Logo-hpp
2022: Two-year Impact Factor: 4.4
Scopus Journal Metrics
CiteScore (2022): 5.3
SNIP(2022):1.389
SJR(2022): 0.78
Platinum
Open Access

Health Promotion Perspectives. 11(2):119-136. doi: 10.34172/hpp.2021.17

Review

Nutritional approach for increasing public health during pandemic of COVID-19: A comprehensive review of antiviral nutrients and nutraceuticals

Vahideh Ebrahimzadeh-Attari 1ORCID logo, Ghodratollah Panahi 2, James R. Hebert 3, 4, Alireza Ostadrahimi 5, Maryam Saghafi-Asl 5, *ORCID logo, Neda Lotfi-Yaghin 6, *ORCID logo, Behzad Baradaran 7
1Department of Nutrition and Food Sciences, Maragheh University of Medical Sciences, Margheh, Iran
2Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
3Department of Epidemiology & Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
4Cancer Prevention and Control Program, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
5Nutrition Research Center, Department of Clinical Nutrition, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
6Student Research Committee, Department of Clinical Nutrition, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
7Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
*Corresponding Authors: Maryam Saghafi- Asl, Email: saghafiaslm@gmail.com, Neda Lotfi-Yaghin, Email: lotfiyagin@gmail.com

Abstract

Background: The novel coronavirus (COVID-19) is considered as the most life-threatening pandemic disease during the last decade. The individual nutritional status, though usually ignored in the management of COVID-19, plays a critical role in the immune function and pathogenesis of infection. Accordingly, the present review article aimed to report the effects of nutrients and nutraceuticals on respiratory viral infections including COVID-19, with a focus on their mechanisms of action.

Methods: Studies were identified via systematic searches of the databases including PubMed/ MEDLINE, ScienceDirect, Scopus, and Google Scholar from 2000 until April 2020, using keywords. All relevant clinical and experimental studies published in English were included.

Results: Protein-energy malnutrition (PEM) is common in severe respiratory infections and should be considered in the management of COVID-19 patients. On the other hand, obesity can be accompanied by decreasing the host immunity. Therefore, increasing physical activity at home and a slight caloric restriction with adequate intake of micronutrients and nutraceuticals are simple aids to boost host immunity and decrease the clinical manifestations of COVID-19.

Conclusion: The most important nutrients which can be considered for COVID-19 management are vitamin D, vitamin C, vitamin A, folate, zinc, and probiotics. Their adequacy should be provided through dietary intake or appropriate supplementation. Moreover, adequate intake of some other dietary agents including vitamin E, magnesium, selenium, alpha linolenic acid and phytochemicals are required to maintain the host immunity.

Keywords: COVID-19, Coronavirus, Immunity, Energy intake, Proteins, Fatty acids, Omega-3 Vitamins, Vitamins, Minerals, Electrolytes

Copyright

© 2021 The Author(s).
This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.


Introduction

The global outbreak of coronavirus disease 2019 (COVID-19) has reached pandemic proportions, heightening public health concern around the world. Coronaviruses (CoVs), the single-stranded RNA viruses, have large, positive and enveloped senses that can affect both humans and animals. 1

COVID-19 is the third serious coronavirus outbreak in the world following severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). According to the latest statistics of World Health Organization (WHO), there are more than 99 million confirmed COVID-19 cases and more than 2 000 000 deaths due to COVID-19 around the world. 2

Clinical manifestations of COVID-19 range from mild symptoms to critical illness and death. 3,4 Frequently reported symptoms of infected patients include fever (77%–98%), cough (46%–82%), myalgia or fatigue (11%–52%), and difficulty of breath (3%-31%) at the onset of illness, which can progress to severe lung injury and respiratory distress syndrome. 3,5-7

Several host-related factors can affect the pathogenicity of COVID-19 including age, genetics, immunity, psychosocial status and especially existing comorbidities. However, another important variable is nutritional status, usually ignored in the epidemiological studies. 8 It was well-acknowledged that malnutrition; e.g., specific dietary deficiencies or severe weight imbalance (including both underweight and overweight/obesity) may affect the consequences of viral infections. 9-16 Actually, the idea that malnutrition makes a subject prone to viral infection first emerged in the West about 200 years ago. 17,18 Researchers indicate that host nutritional status not only affects the immune function of the body, but also may have some direct effects on the viral genome and its virulence. 18 Therefore, assessment of the host nutritional status before and during an infectious disease is of important value that might influence the recovery rate.

Regarding the global pandemic of COVID‐19, preventive policies such as applying public health principles and nutritional support are crucial at this time. Therefore, the present review article aimed to outline nutritional support for the prevention and treatment of COVID-19, focusing on the role of energy, macronutrients, and antiviral micronutrients and nutraceuticals.


Materials and Methods

Searches were performed in PubMed/MEDLINE, ScienceDirect, Scopus, and Google Scholar databases from 2000 until July 2020 using the following keywords based on MeSH terms. Generally, a topic-centric search was conducted to write each section. All relevant clinical and experimental studies published in English were included. The search terms included “immunity” OR “ viral infection” OR “ COVID-19” AND “nutrition” OR “antioxidant”; “vitamin D”; “vitamin A”; “vitamin C”; “folic acid”; “zinc”; “omega 3”; “protein”; “energy”; “probiotic”; “prebiotic”; “phytochemical”. These search items were used for the relevant sections of the review. Data were extracted from the included manuscripts by one author (MSA). Two other authors checked the accuracy of the data extracted (NL, VEA).

Energy

Protein-energy malnutrition (PEM), which is associated with low body weight and loss of both lean body mass and adipose tissue is characterized as “the most common cause of immunosuppression worldwide”. 9 It was reported that if PEM is accompanied by wasting, which is often the case, it can decrease the function of natural killer (NK) cells in both humans and mice 14 and increase the mortality rate in influenza-infected mice. 19

Generally, bacterial and viral infections are usually associated with increasing metabolic rate and energy requirements along with anorexia which exacerbates this energy demand. 9,20 During the COVID-19 pandemic, it has been reported that most of the infection-related mortality occurs in the elderly patients mainly due to malnutrition and other comorbidities. 21 Although aging is associated with a slight decrease in total lymphocytes number and some changes in T-cell response, 22-24 this higher mortality rate due to COVID-19 may be justifiable by the presence of PEM. 22

In this regard, energy requirement of an infected person with COVID-19 increases slightly, like any other viral infection especially during the acute phase of the infection. 9,25-28 It is ideal to measure the energy expenditure using the doubly labeled water as a gold standard method. 29 However, this is not practical in all settings and it is generally recommended that energy requirement be calculated carefully and individually, based upon a slight increase during the fever and acute respiratory distress as there is a hyper-metabolic state in both conditions. 27,28

Current energy recommendations during lower respiratory infections and acute respiratory distress are 30 to 35 kcal/kg ideal body weight (IBW) or resting energy expenditure (REE) ×1.2-1.4 (kcal). Fever also increases REE by approximately13% for each degree above 37°C. 27,28 It should be mentioned that regular nutritional assessment of hospitalized patients is very important to prevent the consequence of over-feeding (e.g., CO2 overproduction and fatty liver) or under-feeding (e.g., increasing morbidity and mortality rate and pulmonary cachexia). 27,28

If the measurement of VCO2 is not possible directly, body weight is used for the estimation of energy requirements, as below: 25–30 kcal/kg/d for non-obese and 21 kcal/kg/d for overweight, 11–14 kcal/kg/d for obese critically ill patients. 30

The adequacy of food intake should be thoroughly assessed in hospitalized patients with COVID-19 pneumonia. 27,28 Parenteral nutrition (PN) should start if enteral feeding is not possible or adequate. On the other hand, the worldwide outbreak of COVID-19 can be unintentionally accompanied by increasing anxiety, overeating and inactivity in the societies which cause people at risk of obesity. It was reported that obese people are at higher risk of developing either respiratory tract or urinary tract infections, compared to the normal weight people which highlights the important of physical fitness. 15 Therefore, as a prevention strategy, it is recommended that people avoid the excessive energy intake and ensure sufficient nutrients supply through healthy food preferences. Overall, avoiding under- nutrition or over- nutrition seems to be essential in case of COVID-19 prevention or treatment.

Macronutrients

It was shown that the amount and kind of macronutrients can change the number and function of immune cells in response to bacterial or viral agents. 31 The other mechanism for their modulation of immune function may be through the changes in the gut microbiota. 32 The roles of each macronutrient in immunity are presented in detail, as below:

Protein

Regarding the hyper-metabolic feature of severe respiratory tract infections like COVID-19, restricting the protein intake can lead to malnutrition which adversely affects lung structure, elasticity, and function. 27,33 The amount of protein intake can also directly impact the immune system. Based on the results of some experimental studies, feeding a low-protein diet, followed by infection with influenza virus was accompanied by decreased number and function of CD8+ T cells as well as NK cells, higher viral titers, and finally, increased mortality of animals. 34-36 It should be noticed that older people are at higher risk of protein deficiency 37-41 which may make them prone to longer hospitalization due to the infection. 41 Therefore, appropriate protein intake of 1.2 to 1.5 g/kg IBW (15%-20% of calories) with at least 50 percent from high biological value (HBV) proteins is recommended for adults and older people to maintain their respiratory muscle strength and support immune function during infection with COVID-19. 26,27 However, diet should be individually planned, considering other comorbidities of patient.

According to American Society for Parenteral and Enteral Nutrition (ASPEN), for critically ill patients with COVID-19 who need enteral nutrition (EN), the recommended protein intake is 1.2–2.0 g/kg/d for non-obese patients (based on actual body weight) and 2-2.5 g/kg/d for obese patients (based on IBW). 42

Carbohydrates

According to limited animal studies, 32,43 significant restriction of carbohydrates and sugar intake can exacerbate viral outcomes. This happens because the immune cells like CD4+ and CD8+ T cells, involved in the pathogen clearance, supply most of their energy from glucose and anaerobic glycolysis. 44 On the other hand, a high-carbohydrate diet increases CO2 production and respiratory quotient. Therefore, a balanced ratio of carbohydrates intake (40% to 50% of calories), mostly as the complex carbohydrates along with decreasing simple sugar is crucial in medical nutrition therapy of COVID-19 pneumonia. In addition, adequate intake of functional fibers (prebiotics such as beta-glucan and fructo-oligosaccharides) from dietary sources (e.g., oat, wheat, banana, onions, garlic, and tomato) has additional health benefits on the gut microbiota and immune defense system. 45

For those with severe COVID-19 symptoms, supportive nutrition may be required. EN is preferred over PN and is usually well-tolerated by the patients. The amount of carbohydrate in enteral feeding formula is dependent upon patient’s state (e.g., glycemic control, ventilator dependency) and usually consists 30%-50% of non-protein calories. 46 However, glucose should not exceed 5 mg/kg/min. 47

Fats

New coronavirus can cause severe lung injury and acute respiratory distress syndrome (ARDS). 48 During this acute-phase infection, increasing dietary fat intake helps to decrease CO 2 production as well as ventilator dependency. According to the dietary recommendations for ARDS and pneumonia, an enteral feeding with 30% to 45% of calories from fat maybe helpful. 27,28 However, the type of fatty acid is very important in the inflammatory and immunomodulatory responses of host to a microbial agent. 49,50

Adequate intake of omega-3 fatty acids may protect host against viral and bacterial infections. 51 However, there are some controversial data about their efficacy in different bacterial and viral infections. 52-55 A recent review article on the current experimental studies and clinical trials reported that intake of long-chain omega-3 fatty acids as docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) was effective in some extracellular infectious agents including Escherichia coli and Streptococcus pneumonia while had adverse effects on the intracellular infectious agents like Mycobacterium tuberculosis, Influenza virus and Herpes simplex virus. It was due to the modulation of immune responses including anti-viral CD8+ T cells and their downstream inflammatory cascades after viral infection. 51,55,56 Therefore, the efficacy of omega-3 long-chain omega-3 polyunsaturated fatty acids (PUFAs) on defense against viral infections is still unclear and needs further well-designed randomized clinical trials.

The other key element in medical nutritional management of COVID-19 infection with severe lung injury and ARDS symptoms is surfactant replacement therapy. 57,58 Dipalmitoyl-phosphatidylcholine (DPPC) is the main component of pulmonary surfactants which consists of two palmitic acids, attached to a phosphatidylcholine (lecithin) head-group. 58 Therefore, in addition to the exogenous surfactant replacement therapy, enteral feeding with dietary sources of lecithin, including egg yolk, meat, milk, and canola oil can be helpful in ARDS. 59

The amount of fat in EN formula depends on patient’s status (e.g., serum triglyceride, ventilator dependency) and usually comprises 50%-70% of non-protein calories. 47 It was reported that long-term deficiency of essential fatty acids (EFAs) including linoleic acid and ɑ-linolenic acid (ALA) can cause the reduction of DPPC level in lung tissue. 60 However, based on two recent systematic reviews, there are some controversies toward the benefits of long-chain omega 3 fatty acids supplementation on the length of mechanical ventilation and mortality rate in adult patients with ARDS. 61,62 Overall, adequate intake of EFAs and appropriate proportion of PUFAs and monounsaturated fatty acids (MUFAs) intake within the range of 30% to 45% of calories from fats can be effective in patients with COVID-19 pneumonia and ARDS symptoms. 63

Micronutrients

Vitamin D

Vitamin D (VitD), also termed cholecalciferol, has a lot of functions in the body including the immunomodulatory effect. 64 The deficiency of the vitamin is more prevalent around the world. 65 Results of some review articles show that low circulating levels of VitD are related to increased risk of respiratory viral infections. 65-67 However, there are some other heterogeneous findings on the protective effects of VitD supplementation for respiratory tract infections like influenza virus. 65,68,69

It is established that VitD (cholecalciferol) has an important role in the transcription of different genes through binding to the nuclear VitD receptors (VDRs). 70 VDRs are expressed by many immune cells, following an immune signaling. Their bindings to VitD result in the modulation of both innate and adaptive immune responses, B-lymphocytes, monocytes, macrophages, dendritic cells (DCs), and T-cells. This immunomodulatory effect on T-cells causes the suppression of the pro-inflammatory Th1 and Th17 cells as well as enhancement of regulatory T cells.

Increased activation, production, and secretion of pro-inflammatory cytokines and chemokines like nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) are the main feature of most respiratory viral infections. 71 Accumulating evidence proposes that a subgroup of patients with severe COVID-19 might have a cytokine storm syndrome. 72 It was reported that one of the main immunomodulatory effects of VitD against respiratory viruses is increasing the NF-κB inhibitor alpha (IκBα) expression. 73 Moreover, the active form of VitD (1, 25(OH)2 D3) can decrease the expression of some cytokines including tumor necrosis factor alpha, interleukin 6 (IL-6), and interferon beta (IFN-β) and increase the production of IL-37 and human beta defensin 2 which have key roles in warding off respiratory viruses. 70,74

VAD is linked to more inflammation and immune activation and low peripheral blood CD4+ T cells. 75 Low levels of VitD in calves have been correlated with increased bovine coronavirus infection. 76 Moreover, it was suggested that VAD can increase the host susceptibility to influenza viral infection. 77-79 It should be noticed that infection with influenza virus, the novel coronavirus, and SARS-CoV have been more abundant in winter, when the circulating level of VitD is the lowest. 77,80,81

Angiotensin (Ang) II-converting enzyme (ACE2) molecule was reported to be the main host cell receptor of COVID-19 and plays a critical role in the entry of virus into the cell to trigger the final infection. 82,83 And, it is closely associated with ARDS, 84 possibly due to an extremely large number of ACE2-expressing cells in the lung, especially in Asian males. 83 More interestingly, chronic VitD deficiency may induce lung fibrosis through the activation of the renin-angiotensin system. 85

Therefore, VitD supplementation seems to inhibit ACE2, consequently, helping to attenuate the lung infection. Overall, regarding the limited dietary values, it seems that VitD supplementation, aside from its wide-spread immunological functions, can be considered both as a preventive strategy and an anti-ACE2 therapeutic agent in the management of COVID-19 infection to maintain its serum level at normal range, but avoid the vitamin toxicity.

Vitamin C

Vitamin C (VitC), also known as ascorbic acid, is an antioxidant and free radical scavenger and serves as an important cofactor for many enzymatic reactions in the body. 86 Severe VitC deficiency leads to scurvy disease, marked by weakness of collagen proteins, poor wound healing, impaired immunity, and increased susceptibility to infections. 87 Studies have shown that VitC can affect the immune system by inhibition of oxidative stress generated by infections. 88,89 In addition, it has immune-modulating effects including phagocytic function, T-cells transformation and interferon production. 90 Clinical trials have shown that treatment by VitC reduced duration and severity of common cold episodes. 91,92 The latest research found that though treatment by VitC in patients with sepsis and ARDS did not improve the organ dysfunction, inflammation and vascular injury. However, mortality rate was significantly decreased. 93

Leukocytes such as neutrophils and monocytes accumulate VitC and have 50- to 100-fold higher concentrations than plasma, indicating its significant functions in the immunity. 94 As VitC attenuates both oxidants generation and NFкB activation in dendritic and neutrophils, 95 the deficiency of the vitamin can result in impaired immunity and higher susceptibility to infections. In turn, infections significantly impact on VitC levels due to enhanced inflammation and metabolic requirements. 96 Moreover, VitC deficiency may affect the ability of neutrophils to migrate to the sites of infection. 97,98

Patients with severe respiratory infections have lower plasma VitC concentrations and treatment by VitC restores the plasma VitC levels and ameliorates the severity of the respiratory symptoms. 99 Additionally, several studies on guinea pigs have demonstrated that supplemental VitC increases serum levels of C1q complement proteins 100-102 as well as antibodies. 103,104 However, no beneficial changes in leukocyte function or production have been reported by VitC treatment. 105-107

While some studies propose VitC as an immune enhancer, human studies published to date are contradictory. According to a Cochrane systematic review, only two of six trials were double-blind, placebo-controlled, and randomized controlled that could be applied to evaluate the effect of VitC supplementation on the prevention or treatment of pneumonia. 108 Overall, compared to placebo, VitC supplementation had a mild beneficial effect. However, firm conclusions cannot be elicited from the available data due to the highly variable characteristics of the study populations and methodological shortcomings. A phase II clinical trial is underway by Peng et al (https://www.clinicaltrials.gov/) from Wuhan to test the efficacy of VitC infusion for the treatment of severe acute respiratory infection associated with the novel coronavirus. They aimed to administer VitC (24 g/d IV) for 7 days to the afflicted patients.

It seems that VitC can help to avoid and cure respiratory and systemic infections via the enhancement of the immunity. At this time, the potential effect of VitC on severe viral respiratory tract infections especially in case of the recent COVID-19 epidemic would seem to be evaluated by further studies.

Vitamin A

Vitamin A (VitA) is one of the fat-soluble retinoids which deficiency is one of the most common nutritional issues in the world. 109 This vitamin has a broad range of immunological functions. VitA deficiency (VAD) is associated with recurrent infections. 110 Since VitA helps maintain the mucosal barriers of the innate immune system. Thus, VAD increases susceptibility to some types of infection, such as respiratory and gastrointestinal infections. 111-114 VAD also results in decreased or altered response of T and B cells to a variety of pathogens; dysregulated development of type IFN; and changes in the number and function of innate immune population, including monocytes, NK cells and DC subsets. 115,116 Serum retinol is inversely associated with serum concentration of interleukin-6, the main regulator in the induction of the acute-phase response. 117 Infections can, in turn, lead to VAD by reducing food intake, impairing vitamin absorption, increasing vitamin excretion, interfering with vitamin utilization, or increasing metabolic requirements of VitA. 111

Genetic studies indicated significantly impaired inflammatory milieu in the infected lungs of VAD calves, with alterations in Th1 and Th17 immune responses, and abnormal mucin production. In chickens fed a diet slightly deficient in VitA, the risk of infection with the infectious bronchitis virus, a kind of coronavirus, was more noticeable than in those fed with a VitA-adequate diet. 118 VitA has been shown to be effective in measles-associated pneumonia in children. 119 However, high-dose vitamin A supplements caused modest adverse effects in children recovering from pneumonia. 120 Therefore, they should not be therapeutically used in such patients unless there is a clinical evidence of VAD.

VitA supplementation was studied as a potential intervention to accelerate recovery, minimize severity, and avoid recurrent episodes of acute infections of the lower respiratory tract. 121,122 However, it is not beneficial in those with lower respiratory infections, such as pneumonia 123 and supplementation may actually aggravate the condition. 120,124,125 As VitA can inhibit viral replication through the up-regulation of the innate immunity, 126 it seems that VitA might be promising in the prevention and treatment of lung infections stemmed from COVID-19. However, due to potential adverse effects, VitA supplements should be administered for those with evidence of VAD. 127

Vitamin E

Vitamin E (VitE), as an antioxidant, protects membranes from oxidative damage by incorporating into cell membranes. 128 The α-tocopherol form of VitE protects against peroxidation of PUFAs which can potentially bring about abnormal immune responses. 129 Therefore, VitE is one of the most effective nutrients known to modulate immune function and reduce the risk of respiratory infections and asthma. Owing to its ability to bind free radicals, VitE plays an important function as an antioxidant. 130,131 Immune cells are enriched in VitE, likely to protect membranes against oxidative damage produced as a result of their high metabolic activity and defensive function. 132

Animal and human studies have demonstrated that VitE deficiency impairs both innate and adaptive immunity. 133 It leads to immune system dysfunction and there is evidence that VitE supplementation, beyond current dietary guidelines, enhances innate immune functions including NK cell activity and macrophage phagocytic capacity. 134 VitE may also exert its influence by modulation of inflammatory mediators like prostaglandin E2 and cytokines. 132

In a trial, daily supplementation of 200 mg of α-tocopherol for 235 days to healthy older adults increased the production of antibodies in response to tetanus and hepatitis B vaccines and improved T lymphocyte-mediated immunity. 135 In another interventional study, supplementation with 200 mg/d of α-tocopherol for three months to older adults significantly enhanced mitogen-induced lymphocyte proliferation and interleukin-2 (IL-2) production and improved NK cytotoxic activity, neutrophil chemotaxis, and phagocytic response, compared to baseline. 136 Daily supplementation of 617 nursing-home residents (≥65 years of age) with 200 IU of synthetic α-tocopherol for one year significantly lowered the risk of affliction with upper respiratory tract infections, particularly the common cold, but had no effect on lower respiratory tract (lung) infections. 135,137-140 Though studies have reported immune effects at dosage of 200–800 mg/d, 141 the optimum intake of VitE required to enhance immune system has not been demonstrated, likely due to prior VitE status and the presence or absence of other conditions. 142

Vitamin B12

Vitamin B12 (VitB12 or cobalamin) acts as a human immunity modulator; it stimulates the T-lymphocytes production involved in cellular immunity, restores the atypically increased ratio of CD4/CD8 and retains the lymphocyte subgroups count in the adequate range. 141,143 It supports both antibody-mediated and cellular immunity. 144 VitB12 deficiency reduces the lymphocytes and CD8+ cells number and the CD4 cells proportion, leading to an unusual high ratio of CD4+/CD8+, immune defense depression against bacteria and viruses and repressed NK cells. 143

Adequate vitamin dosage in deficient individuals leads to increased percent of CD3 and CD7 cells and their absolute number. Additionally, the function of lymphocytes and NK cells are restored, and IgA, IgG, and IgM concentrations are improved after treatment 144 Adults (aged >65 years) with normal immunity but with low serum VitB12 had an abnormal antibody response to pneumococcal polysaccharide vaccine. 145 Supplementation with VitB12 along with folate and VitE in the elderly people increased NK cell cytotoxic activity. 146 These few studies show the importance of a sufficient VitB12 status to maintain an adequate immune response, especially in the elderly who have a high percentage of deficient serum VitB12 level. 147

Vitamin B9 (folate)

Folate (VitB9 or folacin) along with vitamins B6 and B12 plays a vital role in the synthesis of nucleic acid and protein. Insufficient amount of folate leads to considerable alteration in the immune response. 144,148 In folate deficiency, the response of antibody to different antigens is reduced, since it results in lower proteins level involved in immune function activation and regulation, whereas a sufficient proportion with adequate pro-inflammatory cytokines supports an efficacious immune response. 149,150 It was demonstrated that the administration of folate supplements to the elderly improves overall immune function by alteration of the age-associated decrease in NK cell activity supporting a Th1 response; thus, providing protection against infections. 151 Interestingly, the results of a most recent study have proposed folate as a potential compound in the prevention or control of COVID-19 especially in the early stages of disease. In fact, this is the first time that has introduced folate as an effective inhibitor of furin enzyme activity. 152 The furin protein is associated with increasing the pathogenesis of most of bacterial and viral infections. It was proposed that the entrance of coronaviruses into cells is mediated through the spike proteins (S proteins) on their surface after cleavage into the S1 and S2 domains by furin enzyme. It should be mentioned that after this cleavage, the S1 subunit binds to the ACE2 receptor, the main host cell receptor of COVID-19, and then enters the lung cells. Therefore, supplementation with folate can be considered as a safe and promising treatment for patients with COVID-19. 152

Vitamin B6

Vitamin B6 (VitB6) or pyridoxine has a very important role in general cellular metabolism. 153,154 Cytokines and antibodies are made up of amino acids and need vitamin B6 as a coenzyme in their metabolism; therefore, the effects of VitB6 on immune function cannot be overlooked. 144,155 In previous studies, it was revealed that VitB6 deficiency impairs the maturation and growth of lymphocytes, and the production of antibodies and T-cells activity. The mitogenic response of lymphocytes is weakened by depletion of dietary VitB6 in elderly subjects and restored by VitB6 administration. Decreased antibody delayed-type hypersensitivity (DHT) response, NK cell activity, IL-1β, IL-2, IL-2 receptor and lymphocytes proliferation were observed due to deficiency in VitB6 levels. 156-158

Overall, further studies are warranted to evaluate the efficacy of VitB complex intake at dosages higher than the current recommended dietary allowance (RDA) for the prevention and/or reverse of immune system impairments.

Zinc

Zinc (Zn) acts as a powerful antioxidant and anti-inflammatory agent. 159 It has a vital role in both innate and adaptive immune cells, because some cellular functions such as DNA replication, RNA transcription, cell division, and cell activation are Zn-dependent. Zn is also crucial for normal development and function of neutrophils and natural-killer cells. 160 Zn deficiency leads to thymic atrophy, lymphopenia, impaired cellular and antibody-mediated immune responses and increases susceptibility to a variety of pathogens and infectious diseases. 161

Some aspects of immunity can be suppressed by even marginal Zn deficiency, which is more common than severe Zn deficiency, 124 particularly in the elderly due to their inadequate dietary intake. 162,163 In addition, serum Zn levels decline with age. 164,165 Several randomized controlled trials demonstrate that supplementation with low to moderate doses of Zn (ranging from 10 to 45 mg zinc/day) in healthy elderly individuals improves immune function, such as increased number of cytotoxic T lymphocytes and NK cells, reduced number of activated T helper cells, and lower incidence of infections. 166-168 Velthuis et al 169 showed that increasing the intracellular Zn2+ concentration with zinc-ionophores like pyrithione (PT) can efficiently impair the replication of some RNA viruses, including influenza virus. In their study, it was demonstrated that the combination of Zn2+ and PT at low concentrations can inhibit the replication of SARS-CoV in cell culture. 169 Another systematic review demonstrated that zinc supplementation was significantly associated with reducing rates of pneumonia and therefore, recommended supplementing zinc intake in deficient populations. 170

Although several studies favor the vital role of Zn in immunity enhancement, on the other side, there are few preliminary studies, 171,172 implying the possible adverse effects of Zn in cell cultures. For instance, Phillips et al reported that neurovirulent murine coronavirus JHM.SD uses cellular Zn metalloproteases for virus entry and cell-cell fusion. 171 They found that inhibition of matrix metalloproteinase and metalloprotease (ADAM)-family Zn metalloproteases markedly decreased both entry and cell-cell fusion. They concluded that Zn metalloproteases must be considered potential contributors to coronavirus fusion. In this regard, it is worth-mentioning that ACE which COVID-19 as well as SARS-CoV were shown to use as its main entry receptor, 66 belongs to the M2 Zn metalloproteinase family. 173 Moreover, in the study of Tan et al, it was proposed that binding of the 50-untranslated region of coronavirus RNA to Zn finger CCHC-type and RNA-binding motif 1 enhances both viral replication and transcription. 172 Therefore, in the light of current studies, it appears that adequate intake of Zn is essential for proper function of immune system; however, there might be some hazards in case of excessive Zn intake. Overall, precautions should be taken when it comes to supplementation of Zn for the prevention or treatment of infections.

Iron

Iron is an essential micronutrient for cellular energy metabolism, oxygen transport, and several enzymatic reactions. 174 The iron-containing proteins are needed for crucial cellular and organismal actions including mitochondrial respiration, oxygen transport, cell signaling, nucleic acid replication and repair, and host defense. 175 An adequate amount of iron is critical for multiple immune functions, including the differentiation and proliferation of T-cells and generation of reactive oxygen species (ROS) that kill infectious agents. However, iron is also required by most pathogens for replication and survival. During an acute inflammatory response, serum iron concentration depresses, while level of ferritin (the iron storage protein) increases, proposing that sequestration of free iron by host proteins prevents acquisition by pathogens 176 ; this is called nutritional immunity, which is an important host response to infections. 177

Iron ions serve as the catalytic portion of enzymes that mediate redox reactions in key cellular processes, like DNA replication and energy production. 178 Fe (III) prevents replication of DNA and RNA viruses. 179 During viral infection, ROS and superoxide hydrogen production may happen. Iron can be easily oxidized and reduced, making it necessary to be able to catalyze the exchange of hydrogen peroxides to free radicals. 180 Iron-deficient children and iron deficiency anemia has been reported as a risk factor for the development of recurrent acute respiratory tract infections. 181 Furthermore, lactoferrin as the iron-binding glycoprotein shows inhibitory actions against a different range of viruses in vitro. Indeed, it was observed that lactoferrin consumption might protect the host from virus infections by preventing the virus attachment to the cells, the virus replication in the cells, and improvement of systemic immune function. 182

Excessive iron can also be harmful, because of its ability to favor animal viral infections. The metal is required for host cell to synthesize virions which can weaken the cell function defense and raise oxidative stress. In humans and animal models, viral infections can lead to up-regulation of the iron-repressing defense system. Iron chelators are perfect candidates for use in co-infection and excess iron states because they have been effective in inhibition of HIV replication. 183 Overall, it seems essential to restrict iron intake in times of either potential or existing infections to deprive the pathogens of iron, but to have an adequate intake based on RDA to maintain an optimum immune response and avoid the possibility of excess amounts of iron which may induce iron toxicity. 184

Selenium

Selenium (Se) is an important trace element with anti-inflammatory and antioxidant effects. 185 Low serum Se level has been correlated with poor immune function and increased risk of mortality. Adequate Se intake has antiviral effects. 186 The patients diagnosed with influenza virus present a noticeable increase in protein, lipid, and DNA oxidation products in plasma and urine. 187-190 I n some types of RNA viruses, oxidative stress prompts fast mutation rates - frequently to virulence. 191 Se deficiency impairs the innate and acquired immunity unfavorably influencing both cell-mediated immunity and humoral immunity (i.e., antibody production). 192,193 Sequencing of virus genes isolated from Se-deficient and Se-adequate mice showed a strong effect of the Se status on virus mutation. 194 Se deficiency also seems to enhance the virulence or progression of some viral infections. 195,196 However, Se supplementation could ameliorate cell-mediated immunity in deficient individuals and enhance the immune response to viruses; on the other hand, Se supplementation may aggravate allergic asthma and weaken the immune response to parasites. 197,198 Meanwhile, dramatic evidence demonstrates that Se plays a role in the regulation of cytokines and eicosanoids production that adjust the immune response. 199 Overall, Se plays an important role in balancing the redox state, and helping to protect the host from oxidative stress induced by inflammatory reactions and anti-microbial effects of macrophages. Consequently, Se supplementation might be a helpful choice for the prevention or treatment of different types of viruses such as COVID-19 virus. However, Se status of the host is an important factor, when considering Se supplementation. 197

Water and electrolytes

It is usually recommended that higher fluid intake can prevent respiratory infections. Patient with COVID-19 symptoms or pneumonia can also benefit from higher intake of fluid unless contraindicated (such as edema or diarrhea). The recommended amount is usually 1 mL/kcal water or 2-3 L/d, being advised to drink between meals to prevent food reflux or aspirations. 27,28

It was reported that patients with lower respiratory tract infections especially children are at risk of developing hyponatremia due to possible inappropriate antidiuretic hormone secretion (ADH) or excessive administration of free water or lower sodium intake. 200-202 Moreover, as respiratory acidosis may occur following severe respiratory symptoms, serum electrolytes balance must be checked for hospitalized patients with COVID-19. The hypomagnesemia and hypophosphatemia should be avoided because magnesium and phosphorous both act as important intracellular buffers and contribute to the production of adenosine triphosphate (ATP). Their serum levels should be maintained at normal range, since high serum magnesium concentration (higher than 4-5 mmol/L) as well as severe hypophosphatemia (<1 mg/dL) can decrease respiratory function. 27,203-204

Phytochemicals

Plants provide us with a range of therapeutic metabolites that have the ability to prevent viral replication by controlling viral absorption, attaching to cell receptors, and inhibiting virus penetration into the host cell. 205 Flavonoids are examples of metabolites which exhibit antiviral activity. Specially, luteolin, apigenin, quercetin, daidzein, amentoflavone, epigallocatechin, epigallocatechin gallate, gallocatechin gallate, puerarin, and kaempferol were found to hinder the SARS-CoV 3CL pro-proteolytic activity. 206-209 Consequently, the antiviral effect is assumed to be specifically related to repress the SARS-CoV 3CLpro activity in some cases. Additionally, experimental study showed that isobavachalcone, quercetin 3-β-d-glucoside herbacetin, and helichrysetin were reported to block the MERS‐CoV 3CLpro enzymatic activity. Also, some flavonoid derivatives with carbohydrate or hydrophobic molecule in their core structures have been shown to yield a good inhibitory function. 210

Resveratrol as a plant compound with antiviral activity was reported to stimulate ERK1/2 signaling pathway and activate cell proliferation and improve SIR1 signaling, which are associated with DNA repair and cellular survival, following DNA damage. 211-214 Moreover, MERS-CoV infection can lead to the inflammatory cytokines production while, resveratrol might decrease the inflammation via preventing the NF-κB pathway. 215-216 In a study by Lin et al, it was observed that after MERS-CoV infection, the cleaved caspase 3 levels were reduced by resveratrol. 215 They believed that the mentioned changes might be due to the caspase 3 cleavage direct inhibitions by reservation of the cell survival as well as the upstream event inhibition, necessary for caspase 3 cleavages or the decrease in virus-induced apoptosis by resveratrol. 217

Overall, studies support the favorable effects of phytochemicals especially flavonoids and resveratrol in immunity enhancement following viral infections. Though further studies are warranted in case of antiviral impacts of phytochemicals, it appears that their adequate intake can contribute to boost the immune system, particularly in COVID-19.

Probiotics

Probiotics are “live strains of strictly selected microorganisms which, when administered in adequate amounts, confer a health benefit on the host”. 218 Common instances are the Lactobacilli and Bifidobacteria species. Ingested live probiotics can modulate immune functions through the interaction with several receptors on intestinal epithelial cells and other gut-associated immune cells, including M-cells and DCs. 219 Probiotics have positive effects on gastrointestinal disorders and allergic diseases. Also, the effectiveness of probiotics for diseases treatment such as type 2 diabetes 220-221 and obesity has been proven. 222

Probiotics have effects on both the innate and acquired immune systems and have the potential to reduce the infections severity in the upper respiratory as well as gastrointestinal tracts. 223-225 Studies have revealed that probiotics could prevent viral attachment through competitive inhibition, if they were able to bind to viral receptors at the cells surface. 226 In a study by Freitas et al, it was reported that a strain of Bacteroides thetaiotaomicron and the Lactobacillus casei strain DN114001 produce a compound that partly keep epithelial cells from the infection of rotavirus in vitro by modifying the cells apical glycosylation pattern. 227,228 It is also probable that probiotics indirectly affect viruses through changing the cells state, and triggering innate and/or adaptive immunity. 223,229 A study showed that Enterococcus faecium acts as beneficial antiviral agent by inhibiting transmissible gastroenteritis virus replication in swine testicle cells. The following overlapping mechanisms might lead to the detected fall in virus growth: inactivation or absorptive trapping of virus particles by the probiotic bacteria surface components, direct interfering with the attachment of virus and the stimulation of antiviral cytokines IL-6 and IL-8. 226

Interestingly, the intestinal barrier can be reinforced by probiotics through the rise in mucins, the tight junction proteins and the Paneth and Goblet cells. 230 The regeneration of mucosa is improved by mucin ability to inhibit the attachment of virus to epithelial cells and stop the virus replication. Probiotics also have the ability to modify the functions of epithelial cells, CD4+ CD8+ T lymphocytes DCs, NK cells, and induce secretory immunoglobulins synthesis, helping to deactivate a virus. 231,232 Although the scientific evidence is too weak to favor the use of probiotics to reduce respiratory infections and improve vaccination response, especially in the elderly, 226-233 probiotics are projected to be among the rational adjunctive choices for the treatment of several viral diseases.


Conclusion

A summary of energy and macronutrients requirements for the management of COVID-19 is shown in Box 1. According to the studies reviewed, it is recommended to provide a relatively high-energy diet for patients afflicted with COVID-19 pneumonia, due to their increased metabolic demands. On the other hand, for un-afflicted persons, quarantined at home, we recommend to avoid sedentary lifestyle and excessive energy intake which can lead to the obesity and decreased immunity. Overall, avoiding undernutrition or over-nutrition by ensuring sufficient nutrients supply through healthy food preferences seems to be essential in case of COVID-19 prevention or treatment.

Box 1. Summary of energy and macronutrients requirement for the management of COVID-19

  1. Notice regular nutrition assessment of patients considering anthropometry, laboratory, and clinical data to avoid malnutrition.

  2. Design an individualized diet with energy intake of approximately 30 to 35 kcal/kg IBW and 1.2-1.5 g protein/kg IBW to meet hyper-metabolic requirements of patients with COVID-19 pneumonia.

  3. Consider adequate amount of dietary fat (35%-40% of total calories) especially during ARDS and appropriate proportion of fatty acids (EFA, PUFA, and MUFA)

  4. Prevent or correct dehydration using enough fluid intake (2–3 L/d) between meals, unless contraindicated.

  5. Start PN when enteral nutrition has failed or during severe malabsorption.

  6. Provide small, frequent feedings to reduce food reflux and aspirations.

  7. Support lung function and prevent additional concomitant infections with higher intake of antioxidant and anti-microbial nutrients as discussed in detail through dietary sources or supplements, if needed (PN).

  8. Plan a healthy diet during enteral feeding, considering adequate nutraceuticals intake including prebiotics, probiotics, and phytochemicals

Appropriate protein intake of 1.2 to 1.5 g/kg IBW (equal to 15%-20% of calories) with at least 50 percent from HBV proteins for adults is recommended to avoid more respiratory complications and support immunity during infection with COVID-19. However, diet should be individually planned, considering other comorbidities of patients. A balanced ratio of carbohydrates (40% to 50% of calories) mostly as the complex carbohydrates and adequate intake of functional fibers is critical in medical nutrition therapy of COVID-19 pneumonia.

Adequate intake of EFAs and appropriate PUFAs and MUFAs intake within the range of 30% to 45% of calories from dietary fat can be effective in the management of COVID-19 pneumonia with ARDS symptoms by decreasing CO2 production and ventilator dependency. Although omega 3 fatty acids are regarded as anti-inflammatory food agents, the evidence is not suggestive for their administration after viral infection. This is because they can modulate anti-viral CD8+ T cells and their downstream inflammatory cascades.

Patient with COVID-19 symptoms or pneumonia can also benefit from higher intake of fluid, unless contraindicated. The recommended amount is usually 1 mL/kcal water or 2-3 L/d. As respiratory acidosis may occur following severe respiratory symptoms, serum electrolytes balance, especially magnesium, phosphorous and sodium must also be checked.

A summary of micronutrients and nutraceuticals are presented in Table 1. It seems that VitD supplementation can be helpful both as a preventive strategy and an anti-ACE2 therapeutic agent in COVID-19 management to maintain its serum level at normal range, but avoid the vitamin toxicity. VitC can also help avoid and cure respiratory and systemic infections by enhancing the immunity. However, further studies are needed to confirm the potential effect of VitC on severe viral respiratory tract infections especially in case of the recent COVID-19 epidemic. As VitA has a wide range of immunological functions and contributes to the first-line defense of the body, it appears that VitA might be promising in the prevention and treatment of lung infections associated with COVID-19. However, due to potential adverse effects, VitA supplements should be administered for those with evidence of VAD.

Table 1. Significant antiviral micronutrients or nutraceuticals
Nutrient/Nutraceutical RDAs for adults (≥19 years) Major sources b
Vitamin D 19-70 years: 600 IU/day
>71 years: 800 IU/day
Sunshine, salmon, fortified foods like milk
Vitamin A Men: 900 µg/d
Women: 700 µg/d
Liver, milk, egg, turkey meat, sweet potato, carrots, spinach
Vitamin E15 mg/dVegetable oils especially sunflower and canola oil, nuts
Vitamin C Men: 90 mg/d
Women: 75 mg/d
Sweet pepper, citrus fruit, kiwifruit, strawberry
Vitamin B6 19-50 years: 1.3 mg/d
≥51 years:
Men: 1.7 mg/d
Women: 1.5 mg/d
Salmon, turkey, chicken, potato, banana
Vitamin B9400 µg/dLentils, beans, spinach, asparagus, broccoli
Vitamin B122.4 µg/dLiver, clams, beef, oyster, mackerel, skim milk
Selenium55 µg/dBrazil nuts, halibut, tuna, oysters, chicken, egg, rice
Zinc Men: 11 mg/d
Women: 8 mg/d
Oyster, beef, pork, chicken, dairy product, baked bean
Iron Men: 8 mg/d
Women:
19–50 years: 18 mg/d
> 50 years: 8 mg/d
Clams, liver, beef, oyster, baked bean, fortified cereals
Omega 3 fatty acidsa ALA:
Men: 1.6 g/d
Women: 1.1 g/d
DHA and EPA: ND
ALA: Flaxseed, canola oil, soybean oil, walnut, pumpkin seeds
DHA & EPA: Cod liver oil, herring, salmon, sardines
ProbioticsNDFermented foods: yogurt, kefir, sauerkraut, pickles, tempeh
PrebioticsNDOat, wheat, banana, onions, garlic, tomato, asparagus, chicory root
Phytochemicals (flavonoids, resveratrol, quercetin, etc.)NDCoffee, black tea, green tea, most of fresh fruits and vegetables like apple, citrus fruits, berries, grapes, onion and also some herbal plants

Abbreviations: RDA, recommended dietary allowance; ALA, ɑ-Linolenic acid;

aBased on Adequate Intakes (AIs)

bSource: Kathleen Mahan L, Raymond JL, eds. Krause's Food & the Nutrition Care Process. St. Louis, Missouri: Elsevier; 2016

The optimum intake of VitE required for better immunity function has not been reported, likely due to prior VitE status and the person’s comorbidities; though studies have reported effective immune dosage of 200–800 mg/d. Given the effects of VitB complex, especially B6, B9, and B12 on the immunity, further studies are warranted to evaluate the efficacy of VitBs intakes at higher dosages than recommended for the prevention and/or reverse of immune system impairments.

In terms of trace elements, it appears that adequate intake of Zn is essential for proper function of the immune system; however, there might be some health hazards in case of excessive Zn intake. Therefore, precautions should be taken when it comes to supplementation of zinc for the prevention or treatment of infections. It is also essential to restrict iron intake in times of either potential or existing infections, but to have an adequate daily intake, based on the RDA, to maintain an optimum immunity and prevention of viral infections. Moreover, evidence show that adequate selenium intake or its supplementation within safe dose might be promising in the prevention and treatment of respiratory infections including COVID-19.

Based on current limited studies, it seems that adequate intake of alpha linolenic acid, phytochemicals and probiotics may help boost the immune system and prevent or even treat viral infections.

Finally, the present review tried to help the health practitioners to effectively manage patients with COVID-19 by taking nutritional considerations more into account. However, further well-designed clinical trials are needed to confirm the efficacy of nutritional recommendations and determine their effective dose during the outbreak of respiratory viral infections.


Acknowledgments

We wish to express our appreciation to the Research Vice-Chancellor of Tabriz and Maragheh Universities of Medical Sciences.


Funding

The present comprehensive review was performed without any grant and just as the authors’ interest and responsibility.


Competing interests

The authors declare no conflict of interest.


Ethical approval

This review did not need any formal approval from ethical committee.


Authors’ contributions

VEA, MSA and NLY contributed in designing the study, searching for resources and writing the manuscript. GP cooperated in writing the mechanism of action for nutrients. MSA, JRH, AO, and BB each contributed in the revision of the manuscript.


References

  1. Masters PS. The molecular biology of coronaviruses. Adv Virus Res 2006; 66:193-292. doi: 10.1016/S0065-3527(06)66005-3 [Crossref]
  2. World Health Organization. (WHO. Coronavirus disease (COVID-19) pandemic.) Accessed January 2021.
  3. Holshue ML, DeBolt C, Lindquist S, Lofy KH, Wiesman J, Bruce H. First case of 2019 novel coronavirus in the United States. N Engl J Med 2020; 382(10):929-936. doi: 10.1056/NEJMoa2001191 [Crossref]
  4. Chan JF-W, Yuan S, Kok K-H, To KK-W, Chu H, Yang J. A familial cluster of pneumonia associated with the 2019 novel coronavirus indicating person-to-person transmission: a study of a family cluster. The Lancet 2020; 395(10223):514-23. doi: 10.1016/S0140-6736(20)30154-9 [Crossref]
  5. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. The Lancet 2020; 395(10223):497-506. doi: 10.1016/S0140-6736(20)30183-5 [Crossref]
  6. Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus–infected pneumonia in Wuhan, China. JAMA 2020; 323(11):1061-1069. doi: 10.1001/jama.2020.1585 [Crossref]
  7. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. The Lancet 2020; 395(10223):507-13. doi: 10.1016/S0140-6736(20)30211-7 [Crossref]
  8. Field CJ, Johnson IR, Schley PD. Nutrients and their role in host resistance to infection. J Leukoc Biol 2002; 71(1):16-32.
  9. Ritz BW, Gardner EM. Malnutrition and energy restriction differentially affect viral immunity. J Nutr 2006; 136(5):1141-4. doi: 10.1093/jn/136.5.1141 [Crossref]
  10. Soares MP, Teixeira L, Moita LF. Disease tolerance and immunity in host protection against infection. Nat Rev Immunol 2017; 17(2):83-96. doi: 10.1038/nri.2016.136 [Crossref]
  11. Scrimshaw NS, SanGiovanni JP. Synergism of nutrition, infection, and immunity: an overview. Am J Clin Nutr 1997; 66(2):464S-77S. doi: 10.1093/ajcn/66.2.464S [Crossref]
  12. Gavazzi G, Herrmann F, Krause K-H. Aging and infectious diseases in the developing world. Clin Infect Dis 2004; 39(1):83-91. doi: 10.1086/421559 [Crossref]
  13. Redmond HP, Gallagher HJ, Shou J, Daly JM. Antigen presentation in protein-energy malnutrition. Cell Immunol 1995; 163(1):80-7. doi: 10.1006/cimm.1995.1101 [Crossref]
  14. Ingram KG, Croy BA, Woodward BD. Splenic natural killer cell activity in wasted, protein-energy malnourished weanling mice. Nutr Res 1995; 15(2):231-43. doi: 10.1016/0271-5317(95)92589-C [Crossref]
  15. Falagas ME, Kompoti M. Obesity and infection. Lancet Infect Dis 2006; 6(7):438-46. doi: 10.1016/S1473-3099(06)70523-0 [Crossref]
  16. Solomons NW. Malnutrition and infection: an update. Br J Nutr 2007; 98(S1):S5-S10. doi: 10.1017/S0007114507832879 [Crossref]
  17. Keusch GT. The history of nutrition: malnutrition, infection and immunity. J Nutr 2003; 133(1):336S-40S. doi: 10.1093/jn/133.1.336S [Crossref]
  18. Beck MA, Handy J, Levander OA. Host nutritional status: the neglected virulence factor. Trends Microbiol 2004; 12(9):417-23. doi: 10.1016/j.tim.2004.07.007 [Crossref]
  19. Woodward B, Hillyer L, Hunt K. T cells with a quiescent phenotype (CD45RA+) are overabundant in the blood and involuted lymphoid tissues in wasting protein and energy deficiencies. Immunology 1999; 96(2):246. doi: 10.1046/j.1365-2567.1999.00694.x [Crossref]
  20. Kanra GY, Ozen H, Kara A. Infection and anorexia. Turk J Pediatr 2006; 48(4):279-87.
  21. Wang W, Tang J, Wei F. Updated understanding of the outbreak of 2019 novel coronavirus (2019‐nCoV) in Wuhan, China. J Med Virol 2020; 92(4):441-7. doi: 10.1002/jmv.25689 [Crossref]
  22. Lesourd B. Nutrition: a major factor influencing immunity in the elderly. J Nutr Health Aging 2004; 8(1):28-37.
  23. Murasko D GE. Principles of geriatric medicine and gerontology. In: Hazzard WR BJ, Halter JB, Ouslander JG, Tinetti ME, ed. Principles of Geriatric Medicine and Gerontology. New York: McGraw-Hill2003. p. 35–51.
  24. Miller RA. Age-related changes in T cell surface markers: a longitudinal analysis in genetically heterogeneous mice. Mech Ageing Dev 1997; 96(1-3):181-96. doi: 10.1016/s0047-6374(97)01893-9 [Crossref]
  25. Powanda MC, Beisel WR. Metabolic effects of infection on protein and energy status. The J Nutr 2003; 133(1):322S-7S. doi: 10.1093/jn/133.1.322S [Crossref]
  26. Hill K, Vogiatzis I, Burtin C. The importance of components of pulmonary rehabilitation, other than exercise training, in COPD. Eur Respir Rev 2013; 22(129):405-13. doi: 10.1183/09059180.00002913 [Crossref]
  27. Khan SH, Karnik AM. Medical Nutrition Therapy for Pulmonary Disease. In: Kathleen L, Mahan JLR, editors. Krause’s Food & the Nutrition Care Process. St. Louis, Missouri :Elsevier;2016. P. 681-697.
  28. Escott-Stump S. ‘AIDS and immunology, burns, sepsis, and trauma’. In: Escott-Stump S. Nutrition and Diagnosis-Related Care. Philadelphia: Wolters Kluwer, Lippincott Williams & Wilkins. 2015. P. 819- 854.
  29. Berman ES, Swibas T, Kohrt WM, Catenacci VA, Creasy SA, Melanson EL. Maximizing precision and accuracy of the doubly labeled water method via optimal sampling protocol, calculation choices, and incorporation of 17 O measurements. Eur J Clin Nutr 2020; 74(3):454-464. doi: 10.1038/s41430-019-0492-z [Crossref]
  30. Port Port, AM AM, ApovianCM ApovianCM. Metabolic support of the obese intensive care unit patient: A current perspective. Curr Opin Clin Nutr Metab Care 2010; 13(2):184-91. doi: 10.1097/MCO.0b013e328335f1e6 [Crossref]
  31. Balmer ML, Hess C. Starving for survival—how catabolic metabolism fuels immune function. Curr Opin Immunol 2017; 46:8-13. doi: 10.1016/j.coi.2017.03.009 [Crossref]
  32. Varanasi SK, Donohoe D, Jaggi U, Rouse BT. Manipulating glucose metabolism during different stages of viral pathogenesis can have either detrimental or beneficial effects. J Immunol 2017; 199(5):1748-61. doi: 10.4049/jimmunol.1700472 [Crossref]
  33. Collins PF, Stratton RJ, Elia M. Nutritional support in chronic obstructive pulmonary disease: a systematic review and meta-analysis. Am J Clin Nutr 2012; 95(6):1385-95. doi: 10.3945/ajcn.111.023499 [Crossref]
  34. Chatraw JH, Wherry EJ, Ahmed R, Kapasi ZF. Diminished primary CD8 T cell response to viral infection during protein energy malnutrition in mice is due to changes in microenvironment and low numbers of viral-specific CD8 T cell precursors. J Nutr 2008; 138(4):806-12. doi: 10.1093/jn/138.4.806 [Crossref]
  35. Taylor AK, Cao W, Vora KP, Cruz JDL, Shieh W-J, Zaki SR. Protein energy malnutrition decreases immunity and increases susceptibility to influenza infection in mice. J Infect Dis 2013; 207(3):501-10. doi: 10.1093/infdis/jis527 [Crossref]
  36. Pena-Cruz V, Reiss C, McIntosh K. Sendai virus infection of mice with protein malnutrition. J Virol 1989; 63(8):3541-4. doi: 10.1128/JVI.63.8.3541-3544.1989 [Crossref]
  37. Paddon-Jones D, Leidy H. Dietary protein and muscle in older persons. Curr Opin Clin Nutr Metab Care 2014; 17(1):5-11. doi: 10.1097/MCO.0000000000000011 [Crossref]
  38. Volpi E, Campbell WW, Dwyer JT, Johnson MA, Jensen GL, Morley JE. Is the optimal level of protein intake for older adults greater than the recommended dietary allowance?. J Gerontol A Biol Sci Med Sci 2013; 68(6):677-81. doi: 10.1093/gerona/gls229 [Crossref]
  39. Bauer J, Biolo G, Cederholm T, Cesari M, Cruz-Jentoft AJ, Morley JE. Evidence-based recommendations for optimal dietary protein intake in older people: a position paper from the PROT-AGE Study Group. J Am Med Dir Assoc 2013; 14(8):542-59. doi: 10.1016/j.jamda.2013.05.021 [Crossref]
  40. Deutz NE, Bauer JM, Barazzoni R, Biolo G, Boirie Y, Bosy-Westphal A. Protein intake and exercise for optimal muscle function with aging: recommendations from the ESPEN Expert Group. Clin Nutr 2014; 33(6):929-36. doi: 10.1016/j.clnu.2014.04.007 [Crossref]
  41. Deer RR, Volpi E. Protein requirements in critically ill older adults. Nutrients 2018; 10(3):378. doi: 10.3390/nu10030378 [Crossref]
  42. Mechanick JI, Carbone S, Dickerson RN, Hernandez BJD, Hurt RT, Irving SY. ASPEN COVID-19 Task Force on Nutrition Research. Clinical Nutrition Research and the COVID-19 Pandemic: A Scoping Review of the ASPEN COVID-19 Task Force on Nutrition Research. JPEN J Parenter Enteral Nutr 2021; 45(1):13-31. doi: 10.1002/jpen.2036 [Crossref]
  43. Wang A, Huen SC, Luan HH, Yu S, Zhang C, Gallezot J-D. Opposing effects of fasting metabolism on tissue tolerance in bacterial and viral inflammation. Cell 2016; 166(6):1512-25. doi: 10.1016/j.cell.2016.07.026 [Crossref]
  44. O’Neill LA, Kishton RJ, Rathmell J. A guide to immunometabolism for immunologists. Nat Rev Immunol 2016; 16(9):553-65. doi: 10.1038/nri.2016.70 [Crossref]
  45. Shokryazdan P, Jahromi MF, Navidshad B, Liang JB. Effects of prebiotics on immune system and cytokine expression. Med Microbiol Immunol 2017; 206(1):1-9. doi: 10.1007/s00430-016-0481-y [Crossref]
  46. Patel JJ, Martindale RG, McClave SA. Relevant nutrition therapy in COVID-19 and the constraints on its delivery by a unique disease process. Nutr Clin Pract 2020; 35(5):792-9. doi: 10.1002/ncp.10566 [Crossref]
  47. Diabetes (India), National Diabetes Obesity and Cholesterol Foundation (NDOC), and Nutrition Expert Group, India. Balanced nutrition is needed in times of COVID19 epidemic in India: A call for action for all nutritionists and physicians. Diabetes Metab Syndr. 2020; 14(6): 1747–1750. doi: 10.1016/j.dsx.2020.08.030
  48. Rothan HA, Byrareddy SN. The epidemiology and pathogenesis of coronavirus disease (COVID-19) outbreak. J Autoimmun. 2020:102433. 10.1016/j.jaut.2020.102433
  49. Kesteloot H. Epidemiology: past, present and future. Verh K Acad Geneeskd Belg 2004; 66(5-6):384-405.
  50. Das UN. Polyunsaturated fatty acids and sepsis. Nutrition 2019; 65:39-43. doi: 10.1016/j.nut.2019.02.016 [Crossref]
  51. Husson M-O, Ley D, Portal C, Gottrand M, Hueso T, Desseyn J-L. Modulation of host defence against bacterial and viral infections by omega-3 polyunsaturated fatty acids. J Infect 2016; 73(6):523-35. doi: 10.1016/j.jinf.2016.10.001 [Crossref]
  52. Rutting S, Zakarya R, Bozier J, Xenaki D, Horvat JC, Wood LG. Dietary fatty acids amplify inflammatory responses to infection through p38 MAPK signaling. Am J Respir Cell Mol Biol 2019; 60(5):554-68. doi: 10.1165/rcmb.2018-0215OC [Crossref]
  53. Schwerbrock NM, Karlsson EA, Shi Q, Sheridan PA, Beck MA. Fish oil-fed mice have impaired resistance to influenza infection. J Nutr 2009; 139(8):1588-94. doi: 10.3945/jn.109.108027 [Crossref]
  54. Rutting S, Horvat J, Wood L, Hansbro P, Oliver B. The effect of dietary fatty acids on respiratory infection in human lung cells. Eur Respir J. 2018. 10.1183/13993003.congress-2018.PA4983
  55. Kang KW, Kim S, Cho Y-B, Ryu SR, Seo Y-J, Lee S-M. Endogenous n-3 Polyunsaturated Fatty Acids Are Beneficial to Dampen CD8+ T Cell-Mediated Inflammatory Response upon the Viral Infection in Mice. Int J Mol Sci 2019; 20(18):4510. doi: 10.3390/ijms20184510 [Crossref]
  56. Ramana CV, Cheng G-S, Kumar A, Kwon H-J, Enelow RI. Role of alveolar epithelial early growth response-1 (Egr-1) in CD8+ T cell-mediated lung injury. Mol Immunol 2009; 47(2-3):623-31. doi: 10.1016/j.molimm.2009.09.001 [Crossref]
  57. Baer B, Souza LMP, Pimentel AS, Veldhuizen RA. New insights into exogenous surfactant as a carrier of pulmonary therapeutics. Biochem Pharmacol 2019; 164:64-73. doi: 10.1016/j.bcp.2019.03.036 [Crossref]
  58. Niemarkt H, Hütten M, Kramer BW. Surfactant for respiratory distress syndrome: new ideas on a familiar drug with innovative applications. Neonatology 2017; 111(4):408-14. doi: 10.1159/000458466 [Crossref]
  59. Silver MR. Nutritional support in patients with respiratory failure. Clin Pulm Med 1994; 1(6):375-82.
  60. Alam SQ, Alam BS. Lung surfactant and fatty acid composition of lung tissue and lavage of rats fed diets containing different lipids. Lipids 1984; 19(1):38-43. doi: 10.1007/BF02534606 [Crossref]
  61. Langlois PL, D’Aragon F, Hardy G, Manzanares W. Omega-3 polyunsaturated fatty acids in critically ill patients with acute respiratory distress syndrome: A systematic review and meta-analysis. Nutrition 2019; 61:84-92. doi: 10.1016/j.nut.2018.10.026 [Crossref]
  62. Dushianthan A, Cusack R, Burgess VA, Grocott MP, Calder P. Immunonutrition for Adults With ARDS: Results From a Cochrane Systematic Review and Meta-Analysis. Respir Care 2020; 65(1):99-110. doi: 10.4187/respcare.06965 [Crossref]
  63. Turner KL, Moore FA, Martindale R. Nutrition support for the acute lung injury/adult respiratory distress syndrome patient: a review. Nutr Clin Pract 2011; 26(1):14-25. doi: 10.1177/0884533610393255 [Crossref]
  64. Herrmann M, Farrell C-JL, Pusceddu I, Fabregat-Cabello N, Cavalier E. Assessment of vitamin D status–a changing landscape. Clin Chem Lab Med 2017; 55(1):3-26. doi: 10.1515/cclm-2016-0264 [Crossref]
  65. Greiller CL, Martineau AR. Modulation of the immune response to respiratory viruses by vitamin D. Nutrients 2015; 7(6):4240-70. doi: 10.3390/nu7064240 [Crossref]
  66. Zisi D, Challa A, Makis A. The association between vitamin D status and infectious diseases of the respiratory system in infancy and childhood. Hormones 2019; 18(4):353-363. doi: 10.1007/s42000-019-00155-z [Crossref]
  67. Teymoori-Rad M, Shokri F, Salimi V, Marashi SM. The interplay between vitamin D and viral infections. Rev Med Virol 2019; 29(2):e2032. doi: 10.1002/rmv.2032 [Crossref]
  68. Zdrenghea MT, Makrinioti H, Bagacean C, Bush A, Johnston SL, Stanciu LA. Vitamin D modulation of innate immune responses to respiratory viral infections. Rev Med Virol 2017; 27(1):e1909. doi: 10.1002/rmv.1909 [Crossref]
  69. Lee M-D, Lin C-H, Lei W-T, Chang H-Y, Lee H-C, Yeung C-Y. Does vitamin D deficiency affect the immunogenic responses to influenza vaccination? A systematic review and meta-analysis. Nutrients 2018; 10(4):409. doi: 10.3390/nu10040409 [Crossref]
  70. Gruber-Bzura BM. Vitamin D and Influenza—Prevention or Therapy?. Int J Mol Sci 2018; 19(8):2419. doi: 10.3390/ijms19082419 [Crossref]
  71. Baeke F, Takiishi T, Korf H, Gysemans C, Mathieu C. Vitamin D: modulator of the immune system. Curr Opin Pharmacol. 2010 Aug;10(4):482-96. 10.1016/j.coph.2010.04.001
  72. Julkunen I, Melén K, Nyqvist M, Pirhonen J, Sareneva T, Matikainen S. Inflammatory responses in influenza A virus infection. Vaccine 2000; 19:S32-S7. doi: 10.1016/s0264-410x(00)00275-9 [Crossref]
  73. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ. COVID-19: consider cytokine storm syndromes and immunosuppression. The Lancet 2020; 395(10229):1033-1034. doi: 10.1016/S0140-6736(20)30628-0 [Crossref]
  74. Rezaei R, Aslani S, Marashi M, Rezaei F, Sharif-Paghaleh E. Immunomodulatory Effects of Vitamin D in Influenza Infection. Curr Immunol Rev 2018; 14(1):40-9. doi: 10.2174/1573395513666171031162100 [Crossref]
  75. Sadeghi K, Wessner B, Laggner U, Ploder M, Tamandl D, Friedl J. Vitamin D3 down-regulates monocyte TLR expression and triggers hyporesponsiveness to pathogen-associated molecular patterns. Eur J Immunol 2006; 36(2):361-70. doi: 10.1002/eji.200425995 [Crossref]
  76. Jiménez-Sousa MÁ, Martínez I, Medrano LM, Fernández-Rodríguez A, Resino S. Vitamin D in human immunodeficiency virus infection: influence on immunity and disease. Front Immunol 2018; 9:458. doi: 10.3389/fimmu.2018.00458 [Crossref]
  77. Nonnecke BJ, McGill JL, Ridpath JF, Sacco RE, Lippolis JD, Reinhardt TA. Acute phase response elicited by experimental bovine diarrhea virus (BVDV) infection is associated with decreased vitamin D and E status of vitamin-replete preruminant calves. J Dairy Sci 2014; 97(9):5566-79. doi: 10.3168/jds.2014-8293 [Crossref]
  78. Cannell J, Vieth R, Umhau J, Holick M, Grant W, Madronich S. Epidemic influenza and vitamin D. Epidemiol Infect 2006; 134(6):1129-40. doi: 10.1017/S0950268806007175 [Crossref]
  79. Cannell JJ, Vieth R, Willett W, Zasloff M, Hathcock JN, White JH. Cod liver oil, vitamin A toxicity, frequent respiratory infections, and the vitamin D deficiency epidemic. Ann Otol Rhinol Laryngol 2008; 117(11):864-70. doi: 10.1177/000348940811701112 [Crossref]
  80. Cannell JJ, Zasloff M, Garland CF, Scragg R, Giovannucci E. On the epidemiology of influenza. Virol J 2008; 5(1):29. doi: 10.1186/1743-422X-5-29 [Crossref]
  81. Martineau AR, Jolliffe DA, Hooper RL, Greenberg L, Aloia JF, Bergman P. Vitamin D supplementation to prevent acute respiratory tract infections: systematic review and meta-analysis of individual participant data. BMJ 2017; 356:i6583. doi: 10.1136/bmj.i6583 [Crossref]
  82. Yin Y, Wunderink RG. MERS, SARS and other coronaviruses as causes of pneumonia. Respirology 2018; 23(2):130-7. doi: 10.1111/resp [Crossref]
  83. Xu H, Zhong L, Deng J, Peng J, Dan H, Zeng X. High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int J Oral Sci 2020; 12:8. doi: 10.1038/s41368-020-0074-x [Crossref]
  84. Zhao Y, Zhao Z, Wang Y, Zhou Y, Ma Y, Zuo W. Single-cell RNA expression profiling of ACE2, the putative receptor of Wuhan 2019-nCov. BioRxiv 2020; 202(5):756-759.
  85. Xu J, Yang J, Chen J, Luo Q, Zhang Q, Zhang H. Vitamin D alleviates lipopolysaccharide-induced acute lung injury via regulation of the renin-angiotensin system. Mol Med Rep 2017; 16(5):7432-8. doi: 10.3892/mmr.2017.7546 [Crossref]
  86. Young JI , Züchner S . Wang GRegulation of the epigenome by vitamin C. Annu Rev Nutr 2015; 35:545-64. doi: 10.1146/annurev-nutr-071714-034228 [Crossref]
  87. Hemila H. Vitamin C and Infections. Nutrients 2017; 9(11):1211. doi: 10.3390/nu9111211 [Crossref]
  88. Hemilä H. Vitamin C and sex differences in respiratory tract infections. Respir Med 2008; 102(4):625-6. doi: 10.1016/j.rmed.2007.12.011 [Crossref]
  89. Castro SM, Guerrero-Plata A, Suarez-Real G, Adegboyega PA, Colasurdo GN, Khan AM. Antioxidant treatment ameliorates respiratory syncytial virus–induced disease and lung inflammation. Am J Respir Crit Care Med 2006; 174(12):1361-9. doi: 10.1164/rccm.200603-319OC [Crossref]
  90. Hemilä H. Vitamin C and SARS coronavirus. J Antimicrob Chemother 2003; 52(6):1049-50. doi: 10.1093/jac/dkh002 [Crossref]
  91. Hemilä H, Douglas RM. Vitamin C and acute respiratory infections. Int J Tuberc Lung Dis 1999; 3(9):756-61.
  92. Hemilä H. Vitamin C intake and susceptibility to pneumonia. Pediatr Infect Dis J 1997; 16(9):836-7. doi: 10.1097/00006454-199709000-00003 [Crossref]
  93. Fowler AA 3rd, Truwit JD, Hite RD, Morris PE, DeWilde C, Priday A, Fisher B. Effect of vitamin C infusion on organ failure and biomarkers of inflammation and vascular injury in patients with sepsis and severe acute respiratory failure: the CITRIS-ALI randomized clinical trial. JAMA 2019; 322(13):1261-70. doi: 10.1001/jama.2019.11825 [Crossref]
  94. Bergsten P, Amitai G, Kehrl J, Dhariwal KR, Klein HG, Levine M. Millimolar concentrations of ascorbic acid in purified human mononuclear leukocytes. Depletion and reaccumulation. J Biol Chem 1990; 265(5):2584-7.
  95. Tan PH, Sagoo P, Chan C, Yates JB, Campbell J, Beutelspacher SC. Inhibition of NF-kappa B and oxidative pathways in human dendritic cells by antioxidative vitamins generates regulatory T cells. J Immunol 2005; 174(12):7633-44. doi: 10.4049/jimmunol.174.12.7633 [Crossref]
  96. Carr AC, Maggini S. Vitamin C and immune function. Nutrients 2017; 9(11):1211. doi: 10.3390/nu9111211 [Crossref]
  97. Lammermann T. In the eye of the neutrophil swarm-navigation signals that bring neutrophils together in inflamed and infected tissues. J Leukoc Biol 2016; 100(1):55-63. doi: 10.1189/jlb.1MR0915-403 [Crossref]
  98. Goldschmidt MC, Masin WJ, Brown LR, Wyde PR. The effect of ascorbic acid deficiency on leukocyte phagocytosis and killing of actinomyces viscosus. Int J Vitam Nutr Res 1988; 58(3):326-34.
  99. Hunt C, Chakravorty N, Annan G, Habibzadeh N, Schorah C. The clinical effects of vitamin C supplementation in elderly hospitalised patients with acute respiratory infections. Int J Vitam Nutr Res 1994; 64(3):212-9.
  100. Haskell B, Johnston C. Complement component C1q activity and ascorbic acid nutriture in guinea pigs. Am J Clin Nutr 1991; 54(6):1228S-30S. doi: 10.1093/ajcn/54.6.1228s [Crossref]
  101. Johnston CS, Cartee GD, Haskell BE. Effect of ascorbic acid nutriture on protein-bound hydroxyproline in guinea pig plasma. J Nutr 1985; 115(8):1089-93. doi: 10.1093/jn/115.8.1089 [Crossref]
  102. Johnston CS, Kolb WP, Haskell BE. The effect of vitamin C nutriture on complement component C1q concentrations in guinea pig plasma. J Nutr 1987; 117(4):764-8. doi: 10.1093/jn/117.4.764 [Crossref]
  103. Feigen G, Smith B, Dix C, Flynn C, Peterson N, Rosenberg L. Enhancement of antibody production and protection against systemic anaphylaxis by large doses of vitamin C. Res Commun Chem Pathol Pharmacol 1982; 38(2):313-33.
  104. Prinz W, Bloch J, Gilich G, Mitchell G. systematic study of the effect of vitamin C supplementation on the humoral immune response in ascorbate-dependent mammals. I. The antibody response to sheep red blood cells (a T-dependent antigen) in guinea pigs. Int J Vitam Nutr Res 1980; 50(3):294-300.
  105. Shilotri P, Bhat K. Effect of mega doses of vitamin C on bactericidal ativity of leukocytes. The Am J Clin Nutr 1977; 30(7):1077-81. doi: 10.1093/ajcn/30.7.1077 [Crossref]
  106. Delafuente JC, Prendergast JM, Modigh A. Immunologic modulation by vitamin C in the elderly. Int J Immunopharmacol 1986; 8(2):205-11. doi: 10.1016/0192-0561(86)90060-3 [Crossref]
  107. Vogel RI, Lamster IB, Wechsler SA, Macedo B, Hartley LJ, Macedo JA. The effects of megadoses of ascorbic acid on PMN chemotaxis and experimental gingivitis. J Periodontol 1986; 57(8):472-9. doi: 10.1902/jop.1986.57.8.472 [Crossref]
  108. Hemilä H, Louhiala P. Vitamin C for preventing and treating pneumonia. Cochrane Database Syst Rev. 2013; (8):CD005532. doi: 10.1002/14651858.CD005532.pub3.
  109. Imdad A, Mayo-Wilson E, Herzer K, Bhutta ZA. Vitamin A supplementation for preventing morbidity and mortality in children from six months to five years of age. Cochrane Database Syst Rev. 2017; 3(3):CD008524. 10.1002/14651858.CD008524.pub3
  110. Aukrust P, Müller F, Ueland T, Svardal A, Berge R, Frøland S. Decreased vitamin A levels in common variable immunodeficiency: vitamin A supplementation in vivo enhances immunoglobulin production and downregulates inflammatory responses. Eur J Clin Invest 2000; 30(3):252-9. doi: 10.1046/j.1365-2362.2000.00619.x [Crossref]
  111. Stephensen CB. Vitamin A, infection, and immune function. Annu Rev Nutr 2001; 21(1):167-92. doi: 10.1146/annurev.nutr.21.1.167 [Crossref]
  112. McDowell EM, Keenan KP, Huang M. Effects of vitamin A-deprivation on hamster tracheal epithelium. Virchows Arch B Cell Pathol Incl Mol Pathol 1984; 45(1):197-219. doi: 10.1007/BF02889865 [Crossref]
  113. Twining SS, Zhou X, Schulte DP, Wilson PM, Fish B, Moulder J. Effect of vitamin A deficiency on the early response to experimental Pseudomonas keratitis. Invest Ophthalmol Vis Sci 1996; 37(4):511-22.
  114. Sommer A, Katz J, Tarwotjo I. Increased risk of respiratory disease and diarrhea in children with preexisting mild vitamin A deficiency. Am J Clin Nutr 1984; 40(5):1090-5. doi: 10.1093/ajcn/40.5.1090 [Crossref]
  115. Chattha KS, Kandasamy S, Vlasova AN, Saif LJ. Vitamin A deficiency impairs adaptive B and T cell responses to a prototype monovalent attenuated human rotavirus vaccine and virulent human rotavirus challenge in a gnotobiotic piglet model. PloS One 2013; 8(12):e82966. doi: 10.1371/journal.pone.0082966 [Crossref]
  116. Vlasova AN, Chattha KS, Kandasamy S, Siegismund CS, Saif LJ. Prenatally acquired vitamin A deficiency alters innate immune responses to human rotavirus in a gnotobiotic pig model. J Immunol 2013; 190(9):4742-53. doi: 10.4049/jimmunol [Crossref]
  117. Baumann H, Gauldie J. The acute phase response. ImmunoloToday 1994 Feb; 15(2):74-80. doi: 10.1016/0167-5699(94)90137-6 [Crossref]
  118. West CE, Sijtsma SR, Kouwenhoven B, Rombout JH, van der Zijpp AJ. Epithelia-damaging virus infections affect vitamin A status in chickens. J Nutr 1992; 122(2):333-9. doi: 10.1093/jn/122.2.333 [Crossref]
  119. Fawzi WW, Mbise RL, Fataki MR, Herrera MG, Kawau F, Hertzmark E. Vitamin A supplementation and severity of pneumonia in children admitted to the hospital in Dar es Salaam, Tanzania. Am J Clin Nutr 1998; 68(1):187-92. doi: 10.1093/ajcn/68.1.187 [Crossref]
  120. Stephensen CB, Franchi LM, Hernandez H, Campos M, Gilman RH, Alvarez JO. Adverse effects of high-dose vitamin A supplements in children hospitalized with pneumonia. Pediatrics 1998; 101(5):e3. doi: 10.1542/peds.101.5.e3 [Crossref]
  121. Julien MR, da Cruz Gomes A, Varandas L, Rodrigues P, Malveiro F, Aguiar P. A randomized, double‐blind, placebo‐controlled clinical trial of vitamin A in Mozambican children hospitalized with nonmeasles acute lower respiratory tract infections. Trop Med Int Health 1999; 4(12):794-800. doi: 10.1046/j.1365-3156.1999.00493.x [Crossref]
  122. Rodríguez A, Hamer DH, Rivera J, Acosta M, Salgado G, Gordillo M. Effects of moderate doses of vitamin A as an adjunct to the treatment of pneumonia in underweight and normal-weight children: a randomized, double-blind, placebo-controlled trial. Am J Clin Nutr 2005; 82(5):1090-6. doi: 10.1093/ajcn/82.5.1090 [Crossref]
  123. Cameron C, Dallaire F, Vézina C, Muckle G, Bruneau S, Ayotte P. Neonatal vitamin A deficiency and its impact on acute respiratory infections among preschool Inuit children. Can J Public Health 2008; 99(2):102-6. doi: 10.1007/BF03405454 [Crossref]
  124. Imdad A, Yakoob MY, Sudfeld C, Haider BA, Black RE, Bhutta ZA. Impact of vitamin A supplementation on infant and childhood mortality. BMC public health 2011; 11(S3):S20. doi: 10.1186/1471-2458-11-S3-S20 [Crossref]
  125. Sommer A, Rahmathullah L, Underwood B, Milton R, Reddy V, West K. Potential interventions for the prevention of childhood pneumonia in developing countries: a meta-analysis of data from field trials to assess the impact of vitamin A supplementation on pneumonia morbidity and mortality. The Vitamin A and Pneumonia Working Group. Bull World Health Organ 1995; 73(5):609-19.
  126. Trottier C, Colombo M, Mann KK, Miller Jr WH, Ward BJ. Retinoids inhibit measles virus through a type I IFN-dependent bystander effect. The FASEB J 2009; 23(9):3203-12. doi: 10.1096/fj.09-129288 [Crossref]
  127. Villamor E, Fawzi WW. Vitamin A supplementation: implications for morbidity and mortality in children. J Infect Dis 2000; 182(Supplement_1):S122-S33. doi: 10.1086/315921 [Crossref]
  128. Bril F, Biernacki DM, Kalavalapalli S, Lomonaco R, Subbarayan SK, Lai J. Role of Vitamin E for Nonalcoholic Steatohepatitis in Patients With Type 2 Diabetes: A Randomized Controlled Trial. Diabetes Care 2019; 42(8):1481-1488. doi: 10.2337/dc19-0167 [Crossref]
  129. Jiang Q, Christen S, Shigenaga MK, Ames BN. γ-Tocopherol, the major form of vitamin E in the US diet, deserves more attention. Am J Clin Nutr 2001; 74(6):714-22. doi: 10.1093/ajcn/74.6.714 [Crossref]
  130. Krumova K, Friedland S, Cosa G. How lipid unsaturation, peroxyl radical partitioning, and chromanol lipophilic tail affect the antioxidant activity of α-tocopherol: direct visualization via high-throughput fluorescence studies conducted with fluorogenic α-tocopherol analogues. J Am Chem Soc 2012; 134(24):10102-13. doi: 10.1021/ja301680m [Crossref]
  131. Gavazzi G, Krause K-H. Ageing and infection. Lancet Infect Dis 2002; 2(11):659-66. doi: 10.1016/s1473-3099(02)00437-1 [Crossref]
  132. Lewis ED, Meydani SN, Wu D. Regulatory role of vitamin E in the immune system and inflammation. IUBMB life 2019; 71(4):487-94. doi: 10.1002/iub.1976 [Crossref]
  133. Han SN, Meydani SN. Impact of vitamin E on immune function and its clinical implications. Expert Rev Clin Immunol 2006; 2(4):561-7. doi: 10.1586/1744666X.2.4.561 [Crossref]
  134. Moriguchi S, Kobayashi N, Kishino Y. High dietary intakes of vitamin E and cellular immune functions in rats. J Nutr 1990; 120(9):1096-102. doi: 10.1093/jn/120.9.1096 [Crossref]
  135. Meydani SN, Meydani M, Blumberg JB, Leka LS, Siber G, Loszewski R. Vitamin E supplementation and in vivo immune response in healthy elderly subjects: a randomized controlled trial. JAMA 1997; 277(17):1380-6. doi: 10.1001/jama.1997.03540410058031 [Crossref]
  136. De la Fuente M, Hernanz A, Guayerbas N, Manuel Victor V, Arnalich F. Vitamin E ingestion improves several immune functions in elderly men and women. Free Radic Res 2008; 42(3):272-80. doi: 10.1080/10715760801898838 [Crossref]
  137. Wu D, Nikbin Meydani S. Age-associated changes in immune function: impact of vitamin E intervention and the underlying mechanisms. Endocr Metab Immune Disord Drug Targets 2014; 14(4):283-9. doi: 10.2174/1871530314666140922143950 [Crossref]
  138. Meydani SN, Leka LS, Fine BC, Dallal GE, Keusch GT, Singh MF. Vitamin E and respiratory tract infections in elderly nursing home residents: a randomized controlled trial. JAMA 2004; 292(7):828-36. doi: 10.1001/jama.292.7.828 [Crossref]
  139. Miller RA. The aging immune system: primer and prospectus. Science 1996; 273(5271):70-4. doi: 10.1126/science.273.5271.70 [Crossref]
  140. Wang Y, Watson RR. Vitamin E supplementation at various levels alters cytokine production by thymocytes during retrovirus infection causing murine AIDS. Thymus 1994; 22(3):153-65.
  141. Graat JM, Schouten EG, Kok FJ. Effect of daily vitamin E and multivitamin-mineral supplementation on acute respiratory tract infections in elderly persons: a randomized controlled trial. JAMA 2002; 288(6):715-21. doi: 10.1001/jama.288.6.715 [Crossref]
  142. Clarke MW, Burnett JR, Croft KD. Vitamin E in human health and disease. Crit Rev Clin Lab Sci 2008; 45(5):417-50. doi: 10.1080/10408360802118625 [Crossref]
  143. Vellema P, Rutten V, Hoek A, Moll L, Wentink G. The effect of cobalt supplementation on the immune response in vitamin B12 deficient Texel lambs. Vet Immunol Immunopathol 1996; 55(1-3):151-61. doi: 10.1016/s0165-2427(96)05560-2 [Crossref]
  144. Maggini S, Wintergerst ES, Beveridge S, Hornig DH. Selected vitamins and trace elements support immune function by strengthening epithelial barriers and cellular and humoral immune responses. Br J Nutr 2007; 98(S1):S29-S35. doi: 10.1017/S0007114507832971 [Crossref]
  145. Saeed F, Nadeem M, Ahmed RS, Tahir Nadeem M, Arshad MS, Ullah A. Studying the impact of nutritional immunology underlying the modulation of immune responses by nutritional compounds – a review. Food Agr Immunol 2016; 27(2):205-29. doi: 10.1080/09540105.2015.1079600 [Crossref]
  146. Fata FT, Herzlich BC, Schiffman G, Ast AL. Impaired antibody responses to pneumococcal polysaccharide in elderly patients with low serum vitamin B12 levels. Ann Intern Med 1996; 124(3):299-304. doi: 10.7326/0003-4819-124-3-199602010-00003 [Crossref]
  147. Bunout D, Barrera G, Hirsch S, Gattas V, de la Maza MP, Haschke F. Effects of a nutritional supplement on the immune response and cytokine production in free-living Chilean elderly. J Parenter Enteral Nutr 2004; 28(5):348-54. doi: 10.1177/0148607104028005348 [Crossref]
  148. Stabler SP, Lindenbaum J, Allen RH. Vitamin B-12 deficiency in the elderly: current dilemmas. Am J Clin Nutr 1997; 66(4):741-9. doi: 10.1093/ajcn/66.4.741 [Crossref]
  149. Smith AD. Folic acid fortification: the good, the bad, and the puzzle of vitamin B-12. Am J Clin Nutr 2007; 85(1):3-5. doi: 10.1093/ajcn/85.1.3 [Crossref]
  150. Duthie SJ, Horgan G, De Roos B, Rucklidge G, Reid M, Duncan G. Blood folate status and expression of proteins involved in immune function, inflammation, and coagulation: biochemical and proteomic changes in the plasma of humans in response to long-term synthetic folic acid supplementation. J Proteome Res 2010; 9(4):1941-50. doi: 10.1021/pr901103n [Crossref]
  151. Wintergerst ES, Maggini S, Hornig DH. Contribution of selected vitamins and trace elements to immune function. Ann Nutr Metab 2007; 51(4):301-23. doi: 10.1159/000107673 [Crossref]
  152. Sheybani Z, Heydari Dokoohaki, Negahdaripour M, Dehdashti CH, Zolghadr H, Moghadami M, et al. The Role of folic acid in the management of respiratory disease caused by COVID-19.ChemRxiv.2020. Preprint.
  153. Mooney S, Leuendorf J-E, Hendrickson C, Hellmann H. Vitamin B6: a long known compound of surprising complexity. Molecules 2009; 14(1):329-51. doi: 10.3390/molecules14010329 [Crossref]
  154. Hellmann H, Mooney S. Vitamin B6: a molecule for human health?. Molecules 2010; 15(1):442-59. doi: 10.3390/molecules15010442 [Crossref]
  155. Oka T. Modulation of gene expression by vitamin B 6. Nutr Res Rev 2001; 14(2):257-66. doi: 10.1079/NRR200125 [Crossref]
  156. Rail LC, Meydani SN. Vitamin B6 and immune competence. Nutr Rev 1993; 51(8):217-25. doi: 10.1111/j.1753-4887.1993.tb03109.x [Crossref]
  157. Chandra RK, Sudhakaran L. Regulation of immune responses by vitamin B6. Ann N Y Acad Sci 1990; 585(1):404-23. doi: 10.1111/j.1749-6632.1990.tb28073.x [Crossref]
  158. Trakatellis A, Dimitriadou A, Trakatelli M. Pyridoxine deficiency: new approaches in immunosuppression and chemotherapy. Postgrad Med J 1997; 73(864):617-22. doi: 10.1136/pgmj.73.864.617 [Crossref]
  159. Shaker SM, Fathy H, Abdelall EK, Said AS. The effect of zinc and Vitamin A supplements in treating and reducing the incidence of upper respiratory tract infections in children. Natl J Physiol Pharm Pharmacol 2018; 8(7):1010-7. doi: 10.5455/njppp.2018.8.0104206032018 [Crossref]
  160. Shankar AH, Prasad AS. Zinc and immune function: the biological basis of altered resistance to infection. Am J Clin Nutr 1998; 68(2):447S-63S. doi: 10.1093/ajcn/68.2.447S [Crossref]
  161. King LE, Frentzel JW, Mann JJ, Fraker PJ. Chronic zinc deficiency in mice disrupted T cell lymphopoiesis and erythropoiesis while B cell lymphopoiesis and myelopoiesis were maintained. J Am Coll Nutr 2005; 24(6):494-502. doi: 10.1080/07315724.2005.10719495 [Crossref]
  162. Ma J, Betts NM. Zinc and copper intakes and their major food sources for older adults in the 1994–96 continuing survey of food intakes by individuals (CSFII). J Nutr 2000; 130(11):2838-43. doi: 10.1093/jn/130.11.2838 [Crossref]
  163. Briefel RR, Bialostosky K, Kennedy-Stephenson J, McDowell MA, Ervin RB, Wright JD. Zinc intake of the US population: findings from the third National Health and Nutrition Examination Survey, 1988–1994. J Nutr 2000; 130(5):1367S-73S. doi: 10.1093/jn/130.5.1367S [Crossref]
  164. Hotz C, Peerson JM, Brown KH. Suggested lower cutoffs of serum zinc concentrations for assessing zinc status: reanalysis of the second National Health and Nutrition Examination Survey data (1976–1980). Am J Clin Nutr 2003; 78(4):756-64. doi: 10.1093/ajcn/78.4.756 [Crossref]
  165. Ravaglia G, Forti P, Maioli F, Nesi B, Pratelli L, Savarino L. Blood micronutrient and thyroid hormone concentrations in the oldest-old. J Clin Endocrinol Metab 2000; 85(6):2260-5. doi: 10.1210/jcem.85.6.6627 [Crossref]
  166. Prasad AS, Beck FW, Bao B, Fitzgerald JT, Snell DC, Steinberg JD. Zinc supplementation decreases incidence of infections in the elderly: effect of zinc on generation of cytokines and oxidative stress. Am J Clin Nutr 2007; 85(3):837-44. doi: 10.1093/ajcn/85.3.837 [Crossref]
  167. Kahmann L, Uciechowski P, Warmuth S, Plümäkers B, Gressner AM, Malavolta M. Zinc supplementation in the elderly reduces spontaneous inflammatory cytokine release and restores T cell functions. Rejuvenation Res 2008; 11(1):227-37. doi: 10.1089/rej.2007.0613 [Crossref]
  168. Fortes C, Forastiere F, Agabiti N, Fano V, Pacifici R, Virgili F. The effect of zinc and vitamin A supplementation on immune response in an older population. J Am Geriatr Soc 1998; 46(1):19-26. doi: 10.1111/j.1532-5415.1998.tb01008.x [Crossref]
  169. Te Velthuis AJ, van den Worm SH, Sims AC, Baric RS, Snijder EJ, van Hemert MJ. Zn2+ inhibits coronavirus and arterivirus RNA polymerase activity in vitro and zinc ionophores block the replication of these viruses in cell culture. PLoS Pathog 2010; 6(11):e1001176. doi: 10.1371/journal.ppat.1001176 [Crossref]
  170. Lassi ZS, Moin A, Bhutta ZA. Zinc supplementation for the prevention of pneumonia in children aged 2 months to 59 months. Cochrane Database Syst Rev 2016; 12(12):CD005978. doi: 10.1002/14651858.CD005978.pub3 [Crossref]
  171. Phillips JM, Gallagher T, Weiss SR. Neurovirulent murine coronavirus JHM. SD uses cellular zinc metalloproteases for virus entry and cell-cell fusion. J Virol 2017; 91(8):e01564-16. doi: 10.1128/JVI.01564-16 [Crossref]
  172. Tan YW, Hong W, Liu DX. Binding of the 5′-untranslated region of coronavirus RNA to zinc finger CCHC-type and RNA-binding motif 1 enhances viral replication and transcription. Nucleic Acids Res 2012; 40(11):5065-77. doi: 10.1093/nar/gks165 [Crossref]
  173. Ferreira AJ, Raizada MK. Are we poised to target ACE2 for the next generation of antihypertensives?. J Mol Med (Berl) 2008; 86(6):685-90. doi: 10.1007/s00109-008-0339-x [Crossref]
  174. Lal A. Iron in health and disease: an update. Indian J Pediatr 2020; 87(1):58-65. doi: 10.1007/s12098-019-03054-8 [Crossref]
  175. Dev S, Babitt JL. Overview of iron metabolism in health and disease. Hemodial Int 2017; 21:S6-S20. doi: 10.1111/hdi.12542 [Crossref]
  176. Doherty CP. Host-pathogen interactions: the role of iron. J Nutr 2007; 137(5):1341-4. doi: 10.1093/jn/137.5.1341 [Crossref]
  177. Potrykus J, Ballou ER, Childers DS, Brown AJ. Conflicting interests in the pathogen–host tug of war: fungal micronutrient scavenging versus mammalian nutritional immunity. PLoS Pathog 2014; 10(3):e1003910. doi: 10.1371/journal.ppat.1003910 [Crossref]
  178. Verbon EH, Trapet PL, Stringlis IA, Kruijs S, Bakker PAHM, Pieterse CMJ. Iron and Immunity. Annu Rev Phytopathol 2017; 55(1):355-75. doi: 10.1146/annurev-phyto-080516-035537 [Crossref]
  179. Silva B, Faustino P. An overview of molecular basis of iron metabolism regulation and the associated pathologies. Biochim Biophys Acta 2015; 1852(7):1347-59. doi: 10.1016/j.bbadis.2015.03.011 [Crossref]
  180. Lewis JP. Metal uptake in host-pathogen interactions: role of iron in Porphyromonas gingivalis interactions with host organisms. Periodontol 2000 2010; 52(1):94-116. doi: 10.1111/j.1600-0757.2009.00329.x [Crossref]
  181. Jayaweera JAAS, Reyes M, Joseph A. Childhood iron deficiency anemia leads to recurrent respiratory tract infections and gastroenteritis. 2019 9; 12637:1. doi: 10.1038/s41598-019-49122-z [Crossref]
  182. Wakabayashi H, Oda H, Yamauchi K, Abe F. Lactoferrin for prevention of common viral infections. J Infect Chemother 2014; 20(11):666-71. doi: 10.1016/j.jiac.2014.08.003 [Crossref]
  183. Weinberg E. Iron withholding: a defense against viral infections. Biometals 1996; 9(4):393-9. doi: 10.1007/BF00140609 [Crossref]
  184. Oppenheimer SJ. Iron and its relation to immunity and infectious disease. J Nutr 2001; 131(2):616S-33S. doi: 10.1093/jn/131.2.616S [Crossref]
  185. Harthill M. Micronutrient selenium deficiency influences evolution of some viral infectious diseases. Biol Trace Elem Res 2011; 143(3):1325-36. doi: 10.1007/s12011-011-8977-1 [Crossref]
  186. dos Reis A.R., El-Ramady H., Santos E.F., Gratão P.L., Schomburg L. Overview of Selenium Deficiency and Toxicity Worldwide: Affected Areas, Selenium-Related Health Issues, and Case Studies. In: Pilon-Smits E., Winkel L., Lin ZQ. (eds) Selenium in plants. Plant Ecophysiology, vol 11. Springer, Cham. 2017. 10.1007/978-3-319-56249-0_13
  187. Khomich OA, Kochetkov SN, Bartosch B, Ivanov AV. Redox biology of respiratory viral infections. Viruses 2018; 10(8):392. doi: 10.3390/v10080392 [Crossref]
  188. Lim J, Oh E, Kim Y, Jung W, Kim H, Lee J. Enhanced oxidative damage to DNA, lipids, and proteins and levels of some antioxidant enzymes, cytokines, and heat shock proteins in patients infected with influenza H1N1 virus. Acta Virol 2014; 58(3):253-60. doi: 10.4149/av_2014_03_253 [Crossref]
  189. Ng MP, Lee JC, Loke WM, Yeo LL, Quek AM, Lim EC. Does influenza A infection increase oxidative damage? . Antioxid Redox Signal 2014; 21(7):1025-31. doi: 10.1089/ars.2014.5907 [Crossref]
  190. Erkekoğlu P, Aşçı A, Ceyhan M, Kızılgün M, Schweizer U, Ataş C. Selenium levels, selenoenzyme activities and oxidant/antioxidant parameters in H1N1-infected children. Turk J Pediatr 2013; 55(3):271-82.
  191. Sheridan PA, Zhong N, Carlson BA, Perella CM, Hatfield DL, Beck MA. Decreased selenoprotein expression alters the immune response during influenza virus infection in mice. J Nutr 2007; 137(6):1466-71. doi: 10.1093/jn/137.6.1466 [Crossref]
  192. Arthur JR, McKenzie RC, Beckett GJ. Selenium in the immune system. J Nutr 2003; 133(5):1457S-9S. doi: 10.1093/jn/133.5.1457S [Crossref]
  193. Spallholz JE, Boylan LM, Larsen HS. Advances in understanding selenium’s role in the immune system. Ann N Y Acad Sci 1990; 587(1):123-39. doi: 10.1111/j.1749-6632.1990.tb00140.x [Crossref]
  194. Avery JC, Hoffmann PR. Selenium, selenoproteins, and immunity. Nutrients 2018; 10(9):1203. doi: 10.3390/nu10091203 [Crossref]
  195. Kiremidjian-Schumacher L, Roy M, Glickman R, Schneider K, Rothstein S, Cooper J. Selenium and immunocompetence in patients with head and neck cancer. Biological Trace Element Research 2000; 73(2):97-111.
  196. Roy M, Kiremidjian-Schumacher L, Wishe HI, Cohen MW, Stotzky G. Supplementation with selenium and human immune cell functions. Biological trace element research 1994; 41(1-2):103.
  197. Hoffmann PR, Berry MJ. The influence of selenium on immune responses. Molecular nutrition & food research 2008; 52(11):1273-80.
  198. Broome CS, McArdle F, Kyle JA, Andrews F, Lowe NM, Hart CA. An increase in selenium intake improves immune function and poliovirus handling in adults with marginal selenium status. The American journal of clinical nutrition 2004; 80(1):154-62.
  199. Huang Z, Rose AH, Hoffmann PR. The role of selenium in inflammation and immunity: from molecular mechanisms to therapeutic opportunities. Antioxidants & redox signaling 2012; 16(7):705-43.
  200. Chaitra K, Kumar M, Reddy S. Hyponatremia in lower respiratory tract infections. Int J Contemp Pediatr. 2016;3(2):3814. doi.org10.18203/2349-3291.ijcp20161018.
  201. Hanna S, Tibby S, Durward A, Murdoch I. Incidence of hyponatraemia and hyponatraemic seizures in severe respiratory syncytial virus bronchiolitis. Acta Paediatr 2003; 92(4):430-4. doi: 10.1111/j.1651-2227.2003.tb00573.x [Crossref]
  202. Nair V, Niederman MS, Masani N, Fishbane S. Hyponatremia in community-acquired pneumonia. Am J Nephrol 2007; 27(2):184-90. doi: 10.1159/000100866 [Crossref]
  203. Hansen B-A, Bruserud Ø. Hypomagnesemia in critically ill patients. J Intensive Care 2018; 6(1):21. doi: 10.1186/s40560-018-0291-y [Crossref]
  204. Choi HS, Kwon A, Chae HW, Suh J, Kim DH, Kim H-S. Respiratory failure in a diabetic ketoacidosis patient with severe hypophosphatemia. Ann Pediatr Endocrinol Metab 2018; 23(2):103-106. doi: 10.6065/apem.2018.23.2.103 [Crossref]
  205. Naithani R, Huma LC, Holland LE, Shukla D, McCormick DL, Mehta RG. Antiviral activity of phytochemicals: a comprehensive review. Mini Rev Med Chem 2008; 8(11):1106-33. doi: 10.2174/138955708785909943 [Crossref]
  206. Zakaryan H, Arabyan E, Oo A, Zandi K. Flavonoids: promising natural compounds against viral infections. Arch Virol 2017; 162(9):2539-51. doi: 10.1007/s00705-017-3417-y [Crossref]
  207. Ryu YB, Jeong HJ, Kim JH, Kim YM, Park J-Y, Kim D. Biflavonoids from Torreya nucifera displaying SARS-CoV 3CLpro inhibition. Bioorg Med Chem 2010; 18(22):7940-7. doi: 10.1016/j.bmc.2010.09.035 [Crossref]
  208. Nguyen TTH, Woo H-J, Kang H-K, Kim Y-M, Kim D-W, Ahn S-A. Flavonoid-mediated inhibition of SARS coronavirus 3C-like protease expressed in Pichia pastoris. Biotechnol Lett 2012; 34(5):831-8. doi: 10.1007/s10529-011-0845-8 [Crossref]
  209. Schwarz S, Sauter D, Wang K, Zhang R, Sun B, Karioti A. Kaempferol derivatives as antiviral drugs against the 3a channel protein of coronavirus. Planta Med 2014; 80(02/03):177-82. doi: 10.1055/s-0033-1360277 [Crossref]
  210. Jo S, Kim H, Kim S, Shin DH, Kim MS. Characteristics of flavonoids as potent MERS‐CoV 3C‐like protease inhibitors. Chem Biol Drug Des 2019; 94(6):2023-30. doi: 10.1111/cbdd.13604 [Crossref]
  211. Klinge CM, Blankenship KA, Risinger KE, Bhatnagar S, Noisin EL, Sumanasekera WK. Resveratrol and estradiol rapidly activate MAPK signaling through estrogen receptors α and β in endothelial cells. J Biol Chem 2005; 280(9):7460-8. doi: 10.1074/jbc.M411565200 [Crossref]
  212. Li S, Zhao G, Chen L, Ding Y, Lian J, Hong G. Resveratrol protects mice from paraquatinduced lung injury: The important role of SIRT1 and NRF2 antioxidant pathways. Mol Med Rep 2016; 13(2):1833-8. doi: 10.3892/mmr.2015.4710 [Crossref]
  213. Jeong J, Juhn K, Lee H, Kim S-H, Min B-H, Lee K-M. SIRT1 promotes DNA repair activity and deacetylation of Ku70. Exp Mol Med 2007; 39(1):8-13. doi: 10.1038/emm.2007.2 [Crossref]
  214. Kolthur-Seetharam U, Dantzer F, McBurney MW, Murcia1 Gd, Sassone-Corsi P. Control of AIF-mediated cell death by the functional interplay of SIRT1 and PARP-1 in response to DNA damage. Cell cycle 2006; 5(8):873-7. doi: 10.4161/cc.5.8.2690 [Crossref]
  215. Jakus PB, Kalman N, Antus C, Radnai B, Tucsek Z, Gallyas Jr F. TRAF6 is functional in inhibition of TLR4-mediated NF-κB activation by resveratrol. J Nutr Biochem 2013; 24(5):819-23. doi: 10.1016/j.jnutbio.2012.04.017 [Crossref]
  216. Pan W, Yu H, Huang S, Zhu P. Resveratrol protects against TNF-α-induced injury in human umbilical endothelial cells through promoting sirtuin-1-induced repression of NF-KB and p38 MAPK. PloS One 2016; 11(1):e0147034. doi: 10.1371/journal.pone.0147034 [Crossref]
  217. Lin S-C, Ho C-T, Chuo W-H, Li S, Wang TT, Lin C-C. Effective inhibition of MERS-CoV infection by resveratrol. BMC Infect Dis 2017; 17(1):144. doi: 10.1186/s12879-017-2253-8 [Crossref]
  218. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol 2014; 11(8):506-14. doi: 10.1038/nrgastro.2014.66 [Crossref]
  219. Ruemmele F, Bier D, Marteau P, Rechkemmer G, Bourdet-Sicard R, Walker W. Clinical evidence for immunomodulatory effects of probiotic bacteria. J Pediatr Gastroenterol Nutr 2009; 48(2):126-41. doi: 10.1097/MPG.0b013e31817d80ca [Crossref]
  220. Hosseinifard E-S, Morshedi M, Bavafa-Valenlia K, Saghafi-Asl M. The novel insight into anti-inflammatory and anxiolytic effects of psychobiotics in diabetic rats: possible link between gut microbiota and brain regions. Eur J Nutr 2019; 58(8):3361-75. doi: 10.1007/s00394-019-01924-7 [Crossref]
  221. Morshedi M, Valenlia KB, Hosseinifard ES, Shahabi P, Abbasi MM, Ghorbani M. Beneficial psychological effects of novel psychobiotics in diabetic rats: the interaction among the gut, blood and amygdala. J Nutr Biochem 2018; 57:145-52. doi: 10.1016/j.jnutbio.2018.03.022 [Crossref]
  222. Markowiak P, Śliżewska K. Effects of probiotics, prebiotics, and synbiotics on human health. Nutrients 2017; 9(9):1021. doi: 10.3390/nu9091021 [Crossref]
  223. Lehtoranta L, Pitkäranta A, Korpela R. Probiotics in respiratory virus infections. Eur J Clin Microbiol Infect Dis 2014; 33(8):1289-302. doi: 10.1007/s10096-014-2086-y [Crossref]
  224. Guarino A, Guandalini S, Vecchio AL. Probiotics for prevention and treatment of diarrhea. J Clin Gastroenterol 2015; 49 (Suppl 1):S37-45. doi: 10.1097/MCG.0000000000000349 [Crossref]
  225. Park M-K, Vu N, Kwon Y-M, Lee Y-T, Yoo S, Cho Y-H. Lactobacillus plantarum DK119 as a probiotic confers protection against influenza virus by modulating innate immunity. PloS One 2013; 8(10):e75368. doi: 10.1371/journal.pone.0075368 [Crossref]
  226. Chai W, Burwinkel M, Wang Z, Palissa C, Esch B, Twardziok S. Antiviral effects of a probiotic Enterococcus faecium strain against transmissible gastroenteritis coronavirus. Arch Virol 2013; 158(4):799-807. doi: 10.1007/s00705-012-1543-0 [Crossref]
  227. Freitas M, Cayuela C, Antoine JM, Piller F, Sapin C, Trugnan G. A heat labile soluble factor from Bacteroides thetaiotaomicron VPI‐5482 specifically increases the galactosylation pattern of HT29‐MTX cells. Cell Microbiol 2001; 3(5):289-300. doi: 10.1046/j.1462-5822.2001.00113.x [Crossref]
  228. Freitas M, Tavan E, Cayuela C, Diop L, Sapin C, Trugnan G. Host-pathogens cross-talk. Indigenous bacteria and probiotics also play the game. Biol Cell 2003; 95(8):503-6. doi: 10.1016/j.biolcel.2003.08.004 [Crossref]
  229. Clavel T, Haller D. Molecular interactions between bacteria, the epithelium, and the mucosal immune system in the intestinal tract: implications for chronic inflammation. Curr Issues Intest Microbiol 2007; 8(2):25-43.
  230. Colbère-Garapin F, Martin-Latil S, Blondel B, Mousson L, Pelletier I, Autret A. Prevention and treatment of enteric viral infections: possible benefits of probiotic bacteria. Microbes Infect 2007; 9(14-15):1623-31. doi: 10.1016/j.micinf.2007.09.016 [Crossref]
  231. Maldonado Galdeano C, Cazorla SI, Lemme Dumit JM, Vélez E, Perdigón G. Beneficial Effects of Probiotic Consumption on the Immune System. Ann Nutr Metab 2019; 74(2):115-24. doi: 10.1159/000496426 [Crossref]
  232. Kim TH, Lee HK. Differential roles of lung dendritic cell subsets against respiratory virus infection. Immune Netw 2014; 14(3):128-37. doi: 10.4110/in.2014.14.3.128 [Crossref]
  233. Maidens C, Childs C, Przemska A, Dayel IB, Yaqoob P. Modulation of vaccine response by concomitant probiotic administration. Br J Clin Pharmacol 2013; 75(3):663-70. doi: 10.1111/j.1365-2125.2012.04404.x [Crossref]
Submitted: 17 Oct 2020
Accepted: 03 Feb 2021
First published online: 19 May 2021
EndNote EndNote

(Enw Format - Win & Mac)

BibTeX BibTeX

(Bib Format - Win & Mac)

Bookends Bookends

(Ris Format - Mac only)

EasyBib EasyBib

(Ris Format - Win & Mac)

Medlars Medlars

(Txt Format - Win & Mac)

Mendeley Web Mendeley Web
Mendeley Mendeley

(Ris Format - Win & Mac)

Papers Papers

(Ris Format - Win & Mac)

ProCite ProCite

(Ris Format - Win & Mac)

Reference Manager Reference Manager

(Ris Format - Win only)

Refworks Refworks

(Refworks Format - Win & Mac)

Zotero Zotero

(Ris Format - FireFox Plugin)

Abstract View: 1912
PDF Download: 1278
Full Text View: 637