Next Article in Journal
Attenuation of Lipopolysaccharide-Induced Acute Lung Injury by Hispolon in Mice, Through Regulating the TLR4/PI3K/Akt/mTOR and Keap1/Nrf2/HO-1 Pathways, and Suppressing Oxidative Stress-Mediated ER Stress-Induced Apoptosis and Autophagy
Next Article in Special Issue
Effects of Daily Probiotics Supplementation on Anxiety Induced Physiological Parameters among Competitive Football Players
Previous Article in Journal
Human Milk Fatty Acid Composition of Allergic and Non-Allergic Mothers: The Ulm SPATZ Health Study
Previous Article in Special Issue
Lactobacilli Supplemented with Larch Arabinogalactan and Colostrum Stimulates an Immune Response towards Peripheral NK Activation and Gut Tolerance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Can Probiotics and Diet Promote Beneficial Immune Modulation and Purine Control in Coronavirus Infection?

by
Ana H. A. Morais
1,2,3,*,†,
Thais S. Passos
3,†,
Bruna L. L. Maciel
1,3,† and
Juliana K. da Silva-Maia
1,3,†
1
Nutrition Postgraduate Program, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal, RN 59078-970, Brazil
2
Biochemistry Postgraduate Program, Biosciences Center, Federal University of Rio Grande do Norte, Natal, RN 59078-970, Brazil
3
Department of Nutrition, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal, RN 59078-970, Brazil
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2020, 12(6), 1737; https://doi.org/10.3390/nu12061737
Submission received: 3 May 2020 / Revised: 30 May 2020 / Accepted: 5 June 2020 / Published: 10 June 2020

Abstract

:
Infection caused by the SARS-CoV-2 coronavirus worldwide has led the World Health Organization to declare a COVID-19 pandemic. Because there is no cure or treatment for this virus, it is emergingly urgent to find effective and validated methods to prevent and treat COVID-19 infection. In this context, alternatives related to nutritional therapy might help to control the infection. This narrative review proposes the importance and role of probiotics and diet as adjunct alternatives among the therapies available for the treatment of this new coronavirus. This review discusses the relationship between intestinal purine metabolism and the use of Lactobacillus gasseri and low-purine diets, particularly in individuals with hyperuricemia, as adjuvant nutritional therapies to improve the immune system and weaken viral replication, assisting in the treatment of COVID-19. These might be promising alternatives, in addition to many others that involve adequate intake of vitamins, minerals and bioactive compounds from food.

1. Introduction

COVID-19, the disease caused by the new coronavirus SARS-CoV-2, was officially declared a pandemic by the World Health Organization on 11 March 2020 [1]. Coronaviruses (CoVs) belong to the genus coronavirus and the family Coronaviridae. All CoVs are pleomorphic, single-stranded RNA viruses, positive sense, polyadenylated, containing crown shape peplomers with a size of 80–160 nM and 27–32 kb [2].
The initial analysis of the new coronavirus genome showed a typical genomic structure belonging to the β-coronavirus cluster, including SARS-CoV and MERS-CoV [3]. According to Chen et al. [3], CoV recombination rates are very high due to the constant development of RNA dependent DNA polymerase (RpDd) transcription errors, and this has been one of the main targets for the discovery of drugs [4]. These mutations facilitate recognition in the human cell receptors by SARS-CoV-2 [5]. As is the case with other infections, SARS-CoV-2 infection presents specific outcomes, and one of the facts contributing to these variations is SARS-CoV-2’s high mutation rate [6,7].
Coronaviruses (CoVs) are a group of viruses responsible for a broad spectrum of diseases in several species. Human coronaviruses cause various respiratory diseases, some like the common cold, and others more severe, such as pneumonia, bronchitis, severe acute respiratory syndrome, and Middle Eastern respiratory syndrome [7]. In this sense, the infection can range from asymptomatic to oligo symptomatic to a clinical manifestation of the disease, causing symptoms mainly in the respiratory tract, which might lead to intensive care. It might also affect the gastrointestinal, hepatic and neurological systems [6,7,8].
Calculations based on World Health Organization data from Wuhan in China indicate that 14% of infected cases are severe, 5% require intensive care and 4% die [9,10]. The Chinese population was the first affected by the unprecedented outbreak caused by the new coronavirus. However, despite rigorous global efforts to contain the virus through social isolation measures, COVID-19’s incidence keeps increasing, and, currently, affects hundreds of countries worldwide [11].
There is still much to investigate, especially regarding prevalence and mortality estimates, since the real dimension of present and future COVID-19 consequences is not yet well established [12,13]. Nevertheless, the association of COVID-19’s higher mortality with senescence, preexisting diseases, and availability of health care services is well-accepted [14,15,16], and could be different in distinct countries affected by this pandemic. Unfortunately, all determinants of SARS-CoV-2 infection evolution, either asymptomatic or to clinical disease, remain unclear. Due to this easy infection transmissibility, the scientific community has joined efforts to combat COVID-19.
Currently, there are no vaccines or treatments registered for COVID-19. However, several studies have been published which highlight the alternatives related to the general treatment for viral infections [17,18,19,20,21]. Zhang and Liu [22] have added nutritional interventions, showing the virus target and related functions. Among these nutrients are A, C, D, E, and B complex vitamins, especially B2, B3, and B6. The effects of these vitamins, although not consensual for vitamin E [23], are known and related to the improvement of the immune response to viral infections treatment, from in vitro, preclinical, and clinical studies [24,25,26,27,28].
Considering minerals, Zhang and Liu [22] discuss adequate consumption of zinc, selenium, and iron. These minerals can have effects not only on the symptoms related to coronaviruses, such as diarrhea and lower respiratory tract infection but also on COVID-19 itself, with a study demonstrating some mechanisms of action [29,30,31]. Intracellular high levels of Zn2+ can compromise the RNA replication of several viruses that interfere in viral polyprotein processing [29]. Te Velthuis et al. [29] have demonstrated that increased Zn2+ concentrations inhibited the replication of RNA-dependent RNA polymerase (RdRp) from SARS-CoV nsp12. Omega-3 polyunsaturated fatty acids were also emphasized [32]. Another nutritional intervention reported was the administration of prebiotics and probiotics, since imbalance in the intestinal microbiota was observed in patients infected with COVID-19 [33].
Recently, the European Society for Clinical Nutrition and Metabolism (ESPEN) published a document providing concise guidance for the nutritional management of COVID-19 with ten practical recommendations [34]. However, this guideline is directed to patients in intensive care units (ICU), with morbidities and older age. The American Society for Parenteral and Enteral Nutrition (ASPEN) has also published some recommendations regarding mainly hydration, caloric and protein intake for patients with COVID-19 requiring ICU [35]. Although these nutritional therapies are based on scientific evidence, the existing guidelines are still limited to critical patients with COVID-19 [36].
Thus, nutritional recommendations to prevent the evolution of new coronavirus infection and the nutritional status of non-hospitalized patients has not been appropriately considered in the treatment of COVID-19 at this time, as with other viral infectious diseases. Some studies have shown the importance of early nutritional supplementation for non-critical patients hospitalized with COVID-19, emphasizing the importance of a balanced diet, and the necessity of unprocessed and healthy food choices [37,38,39,40]. Nonetheless, measures of primary care to avoid the aggravation of COVID-19 are barely mentioned despite all the attention given to combatting this pandemic combat by United Nations agencies [41].
The interaction between nutrition and infections is well accepted and valued by generations of health-care professionals. Before the use of antibiotics, diet was an integral and essential part of infection management. This strategy needs to be rescued, because understanding of the immune response and nutrition has considerably expanded. However, few studies have assessed the impact of nutritional interventions in the management of infectious diseases, and most observations of the interaction between nutrition and infections come from epidemiological studies [42].
The global pattern of food consumption has been associated with increased inflammation and uncontrolled infectious processes related to lower immune system responsiveness [43,44,45,46,47]. Additionally, the impact of the consumption of ultra-processed foods has been related to imbalances in the human intestinal microbiota [48]. Dysbiosis is closely associated with changes in the dynamics of the immune system [49].
Probiotics can help maintain or restore the balance of the intestinal microbiome and, consequently, improve the immune system’s response against aggression to the human organism [50]. Among the mechanisms of probiotics that corroborate with intestinal and systemic health is the competition with pathogenic microorganisms for adhesion sites and nutritional sources, the secretion of antimicrobial peptides, the action of metabolites, nucleosidase activity, and even immunomodulation by signaling pathways of intestinal and immune cells [51,52,53,54,55,56].
Besides the mechanisms well explored for probiotic action [51,52,53,54,55,56], a metabolic pathway has recently aroused the interest of researchers. This route involves the intestinal purine metabolism, possibly one of the explanations for the benefits achieved with the use of probiotics [57,58], to improve the immune system in cases of viral infections. Studies also show the association between high serum urates and the severity of HIV infection [59], which can help understand how the consumption of purine-rich foods could influence worsening clinical conditions in viral infections, such as those caused by a coronavirus. Thus, curiosity and interest in nutritional therapies to promote the reduction of purine intake have increased, whether using probiotics [33,52,53,57,58] or by dietary restriction advice on source foods [60], and consequently control of serum uric acid concentrations [61]. This behavior can positively influence the health of individuals with viral infections.
Future research may specifically elucidate the effectiveness of adjuvants as part of COVID-19 treatment. This review adds the scientific data available so far, demonstrating the role of the microbiota in the immune response and the application of probiotics in coronavirus infections. Lactobacilli are lactic acid bacteria with probiotic potential. Several health benefits have been attributed to a specific strain of lactobacilli, Lactobacillus gasseri [62]. Studies have shown the action of L. gasseri in different viral infections, including respiratory [63,64]. We also review the importance of other nutritional interventions related to the consumption of food sources of purine. The findings may contribute to combating COVID-19 during this global health emergency.

2. Probiotics and Immunity

Researches have shown that microorganisms with the ability to modulate the intestinal and systemic immune response could be used in bacterial and viral respiratory infections to improve their outcomes [64,65,66,67,68,69].
The gastrointestinal tract (GT) presents a microbiome inhabited by a mass of active bacteria that are important for the maturation of immune cells, affecting human health status [51,70]. These bacteria belong mainly to three phyla: Bacteriodetes, Firmicutes, and Proteobacteria. However, the number and proportion of microorganisms differ according to the GT portion and individual characteristics [71,72].
An important factor that affects both gut microbiota and the immune system is diet. Data have shown that human gut microbiota in industrialized societies is very distinct from the recent ancestral microbiota of humans [48]. Ultra-processed foods modify the gut environment, being trigger factors for low-grade systemic inflammation and oxidative stress [73]. Recent work proposed the term “microbiota insufficiency syndrome”, and this could be linked to many non-communicable chronic diseases [74].
An unbalanced state of the microbiome, called dysbiosis, is characterized by overgrowth of pathobionts, loss of commensals, and lower diversity. Once established, it can disturb the local mucosal and systemic immune cells [49]. Given the association of dysbiosis with the etiology of several diseases, approaches, such as administration of probiotics that control pathogenic microorganism growth and modulate immune response may promote intestinal and systemic health [49,62].
Lactobacillus and Bifidobacteria are the most common microorganisms used as probiotics, but yeast Saccharomyces boulardii and Bacillus species are also widely applied [72]. Probiotic bacteria are mostly members of the gut microbiome and can be incorporated into food or administrated in isolated form. However, probiotic microorganisms are strain-specific, and not all bacteria have this property [51]. Firstly, they have to survive in acid conditions of the stomach to exhibit beneficial health impacts [75]. Then these microorganism strains, such as Lactobacillus, can maintain the ecological balance of the host intestinal microbiota by reinforcing intestinal flora and inhibiting harmful bacteria [70]. Several mechanisms of action can also affect the immune system. Improvements in immune response is a strategy to combat infections, bacterial, and viral infection [50,66,67,68].
Probiotic supplements with lactobacilli and bifidobacterial strains are well-established as part of the treatment of infectious diarrhea caused by rotavirus. Azagra-Boronat and Massot-Cladera [76] showed that probiotics decreased the severity and incidence of diarrhea in the preclinical assay. Furthermore, probiotics and their metabolites have been investigated as adjuvants in immunotherapy for viral hepatitis cirrhosis [77].
Secondary bacterial pneumonia is a significant complication during epidemic and pandemic viral respiratory infections that can increase morbidity and mortality. Virus infection promotes bacterial attachment and colonization, disruption of epithelial barriers, and alteration of the innate immune response in the respiratory tract. A study showed that peptidoglycan from immunobiotic L. rhamnosus CRL1505 improved respiratory antiviral innate immune response, and decreased bacterial transmigration across the lung and pulmonary inflammatory damage in infant mice [65].
A meta-analysis published by Cochrane [69], and other works, demonstrated the efficacy of probiotics in reducing the incidence and duration of acute respiratory tract infections of viral origin and the need for several antibiotic courses [78,79]. A study reported that generalized probiotic intake in the US population for 2017–2018 would have saved 373 million USD in health care for flu-like acute respiratory tract infection [80]. It is essential to avoid the overuse of antibiotics, which can lead to the development of antibiotic-resistant pathogens and alter the composition and functions of the microbiota, causing long-lasting harmful effects for the host [81]. Reasonable care should be applied in antibiotic drug utilization in upper respiratory tract acute infections, assessing severity, clinical signals, and complication risks. In this sense, probiotics could be an adjuvant in respiratory infectious disease treatment, optimizing antibiotic administration [69].

2.1. Probiotic Mechanisms of Action

Although the most widely discussed mechanism of probiotic action is the inhibition of the growth of pathogenic bacteria [51], other mechanisms explain the antagonistic effects of probiotics, among them adhesion and co-aggregation ability, competition for binding sites and nutritional sources, secretion of antimicrobial substances, enhancement of intestinal barrier function by regulation of tight junctions and mucins expression, along with immunomodulation by interaction to receptors of microorganism-associated molecular patterns [51,75,82,83,84].
The intestinal barrier is composed of a mucus layer, intestinal epithelium, and underlying lamina propria. The mucus layer is a physical barrier to avoid direct contact with the intestinal epithelium. Several specialized cell types are present in the intestinal epithelium: enterocytes responsible for absorption, Paneth cells specialized in synthesizing and secreting antimicrobial peptides, and mucus-secreting Goblet and entero-endocrine cells. The mucosal permeability is under the control of the tight junction proteins. The lamina propria consists of dendritic cells, macrophages, plasma cells, and B and T lymphocytes [50,85].
The effectivity of probiotic bacteria depends on their adhesion capacity to the host, to other bacterial cells (co-aggregation), or even to the same species (auto-aggregation). In this way, they can colonize and promote immunomodulatory effects, besides stimulating gut barrier and metabolic functions [82].
Short-chain fatty acids (SCFA) (butyrate, propionate, and acetate) are well-known products from microbiota fermentation with an immunoregulatory function due to the influence in the extracellular and intracellular signaling molecules. SCFA can bind to cell surface G protein-coupled receptors and modulate the immune function indirectly. These compounds can inhibit histone deacetylases, the cell interior, and regulate gene transcription in the immune response. Besides, they can promote B-cell differentiation and antibody synthesis, improving the antibody-antigen response [86]. Other metabolites from microbiota metabolism are essential amino acids, such as tryptophan, which are closely related to the immune system. Tryptophan metabolites attenuate TNF-α-induced activation of NF-κB and reduce expression of the proinflammatory chemokine, besides strengthening the intestinal epithelial barrier by fortifying tight junctions. The intestinal microbiota can also synthesize vitamins from complex B, and vitamins play a vital role in regulating the immune system [72].
Microbiota sensing, by epithelial cells, dendritic cells (DCs) and macrophages, is regulated by pattern recognition receptors (PRRs), which interact with microorganism-associated molecular patterns (MAMPs) driving the cascade of the immune response. Therefore, probiotics can modulate the immune system through the binding of their MAMPS (lipoteichoic acids, peptidoglycan, S-layer proteins, and nucleic acids) with PPRs (toll-like receptors, NOD-like receptors, C-type lectin receptors) expressed in the host intestinal mucosa [62,87]. Interestingly, variations in MAMP profiles have been attributed to differential immunomodulatory capacities of probiotic strains [87]. Human myeloid DC treated with Lactobacillus (either L. gasseri, L. johnsonii and L. reuteri) showed a three-fold induction of TLR-2 transcripts, and induced T cells toward T helper 1 polarization [88].
The response against food and commensal antigens is regulated by T lymphocytes as well as the immune response to combat pathogenic microorganisms [51]. Probiotic functions involve the action on T helper (Th) and regulatory T (TReg) cells in the lamina propria [87]. Depending on the stimulus, naive T cells can be differentiated toward a Th1, Th2, Th17, or Treg response according to the profile of secreted cytokines [89]. Probiotic bacteria can influence mucosal immune responses and T cell differentiation by the induction of different cytokines secretion [51]. This secretion is associated with the equilibrium in DC stimulation and tolerance promoted by the presentation to Lactobacillus cells that, consequently, is reflected in the balance of the Th1/Th2 responses, besides being important to maintain homeostasis in symbiotic bacteria [90].
The anti-inflammatory effect of a probiotic formulation (L. rhamnosus, B. lactis, and B. longum) was demonstrated. There was a significant increase in anti-inflammatory IL-10 production and decrease of proinflammatory cytokines IL-1β and IL-6 in the human macrophage cell line THP1 [91]. Moreover, treatment with probiotics shifted gut microbiota composition, increasing the ratio of Prevotella and Oscillibacter in mice. These bacteria produce anti-inflammatory metabolites and, consequently, decrease Th17 polarization, favoring the differentiation of anti-inflammatory Treg/Type 1 regulatory T cell in the gut of an experimental model [92]. Lactobacilli, including L. gasseri, activate human dendritic cells and lead toward a Type 1 T-helper polarization, as evidenced by higher secretion of IFN-γ and IL12. These cytokines act by switching naive or memory T cells towards Th1 responses, promoting immunity against infections and other diseases [88].
Probiotic bacteria can be adjuvants in vaccine outcomes, acting in T cell responses toward Th1 [88], enhancing B cell and antibody response, besides their protection against infections directly at gut mucosal level and through interactions with the innate immune system [93,94]. Lactic acid bacteria have been assessed as non-parenteral live mucosal vaccine vectors [95,96]. In addition, a clinical trial with 50 volunteers evidenced that oral intake of L. fermentum CECT5716 enhances the effects of influenza vaccination, increasing T-helper type 1 and the vaccine-specific IgA antibodies post-vaccination [97].
Immunoglobulin A (IgA) is part of the humoral response; this antibody is fundamental to adaptive immune response and is necessary to mucosal barrier function. IgA can bind to commensal and pathogen bacteria and toxins, blocking them, which can optimize the DC action and overall immune response. It has been demonstrated that probiotics can induce IgA-secreton, collaborating in maintaining immune surveillance [51,98]. Therefore, probiotics induce protective humoral and cellular immunity.
Probiotics also regulate the expression of multiple intestinal genes. Specific probiotics regulate the expression of tight junction proteins, collaborating to preserve the integrity of the GT epithelium. The Lactobacillus strains were protective against Escherichia coli infection. The epithelial cells treated with the probiotic maintained higher levels of ZO-1 and other tight junction protein expression than those infected with the pathogen alone [84,99,100].
Probiotic bacterial strains (Lactobacilli, Bifidobacteria, and Streptococci) have also been shown to regulate mucins expression, improving the properties of the mucus layer and indirectly supporting the gut immune system [84]. The preserved mucus layer avoids the adherence of pathogenic microorganisms. Probiotics can also regulate gene expression of enterocytes and dendritic cells [72]. Another mechanism of action is related to the stimuli of secretion of antimicrobial peptides (bacteriocin) by gut microbiota, protecting the host against infections. Bacteriocin defends the organism against many pathogenic bacteria, such as Staphylococcus aureus, Escherichia coli, Listeria monocytogenes, and Salmonella [50]. These antimicrobial peptides are released by Paneth cells in response to enteropathogenic bacteria stimulation, contributing to the innate immune response of the GT. These peptides, such as lactocillin and other thiopeptides, have been studied for the production of drug-like molecules to clinical application as antibiotics [101].
In addition to the actions mentioned on immunity, Lactobacillus can influence the nucleotide metabolites in the intestine. L. fermentum ONRIC b0185, L. fermentum ONRIC b0195 e L. pentosus ONRIC b0223, presented in cell, culture high nucleosidase activity converting nucleoside into purine bases [54]. The Lactobacillus DM9218 strain also acts on the purine nucleosides (inosine and guanosine) [55,56]. L. gasseri PA3 has demonstrated an ability to reduce purine in foods and beverages [57,58,61]. Purines are essential to viral RNA synthesis. Reducing purine availability might slow down virus replication, holding down viral infections. Thus, reducing purine availability may be an important pathway to be explored in the use of probiotics during viral infections, besides their immune-modulatory actions.
Concerning the effect on respiratory infections, lactobacilli have been associated with beneficial action in other viral infections. Recently, the anti-HIV effect of lactobacilli was investigated in human cervicovaginal and tonsillar tissues ex vivo, and cell lines were infected with HIV-1. Extracellular vesicles released by L. crispatus BC3 and L. gasseri BC12 protected from HIV-1 infection, a result attributed to the presence of protein and metabolites in these vesicles [102].
Thus, probiotics can help maintain a balance between inflammatory responses to pathogens and natural intestinal homeostasis and function. Promotion of homeostasis in the gut microbiome by probiotics could benefit the immune system as a whole. Consequently, the immune response to infections can be favored, and this may apply to SARS-CoV-2. Specific roles of probiotics in the immune response against viral infections will be discussed, and L. gasseri has been specifically addressed for its effects on immunity and purine control. L. gasseri’s effect on purine metabolism might reduce viral replication, and this should be explored for Sars-CoV-2 infection as discussed below.

2.2. Lactobacillus gasseri

L. gasseri is one of six species in the L. acidophilus complex. L. gasseri has been described as a multifunctional probiotic and one of the dominant species in the human intestine [62]. These microorganisms can be isolated from human feces, breast milk, urogenital tract, and oral cavity [64,103,104]. Several health benefits were attributed to this probiotic through antimicrobial activity and immunomodulation [62].
Recently, lactobacilli were isolated from human feces, and thirteen were identified as L. gasseri by 16 rDNA sequencing, showing the genetic diversity and different probiotic characteristics [105]. The resistance of L. gasseri to the human gastrointestinal tract under difficult conditions is a property that allows this microorganism to colonize the gut, improving the microbiome [103,106]. A recent study showed that L. gasseri PA3 modulated the diversity of the microbiota after in vitro fermentation, increased the relative abundance of Lactobacillus and Escherichia, and decreased Bacteroides and Phascolarctobacterium [70].
Dendritic cells incubated with L. gasseri and bacteria produced more IL-10 and there was a higher level of natural killer cells in the spleen of mice treated with this probiotic [107]. In humans, a combination of L. gasseri CECT5714 and L. coryniformis CECT5711 stimulates NK cells and IgA concentration [108].
A study with L. gasseri 4M13 isolated from infant feces showed higher inhibition of nitric oxide production than other strains and reduced inflammatory mediators (TNF-α, IL-6, IL-1β) in LPS-stimulated macrophages. These results suggest L. gasseri’s potential to modulate proinflammatory cytokines [109].
Regarding its role in viral infections, L. gasseri strains have been intensively studied for the past few years. Human respiratory syncytial virus, an enveloped negative-sense RNA virus, is potentially at high risk of becoming severely symptomatic in elderly and immunocompromised individuals. The oral administration of L. gasseri SBT2055 in mice reduced the virus titer and attenuated the proinflammatory cytokines in the lung, and interferon and interferon-stimulated genes were upregulated [64].
The evolution of other viral respiratory infections was prevented through L. gasseri SBT2055 treatment, including Influenza A and human respiratory syncytial virus infections [63,64]. There was lower virus titer in the lungs, inflammatory cell infiltration, and IL-6 production in mice with Influenza A treated with L. gasseri. The probiotic enhanced the host defense system before the virus infection, increasing the gene expression of Mx1 and Oas1a, critical for the antiviral responses by IFN activation and antiviral immunity, and for enhancement of IgA production [63]. A human trial showed that the probiotic L. gasseri SBT2055 boosted the immune responses in healthy vaccinated subjects that received a trivalent influenza vaccine. This Lactobacillus strain stimulated humoral immunity, and the total IgG and IgA levels in plasma and sIgA production in saliva were also higher in the probiotic-treated group [110].
Other work has demonstrated that oral administration of L. gasseri TMC0356 stimulated local and systemic immune responses protecting against influenza virus infection. Clinical symptom scores were improved, pulmonary virus titers decreased, and mRNA expression of interleukin (IL)-12, IL-15, and IL-21 in Peyer’s patch and the pulmonary mRNA expression of IFN-γ, TNF, IL-12a, IL-12rbl, IL-2rb and perforin-1 increased significantly [111]. Therefore, L. gasseri promoted the down-regulation of viral replication in respiratory infections through the induction of antiviral gene expression [63,64,111].
A study demonstrated that L. gasseri SBT2055 induces transforming growth factor-β (TGF-β) expression in dendritic cells, activates Toll-like receptor-2 (TLR2) signal to produce IgA, and increases the rate of IgA in Peyer’s patch and lamina propria of the mouse small intestine. This probiotic also upregulated the gene expression of retinaldehyde dehydrogenase 2 (RALDH2), an enzyme responsible for the conversion of retinol into retinoic acid. The retinoic acids derived from the dendritic cell can promote IgA secretion via a retinoic acid-dependent mechanism that induces the expression of gut-homing receptors on B cells. Therefore, L. gasseri could enhance host immune responses and protect against infections [112]. Furthermore, L. gasseri OLL2809 promoted an important modulation of several proteins secreted from immature dendritic cells, reinforcing the concept of a protective anti-inflammatory role attributed to this probiotic strain. L. gasseri OLL2809 not only led to modulation in classic immune mediators, but also influenced proteins involved in contractile and desmosome machinery, presenting a new mechanism by which it can modulate the immune response by dendritic cells [113].
Also related to anti-inflammatory activity, another study with distinct Lactobacillus strains found that only L. gasseri Kx110A1 mitigated the action of macrophages stimulated with lipoteichoic acid (LTA) and lipopolysaccharide (LPS), inhibiting the release of the proinflammatory cytokines TNF- alpha and IL-6 through a lower expression of the metalloptroteinase-17 (ADAM17) enzyme (TNF-alpha-converting enzyme). These findings show a new mechanism of L. gasseri in blocking proinflammatory cytokine production [114].
Therefore, L. gasseri can boost both the innate and adaptive immune responses. The mechanisms involved are a stimulus to the production of cytokines from dendritic cells and macrophages and IgA production from B cells, which are associated with Toll-like receptors (TLR)-2 signaling [112,115].
Furthermore, the promotion of homeostasis in the gut microbiome by probiotics could benefit the immune system as a whole. Data illustrate L. gasseri’s potential to contribute to the mucosal gut barrier, microbiome modulation, and human health in general [62]. A clinical study showed that L. gasseri CP2305 administration mitigated the decline of Bifidobacterium ssp. and the increase of Streptococcus ssp. promoted by stress in young adults, favoring a balanced microbiome [116]. This Lactobacillus has also promoted regulatory effects in the gut environment and function according to Sugawara et al. [117]. In this work, individuals treated with L. gasseri CP2305 showed higher Bifidobacterium content, lower Clostridium cluster IV content in feces samples, and fecal p-cresol, a putrefactive product, was significantly decreased in the CP2305 group. The reduced p-cresol in fecal content can suggest that L. gasseri altered the tyrosine metabolism and maintained a healthy intestinal environment, avoiding potentially toxic protein degradation products [117]. An experimental study in chickens has shown that probiotic interventions with Lactobacillus species combined with L. gasseri protected against influenza virus infection through microbiota modulation [118].
The hypothesis that L. gasseri can be potentially beneficial in viral infections has been supported by several studies, including clinical trials [64,110,111,115]. The data are not explicitly related to coronavirus infection. However, the research included viral infections such as influenza, a respiratory viral infection. Therefore, this information might suggest the actions of this lactobacillus strain in COVID-19, improving the innate and adaptive immune systems, and further studies should address this hypothesis.
L. gasseri PA3 can reduce purines, nucleosides, nucleotides and nucleic acids [57,58], including inosine 5′-monophosphate (IMPDH2), inosine, hypoxanthine, guanosine 5′-monophosphate (GMP), and guanosine in rats [52,53]. These effects are important, as recently Gordon et al. [119] cloned, tagged and expressed 26 out of 29 SARS-CoV-2 proteins in human cells, to identify, by affinity purification coupled with mass spectrometry (AP-MS), human proteins that can interact with SARS-CoV-2 proteins. The study showed the host factors that mediate viral infection, helping to elucidate the effective molecular targets for therapies to combat SARS-CoV-2 infection. In the study, purine biosynthesis enzyme IMPDH2, which can be reduced by L. gasseri, could interact with the viral protein nsp14. L. gasseri PA-3 reduces the absorption of the main purines contained in food, and this property has been explored as adjuvant therapy in gout and hyperuricemia [61].
Several studies have revealed and detailed the structure and functions of SARS-CoV-2 polymerases, enabling the understanding and development of potential inhibitors [4,120]. These RNA viruses require RNA-dependent DNA polymerase (RdRPs) for various stages in their life cycle. For this reason, RdRPs are potential targets for drugs and other therapeutic interventions for treating diseases caused by this group of viruses [4,5]. RdRP is the central catalytic subunit in RNA synthesis machinery for CoVs. An RdRP domain catalyzes the replication and transcription of the ~ 30 kb CoV RNA genome in the C-terminal part of nsp12, one of 16 replicase subunits [2,121]. Therefore, this allows the recognition of alternative substrates blocking purine availability for viral polymerase, resulting in errors during viral RNA synthesis [17,122]. Given the high similarity of amino acids that constitute the RdRps of SARR-CoV and SARS-CoV-2, studies have shown nucleotide analogs that can potentially inhibit SARS-CoV-2 polymerase.
According to Maxmen [4], some of these broad-spectrum antiviral agents, with proven efficacy, have been used when considering their viral inhibiting activities. Most of these purine nucleoside analogs block the replication of various RNA and DNA viruses by inhibiting inosine monophosphate dehydrogenase (IMPDH), an essential enzyme involved in guanosine triphosphate (GTP) biosynthesis. Others also inhibit viral RdRP activity, acting like a zinc ionophone, blocking mRNA capping and inducing mutations in RNA-dependent viral replication [4,17,123]. However, clinical application is limited due to serious side effects associated with high doses [124,125,126].
The possibility of using several antiviral drugs in the treatment of COVID-19 emerges, such as Favipiravir (C5H4FN3O2) [122], Alovudine (C10H13FN2O4) [17], Remdesivir (C27H35N6O8P) [127]; Azidotimidine (C10H13N5O4) [17], Atazanavir (C38H52N6O7) [128], Ivermectin (C48H74O14) [129] and Chloroquine (C18H26ClN3) [21,130]. These agents are known to be effective against other RNA viruses, including Influenza, HIV, Ebola, MERS-CoV, SARS-CoV, and SARS-CoV 2 in in vitro, experimental and clinical studies. Interestingly, some of these drugs influence the purine pathways [17,122,127].
Ahn et al. [131] proposed that SARS-CoV nsp12 can use both primer-dependent and primer-independent RNA synthesis activities, concluding that purine nucleotides are preferably utilized by nsp12 to restart RNA synthesis. Furthermore, the poly (A) tail at the 3ʹ end of the viral genome plays a regulatory role in the initiation during the minus-strand RNA synthesis. Finally, nsp12 needs a small region with less than 40 nucleotides at the ends of the viral genome and its complementary minus-strand RNA to initiate RNA synthesis. These results guide further modifications of these purine nucleoside analogs to generate broader and more potent anti-coronavirus agents.

3. The Role of Low-Purine Diets

Biological purines are small molecules found inside and outside cells. They consist of organic, aromatic, and heterocyclic compounds formed by a pyrimidine ring attached to an imidazole ring [132]. Purine bases are present in all plant and animal cells, are essential for life, as components of cellular energy systems (ATP and NAD), signaling (GTP, cAMP, and cGMP), and are associated with pyrimidines form RNA and DNA (adenine and guanine) [133]. There are two synthesis pathways for purine nucleotides: De Novo synthesis, and the salvage pathway. The first is the main synthesis pathway and the second is essential for brain and bone marrow [134].
Foods of animal and vegetable origin contain different amounts of purines, that when degraded generate uric acid, of which approximately 80% of excretion occurs through the kidneys [135]. Uric acid is considered an antioxidant due to the ability of urates to scavenge free radicals. However, the consumption of diets rich in purine can cause the overproduction of uric acid, which has low solubility in biological fluids, leading to the accumulation of serum urate in the blood above the saturation level (hyperuricemia). This clinical condition is responsible for causing health problems such as gout and is a risk factor for cardiovascular, renal, and metabolic syndrome [134,135,136].
Additionally, the main disorder related to purine metabolism is characterized by a deficiency of the hypoxanthine-guanine enzyme phosphoribosyl transferase (HPRT). This clinical condition is associated with the overproduction of uric acid, which can impair the hematological, immune, renal, and neurological systems. Thus, the genes associated with purine metabolism must be related to the metabolism of drugs [137].
When infected by viruses, human cells increase the demand for purine nucleotides, which are necessary for the synthesis of RNA and viral DNA. As a result, viruses obtain purine nucleotides through the action of the enzyme Inosine Monophosphate Dehydrogenase (IMPDH), which is essential for viral cell growth and differentiation. Therefore, IMPDH also becomes a target for antiviral pharmacological treatment [4,132].
According to studies, patients with HIV, for example, have an incidence of hyperuricemia higher than in the population of healthy individuals. This increase occurs because uncontrolled virus replication increases serum urate concentrations in response to increased cell turnover [59]. Carlucci et al. [138] concluded that the HIV-1 virus strongly influences the metabolism of the purine nucleotides in host cells. Considering these data documented for HIV, it is likely that the same preference for purine nucleotides may occur during the growth and differentiation of SARS-COV-2 cells.
Thus, COVID-19 may be worsened in patients who ingest foods with high purine content, promoting an increase in uric acid concentrations. Probably, people with hyperuricemia or gout can be considered a risk group in cases of COVID-19. This proposition is reinforced by Ahn et al. [131], who suggested that coronaviruses use purine nucleotides to promote RNA synthesis. Lewandowski and Hsieh [139] mentioned rheumatologists’ concern about the increased risk of infection and death among patients with rheumatic disease, due to the increasing number of pre-admissions of COVID-19 patients with rheumatic disease. Due to the global urgency to answer this clinical question, the international rheumatology community created the COVID-19 Global Rheumatology Alliance to allow healthcare professionals from around the world to exchange information on case reports of patients with rheumatic disease diagnosed with COVID-19 to assist healthcare providers [140]. Currently, an anti-inflammatory widely used to treat patients with gout, colchicine [141], is undergoing clinical tests for the treatment of COVID-19. In addition to other drugs commonly prescribed by rheumatologists, hydroxychloroquine, glucocorticoids, intravenous immunoglobulin, anti-interleukin (IL) -1 and anti-IL-6 therapies, and Janus kinase inhibitors have also been tested [140].
Nutritional therapy is a promising adjunct to drug therapy, helping to reduce the intake of purines and control the concentrations of uric acid in the blood. This strategy could improve the clinical condition of patients with hyperuricemia resulting from viral infections, such as in AIDS [59], and probably COVID-19. To reduce purines from the diet, it is of fundamental importance to reduce the consumption of food sources containing a high concentration of these organic compounds, such as alcoholic beverages, sweetbreads, anchovies, sardines, liver, kidneys, brains, herring, mackerel and scallops [60,142]. Controlled consumption of sources with moderate concentrations of purine, for example, beef, pork, poultry, fish and seafood, asparagus, cauliflower, spinach, mushrooms, peas, lentils, dried peas, beans, oats, bran wheat, and wheat germ could also be beneficial [132].
According to the review study by Wu et al. [60], the purine content in food can change due to the type of processing, cooking, and storage. Based on the literature, the authors stated that lower storage temperatures could reduce enzyme activity, slowing down the nucleotide degradation pathways, affecting the content of purine bases. Besides, during industrial processing, food washing processes can reduce the content of hypoxanthine and adenine by around 60%, due to the higher solubility of these molecules [60]. This information can help individuals with hyperuricemia diagnosis to understand that some technological processes also help to reduce the purine content present in food.
On the other hand, modifying eating habits is a great challenge. It is also observed in cases of hyperuricemia; some research has shown innovative alternatives, which consist of developing foods and beverages with low purine content, using enzymatic degradation [143,144]. In this context, Purine Nucleoside Phosphorylase (PNP) is a crucial enzyme related to the purine degradation process, because it catalyzes the reversible phosphorolysis of purine nucleosides to the respective bases and sugars (ribose or deoxyribose) 1-phosphate. PNP acts on the salvage pathway allowing cells to reuse purine bases for nucleotide synthesis [145]. Therefore, this enzyme regulates nucleoside concentrations within living cells and is, therefore, the target of several types of pharmacological treatments, such as gene therapy for inherited immune deficiencies and solid tumors [146]. Although this idea is promising to help patients with hyperuricemia, Jankowska et al. [135] reinforced the idea that to be feasible it is necessary to overcome the technological challenge, which consists of producing enzymes in a way that preserves activity in the food and allows storage for long periods.
The use of probiotics as an alternative therapy to the use of drugs in order to treat hyperuricemia was discussed in the review by Prasad et al. [147]. They pointed out that, because pharmacological treatments for hyperuricemia have several side effects, researches have sought therapeutic alternatives less aggressive to the human body and effective in promoting the reduction of uric acid levels. In this context, research has involved the manipulation of the intestinal microbiome using probiotics. Therefore, probiotics can influence the metabolism of purines, breaking down the products inosine and guanosine to directly influence the reduction of uric acid levels. Also, Li et al. [56] investigated a strain of probiotics isolated from Chinese sauerkraut (DM9218-A) intra-gastrically administered in a rat model with diet-induced hyperuricemia. They observed that serum uric acid levels in animals were significantly reduced, suggesting that DM9218-A may be a promising candidate as an adjunctive treatment in patients with hyperuricemia during the initial period, besides its potential as probiotic acting in the prevention of hyperuricemia in the population. So, the use of probiotics is an interesting strategy, in cases of viral infections such as SARS-CoV-2, to reduce the purine nucleotides, and hinder the synthesis of RNA and viral DNA.
Thus, this study highlights that one of the promising nutritional strategies to be included for SARS-COV-2 infection is the ingestion of probiotics, especially L. gasseri, which can have beneficial effects on the host’s health, activating the immune response in cases of infection. Besides, studies demonstrated that L. gasseri PA-3 promoted the absorption and incorporation of inosine and compounds related to purine metabolism in vitro and reduced intestinal purine absorption in clinical studies [52,53,57,58,61].
Studies on diverse aspects of COVID-19 are emerging now following the disease spread, such as a paper related to the treatment protocol established in China. The researchers have found that some patients with COVID-19 showed dysbiosis characterized by a lower level of Lactobacillus and Bifidobacterium. Thus, to regulate intestinal microbiota, the medical team included prebiotics or probiotics for these patients to reduce the risk of secondary infection due to bacterial translocation [33]. Future studies will elucidate the mechanisms and applications of probiotics in COVID-19 treatment. Nonetheless, there is already scientific data to support the action of probiotics enhancing the immune response in infection cases such as viral infections. So far, adjuvant therapies cannot substitute treatment with specific medication. However, the incorporation of adjuvant therapeutic factors in food could be a means of providing preventive medicine or better treatment.

Author Contributions

Literature review: A.H.A.M., T.S.P. and J.K.d.S.-M; original draft preparation: A.H.A.M., T.S.P. and J.K.d.S.-M; writing, critically review and editing: A.H.A.M., T.S.P., B.L.L.M., and J.K.d.S.-M.; supervision: A.H.A.M. All authors have read and agreed to the published version of the manuscript.

Funding

This study was financed in part by the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior—Brasil (CAPES)—Finance Code 001.

Acknowledgments

The authors thank the Federal University of Rio Grande do Norte (UFRN), especially the Pro-Rectory of Postgraduate and the Pro-Rectory of Research, for all efforts dedicated to supporting the research in our institution.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. Virtual press conference on COVID-19—11 March 2020. Available online: https://www.who.int/docs/default-source/coronaviruse/transcripts/who-audio-emergencies-coronavirus-press-conference-full-and-final-11mar2020.pdf?sfvrsn=cb432bb3_2 (accessed on 30 March 2020).
  2. Banerjee, A.; Kulcsar, K.; Misra, V.; Frieman, M.; Mossman, K. Bats and coronaviruses. Viruses 2019, 11, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Chen, Y.; Liu, Q.; Guo, D. Emerging coronaviruses: Genome structure, replication, and pathogenesis. J. Med. Virol. 2020, 92, 418–423. [Google Scholar] [CrossRef] [PubMed]
  4. Maxmen, A. More than 80 clinical trials launch to test coronavirus treatments. Nature 2020, 578, 347–348. [Google Scholar] [CrossRef] [Green Version]
  5. Shang, J.; Ye, G.; Shi, K.; Wan, Y.; Luo, C.; Aihara, H.; Geng, Q.; Auerbach, A.; Li, F. Structural basis of receptor recognition by SARS-CoV-2. Nature 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Zumla, A.; Hui, D.S.; Perlman, S. Middle East respiratory syndrome. Lancet 2015, 386, 995–1007. [Google Scholar] [CrossRef] [Green Version]
  7. Cascella, M.; Rajnik, M.; Cuomo, A.; Dulebohn, S.C.; Napoli, R.D. Evaluation and Treatment Coronavirus (COVID-19) [Updated 2020 Mar 20]. In StatPearls; StatPearls: Petersburg, FL, USA, 2020. [Google Scholar]
  8. Pan, L.; Mu, M.; Mu, M.; Sun, Y.; Wang, R.; Yan, J.; Li, P.; Hu, B.; Wang, J.; Hu, C.; et al. Clinical characteristics of COVID-19 patients with digestive symptoms in Hubei, China: A descriptive, cross-sectional, multicenter study. Am. J. Gastroenterol. 2020. [Google Scholar] [CrossRef]
  9. Grech, V. Unknown unknowns—COVID-19 and potential global mortality. Early Hum. Dev. 2020, 144, 105026. [Google Scholar] [CrossRef]
  10. Centers for Disease Control and Prevention. Available online: https://www.cdc.gov/ (accessed on 30 April 2020).
  11. Sohrabi, C.; Alsafi, Z.; O’Neill, N.; Khan, M.; Kerwan, A.; Al-Jabir, A.; Iosifidis, C.; Agha, R. World Health Organization declares global emergency: A review of the 2019 novel coronavirus (COVID-19). Int. J. Surg. 2020, 76, 71–76. [Google Scholar] [CrossRef] [PubMed]
  12. Baud, D.; Qi, X.; Nielsen-Saines, K.; Musso, D.; Pomar, L.; Favre, G. Real estimates of mortality following COVID-19 infection. Lancet Infect. Dis. 2020. [Google Scholar] [CrossRef] [Green Version]
  13. Adams, J.G.; Walls, R.M. Supporting the health care workforce during the COVID-19 global epidemic. JAMA 2020, 323, 1439–1440. [Google Scholar] [CrossRef] [Green Version]
  14. Ji, Y.; Ma, Z.; Peppelenbosch, M.P.; Pan, Q. Potential association between COVID-19 mortality and health-care resource availability. Lancet Glob. Health 2020, 8, e480. [Google Scholar] [CrossRef] [Green Version]
  15. Wang, B.; Li, R.; Lu, Z.; Huang, Y. Does comorbidity increase the risk of patients with COVID-19: Evidence from meta-analysis. Aging 2020, 12, 6049–6057. [Google Scholar] [CrossRef] [PubMed]
  16. Landi, F.; Barillaro, C.; Bellieni, A.; Brandi, V.; Carfì, A.; D’Angelo, M.; Fusco, D.; Landi, G.; Lo Monaco, R.; Martone, A.M.; et al. The new challenge of geriatrics: Saving frail older people from the SARS-CoV-2 pandemic infection. J. Nutr. Health Aging 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Ju, J.; Li, X.; Kumar, S.; Jockusch, S.; Chien, M.; Tao, C.; Morozova, I.; Kalachikov, S.; Kirchdoerfer, R.N.; Russo, J.J. Nucleotide analogues as inhibitors of SARS-CoV polymerase. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  18. Liu, C.; Zhou, Q.; Li, Y.; Garner, L.V.; Watkins, S.P.; Carter, L.J.; Smoot, J.; Gregg, A.C.; Daniels, A.D.; Jervey, S.; et al. Research and development on therapeutic agents and vaccines for COVID-19 and related human coronavirus diseases. ACS Cent. Sci. 2020, 6, 315–331. [Google Scholar] [CrossRef] [PubMed]
  19. Monteil, V.; Kwon, H.; Prado, P.; Hagelkrüys, A.; Wimmer, R.A.; Stahl, M.; Leopoldi, A.; Garreta, E.; Pozo, C.H.d.; Prosper, F.; et al. Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell 2020. [Google Scholar] [CrossRef] [PubMed]
  20. Popov, D. Treatment of Covid-19 Infection. A rationale for current and future pharmacological approach. EC Pulmonol. Respir. Med. 2020, 9, 38–58. [Google Scholar]
  21. Kearney, J. Chloroquine as a potential treatment and prevention measure for the 2019 novel coronavirus: A review. Preprints 2020, 1. [Google Scholar] [CrossRef] [Green Version]
  22. Zhang, L.; Liu, Y. Potential interventions for novel coronavirus in China: A systematic review. J. Med. Virol. 2020, 92, 479–490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Miller, S.M. Vitamin E may worsen acute respiratory tract infections in the elderly. J. Fam. Pr. 2002, 51, 925. [Google Scholar]
  24. Villamor, E.; Mbise, R.; Spiegelman, D.; Hertzmark, E.; Fataki, M.; Peterson, K.E.; Ndossi, G.; Fawzi, W.W. Vitamin A supplements ameliorate the adverse effect of HIV-1, malaria, and diarrheal infections on child growth. Pediatrics 2002, 109, E6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Keil, S.D.; Bowen, R.; Marschner, S. Inactivation of Middle East respiratory syndrome coronavirus (MERS-CoV) in plasma products using a riboflavin-based and ultraviolet light-based photochemical treatment. Transfusion 2016, 56, 2948–2952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Hemila, H. Vitamin C intake and susceptibility to pneumonia. Pediatr. Infect. Dis. J. 1997, 16, 836–837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Martineau, A.R.; Jolliffe, D.A.; Hooper, R.L.; Greenberg, L.; Aloia, J.F.; Bergman, P.; Dubnov-Raz, G.; Esposito, S.; Ganmaa, D.; Ginde, A.A.; et al. Vitamin D supplementation to prevent acute respiratory tract infections: Systematic review and meta-analysis of individual participant data. BMJ 2017, 356, i6583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Lewis, E.D.; Meydani, S.N.; Wu, D. Regulatory role of vitamin E in the immune system and inflammation. Iubmb Life 2019, 71, 487–494. [Google Scholar] [CrossRef] [PubMed]
  29. Te Velthuis, A.J.; Van den Worm, S.H.; Sims, A.C.; Baric, R.S.; Snijder, E.J.; Van Hemert, M.J. Zn(2+) inhibits coronavirus and arterivirus RNA polymerase activity in vitro and zinc ionophores block the replication of these viruses in cell culture. PLoS Pathog. 2010, 6, e1001176. [Google Scholar] [CrossRef] [PubMed]
  30. Harthill, M. Review: Micronutrient selenium deficiency influences evolution of some viral infectious diseases. Biol. Trace Elem. Res. 2011, 143, 1325–1336. [Google Scholar] [CrossRef] [PubMed]
  31. Jayaweera, J.; Reyes, M.; Joseph, A. Childhood iron deficiency anemia leads to recurrent respiratory tract infections and gastroenteritis. Sci. Rep. 2019, 9, 12637. [Google Scholar] [CrossRef] [Green Version]
  32. Leu, G.Z.; Lin, T.Y.; Hsu, J.T. Anti-HCV activities of selective polyunsaturated fatty acids. Biochem Biophys Res. Commun. 2004, 318, 275–280. [Google Scholar] [CrossRef]
  33. Xu, K.; Cai, H.; Shen, Y.; Ni, Q.; Chen, Y.; Hu, S.; Li, J.; Wang, H.; Yu, L.; Huang, H.; et al. Management of corona virus disease-19 (COVID-19): The Zhejiang experience. J. Zhejiang Univ. Med Sci. 2020, 49. [Google Scholar] [CrossRef]
  34. Barazzoni, R.; Bischoff, S.C.; Breda, J.; Wickramasinghe, K.; Krznaric, Z.; Nitzan, D.; Pirlich, M.; Singer, P.; endorsed by the, E.C. ESPEN expert statements and practical guidance for nutritional management of individuals with SARS-CoV-2 infection. Clin. Nutr. 2020. [Google Scholar] [CrossRef] [PubMed]
  35. American Society for Parenteral and Enteral Nutrition—Resources for Clinicians Caring for Patients with Coronavirus. Available online: https://www.nutritioncare.org/Guidelines_and_Clinical_Resources/Resources_for_Clinicians_Caring_for_Patients_with_Coronavirus/ (accessed on 30 April 2020).
  36. Romano, L.; Bilotta, F.; Dauri, M.; Macheda, S.; Pujia, A.; De Santis, G.L.; Tarsitano, M.G.; Merra, G.; Di Renzo, L.; Esposito, E.; et al. Short Report—Medical nutrition therapy for critically ill patients with COVID-19. Eur. Rev. Med. Pharm. Sci. 2020, 24, 4035–4039. [Google Scholar] [CrossRef]
  37. Butler, M.J.; Barrientos, R.M. The impact of nutrition on COVID-19 susceptibility and long-term consequences. Brain Behav. Immun. 2020. [Google Scholar] [CrossRef] [PubMed]
  38. Muscogiuri, G.; Barrea, L.; Savastano, S.; Colao, A. Nutritional recommendations for CoVID-19 quarantine. Eur. J. Clin. Nutr. 2020. [Google Scholar] [CrossRef] [PubMed]
  39. Laviano, A.; Koverech, A.; Zanetti, M. Nutrition support in the time of SARS-CoV-2 (COVID-19). Nutrition 2020. [Google Scholar] [CrossRef]
  40. Caccialanza, R.; Laviano, A.; Lobascio, F.; Montagna, E.; Bruno, R.; Ludovisi, S.; Corsico, A.G.; Di Sabatino, A.; Belliato, M.; Calvi, M.; et al. Early nutritional supplementation in non-critically ill patients hospitalized for the 2019 novel coronavirus disease (COVID-19): Rationale and feasibility of a shared pragmatic protocol. Nutrition 2020, 110835. [Google Scholar] [CrossRef]
  41. Agencies, U.N. Joint statement on nutrition in the context of the COVID-19 pandemic in Asia and the Pacific. Available online: https://www.humanitarianresponse.info/en/operations/bangladesh/document/joint-statement-nutrition-context-covid-19-pandemic-asia-and-pacific (accessed on 30 April 2020).
  42. Krawinkel, M.B. Interaction of nutrition and infections globally: An overview. Ann Nutr Metab 2012, 61 (Suppl. 1), 39–45. [Google Scholar] [CrossRef]
  43. Childs, C.E.; Calder, P.C.; Miles, E.A. Diet and immune function. Nutrients 2019, 11, 1933. [Google Scholar] [CrossRef] [Green Version]
  44. Monteiro, C.A.; Cannon, G.; Moubarac, J.C.; Levy, R.B.; Louzada, M.L.C.; Jaime, P.C. The UN Decade of Nutrition, the NOVA food classification and the trouble with ultra-processing. Public Health Nutr. 2018, 21, 5–17. [Google Scholar] [CrossRef] [Green Version]
  45. Gibney, M.J.; Forde, C.G.; Mullally, D.; Gibney, E.R. Ultra-processed foods in human health: A critical appraisal. Am. J. Clin. Nutr. 2017, 106, 717–724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Monteiro, C.A.; Moubarac, J.C.; Cannon, G.; Ng, S.W.; Popkin, B. Ultra-processed products are becoming dominant in the global food system. Obes. Rev. 2013, 14 (Suppl. 2), 21–28. [Google Scholar] [CrossRef]
  47. Myles, I.A. Fast food fever: Reviewing the impacts of the Western diet on immunity. Nutr. J. 2014, 13, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Sonnenburg, E.D.; Sonnenburg, J.L. The ancestral and industrialized gut microbiota and implications for human health. Nat. Rev. Microbiol. 2019, 17, 383–390. [Google Scholar] [CrossRef] [PubMed]
  49. Levy, M.; Kolodziejczyk, A.A.; Thaiss, C.A.; Elinav, E. Dysbiosis and the immune system. Nat. Rev. Immunol. 2017, 17, 219–232. [Google Scholar] [CrossRef] [PubMed]
  50. La Fata, G.; Weber, P.; Mohajeri, M.H. Probiotics and the gut immune system: Indirect regulation. Probiotics Antimicrob. Proteins 2018, 10, 11–21. [Google Scholar] [CrossRef] [PubMed]
  51. Maldonado Galdeano, C.; Cazorla, S.I.; Lemme Dumit, J.M.; Velez, E.; Perdigon, G. Beneficial effects of probiotic consumption on the immune system. Ann. Nutr. Metab. 2019, 74, 115–124. [Google Scholar] [CrossRef] [PubMed]
  52. Yamada, N.; Saito-Iwamoto, C.; Nakamura, M.; Soeda, M.; Chiba, Y.; Kano, H.; Asami, Y. Lactobacillus gasseri PA-3 Uses the purines IMP, inosine and hypoxanthine and reduces their absorption in rats. Microorganisms 2017, 5, 10. [Google Scholar] [CrossRef] [PubMed]
  53. Yamada, N.; Saito, C.; Murayama-Chiba, Y.; Kano, H.; Asami, Y.; Itoh, H. Lactobacillus gasseri PA-3 utilizes the purines GMP and guanosine and decreases their absorption in rats. Nucleosides Nucleotides Nucleic Acids 2018, 37, 307–315. [Google Scholar] [CrossRef]
  54. Ogawa, J. Analysis of Microbial Purine Metabolism and Its Application for Hyperuricemia Prevention; Institute for Scientific Research GRANT: Noda, Japan, 2006. [Google Scholar]
  55. Wang, H.; Mei, L.; Deng, Y.; Liu, Y.; Wei, X.; Liu, M.; Zhou, J.; Ma, H.; Zheng, P.; Yuan, J.; et al. Lactobacillus brevis DM9218 ameliorates fructose-induced hyperuricemia through inosine degradation and manipulation of intestinal dysbiosis. Nutrition 2019, 62, 63–73. [Google Scholar] [CrossRef]
  56. Li, M.; Yang, D.; Mei, L.; Yuan, L.; Xie, A.; Yuan, J. Screening and characterization of purine nucleoside degrading lactic acid bacteria isolated from Chinese sauerkraut and evaluation of the serum uric acid lowering effect in hyperuricemic rats. PLoS ONE 2014, 9, e105577. [Google Scholar] [CrossRef]
  57. Yamada, N.; Iwamoto, C.; Nakamura, M.; Soeda, M.; Tsuboi, H.; Kano, H.; Asami, Y. Reducing effect of Lactobacillus gasseri PA-3 on the absorption of food-derived purines. Milk Sci. 2016, 65, 25–31. [Google Scholar] [CrossRef]
  58. Yamada, N.; Iwamoto, C.; Kano, H.; Yamaoka, N.; Fukuuchi, T.; Kaneko, K.; Asami, Y. Evaluation of purine utilization by Lactobacillus gasseri strains with potential to decrease the absorption of food-derived purines in the human intestine. Nucleosides Nucleotides Nucleic Acids 2016, 35, 670–676. [Google Scholar] [CrossRef] [PubMed]
  59. Walker, U.A.; Tyndall, A.; Daikeler, T. Rheumatic conditions in human immunodeficiency virus infection. Rheumatology 2008, 47, 952–959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Wu, B.; Roseland, J.M.; Haytowitz, D.B.; Pehrsson, P.R.; Ershow, A.G. Availability and quality of published data on the purine content of foods, alcoholic beverages, and dietary supplements. J. Food Compos. Anal. 2019, 84, 103281. [Google Scholar] [CrossRef]
  61. Yamanaka, H.; Taniguchi, A.; Tsuboi, H.; Kano, H.; Asami, Y. Hypouricaemic effects of yoghurt containing Lactobacillus gasseri PA-3 in patients with hyperuricaemia and/or gout: A randomised, double-blind, placebo-controlled study. Mod. Rheumatol. 2019, 29, 146–150. [Google Scholar] [CrossRef] [PubMed]
  62. Selle, K.; Klaenhammer, T.R. Genomic and phenotypic evidence for probiotic influences of Lactobacillus gasseri on human health. FEMS Microbiol. Rev. 2013, 37, 915–935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Nakayama, Y.; Moriya, T.; Sakai, F.; Ikeda, N.; Shiozaki, T.; Hosoya, T.; Nakagawa, H.; Miyazaki, T. Oral administration of Lactobacillus gasseri SBT2055 is effective for preventing influenza in mice. Sci. Rep. 2014, 4, 4638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Eguchi, K.; Fujitani, N.; Nakagawa, H.; Miyazaki, T. Prevention of respiratory syncytial virus infection with probiotic lactic acid bacterium Lactobacillus gasseri SBT2055. Sci. Rep. 2019, 9, 4812. [Google Scholar] [CrossRef] [PubMed]
  65. Clua, P.; Kanmani, P.; Zelaya, H.; Tada, A.; Kober, A.K.M.H.; Salva, S.; Alvarez, S.; Kitazawa, H.; Villena, J. Peptidoglycan from immunobiotic lactobacillus rhamnosus improves resistance of infant mice to respiratory syncytial viral infection and secondary pneumococcal pneumonia. Front. Immunol. 2017, 8, 948. [Google Scholar] [CrossRef] [PubMed]
  66. De Vrese, M.; Winkler, P.; Rautenberg, P.; Harder, T.; Noah, C.; Laue, C.; Ott, S.; Hampe, J.; Schreiber, S.; Heller, K.; et al. Probiotic bacteria reduced duration and severity but not the incidence of common cold episodes in a double blind, randomized, controlled trial. Vaccine 2006, 24, 6670–6674. [Google Scholar] [CrossRef]
  67. De Vrese, M.; Winkler, P.; Rautenberg, P.; Harder, T.; Noah, C.; Laue, C.; Ott, S.; Hampe, J.; Schreiber, S.; Heller, K.; et al. Effect of Lactobacillus gasseri PA 16/8, Bifidobacterium longum SP 07/3, B. bifidum MF 20/5 on common cold episodes: A double blind, randomized, controlled trial. Clin. Nutr. 2005, 24, 481–491. [Google Scholar] [CrossRef] [PubMed]
  68. Winkler, P.; de Vrese, M.; Laue, C.; Schrezenmeir, J. Effect of a dietary supplement containing probiotic bacteria plus vitamins and minerals on common cold infections and cellular immune parameters. Int. J. Clin. Pharm. 2005, 43, 318–326. [Google Scholar] [CrossRef] [PubMed]
  69. Hao, Q.; Dong, B.R.; Wu, T. Probiotics for preventing acute upper respiratory tract infections. Cochrane Database Syst. Rev. 2015. [Google Scholar] [CrossRef] [PubMed]
  70. Xiang, S.; Fu, J.; Ye, K.; Zheng, Y.; Zhu, X.; Chen, J.; Chen, Y. Effect of Lactobacillus gasseri PA3 on gut microbiota in an in vitro colonic simulation. Food Sci. Nutr. 2019, 7, 3883–3891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Spor, A.; Koren, O.; Ley, R. Unravelling the effects of the environment and host genotype on the gut microbiome. Nat. Rev. Microbiol. 2011, 9, 279–290. [Google Scholar] [CrossRef]
  72. Zhang, C.X.; Wang, H.Y.; Chen, T.X. Interactions between Intestinal Microflora/Probiotics and the Immune System. Biomed. Res. Int. 2019, 2019, 6764919. [Google Scholar] [CrossRef] [Green Version]
  73. Martinez Leo, E.E.; Segura Campos, M.R. Effect of ultra-processed diet on gut microbiota and thus its role in neurodegenerative diseases. Nutrition 2020, 71, 110609. [Google Scholar] [CrossRef] [PubMed]
  74. Martinez Steele, E.; Baraldi, L.G.; Louzada, M.L.; Moubarac, J.C.; Mozaffarian, D.; Monteiro, C.A. Ultra-processed foods and added sugars in the US diet: Evidence from a nationally representative cross-sectional study. BMJ Open 2016, 6, e009892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Wan, M.L.Y.; Forsythe, S.J.; El-Nezami, H. Probiotics interaction with foodborne pathogens: A potential alternative to antibiotics and future challenges. Crit. Rev. Food Sci. Nutr. 2019, 59, 3320–3333. [Google Scholar] [CrossRef]
  76. Azagra-Boronat, I.; Massot-Cladera, M. Strain-Specific probiotic properties of bifidobacteria and lactobacilli for the prevention of diarrhea caused by rotavirus in a preclinical model. Nutrients 2020, 12, 498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Wang, Y.; Pan, C.Q.; Xing, H. Advances in gut microbiota of viral hepatitis cirrhosis. Biomed. Res. Int. 2019, 2019, 9726786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. King, S.; Tancredi, D.; Lenoir-Wijnkoop, I.; Gould, K.; Vann, H.; Connors, G.; Sanders, M.E.; Linder, J.A.; Shane, A.L.; Merenstein, D. Does probiotic consumption reduce antibiotic utilization for common acute infections? A systematic review and meta-analysis. Eur. J. Public Health 2019, 29, 494–499. [Google Scholar] [CrossRef]
  79. Lei, W.T.; Shih, P.C.; Liu, S.J. Effect of probiotics and prebiotics on immune response to influenza vaccination in adults: A systematic review and meta-analysis of randomized controlled trials. Nutrients 2017, 9, 1175. [Google Scholar] [CrossRef]
  80. Lenoir-Wijnkoop, I.; Merenstein, D.; Korchagina, D.; Broholm, C.; Sanders, M.E.; Tancredi, D. Probiotics reduce health care cost and societal impact of flu-like respiratory tract infections in the USA: An economic modeling study. Front. Pharm. 2019, 10, 980. [Google Scholar] [CrossRef] [PubMed]
  81. Becattini, S.; Taur, Y.; Pamer, E.G. Antibiotic-induced changes in the intestinal microbiota and disease. Trends Mol. Med. 2016, 22, 458–478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Monteagudo-Mera, A.; Rastall, R.A.; Gibson, G.R.; Charalampopoulos, D.; Chatzifragkou, A. Adhesion mechanisms mediated by probiotics and prebiotics and their potential impact on human health. Appl. Microbiol. Biotechnol. 2019, 103, 6463–6472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Qin, H.; Zhang, Z.; Hang, X.; Jiang, Y.L. plantarum prevents enteroinvasive Escherichia coli-induced tight junction proteins changes in intestinal epithelial cells. BMC Microbiol 2009, 9, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Caballero-Franco, C.; Keller, K.; De Simone, C.; Chadee, K. The VSL#3 probiotic formula induces mucin gene expression and secretion in colonic epithelial cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2007, 292, G315–G322. [Google Scholar] [CrossRef] [PubMed]
  85. Schenk, M.; Mueller, C. The mucosal immune system at the gastrointestinal barrier. Best Pr. Res. Clin. Gastroenterol. 2008, 22, 391–409. [Google Scholar] [CrossRef] [PubMed]
  86. Vinolo, M.A.R.; Rodrigues, H.G.; Nachbar, R.T.; Curi, R. Regulation of inflammation by short chain fatty acids. Nutrients 2011, 3, 858–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Bron, P.A.; van Baarlen, P.; Kleerebezem, M. Emerging molecular insights into the interaction between probiotics and the host intestinal mucosa. Nat. Rev. Microbiol. 2012, 10, 66–78. [Google Scholar] [CrossRef] [PubMed]
  88. Mohamadzadeh, M.; Olson, S.; Kalina, W.V.; Ruthel, G.; Demmin, G.L.; Warfield, K.L.; Bavari, S.; Klaenhammer, T.R. Lactobacilli activate human dendritic cells that skew T cells toward T helper 1 polarization. Proc. Natl. Acad. Sci. USA 2005, 102, 2880–2885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Kim, H.W.; Hong, R.; Choi, E.Y.; Yu, K.; Kim, N.; Hyeon, J.Y.; Cho, K.K.; Choi, I.S.; Yun, C.-H. A probiotic mixture regulates t cell balance and reduces atopic dermatitis symptoms in mice. Front. Microbiol. 2018, 9, 2414. [Google Scholar] [CrossRef] [PubMed]
  90. Rescigno, M. Dendritic cells in oral tolerance in the gut. Cell Microbiol. 2011, 13, 1312–1318. [Google Scholar] [CrossRef] [PubMed]
  91. Sichetti, M.; De Marco, S.; Pagiotti, R.; Traina, G.; Pietrella, D. Anti-inflammatory effect of multistrain probiotic formulation (L. rhamnosus, B. lactis, and B. longum). Nutrition 2018, 53, 95–102. [Google Scholar] [CrossRef] [PubMed]
  92. Li, J.; Sung, C.Y.; Lee, N.; Ni, Y.; Pihlajamaki, J.; Panagiotou, G.; El-Nezami, H. Probiotics modulated gut microbiota suppresses hepatocellular carcinoma growth in mice. Proc. Natl. Acad. Sci. USA 2016, 113, E1306–E1315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Abdo, Z.; LeCureux, J.; LaVoy, A.; Eklund, B.; Ryan, E.P.; Dean, G.A. Impact of oral probiotic Lactobacillus acidophilus vaccine strains on the immune response and gut microbiome of mice. PLoS ONE 2019, 14, e0225842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Kandasamy, S.; Chattha, K.S.; Vlasova, A.N.; Rajashekara, G.; Saif, L.J. Lactobacilli and Bifidobacteria enhance mucosal B cell responses and differentially modulate systemic antibody responses to an oral human rotavirus vaccine in a neonatal gnotobiotic pig disease model. Gut Microbes 2014, 5, 639–651. [Google Scholar] [CrossRef] [PubMed]
  95. Diaz-Dinamarca, D.A.; Hernandez, C.; Escobar, D.F.; Soto, D.A.; Munoz, G.A.; Badilla, J.F.; Manzo, R.A.; Carrion, F. Mucosal vaccination with lactococcus lactis-secreting surface immunological protein induces humoral and cellular immune protection against group b streptococcus in a murine model. Vaccines 2020, 8, 146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. LeCureux, J.S.; Dean, G.A. Lactobacillus mucosal vaccine vectors: Immune responses against bacterial and viral antigens. mSphere 2018, 3, e00061-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Olivares, M.; Diaz-Ropero, M.P.; Sierra, S.; Lara-Villoslada, F.; Fonolla, J.; Navas, M.; Rodriguez, J.M.; Xaus, J. Oral intake of Lactobacillus fermentum CECT5716 enhances the effects of influenza vaccination. Nutrition 2007, 23, 254–260. [Google Scholar] [CrossRef] [PubMed]
  98. Lemme-Dumit, J.M.; Polti, M.A.; Perdigon, G.; Galdeano, C.M. Probiotic bacteria cell walls stimulate the activity of the intestinal epithelial cells and macrophage functionality. Benef. Microbes 2018, 9, 153–164. [Google Scholar] [CrossRef] [PubMed]
  99. Johnson-Henry, K.C.; Donato, K.A.; Shen-Tu, G.; Gordanpour, M.; Sherman, P.M. Lactobacillus rhamnosus Strain GG Prevents Enterohemorrhagic Escherichia coli O157:H7-Induced Changes in Epithelial Barrier Function. Infect. Immun. 2008, 76, 1340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Parassol, N.; Freitas, M.; Thoreux, K.; Dalmasso, G.; Bourdet-Sicard, R.; Rampal, P. Lactobacillus casei DN-114 001 inhibits the increase in paracellular permeability of enteropathogenic Escherichia coli-infected T84 cells. Res. Microbiol. 2005, 156, 256–262. [Google Scholar] [CrossRef] [PubMed]
  101. Donia, M.S.; Cimermancic, P.; Schulze, C.J.; Wieland Brown, L.C.; Martin, J.; Mitreva, M.; Clardy, J.; Linington, R.G.; Fischbach, M.A. A systematic analysis of biosynthetic gene clusters in the human microbiome reveals a common family of antibiotics. Cell 2014, 158, 1402–1414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Nahui Palomino, R.A.; Vanpouille, C.; Laghi, L. Extracellular vesicles from symbiotic vaginal lactobacilli inhibit HIV-1 infection of human tissues. Nat. Commun. 2019, 10, 5656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Pan, M.; Hidalgo-Cantabrana, C.; Goh, Y.J.; Sanozky-Dawes, R.; Barrangou, R. Comparative analysis of lactobacillus gasseri and lactobacillus crispatus isolated from human urogenital and gastrointestinal tracts. Front. Microbiol. 2019, 10, 3146. [Google Scholar] [CrossRef] [PubMed]
  104. Garcia-Gutierrez, E.; O’Connor, P.M.; Colquhoun, I.J.; Vior, N.M.; Rodríguez, J.M.; Mayer, M.J.; Cotter, P.D.; Narbad, A. Production of multiple bacteriocins, including the novel bacteriocin gassericin M, by Lactobacillus gasseri LM19, a strain isolated from human milk. Appl. Microbiol. Biotechnol. 2020. [Google Scholar] [CrossRef] [Green Version]
  105. Zhou, X.; Yang, B.; Stanton, C.; Ross, R.P.; Zhao, J.; Zhang, H.; Chen, W. Comparative analysis of Lactobacillus gasseri from Chinese subjects reveals a new species-level taxa. BMC Genom. 2020, 21, 119. [Google Scholar] [CrossRef] [PubMed]
  106. Takahashi, H.; Fujita, T.; Suzuki, Y.; Benno, Y. Monitoring and survival of Lactobacillus gasseri SBT2055 in the human intestinal tract. Microbiol. Immunol. 2006, 50, 867–870. [Google Scholar] [CrossRef] [Green Version]
  107. Stoeker, L.; Nordone, S.; Gunderson, S.; Zhang, L.; Kajikawa, A.; LaVoy, A.; Miller, M.; Klaenhammer, T.R.; Dean, G.A. Assessment of Lactobacillus gasseri as a candidate oral vaccine vector. Clin. Vaccine Immunol. 2011, 18, 1834–1844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Olivares, M.; Diaz-Ropero, M.P.; Gomez, N.; Lara-Villoslada, F.; Sierra, S.; Maldonado, J.A.; Martin, R.; Rodriguez, J.M.; Xaus, J. The consumption of two new probiotic strains, Lactobacillus gasseri CECT 5714 and Lactobacillus coryniformis CECT 5711, boosts the immune system of healthy humans. Int. Microbiol. 2006, 9, 47–52. [Google Scholar] [PubMed]
  109. Oh, N.S.; Joung, J.Y.; Lee, J.Y.; Kim, Y. Probiotic and anti-inflammatory potential of Lactobacillus rhamnosus 4B15 and Lactobacillus gasseri 4M13 isolated from infant feces. PLoS ONE 2018, 13, e0192021. [Google Scholar] [CrossRef] [PubMed]
  110. Nishihira, J.; Moriya, T.; Sakai, F.; Kabuki, T.; Kawasaki, Y.; Nishimura, M. Lactobacillus gasseri SBT2055 stimulates immunoglobulin production and innate immunity after influenza vaccination in healthy adult volunteers. Funct. Foods Health Dis. 2016, 6, 544–568. [Google Scholar] [CrossRef]
  111. Kawase, M.; He, F.; Kubota, A.; Yoda, K.; Miyazawa, K.; Hiramatsu, M. Heat-killed Lactobacillus gasseri TMC0356 protects mice against influenza virus infection by stimulating gut and respiratory immune responses. FEMS Immunol. Med. Microbiol. 2012, 64, 280–288. [Google Scholar] [CrossRef]
  112. Sakai, F.; Hosoya, T.; Ono-Ohmachi, A.; Ukibe, K.; Ogawa, A.; Moriya, T.; Kadooka, Y.; Shiozaki, T.; Nakagawa, H.; Nakayama, Y.; et al. Lactobacillus gasseri SBT2055 induces TGF-beta expression in dendritic cells and activates TLR2 signal to produce IgA in the small intestine. PLoS ONE 2014, 9, e105370. [Google Scholar] [CrossRef] [PubMed]
  113. Mazzeo, M.F.; Luongo, D.; Sashihara, T.; Rossi, M.; Siciliano, R.A. Secretome analysis of mouse dendritic cells interacting with a probiotic strain of lactobacillus gasseri. Nutrients 2020, 12, 555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Gebremariam, H.G.; Qazi, K.R.; Somiah, T.; Pathak, S.K.; Sjolinder, H.; Sverremark Ekstrom, E.; Jonsson, A.B. Lactobacillus gasseri suppresses the production of proinflammatory cytokines in helicobacter pylori-infected macrophages by inhibiting the expression of ADAM17. Front. Immunol. 2019, 10, 2326. [Google Scholar] [CrossRef]
  115. Nishihira, J.; Nishimura, M.; Moriya, T.; Sakai, F.; Kabuki, T.; Kawasaki, Y. Chapter 20—Lactobacillus gasseri potentiates immune response against influenza virus infection. In Immunity and Inflammation in Health and Disease; Chatterjee, S., Jungraithmayr, W., Bagchi, D., Eds.; Academic Press: Cambridge, MA, USA, 2018; pp. 249–255. [Google Scholar] [CrossRef]
  116. Nishida, K.; Sawada, D. Health Benefits of Lactobacillus gasseri CP2305 Tablets in young adults exposed to chronic stress: A randomized, double-blind, placebo-controlled study. Nutrients 2019, 11, 1859. [Google Scholar] [CrossRef] [Green Version]
  117. Sugawara, T.; Sawada, D.; Ishida, Y.; Aihara, K.; Aoki, Y.; Takehara, I.; Takano, K.; Fujiwara, S. Regulatory effect of paraprobiotic Lactobacillus gasseri CP2305 on gut environment and function. Microb. Ecol. Health Dis. 2016, 27, 30259. [Google Scholar] [CrossRef]
  118. Yitbarek, A.; Taha-Abdelaziz, K.; Hodgins, D.C.; Read, L.; Nagy, É.; Weese, J.S.; Caswell, J.L.; Parkinson, J.; Sharif, S. Gut microbiota-mediated protection against influenza virus subtype H9N2 in chickens is associated with modulation of the innate responses. Sci. Rep. 2018, 8, 13189. [Google Scholar] [CrossRef] [PubMed]
  119. Gordon, D.E.; Jang, G.M.; Bouhaddou, M.; Xu, J.; Obernier, K.; O’Meara, M.J.; Guo, J.Z.; Swaney, D.L.; Tummino, T.A.; Huettenhain, R.; et al. A SARS-CoV-2-Human protein-protein interaction map reveals drug targets and potential drug-repurposing. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  120. Guo, D. Old weapon for new enemy: Drug repurposing for treatment of newly emerging viral diseases. Virol. Sin. 2020, 1–3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Sexton, N.R.; Smith, E.C.; Blanc, H.; Vignuzzi, M.; Peersen, O.B.; Denison, M.R. Homology-based identification of a mutation in the coronavirus RNA-Dependent RNA polymerase that confers resistance to multiple mutagens. J. Virol. 2016, 90, 7415–7428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Mifsud, E.J.; Hayden, F.G.; Hurt, A.C. Antivirals targeting the polymerase complex of influenza viruses. Antivir. Res. 2019, 169, 104545. [Google Scholar] [CrossRef] [PubMed]
  123. Xue, J.; Moyer, A.; Peng, B.; Wu, J.; Hannafon, B.N.; Ding, W.-Q. Chloroquine is a zinc ionophore. PLoS ONE 2014, 9, e109180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Cheng, V.C.; Chan, J.F.; To, K.K.; Yuen, K.Y. Clinical management and infection control of SARS: Lessons learned. Antivir. Res. 2013, 100, 407–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Peters, H.L.; Jochmans, D.; de Wilde, A.H.; Posthuma, C.C.; Snijder, E.J.; Neyts, J.; Seley-Radtke, K.L. Design, synthesis and evaluation of a series of acyclic fleximer nucleoside analogues with anti-coronavirus activity. Bioorg. Med. Chem. Lett. 2015, 25, 2923–2926. [Google Scholar] [CrossRef] [PubMed]
  126. Chatre, C.; Roubille, F.; Vernhet, H.; Jorgensen, C.; Pers, Y.M. Cardiac complications attributed to chloroquine and hydroxychloroquine: A systematic review of the literature. Drug Saf. 2018, 41, 919–931. [Google Scholar] [CrossRef] [PubMed]
  127. Martinez, M.A. Compounds with therapeutic potential against novel respiratory 2019 coronavirus. Antimicrob. Agents Chemother. 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Fintelman-Rodrigues, N.; Sacramento, C.Q.; Ribeiro Lima, C.; Souza da Silva, F.; Ferreira, A.; Mattos, M.; de Freitas, C.S.; Cardoso Soares, V.; da Silva Gomes Dias, S.; Temerozo, J.R.; et al. Atazanavir inhibits SARS-CoV-2 replication and pro-inflammatory cytokine production. bioRxiv 2020. [Google Scholar] [CrossRef]
  129. Caly, L.; Druce, J.D.; Catton, M.G.; Jans, D.A.; Wagstaff, K.M. The FDA-approved Drug Ivermectin inhibits the replication of SARS-CoV-2 in vitro. Antivir. Res. 2020. [Google Scholar] [CrossRef] [PubMed]
  130. Cortegiani, A.; Ingoglia, G.; Ippolito, M.; Giarratano, A.; Einav, S. A systematic review on the efficacy and safety of chloroquine for the treatment of COVID-19. J. Crit. Care 2020. [Google Scholar] [CrossRef] [PubMed]
  131. Ahn, D.G.; Choi, J.K.; Taylor, D.R.; Oh, J.W. Biochemical characterization of a recombinant SARS coronavirus nsp12 RNA-dependent RNA polymerase capable of copying viral RNA templates. Arch. Virol 2012, 157, 2095–2104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Dinesh, S.; Shikha, G.; Gangwar, B.; Nidhi, S.; Dileep, S. Biological activities of purine analogues: A review. J. Pharm. Sci. Innov. 2012, 1, 29–34. [Google Scholar]
  133. Li, B.-x.; Zhang, H.; Liu, Y.; Li, Y.; Zheng, J.-j.; Li, W.-X.; Feng, K.; Sun, M.; Dai, S.-X. Novel pathways of HIV latency reactivation revealed by integrated analysis of transcriptome and target profile of bryostatin. Sci. Rep. 2020, 10, 3511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Chukwuma Sr, C. Functionalities of purines in health and disease. Mat. J. 2019, 1, 29–40. [Google Scholar]
  135. Jankowska, D.A.; Trautwein-Schult, A.; Cordes, A.; Bode, R.; Baronian, K.; Kunze, G. A novel enzymatic approach in the production of food with low purine content using Arxula adeninivorans endogenous and recombinant purine degradative enzymes. Bioengineered 2015, 6, 20–25. [Google Scholar] [CrossRef] [Green Version]
  136. Vitart, V.; Rudan, I.; Hayward, C.; Gray, N.K.; Floyd, J.; Palmer, C.N.A.; Knott, S.A.; Kolcic, I.; Polasek, O.; Graessler, J.; et al. SLC2A9 is a newly identified urate transporter influencing serum urate concentration, urate excretion and gout. Nat. Genet. 2008, 40, 437–442. [Google Scholar] [CrossRef]
  137. Nyhan, W.L. Disorders of purine and pyrimidine metabolism. Mol. Genet. Metab. 2005, 86, 25–33. [Google Scholar] [CrossRef] [PubMed]
  138. Carlucci, F.; Tabucchi, A.; Perrett, D.; Pizzichini, M.; Rosi, F.; Pagani, R.; Marinello, E. Purine metabolism in HIV-1 virus-infected T lymphocyte population. Biomed. Pharm. 1996, 50, 505–509. [Google Scholar] [CrossRef]
  139. Lewandowski, L.B.; Hsieh, E. Global rheumatology in the time of COVID-19. Lancet Rheumatol. 2020, 2, e254–e255. [Google Scholar] [CrossRef]
  140. Gianfrancesco, M.A.; Hyrich, K.L.; Gossec, L.; Strangfeld, A.; Carmona, L.; Mateus, E.F.; Sufka, P.; Grainger, R.; Wallace, Z.; Bhana, S.; et al. Rheumatic disease and COVID-19: Initial data from the COVID-19 Global Rheumatology Alliance provider registries. Lancet Rheumatol 2020, 2, e250–e253. [Google Scholar] [CrossRef]
  141. Dalbeth, N.; Choi, H.K.; Joosten, L.A.B.; Khanna, P.P.; Matsuo, H.; Perez-Ruiz, F.; Stamp, L.K. Gout. Nat. Rev. Dis. Primers 2019, 5, 69. [Google Scholar] [CrossRef] [PubMed]
  142. Chen, C.; Lü, J.-M.; Yao, Q. Hyperuricemia-Related Diseases and Xanthine Oxidoreductase (XOR) Inhibitors: An Overview. Med. Sci. Monit. 2016, 22, 2501–2512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Trautwein-Schult, A.; Jankowska, D.; Cordes, A.; Hoferichter, P.; Klein, C.; Matros, A.; Mock, H.P.; Baronian, K.; Bode, R.; Kunze, G. Arxula adeninivorans recombinant guanine deaminase and its application in the production of food with low purine content. J. Mol. Microbiol. Biotechnol. 2014, 24, 67–81. [Google Scholar] [CrossRef] [PubMed]
  144. Mahor, D.; Priyanka, A.; Prasad, G.S.; Thakur, K.G. Functional and structural characterization of purine nucleoside phosphorylase from kluyveromyces lactis and its potential applications in reducing purine content in food. PLoS ONE 2016, 11, e0164279. [Google Scholar] [CrossRef] [PubMed]
  145. Kang, Y.N.; Zhang, Y.; Allan, P.W.; Parker, W.B.; Ting, J.W.; Chang, C.Y.; Ealick, S.E. Structure of grouper iridovirus purine nucleoside phosphorylase. Acta Cryst. D Biol. Cryst. 2010, 66, 155–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Stachelska-Wierzchowska, A.; Wierzchowski, J.; Gorka, M.; Bzowska, A.; Stolarski, R.; Wielgus-Kutrowska, B. Tricyclic nucleobase analogs and their ribosides as substrates and inhibitors of purine-nucleoside phosphorylases III. Aminopurine derivatives. Molecules 2020, 25, 681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Prasad, C.; Iqbal, U.; Westfall, S.; Prakash, S. Management of hyperuricemia and gout by prebiotics and probiotics: Potentials and limitations. Int. J. Probiotics Prebiotics 2017, 12, 5–15. [Google Scholar]

Share and Cite

MDPI and ACS Style

Morais, A.H.A.; Passos, T.S.; Maciel, B.L.L.; da Silva-Maia, J.K. Can Probiotics and Diet Promote Beneficial Immune Modulation and Purine Control in Coronavirus Infection? Nutrients 2020, 12, 1737. https://doi.org/10.3390/nu12061737

AMA Style

Morais AHA, Passos TS, Maciel BLL, da Silva-Maia JK. Can Probiotics and Diet Promote Beneficial Immune Modulation and Purine Control in Coronavirus Infection? Nutrients. 2020; 12(6):1737. https://doi.org/10.3390/nu12061737

Chicago/Turabian Style

Morais, Ana H. A., Thais S. Passos, Bruna L. L. Maciel, and Juliana K. da Silva-Maia. 2020. "Can Probiotics and Diet Promote Beneficial Immune Modulation and Purine Control in Coronavirus Infection?" Nutrients 12, no. 6: 1737. https://doi.org/10.3390/nu12061737

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop