exhaustion of CD4+ as well as CD8+ T cells, as detected by elevated expression of Tim-3 and PD-1, is frequently described and observed early during disease(Reference Diao, Wang and Tan36,Reference Liu, Zhang and He37) . The recovery of T cell numbers in severely ill patients was paralleled with the improvement of the symptoms and with positive prognosis and survival(Reference Liu, Li and Liu38).

Available data on the effects of SARS-CoV-2 on CD4+ compared with CD8+ T cells are somewhat controversial. While, in one study, no significant difference in the CD4+:CD8+ ratio but increased expression of CD8+ was found(Reference Ganji, Farahani and Khansarinejad39), other studies have reported a decrease particularly of CD8+ T cells, or a significantly elevated CD4:CD8 ratio in COVID-19 patients(Reference Liu, Zhang and He37,Reference Liu, Li and Liu38,Reference Jiang, Guo and Luo40) . As high levels of either perforin or granulysin, or both, were detected in CD8+ T cells(Reference Xu, Shi and Wang41), it can be assumed that CD8+ cells are overreacting initially and are subject to exhaustion and apoptosis at later stages. However, this hypothesis remains to be addressed. In contrast, B cell numbers and serum levels of Ig (IgA, IgG and IgM) have been reported to be rather weakly affected during COVID-19(Reference Liu, Li and Liu38).

Haematopoiesis is severely disturbed during both severe and mild Zn deficiency, which was found in human and animal studies as illustrated in Fig. 2. Especially, a loss in pre-B cells and immature B cells, as well as early developmental T cells, including CD4/CD8 double positive and pre-T cells, was described for humans and rodents with Zn deficiency as diagnosed by low plasma Zn levels. These effects can be corrected by Zn supplementation, as shown in subjects over 65 years of age suffering from mild serum Zn deficiency and in obese subjects with decreased serum Zn levels(Reference Rios-Lugo, Madrigal-Arellano and Gaytán-Hernández5,Reference Prasad16,Reference Kahmann, Uciechowski and Warmuth27,Reference Prasad, Rabbani and Abbasii42Reference Hönscheid, Rink and Haase44) . Several mechanisms were described to underlie this decrease in cell numbers. Most importantly, thymus atrophy and decreased serum concentration of thymulin, which is necessary especially during maturation of T cells(Reference King, Osati-Ashtiani and Fraker45,Reference Mocchegiani, Giacconi and Costarelli46) , and lower levels of growth factors such as IL-2 (T cells) were reported in individuals with decreased serum Zn levels, and a disruption of IL-2 signalling was found when analysing cell cultures, where cellular Zn was depleted using a Zn chelator(Reference Kaltenberg, Plum and Ober-Blöbaum47). During dietary Zn deprivation in humans and rodents, a decreased ratio of type 1:type 2 T-helper cells, with reduced production of T-helper type 1 cytokines like interferon γ, is observed due to increased apoptosis(Reference Prasad16,Reference Truong-Tran, Carter and Ruffin30,Reference Hönscheid, Rink and Haase44) . Assuming that a Zn-deficient individual has fewer B cells compared with a person with a balanced Zn homoeostasis, a decreased generation of pathogen-specific antibodies can be expected. This might suggest that individuals, especially with a pre-existing severe Zn deficiency, would not be able to generate a sufficiently strong antibody response against SARS-CoV-2(Reference Anzilotti, Swan and Boisson48,Reference Fraker and King49) .

Fig. 2 Alterations in haematopoiesis are reported during zinc deficiency as well as in Corona Virus Disease 19 (COVID-19). During zinc deficiency, indicated by the red arrow, differentiation of myeloid cells, including polymorphonuclear neutrophils (PMN) and monocytes (Mo), is prioritised over development of adaptive immune cells, this especially impacts T cells (T). Amongst others, the prioritisation of myeloid cells may be explained by changes in growth factor expression: granulocyte-macrophage colony-stimulating factor (GM-CSF) and granulocyte-colony-stimulating factor (G-CSF) were described to be highly expressed, while levels of IL-2 are decreased during zinc deficiency. Furthermore, the T helper cell (TH)1:TH2 ratio is imbalanced during zinc deficiency, Th17 cell numbers are increased, while regulatory T cell (Treg) numbers were described as decreased as well as their functions. Most of those haematopoietic disturbances found during zinc deficiency are generally described for COVID-19 patients, as detailed in the text. B, B cell; BCP, B-cell progenitor; E, erythrocyte; EPO, erythropoietin; GM, granulocyte-macrophage progenitor; HSC, hematopoietic stem cell; MEP, megakaryocyte–erythroid progenitor; NK, natural killer cell; Pl, platelets; SCF, stem cell factor; TC: cytotoxic T cell; TNK, T and NK cell progenitor; TPO, Thrombopoietin.

In COVID-19 patients, especially those with severe symptoms, TH17 cell numbers were elevated, which is in line with the hyperinflammatory status of the immune system(Reference Xu, Shi and Wang41). Recent studies underline that differentiation into the main CD4+ cell subtypes is disturbed when Zn supply is low. In vivo data from patients with allergic asthma reveal that impaired Treg-mediated suppression can be correlated with decreased serum Zn levels(Reference Nurmatov, Nwaru and Devereux50). Data from in vitro differentiation experiments, using Zn-deficient compared with Zn-adequate culture medium, further strengthen the hypothesis, that development of proinflammatory TH17 cells is supported as a consequence of Zn deficiency(Reference Kulik, Maywald and Kloubert51).

In contrast to the consistently observed lymphopenia in COVID-19 patients, the numbers of myeloid cells in the blood and in lung tissue were strongly elevated. Neutrophilia was associated with the progression of severe disease to ARDS and with increased mortality therefrom, similarly to that described for bacteria-induced lung injury(Reference Huang, Wang and Li52Reference Liu, Liu and Xiang55). As blood analyses of non-survivors revealed severe lymphopenia combined with significantly elevated numbers of neutrophils, the neutrophil:lymphocyte ratio was suggested as a prognostic marker for COVID-19 patients(Reference Liu, Li and Liu38,Reference Liu, Liu and Xiang55) . Similar to the neutrophil:lymphocyte ratio shift in COVID-19 patients, the balance between adaptive and innate immune cells is shifted towards the latter during Zn deficiency. Investigation of severely Zn-deficient rodents, that were fed on a low-Zn diet, showed high numbers of neutrophils and their specific products in the bone marrow and blood compared with Zn-adequate animals(Reference Fraker and King49,Reference Fraker, King and Laakko56,Reference Sakakibara, Sato and Kawashima57) . Our own unpublished results suggest that maturation of myeloid precursors into granulocytes in Zn-deficient human cell cultures is also increased compared with cells that differentiate in Zn-adequate cell cultures, while Zn supplementation attenuates the development into mature neutrophils(Reference Tillmann, Rink and Wessels58). As an underlying mechanism, the increased response to growth factors, for example, to the granulocyte-macrophage colony-stimulating factor and granulocyte-colony-stimulating factor, can be named, as was determined in cell culture experiments where Zn was added to the culture medium(Reference Wessels, Pupke and Trotha54,Reference Aster, Barth and Rink59) .

Thrombocytopenia, which we will come back to later, and a decline in Hb are also common in COVID-19 patients(Reference Liu, Zhang and He37). Lower Hb and erythrocyte counts were found in severe COVID-19 cases compared with moderate cases. Furthermore, higher ferritin was found in severe COVID-19 cases, and a significant difference in the mean ferritin levels was found between survivors and non-survivors(Reference Taneri, Gómez-Ochoa and Llanaj60). Additional research is necessary to prove the suggested important role of anaemia and a disturbed Fe in severe cases of COVID-19. This might uncover new treatment options.

Alterations in bone marrow metabolism were related to decreased serum Zn concentrations in humans(Reference Prasad21,Reference Mir, Hossein-nezhad and Bahrami61) . This finding together with the observation that the osmotic fragility of erythrocyte membranes is elevated in animals with dietary Zn deficiency, as are levels of lipid peroxidation in mitochondrial and microsomal membranes, suggests that there might be some interconnection between Zn deficiency and anaemia(Reference Vallee and Falchuk62). Indeed, serum hypozincaemia is commonly observed in anaemic subjects(Reference Mir, Hossein-nezhad and Bahrami61,Reference Abdelhaleim, Abdo Soliman and Amer63Reference Shweta, Prantesh and Shashvat66) . However, importantly, a causal association between Zn deficiency and anaemia has so far not been established clearly and is discussed controversially. For example, serum Zn concentration was correlated with serum ferritin concentration in patients undergoing peritoneal dialysis(Reference Kaneko, Morino and Minato67). Morover, lower serum ferritin was significantly correlated with smaller sizes of Zn pools in premenopausal women, although without anaemia(Reference Yokoi, Sandstead and Egger68). Regarding the effects of adjuvant Zn therapy for improving anaemia in haemodialysis patients, Hb levels were found to increase significantly in Zn-supplemented patients compared with patients not supplemented with Zn. The authors suggest a ‘zinc deficiency anemia’, which needs further evaluation(Reference Fukushima, Horike and Fujiki64). In this regard, it should be pointed out that nutritional deficits often include several elements concomitantly, as shown for Zn and Fe, Se and others(Reference Heller, Sun and Hackler10,Reference Abdelhaleim, Abdo Soliman and Amer63,Reference Houghton, Parnell and Thomson65,Reference Gombart, Pierre and Maggini69) . Since the association of anaemia with an increased risk and severe progression of COVID-19 has not been clearly established, this will not be discussed further in this article. However, as anaemia is generally related to poor outcomes of infectious diseases(Reference Viana70), possible nutritional deficits in COVID-19 risk groups should be addressed and not only Zn but also Fe, Se and other elements might need to be supplemented if applicable.

Comparing the disturbance of haematopoiesis observed in individuals with low serum Zn levels or with COVID-19, various congruencies become apparent. As lymphopenia, neutrophilia and a decline in Hb are associated with progression to severe COVID-19, it can be hypothesised that a pre-existing severe Zn deficiency will predispose patients to stronger progression of infections with SARS-CoV-2 and that even a mild Zn deficiency should be corrected to prevent more severe progression of the viral infection. A pre-existing elevated neutrophil:lymphocyte ratio, even one of low magnitude, as during Zn deficiency, might be detrimental in the case of severe and aggressive infections such as COVID-19. At first sight, elevated numbers of innate immune cells as a first line of defence might appear beneficial. However, they are easily overrun during viral infections as a specific response, and the release of anti-viral factors and antibodies, especially by adaptive immune cells, is of major importance here. The elevated numbers of hyperactivated innate immune cells can even lead to high levels of inflammatory factors and oxidative stress causing destruction of host tissue. In SARS-CoV-infected mice, the recruitment of high numbers of monocytes and macrophages to the lungs was observed, secreting high numbers of proinflammatory cytokines and chemokines, which are associated with vascular leakage, underlining the detrimental effect of highly reactive immune cells(Reference Channappanavar, Fehr and Vijay71). Similar scenarios are suggested for SARS-CoV-2 in humans(Reference Henderson, Canna and Schulert72Reference Chen, Di and Guo74).

The next chapters will show that the alterations in immune cell counts are not the only indication of an association between Zn deficiency and COVID-19.

Pre-existing zinc deficiency could prime for the cytokine release syndrome

A frequent complication among patients with severe COVID-19 is the cytokine release syndrome, which spreads throughout the body from the focal infected area and may lead to death because of subsequent ARDS or multiple organ dysfunction syndrome and other complications(Reference Ye, Wang and Mao75,Reference Chousterman, Swirski and Weber76) . It was reported that among the COVID-19 patients, the classic serum proinflammatory cytokines like TNF-α, IL-2, IL-6, IL-7, IL-8, IL-10, granulocyte-colony-stimulating factor and C-reactive protein are elevated(Reference Henderson, Canna and Schulert72,Reference Conti, Ronconi and Caraffa73,Reference Chousterman, Swirski and Weber76) . IL-6 in particular, which is produced by lung resident macrophages and circulating immune cells(Reference Guan, Ni and Hu77Reference Vinciguerra, Romiti and Fattouch79), has been associated with severe COVID-19 and increased mortality(Reference Aziz, Fatima and Assaly80). Moreover, D-dimers, ferritin, lactate dehydrogenase, aspartate aminotransferase, alanine aminotransferase and soluble CD25 (IL-2 receptor) are increased, while fibrinogen is decreased(Reference Henderson, Canna and Schulert72).

Although there are planned and ongoing trials to counter the cytokine storm using approved antibodies such as tocilizumab (anti-IL-6 receptor), anakinra (IL-1 receptor antagonist, IL-1RA) and anti-TNF antibodies used to treat other hyperinflammatory conditions, and in spite of some benefits, so far their efficacy was not proven in large-scale, randomised controlled trials(Reference Henderson, Canna and Schulert72,Reference Di Giambenedetto, Ciccullo and Borghetti81Reference Feldmann, Maini and Woody83) , and therefore, therapeutic options are still limited.

In recent years, in vivo and in vitro data supporting the hypothesis that a pre-existing Zn deficiency augments the activation-induced inflammatory response, and results characterizing the possible underlying mechanisms, are constantly accumulating. Serum hypozincaemia was correlated with increased serum levels of, amongst others, IL-1β, IL-6, TNFα, IL-8, granulocyte-colony-stimulating factor, IL-10, IL-1RA, IL-17, C-reactive protein and calprotectin; thus, a whole battery of proinflammatory mediators is increased, especially during severe Zn deficiency, and particularly in combination with the inflammatory response to a pathogen(Reference Wessels, Pupke and Trotha54,Reference Reda, Abbas and Mohammed84Reference Wessels, Haase and Engelhardt88) . In the case of IL-1β and TNFα, Zn chelation was shown to induce epigenetic changes in the promoters of both genes in cell culture experiments. More specifically, the accessibility of regions in the DNA close to the transcriptional start site was significantly increased so that after inflammatory activation of the cells, by, for example, lipopolysaccharide, gene expression was augmented(Reference Wessels, Haase and Engelhardt88). Moreover, activation of NFκB a central player in the signalling pathways involved in the generation of inflammatory factors is increased when Zn is limiting, as found in mice with diet-induced serum hypozincaemia(Reference Bao, Liu and Lee87). Cell culture experiments using Zn-depleted medium revealed increased expression of calprotectin in myeloid precursors and mature monocytic cells(Reference Lienau, Rink and Wessels89)

Activated T cells express lower IL-2 and interferon γ mRNA levels, as was shown in vitro and observed in individuals with low serum Zn(Reference Prasad, Meftah and Abdallah90). IL-2 is essential for natural killer cell and cytotoxic T lymphocyte activity. Interferon γ is essential for killing viruses, parasites and bacteria. Thus, the decreased efficiency of the immune response in Zn-deficient subjects is easily explained(Reference Beck, Prasad and Kaplan14,Reference Prasad, Meftah and Abdallah90,Reference Tapazoglou, Prasad and Hill91) . Defects in T cell function as a consequence of Zn deficiency can also be explained by the accumulation of deoxyguanosine, which results from decreased Zn-dependent nucleoside phosphorylase activity in human lymphocytes, derived from human Zn-deficient volunteers before and after Zn supplementation(Reference Meftah and Prasad92). Serum Zn deficiency strongly affects Th1 cells, while Th2 cells are largely unaffected, and production of IL-4, IL-6 and IL-10 (Th2 cytokines) remains rather stable. However, production of interferon γ and IL-2 (Th1 cytokine) is decreased(Reference Prasad93).

Although Treg cell numbers might be constant, or even elevated, during in vitro differentiation under Zn deficiency, it was suggested that their function is disturbed(Reference Kulik, Maywald and Kloubert51). In vivo data are so far scarce, but some studies in mice suggest decreased transforming growth factor β (Treg cytokine) levels during Zn deficiency, pointing to a malfunctioning of Treg cells and thus imbalance of the immune response(Reference Finamore, Roselli and Merendino94). As Treg cells are important master regulators within the immune system, essential for tolerance and balance and differentiation of the remaining CD4+ T cell subtypes, a disturbed immune response can be expected.

Treatment of cell cultures with a Zn chelator disturbed the cytotoxic activity of natural killer cells(Reference Rolles, Maywald and Rink95,Reference Wessels, Maywald and Rink96) . Similar effects were reported for rats fed on a Zn-deficient diet(Reference Öztürk, Erbas and Imir97). This effect might decrease the killing of host cells which become infected by the virus.

A number of effects described above are due to the requirement of Zn for intracellular signal transduction and the consequent disruption of a multitude of signalling pathways when Zn supply is limited. Zn’s effect on phosphatases and kinases is central here, as is its ability to induce changes in membrane fluidity and thus receptor expression and dimerization as found in vitro and in vivo (Reference Kaltenberg, Plum and Ober-Blöbaum47,Reference Aster, Barth and Rink59,Reference Sadighi, Roshan and Moradi98) . Finally, epigenetic changes occur during Zn deficiency as described above as found in various models of Zn deficiency(Reference Wessels99,Reference Rosenkranz, Metz and Maywald100) .

In the case of IL-6, another connection to Zn deficiency has been described. A SNP was found in the IL-6 gene at position −174. It is associated with a disturbed age-related Zn deficiency, and it seems to be relevant during the regulation of Zn-related genes such as metallothioneins. The frequency of this polymorphism increases with age and offers an additional explanation for the high risk of Zn deficiency described for the elderly(Reference Mocchegiani, Giacconi and Costarelli46,Reference Wessels and Cousins101) . Interestingly, the IL-6–174 SNP was also associated with an increased risk for severe progression of and mortality from COVID-19, as was suggested previously for sepsis, but never proven up to now(Reference Kirtipal and Bharadwaj102Reference Mayor-Ibarguren, Busca-Arenzana and Robles-Marhuenda104). Individuals with this SNP could be actively supplemented with Zn, not only to help prevent severe COVID-19 but also to enable a balanced immune response in general(Reference Mocchegiani, Giacconi and Costarelli46,Reference Mariani, Neri and Cattini105) .

Glucocorticoids were suggested as a means to attenuate the cytokine storm and proposed as a treatment option during the hyperinflammatory phase of COVID-19(Reference Henderson, Canna and Schulert72). On the other hand, chronically increased glucocorticoids may augment lymphopenia(Reference Grant, Rotstein and Liu106,Reference Olnes, Kotliarov and Biancotto107) . Serum Zn deficiency was associated with chronically elevated levels of glucocorticoids, especially corticosteroids. However, data are not clear in this regard yet, and studies have been published not recommending the use of glucocorticoids during COVID-19 treatment, or at least recommend caution. Criticism of glucocorticoid use is largely based on data on SARS from 2003, where improper use of systemic corticosteroids increased the risk of osteonecrosis of the femoral head, which is, however, a classical side effect of glucocorticoid therapy and not related to the virus(Reference Chen, Tang and Tan108Reference Isidori, Arnaldi and Boscaro111). At first sight, this is one of the only consequences of Zn deficiency that might be viewed as an advantage in terms of COVID-19. In this regard, it should also be mentioned that the chronically increased glucocorticoid levels were suggested to be associated with the increased apoptosis of lymphocytes and probably also of cells of the thymus, thus explaining thymic atrophy in mice and humans with decreased serum Zn levels(Reference Garvy, King and Telford112Reference Taub and Longo114). However, those suggestions require experimental verification.

The cytokine storm is central to the progression from mild or severe disease to complications and critical illness associated with COVID-19 and should be prevented by any possible means. The hyper-inflammation is largely involved in damaging various organs, including the lung, heart, liver, kidney and probably also the intestine and the brain. Interestingly, the central nervous system, the gastrointestinal tract, lungs, liver, the epidermal, reproductive and skeletal system are clinically affected by severe Zn deficiency which causes elevation of inflammatory markers(Reference Prasad16,Reference Vallee and Falchuk62) . As the treatment of the cytokine storm is complex and the individual patient response to certain treatments is almost impossible to predict, the best option is to prevent the cytokine storm. Thus, groups that are at risk of Zn deficiency should be supplemented routinely. Of course, individuals with severe pre-existing Zn deficiency will benefit the most; however, adjusting mild Zn deficiencies is also of importance especially in individuals from COVID-19 risk groups such as the elderly, diabetic patients and individuals with heart and vascular co-morbidities.

Additional roles of Zn in the regulation of immune cell function, but perhaps not obviously relevant for what is known of SARS-CoV-2 infection, have been reviewed extensively elsewhere(Reference Wessels and Rink25,Reference Beyersmann and Haase29,Reference Hönscheid, Rink and Haase44,Reference Kulik, Maywald and Kloubert51,Reference Bao, Liu and Lee87,Reference Wessels, Maywald and Rink96,Reference Wessels99,Reference Haase and Schomburg115,Reference Gammoh and Rink116) .

Zinc deficiency and vascular complications: possible association with complications affecting multiple organs

Cardiovascular complications are frequently reported during COVID-19, especially in patients with pre-existing pathologies of the heart and vascular system, such as atherosclerosis(Reference Vinciguerra, Romiti and Fattouch79,Reference Gavriilaki and Brodsky117) . Venous, arterial and microvascular thromboses are increased in patients with COVID-19. Moreover, COVID-19-associated hypercoagulopathy closely resembles the pathophysiology and phenotype of complement-mediated thrombotic microangiopathy(Reference Gavriilaki and Brodsky117). An increase of proinflammatory cytokines, increased complement activation, endothelial dysfunction and immunothrombosis are considered to be key mechanisms of hypercoagulopathy. For instance, venous thromboembolisms, also driven by a hyperinflammatory milieu, were described in 20–31 % of severe COVID-19 cases(Reference Liu, Zhang and He37,Reference Middeldorp, Coppens and van Haaps118Reference Levi, Thachil and Iba121) . Moreover, an increased number of especially polymorphonuclear neutrophils (PMN) together with high amounts of neutrophil extracellular traps were observed in the thrombi of COVID-19 patients(Reference Nicolai, Leunig and Brambs122). Arterial embolism, including acute pulmonary embolism, ischaemic stroke and acute myocardial injury, was also increased in patients with severe SARS-CoV-2 infection(Reference Kashi, Jacquin and Dakhil123Reference Bavishi, Bonow and Trivedi126). Subsequent thrombocytopenia was associated with poor prognosis for COVID-19 patients. Concerning the endothelial dysfunction, it was proposed that direct endothelial damage can lead to an increased thrombogenic effect in the microcirculation(Reference Ziegler, Allon and Nyquist127). An impaired microcirculation can cause complications in various organs including the lung, the kidneys, the heart, the brain, the liver or the pancreas.

As already indicated, an increased activation and tissue recruitment of PMN in Zn-deficient individuals are likely(Reference Wessels, Pupke and Trotha54,Reference Knoell, Julian and Bao128,Reference Knoell, Smith and Sapkota129) . Thus, pre-existing Zn deficiency may be indirectly associated with thrombus formation. The association of pre-existing Zn deficiency with hyperinflammation was already described in this article and can also be related to an increased risk for thromboembolism. In addition, Zn is essential for various aspects of physiological coagulation and might impact thrombogenesis as well as fibrinolysis(Reference Vu, Fredenburgh and Weitz130,Reference Taylor and Pugh131) . However, Zn’s effects seem to depend on the microenvironment and might be locally restricted and temporary(Reference Vu, Fredenburgh and Weitz130). For example, Zn can be secreted by activated platelets resulting in locally increased Zn concentrations in the vicinity of a thrombus, while the systemic Zn homoeostasis remains probably rather stable. The direct effects of pre-existing Zn deficiency on coagulation are not entirely clear. Studies in Zn-deficient humans, rodents and guinea pigs revealed clotting abnormalities, impaired platelet function as well as an increased and prolonged bleeding tendency(Reference Emery, Browning and O’Dell132,Reference Gordon and O’Dell133) . Those Zn-deficiency-induced defects were reversible by Zn supplementation(Reference Marx, Krugliak and Shaklai134). In a recent in vitro study, Zn deficiency inhibited the agonist-activated production of reactive oxygen species (ROS) by platelets and decreased glutathione levels and glutathione peroxidase activity, which might result in altered thrombus formation(Reference Lopes-Pires, Ahmed and Vara135). Due to the lack of detailed and consitent data, a clear conclusion on the effects of Zn deficiency regarding fibrinolysis and coagulation cannot be drawn. However, as with many topics discussed in this review, a well-balanced Zn homoeostasis seems to be key to a physiological balance also in the example of coagulation and fibrinolysis.

The risk of developing an acute coronary syndrome during SARS-CoV-2 infection is especially increased in patients with atherosclerotic vascular disease(Reference Bonow, Fonarow and O’Gara136). The development and subsequent rupture of vulnerable plaques can result in heart attack or stroke, and subsequent heart failure and death(Reference Lopez, Keen and Lanoue137,Reference Libby138) . During the development of atherosclerosis, up-regulation of adhesion molecules on endothelial cells is one of the central events, largely involving the transcription factor NFκB. An increased activation and DNA binding of selected transcription factors during Zn deficiency were established in vitro (Reference Connell, Young and Toborek139,Reference Hennig, Meerarani and Toborek140) . In addition, the role of Zn in NFκB-related signalling has been described in various studies(Reference Bao, Liu and Lee87,Reference Wessels, Maywald and Rink96) . The association of severe pre-existing serum Zn deficiency in mice with an increased risk of atherosclerosis was additionally explained by the Zn-dependent alteration of endothelial surface markers, changes of the plasma lipid composition and the promotion of the proinflammatory milieu(Reference Bao, Prasad and Beck85,Reference Wessels, Maywald and Rink96,Reference Reiterer, MacDonald and Browning141) . Results from various in vivo and in vitro studies as summarised by Choi et al. indicate that Zn supplementation may reduce the risk of atherosclerosis and protect against myocardial infarction as well as ischaemia/reperfusion injury(Reference Choi, Liu and Pan23). The vasculitis described in COVID-19 patients resembles the reaction to infections with Varicella zoster virus, where the viral replication in the cerebral arterial wall directly triggered local inflammation(Reference Gilden, Cohrs and Mahalingam142). Zn supplementation in cell culture experiments was shown to decrease viral replication(Reference te Velthuis, van den Worm and Sims143), and Zn supplementation might thus attenuate virus-induced vasculitis. Recently, a molecular modelling study predicted an interaction of Zn with RNA-dependent, RNA-polymerase and 3C-like proteinase enzymes of SARS-CoV-2, which awaits experimental verification.

Associations of diseases such as arterial hypertension, atherosclerosis, congestive heart failure and CHD are described in both Zn deficiency and COVID-19(Reference Bao, Prasad and Beck85,Reference Tomat, de los Ángeles Costa and Arranz144Reference Little, Bhattacharya and Moreyra150) , but a causal link between Zn deficiency and the observations in COVID-19 remains to be established.

Pre-existing zinc deficiency is associated with severe progression of respiratory diseases

SARS-CoV-2 enters the human body predominantly via the respiratory tract. In healthy individuals, viral entry is hampered by the mucous-coated membrane of the alveoli as well as the immune cells and their anti-viral products protecting the lungs(Reference Carrillo, Rodríguez and Coronado151). When SARS-CoV-2 has crossed the epithelial barrier, it can elicit extensive alveolar injury and pulmonary fibrosis, which are irreversible pathological changes. The progression of mild COVID-19 to pneumonia, acute lung injury and subsequently to ARDS is the leading cause of mortality, affecting 5–10 % of the COVID-19 patients worldwide(Reference Ruan, Yang and Wang152,Reference Chen, Zhou and Dong153) .

As illustrated in Fig. 3, the expression of tight-junction proteins is decreased under Zn-deficient conditions. This as well as reduced expression of adherens junction proteins reduces the integrity of the endothelial barrier and might facilitate viral entry, as shown in a variety of studies investigating human and rodent tissue in vivo, ex vivo and in vitro (Reference Wessels, Pupke and Trotha54,Reference Reiterer, MacDonald and Browning141,Reference Roscioli, Jersmann and Lester154Reference Bao and Knoell157) . Experiments using an ex vivo model of differentiated human airway epithelium showed that exposure to Zn-depleted medium significantly augmented the down-regulation of the tight junction proteins such as Zonula Occludens-1 and Claudin-1 that was induced by cigarette smoke extract(Reference Roscioli, Jersmann and Lester154). Another study, which investigated primary human upper airway and type I/II alveolar epithelial cells that were grown in Zn-depleted compared with Zn-adequate medium, revealed that Zn deprivation augmented activation-induced proteolysis of E-cadherin and β-catenin, both adherens junction proteins(Reference Bao and Knoell157). Since intracellular Zn levels of endothelial cells largely depend on the protein-bound Zn pool in the blood serum, the cells are deprived of Zn during serum hypozincaemia. Low endothelial Zn disturbs cellular metabolism and is associated with oxidative stress. Increased serum levels of oxidised LDL and high amounts of inflammatory cytokines derived from activated monocytes are frequently observed in individuals with serum Zn deficiency, and together with the high oxidative stress, this leads to increased apoptosis of epithelial cells. Consequently, mild pre-existing Zn deficiency combined with inflammation-induced serum hypozincaemia may exacerbate epithelial barrier permeability of the lung in COVID-19 patients.

Fig. 3 Pulmonary effects observed in Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) infected patients with pre-existing zinc deficiency as compared with patients with a balanced zinc homoeostasis. Pre-existing zinc deficiency (left) was suggested to increase the number, recruitment and inflammatory potential of especially PMN to the insides of the bronchi. Lymphocyte numbers are generally decreased, most prominently affecting T helper cell (TH) cells. The zinc deficiency-related decrease in tight junction expression and the increase in endothelial cell apoptosis have several consequences. Thus, infiltration of the lung by host cells, as well as the leakage of pathogens such as SARS-CoV-2 and secondary pathogens such as Streptococcus pneumoniae into the vascular system, is frequently observed during zinc deficiency. Detailed explanations can be found in the text. For comparison, the characteristics of zinc-adequate individual are indicated on the right. Ab, antibody; B, B cell; E, erythrocyte; G-CSF, granulocyte colony-stimulating factor; GC, glucocorticoid; GM-CSF, granulocyte-macrophage CSF; MMP, matrix metalloproteinase; Mo, monocyte; Mϕ, macrophage; NET, neutrophil extracellular trap; NK, natural killer cell; Pl, platelet; PMN, polymorphonuclear neutrophil; ROS, reactive oxygen species; Tc, cytotoxic T cell; TJ, tight junction; ZA, zinc adequate; ZD, zinc deficient.

Previous investigations on SARS-CoV-1 infections revealed that phagocytic cells largely contributed to the antibody-mediated elimination of the virus(Reference Yasui, Kohara and Kitabatake158). Amongst the phagocytes, resident macrophages are constantly patrolling the lung, while high numbers of PMN are recruited during infections, abundant ones. PMN are highly reactive cells, equipped with their complete anti-microbial weaponry when they leave the bone marrow. Upon activation, they release their granular content which includes highly reactive mediators such as ROS, reactive nitrogen species, antimicrobial peptides, matrix metalloproteases that degrade extracellular matrix and more(Reference Wessels, Jansen and Rink159,Reference Cowland and Borregaard160) . Those factors are primarily secreted to destroy invading pathogens. However, if secreted in excessively high amounts, they can destroy the host tissue as well(Reference Grommes and Soehnlein161), as was suggested to explain tissue injury in SARS-CoV-2 infections.

With respect to PMN activity, the effects of Zn deficiency are not clearly defined. While some studies describe attenuated motility of PMN in moderately Zn-deficient individuals(Reference Briggs, Pedersen and Mahajan162,Reference Hasan, Rink and Haase163) , the numbers of PMN found in the infected tissues of animals with pre-existing Zn deficiency are higher compared with animals with adequate Zn supply(Reference Knoell, Julian and Bao128,Reference Knoell, Smith and Sapkota129) . Whether the defect in chemotaxis is compensated by the elevated numbers of PMN observed in Zn-deficient rodents, remains to be investigated(Reference Fraker and King49). The formation of ROS and neutrophil extracellular traps by PMN was reported to be decreased in Zn-deficient cells in culture(Reference Hasan, Rink and Haase164). Surprisingly, Zn supplementation of mice in vivo, or of human neutrophils in cell culture, also decreased activation-induced neutrophil extracellular trap formation(Reference Wessels, Pupke and Trotha54). In this context, we would like to mention that it was shown for various cell types, mostly in cell culture models, that Zn deficiency alters redox metabolism and results in oxidative stress(Reference Kloubert and Rink165Reference O’Dell169). There are several suggestions for the mechanisms responsible for the elevated ROS levels in Zn-deficient conditions, as summarised in Fig. 4. First, Zn deficiency was related to the decreased activity of enzymes which are central to ROS metabolism, such as the Cu/Zn superoxide dismutase in vitro. Here, the inactivation of enzymes due to the lack of Zn in their catalytic centre was described(Reference Lee168,Reference Ramirez, Gomez-Mejiba and Corbett170,Reference Taylor, Bettger and Bray171) . Second, expression of metallothioneins, not only the major intracellular Zn binding proteins but also an important free radical scavenger, is decreased during Zn deficiency, which was shown using various models of Zn deficiency and was recently summarised(Reference Lee168). As a third mechanism, Zn is necessary to protect the free sulfhydryl groups in proteins from oxidation. A lack of Zn might also alter the formation of intramolecular disulphide bonds, causing steric hindrance and conformational changes, which can be associated with increased activity or the loss of function of molecules involved in balancing the redox state of the cells, determined in cell culture experiments and suggested by in vivo examination of Zn-deficient animals(Reference Bray and Bettger172). In Zn-adequate conditions, Zn competes with other redox-active metal ions with similar coordination chemistry such as Cu or Fe for protein binding. The lack of Zn as competitor is a fourth suggested mechanism explaining the increased oxidative stress when Zn is limited. This was investigated for the oxidation of myoglobin and the activity of superoxide dismutase(Reference Ramirez, Gomez-Mejiba and Corbett170,Reference Hegetschweiler, Saltman and Dalvit173) . Zn also competes with Fe and Cu for binding to the NADPH oxidase and usually inhibits NADPH oxidase activity. Increased NADPH oxidase activity was reported for neuronal cells cultured in the Zn-depleted medium(Reference Aimo, Cherr and Oteiza174). In this context, Zn can bind NADPH, but not NADH, and thus inhibits NADPH-dependent enzymes in vitro (Reference Ludwig, Misiorowski and Chvapil175,Reference Verstraeten, Nogueira and Schreier176) . Moreover, Zn interferes with the Fenton reaction in vitro suppressing lipid peroxidation(Reference Friedrich, Mendes and Silva177,Reference Zago, Verstraeten and Oteiza178) . As a fifth point, Zn deprivation was associated with dysfunctions of mitochondria and the endoplasmic reticulum. Finally, Zn’s effect on gene expression might affect redox metabolism. Zn was shown to be involved in the up-regulation of several transcription factors, and some antioxidant molecules such as glutathione and detoxifying enzymes such as glutathione S-transferase and haemeoxygenase-1 mostly investigated using Zn-deficient cell cultures(Reference Verstraeten, Nogueira and Schreier176,Reference Jarosz, Olbert and Wyszogrodzka179) . The nuclear factor erythroid 2-related factor 2 can be induced by Zn, as was investigated in rats fed on a low-Zn, Zn-adequate or high-Zn diet(Reference Verstraeten, Nogueira and Schreier176,Reference Jarosz, Olbert and Wyszogrodzka179,Reference Wang, Nie and Lu180) . Whether Zn deficiency has the opposite effect to Zn supplementation remains to be explored, but in summary, the multiple mechanisms described above can explain the overall increase in ROS during Zn deficiency, which was consistently found in various models of Zn deficiency. We thus hypothesise that in combination with the infection-induced inflammation observed in COVID-19 patients, pre-existing Zn deficiency might augment the formation of ROS and reactive nitrogen species causing severe tissue damage. On the other hand, the anti-oxidative properties of Zn are widely described and accepted(Reference Choi, Liu and Pan23,Reference Hennig, Meerarani and Toborek140,Reference Abdulhamid, Beck and Millard181,Reference Cao, Duan and Zhang182) , suggesting benefits of Zn supplementation for COVID-19 patients.

Fig. 4 Effects of zinc deficiency on stress-induced changes in redox metabolism. Green arrows indicate zinc-dependent cellular functions. Red arrows illustrate the effects of zinc deficiency. A detailed description of the mechanisms underlying disturbed redox metabolism during zinc deficiency can be found in the text. AP-1, Activator protein 1; Bcl-2, B-cell lymphoma 2; CAT, catalase; COX, Cyclo-oxygenase; CRP, C-reactive protein; ER, endoplasmic reticulum; GPx, glutathione peroxidase; ICAM, intercellular adhesion molecule-1; iNOS, inducible nitric oxide synthase; MT, metallothionein; MTF, metal-responsive transcription factor-1; Ox, oxidated; MCP, monocyte chemoattractant protein; NIK, NFκB-Inducing Kinase; ROS, reactive oxygen species; SOD, superoxide dismutase; VCAM, vascular cell adhesion molecule.

COVID-19 often shows systemic effects in the patient’s tissues and organs, often resulting in multi-organ failure and high death rates(Reference Bencivenga, Rengo and Varricchi3,Reference Chen, Di and Guo74,Reference Cavalli, Luca and Campochiaro82,Reference Petrilli, Jones and Yang183) . Furthermore, ‘septic shock’ is another cause of mortality from SARS-CoV-2 and is currently observed in 4–8 % of COVID-19 patients(Reference Huang, Wang and Li52,Reference Wang, Hu and Hu53,Reference Rodriguez-Morales, Cardona-Ospina and Gutiérrez-Ocampo184) . When reading all the articles on COVID-19 discussing the symptoms in individuals undergoing mild compared with severe viral disease, one cannot help but notice the parallels to mild bacterial infections compared with bacterial sepsis and its progression to ARDS(Reference Liu, Zhang and He37,Reference Grommes and Soehnlein161,Reference Matthay, Zemans and Zimmerman185) . Regarding bacterial sepsis, various studies in animals and humans describe an association of disease progression and mortality in relation to the Zn status, which could be extrapolated to COVID-19. It was shown that pre-existing Zn deficiency was a prerequisite for the progression from mild inflammation to pneumonia and severe sepsis in mice. Severity of disease was monitored by analysing the serum levels of proinflammatory cytokines (i.e. assessment of the cytokine storm) and damage to the lungs, the liver and the kidney. Also, serum Zn concentrations were inversely correlated with sepsis severity. Thus, serum Zn was suggested as a prognostic marker for mortality in septic mice, pigs, adult humans and infants. In critically ill children, complications of sepsis, the necessity for mechanical ventilation and resulting mortality rates were correlated with low serum Zn levels.(Reference Wessels and Cousins101,Reference Knoell, Julian and Bao128,Reference Knoell, Smith and Sapkota129,Reference Hoeger, Simon and Beeker186Reference Alker and Haase193) . Moreover, Boudreault et al. revealed that pre-existing Zn deficiency primes the lungs for severe complications derived from mechanical ventilation, including the progression from acute lung injury to ARDS(Reference Boudreault, Pinilla-Vera and Englert194). In cystic fibrosis, Zn deficiency, caused by a splice switch in the Zn Importer ZIP2, caused hypersecretion of the glycoprotein mucin in airway epithelial cells, significantly increasing disease severity(Reference Kamei, Fujikawa and Nohara195). Pre-existing serum Zn deficiency was implicated to be responsible for the high incidence of pneumonia in elderly, hospitalised patients(Reference Barnett, Hamer and Meydani192,Reference Bhat, Rather and Dhobi196,Reference Meydani, Barnett and Dallal197) . Enhanced infection and virulence of Streptococcus pneumoniae in Zn-deficient mice were reported. In addition to disrupted epithelial barriers and inadequate immune response, the enhanced virulence was explained by the sensitivity of S. pneumonia to Zn intoxication, reduced during Zn deficiency(Reference Eijkelkamp, Morey and Neville189). Direct effects of Zn deficiency on viral replication have not been addressed to date. Finally, the correlation between Zn deficiency and infection severity may be due to reverse causality, that is, the negative effects that inflammation has on serum Zn concentration. We thus suggest that when the serum Zn levels fall below a certain threshold, the inflammatory response will be self-perpetuating. Again, most tissue damage and detrimental consequences can be expected for patients with pre-existing severe Zn deficiency, but in view of the manifold effects of already mild deficiency, normalising the Zn status offers an easy and cost-efficient approach to reduce disease symptoms.

The hypothesis that Zn deficiency is a risk factor for severe COVID-19 progression and the development of pneumonia and ARDS is supported by successful supplementation studies using Zn to prevent or attenuate respiratory diseases, as we summarised previously(Reference Wessels, Rolles and Rink6). Moreover, first data indicating the congruency of low-Zn status of COVID-19 patients as well as the inverse correlation between serum Zn levels and COVID-19 severity were recently published(Reference Heller, Sun and Hackler10,Reference Vogel-González, Talló-Parra and Herrera-Fernández198) . However, the low serum Zn levels might, once more, be the result of the severe inflammatory response elicited by the virus(Reference McDonald, Suchdev and Krebs18). Clear data on possible pre-existing serum Zn defiicencies are still lacking.

Disrupted epithelial barrier integrity during zinc deficiency: opening the way for Severe Acute Respiratory Syndrome-Coronavirus-2 and co-infections

Evidence is accumulating that, in addition to attacking the lungs and the respiratory tract, SARS-CoV-2 frequently damages other organs (heart, vessels, nerves/brain, kidneys and skin). Disruption of tissue barriers is an integral part of the pathophysiology of infectious diseases, as it facilitates distribution of the pathogen within the body(Reference Carrillo, Rodríguez and Coronado151). The effects of Zn deficiency, described above regarding the lung endothelial barrier, were similarly described for other endothelial layers, including those of kidney, liver, intestine and brain.

It should also be mentioned that the expression of ACE2, lately also called ‘SARS-CoV-2 receptor’, is not limited to the lungs, that is, the goblet and ciliated epithelial cells of the upper airways, alveolar (Type II) epithelial cells and cells of the pulmonary vasculature. ACE2 is also expressed on migratory angiogenic cells, and vascular smooth muscle cells, cardiofibroblasts, cardiomyocytes, pericytes and epicardial adipose cells of the heart; glomerular endothelial cells, podocytes and proximal tubule epithelial cells of the kidneys; cholangiocytes and hepatocytes of the liver; pigmented epithelial cells, rod and cone photoreceptor cells and Müller glial cells of the retina; enterocytes of the intestines and on cells from circumventricular organs of the central nervous system. Binding of SARS-CoV-2 was claimed to result in the loss of ACE2 from the cell surface due to receptor endocytosis and proteolytic cleavage(Reference Gheblawi, Wang and Viveiros199). On the other hand, ADAM17-mediated ACE2 shedding facilitates SARS-CoV-1 entry and induces tissue damage by TNF-α production, which remains to be shown for SARS-CoV-2(Reference Haga, Yamamoto and Nakai-Murakami200). However, disturbed ACE2 expression levels on the cell surface and increased viral entry result from both scenarios. Amongst the normal physiological functions of the ACE2 system are protection against heart failure, myocardial infarction and hypertension. This can explain heart-related COVID-19 complications. Furthermore, defects in the ACE2 system were associated with lung disease, diabetes mellitus and gut dysfunction(Reference Gheblawi, Wang and Viveiros199). ACE2 is a Zn-metalloenzyme, and its normal function is therefore Zn-dependent. Thus, a likely explanation for the association of pre-existing Zn deficiency with COVID-19 complications is the decreased ACE2 activity reported for animals fed on a low-Zn diet(Reference Reeves and O’Dell201,Reference Apgar and Everett202) . ACE activity was even suggested as a biomarker for moderate Zn deficiency in patients with idiopathic taste impairment(Reference Takeda, Takaoka and Ueda203). A Zn deficiency-related, mildly restricted ACE2 activity might not result in clinical symptoms. However, if ACE2 activity is further impaired by the virus, it might fall below a certain threshold and cause vascular complications, heart problems, gut disturbances and so on. Conversely, one study reported increased ACE2 activity in the lung tissue of Zn-deficient rats(Reference Reeves and O’Dell204). Thus, further clarification is needed before conclusions can be drawn regarding the relation to COVID-19. As Zn is a structural element of ACE2, the receptor’s conformation and subsequent affinity for the virus might be altered in patients with pre-existing Zn deficiency, which remains to be tested(Reference Reeves and O’Dell201). Furthermore, Zn deficiency might impair ACE2 expression, as was reported for other Zn-containing metalloenzymes(Reference Cao, Duan and Zhang182). Zn supplementation led to decreased Sirtuin-1 activity as found in cell culture. Interestingly, Zn removal from the closely related Plasmodium falciparum Sirtuin-2 deacetylase, resulted in structural collapse and malfunction of the enzyme. Since Sirtuin-1 is involved in regulating ACE2 transcription, this might result in disturbed ACE2 expression in patients with a Zn imbalance(Reference Rosenkranz, Metz and Maywald100,Reference Min, Landry and Sternglanz205,Reference Chakrabarty and Balaram206) .

In summary, pre-existing Zn deficiency might alter ACE2 expression, structure and/or activity in a tissue-specific manner, which could affect viral entry and pre-dispose to virus-induced tissue damage, but more and detailed studies are necessary to verify those speculation.

Acute kidney injury is another complication that can cause high mortality in COVID-19 patients(Reference Chen, Shao and Hsu207). The total incidence of acute kidney injury in COVID-19 patients is estimated to be about 4–9 %, while in a retrospective study, it was demonstrated that the percentage of patients with complications can reach 37–78 %(Reference Chen, Shao and Hsu207,Reference Argenziano, Bruce and Slater208) . In addition to increased epithelial barrier permeability and the infection with the virus via ACE2, Zn deficiency was associated with renal insufficiency(Reference Bao, Prasad and Beck85). Although described for rats, severe Zn deficiency that was observed in parallel decreased the glomerular filtration rates and renal blood flow, while renal vascular resistance increased(Reference Kurihara, Yanagisawa and Sato209). The resulting renal insufficiency might be a pre-requisite for acute kidney injury and kidney failure during COVID-19. This hypothesis is supported by the finding that the role of Zn in renal function seems to be more crucial in diseased animals than in healthy ones. Tubulointerstitial nephropathy and glomerular haemodynamics were, for example, aggravated in rats with pre-existing Zn deficiency that were suffering from unilateral ureteral obstruction. Zn deficiency further increased the disease-related high expression of endothelin-1 in the glomeruli of the obstructed kidneys(Reference Yanagisawa, Moridaira and Wada210,Reference Yanagisawa, Nodera and Wada211) . Since during kidney diseases and dialysis, Zn loss is increased, Zn deficiency is self-perpetuating and a vicious circle develops causing more severe disease(Reference Cabral, Diniz and Arruda212).

Diarrhoea was reported as a consequence of COVID-19 in a high number of cases(Reference Guan, Ni and Hu77). The association of Zn deficiency with intestinal alterations and a leaky gut are well described in clinical investigations and Zn supplementation studies, and there are excellent reviews focusing on the underlying mechanism(Reference Skrovanek213Reference Maares and Haase215).

Infection routes of COVID-19 may not include the intestinal tract. However, the leaky gut increases the risk of secondary infections, and intestinal morbidities as commensals are able to enter the human body(Reference Maguire and Maguire216,Reference Anders, Andersen and Stecher217) , especially if the immune system is otherwise occupied by the response to SARS-CoV-2. During renal diseases, nutrients not only Zn but also other elements important for an effective immune response can be lost from the body together with fluids. Consequently, dehydration and deficiency of various minerals can be expected(Reference Skrovanek213,Reference Sturniolo, D’Inca and Parisi218) .

Finally, it is not without reason that Zn supplementation, especially of children in developing countries, is recommended by the WHO to prevent and treat diarrhoea, underlining the relevance of Zn for preserving a healthy gut, as a basic step towards improving the overall health status of individuals(219).

Pre-existing Zn deficiency decreases wound healing and tissue regeneration

Long-term consequences of COVID-19 including the damage to multiple organs are becoming more and more apparent. This is of course partly due to the severe damage caused by the virus but also due to slow and inefficient recovery and healing. Again, there are striking parallels between COVID-19 symptoms and impaired healing observed in Zn deficiency(Reference Kogan, Sood and Garnick220,Reference Khorasani, Hosseinimehr and Kaghazi221) , as found during ex vivo investigation of differentiated human airway epithelium and described by several research groups(Reference Little, Bhattacharya and Moreyra150,Reference Roscioli, Jersmann and Lester154) .

In Zn-deficient rats, intestinal cell proliferation and the quality of intestinal wound healing after intestinal surgery were decreased compared with Zn-adequate controls. This was explained by higher expression of matrix metalloproteinases 2, 9 and 13 and decreased expression of Ki67 (proliferation marker). In addition, the collagen type I:III ratio was reduced in the Zn-deficient animals(Reference Binnebösel, Grommes and Koenen222). Whereas collagen type III dominates the early phase of wound healing, collagen type I rather represents late phase wound healing.

When the influx of Zn into the liver after partial hepatectomy was inhibited in a knock-out mutant of the Zn importer Zip14 in mice, proliferation of hepatocytes was significantly decreased(Reference Aydemir, Sitren and Cousins223). Pre-existing Zn deficiency had similar effects regarding regeneration of heart and lungs. Moreover, Zn supplementation improved the recovery from ischaemia as for example shown in rats where Zn was added during re-perfusion or to the diet(Reference Roscioli, Jersmann and Lester154,Reference Karagulova, Yue and Moreyra224,Reference Turan and Tuncay225) .

A Zn-adequate nutrition may thus also be relevant during recovery from COVID-19.

Zinc deficiency as pre-requisite to virus-induced neuronal damage and loss in smell and taste

In healthy and Zn-adequate individuals, the brain is usually separated from most of the immune cells by the blood–brain barrier. If the blood–brain barrier is damaged, for example, due to high levels of matrix metalloproteinase-9 or other matrix-degrading factors, the brain can easily be infiltrated by immune cells as well as by pathogens, causing neuronal damage(Reference Choi, Jung and Suh226). Thus, entry of a virus into the brain and subsequent damage of the neuronal system culminating in disturbances of their sensory function might be expected during severe Zn deficiency.

Neurological complications of COVID-19 include meningitis and encephalitis, followed by delirium and coma, acute disseminated encephalomyelitis, myelitis, Guillain-Barré syndrome and cerebrovascular complications (stroke, transient ischaemic attack, central nervous system vasculitis)(Reference Ellul, Benjamin and Singh227). However, in comparison with patients with respiratory complications, the proportion of patients with neurological manifestations of COVID-19 might be rather small. Since a high percentage of the world’s population is likely to be infected with the virus, the total number of patients with neuronal complication might be expected to be high. Moreover, lifelong disabilities can result from encephalitis and stroke. Psychosis and paralysis are also discussed as COVID-19-related(Reference Ellul, Benjamin and Singh227,Reference Paterson, Brown and Benjamin228) . Subsequent health, social, care and economic costs to society will be high(Reference Ellul, Benjamin and Singh227,Reference Paterson, Brown and Benjamin228) . Although the exact mechanisms underlying the neurological disturbances in COVID-19 patients are so far not clearly defined, a combination of direct viral invasion with secondary effects of the over-responding immune system is likely.

Serum Zn deficiency has been related to neuronal conditions such as autism, depression, psychosis, Alzheimer’s disease, stroke and schizophrenia. Disturbed neurogenesis and elevated apoptosis of neuronal cells, which can result in defects in learning and memory, were described during Zn deficiency, as was shown in animals fed on a Zn-deficient diet. Retrospective studies on stroke patients also suggest a clinical significance for serum Zn deficiency(Reference Andrews229Reference Pochwat, Domin and Rafało-Ulińska235). The increase in neuronal apoptosis might involve mitochondrial p53 as well as p53-dependent caspase-mediated mechanisms as shown in vitro (Reference Seth, Corniola and Gower-Winter236). Moreover, a deficiency in synaptic Zn, achieved by Zn chelation, elevated the susceptibility to epileptic seizures in rodents(Reference Cole, Robbins and Wenzel237,Reference Blasco-Ibáñez, Poza-Aznar and Crespo238) . Also, Zn deficiency reduces the amount of Zn available for signal transmission and processing of information, considering that Zn functions as a neurotransmitter, as reviewed in detail elsewhere(Reference Weiss, Sensi and Koh239). Zn is usually packaged into synaptic vesicles of a large sub-population of excitatory neurons for the purpose of neurotransmission. In addition, Zn functions as an important neuromodulator in the olfactory bulbs in rodents(Reference Horning and Trombley240,Reference Sekler, Moran and Hershfinkel241) . Restricting the release of Zn by knocking out the Zn exporter ZnT3 inhibited cell proliferation and neuronal differentiation in the adult hippocampus in mice(Reference Choi, Hong and Jeong242). Surprisingly, Zn in the brain remains unaltered or might even be elevated and involved in Alzheimer-related plaque formation in Zn-deficient animals and humans(Reference Chowanadisai, Kelleher and Lönnerdal243Reference Datki, Galik-Olah and Janosi-Mozes245). Thus, the relevance of direct effects of Zn deficiency to explain neuronal damage and defects in brain function awaits further data to assist verification. However, ROS, reactive nitrogen species and matrix metalloproteinase-9, which can cross the blood–brain barrier, are elevated during Zn deficiency and affect blood–brain barrier integrity, thus explaining the neuronal damage that has been found in vitro and in vivo (Reference Kumar246,Reference Gilgun-Sherki, Melamed and Offen247) .

Although not in itself life threatening, descriptions of disturbed sense of smell or taste, or both, in COVID-19 patients have accumulated(Reference Beltran-Corbellini, Chico-Garcia and Martinez-Poles248Reference Russell, Moss and Rigg251). An association between Zn deficiency and the (partial) loss of smell, taste or both has been described in several studies(Reference Russell, Cox and Solomons252,Reference Lyckholm, Heddinger and Parker253) . However, underlying mechanisms are so far not clear. Thus, a connection between Zn deficiency and the disturbances in taste and smell in COVID-19 patients must be carefully analysed in future studies. Extrapolating from the literature, however, still suggests some logical associations.

The elderly: a risk group not only for Zn deficiency

The above-discussed consequences of Zn deficiency are relevant for all age groups. However, in a large number of subjects older than 65 years, co-morbidities may exist. Thus, the association of the age-related decline of serum Zn with the high susceptibility of the elderly for severe COVID-19 is hard to estimate. Instead, we would like to point out that although this article’s focus is Zn, a deficiency in other nutritional elements could also worsen COVID-19 prognosis(Reference Bencivenga, Rengo and Varricchi3,Reference Calder, Carr and Gombart254,Reference Handu, Moloney and Rozga255) . Especially, the elderly suffer not only from Zn deficiency but often from inadequate nutrition. Thus, their nutritional status should generally be checked regularly. It was estimated that the prevalence of inappropriate nutrition risk in Europe is 8·5 % in the community setting, 17·5 % in residential care and 28 % in hospitalisation for individuals ≥65 years(Reference Leij-Halfwerk, Verwijs and van Houdt256). The evidence of the relationship between inappropriate nutrition, immunosenescence and the higher morbidity and mortality from COVID-19 in elderly patients was recently discussed(Reference Bencivenga, Rengo and Varricchi3,Reference Calder, Carr and Gombart254) . Those articles may be consulted in regard to options especially for supporting the aged population in addition to Zn supplementation. The articles provide an elaboration on the impact of malnutrition on the immune system specifically of older subjects including cell-mediated immunity, cytokine production and phagocytic function(Reference Bencivenga, Rengo and Varricchi3,Reference Wessels, Rolles and Rink6,Reference Skalny, Rink and Ajsuvakova7,Reference Calder, Carr and Gombart254) .

However, we believe that Zn supplementation of groups at risk of Zn deficiency and especially in case of the elderly can significantly reduce the severity of infectious diseases such as COVID-19, especially when combined with a generally optimised and nutritious diet, and physical exercise(Reference Wessels, Rolles and Rink6,Reference Skalny, Rink and Ajsuvakova7,Reference Calder, Carr and Gombart254) .

Next step: clinical trials

Based on the available literature, this article suggests a multitude of mechanisms as to how pre-existing Zn deficiency poses a risk of higher susceptibility to SARS-CoV-2 infections and a more severe progression of disease. To test this hypothesis, clinical studies are necessary and some are already registered(257) (online Supplementary Table S1). Moreover, first clinical data support the hypothesis that serum Zn levels are decreased in COVID-19 patients and that disease severity and mortality might be inversely correlated with serum Zn concentration(Reference Heller, Sun and Hackler10,Reference Jothimani, Kailasam and Danielraj258) . A decreased serum Zn level might perhaps be expected in COVID-19 patients due to the strong inflammatory response. Indeed, in serum samples from thirty-five patients with COVID-19, Zn levels were below those from healthy controls(Reference Heller, Sun and Hackler10). Furthermore, in a study of pregnant women, COVID-19 was associated with lower serum Zn levels and serum Zn was negatively correlated with inflammatory markers(Reference Anuk, Polat and Akdas259). Thus, subjects starting out with an inherent Zn deficiency might be expected to be less well prepared for a COVID-19-induced decrease in serum Zn. In this regard, serum samples from non-survivors of COVID-19, taken at various time points, showed that the majority were below the threshold categorised as Zn-deficient. This was also noted for half of the surviving patients. The same study also found a Se deficiency in the majority of patients. The levels of Selenoprotein P and Zn in relation to the age of the subject were identified as reliable prognostic indicators for COVID-19 survival(Reference Heller, Sun and Hackler10). Analysis and correction of Se and Zn status were recommended. Another study also found that a significant number of COVID-19 patients were Zn deficient. Here, Zn-deficient patients revealed more complications, a prolonged hospital stay and higher mortality(Reference Jothimani, Kailasam and Danielraj258). Low Zn levels in COVID-19 patients at clinical admission were associated with poor disease outcomes(Reference Vogel-González, Talló-Parra and Herrera-Fernández198). Finally, in Sakai City Medical Center (Osaka, Japan), most severely ill patients with COVID-19 showed Zn deficiency. Regarding those patients, critical illness could be predicted by serum Zn values. The authors thus suggest serum Zn levels as a predictive factor for a critical illness of COVID-19(Reference Yasui, Yasui and Suzuki260). Additional studies on correlating serum Zn levels with disease severity are ongoing(261).

The data we present strongly suggest that individuals with severe pre-existing Zn deficiency should be included in potential risk groups for COVID-19. We also suggest that prophylactic Zn supplementation, addressing mild pre-existing Zn deficiency, would be more promising than using Zn for the treatment of active disease. In several registered studies, Zn supplementation of groups with high risk of close contact with SARS-CoV-2, including medical or military personnel, is being investigated. Finally, the use of Zn supplementation alone or in combination with other treatment strategies is being tested in clinical studies. First data on the benefits of Zn supplementation as monitored by improved disease status in four confirmed cases of COVID-19 which were supplemented with up to 200 mg of elemental Zn per d were recently published(Reference Finzi262). However, only a minimal effect of Zn on the survival of Zn treated (100 mg elemental Zn per day) v. untreated COVID-19 patients was found by others(Reference Yao, Paguio and Dee263). Supplementation studies using Zn together with the ionophore chloroquine have so far produced contradictory results(Reference Frontera, Rahimian and Yaghi264Reference Carlucci, Ahuja and Petrilli268).

Combining the Zn-related data from descriptive, preventive and treatment studies will be necessary to increase our knowledge of the importance of Zn homoeostasis during COVID-19 infections and for developing optimal Zn-based supplementation strategies.

Conclusion

Zn is not without reason called an ‘essential’ trace element. Although its single actions on the various cells of the human body might be small and the symptoms of mild to moderate Zn deficiency are rather subtle, the pre-existing lack of Zn in combination with a pathogen such as SARS-CoV-2 can be detrimental and life threatening. Unfortunately, the current data for COVID-19 patients do not allow to distinguish, whether the low serum Zn levels repeatedly found are elicited by virus-induced inflammation, or are reflecting a pre-existing Zn deficiency which cause a more severe disease. Irrespective of this ambiguity, it is quite obvious that groups at risk of Zn deficiency may also be at risk of severe progression of COVID-19, in which the literature on the effects of Zn deficiency, summarised in this article, emphasises. Still, this hypothesis needs to be tested experimentally in clinical studies, some of which are currently in progress. At present, the hypothesis is only supported by data derived largely from animal and cell culture models of Zn deficiency.

This article underlines the various ways as to how a vicious circle of pre-existing, low-grade Zn deficiency and mild pathogen-induced symptoms, followed by increased loss of Zn from the body and the switch to more severe symptoms and serious complications, can be generated. Especially since Zn and its deficiency can have a wide variety of individually very different effects, the consequences of pre-existing Zn deficiency in combination with a pathogen like SARS-CoV-2 that causes so many different symptoms and complications by itself are almost impossible to predict. However, as a conclusion, it can be assumed that Zn deficiency represents a risk for severe progression of SARS-CoV-2-induced disease and a high mortality therefrom. As Zn supplementation is cost-effective and can be regarded as safe, it is highly recommended to supplement individuals who are at risk of Zn deficiency. Finally, we would like to add that more attention should be paid to monitoring nutritional status, since minerals and trace elements are inevitably associated with an efficient immune response. Collaborations between the wide range of clinical and research expertise from the nutritional field along with those involved in intensive care treatment, forming a COVID-19 Nutrition Network is desirable.

Acknowledgements

We thank Wenlei Liu for great support in designing the Figures and the Table. L. Rink is a member of Zinc-Net.

This research received no specific grant from any funding agency, commercial or not-for-profit sectors.

I. W., B. R. and L. R. drafted the original manuscript and figures. A. J. S. edited language and style and contributed substantially to the revised manuscript. All authors supported writing the manuscript and designing figures and table. The manuscript, figures and the table were critically proofread by all authors.

The authors declare that there are no conflicts of interest.

Supplementary material

For supplementary material referred to in this article, please visit https://doi.org/10.1017/S0007114521000738

References

World Health Organization (2020) WHO Director-General’s opening remarks at the media briefing on COVID-19–11 March 2020. https://www.who.int/dg/speeches/detail/who-director-general-s-opening-remarks-at-the-media-briefing-on-covid-19%2D%2D-11-march-2020 (accessed July 2020).Google Scholar
World Health Organization (2020) Clinical Management of COVID-19 Interim Guidance 27 May 2020, 2019th ed. Geneva: WHO.Google Scholar
Bencivenga, L, Rengo, G & Varricchi, G (2020) Elderly at time of Coronavirus disease 2019 (COVID-19): possible role of immunosenescence and malnutrition. Geroscience 42, 10891092.CrossRefGoogle ScholarPubMed
World Health Organization (2020) Malnutrition. https://www.who.int/news-room/fact-sheets/detail/malnutrition (accessed July 2020).Google Scholar
Rios-Lugo, MJ, Madrigal-Arellano, C, Gaytán-Hernández, D, et al. (2020) Association of serum zinc levels in overweight and obesity. Biol Trace Elem Res 198, 5157.10.1007/s12011-020-02060-8CrossRefGoogle ScholarPubMed
Wessels, I, Rolles, B & Rink, L (2020) The potential impact of zinc supplementation on COVID-19 pathogenesis. Front Immunol 11, 1712.CrossRefGoogle ScholarPubMed
Skalny, AV, Rink, L, Ajsuvakova, OP, et al. (2020) Zinc and respiratory tract infections: perspectives for COVID19 (Review). Int J Mol Med 46, 1726.Google Scholar
Sandstead, HH & Freeland-Graves, JH (2014) Dietary phytate, zinc and hidden zinc deficiency. J Trace Elem Med Biol 28, 414417 CrossRefGoogle ScholarPubMed
Lowe, NM, Fekete, K & Decsi, T (2009) Methods of assessment of zinc status in humans: a systematic review. Am J Clin Nutr 89, 2040S2051S.10.3945/ajcn.2009.27230GCrossRefGoogle ScholarPubMed
Heller, RA, Sun, Q, Hackler, J, et al. (2020) Prediction of survival odds in COVID-19 by zinc, age and selenoprotein P as composite biomarker. Redox Biol 38, 101764.CrossRefGoogle ScholarPubMed
Guillard, O, Piriou, A, Gombert, J, et al. (1979) Diurnal variations of zinc, copper and magnesium in the serum of normal fasting adults. Biomedicine 31, 193194.Google ScholarPubMed
King, LE, Frentzel, JW, Mann, JJ, et al. (2005) Chronic zinc deficiency in mice disrupted T cell lymphopoiesis and erythropoiesis while B cell lymphopoiesis and myelopoiesis were maintained. J Am Coll Nutr 24, 494502.10.1080/07315724.2005.10719495CrossRefGoogle Scholar
King, LE & Fraker, PJ (2002) Zinc deficiency in mice alters myelopoiesis and hematopoiesis. J Nutr 132, 33013307.CrossRefGoogle ScholarPubMed
Beck, FW, Prasad, AS, Kaplan, J, et al. (1997) Changes in cytokine production and T cell subpopulations in experimentally induced zinc-deficient humans. Am J Physiol 272, E1002E1007.Google Scholar
Trame, S, Wessels, I, Haase, H, et al. (2018) A short 18 items food frequency questionnaire biochemically validated to estimate zinc status in humans. J Trace Elem Med Biol 49, 285295.10.1016/j.jtemb.2018.02.020CrossRefGoogle Scholar
Prasad, AS (2020) Lessons learned from experimental human model of zinc deficiency. J Immunol Res 2020, 9207279.10.1155/2020/9207279CrossRefGoogle ScholarPubMed
King, JC, Brown, KH, Gibson, RS, et al. (2015) Biomarkers of nutrition for development (BOND)-Zinc review. J Nutr 146, 858S885S.10.3945/jn.115.220079CrossRefGoogle ScholarPubMed
McDonald, CM, Suchdev, PS, Krebs, NF, et al. (2020) Adjusting plasma or serum zinc concentrations for inflammation: biomarkers Reflecting Inflammation and Nutritional Determinants of Anemia (BRINDA) project. Am J Clin Nutr 111, 927937.10.1093/ajcn/nqz304CrossRefGoogle ScholarPubMed
Likoswe, BH, Phiri, FP, Broadley, MR, et al. (2020) Inflammation adjustment by two methods decreases the estimated prevalence of zinc deficiency in Malawi. Nutrients 12, 6.10.3390/nu12061563CrossRefGoogle ScholarPubMed
Sandstead, HH (2012) Subclinical zinc deficiency impairs human brain function. J Trace Elem Med Biol 26, 7073.10.1016/j.jtemb.2012.04.018CrossRefGoogle ScholarPubMed
Prasad, AS (1985) Clinical and biochemical manifestation zinc deficiency in human subjects. J Pharmacol 16, 344352.Google ScholarPubMed
Ackland, ML & Michalczyk, A (2006) Zinc deficiency and its inherited disorders: a review. Genes Nutr 1, 4149.10.1007/BF02829935CrossRefGoogle ScholarPubMed
Choi, S, Liu, X & Pan, Z (2018) Zinc deficiency and cellular oxidative stress: prognostic implications in cardiovascular diseases. Acta Pharmacol Sin 39, 11201132.10.1038/aps.2018.25CrossRefGoogle ScholarPubMed
Hambidge, KM & Krebs, NF (2007) Zinc deficiency: a special challenge. J Nutr 137, 11011105.10.1093/jn/137.4.1101CrossRefGoogle ScholarPubMed
Wessels, I & Rink, L (2020) Micronutrients in autoimmune diseases: possible therapeutic benefits of zinc and vitamin D. J Nutr Biochem 77, 108240.10.1016/j.jnutbio.2019.108240CrossRefGoogle ScholarPubMed
Barnes, PM & Moynahan, EJ (1973) Zinc deficiency in acrodermatitis enteropathica: multiple dietary intolerance treated with synthetic diet. Proc R Soc Med 66, 327329.Google ScholarPubMed
Kahmann, L, Uciechowski, P, Warmuth, S, et al. (2008) Zinc supplementation in the elderly reduces spontaneous inflammatory cytokine release and restores T cell functions. Rejuvenation Res 11, 227237.10.1089/rej.2007.0613CrossRefGoogle ScholarPubMed
Roohani, N, Hurrell, R, Kelishadi, R, et al. (2013) Zinc and its importance for human health: an integrative review. J Res Med Sci 18, 144157.Google Scholar
Beyersmann, D & Haase, H (2001) Functions of zinc in signaling, proliferation and differentiation of mammalian cells. Biometals 14, 331341.10.1023/A:1012905406548CrossRefGoogle ScholarPubMed
Truong-Tran, AQ, Carter, J, Ruffin, RE, et al. (2001) The role of zinc in caspase activation and apoptotic cell death. Biometals 14, 315330.10.1023/A:1012993017026CrossRefGoogle ScholarPubMed
Leon, O & Roth, M (2000) Zinc fingers: DNA binding and protein–protein interactions. Biol Res 33, 2130.CrossRefGoogle ScholarPubMed
Yamasaki, S, Sakata-Sogawa, K, Hasegawa, A, et al. (2007) Zinc is a novel intracellular second messenger. J Cell Biol 177, 637645.10.1083/jcb.200702081CrossRefGoogle ScholarPubMed
Tóth, K (2011) Zinc in neurotransmission. Annu Rev Nutr 31, 139153.10.1146/annurev-nutr-072610-145218CrossRefGoogle ScholarPubMed
Caulfield, LE, Onis, M de, Blössner, M, et al. (2004) Undernutrition as an underlying cause of child deaths associated with diarrhea, pneumonia, malaria, and measles. Am J Clin Nutr 80, 193198.CrossRefGoogle Scholar
Fischer Walker, CL, Ezzati, M & Black, RE (2009) Global and regional child mortality and burden of disease attributable to zinc deficiency. Eur J Clin Nutr 63, 591597.10.1038/ejcn.2008.9CrossRefGoogle ScholarPubMed
Diao, b, Wang, c, Tan, y, et al. (2020) reduction and functional exhaustion of t cells in patients with Coronavirus disease 2019 (COVID-19). Front Immunol 11, 17.10.3389/fimmu.2020.00827CrossRefGoogle Scholar
Liu, X, Zhang, R & He, G (2020) Hematological findings in coronavirus disease 2019: indications of progression of disease. Ann Hematol 99, 14211428.10.1007/s00277-020-04103-5CrossRefGoogle ScholarPubMed
Liu, J, Li, S, Liu, J, et al. (2020) Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine 55, 102763.CrossRefGoogle ScholarPubMed
Ganji, A, Farahani, I, Khansarinejad, B, et al. (2020) Increased expression of CD8 marker on T-cells in COVID-19 patients. Blood Cells Mol Dis 83, 102437.CrossRefGoogle ScholarPubMed
Jiang, M, Guo, Y, Luo, Q, et al. (2020) T-Cell subset counts in peripheral blood can be used as discriminatory biomarkers for diagnosis and severity prediction of coronavirus disease 2019. J Infect Dis 222, 198202.10.1093/infdis/jiaa252CrossRefGoogle ScholarPubMed
Xu, Z, Shi, L, Wang, Y, et al. (2020) Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med 8, 420422.CrossRefGoogle ScholarPubMed
Prasad, AS, Rabbani, P, Abbasii, A, et al. (1978) Experimental zinc deficiency in humans. Ann Intern Med 89, 483490.10.7326/0003-4819-89-4-483CrossRefGoogle ScholarPubMed
Iwata, T, Incefy, GS, Tanaka, T, et al. (1979) Circulating thymic hormone levels in zinc deficiency. Cell Immunol 47, 100105.10.1016/0008-8749(79)90318-6CrossRefGoogle ScholarPubMed
Hönscheid, A, Rink, L & Haase, H (2009) T-lymphocytes: a target for stimulatory and inhibitory effects of zinc ions. Endocr Metab Immune Disord Drug Targets 9, 132144.10.2174/187153009788452390CrossRefGoogle ScholarPubMed
King, LE, Osati-Ashtiani, F & Fraker, PJ (2002) Apoptosis plays a distinct role in the loss of precursor lymphocytes during zinc deficiency in mice. J Nutr 132, 974979.10.1093/jn/132.5.974CrossRefGoogle Scholar
Mocchegiani, E, Giacconi, R, Costarelli, L, et al. (2008) Zinc deficiency and IL-6 -174G/C polymorphism in old people from different European countries: effect of zinc supplementation. ZINCAGE study. Exp Gerontol 43, 433444.CrossRefGoogle ScholarPubMed
Kaltenberg, J, Plum, LM, Ober-Blöbaum, JL, et al. (2010) Zinc signals promote IL-2-dependent proliferation of T cells. Eur J Immunol 40, 14961503.10.1002/eji.200939574CrossRefGoogle ScholarPubMed
Anzilotti, C, Swan, DJ, Boisson, B, et al. (2019) An essential role for the Zn2+ transporter ZIP7 in B cell development. Nat Immunol 20, 350361.CrossRefGoogle ScholarPubMed
Fraker, PJ & King, LE (2004) Reprogramming of the immune system during zinc deficiency. Annu Rev Nutr 24, 277298.10.1146/annurev.nutr.24.012003.132454CrossRefGoogle ScholarPubMed
Nurmatov, U, Nwaru, BI, Devereux, G, et al. (2012) Confounding and effect modification in studies of diet and childhood asthma and allergies. Allergy 67, 10411059.10.1111/j.1398-9995.2012.02858.xCrossRefGoogle ScholarPubMed
Kulik, L, Maywald, M, Kloubert, V, et al. (2019) Zinc deficiency drives Th17 polarization and promotes loss of Treg cell function. J Nutr Biochem 63, 1118.CrossRefGoogle ScholarPubMed
Huang, C, Wang, Y, Li, X, et al. (2020) Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 395, 497506.10.1016/S0140-6736(20)30183-5CrossRefGoogle ScholarPubMed
Wang, D, Hu, B, Hu, C, et al. (2020) Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus–Infected Pneumonia in Wuhan, China. JAMA 323, 10611069.10.1001/jama.2020.1585CrossRefGoogle ScholarPubMed
Wessels, I, Pupke, JT, Trotha, K-T von, et al. (2020) Zinc supplementation ameliorates lung injury by reducing neutrophil recruitment and activity. Thorax 75, 253261.10.1136/thoraxjnl-2019-213357CrossRefGoogle ScholarPubMed
Liu, J, Liu, Y, Xiang, P, et al. (2020) Neutrophil-to-lymphocyte ratio predicts critical illness patients with 2019 coronavirus disease in the early stage. J Transl Med 18, 206.10.1186/s12967-020-02374-0CrossRefGoogle ScholarPubMed
Fraker, PJ, King, LE, Laakko, T, et al. (2000) The dynamic link between the integrity of the immune system and zinc status. J Nutr 130, 1399S1400S.10.1093/jn/130.5.1399SCrossRefGoogle ScholarPubMed
Sakakibara, Y, Sato, S, Kawashima, Y, et al. (2011) Different recovery responses from dietary zinc-deficiency in the distribution of rat granulocytes. J Nutr Sci Vitaminol 57, 197201.10.3177/jnsv.57.197CrossRefGoogle ScholarPubMed
Tillmann, N, Rink, L & Wessels, I The role of zinc in granulopoiesis. Submitted thesis. RWTH Aachen University, Medical Faculty (unpublished data).Google Scholar
Aster, I, Barth, L-M, Rink, L, et al. (2019) Alterations in membrane fluidity are involved in inhibition of GM-CSF-induced signaling in myeloid cells by zinc. J Trace Elem Med Biol 54, 214220.10.1016/j.jtemb.2019.04.018CrossRefGoogle ScholarPubMed
Taneri, PE, Gómez-Ochoa, SA, Llanaj, E, et al. (2020) Anemia and iron metabolism in COVID-19: a systematic review and meta-analysis. Eur J Epidemiol 35, 763773.10.1007/s10654-020-00678-5CrossRefGoogle ScholarPubMed
Mir, E, Hossein-nezhad, E, Bahrami, A, et al. (2007) Serum zinc concentration could predict bone mineral density and protect osteoporosis n healthy men. Iranian J Public Health 3036.Google Scholar
Vallee, BL & Falchuk, KH (1993) The biochemical basis of zinc physiology. Physiol Rev 73, 79118.CrossRefGoogle ScholarPubMed
Abdelhaleim, AF, Abdo Soliman, JS & Amer, AY (2019) Association of zinc deficiency with iron deficiency anemia and its symptoms: results from a case-control study. Cureus 11, e3811.Google ScholarPubMed
Fukushima, T, Horike, H, Fujiki, S, et al. (2009) Zinc deficiency anemia and effects of zinc therapy in maintenance hemodialysis patients. Ther Apher Dial 13, 213219.CrossRefGoogle ScholarPubMed
Houghton, LA, Parnell, WR, Thomson, CD, et al. (2016) Serum zinc is a major predictor of anemia and mediates the effect of selenium on hemoglobin in school-aged children in a Nationally Representative Survey in New Zealand. J Nutr 146, 16701676.CrossRefGoogle Scholar
Shweta, G, Prantesh, J & Shashvat, S (2014) Isolated zinc deficiency causing severe microcytosis and sideroblastic anemia. Turk J Haematol 31, 339340.10.4274/Tjh.2012.0145CrossRefGoogle ScholarPubMed
Kaneko, S, Morino, J, Minato, S, et al. (2020) Serum zinc concentration correlates with ferritin concentration in patients undergoing peritoneal dialysis: a cross-sectional study. Front Med 7, 537586.10.3389/fmed.2020.537586CrossRefGoogle ScholarPubMed
Yokoi, K, Sandstead, HH, Egger, NG, et al. (2007) Association between zinc pool sizes and iron stores in premenopausal women without anaemia. Br J Nutr 98, 12141223.10.1017/S0007114507803394CrossRefGoogle ScholarPubMed
Gombart, AF, Pierre, A & Maggini, S (2020) A review of micronutrients and the immune system-working in harmony to reduce the risk of infection. Nutrients 12, 1.10.3390/nu12010236CrossRefGoogle Scholar
Viana, MB (2011) Anemia and infection: a complex relationship. Rev Bras Hematol Hemoter 33, 9092.10.5581/1516-8484.20110024CrossRefGoogle ScholarPubMed
Channappanavar, R, Fehr, AR, Vijay, R, et al. (2016) Dysregulated type i interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice. Cell Host Microbe 19, 181193.10.1016/j.chom.2016.01.007CrossRefGoogle ScholarPubMed
Henderson, LA, Canna, SW, Schulert, GS, et al. (2020) On the alert for cytokine storm: immunopathology in COVID-19. Arthritis Rheumatol 72, 10591063.CrossRefGoogle ScholarPubMed
Conti, P, Ronconi, G, Caraffa, A, et al. (2020) Induction of pro-inflammatory cytokines (IL-1 and IL-6) and lung inflammation by Coronavirus-19 (COVI-19 or SARS-CoV-2): anti-inflammatory strategies. J Biol Regul Homeost Agents 34, 2.Google ScholarPubMed
Chen, G, Di, Wu, Guo, W, et al. (2020) Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest 130, 26202629.10.1172/JCI137244CrossRefGoogle ScholarPubMed
Ye, Q, Wang, B & Mao, J (2020) The pathogenesis and treatment of the ‘Cytokine Storm’ in COVID-19. J Infect 80, 607613.CrossRefGoogle ScholarPubMed
Chousterman, BG, Swirski, FK & Weber, GF (2017) Cytokine storm and sepsis disease pathogenesis. Semin Immunopathol 39, 517528.CrossRefGoogle ScholarPubMed
Guan, W-J, Ni, Z-Y, Hu, Y, et al. (2020) Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med 382, 17081720.CrossRefGoogle ScholarPubMed
Mehta, P, McAuley, DF, Brown, M, et al. (2020) COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet 395, 10331034.10.1016/S0140-6736(20)30628-0CrossRefGoogle ScholarPubMed
Vinciguerra, M, Romiti, S, Fattouch, K, et al. (2020) Atherosclerosis as pathogenetic substrate for Sars-Cov2 cytokine storm. J Clin Med 9, 7.CrossRefGoogle ScholarPubMed
Aziz, M, Fatima, R & Assaly, R (2020) Elevated interleukin-6 and severe COVID-19: a meta-analysis. J Med Virol 92, 22832285.CrossRefGoogle ScholarPubMed
Di Giambenedetto, S, Ciccullo, A, Borghetti, A, et al. (2020) Off-label use of tocilizumab in patients with SARS-CoV-2 infection. J Med Virol 92, 17871788.CrossRefGoogle Scholar
Cavalli, G, Luca, G de, Campochiaro, C, et al. (2020) Interleukin-1 blockade with high-dose anakinra in patients with COVID-19, acute respiratory distress syndrome, and hyperinflammation: a retrospective cohort study. Lancet Rheumatol 2, e325e331.10.1016/S2665-9913(20)30127-2CrossRefGoogle ScholarPubMed
Feldmann, M, Maini, RN, Woody, JN, et al. (2020) Trials of anti-tumour necrosis factor therapy for COVID-19 are urgently needed. Lancet 395, 14071409.10.1016/S0140-6736(20)30858-8CrossRefGoogle ScholarPubMed
Reda, R, Abbas, AA, Mohammed, M, et al. (2015) The interplay between Zinc, Vitamin D and, IL-17 in patients with Chronic Hepatitis C Liver disease. J Immunol Res 2015, 846348.10.1155/2015/846348CrossRefGoogle ScholarPubMed
Bao, B, Prasad, AS, Beck, FWJ, et al. (2010) Zinc decreases C-reactive protein, lipid peroxidation, and inflammatory cytokines in elderly subjects: a potential implication of zinc as an atheroprotective agent. Am J Clin Nutr 91, 16341641.10.3945/ajcn.2009.28836CrossRefGoogle ScholarPubMed
Bao, B, Prasad, AS, Beck, FWJ, et al. (2003) Zinc modulates mRNA levels of cytokines. Am J Physiol Endocrinol Metab 285, E1095102.10.1152/ajpendo.00545.2002CrossRefGoogle ScholarPubMed
Bao, S, Liu, M-J, Lee, B, et al. (2010) Zinc modulates the innate immune response in vivo to polymicrobial sepsis through regulation of NF-kappaB. Am J Physiol Lung Cell Mol Physiol 298, L744L754.10.1152/ajplung.00368.2009CrossRefGoogle ScholarPubMed
Wessels, I, Haase, H, Engelhardt, G, et al. (2013) Zinc deficiency induces production of the proinflammatory cytokines IL-1β and TNFα in promyeloid cells via epigenetic and redox-dependent mechanisms. J Nutr Biochem 24, 289297.10.1016/j.jnutbio.2012.06.007CrossRefGoogle ScholarPubMed
Lienau, S, Rink, L & Wessels, I (2018) The role of zinc in calprotectin expression in human myeloid cells. J Trace Elem Med Biol 49, 106112.CrossRefGoogle ScholarPubMed
Prasad, AS, Meftah, S, Abdallah, J, et al. (1988) Serum thymulin in human zinc deficiency. J Clin Invest 82, 12021210.CrossRefGoogle ScholarPubMed
Tapazoglou, E, Prasad, AS, Hill, G, et al. (1985) Decreased natural killer cell activity in patients with zinc deficiency with sickle cell disease. J Lab Clin Med 105, 1922.Google ScholarPubMed
Meftah, S & Prasad, AS (1989) Nucleotides in lymphocytes of human subjects with zinc deficiency. J Lab Clin Med 114, 114119.Google ScholarPubMed
Prasad, AS (2000) Effects of zinc deficiency on Th1 and Th2 cytokine shifts. J Infect Dis 182, S62S68.CrossRefGoogle ScholarPubMed
Finamore, A, Roselli, M, Merendino, N, et al. (2003) Zinc deficiency suppresses the development of oral tolerance in rats. J Nutr 133, 191198.10.1093/jn/133.1.191CrossRefGoogle ScholarPubMed
Rolles, B, Maywald, M & Rink, L (2018) Influence of zinc deficiency and supplementation on NK cell cytotoxicity. J Funct Foods 48, 322328.CrossRefGoogle Scholar
Wessels, I, Maywald, M & Rink, L (2017) Zinc as a gatekeeper of immune function. Nutrients 9, 12.CrossRefGoogle ScholarPubMed
Öztürk, G, Erbas, D, Imir, T, et al. (1994) Decreased natural killer (NK) cell activity in zinc-deficient rats. Gen Pharmacol 25, 14991503.CrossRefGoogle ScholarPubMed
Sadighi, A, Roshan, MM, Moradi, A, et al. (2008) The effects of zinc supplementation on serum zinc, alkaline phosphatase activity and fracture healing of bones. Saudi Med J 29, 12761279.Google ScholarPubMed
Wessels, I (2017) Epigenetics and Minerals: an Overview. Patel, Preedy (Hg.) 2017 – Handbook of Nutrition. Basel: Springer International Publishing.Google Scholar
Rosenkranz, E, Metz, CHD, Maywald, M, et al. (2016) Zinc supplementation induces regulatory T cells by inhibition of Sirt-1 deacetylase in mixed lymphocyte cultures. Mol Nutr Food Res 60, 661671.CrossRefGoogle ScholarPubMed
Wessels, I & Cousins, RJ (2015) Zinc dyshomeostasis during polymicrobial sepsis in mice involves zinc transporter Zip14 and can be overcome by zinc supplementation. Am J Physiol Gastrointest Liver Physiol 309, G768G778.CrossRefGoogle ScholarPubMed
Kirtipal, N & Bharadwaj, S (2020) Interleukin 6 polymorphisms as an indicator of COVID-19 severity in humans. J Biomol Struct Dyn 13.Google ScholarPubMed
Chen, Y, Hu, Y & Song, Z (2019) The association between interleukin-6 gene -174G/C single nucleotide polymorphism and sepsis: an updated meta-analysis with trial sequential analysis. BMC Med Genet 20, 35.CrossRefGoogle ScholarPubMed
Mayor-Ibarguren, A, Busca-Arenzana, C & Robles-Marhuenda, Á (2020) A hypothesis for the possible role of zinc in the immunological pathways related to COVID-19 infection. Front Immunol 11, 1736.CrossRefGoogle ScholarPubMed
Mariani, E, Neri, S, Cattini, L, et al. (2008) Effect of zinc supplementation on plasma IL-6 and MCP-1 production and NK cell function in healthy elderly: interactive influence of +647 MT1a and -174 IL-6 polymorphic alleles. Exp Gerontol 43, 462471.CrossRefGoogle ScholarPubMed
Grant, RC, Rotstein, C, Liu, G, et al. (2020) Reducing dexamethasone antiemetic prophylaxis during the COVID-19 pandemic: recommendations from Ontario, Canada. Support Care Cancer 28, 50315036.10.1007/s00520-020-05588-6CrossRefGoogle ScholarPubMed
Olnes, MJ, Kotliarov, Y, Biancotto, A, et al. (2016) Effects of systemically administered hydrocortisone on the human immunome. Sci Rep 6, 23002.CrossRefGoogle ScholarPubMed
Chen, R-C, Tang, X-P, Tan, S-Y, et al. (2006) Treatment of severe acute respiratory syndrome with glucosteroids: the Guangzhou experience. Chest 129, 14411452.CrossRefGoogle ScholarPubMed
Guo, KJ, Zhao, FC, Guo, Y, et al. (2014) The influence of age, gender and treatment with steroids on the incidence of osteonecrosis of the femoral head during the management of severe acute respiratory syndrome: a retrospective study. Bone Joint J 96, 259262.CrossRefGoogle ScholarPubMed
Tang, C, Wang, Y, Lv, H, et al. (2020) Caution against corticosteroid-based COVID-19 treatment. Lancet 395, 17591760.CrossRefGoogle ScholarPubMed
Isidori, AM, Arnaldi, G, Boscaro, M, et al. (2020) COVID-19 infection and glucocorticoids: update from the Italian Society of Endocrinology Expert Opinion on steroid replacement in adrenal insufficiency. J Endocrinol Invest 43, 11411147.CrossRefGoogle Scholar
Garvy, BA, King, LE, Telford, WG, et al. (1993) Chronic elevation of plasma corticosterone causes reductions in the number of cycling cells of the B lineage in murine bone marrow and induces apoptosis. Immunology 80, 587592.Google Scholar
Cook, RT, Schlueter, AJ, Coleman, RA, et al. (2007) Thymocytes, pre-B cells, and organ changes in a mouse model of chronic ethanol ingestion--absence of subset-specific glucocorticoid-induced immune cell loss. Alcohol: Clin Exp Res 31, 17461758.CrossRefGoogle Scholar
Taub, DD & Longo, DL (2005) Insights into thymic aging and regeneration. Immunol Rev 205, 7293.CrossRefGoogle ScholarPubMed
Haase, H & Schomburg, L (2019) You’d better zinc-trace element homeostasis in infection and inflammation. Nutrients 11, 9.CrossRefGoogle ScholarPubMed
Gammoh, NZ & Rink, L (2017) Zinc in Infection and Inflammation. Nutrients 9, 6.10.3390/nu9060624CrossRefGoogle ScholarPubMed
Gavriilaki, E & Brodsky, RA (2020) Severe COVID-19 infection and thrombotic microangiopathy: success doesn’t come easily. Br J Haematol 189, e227e230.CrossRefGoogle Scholar
Middeldorp, S, Coppens, M, van Haaps, TF, et al. (2020) Incidence of venous thromboembolism in hospitalized patients with COVID-19. J Thromb Haemost 18, 19952002.10.1111/jth.14888CrossRefGoogle ScholarPubMed
Cui, S, Chen, S, Li, X, et al. (2020) Prevalence of venous thromboembolism in patients with severe novel coronavirus pneumonia. J Thromb Haemost 18, 14211424.10.1111/jth.14830CrossRefGoogle ScholarPubMed
Lefrançais, E, Ortiz-Muñoz, G, Caudrillier, A, et al. (2017) The lung is a site of platelet biogenesis and a reservoir for haematopoietic progenitors. Nature 544, 105109.CrossRefGoogle Scholar
Levi, M, Thachil, J, Iba, T, et al. (2020) Coagulation abnormalities and thrombosis in patients with COVID-19. Lancet Haematol 7, e438e440.10.1016/S2352-3026(20)30145-9CrossRefGoogle ScholarPubMed
Nicolai, L, Leunig, A, Brambs, S, et al. (2020) Immunothrombotic dysregulation in COVID-19 pneumonia is associated with respiratory failure and coagulopathy. Circulation 142, 11761189.10.1161/CIRCULATIONAHA.120.048488CrossRefGoogle ScholarPubMed
Kashi, M, Jacquin, A, Dakhil, B, et al. (2020) Severe arterial thrombosis associated with Covid-19 infection. Thromb Res 192, 7577.CrossRefGoogle ScholarPubMed
Martin, AI & Rao, G (2020) COVID-19: a potential risk factor for acute pulmonary embolism. Methodist Debakey Cardiovasc J 16, 155157.10.14797/mdcj-16-2-155CrossRefGoogle ScholarPubMed
Beyrouti, R, Adams, ME, Benjamin, L, et al. (2020) Characteristics of ischaemic stroke associated with COVID-19. J Neurol Neurosurg Psychiatr 91, 889891.10.1136/jnnp-2020-323586CrossRefGoogle ScholarPubMed
Bavishi, C, Bonow, RO, Trivedi, V, et al. (2020) Acute myocardial injury in patients hospitalized with COVID-19 infection: a review. Prog Cardiovasc Dis 63, 682689.10.1016/j.pcad.2020.05.013CrossRefGoogle ScholarPubMed
Ziegler, CG, Allon, SJ, Nyquist, SK, et al. (2020) SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell 181, 10161035.10.1016/j.cell.2020.04.035CrossRefGoogle ScholarPubMed
Knoell, DL, Julian, MW, Bao, S, et al. (2009) Zinc deficiency increases organ damage and mortality in a murine model of polymicrobial sepsis. Crit Care Med 37, 13801388.CrossRefGoogle Scholar
Knoell, DL, Smith, DA, Sapkota, M, et al. (2019) Insufficient zinc intake enhances lung inflammation in response to agricultural organic dust exposure. J Nutr Biochem 70, 5664.10.1016/j.jnutbio.2019.04.007CrossRefGoogle ScholarPubMed
Vu, TT, Fredenburgh, JC & Weitz, JI (2013) Zinc: an important cofactor in haemostasis and thrombosis. Thromb Haemost 109, 421430.CrossRefGoogle ScholarPubMed
Taylor, KA & Pugh, N (2016) The contribution of zinc to platelet behaviour during haemostasis and thrombosis. Metallomics 8, 144155.10.1039/C5MT00251FCrossRefGoogle ScholarPubMed
Emery, MP, Browning, JD & O’Dell, BL (1990) Impaired hemostasis and platelet function in rats fed low zinc diets based on egg white protein. J Nutr 120, 10621067.CrossRefGoogle ScholarPubMed
Gordon, PR & O’Dell, BL (1983) Zinc deficiency and impaired platelet aggregation in guinea pigs. J Nutr 113, 239245.CrossRefGoogle ScholarPubMed
Marx, G, Krugliak, J & Shaklai, M (1991) Nutritional zinc increases platelet reactivity. Am J Hematol 38, 161165.CrossRefGoogle ScholarPubMed
Lopes-Pires, ME, Ahmed, NS, Vara, D, et al. (2020) Zinc regulates reactive oxygen species generation in platelets. Platelets 110.Google Scholar
Bonow, RO, Fonarow, GC, O’Gara, PT, et al. (2020) Association of coronavirus disease 2019 (covid-19) with myocardial injury and mortality. JAMA Cardiol 5, 751753.CrossRefGoogle ScholarPubMed
Lopez, V, Keen, CL & Lanoue, L (2008) Prenatal zinc deficiency: influence on heart morphology and distribution of key heart proteins in a rat model. Biol Trace Elem Res 122, 238255.CrossRefGoogle ScholarPubMed
Libby, P (2006) Inflammation and cardiovascular disease mechanisms. Am J Clin Nutr 83, 456S460S.CrossRefGoogle ScholarPubMed
Connell, P, Young, VM, Toborek, M, et al. (1997) Zinc attenuates tumor necrosis factor-mediated activation of transcription factors in endothelial cells. J Am Coll Nutr 16, 411417.10.1080/07315724.1997.10718706CrossRefGoogle ScholarPubMed
Hennig, B, Meerarani, P, Toborek, M, et al. (1999) Antioxidant-like properties of zinc in activated endothelial cells. J Am Coll Nutr 18, 152158.CrossRefGoogle ScholarPubMed
Reiterer, G, MacDonald, R, Browning, JD, et al. (2005) Zinc deficiency increases plasma lipids and atherosclerotic markers in LDL-receptor-deficient mice. J Nutr 135, 21142118.CrossRefGoogle ScholarPubMed
Gilden, D, Cohrs, RJ, Mahalingam, R, et al. (2009) Varicella zoster virus vasculopathies: diverse clinical manifestations, laboratory features, pathogenesis, and treatment. Lancet Neurol 8, 731740.CrossRefGoogle ScholarPubMed
te Velthuis, AJW, van den Worm, SHE, Sims, AC, et al. (2010) Zn(2+) inhibits coronavirus and arterivirus RNA polymerase activity in vitro and zinc ionophores block the replication of these viruses in cell culture. PLoS Pathog 6, e1001176.CrossRefGoogle ScholarPubMed
Tomat, AL, de los Ángeles Costa, M & Arranz, CT (2011) Zinc restriction during different periods of life: influence in renal and cardiovascular diseases. Nutrition 27, 392398.CrossRefGoogle ScholarPubMed
Tubek, S (2005) Zinc content in lymphocytes and the activity of zinc ion efflux from lymphocytes in primary arterial hypertension. BTER 107, 89100.CrossRefGoogle ScholarPubMed
Tubek, S (2007) Role of zinc in regulation of arterial blood pressure and in the etiopathogenesis of arterial hypertension. Biol Trace Elem Res 117, 3951.10.1007/BF02698082CrossRefGoogle ScholarPubMed
Eide, DJ (2011) The oxidative stress of zinc deficiency. Metallomics 3, 11241129.CrossRefGoogle ScholarPubMed
Prasad, AS (2009) Impact of the discovery of human zinc deficiency on health. J Am Coll Nutr 28, 257265.CrossRefGoogle ScholarPubMed
Cohen, N & Golik, A (2006) Zinc balance and medications commonly used in the management of heart failure. Heart Fail Rev 11, 1924.10.1007/s10741-006-9189-1CrossRefGoogle ScholarPubMed
Little, PJ, Bhattacharya, R, Moreyra, AE, et al. (2010) Zinc and cardiovascular disease. Nutr 26, 10501057.CrossRefGoogle ScholarPubMed
Carrillo, JLM, Rodríguez, FPC, Coronado, OG, et al. (2017) Physiology and Pathology of Innate Immune Response Against Pathogens. Physiology and Pathology of Immunology. London: InTechOpen.Google Scholar
Ruan, Q, Yang, K, Wang, W, et al. (2020) Correction to: clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med 46, 12941297.CrossRefGoogle Scholar
Chen, N, Zhou, M, Dong, X, et al. (2020) Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet 395, 507513.CrossRefGoogle ScholarPubMed
Roscioli, E, Jersmann, HP, Lester, S, et al. (2017) Zinc deficiency as a codeterminant for airway epithelial barrier dysfunction in an ex vivo model of COPD. Int J Chron Obstruct Pulmon Dis 12, 35033510.CrossRefGoogle Scholar
Hennig, B, Wang, Y, Ramasamy, S, et al. (1992) Zinc deficiency alters barrier function of cultured porcine endothelial cells. J Nutr 122, 12421247.CrossRefGoogle ScholarPubMed
Hennig, B, Wang, Y, Ramasamy, S, et al. (1993) Zinc protects against tumor necrosis factor-induced disruption of porcine endothelial cell monolayer integrity. J Nutr 123, 10031009.Google ScholarPubMed
Bao, S & Knoell, DL (2006) Zinc modulates cytokine-induced lung epithelial cell barrier permeability. Am J Physiol Lung Cell Mol Physiol 291, L113241.CrossRefGoogle ScholarPubMed
Yasui, F, Kohara, M, Kitabatake, M, et al. (2014) Phagocytic cells contribute to the antibody-mediated elimination of pulmonary-infected SARS coronavirus. Virol 454–455, 157168.CrossRefGoogle Scholar
Wessels, I, Jansen, J, Rink, L, et al. (2010) Immunosenescence of polymorphonuclear neutrophils. Sci World J 10, 145160.CrossRefGoogle ScholarPubMed
Cowland, JB & Borregaard, N (2016) Granulopoiesis and granules of human neutrophils. Immunol Rev 273, 1128.CrossRefGoogle ScholarPubMed
Grommes, J & Soehnlein, O (2011) Contribution of neutrophils to acute lung injury. Mol Med 17, 293307.CrossRefGoogle ScholarPubMed
Briggs, WA, Pedersen, MM, Mahajan, SK, et al. (1982) Lymphocyte and granulocyte function in zinc-treated and zinc-deficient hemodialysis patients. Kidney Int 21, 827832.CrossRefGoogle ScholarPubMed
Hasan, R, Rink, L & Haase, H (2016) Chelation of free Zn(2)(+) impairs chemotaxis, phagocytosis, oxidative burst, degranulation, and cytokine production by neutrophil granulocytes. Biol Trace Elem Res 171, 7988.CrossRefGoogle ScholarPubMed
Hasan, R, Rink, L & Haase, H (2013) Zinc signals in neutrophil granulocytes are required for the formation of neutrophil extracellular traps. Innate Immun 19, 253264.CrossRefGoogle ScholarPubMed
Kloubert, V & Rink, L (2015) Zinc as a micronutrient and its preventive role of oxidative damage in cells. Food Funct 6, 31953204.CrossRefGoogle ScholarPubMed
Maret, W (2009) Molecular aspects of human cellular zinc homeostasis: redox control of zinc potentials and zinc signals. Biometals 22, 149157.CrossRefGoogle ScholarPubMed
Prasad, AS, Bao, B, Beck, FWJ, et al. (2004) Antioxidant effect of zinc in humans. Free Radic Biol Med 37, 11821190.10.1016/j.freeradbiomed.2004.07.007CrossRefGoogle ScholarPubMed
Lee, SR (2018) Critical role of zinc as either an antioxidant or a prooxidant in cellular systems. Oxid Med Cell Longev 2018, 9156285.CrossRefGoogle ScholarPubMed
O’Dell, BL (2000) Role of zinc in plasma membrane function. J Nutr 130, 1432S1436S.10.1093/jn/130.5.1432SCrossRefGoogle ScholarPubMed
Ramirez, DC, Gomez-Mejiba, SE, Corbett, JT, et al. (2009) Cu, Zn-Superoxide dismutase-driven free radical modifications: copper- and carbonate radical anion-initiated protein radical chemistry. Biochem J 417, 341353.CrossRefGoogle ScholarPubMed
Taylor, CG, Bettger, WJ & Bray, TM (1988) Effect of dietary zinc or copper deficiency on the primary free radical defense system in rats. J Nutr 118, 613621.CrossRefGoogle ScholarPubMed
Bray, TM & Bettger, WJ (1990) The physiological role of zinc as an antioxidant. Free Radical Biol Med 8, 281291.10.1016/0891-5849(90)90076-UCrossRefGoogle ScholarPubMed
Hegetschweiler, K, Saltman, P, Dalvit, C, et al. (1987) Kinetics and mechanisms of the oxidation of myoglobin by Fe(III) and Cu(II) complexes. Biochim Biophys Acta 912, 384397.CrossRefGoogle ScholarPubMed
Aimo, L, Cherr, GN & Oteiza, PI (2010) Low extracellular zinc increases neuronal oxidant production through nadph oxidase and nitric oxide synthase activation. Free Radic Biol Med 48, 15771587.10.1016/j.freeradbiomed.2010.02.040CrossRefGoogle ScholarPubMed
Ludwig, JC, Misiorowski, RL, Chvapil, M, et al. (1980) Interaction of zinc ions with electron carrying coenzymes NADPH and NADH. Chem Biol Interact 30, 2534.10.1016/0009-2797(80)90111-8CrossRefGoogle ScholarPubMed
Verstraeten, SV, Nogueira, LV, Schreier, S, et al. (1997) Effect of trivalent metal ions on phase separation and membrane lipid packing: role in lipid peroxidation. Arch Biochem Biophys 338, 121127.CrossRefGoogle ScholarPubMed
Friedrich, LC, Mendes, MA, Silva, VO, et al. (2012) Mechanistic implications of zinc(II) ions on the degradation of phenol by the fenton reaction. J Braz Chem Soc 23, 13721377.10.1590/S0103-50532012000700022CrossRefGoogle Scholar
Zago, MP, Verstraeten, SV & Oteiza, PI (2000) Zinc in the prevention of Fe2+-initiated lipid and protein oxidation. Biol Res 33, 143150.CrossRefGoogle ScholarPubMed
Jarosz, M, Olbert, M, Wyszogrodzka, G, et al. (2017) Antioxidant and anti-inflammatory effects of zinc. Zinc-dependent NF-κB signaling. Inflammopharmacology 25, 1124.CrossRefGoogle ScholarPubMed
Wang, S, Nie, P, Lu, X, et al. (2020) Nrf2 participates in the anti-apoptotic role of zinc in Type 2 diabetic nephropathy through Wnt/β-catenin signaling pathway. J Nutr Biochem 84, 108451.CrossRefGoogle ScholarPubMed
Abdulhamid, I, Beck, FWJ, Millard, S, et al. (2008) Effect of zinc supplementation on respiratory tract infections in children with cystic fibrosis. Pediatr Pulmonol 43, 281287.CrossRefGoogle ScholarPubMed
Cao, J-W, Duan, S-Y, Zhang, H-X, et al. (2019) Zinc deficiency promoted fibrosis via ROS and TIMP/MMPs in the myocardium of mice. Biol Trace Elem Res 196, 145152.CrossRefGoogle ScholarPubMed
Petrilli, CM, Jones, SA, Yang, J, et al. (2020) Factors associated with hospital admission and critical illness among 5279 people with coronavirus disease 2019 in New York City: prospective cohort study. BMJ 369, m1966.CrossRefGoogle ScholarPubMed
Rodriguez-Morales, AJ, Cardona-Ospina, JA, Gutiérrez-Ocampo, E, et al. (2020) Clinical, laboratory and imaging features of COVID-19: a systematic review and meta-analysis. Travel Med Infect Dis 34, 101623.CrossRefGoogle ScholarPubMed
Matthay, MA, Zemans, RL, Zimmerman, GA, et al. (2019) Acute respiratory distress syndrome. Nat Rev Dis Primers 5, 18.CrossRefGoogle ScholarPubMed
Hoeger, J, Simon, T-P, Beeker, T, et al. (2017) Persistent low serum zinc is associated with recurrent sepsis in critically ill patients: a pilot study. PLoS One 12, e0176069.10.1371/journal.pone.0176069CrossRefGoogle ScholarPubMed
Saleh, NY & Abo El Fotoh, WMM (2018) Low serum zinc level: the relationship with severe pneumonia and survival in critically ill children. Int J Clin Pract 72, e13211.CrossRefGoogle ScholarPubMed
Besecker, BY, Exline, MC, Hollyfield, J, et al. (2011) A comparison of zinc metabolism, inflammation, and disease severity in critically ill infected and noninfected adults early after intensive care unit admission. Am J Clin Nutr 93, 13561364.CrossRefGoogle ScholarPubMed
Eijkelkamp, BA, Morey, JR, Neville, SL, et al. (2019) Dietary zinc and the control of Streptococcus pneumoniae infection. PLoS Pathog 15, e1007957.CrossRefGoogle ScholarPubMed
Visalakshy, J, Surendran, S, Pillai, MPG, et al. (2017) Could plasma zinc be a predictor for mortality and severity in sepsis syndrome? Int J Res Med Sci 5, 3929.CrossRefGoogle Scholar
Saner, G, Uğur Baysal, S, Ünüvar, E, et al. (2000) Serum zinc, copper levels, and copper/zinc ratios in infants with sepsis syndrome. J Trace Elem Exp Med 13, 265270.3.0.CO;2-D>CrossRefGoogle Scholar
Barnett, JB, Hamer, DH & Meydani, SN (2010) Low zinc status: a new risk factor for pneumonia in the elderly? Nutr Rev 68, 3037.10.1111/j.1753-4887.2009.00253.xCrossRefGoogle ScholarPubMed
Alker, W & Haase, H (2018) Zinc and Sepsis. Nutrients 10, 8.CrossRefGoogle ScholarPubMed
Boudreault, F, Pinilla-Vera, M, Englert, JA, et al. (2017) Zinc deficiency primes the lung for ventilator-induced injury. JCI Insight 2, 11.CrossRefGoogle ScholarPubMed
Kamei, S, Fujikawa, H, Nohara, H, et al. (2018) Zinc deficiency via a splice switch in zinc importer ZIP2/SLC39A2 causes cystic fibrosis-associated MUC5AC hypersecretion in airway epithelial cells. EBioMedicine 27, 304316.CrossRefGoogle Scholar
Bhat, MH, Rather, AB, Dhobi, GN et al. (2016) Zinc Levels in community acquired pneumonia in hospitalized patients; a case control study. Egypt J Chest Dis Tuberculosis 65, 485489.10.1016/j.ejcdt.2015.12.020CrossRefGoogle Scholar
Meydani, SN, Barnett, JB, Dallal, GE, et al. (2007) Serum zinc and pneumonia in nursing home elderly. Am J Clin Nutr 86, 11671173.CrossRefGoogle ScholarPubMed
Vogel-González, M, Talló-Parra, M, Herrera-Fernández, V, et al. (2021) Low zinc levels at admission associates with poor clinical outcomes in SARS-CoV-2 infection. Nutrients 13, 562.CrossRefGoogle ScholarPubMed
Gheblawi, M, Wang, K, Viveiros, A, et al. (2020) Angiotensin-Converting enzyme 2: SARS-CoV-2 receptor and regulator of the renin-angiotensin system: celebrating the 20th anniversary of the discovery of ACE2. Circ Res 126, 14561474.10.1161/CIRCRESAHA.120.317015CrossRefGoogle ScholarPubMed
Haga, S, Yamamoto, N, Nakai-Murakami, C, et al. (2008) Modulation of TNF-alpha-converting enzyme by the spike protein of SARS-CoV and ACE2 induces TNF-α production and facilitates viral entry. Proc Natl Acad Sci USA 105, 78097814.CrossRefGoogle ScholarPubMed
Reeves, PG & O’Dell, BL (1986) Effects of dietary zinc deprivation on the activity of angiotensin-converting enzyme in serum of rats and guinea pigs. J Nutr 116, 128134.CrossRefGoogle ScholarPubMed
Apgar, J & Everett, GA (1991) Low zinc intake affects maintenance of pregnancy in guinea pigs. J Nutr 121, 192200.CrossRefGoogle ScholarPubMed
Takeda, N, Takaoka, T, Ueda, C, et al. (2004) Zinc deficiency in patients with idiopathic taste impairment with regard to angiotensin converting enzyme activity. Auris Nasus Larynx 31, 425428.CrossRefGoogle ScholarPubMed
Reeves, PG & O’Dell, BL (1988) Zinc deficiency in rats and angiotensin-converting enzyme activity: comparative effects on lung and testis. J Nutr 118, 622626.10.1093/jn/118.5.622CrossRefGoogle ScholarPubMed
Min, J, Landry, J, Sternglanz, R, et al. (2001) Crystal structure of a SIR2 Homolog–NAD complex. Cell 105, 269279.CrossRefGoogle ScholarPubMed
Chakrabarty, SP & Balaram, H (2010) Reversible binding of zinc in Plasmodium falciparum Sir2: structure and activity of the apoenzyme. Biochim Biophys Acta 1804, 17431750.CrossRefGoogle ScholarPubMed
Chen, Y-T, Shao, S-C, Hsu, C-K, et al. (2020) Incidence of acute kidney injury in COVID-19 infection: a systematic review and meta-analysis. Crit Care 24, 346.CrossRefGoogle ScholarPubMed
Argenziano, MG, Bruce, SL, Slater, CL, et al. (2020) Characterization and clinical course of 1000 patients with coronavirus disease 2019 in New York: retrospective case series. BMJ 34, m1996.CrossRefGoogle Scholar
Kurihara, N, Yanagisawa, H, Sato, M, et al. (2002) Increased renal vascular resistance in zinc-deficient rats: role of nitric oxide and superoxide. Clin Exp Pharmacol Physiol 29, 10961104.CrossRefGoogle ScholarPubMed
Yanagisawa, H, Moridaira, K & Wada, O (2000) Zinc deficiency further increases the enhanced expression of endothelin-1 in glomeruli of the obstructed kidney. Kidney Int 58, 575586.CrossRefGoogle ScholarPubMed
Yanagisawa, H, Nodera, M & Wada, O (1998) Zinc deficiency aggravates tubulointerstitial nephropathy caused by ureteral obstruction. Biol Trace Elem Res 65, 16.CrossRefGoogle ScholarPubMed
Cabral, PC, Diniz, AdS & Arruda, IKG de (2005) Vitamin A and zinc status in patients on maintenance haemodialysis. Nephrology 10, 459463.CrossRefGoogle Scholar
Skrovanek, S (2014) Zinc and gastrointestinal disease. WJGP 5, 496.CrossRefGoogle ScholarPubMed
Wapnir, RA (2000) Zinc deficiency, malnutrition and the gastrointestinal tract. J Nutr 130, 1388S1392S.CrossRefGoogle ScholarPubMed
Maares, M & Haase, H (2020) A guide to human zinc absorption: general overview and recent advances of In Vitro intestinal models. Nutrients 12, 3.CrossRefGoogle ScholarPubMed
Maguire, M & Maguire, G (2019) Gut dysbiosis, leaky gut, and intestinal epithelial proliferation in neurological disorders: towards the development of a new therapeutic using amino acids, prebiotics, probiotics, and postbiotics. Rev Neurosci 30, 179201.CrossRefGoogle ScholarPubMed
Anders, H-J, Andersen, K & Stecher, B (2013) The intestinal microbiota, a leaky gut, and abnormal immunity in kidney disease. Kidney Int 83, 10101016.CrossRefGoogle ScholarPubMed
Sturniolo, GC, D’Inca, R, Parisi, G, et al. (1992) Taste alterations in liver cirrhosis: are they related to zinc deficiency? J Trace Elem Electrolytes Health Dis 6, 1519.Google ScholarPubMed
World Health Organization (2006) Implementing the New Recommendations of the Clinical Management of Diarrhoea. Geneva: WHO.Google Scholar
Kogan, S, Sood, A & Garnick, MS (2017) Zinc and wound healing: a review of zinc physiology and clinical applications. Wounds 29, 102106.Google ScholarPubMed
Khorasani, G, Hosseinimehr, SJ & Kaghazi, Z (2008) The alteration of plasma’s zinc and copper levels in patients with burn injuries and the relationship to the time after burn injuries. Singap Med J 49, 627630.Google Scholar
Binnebösel, M, Grommes, J, Koenen, B, et al. (2010) Zinc deficiency impairs wound healing of colon anastomosis in rats. Int J Colorectal Dis 25, 251257.CrossRefGoogle ScholarPubMed
Aydemir, TB, Sitren, HS & Cousins, RJ (2012) The zinc transporter Zip14 influences c-Met phosphorylation and hepatocyte proliferation during liver regeneration in mice. Gastroenterology 142, 15361546.e5.CrossRefGoogle ScholarPubMed
Karagulova, G, Yue, Y, Moreyra, A, et al. (2007) Protective role of intracellular zinc in myocardial ischemia/reperfusion is associated with preservation of protein kinase C isoforms. J Pharmacol Exp Ther 321, 517525.CrossRefGoogle ScholarPubMed
Turan, B & Tuncay, E (2017) Impact of labile zinc on heart function: from physiology to pathophysiology. Int J Mol Sci 18, 11.CrossRefGoogle ScholarPubMed
Choi, BY, Jung, JW & Suh, SW (2017) The emerging role of zinc in the pathogenesis of multiple sclerosis. Int J Mol Sci 18, 10.CrossRefGoogle ScholarPubMed
Ellul, MA, Benjamin, L, Singh, B, et al. (2020) Neurological associations of COVID-19. Lancet Neurol 19, 767783.CrossRefGoogle ScholarPubMed
Paterson, RW, Brown, RL, Benjamin, L, et al. (2020) The emerging spectrum of COVID-19 neurology: clinical, radiological and laboratory findings. Brain 143, 31043120.CrossRefGoogle ScholarPubMed
Andrews, RC (1992) An update of the zinc deficiency theory of schizophrenia. Identification of the sex determining system as the site of action of reproductive zinc deficiency. Med Hypotheses 38, 284291.CrossRefGoogle ScholarPubMed
Szewczyk, B, Kubera, M & Nowak, G (2011) The role of zinc in neurodegenerative inflammatory pathways in depression. Prog Neuro-Psychopharmacol Biol Psychiatr 35, 693701.CrossRefGoogle ScholarPubMed
Brewer, GJ, Kanzer, SH, Zimmerman, EA, et al. (2010) Subclinical zinc deficiency in Alzheimer’s disease and Parkinson’s disease. Am J Alzheimers Dis Other Demen 25, 572575.CrossRefGoogle ScholarPubMed
Roy, A, Evers, SE, Avison, WR, et al. (2010) Higher zinc intake buffers the impact of stress on depressive symptoms in pregnancy. Nutr Res 30, 695704.CrossRefGoogle ScholarPubMed
Munshi, A, Babu, S, Kaul, S, et al. (2010) Depletion of serum zinc in ischemic stroke patients. Methods Find Exp Clin Pharmacol 32, 433436.10.1358/mf.2010.32.6.1487084CrossRefGoogle ScholarPubMed
Bhatt, A, Farooq, MU, Enduri, S, et al. (2011) Clinical significance of serum zinc levels in cerebral ischemia. Stroke Res Treat 2010, 245715.Google ScholarPubMed
Pochwat, B, Domin, H, Rafało-Ulińska, A, et al. (2020) Ketamine and Ro 25–6981 reverse behavioral abnormalities in rats subjected to dietary zinc restriction. Int J Mol Sci 21, 13.CrossRefGoogle ScholarPubMed
Seth, R, Corniola, RS, Gower-Winter, SD, et al. (2015) Zinc deficiency induces apoptosis via mitochondrial p53- and caspase-dependent pathways in human neuronal precursor cells. J Trace Elem Med Biol 30, 5965.CrossRefGoogle ScholarPubMed
Cole, TB, Robbins, CA, Wenzel, HJ, et al. (2000) Seizures and neuronal damage in mice lacking vesicular zinc. Epilepsy Res 39, 153169.10.1016/S0920-1211(99)00121-7CrossRefGoogle ScholarPubMed
Blasco-Ibáñez, J-M, Poza-Aznar, J, Crespo, C, et al. (2004) Chelation of synaptic zinc induces overexcitation in the hilar mossy cells of the rat hippocampus. Neurosci Lett 355, 101104.CrossRefGoogle ScholarPubMed
Weiss, JH, Sensi, SL & Koh, JY (2000) Zn2+: a novel ionic mediator of neural injury in brain disease. Trends Pharmacol Sci 21, 395401.CrossRefGoogle Scholar
Horning, MS & Trombley, PQ (2001) Zinc and copper influence excitability of rat olfactory bulb neurons by multiple mechanisms. J Neurophysiol 86, 16521660.CrossRefGoogle ScholarPubMed
Sekler, I, Moran, A, Hershfinkel, M, et al. (2002) Distribution of the zinc transporter ZnT-1 in comparison with chelatable zinc in the mouse brain. J Comp Neurol 447, 201209.CrossRefGoogle ScholarPubMed
Choi, BY, Hong, DK, Jeong, JH, et al. (2020) Zinc transporter 3 modulates cell proliferation and neuronal differentiation in the adult hippocampus. Stem Cells 38, 9941006.CrossRefGoogle ScholarPubMed
Chowanadisai, W, Kelleher, SL & Lönnerdal, B (2005) Zinc deficiency is associated with increased brain zinc import and LIV-1 expression and decreased ZnT-1 expression in neonatal rats. J Nutr 135, 10021007.CrossRefGoogle ScholarPubMed
Stoltenberg, M, Bush, AI, Bach, G, et al. (2007) Amyloid plaques arise from zinc-enriched cortical layers in APP/PS1 transgenic mice and are paradoxically enlarged with dietary zinc deficiency. Neuroscience 150, 357369.CrossRefGoogle ScholarPubMed
Datki, Z, Galik-Olah, Z, Janosi-Mozes, E, et al. (2020) Alzheimer risk factors age and female sex induce cortical Aβ aggregation by raising extracellular zinc. Mol Psychiatry 25, 27282741.CrossRefGoogle ScholarPubMed
Kumar, V (2016) Zinc deficiency and its effect on the brain: an update. Int J Mol Genet Gene Ther 1, 1.Google Scholar
Gilgun-Sherki, Y, Melamed, E & Offen, D (2004) The role of oxidative stress in the pathogenesis of multiple sclerosis: the need for effective antioxidant therapy. J Neurol 251, 261268.Google ScholarPubMed
Beltran-Corbellini, A, Chico-Garcia, JL, Martinez-Poles, J, et al. (2020) Acute-onset smell and taste disorders in the context of Covid-19: a pilot multicenter PCR-based case-control study. Eur J Neurol 27, e33.Google Scholar
Lechien, JR, Hopkins, C & Saussez, S (2020) Sniffing out the evidence; It’s now time for public health bodies recognize the link between COVID-19 and smell and taste disturbance. Rhinology 58, 402403.Google ScholarPubMed
Moein, ST, Hashemian, SMR, Mansourafshar, B, et al. (2020) Smell dysfunction: a biomarker for COVID-19. Int Forum Allergy Rhinol 10, 944995.CrossRefGoogle ScholarPubMed
Russell, B, Moss, C, Rigg, A, et al. (2020) Anosmia and ageusia are emerging as symptoms in patients with COVID-19: what does the current evidence say? Ecancermedicalscience 14, ed98.CrossRefGoogle ScholarPubMed
Russell, RM, Cox, ME & Solomons, N (1983) Zinc and the special senses. Ann Intern Med 99, 227239.CrossRefGoogle ScholarPubMed
Lyckholm, L, Heddinger, SP, Parker, G, et al. (2012) A randomized, placebo controlled trial of oral zinc for chemotherapy-related taste and smell disorders. J Pain Palliat Care Pharmacother 26, 111114.CrossRefGoogle ScholarPubMed
Calder, PC, Carr, AC, Gombart, AF, et al. (2020) Optimal nutritional status for a well-functioning immune system is an important factor to protect against viral infections. Nutrients 12, 4.CrossRefGoogle ScholarPubMed
Handu, D, Moloney, L, Rozga, M, et al. (2020) Malnutrition care during the COVID-19 pandemic: considerations for registered dietitian nutritionists. J Acad Nutr Diet (Epublication ahead of print version 14 May 2020).Google ScholarPubMed
Leij-Halfwerk, S, Verwijs, MH, van Houdt, S, et al. (2019) Prevalence of protein-energy malnutrition risk in European older adults in community, residential and hospital settings, according to 22 malnutrition screening tools validated for use in adults ≥ 65 years: a systematic review and meta-analysis. Maturitas 126, 8089.CrossRefGoogle Scholar
ctri.nic.in/ (2020) To Determine the Efficacy of An Ayurvedic Preparation Raj Nirwan Bati (RNB) on symptomatic COVID-19 Patients: A Double-Blind Randomized Controlled Trial: efficacy of An Ayurvedic Preparation Raj Nirwan Bati (RNB) on symptomatic COVID-19 Patient. http://ctri.nic.in/Clinicaltrials/showallp.php?mid1=44707&EncHid=&userName=2020/06/025998 (accessed July 2020).Google Scholar
Jothimani, D, Kailasam, E, Danielraj, S, et al. (2020) COVID-19: poor outcomes in patients with zinc deficiency. Int J Infect Dis 100, 343349.CrossRefGoogle ScholarPubMed
Anuk, AT, Polat, N, Akdas, S, et al. (2020) The relation between trace element status (zinc, copper, magnesium) and clinical outcomes in COVID-19 infection during pregnancy. Biol Trace Elem Res 110.Google ScholarPubMed
Yasui, Y, Yasui, H, Suzuki, K, et al. (2020) Analysis of the predictive factors for a critical illness of COVID-19 during treatment – relationship between serum zinc level and critical illness of COVID-19. Int J Infect Dis 100, 230236.CrossRefGoogle ScholarPubMed
ClinicalTrials.gov (2020) Evaluation of the Relationship Between Zinc Vitamin D and b12 Levels in the Covid-19 Positive Pregnant Women. https://ClinicalTrials.gov/show/NCT04407572 (accessed May 2020).Google Scholar
Finzi, E (2020) Treatment of SARS-CoV-2 with high dose oral zinc salts: a report on four patients. Int J Infect Dis 99, 307309.CrossRefGoogle ScholarPubMed
Yao, JS, Paguio, JA, Dee, EC, et al. (2020) The minimal effect of zinc on the survival of hospitalized patients with Covid-19: an observational study. Chest 159, 108111.CrossRefGoogle ScholarPubMed
Frontera, JA, Rahimian, JO, Yaghi, S, et al. (2020) Treatment with Zinc is associated with reduced in-hospital mortality among COVID-19 patients: a multi-center cohort study. Res Sq (Epublication ahead of print version 26 October 2020).Google ScholarPubMed
Kamran, SM, Mirza, Z-e-H, Naseem, A, et al. (2020) Clearing the fog: is Hydroxychloroquine effective in reducing Corona virus disease-2019 progression: a randomized controlled trial. medRxiv (Epublication ahead of print version 11 October 2020).Google Scholar
Abd-Elsalam, S, Soliman, S, Esmail, ES, et al. (2020) Do zinc supplements enhance the clinical efficacy of hydroxychloroquine?: a randomized, multicenter trial. Biol Trace Elem Res, 15 (Epublication ahead of print version 27 Nov 2020).Google ScholarPubMed
Derwand, R & Scholz, M (2020) Does zinc supplementation enhance the clinical efficacy of chloroquine/hydroxychloroquine to win todays battle against COVID-19? Med Hypotheses 142, 109815.CrossRefGoogle Scholar
Carlucci, PM, Ahuja, T, Petrilli, C, et al. (2020) Zinc sulfate in combination with a zinc ionophore may improve outcomes in hospitalized COVID-19 patients. J Med Microbiol 69, 12281234.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1 Summary of complications that can be expected in patients with pre-existing zinc deficiency, when challenged by Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2). A patient with no co-morbidities and a balanced zinc homoeostasis will most likely develop no or mild symptoms or complications if infected with SARS-CoV-2 because immune cell numbers and functions are balanced, as are the other parameters listed in the Figure. However, zinc deficiency alone will result in the alterations indicated in the Figure. Preconditions resulting from zinc deficiency may result in the development of severe symptoms, critical illness and even death if the patient becomes infected with SARS-CoV-2. ARDS, acute respiratory distress syndrome; CNS, central nervous system; IFN, interferon; MMP, matrix metalloproteinase; TH, T helper cell; Treg, regulatory T cell; ZA, zinc adequate; ZD: zinc deficient.

Figure 1

Fig. 2 Alterations in haematopoiesis are reported during zinc deficiency as well as in Corona Virus Disease 19 (COVID-19). During zinc deficiency, indicated by the red arrow, differentiation of myeloid cells, including polymorphonuclear neutrophils (PMN) and monocytes (Mo), is prioritised over development of adaptive immune cells, this especially impacts T cells (T). Amongst others, the prioritisation of myeloid cells may be explained by changes in growth factor expression: granulocyte-macrophage colony-stimulating factor (GM-CSF) and granulocyte-colony-stimulating factor (G-CSF) were described to be highly expressed, while levels of IL-2 are decreased during zinc deficiency. Furthermore, the T helper cell (TH)1:TH2 ratio is imbalanced during zinc deficiency, Th17 cell numbers are increased, while regulatory T cell (Treg) numbers were described as decreased as well as their functions. Most of those haematopoietic disturbances found during zinc deficiency are generally described for COVID-19 patients, as detailed in the text. B, B cell; BCP, B-cell progenitor; E, erythrocyte; EPO, erythropoietin; GM, granulocyte-macrophage progenitor; HSC, hematopoietic stem cell; MEP, megakaryocyte–erythroid progenitor; NK, natural killer cell; Pl, platelets; SCF, stem cell factor; TC: cytotoxic T cell; TNK, T and NK cell progenitor; TPO, Thrombopoietin.

Figure 2

Fig. 3 Pulmonary effects observed in Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) infected patients with pre-existing zinc deficiency as compared with patients with a balanced zinc homoeostasis. Pre-existing zinc deficiency (left) was suggested to increase the number, recruitment and inflammatory potential of especially PMN to the insides of the bronchi. Lymphocyte numbers are generally decreased, most prominently affecting T helper cell (TH) cells. The zinc deficiency-related decrease in tight junction expression and the increase in endothelial cell apoptosis have several consequences. Thus, infiltration of the lung by host cells, as well as the leakage of pathogens such as SARS-CoV-2 and secondary pathogens such as Streptococcus pneumoniae into the vascular system, is frequently observed during zinc deficiency. Detailed explanations can be found in the text. For comparison, the characteristics of zinc-adequate individual are indicated on the right. Ab, antibody; B, B cell; E, erythrocyte; G-CSF, granulocyte colony-stimulating factor; GC, glucocorticoid; GM-CSF, granulocyte-macrophage CSF; MMP, matrix metalloproteinase; Mo, monocyte; Mϕ, macrophage; NET, neutrophil extracellular trap; NK, natural killer cell; Pl, platelet; PMN, polymorphonuclear neutrophil; ROS, reactive oxygen species; Tc, cytotoxic T cell; TJ, tight junction; ZA, zinc adequate; ZD, zinc deficient.

Figure 3

Fig. 4 Effects of zinc deficiency on stress-induced changes in redox metabolism. Green arrows indicate zinc-dependent cellular functions. Red arrows illustrate the effects of zinc deficiency. A detailed description of the mechanisms underlying disturbed redox metabolism during zinc deficiency can be found in the text. AP-1, Activator protein 1; Bcl-2, B-cell lymphoma 2; CAT, catalase; COX, Cyclo-oxygenase; CRP, C-reactive protein; ER, endoplasmic reticulum; GPx, glutathione peroxidase; ICAM, intercellular adhesion molecule-1; iNOS, inducible nitric oxide synthase; MT, metallothionein; MTF, metal-responsive transcription factor-1; Ox, oxidated; MCP, monocyte chemoattractant protein; NIK, NFκB-Inducing Kinase; ROS, reactive oxygen species; SOD, superoxide dismutase; VCAM, vascular cell adhesion molecule.

Supplementary material: File

Wessels et al. Supplementary Material

Supplementary Table 1

Download Wessels et al. Supplementary Material(File)
File 81.9 KB