Next Article in Journal
Investigation into In Vitro and In Vivo Caenorhabditis elegans Models to Select Cheese Yeasts as Probiotic Candidates for their Preventive Effects against Salmonella Typhimurium
Next Article in Special Issue
Longitudinal Investigation of the Gut Microbiota in Goat Kids from Birth to Postweaning
Previous Article in Journal
Pontiella desulfatans gen. nov., sp. nov., and Pontiella sulfatireligans sp. nov., Two Marine Anaerobes of the Pontiellaceae fam. nov. Producing Sulfated Glycosaminoglycan-like Exopolymers
Previous Article in Special Issue
Beyond Just Bacteria: Functional Biomes in the Gut Ecosystem Including Virome, Mycobiome, Archaeome and Helminths
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microbiota Modulating Nutritional Approaches to Countering the Effects of Viral Respiratory Infections Including SARS-CoV-2 through Promoting Metabolic and Immune Fitness with Probiotics and Plant Bioactives

1
Centre for Food Innovation, Tasmanian Institute of Agriculture, University of Tasmania, Launceston, TAS 7250, Australia
2
Gut Health Research Group, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS 7250, Australia
3
Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, and University of Technology Sydney, Faculty of Science, Ultimo, NSW 2007, Australia
4
Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS 7248, Australia
5
Dingle Wellness, South Fremantle, WA 6162, Australia
*
Authors to whom correspondence should be addressed.
Submission received: 28 May 2020 / Revised: 16 June 2020 / Accepted: 16 June 2020 / Published: 18 June 2020
(This article belongs to the Special Issue Gut Microbiome and Aging)

Abstract

:
Viral respiratory infections (VRIs) can spread quickly and cause enormous morbidity and mortality worldwide. These events pose serious threats to public health due to time lags in developing vaccines to activate the acquired immune system. The high variability of people’s symptomatic responses to viral infections, as illustrated in the current COVID-19 pandemic, indicates the potential to moderate the severity of morbidity from VRIs. Growing evidence supports roles for probiotic bacteria (PB) and prebiotic dietary fiber (DF) and other plant nutritional bioactives in modulating immune functions. While human studies help to understand the epidemiology and immunopathology of VRIs, the chaotic nature of viral transmissions makes it difficult to undertake mechanistic study where the pre-conditioning of the metabolic and immune system could be beneficial. However, recent experimental studies have significantly enhanced our understanding of how PB and DF, along with plant bioactives, can significantly modulate innate and acquired immunity responses to VRIs. Synbiotic combinations of PB and DF potentiate increased benefits primarily through augmenting the production of short-chain fatty acids (SCFAs) such as butyrate. These and specific plant polyphenolics help to regulate immune responses to both restrain VRIs and temper the neutrophil response that can lead to acute respiratory distress syndrome (ARDS). This review highlights the current understanding of the potential impact of targeted nutritional strategies in setting a balanced immune tone for viral clearance and reinforcing homeostasis. This knowledge may guide the development of public health tactics and the application of functional foods with PB and DF components as a nutritional approach to support countering VRI morbidity.

1. Introduction

Viral respiratory infections (VRIs) are a major public health issue. Annually, they induce infectious diseases that result in enormous severe morbidity and mortality globally. Major viral pathogens include the influenza virus, respiratory syncytial virus (RSV), coronavirus, adenovirus, and rhinovirus [1]. The influenza virus is the major cause of seasonal respiratory infections, causing annual epidemics with an estimated 3 to 5 million severe illnesses and 290,000 to 600,000 deaths worldwide [2]. However, other viruses that are associated with lower mortality also cause a huge economic burden owing to their high morbidity [1]. The constant threat of the emergence of novel subtypes and strains of viruses creates an even greater risk to society. This has been clearly demonstrated by the recent pandemic, with the novel coronavirus (CoV)-2 causing severe acute respiratory syndrome (SARS-CoV-2) in susceptible people. First reported in late December 2019 in Wuhan, China, and spreading rapidly across the globe, >3.5 million cases have been confirmed and >250,000 deaths have been reported [3]. The human-to-human transmission of the novel virus, the induction of COVID-19 pneumonia and acute respiratory distress syndrome (ARDS) [4], and its continual geographic expansion serves as an important reminder of our vulnerability to emerging viral infections.
Vaccination to engage the acquired immune system is considered the most effective available means of protecting populations against viral diseases in general. However, rapid antigenic changes in viruses, leading to the emergence of novel strains, hampers efforts to provide adequate strain-specific timely protection against viral diseases. Moreover, the low to moderate effectiveness of vaccines in at-risk populations—for instance, against influenza [5,6]—accounts for its high annual mortality. Immunocompromised individuals show a higher propensity towards acquiring severe respiratory infections and mortality [7,8]. Sub-optimal immune health can be related to age [9], chronic inflammatory conditions [10], metabolic dysfunction [11], or stress [12]. The decline in function compromises the ability of immune cells to counter infections and can lead to the dysregulation of the immune response. Most patients with SARS-CoV-2 infections have mild to moderate illness accompanied by shortness of breath after one week [13]. In severely ill patients, the infection progresses rapidly to acute respiratory failure, ARDS, metabolic acidosis, and septic shock [13]. The shortage of key equipment, including ventilators needed to care for critically ill patients during the COVID-19 pandemic [14], has also led to the need for the rationing of medical equipment and interventions [15]. This underscores the urgent need for directing research efforts towards effective approaches for blunting acute responses to infections particularly in at-risk populations.
There is an increasing consensus on the roles of commensal microbiota in shaping host immunity [16,17,18,19,20,21]. This highlights the opportunity for the modulation of microbiota and its functions to regulate host immune responses against viral infections. Considerable evidence supports the potential of functional food components, such as probiotic bacteria (PB) and prebiotic dietary fiber (DF), in inducing beneficial changes in microbial composition and metabolic function. Thus, the application of PB, DF, and other plant bioactives to confer metabolic and immune benefits could be a pragmatic prophylactic nutritional strategy capable of being widely implemented. This review aims to identify evidence relating to potential mechanisms that could be deployed as preventive strategies and adjuvants to impart immune fitness against the immunopathology of VRIs.

2. Immunopathogenesis of VRIs

Understanding the immunopathological features of VRIs is crucial to facilitating the development of effective strategies to confer protection and designing specific treatments targeted at improving immune functions in response to VRIs and associated damage (Figure 1). Evidence from influenza virus infections and three major beta coronavirus (CoV) pandemic infections—Middle East Respiratory Syndrome (MERS), Severe Acute Respiratory Syndrome (SARS)-CoV-1, and COVID-19—underscores some key immunopathological features of the diseases [22,23]. These include the dysregulation of acute inflammatory responses involving macrophages, neutrophils, dendritic cells (DCs), toll-like receptors (TLRs), cytokines, chemokines, and CD4+ and CD8+ T-cells, as well as tissue remodeling processes and the role of bacterial superinfection [22]. Infection with the influenza virus induces heightened immunological responses that are necessary for viral clearance, including the influx of innate immune cells and the overproduction of cytokines and chemokines. While triggering cytotoxic mechanisms to destroy viral-infected cells is also necessary, this can be detrimental, leading to pulmonary immunopathology [22,24,25]. Tissue damage induced by uncontrolled innate immune responses and excessive neutrophil infiltration upon influenza virus infection has been linked to severe morbidity and mortality [22]. Elevated myeloperoxidase (MPO) activity (congruent with increased neutrophil influx) is confirmed to cause tissue injury and endothelial damage in the lungs [26]. The development of fibrosis in SARS-CoV-2-infected lungs in COVID-19 pneumonia has also been reported [27]. Marked neutrophil infiltration in the lungs of COVID-19 patients has been implicated in inducing a “cytokine storm” [28,29]. The neutrophil-to-lymphocyte ratio (NLR) has been reported to predict severe illness in patients with COVID-19 in the early stages of infection [13]. This agrees with another study that confirmed a strong association between elevated NLR with increased abdominal obesity-induced systemic inflammation in the older Spanish population [30]. Thus, NLR may be used as a prognostic factor for disease severity in VRIs that could facilitate an early detection of severity and the rapid implementation of interventions to aid patient responses to infection.
Virus-induced oxidative stress via the generation of reactive oxygen species (ROS) is also a factor that increases the expression of proinflammatory cytokines and chemokines [31] and sensitizes lung cells to bacteria toxin-mediated necroptosis [32]. The heightened neutrophil influx and cytokine storm coupled with subsequent ARDS correlates with multi-organ damage and mortality among COVID-19 patients [28]. The markedly increased serum concentrations of interleukin (IL)-2R, IL-6, tumor necrosis factor (TNF)-α, and IL-10 that occur more in patients with severe COVID-19 than in moderate cases also suggests an association between the cytokine storm and the disease severity [29]. The dysregulated responses of other immune cells, including macrophages and DCs, are also associated with the pulmonary immunopathology of influenza [22]. These observations indicate that the over-exuberant activation of innate immunity can be detrimental, and strategies to regulate their optimal functioning should be targeted.
Adaptive immune components and CD4+ and CD8+ T-cells are essential for virus clearance and immune regulation. SARS-Co-V2 infection has been reported to cause significant alterations in circulating lymphocytes and T-cell subsets [29]. Lymphopenia, particularly in CD4+ and CD8+ T-cells, as well as reduced interferon (IFN)-γ production by CD4+ T-cells but not CD8+ T cells or natural killer cells are associated with the increased severity of COVID-19 [29]. CD4+ T-cells regulate the immune response by orchestrating the deletion and amplification of immune cells, especially CD8+ T-cells. CD4+ T-cells mediate virus-specific antibody production via the T-cell dependent activation of B cells [33]. For influenza infections, it was shown that the production of IFN-γ by CD4+ T-cells was required for viral clearance, thus ameliorating the immunopathology [22]. In contrast, CD8+ T-cells have cytotoxic effects through cytolytic activities against the target cells or the secretion of cytokines and chemokines [34]. MERS-CoV infects T-cells from peripheral blood and human lymphoid organs and induces their apoptosis, thus contributing to the lymphopenia [35], which is also observed for SARS-CoV-1 [36]. Hence, strategies that promote tight immune regulation and effectively mount adequate levels of innate and adaptive immune responses for viral clearance and the restoration of immune homeostasis should be prioritized. In addition, considering sub-optimal immune activity in immunocompromised patients, developing treatments and prophylactic approaches that promote sufficient immune responses for viral clearance without causing additional damage should be pursued.

3. Sub-Optimal Immunity Driven by Microbial Dysbiosis

A well-functioning immune system determines the host immune fitness, which dictates protection against invading pathogens. The immune surveillance system continually monitors for signs of invasion and signals for appropriate immune responses to mount a defense against specific pathogens. Appropriate immune responses maintain strength and/or return to immune homeostasis after an external challenge [37]. Reponses to gut microbiota are known to influence homoeostasis at distant mucosal sites, including the respiratory tract, through a common mucosal immune system [16,38,39,40]. The immunological health of the gut, primarily mediated by the microbiota, influences lung health via the “gut-lung axis” [39,40,41,42,43]. In addition, microbial communities inhabiting the mucosal surfaces of the respiratory tract also contribute towards host defense against VRIs [17,18]. Acute VRIs are associated with microbial dysbiosis in these communities, thus affecting the optimal functioning of the immune system. Alterations in the microbiota during influenza virus infection contributes to the pathogenesis of secondary bacterial infections, thus increasing the severity of the clinical course in the absence of appropriate immune responses [44].
Additionally, alterations in immune functions associated with chronic inflammation and related metabolic dysfunctions are also known to drive impairment in innate and acquired immune functions in the host [10,11,45]. The failure to effectively mount appropriate immune responses against pathogens renders the host susceptible to infection. The prevalence of comorbidities (including chronic pulmonary diseases, diabetes, hypertension, and cardiovascular diseases) and old age predispose one to infection, the development of ARDS and pneumonia, and a high mortality in COVID-19 patients [46]. Similar factors have been noted for previous infections, such as with influenza virus [7,8,47]. Chronic inflammation and antibiotic use are known to accompany disturbances in the gut microbiota, resulting in dysbiosis and thus aggravating immune dysfunctions [17,48]. Moreover, ageing-related changes in the composition and function of gut microbiota are associated with immunosenescence, a state of gradual deterioration of the immune system [9]. Immunosenescence affects both the innate and adaptive arms of the immune system. This results in a progressive reduction in the ability to mount effective cellular and antibody responses against infections and vaccination in the elderly, increasing their vulnerability to infectious agents [49]. Children and infants are more prone to VRIs due to their immature immune systems. The early-life microbial colonization of the gut and airways influences their susceptibility to severe VRIs [50]. For adults, lifestyle factors including cigarette smoking [51,52,53] and the consumption of a Westernized diet, characterized by low dietary fiber content [54,55] and high fat intake [56], have been associated with promoting the exacerbations of viral infections due to impairment in immune functions. Smoking upregulates the angiotensin-converting enzyme-2 (ACE2) receptor, which has been reported to be the receptor for SARS-CoV-1 and SARS-CoV-2 [53,57]. Smoking is established as the primary etiological factor for chronic obstructive pulmonary disease (COPD) and the patients with COPD are significantly vulnerable to the increased frequency and progression of respiratory viral-induced exacerbations and mortality [24]. In addition, stress-related dysbiosis also has a role in rendering the host susceptible to VRIs. Psychological, intensive physical training, sleep deprivation, and travel-related stressors can cause gut dysbiosis and are associated with respiratory infections among travelers, military troops, astronauts, and elite athletes [12,58,59,60]. Thus, perturbations in gut and respiratory microbial communities are a major contributor to the deterioration of immune homeostasis and resilience that is critical for protection against VRIs.

4. Functional Foods for Immune Fitness

Given the strong links between the impairment of immune functions and severity of viral infections, the development of strategies to support optimal immune function may be effective in protection and prevention of severe morbidity. Increasing evidence exists that supports the roles of gut microbiota and diet in shaping immunity [16,17,18,19,20,21,61,62]. Modulating the composition and metabolic capacity of the microbiome by specific dietary components is a promising strategy to influence immune responses against VRIs. Functional food components including PB, prebiotic DFs and other plant-based bioactive component have been associated with immune benefits, primarily via microbiota modulation and impacting oxidative stress [63,64]. Despite the evidence in promoting health benefits, functional foods have received less attention relative to pharmaceutical agents for application to human VRIs in clinical settings [65]. While functional foods are largely prophylactic and not treatment options for acute medical conditions, there is also uncertainty and even skepticism about their efficacy. This could be principally attributed to variations in the study outcomes of functional food bio-actives. However, while variation in study outcomes is expected from the wide range of functional food components and different intervention study designs, it does not preclude the efficacy of application in specific cases. The experimental evidence elucidating the immune mechanistic efficacy of particular bioactive components and/or combinations has huge potential in guiding public health strategies to develop effective dietary approaches to prevent and/or treat VRIs via promoting immune fitness.

5. Probiotics

Probiotics are “live microorganisms that when administered in appropriate doses, confer a benefit to the health of the host” [66]. Many probiotic bacteria are members of the gut microbiota, and some are being increasingly incorporated into foods to improve gut health and wellbeing. Recently, the immunogenic potential of even non-viable probiotic cells, bacterial exopolysaccharides, and spores has also been noted [67,68,69,70,71,72]. Additionally, the ingestion of probiotic-rich food or supplements has been shown to influence immune functions, partly by driving changes in the metabolic activities of endogenous microbiota [73].
The ability of PBs to induce immunomodulation could be mediated either directly through interaction with immune cells or indirectly by supporting the challenged commensal microbiota [74]. Ingested PBs stimulate the immune system and initiate a network of signals mediated by the whole bacteria or their cell wall components. Once administered, they interact with intestinal epithelial cells (IECs) or immune cells associated with the lamina propria through TLRs or other microbial pattern recognition receptors (PRRs) and trigger the production of an array of cytokines and chemokines. These molecules then interact with other immune cells through a complex network of signaling pathways, leading to the activation of the mucosal immune systems. Specific PBs have been demonstrated to enhance T-helper 1 (Th1) and regulatory T-cell (Treg) function [75] and strengthen the epithelial barrier function by increasing mucin production, tight junction proteins, and goblet cells [76,77,78]. In contrast to some earlier reports [79,80,81], current studies support the potency of specific PBs in inducing changes in gut bacterial diversity [82,83,84]. Probiotic-driven benefits are also linked with their capacity to cross-feed other beneficial members of the gut microbiota by digestive activities that release nutrients. They can therefore promote metabolic shifts, including the increased production of SCFAs [77,78,85]. This can have substantial health benefits, as microbiota-derived SCFAs are known to play a significant role in maintaining gut physiology and influencing metabolic balance [86,87]. Thus, PB application offers potential as a strategy for supporting a healthy immune system.
Experimental studies indicate that there can be a substantial potency of specific PB strains in protecting against VRIs. The effects of PBs on the mucosal system are not limited to the intestinal tract, with cross-modulatory effects confirmed in other locations, including the upper respiratory tract. Protection is mediated by the induction of systemic and/or local cellular immune responses [88]. Specific PBs are also known to stimulate the humoral immune response by increasing the numbers of IgA-secreting cells that migrate from Peyer’s Patches to distant mucosal sites such as respiratory glands [89]. Mice models have been instrumental in demonstrating the potential of mechanisms through which probiotics could confer benefits. The oral administration of live or heat-killed probiotic strains of Lactobacillus [90,91,92,93,94,95] and Bifidobacterium [96] in mice has been demonstrated to improve cytokine production against viruses in the lungs or serum. In addition, many studies support the potency of intranasally administered PBs to protect mice against VRIs by stimulating local innate immune responses directly in the respiratory epithelium [92,97,98,99,100,101]. Some studies have also confirmed the excellent efficacy of certain heat-killed strains of Lactobacillus and Bifidobacterium to stimulate the synthesis of virus-specific immunoglobulins and cellular immune responses in respiratory secretions and serum in mice [67,68,69,70]. Similarly, the oral administration of mice with Bifidobacterium breve YIT4064 increased anti-influenza virus IgG antibodies in serum and protected against infection [102]. The sublingual administration of L. rhamnosus in influenza-infected mice enhanced mucosal secretory IgA production, T and NK cell activities, and IL-12 levels in the lungs [103], thus supporting local cellular and humoral immune functions. However, while the mice models are indicative, the differences in the evolved biology of humans compared to mice requires caution in translation of the results to impacts on human disease [104]. Direct human trials will be needed to validate the probiotic effects.
Most evidence of PB-related benefits has been demonstrated in the treatment or management of specific pathologies [105,106,107,108]. Studies evaluating the immunomodulatory efficacy and potential prophylactic activity of PBs in non-diseased subjects are minimal. PB supplementation with influenza vaccination improved the vaccine efficacy in healthy adults [109,110], supporting the role of PBs in VRIs. However, the systematic reviews of randomized controlled trials of probiotic supplementation in healthy subjects are not conclusive. Studies and meta-analysis of trials report mixed results [111,112,113]. Differences in the duration of intervention, age of subjects (children, adults, elderly), doses (106–1010 cfu), or matrices (milk, yoghurt, capsule) may partly account for the conflicting results [88]. Differences in susceptibility, or other unaccounted risk factors in otherwise healthy populations, may also explain variations in PB efficacy on influencing health outcomes found in the trials. While changes in immune responses induced by interventions in healthy subjects with generally well-functioning immune systems are difficult to demonstrate, the elderly can have sub-optimal immunity. Infectious diseases are common in elderly populations due to the age-related decline in immune efficacy referred to as immunosenescence [9]. Furthermore, ageing-related microbial dysbiosis is also known to fuel inflammation in the gut, affecting immune regulation abilities [49]. Daily supplementation with B. lactis HN019 was reported to enhance NK cell tumoricidal activity and polymorphonuclear (PMN) phagocytic capacity, reinforcing immune resilience against viral infections in healthy elderly subjects. In addition to the decline in the functions and proportions of T and B cells [9], age-related changes have also been reported for innate immune components, including PMN and NK cells [109]. Neutrophils that account for 90–95% of PMN cells in the blood have been confirmed to have reduced chemotaxis and phagocytic activity in the elderly [110]. Despite the increase in the numbers of NK cells with age, their signaling capacity, cytokine production, and up-regulation of co-stimulatory molecules is reported to deteriorate [114]. NK cell immunosenescence has been linked to the higher incidence of viral infections in the elderly [115,116].
In addition to directly influencing immune cell functions, certain PBs may also induce beneficial modulations in gut microbiota that in turn have an impact on immune status. B. longum strains have been demonstrated to cause changes in the level of specific Bifidobacterium species in the elderly that correlated with TNF-α, TGF-β, and IL-10 levels in serum [117]. The daily consumption of Bacillus coagulans BC30 PB spores has been shown to significantly increase the populations of Faecalibacterium prausnitzii, with concomitant increases in anti-inflammatory IL-10 levels in elderly subjects [118], supporting the direct correlation confirmed by another study [119]. B. coagulans BC30 has been confirmed in ex vivo experiments to increase the T-cell production of TNF-α in response to specific adenovirus and influenza virus exposures [71]. L. casei [120] and L. plantaurm [121] have also been evaluated and shown to improve influenza virus vaccination in the elderly
Age-related microbial dysbiosis also affects the metabolic capacity of gut microbiota in producing beneficial metabolites, including SCFAs [49,109]. Declines in the populations of Bacteroides group, Bifidobacterium, Faecalibacterium, Akkermansia, or Clostridium cluster XIVa in the elderly gut is commonly reported [49]. The supplementation of aged mice with L. acidophilus DDS-1 resulted in increased relative abundances of beneficial Akkermansia and Lactobacillus species and enhanced butyrate levels while concomitantly downregulating pro-inflammatory cytokines [82]. Taken together, these studies show that PB application could encourage the restoration of immune functions in the elderly, however more research on the strain-specific effects of PBs is required.
PB-induced benefits in respiratory infections have also been shown in infants and children. Some studies have found specific Lactobacillus and Bifidobacterium PB strains to be beneficial in reducing the number of participants experiencing respiratory infection episodes, their mean duration, antibiotic use, and school or childcare absences [122,123,124]. However, some reported no clear benefit [125,126]. Childcare exposure has been associated with the increased risk of upper respiratory infections along with the immature immune system of children [126]. A meta-analysis confirmed that infants and children who received PBs to prevent common acute illness had a reduced risk of being prescribed antibiotics [124]. A recent systematic review on the benefits of PB-supplemented infant formula concluded that some beneficial effects are possible; however, a lack of existing robust evidence to recommend their routine use was reported [127]. The small amount of data on specific PB strains and outcomes, rather than a genuine lack of effects, was acknowledged. In infants, a PB-supplemented formula (B. infantis R0033, B. bifidum R0071, and L. helveticus R0052) was confirmed to sustain the development of mucosal immunity through effects on secretory IgA (sIgA) production [128]. sIgA accounts for 90% of the immunoglobulins in breast milk and protects the intestinal mucosa and prevents infections mainly by blocking the contact of pathogens with epithelial layer and trapping them within mucus layers [129]. sIgA in the intestine maintains controlled microbial colonization while preserving mucosal homeostasis in newborns [128]. In this context, PBs may benefit the immature immune system of formula-fed newborns that lack the vital first line of defense against pathogens.

6. Prebiotic DF

The lower consumption of fruits and vegetables among adults in Western countries is congruent with the epidemic rise in chronic conditions, including respiratory conditions, obesity, diabetes, cardiovascular diseases, and even cancer [130,131]. A “Westernized diet” is strongly associated with chronic local and systemic inflammation, leading to microbial dysbiosis, altered immunity, and weakened gut barrier functions [63,132]. This diet is generally high in refined carbohydrates and fat, with a low DF content from fruits and vegetables. The underlying chronic conditions were noted to account for high risk of morbidity and mortality for the recent MERS, SARS, and COVID-19 major beta coronavirus pandemics [133,134]. Conversely, diets rich in fruits and vegetables have been shown to lower the risk of cardiovascular diseases, metabolic disorders, and gastrointestinal disorders [135,136,137]. Thus, plant-based diets, functional foods, and supplements present a promising strategy for protecting against respiratory infections.
Prebiotic DF from fruits and vegetables is well-established to modulate the gut microbiota, and numerous benefits have been reported in chronic inflammatory and metabolic conditions [138]. Moreover, increased DF consumption is linked to reduced mortality rates in respiratory-related diseases [139] and improved lung function [140]. A few studies report the ability of prebiotic supplements to confer immune response benefits against viral vaccinations. A prebiotic nutritional formula supplemented with fructo-oligosaccharides (FOS), triglycerols, vitamins, and minerals was shown to improve immune function and reduce the duration of upper respiratory infections in elderly subjects in nursing homes [141,142]. The immunogenic benefits were indicated by enhanced responses to the influenza vaccine (activation of specific T-lymphocyte subsets), reduced fever, and a reduction in newly prescribed antibiotics [141]. In contrast, feeding a prebiotic supplement containing FOS (70% Raftilose® and 30% Raftiline®) to free-living seniors was confirmed to have no clear benefit on immune responses to vaccination [143]. A specific combination of long-chain inulin and oligofructose (Synergy1®) was also shown to impart limited effects on antibody responses to influenza vaccination in middle-aged subjects [144]. The variations in the formulation of prebiotic supplements in these studies could account for the discrepancies in the status of participants and outcomes. The type of prebiotic fiber, its complexity (purified or whole-plant) and added bio-actives tend to considerably influence the benefits achieved [77,78].
The established capacity of DF to influence gut microbiota and influence immune modulation indicates a promising protective potential against viral infections. The ability of DF to influence microbiota composition and undergo microbial fermentation to produce SCFAs, such as butyrate, acetate, and propionate, is frequently cited as the mechanism of action [87]. Mice fed a high-fiber diet rich in inulin had prolonged survival, reduced influenza virus-induced immunopathology, and improved anti-viral T-cell responses [55]. These dual effects strongly correlated to butyrate levels. A high-fiber diet was also confirmed to alter bone marrow hematopoiesis, leading to the accumulation of alternatively activated macrophages in the lungs of influenza virus-infected mice. The macrophages generated less CXCL1 chemokine, reducing early neutrophil influx into the airways and, thus, avoiding exaggerated tissue damage. The fermentation of DF also enhanced CD8+ T-cell metabolism, shown by increased activation, migration, cytotoxic activity, and viral clearance in high fiber-fed infected mice [55,145]. A protective role of SCFAs in modulating neutrophil migration in acute inflammation in the colon was also demonstrated in another study using acetate [146]. SCFAs modulated neutrophil migration, with acetate failing to suppress the accumulation of neutrophils in the intestinal tissues of GPR43−/− mice. The benefits of microbially-fermented SCFAs are suggested to be mediated through the direct actions of G-protein-coupled receptors (GPRs) expressed on the gut epithelium; adipose tissues; and immune cells, including monocytes and neutrophils [147]. These studies highlight the efficacy of DF supplementation in balancing the discrete innate, adaptive, and humoral immune components via SCFAs and setting an optimal immune tone in the airways to facilitate efficient viral clearance while avoiding exaggerated tissue damage [55].
The output of SCFAs varies widely depending on the type of DF as well as the presence of fiber-fermenting bacteria in the gut. In a study comparing the effect of three fermentable fibers on microbiota modulation and SCFA levels in humans, resistant potato starch was noted to be highly butyrogenic compared to inulin and resistant starch from maize [148]. Different bacterial changes were induced with potato resistant starch favoring the abundance of Bifidobacteria populations and butyrate-producing species. However, commonly studied purified DFs represent limited biochemical complexity compared to that occurring in fruits and vegetables [149]. Whole-plant prebiotic sugarcane fiber and green banana resistant starch flour supplements with natural fiber complexity have been shown to elevate SCFA levels and reduce local inflammation in acute colitis mice [77,78]. Non-purified DF supplements contain both soluble and insoluble fibers with rapid and slow-fermentable fractions and at ratios that more accurately represent those occurring in natural whole-plant foods [77]. DF supplementation could therefore be implemented in public health strategies for preventing and protecting against acute infections in addition to vaccination.

7. Synbiotics

The combination of PBs and prebiotic DFs, known as synbiotics, is receiving increased attention for its ability to confer augmented health benefits. A synbiotic is expected to impart augmented benefits to the host, owing to either complementary and/or synergistic functions [150]. The success of achieving the desired health effects with either PB or prebiotic DFs is dependent on several factors. PB benefits are highly species- and strain-specific. Moreover, regardless of the species/strain and the potential effects, the administered PB must survive gastric and bile acids and reach the intestine to impart the benefits associated with live organisms [75,151]. For sustained positive effects, PBs also need to be present from either continued ingestion or from having an effective prolonged residence time and replication in the gut. Furthermore, the ability of DF to induce its benefits via SCFA production is strictly dependent on the presence of fiber-fermenting gut microbiota [152]. Thus, the induction of substantial modulations in the complex network of microbiota and its metabolic capacity by specific PBs or prebiotics would be improved with a synbiotic. This involves simultaneous supplementation of the PB and prebiotic DF in combination to improve efficacy and health outcomes [77,78].
There is clinical evidence for synbiotic efficacy against VRIs that has been demonstrated in elderly populations, but the outcomes of this limited number of studies vary. A synbiotic with prebiotic galacto-oligosaccharide (GOS) and a bifidogenic growth factor in combination with heat-treated lactic acid-bacteria-fermented milk in elderly subjects (with history of stroke) enhanced vaccination response against influenza [153]. Synbiotic intervention in that study influenced Bifidobacterium counts and facilitated the sustainment of seroprotective effects for a longer period to the vaccine. In contrast, in another study, B. longum bv. Infantis CCUG 52486, combined with a prebiotic GOS, failed to reduce age-related changes in the NK cell response to seasonal influenza vaccination in elderly compared to young individuals [154]. There were only insignificant effects on B- and T-cell profiles in older subjects compared to those of younger counterparts [155]. The synbiotic combination increased memory IgA+, memory IgG+ and total IgG+ B cell levels in young subjects but failed to reproduce these effects in older participants and also ineffective in significantly altering T-cell subsets. These observations highlight the critical nature of immunosenescence, characterized by significant alteration in immune functions and gut dysbiosis in influencing the outcomes of interventions [156]. This might suggest that a more precise approach based on further research is required when selecting ingredients for synbiotic application in ageing. The application of senolytics, compounds that induce apoptosis in senescent cells, are currently undergoing clinical trials in humans for treating a variety of age-related pathologies [157,158]. Many senolytics, such as quercetin, fisetin, piperlongumine, and curcumin, have been identified as natural phytochemical compounds in foods [159]. Research efforts to determine the efficacy of combining senolytics and synbiotics in improving the age-related decline in immune functions could be explored.
Developing ARDS is responsible for high mortality among the elderly and individuals with chronic conditions and COVID-19 pneumonia [46]. Therefore, the designing of prophylactic nutritional strategies that facilitate mounting appropriate immune responses in at-risk individuals is prudent. The augmentation of SCFA production, for instance, could be a critical mechanism to support the development of more effective immunity (Figure 2). Gut-derived SCFAs have been shown to influence the functions of innate immune cells as well as impact acquired immune components. Notably, SCFAs are not restricted to the intestinal tract and are also disseminated through the circulatory system [160]. SCFAs produced by the bacteria are absorbed in the colon and either used by colonocytes or transported via the portal vein to reach the blood circulation and other organs. High fiber-induced SCFA production alters bone marrow hematopoiesis, characterized by the enhanced generation of macrophages and DC precursors and the subsequent seeding of the lungs by highly phagocytic DCs [145]. This type of DC was reported to have an impaired ability to induce Th2 cell effector function, thus resolving allergic airway inflammation in fiber-fed mice. In another study, the SCFA butyrate inhibited histone deacetylase 3 (HDAC3) to confer macrophages with non-inflammatory enhanced antimicrobial activity [161]. A shift in the macrophage metabolism, shown by the reduced glycolysis and inhibition of the mechanistic target of rapamycin (mTOR) activity, accounted for the butyrate-induced antimicrobial activity of the macrophages. Similarly, the ability of sodium butyrate to alleviate acute lung injury in mice has been shown through suppression of high-mobility group box 1 (HMGB1) release and NF-κB activation [162]. While the NF-κB activation promotes the heightened expression of inflammatory mediators in response to injury and inflammation, HMGB1 participates in the downstream development of acute lung injury as a late pro-inflammatory mediator. In addition, significant experimental evidence also supports the role of SCFAs in regulating the activities and differentiation of T-cells in impacting tissue inflammation [160]. Furthermore, SCFAs also accelerate cellular metabolism and regulate gene expression to promote plasma B cell differentiation in antibody-producing cells [54]. Hence, boosting SCFAs via microbial fermentation has promising potential in improving appropriate mucosal and systemic innate and acquired immune responses to control inflammation during infections and reinforce homeostasis.
Selecting a symbiotic combination using PBs that can potentiate elevated SCFAs by efficiently metabolizing the co-administered prebiotic DF, supporting other gut microbial fermenters, or prolonging the residence time of the PB in the gut should be considered. The declines in populations of primary and secondary fermenters in the inflamed gut is one of the prominent features of dysbiosis. In such conditions, the supplementation of only DF is insufficient to produce significant levels of SCFAs to impart benefits [152]. Similarly, probiotic intervention alone may be of limited efficacy in the absence of fiber substrate to influence the growth and activity of the administered probiotic. Probiotic spores, in synbiotic combination with whole-plant prebiotic DF, augmented butyrate production along the entire colon length [77,78]. In contrast, neither PB B. coagulans or prebiotic alone could produce the same level of this effect in a chemically induced mice model of colitis. The ability of B. coagulans to metabolize a variety of plant fibers to produce SCFAs and lactic acid has been previously determined [163,164,165]. B. coagulans spores in synbiotic combination with whole-plant sugar cane fibre [77] or green banana resistant starch [78] induced protection against chemically induced damage to the colonic epithelium by reducing inflammatory infiltrates and preventing the disruption of tight junction proteins, along with augmentation of SCFA levels. The administration of B. coagulans, in combination with FOS and GOS in the elderly, has also been shown to result in beneficial microbial changes in fecal microbiota ex vivo [166], while another in vivo study [167] identified a suitable synbiotic combination to enhance the butyrate production in swine, inducing improved immune functions. Potato resistant starch was reported to stimulate lactic acid bacteria and secondary fermenters, such as Anaerostipes hadrus, in the GI tract of swine, increasing the SCFA production. Thus, a synbiotic that can deliver a fermentable DF and a PB that can metabolize it is an attractive strategy to support optimal immune responses against the immunopathology of VRIs (Figure 2).

8. Polyphenolic Plant Bioactives

In addition to DF, phytochemicals including vitamins, micronutrients, and polyphenols present in fruits and vegetables are ingested as a part of the diet. Phytochemicals have similarly been shown to be of considerable importance in nutritional strategies for addressing the severity of viral respiratory diseases [62,168,169]. Apart from the ability of certain polyphenols to influence microbiota composition, they also impart antioxidant, anti-inflammatory, and anti-viral effects [170,171,172]. The antiviral effects of polyphenols have been demonstrated to be mediated either by direct inhibitory effects on virus replication or through the induction of immunomodulatory/antioxidant responses [170,171]. Oxidative stress has been implicated in lung tissue injury and epithelial barrier dysfunction in acute respiratory viral infections [31,32]. Accumulating evidence has revealed the excellent immunomodulatory effects of epigallocatechin-3-gallate (EGCG), abundant in green tea on both innate and adaptive immune responses [173]. EGCG supplementation has been proven experimentally to alleviate acute lung injury induced by swine influenza virus via the inhibition of TLR4 signaling and reducing inflammatory cell migration [174]. Green tea catechin metabolites have been shown to enhance CD4+ T-cell and NK cell activities in vivo [175]. Moreover, polyphenols upon reaching the colon are also known to interact with gut microbiota [176], thus influencing their metabolic and immune capabilities [177]. While this evidence from experimental studies indicates the substantial capacity of plant-based bioactives in influencing immune functions and microbiota, the limited studies in viral-related human disease highlights the need for more research in this area.

9. Conclusions

There is strong evidence to support the integral role of the microbiota in shaping host immunity. The severe immunopathology of VRIs, including tissue damage, pneumonia, ARDS, multiorgan failure, and death, is induced by dysregulated immune responses and exacerbated inflammation. Moreover, underlying chronic inflammation and associated dysbiosis increases the risk of morbidity and mortality. Employing combined PB, DF, and nutritionally sourced plant bioactives to modulate bacterial composition and activities is a pragmatic approach for enhanced protection against the acute morbidities associated with VRIs. The use of selected synergistic combinations of compatible PB, DF, and bioactives might be capable of improving consistency in balancing innate and acquired immune responses at appropriate levels for viral clearance and to reinforce homeostasis. The evidence from experimental studies on their ability to influence immune health and disease severity in VRIs warrants more defined clinical studies to confirm the benefits in humans. This knowledge will potentially guide public health recommendations and the development of dietary strategies targeted at enhanced immune fitness and wellbeing in response to acute viral challenges.

Author Contributions

T.S. and R.S. conceived the review, T.S. drafted the manuscript, and all the authors reviewed the manuscript for important intellectual content and agreed to the published version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding. P.M.H. is funded by a fellowship and grants from the National Health and Medical Research Council (NHMRC) of Australia (1175134, 1138004). S.S.S. is funded by grants from Clifford Craig Foundation Launceston General Hospital and Rebecca L. Cooper Medical Research Foundation.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analysis, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Zhang, N.; Wang, L.; Deng, X.; Liang, R.; Su, M.; He, C.; Hu, L.; Su, Y.; Ren, J.; Yu, F. Recent advances in the detection of respiratory virus infection in humans. J. Med. Virol. 2020, 92, 408–417. [Google Scholar] [CrossRef] [PubMed]
  2. Iuliano, A.D.; Roguski, K.M.; Chang, H.H.; Muscatello, D.J.; Palekar, R.; Tempia, S.; Cohen, C.; Gran, J.M.; Schanzer, D.; Cowling, B.J.; et al. Estimates of global seasonal influenza-associated respiratory mortality: A modelling study. Lancet 2018, 391, 1285–1300. [Google Scholar] [CrossRef]
  3. WHO. Coronavirus disease 2019 (Covid-19): Situation report, 80. Available online: https://www.Who.Int/docs/default-source/coronaviruse/situation-reports/20200409-sitrep-80-covid-19.Pdf?Sfvrsn=1b685d64_4 (accessed on 9 April 2020).
  4. Li, Q.; Guan, X.; Wu, P.; Wang, X.; Zhou, L.; Tong, Y.; Ren, R.; Leung, K.S.M.; Lau, E.H.Y.; Wong, J.Y.; et al. Early transmission dynamics in wuhan, china, of novel coronavirus–infected pneumonia. N. Engl. J. Med. 2020, 382, 1199–1207. [Google Scholar] [CrossRef] [PubMed]
  5. Paules, C.I.; Sullivan, S.G.; Subbarao, K.; Fauci, A.S. Chasing seasonal influenza—the need for a universal influenza vaccine. N. Engl. J. Med. 2018, 378, 7–9. [Google Scholar] [CrossRef] [PubMed]
  6. Belongia, E.A.; Simpson, M.D.; King, J.P.; Sundaram, M.E.; Kelley, N.S.; Osterholm, M.T.; McLean, H.Q. Variable influenza vaccine effectiveness by subtype: A systematic review and meta-analysis of test-negative design studies. Lancet Infect. Dis. 2016, 16, 942–951. [Google Scholar] [CrossRef]
  7. Chen-Yu Hsu, A.; Starkey, M.R.; Hanish, I.; Parsons, K.; Haw, T.J.; Howland, L.J.; Barr, I.; Mahony, J.B.; Foster, P.S.; Knight, D.A. Targeting pi3k-p110α suppresses influenza virus infection in chronic obstructive pulmonary disease. Am. J. Resp. Crit. Care Med. 2015, 191, 1012–1023. [Google Scholar] [CrossRef]
  8. Starkey, M.R.; Jarnicki, A.G.; Essilfie, A.-T.; Gellatly, S.L.; Kim, R.Y.; Brown, A.C.; Foster, P.S.; Horvat, J.C.; Hansbro, P.M. Murine models of infectious exacerbations of airway inflammation. Curr. Opin. Pharmacol. 2013, 13, 337–344. [Google Scholar] [CrossRef] [PubMed]
  9. Aiello, A.; Farzaneh, F.; Candore, G.; Caruso, C.; Davinelli, S.; Gambino, C.M.; Ligotti, M.E.; Zareian, N.; Accardi, G. Immunosenescence and its hallmarks: How to oppose aging strategically? A review of potential options for therapeutic intervention. Front. Immunol. 2019, 10, 2247. [Google Scholar] [CrossRef] [Green Version]
  10. Hijano, D.R.; Maron, G.; Hayden, R.T. Respiratory viral infections in patients with cancer or undergoing hematopoietic cell transplant. Front. Microbiol. 2018, 9, 3097. [Google Scholar] [CrossRef]
  11. Honce, R.R.; Schultz-Cherry, S. Impact of obesity on influenza a virus pathogenesis, immune response, and evolution. Front. Immunol. 2019, 10, 1071. [Google Scholar] [CrossRef]
  12. Tiollier, E.; Chennaoui, M.; Gomez-Merino, D.; Drogou, C.; Filaire, E.; Guezennec, C.Y. Effect of a probiotics supplementation on respiratory infections and immune and hormonal parameters during intense military training. Mil. Med. 2007, 172, 1006–1011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Liu, J.; Liu, Y.; Xiang, P.; Pu, L.; Xiong, H.; Li, C.; Zhang, M.; Tan, J.; Xu, Y.; Song, R.; et al. Neutrophil-to-lymphocyte ratio predicts severe illness patients with 2019 novel coronavirus in the early stage. medRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  14. Ranney, M.L.; Griffeth, V.; Jha, A.K. Critical supply shortages—the need for ventilators and personal protective equipment during the covid-19 pandemic. N. Engl. J. Med. 2020, 382, e41. [Google Scholar] [CrossRef]
  15. Emanuel, E.J.; Persad, G.; Upshur, R.; Thome, B.; Parker, M.; Glickman, A.; Zhang, C.; Boyle, C.; Smith, M.; Phillips, J.P. Fair allocation of scarce medical resources in the time of covid-19. N. Engl. J. Med. 2020, 382, 2049–2055. [Google Scholar] [CrossRef] [PubMed]
  16. Chotirmall, S.H.; Gellatly, S.L.; Budden, K.F.; Mac Aogáin, M.; Shukla, S.D.; Wood, D.L.A.; Hugenholtz, P.; Pethe, K.; Hansbro, P.M. Microbiomes in respiratory health and disease: An asia-pacific perspective. Respirology 2017, 22, 240–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Budden, K.F.; Shukla, S.D.; Rehman, S.F.; Bowerman, K.L.; Keely, S.; Hugenholtz, P.; Armstrong-James, D.P.; Adcock, I.M.; Chotirmall, S.H.; Chung, K.F.; et al. Functional effects of the microbiota in chronic respiratory disease. Lancet Resp. Med. 2019, 7, 907–920. [Google Scholar] [CrossRef]
  18. Lee, K.H.; Gordon, A.; Shedden, K.; Kuan, G.; Ng, S.; Balmaseda, A.; Foxman, B. The respiratory microbiome and susceptibility to influenza virus infection. PLoS ONE 2019, 14, e0207898. [Google Scholar] [CrossRef] [Green Version]
  19. Li, N.; Ma, W.-T.; Pang, M.; Fan, Q.-L.; Hua, J.-L. The commensal microbiota and viral infection: A comprehensive review. Front. Immunol. 2019, 10, 1551. [Google Scholar] [CrossRef]
  20. Wilks, J.; Golovkina, T. Influence of microbiota on viral infections. PLoS Pathog. 2012, 8, e1002681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Ichinohe, T.; Pang, I.K.; Kumamoto, Y.; Peaper, D.R.; Ho, J.H.; Murray, T.S.; Iwasaki, A. Microbiota regulates immune defense against respiratory tract influenza a virus infection. Proc. Natl. Acad. Sci. USA 2011, 108, 5354–5359. [Google Scholar] [CrossRef] [Green Version]
  22. Damjanovic, D.; Small, C.-L.; Jeyananthan, M.; McCormick, S.; Xing, Z. Immunopathology in influenza virus infection: Uncoupling the friend from foe. Clin. Immunol. 2012, 144, 57–69. [Google Scholar] [CrossRef] [PubMed]
  23. Dandekar, A.A.; Perlman, S. Immunopathogenesis of coronavirus infections: Implications for sars. Nat. Rev. Immunol. 2005, 5, 917–927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Atto, B.; Eapen, M.S.; Sharma, P.; Frey, U.; Ammit, A.J.; Markos, J.; Chia, C.; Larby, J.; Haug, G.; Weber, H.C. New therapeutic targets for the prevention of infectious acute exacerbations of copd: Role of epithelial adhesion molecules and inflammatory pathways. Clin. Sci. 2019, 133, 1663–1703. [Google Scholar] [CrossRef] [PubMed]
  25. Eapen, M.S.; Sohal, S.S. Understanding novel mechanisms of microbial pathogenesis in chronic lung disease: Implications for new therapeutic targets. Clin. Sci. 2018, 132, 375–379. [Google Scholar] [CrossRef]
  26. Narasaraju, T.; Yang, E.; Samy, R.P.; Ng, H.H.; Poh, W.P.; Liew, A.-A.; Phoon, M.C.; van Rooijen, N.; Chow, V.T. Excessive neutrophils and neutrophil extracellular traps contribute to acute lung injury of influenza pneumonitis. Am. J. Pathol. 2011, 179, 199–210. [Google Scholar] [CrossRef]
  27. Shi, H.; Han, X.; Jiang, N.; Cao, Y.; Alwalid, O.; Gu, J.; Fan, Y.; Zheng, C. Radiological findings from 81 patients with covid-19 pneumonia in wuhan, china: A descriptive study. Lancet Infect. Dis. 2020, 20, 425–434. [Google Scholar] [CrossRef]
  28. Barnes, B.J.; Adrover, J.M.; Baxter-Stoltzfus, A.; Borczuk, A.; Cools-Lartigue, J.; Crawford, J.M.; Daßler-Plenker, J.; Guerci, P.; Huynh, C.; Knight, J.S.; et al. Targeting potential drivers of covid-19: Neutrophil extracellular traps. J. Exp. Med. 2020, 217. [Google Scholar] [CrossRef]
  29. Chen, G.; Wu, D.; Guo, W.; Cao, Y.; Huang, D.; Wang, H.; Wang, T.; Zhang, X.; Chen, H.; Yu, H.; et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J. Clin. Investig. 2020, 130. [Google Scholar] [CrossRef] [Green Version]
  30. Rodríguez-Rodríguez, E.; López-Sobaler, A.M.; Ortega, R.M.; Delgado-Losada, M.L.; López-Parra, A.M.; Aparicio, A. Association between neutrophil-to-lymphocyte ratio with abdominal obesity and healthy eating index in a representative older Spanish population. Nutrients 2020, 12, 855. [Google Scholar] [CrossRef] [Green Version]
  31. Hosakote, Y.M.; Rayavara, K. Respiratory syncytial virus-induced oxidative stress in lung pathogenesis. In Oxidative Stress in Lung Diseases; Chakraborti, S., Parinandi, N., Ghosh, R., Ganguly, N., Chakraborti, T., Eds.; Springer: Singapore, 2020; pp. 297–330. [Google Scholar]
  32. Gonzalez-Juarbe, N.; Riegler, A.N.; Jureka, A.S.; Gilley, R.P.; Brand, J.; Trombley, J.E.; Scott, N.R.; Dube, P.H.; Petit, C.M.; Harrod, K.S.; et al. Influenza-induced oxidative stress sensitizes lung cells to bacterial toxin-mediated necroptosis. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  33. Xu, X.; Gao, X.-M. Immunological responses against SARS-coronavirus infection in humans. Cell. Mol. Immunol. 2004, 1, 119–122. [Google Scholar] [PubMed]
  34. Frasca, L.; Piazza, C.; Piccolella, E. CD4+ T cells orchestrate both amplification and deletion of CD8+ T cells. Crit. Rev. Immunol. 1998, 18, 569–594. [Google Scholar] [CrossRef] [PubMed]
  35. Chu, H.; Zhou, J.; Wong, B.H.-Y.; Li, C.; Chan, J.F.-W.; Cheng, Z.-S.; Yang, D.; Wang, D.; Lee, A.C.-Y.; Li, C. Middle east respiratory syndrome coronavirus efficiently infects human primary t lymphocytes and activates the extrinsic and intrinsic apoptosis pathways. J. Infect. Dis. 2016, 213, 904–914. [Google Scholar] [CrossRef] [Green Version]
  36. He, Z.; Zhao, C.; Dong, Q.; Zhuang, H.; Song, S.; Peng, G.; Dwyer, D.E. Effects of severe acute respiratory syndrome (sars) coronavirus infection on peripheral blood lymphocytes and their subsets. Int. J. Infect. Dis. 2005, 9, 323–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Sompayrac, L.M. How the Immune System Work; John Wiley & Sons: Hoboken, NJ, USA, 2019. [Google Scholar]
  38. Domínguez-Díaz, C.; García-Orozco, A.; Riera-Leal, A.; Padilla-Arellano, J.R.; Fafutis-Morris, M. Microbiota and its role on viral evasion: Is it with us or against us? Front. Cell Infect. Microbiol. 2019, 9, 256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Budden, K.F.; Gellatly, S.L.; Wood, D.L.; Cooper, M.A.; Morrison, M.; Hugenholtz, P.; Hansbro, P.M. Emerging pathogenic links between microbiota and the gut–lung axis. Nat. Rev. Microbiol. 2017, 15, 55–63. [Google Scholar] [CrossRef] [PubMed]
  40. Shukla, S.D.; Budden, K.F.; Neal, R.; Hansbro, P.M. Microbiome effects on immunity, health and disease in the lung. Clin. Transl. Immunol. 2017, 6, e133. [Google Scholar] [CrossRef]
  41. Fricker, M.; Goggins, B.J.; Mateer, S.; Jones, B.; Kim, R.Y.; Gellatly, S.L.; Jarnicki, A.G.; Powell, N.; Oliver, B.G.; Radford-Smith, G.; et al. Chronic cigarette smoke exposure induces systemic hypoxia that drives intestinal dysfunction. JCI Insight 2018, 3, e94040. [Google Scholar] [CrossRef] [Green Version]
  42. Mateer, S.W.; Mathe, A.; Bruce, J.; Liu, G.; Maltby, S.; Fricker, M.; Goggins, B.J.; Tay, H.L.; Marks, E.; Burns, G.; et al. Il-6 drives neutrophil-mediated pulmonary inflammation associated with bacteremia in murine models of colitis. Am. J. Pathol. 2018, 188, 1625–1639. [Google Scholar] [CrossRef] [Green Version]
  43. Liu, G.; Mateer, S.W.; Hsu, A.; Goggins, B.J.; Tay, H.; Mathe, A.; Fan, K.; Neal, R.; Bruce, J.; Burns, G.; et al. Platelet activating factor receptor regulates colitis-induced pulmonary inflammation through the nlrp3 inflammasome. Mucosal Immunol. 2019, 12, 862–873. [Google Scholar] [CrossRef]
  44. Hanada, S.; Pirzadeh, M.; Carver, K.Y.; Deng, J.C. Respiratory viral infection-induced microbiome alterations and secondary bacterial pneumonia. Front. Immunol. 2018, 9, 2640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Hansbro, P.M.; Kim, R.Y.; Starkey, M.R.; Donovan, C.; Dua, K.; Mayall, J.R.; Liu, G.; Hansbro, N.G.; Simpson, J.L.; Wood, L.G. Mechanisms and treatments for severe, steroid-resistant allergic airway disease and asthma. Immunol. Rev. 2017, 278, 41–62. [Google Scholar] [CrossRef] [PubMed]
  46. Wu, C.; Chen, X.; Cai, Y.; Xia, J.a.; Zhou, X.; Xu, S.; Huang, H.; Zhang, L.; Zhou, X.; Du, C.; et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern. Med. 2020, e200994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Liu, S.; Sun, J.; Cai, J.; Miao, Z.; Lu, M.; Qin, S.; Wang, X.; Lv, H.; Yu, Z.; Amer, S. Epidemiological, clinical and viral characteristics of fatal cases of human avian influenza a (h7n9) virus in Zhejiang province, China. J. Infect. 2013, 67, 595–605. [Google Scholar] [CrossRef] [PubMed]
  48. 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]
  49. Salazar, N.; Arboleya, S.; Fernández-Navarro, T.; de los Reyes-Gavilán, C.G.; Gonzalez, S.; Gueimonde, M. Age-associated changes in gut microbiota and dietary components related with the immune system in adulthood and old age: A cross-sectional study. Nutrients 2019, 11, 1765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Lynch, J.P.; Sikder, M.A.A.; Curren, B.F.; Werder, R.B.; Simpson, J.; Cuív, P.Ó.; Dennis, P.G.; Everard, M.L.; Phipps, S. The influence of the microbiome on early-life severe viral lower respiratory infections and asthma—food for thought? Front. Immunol. 2017, 8, 156. [Google Scholar] [CrossRef] [Green Version]
  51. Lawrence, H.; Hunter, A.; Murray, R.; Lim, W.S.; McKeever, T. Cigarette smoking and the occurrence of influenza -systematic review. J. Infect. 2019, 79, 401–406. [Google Scholar] [CrossRef]
  52. Bauer, C.M.T.; Morissette, M.C.; Stämpfli, M.R. The influence of cigarette smoking on viral infections: Translating bench science to impact copd pathogenesis and acute exacerbations of copd clinically. Chest 2013, 143, 196–206. [Google Scholar] [CrossRef]
  53. Brake, S.J.; Barnsley, K.; Lu, W.; McAlinden, K.D.; Eapen, M.S.; Sohal, S.S. Smoking upregulates angiotensin-converting enzyme-2 receptor: A potential adhesion site for novel coronavirus SARS-CoV-2 (Covid-19). J. Clin. Med. 2020, 9, 841. [Google Scholar] [CrossRef] [Green Version]
  54. Kim, M.; Qie, Y.; Park, J.; Kim, C.H. Gut microbial metabolites fuel host antibody responses. Cell Host Microbe 2016, 20, 202–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Trompette, A.; Gollwitzer, E.S.; Pattaroni, C.; Lopez-Mejia, I.C.; Riva, E.; Pernot, J.; Ubags, N.; Fajas, L.; Nicod, L.P.; Marsland, B.J. Dietary fiber confers protection against flu by shaping ly6− patrolling monocyte hematopoiesis and CD8+ T cell metabolism. Immunity 2018, 48, 992–1005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Kosaraju, R.; Guesdon, W.; Crouch, M.J.; Teague, H.L.; Sullivan, E.M.; Karlsson, E.A.; Schultz-Cherry, S.; Gowdy, K.; Bridges, L.C.; Reese, L.R. B cell activity is impaired in human and mouse obesity and is responsive to an essential fatty acid upon murine influenza infection. J. Immunol. 2017, 198, 4738–4752. [Google Scholar] [CrossRef] [Green Version]
  57. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.-H.; Nitsche, A.; et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 2020, 181, 271–280.e278. [Google Scholar] [CrossRef]
  58. Crucian, B.; Babiak-Vazquez, A.; Johnston, S.; Pierson, D.L.; Ott, C.M.; Sams, C. Incidence of clinical symptoms during long-duration orbital spaceflight. Int. J. Gen. Med. 2016, 9, 383–391. [Google Scholar] [CrossRef] [Green Version]
  59. Colbey, C.; Cox, A.J.; Pyne, D.B.; Zhang, P.; Cripps, A.W.; West, N.P. Upper respiratory symptoms, gut health and mucosal immunity in athletes. Sports Med. 2018, 48, 65–77. [Google Scholar] [CrossRef] [Green Version]
  60. Trimble, A.; Moffat, V.; Collins, A.M. Pulmonary infections in the returned traveller. Pneumonia 2017, 9, 1. [Google Scholar] [CrossRef] [Green Version]
  61. Hughes, D.A. Dietary antioxidants and human immune function. Nutr. Bull. 2000, 25, 35–41. [Google Scholar] [CrossRef]
  62. Gombart, A.F.; Pierre, A.; Maggini, S. A review of micronutrients and the immune system–working in harmony to reduce the risk of infection. Nutrients 2020, 12, 236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Maslowski, K.M.; Mackay, C.R. Diet, gut microbiota and immune responses. Nat. Immunol. 2011, 12, 5–9. [Google Scholar] [CrossRef] [PubMed]
  64. Sgarbanti, R.; Amatore, D.; Celestino, I.; Elena Marcocci, M.; Fraternale, A.; Ciriolo, M.R.; Magnani, M.; Saladino, R.; Garaci, E.; Teresa Palamara, A. Intracellular redox state as target for anti-influenza therapy: Are antioxidants always effective? Curr. Top. Med. Chem. 2014, 14, 2529–2541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. De Clercq, E.; Li, G. Approved antiviral drugs over the past 50 years. Clin. Microbiol. Rev. 2016, 29, 695–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S. Expert consensus document: 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, 506–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Kobayashi, N.; Saito, T.; Uematsu, T.; Kishi, K.; Toba, M.; Kohda, N.; Suzuki, T. Oral administration of heat-killed lactobacillus pentosus strain b240 augments protection against influenza virus infection in mice. Int. Immunopharmacol. 2011, 11, 199–203. [Google Scholar] [CrossRef]
  68. Nagai, T.; Makino, S.; Ikegami, S.; Itoh, H.; Yamada, H. Effects of oral administration of yogurt fermented with lactobacillus delbrueckii ssp. Bulgaricus oll1073r-1 and its exopolysaccharides against influenza virus infection in mice. Int. Immunopharmacol. 2011, 11, 2246–2250. [Google Scholar] [CrossRef]
  69. Jung, Y.-J.; Lee, Y.-T.; Le Ngo, V.; Cho, Y.-H.; Ko, E.-J.; Hong, S.-M.; Kim, K.-H.; Jang, J.-H.; Oh, J.-S.; Park, M.-K. Heat-killed lactobacillus casei confers broad protection against influenza a virus primary infection and develops heterosubtypic immunity against future secondary infection. Sci. Rep. 2017, 7, 1–12. [Google Scholar] [CrossRef] [Green Version]
  70. Tonetti, F.R.; Islam, M.A.; Vizoso-Pinto, M.G.; Takahashi, H.; Kitazawa, H.; Villena, J. Nasal priming with immunobiotic lactobacilli improves the adaptive immune response against influenza virus. Int. Immunopharmacol. 2020, 78, 106115. [Google Scholar] [CrossRef]
  71. Baron, M. A patented strain of Bacillus coagulans increased immune response to viral challenge. Postgrad. Med. 2009, 121, 114–118. [Google Scholar] [CrossRef]
  72. Shinde, T.; Vemuri, R.; Shastri, M.D.; Perera, A.P.; Tristram, S.; Stanley, R.; Eri, R. Probiotic Bacillus coagulans mtcc 5856 spores exhibit excellent in-vitro functional efficacy in simulated gastric survival, mucosal adhesion and immunomodulation. J. Funct. Foods 2019, 52, 100–108. [Google Scholar] [CrossRef]
  73. Galdeano, C.M.; Cazorla, S.I.; Dumit, J.M.L.; Vélez, E.; Perdigón, G. Beneficial effects of probiotic consumption on the immune system. Ann. Nutr. Metab. 2019, 74, 115–124. [Google Scholar] [CrossRef]
  74. Wieërs, G.; Belkhir, L.; Enaud, R.; Leclercq, S.; Philippart de Foy, J.-M.; Dequenne, I.; de Timary, P.; Cani, P.D. How probiotics affect the microbiota. Front. Cell Infect. Microbiol. 2020, 9, 454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Smelt, M.J.; de Haan, B.J.; Bron, P.A.; van Swam, I.; Meijerink, M.; Wells, J.M.; Faas, M.M.; de Vos, P. Probiotics can generate FoxP3 T-cell responses in the small intestine and simultaneously inducing CD4 and CD8 T cell activation in the large intestine. PLoS ONE 2013, 8, e68952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. de LeBlanc, A.d.M.; Dogi, C.A.; Galdeano, C.M.; Carmuega, E.; Weill, R.; Perdigón, G. Effect of the administration of a fermented milk containing lactobacillus casei dn-114001 on intestinal microbiota and gut associated immune cells of nursing mice and after weaning until immune maturity. BMC Immunol. 2008, 9, 27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Shinde, T.; Perera, A.P.; Vemuri, R.; Gondalia, S.V.; Karpe, A.V.; Beale, D.J.; Shastri, S.; Southam, B.; Eri, R.; Stanley, R. Synbiotic supplementation containing whole plant sugar cane fibre and probiotic spores potentiates protective synergistic effects in mouse model of ibd. Nutrients 2019, 11, 818. [Google Scholar] [CrossRef] [Green Version]
  78. Shinde, T.; Perera, A.P.; Vemuri, R.; Gondalia, S.V.; Beale, D.J.; Karpe, A.V.; Shastri, S.; Basheer, W.; Southam, B.; Eri, R.; et al. Synbiotic supplementation with prebiotic green banana resistant starch and probiotic bacillus coagulans spores ameliorates gut inflammation in mouse model of inflammatory bowel diseases. Eur. J. Nutr. 2020. [Google Scholar] [CrossRef] [Green Version]
  79. Kristensen, N.B.; Bryrup, T.; Allin, K.H.; Nielsen, T.; Hansen, T.H.; Pedersen, O. Alterations in fecal microbiota composition by probiotic supplementation in healthy adults: A systematic review of randomized controlled trials. Genome Med. 2016, 8, 52. [Google Scholar] [CrossRef] [Green Version]
  80. McNulty, N.P.; Yatsunenko, T.; Hsiao, A.; Faith, J.J.; Muegge, B.D.; Goodman, A.L.; Henrissat, B.; Oozeer, R.; Cools-Portier, S.; Gobert, G.; et al. The impact of a consortium of fermented milk strains on the gut microbiome of gnotobiotic mice and monozygotic twins. Sci. Transl. Med. 2011, 3, 106ra106. [Google Scholar] [CrossRef] [Green Version]
  81. Larsen, N.; Vogensen, F.K.; Gøbel, R.; Michaelsen, K.F.; Al-Soud, W.A.; Sørensen, S.J.; Hansen, L.H.; Jakobsen, M. Predominant genera of fecal microbiota in children with atopic dermatitis are not altered by intake of probiotic bacteria Lactobacillus acidophilus ncfm and Bifidobacterium animalis subsp. Lactis bi-07. FEMS Microbiol. Ecol. 2011, 75, 482–496. [Google Scholar] [CrossRef]
  82. Vemuri, R.; Gundamaraju, R.; Shinde, T.; Perera, A.P.; Basheer, W.; Southam, B.; Gondalia, S.V.; Karpe, A.V.; Beale, D.J.; Tristram, S.; et al. Lactobacillus acidophilus dds-1 modulates intestinal-specific microbiota, short-chain fatty acid and immunological profiles in aging mice. Nutrients 2019, 11, 1297. [Google Scholar] [CrossRef] [Green Version]
  83. Tankou, S.K.; Regev, K.; Healy, B.C.; Tjon, E.; Laghi, L.; Cox, L.M.; Kivisäkk, P.; Pierre, I.V.; Hrishikesh, L.; Gandhi, R.; et al. A probiotic modulates the microbiome and immunity in multiple sclerosis. Ann. Neurol. 2018, 83, 1147–1161. [Google Scholar] [CrossRef]
  84. Korpela, K.; Salonen, A.; Vepsäläinen, O.; Suomalainen, M.; Kolmeder, C.; Varjosalo, M.; Miettinen, S.; Kukkonen, K.; Savilahti, E.; Kuitunen, M.; et al. Probiotic supplementation restores normal microbiota composition and function in antibiotic-treated and in caesarean-born infants. Microbiome 2018, 6, 182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Sánchez, B.; Delgado, S.; Blanco-Míguez, A.; Lourenço, A.; Gueimonde, M.; Margolles, A. Probiotics, gut microbiota, and their influence on host health and disease. Mol. Nutr. Food Res. 2017, 61, 1600240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Venegas, D.P.; Marjorie, K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.; Faber, K.N.; Hermoso, M.A. Short chain fatty acids (scfas)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef] [Green Version]
  88. Lehtoranta, L.; Pitkäranta, A.; Korpela, R. Probiotics in respiratory virus infections. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 1289–1302. [Google Scholar] [CrossRef]
  89. Perdigón, G.; Fuller, R.; Raya, R. Lactic acid bacteria and their effect on the immune system. Curr. Issues Intest. Microbiol. 2001, 2, 27–42. [Google Scholar]
  90. Yasui, H.; Kiyoshima, J.; Hori, T. Reduction of influenza virus titer and protection against influenza virus infection in infant mice fed Lactobacillus casei shirota. Clin. Vaccine Immunol. 2004, 11, 675–679. [Google Scholar] [CrossRef] [Green Version]
  91. Takeda, S.; Takeshita, M.; Kikuchi, Y.; Dashnyam, B.; Kawahara, S.; Yoshida, H.; Watanabe, W.; Muguruma, M.; Kurokawa, M. Efficacy of oral administration of heat-killed probiotics from mongolian dairy products against influenza infection in mice: Alleviation of influenza infection by its immunomodulatory activity through intestinal immunity. Int. Immunopharmacol. 2011, 11, 1976–1983. [Google Scholar] [CrossRef]
  92. Park, M.-K.; Vu, N.; Kwon, Y.-M.; Lee, Y.-T.; Yoo, S.; Cho, Y.-H.; Hong, S.-M.; Hwang, H.S.; Ko, E.-J.; Jung, Y.-J. Lactobacillus plantarum dk119 as a probiotic confers protection against influenza virus by modulating innate immunity. PLoS ONE 2013, 8, e75368. [Google Scholar] [CrossRef] [Green Version]
  93. 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]
  94. Waki, N.; Yajima, N.; Suganuma, H.; Buddle, B.; Luo, D.; Heiser, A.; Zheng, T. Oral administration of Lactobacillus brevis kb 290 to mice alleviates clinical symptoms following influenza virus infection. Lett. Appl. Microbiol. 2014, 58, 87–93. [Google Scholar] [CrossRef] [PubMed]
  95. Goto, H.; Sagitani, A.; Ashida, N.; Kato, S.; Hirota, T.; Shinoda, T.; Yamamoto, N. Anti-influenza virus effects of both live and non-live lactobacillus acidophilus l-92 accompanied by the activation of innate immunity. Br. J. Nutr. 2013, 110, 1810–1818. [Google Scholar] [CrossRef] [Green Version]
  96. Iwabuchi, N.; Xiao, J.-Z.; Yaeshima, T.; Iwatsuki, K. Oral administration of Bifidobacterium longum ameliorates influenza virus infection in mice. Biol. Pharm. Bull. 2011, 34, 1352–1355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Harata, G.; He, F.; Hiruta, N.; Kawase, M.; Kubota, A.; Hiramatsu, M.; Yausi, H. Intranasal administration of lactobacillus rhamnosus gg protects mice from h1n1 influenza virus infection by regulating respiratory immune responses. Lett. Appl. Microbiol. 2010, 50, 597–602. [Google Scholar] [CrossRef] [PubMed]
  98. Izumo, T.; Maekawa, T.; Ida, M.; Noguchi, A.; Kitagawa, Y.; Shibata, H.; Yasui, H.; Kiso, Y. Effect of intranasal administration of lactobacillus pentosus s-pt84 on influenza virus infection in mice. Int. Immunopharmacol. 2010, 10, 1101–1106. [Google Scholar] [CrossRef] [PubMed]
  99. Gabryszewski, S.J.; Bachar, O.; Dyer, K.D.; Percopo, C.M.; Killoran, K.E.; Domachowske, J.B.; Rosenberg, H.F. Lactobacillus-mediated priming of the respiratory mucosa protects against lethal pneumovirus infection. J. Immunol. 2011, 186, 1151–1161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Garcia-Crespo, K.E.; Chan, C.C.; Gabryszewski, S.J.; Percopo, C.M.; Rigaux, P.; Dyer, K.D.; Domachowske, J.B.; Rosenberg, H.F. Lactobacillus priming of the respiratory tract: Heterologous immunity and protection against lethal pneumovirus infection. Antivir. Res. 2013, 97, 270–279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Hori, T.; Kiyoshima, J.; Shida, K.; Yasui, H. Augmentation of cellular immunity and reduction of influenza virus titer in aged mice fed lactobacillus casei strain shirota. Clin. Vaccine Immunol. 2002, 9, 105–108. [Google Scholar] [CrossRef] [Green Version]
  102. Yasui, H.; Kiyoshima, J.; Hori, T.; Shida, K. Protection against influenza virus infection of mice fed bifidobacterium breve yit4064. Clin. Vaccine Immunol. 1999, 6, 186–192. [Google Scholar] [CrossRef] [Green Version]
  103. Lee, Y.-N.; Youn, H.-N.; Kwon, J.-H.; Lee, D.-H.; Park, J.-K.; Yuk, S.-S.; Erdene-Ochir, T.-O.; Kim, K.-T.; Lee, J.-B.; Park, S.-Y. Sublingual administration of Lactobacillus rhamnosus affects respiratory immune responses and facilitates protection against influenza virus infection in mice. Antivir. Res. 2013, 98, 284–290. [Google Scholar] [CrossRef]
  104. Nguyen, T.L.A.; Vieira-Silva, S.; Liston, A.; Raes, J. How informative is the mouse for human gut microbiota research? Dis. Model. Mech. 2015, 8, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Tiderencel, K.A.; Hutcheon, D.A.; Ziegler, J. Probiotics for the treatment of type 2 diabetes: A review of randomized controlled trials. Diabetes Metab. Res. Rev. 2020, 36, e3213. [Google Scholar] [CrossRef] [PubMed]
  106. Cerdó, T.; García-Santos, J.A.; Bermúdez, M.G.; Campoy, C. The role of probiotics and prebiotics in the prevention and treatment of obesity. Nutrients 2019, 11, 635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Celiberto, L.S.; Bedani, R.; Rossi, E.A.; Cavallini, D.C.U. Probiotics: The scientific evidence in the context of inflammatory bowel disease. Crit. Rev. Food Sci. Nutr. 2017, 57, 1759–1768. [Google Scholar] [CrossRef] [PubMed]
  108. Helmink, B.A.; Khan, M.W.; Hermann, A.; Gopalakrishnan, V.; Wargo, J.A. The microbiome, cancer, and cancer therapy. Nat. Med. 2019, 25, 377–388. [Google Scholar] [CrossRef]
  109. Miller, L.E.; Lehtoranta, L.; Lehtinen, M.J. The effect of bifidobacterium animalis ssp. Lactis hn019 on cellular immune function in healthy elderly subjects: Systematic review and meta-analysis. Nutrients 2017, 9, 191. [Google Scholar] [CrossRef]
  110. Shaw, A.C.; Joshi, S.; Greenwood, H.; Panda, A.; Lord, J.M. Aging of the innate immune system. Curr. Opin. Immunol. 2010, 22, 507–513. [Google Scholar] [CrossRef] [Green Version]
  111. Mohr, A.E.; Basile, A.J.; Crawford, M.s.S.; Sweazea, K.L.; Carpenter, K.C. Probiotic supplementation has a limited effect on circulating immune and inflammatory markers in healthy adults: A systematic review of randomized controlled trials. J. Acad. Nutr. Dietet. 2020, 120, 548–564. [Google Scholar] [CrossRef]
  112. Khalesi, S.; Bellissimo, N.; Vandelanotte, C.; Williams, S.; Stanley, D.; Irwin, C. A review of probiotic supplementation in healthy adults: Helpful or hype? Eur. J. Clin. Nutr. 2019, 73, 24–37. [Google Scholar] [CrossRef]
  113. King, S.; Glanville, J.; Sanders, M.E.; Fitzgerald, A.; Varley, D. Effectiveness of probiotics on the duration of illness in healthy children and adults who develop common acute respiratory infectious conditions: A systematic review and meta-analysis. Br. J. Nutr. 2014, 112, 41–54. [Google Scholar] [CrossRef]
  114. Hazeldine, J.; Lord, J.M. The impact of ageing on natural killer cell function and potential consequences for health in older adults. Ageing Res. Rev. 2013, 12, 1069–1078. [Google Scholar] [CrossRef] [PubMed]
  115. Rukavina, D.; Laskarin, G.; Rubesa, G.; Strbo, N.; Bedenicki, I.; Manestar, D.; Glavas, M.; Christmas, S.E.; Podack, E.R. Age-related decline of perforin expression in human cytotoxic T lymphocytes and natural killer cells. Blood 1998, 92, 2410–2420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Hayhoe, R.P.G.; Henson, S.M.; Akbar, A.N.; Palmer, D.B. Variation of human natural killer cell phenotypes with age: Identification of a unique klrg1-negative subset. Hum. Immunol. 2010, 71, 676–681. [Google Scholar] [CrossRef]
  117. Ouwehand, A.C.; Bergsma, N.; Parhiala, R.; Lahtinen, S.; Gueimonde, M.; Finne-Soveri, H.; Strandberg, T.; Pitkälä, K.; Salminen, S. Bifidobacterium microbiota and parameters of immune function in elderly subjects. FEMS Immunol. Med. Microbiol. 2008, 53, 18–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Nyangale, E.P.; Farmer, S.; Cash, H.A.; Keller, D.; Chernoff, D.; Gibson, G.R. Bacillus coagulans gbi-30, 6086 modulates Faecalibacterium prausnitzii in older men and women. J. Nutr. 2015, 145, 1446–1452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Alameddine, J.; Godefroy, E.; Papargyris, L.; Sarrabayrouse, G.; Tabiasco, J.; Bridonneau, C.; Yazdanbakhsh, K.; Sokol, H.; Altare, F.; Jotereau, F. Faecalibacterium prausnitzii skews human dc to prime il10-producing t cells through tlr2/6/jnk signaling and il-10, il-27, cd39, and ido-1 induction. Front. Immunol. 2019, 10. [Google Scholar] [CrossRef] [Green Version]
  120. Boge, T.; Rémigy, M.; Vaudaine, S.; Tanguy, J.; Bourdet-Sicard, R.; van der Werf, S. A probiotic fermented dairy drink improves antibody response to influenza vaccination in the elderly in two randomised controlled trials. Vaccine 2009, 27, 5677–5684. [Google Scholar] [CrossRef]
  121. Bosch, M.; Méndez, M.; Pérez, M.; Farran, A.; Fuentes, M.C.; Cuñé, J. Lactobacillus plantarum cect7315 and cect7316 stimulate immunoglobulin production after influenza vaccination in elderly. Nutr. Hosp. 2012, 27, 504–509. [Google Scholar] [CrossRef]
  122. Gutierrez-Castrellon, P.; Weizman, Z.; Cruchet, S.; Dinleyci, E.C.; Jimenez-Gutierrez, C.; Lopez-Velazquez, G. Role of probiotics to prevent and reduce the duration of upper respiratory infections in ambulatory children: Systematic review with network-meta analysis. Preprints 2018. [Google Scholar] [CrossRef]
  123. Hojsak, I.; Snovak, N.; Abdović, S.; Szajewska, H.; Mišak, Z.; Kolaček, S. Lactobacillus gg in the prevention of gastrointestinal and respiratory tract infections in children who attend day care centers: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2010, 29, 312–316. [Google Scholar] [CrossRef]
  124. 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 2018, 29, 494–499. [Google Scholar] [CrossRef] [PubMed]
  125. Laursen, R.P.; Larnkjær, A.; Ritz, C.; Hauger, H.; Michaelsen, K.F.; Mølgaard, C. Probiotics and child care absence due to infections: A randomized controlled trial. Pediatrics 2017, 140, e20170735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Laursen, R.P.; Hojsak, I. Probiotics for respiratory tract infections in children attending day care centers—A systematic review. Eur. J. Pediatr. 2018, 177, 979–994. [Google Scholar] [CrossRef] [PubMed]
  127. Skórka, A.; Piescik-Lech, M.; Kolodziej, M.; Szajewska, H. To add or not to add probiotics to infant formulae? An updated systematic review. Benef. Microbes 2017, 8, 717–725. [Google Scholar] [CrossRef]
  128. Xiao, L.; Gong, C.; Ding, Y.; Ding, G.; Xu, X.; Deng, C.; Ze, X.; Malard, P.; Ben, X. Probiotics maintain intestinal secretory immunoglobulin a levels in healthy formula-fed infants: A randomised, double-blind, placebo-controlled study. Benef. Microbes 2019, 10, 729–739. [Google Scholar] [CrossRef]
  129. Cacho, N.T.; Lawrence, R.M. Innate immunity and breast milk. Front. Immunol. 2017, 8. [Google Scholar] [CrossRef] [Green Version]
  130. Moore, L.V.; Thompson, F.E. Adults meeting fruit and vegetable intake recommendations—United States, 2013. MMWR Morb. Mortal Wkly. Rep. 2015, 64, 709–713. [Google Scholar]
  131. Nour, M.; Sui, Z.; Grech, A.; Rangan, A.; McGeechan, K.; Allman-Farinelli, M. The fruit and vegetable intake of young australian adults: A population perspective. Public Health Nutr. 2017, 20, 2499–2512. [Google Scholar] [CrossRef] [Green Version]
  132. Wood, L.G.; Li, Q.; Scott, H.A.; Rutting, S.; Berthon, B.S.; Gibson, P.G.; Hansbro, P.M.; Williams, E.; Horvat, J.; Simpson, J.L. Saturated fatty acids, obesity, and the nucleotide oligomerization domain–like receptor protein 3 (nlrp3) inflammasome in asthmatic patients. J. Allergy Clin. Immunol. 2019, 143, 305–315. [Google Scholar] [CrossRef] [Green Version]
  133. Peeri, N.C.; Shrestha, N.; Rahman, M.S.; Zaki, R.; Tan, Z.; Bibi, S.; Baghbanzadeh, M.; Aghamohammadi, N.; Zhang, W.; Haque, U. The SARS, MERS and novel coronavirus (Covid-19) epidemics, the newest and biggest global health threats: What lessons have we learned? Int. J. Epidemiol. 2020. [Google Scholar] [CrossRef] [Green Version]
  134. Zhou, F.; Yu, T.; Du, R.; Fan, G.; Liu, Y.; Liu, Z.; Xiang, J.; Wang, Y.; Song, B.; Gu, X.; et al. Clinical course and risk factors for mortality of adult inpatients with covid-19 in wuhan, china: A retrospective cohort study. Lancet 2020, 395, 1054–1062. [Google Scholar] [CrossRef]
  135. Kim, H.; Caulfield Laura, E.; Garcia-Larsen, V.; Steffen Lyn, M.; Coresh, J.; Rebholz Casey, M. Plant-based diets are associated with a lower risk of incident cardiovascular disease, cardiovascular disease mortality, and all-cause mortality in a general population of middle-aged adults. J. Am. Heart Assoc. 2019, 8, e012865. [Google Scholar] [CrossRef] [PubMed]
  136. Kopf, J.C.; Suhr, M.J.; Clarke, J.; Eyun, S.-i.; Riethoven, J.-J.M.; Ramer-Tait, A.E.; Rose, D.J. Role of whole grains versus fruits and vegetables in reducing subclinical inflammation and promoting gastrointestinal health in individuals affected by overweight and obesity: A randomized controlled trial. Nutr. J. 2018, 17, 72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Li, F.; Liu, X.; Wang, W.; Zhang, D. Consumption of vegetables and fruit and the risk of inflammatory bowel disease: A meta-analysis. Eur. J. Gastroenterol. Hepatol. 2015, 27, 623–630. [Google Scholar] [CrossRef]
  138. Cui, J.; Lian, Y.; Zhao, C.; Du, H.; Han, Y.; Gao, W.; Xiao, H.; Zheng, J. Dietary fibers from fruits and vegetables and their health benefits via modulation of gut microbiota. Compr. Rev. Food Sci. F. 2019, 18, 1514–1532. [Google Scholar] [CrossRef] [Green Version]
  139. Park, Y.; Subar, A.F.; Hollenbeck, A.; Schatzkin, A. Dietary fiber intake and mortality in the nih-aarp diet and health study. Arch. Intern. Med. 2011, 171, 1061–1068. [Google Scholar] [CrossRef] [Green Version]
  140. Hanson, C.; Lyden, E.; Rennard, S.; Mannino, D.M.; Rutten, E.P.; Hopkins, R.; Young, R. The relationship between dietary fiber intake and lung function in the national health and nutrition examination surveys. Ann. Am. Thorac. Soc. 2016, 13, 643–650. [Google Scholar] [CrossRef] [PubMed]
  141. Langkamp-Henken, B.; Wood, S.M.; Herlinger-Garcia, K.A.; Thomas, D.J.; Stechmiller, J.K.; Bender, B.S.; Gardner, E.M.; DeMichele, S.J.; Schaller, J.P.; Murasko, D.M. Nutritional formula improved immune profiles of seniors living in nursing homes. J. Am. Geriatr. Soc. 2006, 54, 1861–1870. [Google Scholar] [CrossRef]
  142. Langkamp-henken, B.; Bender, B.S.; Gardner, E.M.; Herrlinger-garcia, K.A.; Kelley, M.J.; Murasko, D.M.; Schaller, J.P.; Stechmiller, J.K.; Thomas, D.J.; Wood, S.M. Nutritional formula enhanced immune function and reduced days of symptoms of upper respiratory tract infection in seniors. J. Am. Geriatr. Soc. 2004, 52, 3–12. [Google Scholar] [CrossRef]
  143. Bunout, D.; Hirsch, S.; de la Maza, M.; Munoz, C.; Haschke, F.; Steenhout, P.; Klassen, P.; Barrera, G.; Gattas, V.; Petermann, M. Effects of prebiotics on the immune response to vaccination in the elderly. J. Parenter. Enteral Nutr. 2002, 26, 372–376. [Google Scholar] [CrossRef]
  144. Lomax, A.R.; Cheung, L.V.Y.; Noakes, P.S.; Miles, E.A.; Calder, P.C. Inulin-type β2-1 fructans have some effect on the antibody response to seasonal influenza vaccination in healthy middle-aged humans. Front. Immunol. 2015, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Trompette, A.; Gollwitzer, E.S.; Yadava, K.; Sichelstiel, A.K.; Sprenger, N.; Ngom-Bru, C.; Blanchard, C.; Junt, T.; Nicod, L.P.; Harris, N.L. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 2014, 20, 159–166. [Google Scholar] [CrossRef] [PubMed]
  146. Kamp, M.E.; Shim, R.; Nicholls, A.J.; Oliveira, A.C.; Mason, L.J.; Binge, L.; Mackay, C.R.; Wong, C.H.Y. G protein-coupled receptor 43 modulates neutrophil recruitment during acute inflammation. PLoS ONE 2016, 11, e0163750. [Google Scholar] [CrossRef]
  147. Ang, Z.; Ding, J.L. Gpr41 and gpr43 in obesity and inflammation - protective or causative? Front. Immunol. 2016, 7, 28. [Google Scholar] [CrossRef] [Green Version]
  148. Baxter, N.T.; Schmidt, A.W.; Venkataraman, A.; Kim, K.S.; Waldron, C.; Schmidt, T.M. Dynamics of human gut microbiota and short-chain fatty acids in response to dietary interventions with three fermentable fibers. MBio 2019, 10, e02566–02518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Williams, B.A.; Grant, L.J.; Gidley, M.J.; Mikkelsen, D. Gut fermentation of dietary fibres: Physico-chemistry of plant cell walls and implications for health. Int. J. Mol. Sci. 2017, 18, 2203. [Google Scholar] [CrossRef] [Green Version]
  150. Kolida, S.; Gibson, G.R. Synbiotics in health and disease. Annu. Rev. Food Sci. Technol. 2011, 2, 373–393. [Google Scholar] [CrossRef] [Green Version]
  151. Weiss, G.; Christensen, H.R.; Zeuthen, L.H.; Vogensen, F.K.; Jakobsen, M.; Frøkiær, H. Lactobacilli and bifidobacteria induce differential interferon-β profiles in dendritic cells. Cytokine 2011, 56, 520–530. [Google Scholar] [CrossRef]
  152. Donohoe, D.R.; Holley, D.; Collins, L.B.; Montgomery, S.A.; Whitmore, A.C.; Hillhouse, A.; Curry, K.P.; Renner, S.W.; Greenwalt, A.; Ryan, E.P.; et al. A gnotobiotic mouse model demonstrates that dietary fiber protects against colorectal tumorigenesis in a microbiota- and butyrate-dependent manner. Cancer Discov. 2014, 4, 1387–1397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Akatsu, H.; Nagafuchi, S.; Kurihara, R.; Okuda, K.; Kanesaka, T.; Ogawa, N.; Kanematsu, T.; Takasugi, S.; Yamaji, T.; Takami, M.; et al. Enhanced vaccination effect against influenza by prebiotics in elderly patients receiving enteral nutrition. Geriat. Gerontol. Int. 2016, 16, 205–213. [Google Scholar] [CrossRef]
  154. Przemska-Kosicka, A.; Childs, C.E.; Maidens, C.; Dong, H.; Todd, S.; Gosney, M.A.; Tuohy, K.M.; Yaqoob, P. Age-related changes in the natural killer cell response to seasonal influenza vaccination are not influenced by a synbiotic: A randomised controlled trial. Front. Immunol. 2018, 9, 591. [Google Scholar] [CrossRef] [PubMed]
  155. Enani, S.; Przemska-Kosicka, A.; Childs, C.E.; Maidens, C.; Dong, H.; Conterno, L.; Tuohy, K.; Todd, S.; Gosney, M.; Yaqoob, P. Impact of ageing and a synbiotic on the immune response to seasonal influenza vaccination; a randomised controlled trial. Clin. Nutr. 2018, 37, 443–451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Przemska-Kosicka, A.; Childs, C.E.; Enani, S.; Maidens, C.; Dong, H.; Dayel, I.B.; Tuohy, K.; Todd, S.; Gosney, M.A.; Yaqoob, P. Effect of a synbiotic on the response to seasonal influenza vaccination is strongly influenced by degree of immunosenescence. Immun. Ageing 2016, 13. [Google Scholar] [CrossRef] [Green Version]
  157. Hickson, L.J.; Langhi Prata, L.G.P.; Bobart, S.A.; Evans, T.K.; Giorgadze, N.; Hashmi, S.K.; Herrmann, S.M.; Jensen, M.D.; Jia, Q.; Jordan, K.L.; et al. Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of dasatinib plus quercetin in individuals with diabetic kidney disease. EBioMedicine 2019, 47, 446–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Thoppil, H.; Riabowol, K. Senolytics: A translational bridge between cellular senescence and organismal aging. Front. Cell Dev. Biol. 2020, 7, 367. [Google Scholar] [CrossRef]
  159. Li, W.; Qin, L.; Feng, R.; Hu, G.; Sun, H.; He, Y.; Zhang, R. Emerging senolytic agents derived from natural products. Mech. Ageing Dev. 2019, 181, 1–6. [Google Scholar] [CrossRef] [PubMed]
  160. Kim, C.H.; Park, J.; Kim, M. Gut microbiota-derived short-chain fatty acids, T cells, and inflammation. Immune Netw. 2014, 14, 277–288. [Google Scholar] [CrossRef] [Green Version]
  161. Schulthess, J.; Pandey, S.; Capitani, M.; Rue-Albrecht, K.C.; Arnold, I.; Franchini, F.; Chomka, A.; Ilott, N.E.; Johnston, D.G.; Pires, E. The short chain fatty acid butyrate imprints an antimicrobial program in macrophages. Immunity 2019, 50, 432–445.e437. [Google Scholar] [CrossRef] [Green Version]
  162. Li, N.; Liu, X.-X.; Hong, M.; Huang, X.-Z.; Chen, H.; Xu, J.-H.; Wang, C.; Zhang, Y.-X.; Zhong, J.-X.; Nie, H. Sodium butyrate alleviates lps-induced acute lung injury in mice via inhibiting hmgb1 release. Int. Immunopharmacol. 2018, 56, 242–248. [Google Scholar] [CrossRef]
  163. Majeed, M.; Nagabhushanam, K.; Arumugam, S.; Natarajan, S.; Majeed, S.; Pande, A.; Beede, K.; Ali, F. Cranberry seed fibre: A promising prebiotic fibre and its fermentation by the probiotic bacillus coagulans mtcc 5856. Int. J. Food Sci. Technol. 2018, 53, 1640–1647. [Google Scholar] [CrossRef] [Green Version]
  164. Majeed, M.; Majeed, S.; Nagabhushanam, K.; Arumugam, S.; Natarajan, S.; Beede, K.; Ali, F. Galactomannan from trigonella foenum-graecum l. Seed: Prebiotic application and its fermentation by the probiotic bacillus coagulans strain mtcc 5856. Food Sci. Nutr. 2018, 6, 666–673. [Google Scholar] [CrossRef] [PubMed]
  165. Walton, S.L.; Bischoff, K.M.; van Heiningen, A.R.; van Walsum, G.P. Production of lactic acid from hemicellulose extracts by Bacillus coagulans MXL-9. J. Ind. Microbiol. Biotechnol. 2010, 37, 823–830. [Google Scholar] [CrossRef] [PubMed]
  166. Nyangale, E.P.; Farmer, S.; Keller, D.; Chernoff, D.; Gibson, G.R. Effect of prebiotics on the fecal microbiota of elderly volunteers after dietary supplementation of bacillus coagulans gbi-30, 6086. Anaerobe 2014, 30, 75–81. [Google Scholar] [CrossRef] [PubMed]
  167. Trachsel, J.; Briggs, C.; Gabler, N.K.; Allen, H.K.; Loving, C.L. Dietary resistant potato starch alters intestinal microbial communities and their metabolites, and markers of immune regulation and barrier function in swine. Front. Immunol. 2019, 10. [Google Scholar] [CrossRef] [Green Version]
  168. Denaro, M.; Smeriglio, A.; Barreca, D.; De Francesco, C.; Occhiuto, C.; Milano, G.; Trombetta, D. Antiviral activity of plants and their isolated bioactive compounds: An update. Phytother. Res. 2020, 34, 742–768. [Google Scholar] [CrossRef]
  169. Calder, P.C.; Carr, A.C.; Gombart, A.F.; Eggersdorfer, M. Optimal nutritional status for a well-functioning immune system is an important factor to protect against viral infections. Nutrients 2020, 12, 1181. [Google Scholar] [CrossRef] [Green Version]
  170. Bahramsoltani, R.; Sodagari, H.R.; Farzaei, M.H.; Abdolghaffari, A.H.; Gooshe, M.; Rezaei, N. The preventive and therapeutic potential of natural polyphenols on influenza. Expert Rev. Anti. Infect. Ther. 2016, 14, 57–80. [Google Scholar] [CrossRef]
  171. Mohammadi Pour, P.; Fakhri, S.; Asgary, S.; Farzaei, M.H.; Echeverría, J. The signaling pathways, and therapeutic targets of antiviral agents: Focusing on the antiviral approaches and clinical perspectives of anthocyanins in the management of viral diseases. Front. Pharmacol. 2019, 10, 1207. [Google Scholar] [CrossRef] [Green Version]
  172. Kawabata, K.; Yoshioka, Y.; Terao, J. Role of intestinal microbiota in the bioavailability and physiological functions of dietary polyphenols. Molecules 2019, 24, 370. [Google Scholar] [CrossRef] [Green Version]
  173. Pae, M.; Wu, D. Immunomodulating effects of epigallocatechin-3-gallate from green tea: Mechanisms and applications. Food Funct. 2013, 4, 1287–1303. [Google Scholar] [CrossRef]
  174. Xu, M.-J.; Liu, B.-J.; Wang, C.-L.; Wang, G.-H.; Tian, Y.; Wang, S.-H.; Li, J.; Li, P.-Y.; Zhang, R.-H.; Wei, D.; et al. Epigallocatechin-3-gallate inhibits tlr4 signaling through the 67-kda laminin receptor and effectively alleviates acute lung injury induced by h9n2 swine influenza virus. Int. Immunopharmacol. 2017, 52, 24–33. [Google Scholar] [CrossRef] [PubMed]
  175. Kim, Y.H.; Won, Y.-S.; Yang, X.; Kumazoe, M.; Yamashita, S.; Hara, A.; Takagaki, A.; Goto, K.; Nanjo, F.; Tachibana, H. Green tea catechin metabolites exert immunoregulatory effects on cd4+ t cell and natural killer cell activities. J. Agric. Food Chem. 2016, 64, 3591–3597. [Google Scholar] [CrossRef] [PubMed]
  176. Ozdal, T.; Sela, D.A.; Xiao, J.; Boyacioglu, D.; Chen, F.; Capanoglu, E. The reciprocal interactions between polyphenols and gut microbiota and effects on bioaccessibility. Nutrients 2016, 8, 78. [Google Scholar] [CrossRef] [PubMed]
  177. Nash, V.; Ranadheera, C.S.; Georgousopoulou, E.N.; Mellor, D.D.; Panagiotakos, D.B.; McKune, A.J.; Kellett, J.; Naumovski, N. The effects of grape and red wine polyphenols on gut microbiota—A systematic review. Food Res. Int. 2018, 113, 277–287. [Google Scholar] [CrossRef]
Figure 1. Immunopathological features and risk factors associated with viral respiratory infections.
Figure 1. Immunopathological features and risk factors associated with viral respiratory infections.
Microorganisms 08 00921 g001
Figure 2. Potential mechanism by which synergistic synbiotic-induced short-chain fatty acids (SCFAs) can regulate the innate and acquired immune responses to attenuate viral respiratory infection.
Figure 2. Potential mechanism by which synergistic synbiotic-induced short-chain fatty acids (SCFAs) can regulate the innate and acquired immune responses to attenuate viral respiratory infection.
Microorganisms 08 00921 g002

Share and Cite

MDPI and ACS Style

Shinde, T.; Hansbro, P.M.; Sohal, S.S.; Dingle, P.; Eri, R.; Stanley, R. Microbiota Modulating Nutritional Approaches to Countering the Effects of Viral Respiratory Infections Including SARS-CoV-2 through Promoting Metabolic and Immune Fitness with Probiotics and Plant Bioactives. Microorganisms 2020, 8, 921. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms8060921

AMA Style

Shinde T, Hansbro PM, Sohal SS, Dingle P, Eri R, Stanley R. Microbiota Modulating Nutritional Approaches to Countering the Effects of Viral Respiratory Infections Including SARS-CoV-2 through Promoting Metabolic and Immune Fitness with Probiotics and Plant Bioactives. Microorganisms. 2020; 8(6):921. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms8060921

Chicago/Turabian Style

Shinde, Tanvi, Philip M Hansbro, Sukhwinder Singh Sohal, Peter Dingle, Rajaraman Eri, and Roger Stanley. 2020. "Microbiota Modulating Nutritional Approaches to Countering the Effects of Viral Respiratory Infections Including SARS-CoV-2 through Promoting Metabolic and Immune Fitness with Probiotics and Plant Bioactives" Microorganisms 8, no. 6: 921. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms8060921

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