Next Article in Journal
Effects of HCV Clearance with Direct-Acting Antivirals (DAAs) on Liver Stiffness, Liver Fibrosis Stage and Metabolic/Cellular Parameters
Next Article in Special Issue
Scoping Review of Clinical Presentations and Outcomes in Patients with Concomitant COVID-19 Infection and Acute Mesenteric Ischaemia
Previous Article in Journal
Antiviral and Virucidal Activities of Uncaria tomentosa (Cat’s Claw) against the Chikungunya Virus
Previous Article in Special Issue
A Systematic Review of the Prevalence of Persistent Gastrointestinal Symptoms and Incidence of New Gastrointestinal Illness after Acute SARS-CoV-2 Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Interplay between COVID-19 and Gut Health: The Potential Role of Prebiotics and Probiotics in Immune Support

by
Marta Giovanetti
1,2,3,*,
Gianfranco Pannella
1,4,
Annamaria Altomare
1,5,
Giulia Rocchi
5,
Michele Guarino
5,6,
Massimo Ciccozzi
7,
Elisabetta Riva
8,9 and
Giovanni Gherardi
9,*
1
Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
2
Climate Amplified Diseases and Epidemics (CLIMADE), Brasilia 70070-130, Brazil
3
Instituto Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil
4
Department of Agricultural, Enviromental and Food Science, University of Molise, 86100 Campobasso, Italy
5
Research Unit of Gastroenterology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
6
Operative Research Unit of Gastroenterology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
7
Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, 00128 Roma, Italy
8
Unit of Virology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
9
Applied Bacteriological Sciences Unit, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
*
Authors to whom correspondence should be addressed.
Viruses 2024, 16(3), 370; https://doi.org/10.3390/v16030370
Submission received: 15 January 2024 / Revised: 23 February 2024 / Accepted: 26 February 2024 / Published: 27 February 2024
(This article belongs to the Special Issue COVID-19 and Gastrointestinal Symptoms)

Abstract

:
The COVID-19 pandemic has profoundly impacted global health, leading to extensive research focused on developing strategies to enhance outbreak response and mitigate the disease’s severity. In the aftermath of the pandemic, attention has shifted towards understanding and addressing long-term health implications, particularly in individuals experiencing persistent symptoms, known as long COVID. Research into potential interventions to alleviate long COVID symptoms has intensified, with a focus on strategies to support immune function and mitigate inflammation. One area of interest is the gut microbiota, which plays a crucial role in regulating immune responses and maintaining overall health. Prebiotics and probiotics, known for their ability to modulate the gut microbiota, have emerged as potential therapeutic agents in bolstering immune function and reducing inflammation. This review delves into the intricate relationship between long COVID, the gut microbiota, and immune function, with a specific focus on the role of prebiotics and probiotics. We examine the immune response to long COVID, emphasizing the importance of inflammation and immune regulation in the persistence of symptoms. The potential of probiotics in modulating immune responses, including their mechanisms in combating viral infections such as COVID-19, is discussed in detail. Clinical evidence supporting the use of probiotics in managing long COVID symptoms is summarized, highlighting their role as adjunctive therapy in addressing various aspects of SARS-CoV-2 infection and its aftermath.

1. Introduction

1.1. Background on the COVID-19 Pandemic and Its Impact on Global Health

The COVID-19 pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has dramatically impacted global health, leading to extensive illness, death, and significant societal and economic disruptions [1]. First identified in Wuhan, China, in December 2019, the SARS-CoV-2 is a novel coronavirus and part of a larger family that includes pathogens causing mild conditions like the common cold as well as severe diseases such as Middle East Respiratory Syndrome (MERS) and Severe Acute Respiratory Syndrome (SARS) [2]. Transmission primarily occurs through respiratory droplets during coughing, sneezing, or talking, though contact with contaminated surfaces is also a possible, albeit secondary, route [3,4].
The symptoms of COVID-19 vary widely, ranging from mild respiratory issues to severe complications like pneumonia, acute respiratory distress syndrome, organ failure, and death, especially in older individuals and those with pre-existing conditions [5]. Common symptoms include fever, cough, and breathing difficulties. Uniquely, the virus can also cause atypical symptoms affecting various body systems, such as gastrointestinal infections, diarrhea, and ulcerative colitis. Notably, patients with gastrointestinal manifestations often experience more severe respiratory complications [6], possibly due to a microbial imbalance characterized by a reduction in beneficial bacteria like Lactobacillus and Bifidobacterium [7].
The pathogenesis of COVID-19 begins when the SARS-CoV-2 virus binds to angiotensin-converting enzyme 2 (ACE2) receptors found on human cells, predominantly in the lungs, heart, kidneys, and intestines (Figure 1) [8]. This interaction is critical to understanding the disease’s progression and its diverse impact on human health.
The virus’s spike protein plays a crucial role in facilitating its binding to host cells. Upon attachment, the virus fuses with the host cell membrane, a process potentially aided by cellular enzymes, leading to the release of its RNA into the cytoplasm [9]. The host cell then employs its own mechanisms to translate this viral RNA into two large polypeptides, which the virus’s proteases further cleave into smaller, functional proteins. Among these, some form the replication–transcription complex (RTC), vital for the replication of viral RNA. Subsequently, the newly synthesized viral RNA and structural proteins are transported to the endoplasmic reticulum and Golgi apparatus, where they assemble into new virus particles [9]. These virions then bud into the lumen of the ER–Golgi intermediate compartment (ERGIC), get encased in vesicles, and are finally released from the host cell through exocytosis [9]. The virus triggers an immune response which is essential for combating the infection but can also cause tissue damage. In severe cases, this response may become hyperactive, leading to a cytokine storm, potentially resulting in acute respiratory distress syndrome (ARDS) and multi-organ failure [10]. Understanding these mechanisms is key in developing effective treatments and preventative strategies. Beyond the immediate health impact, COVID-19 has profoundly affected global healthcare systems, disrupted routine medical services, and exerted a psychological and socioeconomic toll due to containment measures like lockdowns and travel restrictions. The pandemic has highlighted the interconnectedness of global health and the importance of international collaboration in facing such challenges.

1.2. Overview of the Immune Response to COVID-19 and the Role of Inflammation

The immune response to COVID-19 plays a crucial role in determining the course and severity of the disease [11]. When the body is infected by SARS-CoV-2, it triggers an immune response starting with the innate immune system, the body’s first line of defense [12]. This system includes physical barriers such as the skin and mucosal linings and immune cells like macrophages and dendritic cells that respond to pathogens in a non-specific way [13]. The presence of the virus activates these innate immune cells, which attempt to eliminate it through processes like phagocytosis and the release of various cytokines. These cytokines are critical in managing the infection but can also lead to inflammation [14]. SARS-CoV-2 has developed mechanisms to evade immune detection, including reducing the interferon (IFN) response, leading to lower levels of type I and II IFNs and IFN-stimulated genes (ISGs) in the early stages of infection [14]. IFNs typically help clear infections by promoting ISG transcription and producing antiviral proteins and cytokines. However, in severe COVID-19 cases, dysregulated cytokine and IFN feedback loops can exacerbate the cytokine storm, resulting in hyperinflammation, multi-organ failure, and death [11,12,13,14]. Following the innate response, the adaptive immune system kicks in, offering a more targeted attack against the virus. This includes B cells producing antibodies to neutralize the virus and prevent cell invasion. T cells, another component of the adaptive system, can destroy virus-infected cells and help coordinate the immune response. A key factor in severe COVID-19 cases is the overproduction of cytokines, leading to the notorious ‘cytokine storm’ [15]. This excessive inflammatory response can cause significant tissue damage, ARDS, and multi-organ failure [16], contributing to the high morbidity and mortality rates in severe COVID-19 cases. Moreover, the immune response to COVID-19 varies significantly among individuals. Some exhibit efficient responses, eliminating the virus with minimal symptoms, while others have delayed or inadequate responses, leading to severe illness. The reasons for this variability are not fully understood but may include genetic factors, pre-existing health conditions, age, and possibly previous exposure to other coronaviruses (though this aspect remains under debate). Understanding the intricate details of the immune response to COVID-19, particularly the role of inflammation, is vital in developing effective treatments and preventive strategies.

1.3. Overview of the Importance of the Immune System and Gut Microbiota in Maintaining Health

The immune system and gut microbiota are fundamental in maintaining overall health, each playing a critical role in disease prevention and normal bodily functions [17]. The immune system functions as the body’s protective mechanism against pathogens and other potential dangers. The immune system carries out a sequence of actions called the immune response to combat invading organisms and substances that infiltrate the body and lead to sickness [18]. This system comprised an intricate network of cells, tissues, and organs working together to protect the organism. Central to the immune response are leukocytes, or white blood cells, which are categorized into two main types: innate immune cells, the body’s first line of defense, and adaptive immune cells, which provide a more specific response to particular pathogens [17,18,19]. The gut microbiota, consisting of a diverse array of microorganisms like bacteria, viruses, fungi, and protozoa inhabiting the gastrointestinal system, significantly influences the body’s immune response. An optimal gut microbiota is crucial for immune system maturation, maintaining its balance, and protecting the host from harmful microbes [19]. These microorganisms aid in immune cell development, the production of antimicrobial substances, nutrient breakdown and absorption, vitamin synthesis, and the generation of short-chain fatty acids, vital for gut health [17,18,19]. The interaction between gut bacteria and the immune system is complex and ongoing, involving a mutual exchange where the microbiota helps maintain tolerance towards beneficial microbes and enables the immune response against pathogens. This communication is facilitated by microbial-associated molecular patterns (MAMPs) recognized by pattern recognition receptors (PRRs) on immune cells [20]. Dysbiosis, or an imbalance in the gut microbiota composition, has been linked to various conditions, including inflammatory bowel disease, functional gastrointestinal disorders, viral infections, allergies, obesity, and even mental health issues [21]. Thus, a healthy gut microbiota is crucial not just for gastrointestinal health but also for broader health implications. Maintaining a healthy gut involves mindful dietary choices, considering lifestyle factors like physical activity, smoking, and alcohol consumption, and avoiding unnecessary antibiotic use, which can disrupt microbial balance [21]. Probiotics and prebiotics are used to improve gut microbiota composition, potentially enhancing immune function and overall health [21].

2. Prebiotics and Probiotics

Clear definitions are vital for terms such as ‘probiotic’, ‘prebiotic’, ‘synbiotic’, and other terms recently introduced in the functional foods’ domain [22,23]. Probiotics, a key focus in this field, are live microorganisms that, when consumed in adequate amounts, confer health benefits to the host. Werner Kollath was the first to use the term ‘probiotic’ in 1953, defining it as organisms that significantly contribute to health improvement [23,24,25]. The FAO and WHO describe probiotics as live microorganisms that, when ingested in sufficient quantities, have a positive impact on the host’s health. The ISAPP defines a prebiotic as ‘a substrate selectively utilized by host microorganisms conferring a health benefit’ [26]. To be classified as a prebiotic, a substance must be resistant to gastric acidity, hydrolysis by mammalian enzymes, and gastrointestinal absorption; fermentable by the gut microbiota; and selectively stimulate the growth and/or activity of gut bacteria linked to health [26]. Although the FDA has not officially recognized the term ‘prebiotic’, many substances identified as prebiotics by the ISAPP are categorized by the FDA as ‘non-digestible carbohydrates with a physiological effect’ [26,27,28,29]. These include various fibers, such as non-fiber prebiotics (e.g., cellulose, pectin, guar gum, psyllium husk) and fiber prebiotics like inulin, inulin-type fructans (ITF), and galacto-oligosaccharides (GOS) [30,31]. Recent years have seen the emergence of other substances like lactulose being recognized as non-fiber prebiotics or prebiotic candidates (Table 1). While prebiotics are predominantly known for enhancing beneficial bacteria like bifidobacteria and lactobacilli, the research on their broader effects on the gut microbiota in humans and animals is still developing. Studies on substances such as galacto- and fructo-oligosaccharides have demonstrated their potential antidepressant and anxiolytic properties, including their ability to mitigate the effects of chronic stress [32].
Furthermore, new terms such as synbiotics, postbiotics, and paraprobiotics have emerged in this field. Synbiotics, which combine prebiotics and probiotics, work to enhance the viability and efficacy of probiotic microorganisms, thereby boosting the presence of beneficial microbes in the gastrointestinal tract. Studies suggest that synbiotics can be more effective than using either probiotics or prebiotics alone [33,34,35]. Postbiotics, also referred to as metabiotics, biogenics, or cell-free supernatants (CFSs), are composed of metabolic byproducts and soluble factors produced during bacterial fermentation. These include short-chain fatty acids (SCFAs), enzymes, antimicrobial peptides (AMPs), teichoic acids, endo- and exopolysaccharides, cell surface proteins, vitamins, plasmalogens, organic acids, and other bioactive compounds, originating from live or lysed bacteria [35]. Paraprobiotics, distinct in their category, are nonviable or inactivated microbial cells that, when administered in adequate quantities, can induce beneficial biological activities in the host [35].

2.1. Probiotics and Their Role in Gut Microbiota and Immune System

The complex link between microbiota composition and human health is evident from the significant variations in gut microbiota observed between healthy individuals and those with various diseases and pathologies. These differences have implications not just for the intestine but also for mucosal tissues and the entire body [36,37]. Probiotics are key in modulating the gut microbiota and the immune system (Figure 2). Their roles include exerting antiallergic effects, anticancer activities, influencing inflammatory intestinal diseases, and impacting neurological disorders [38]. The metabolites produced by probiotics are gaining recognition for their importance in facilitating host–microbe interactions, which can be nutritionally influenced. These metabolites can improve intestinal barrier function, promote gastric motility and hormonal secretion, offer anti-inflammatory and antioxidant benefits, synthesize neurotransmitters, and alter the gut microbiome’s composition and metabolism [38,39,40]. Additionally, probiotics can prevent pathogens from adhering to mucosal cells and modulate the host’s immune response, further strengthening the intestinal barrier [41].
Strategies like probiotic administration are employed to correct dysbiosis in the gut microbiome, aiming to restore its equilibrium [42]. Probiotics can increase microbial diversity, boost lactase enzyme production, improve the immune microenvironment, and enhance intestinal permeability [43,44]. The human gastrointestinal tract harbors a vast array of symbiotic bacteria, forming a dynamic ecosystem with substantial physiological influence [45]. This microbiome plays a critical role in early life, where initial microbial colonization can shape the future development of allergies, obesity, and inflammatory conditions [46,47]. The establishment of gut microbiota in infancy is crucial for the development of the mucosal immune system [48]. Influences such as delivery method and breast milk composition can impact neonatal microbial colonization and mucosal immunity development, with long-term effects on immune-mediated diseases [49]. The timing of bacterial colonization in early life is pivotal for proper immune development, and ongoing research is illuminating aspects of maternal-to-infant microbial transmission during pregnancy and childbirth [50]. Intriguingly, the human placenta contains DNA from probiotic Bifidobacteria and Lactobacilli, while exposure to pathogenic bacteria like Ureaplasma species can disrupt immune development, leading to significant complications [51]. The mucosal immune system and commensal bacteria have co-evolved, particularly at mucosal sites, to maintain a balance between stability and pathogen defense [52]. Key immune cells like dendritic cells and macrophages, found throughout the body, regulate immune responses and maintain tissue homeostasis [53]. Innate lymphoid cells (ILCs), abundant in mucosal tissues and similar to phagocytes, interact with commensal microbiota, affecting cytokine production and various immune functions, highlighting their role in mucosal infection responses [54]. The host’s immune system shapes the microbiota, especially through secretory immunoglobulin A (sIgA), which binds to the gut microbiota, mediating effects like immune exclusion and microbial growth modulation [55]. The microbiome’s role in developing atopic diseases is well established [56]. Probiotics and their derivatives stimulate the immune system and modulate immunological responses, affecting dendritic cells, macrophages, B cells, and T cells [57]. They promote anti-inflammatory cytokine production, interact with gut–brain-axis neurotransmitters, and influence stress-related pathways [58,59,60]. The benefits of probiotics extend to reducing nutritional intolerances, improving nutrient availability, and alleviating allergies [61]. By targeting the gut microbiome, they manage allergy symptoms through modulating immune and inflammatory responses, believed to improve mucosal barrier functions and cytokine expression, thereby balancing Th1 and Th2 immune responses [62,63,64,65,66].

2.2. Probiotics and Their Roles on Viral Infections

Viral infectious diseases are a major contributor to the global burden of death and disability in both developed and developing countries [67,68]. A healthy and diverse microbial population, predominantly situated in the intestines, can protect the human host from various pathogenic diseases through a range of mechanisms, exerting substantial inhibitory effects [69]. Consequently, probiotics serve as a supportive approach, offering beneficial effects against viral infections by bolstering the immune response, maintaining the integrity of the protective cellular layer, and interacting with harmful microorganisms to impede their attachment (Figure 3).
The antiviral properties of specific Lactobacillus and Bifidobacterium strains against gastrointestinal and respiratory viruses have been extensively studied [70]. Numerous research studies highlight the effectiveness of probiotics as complementary treatments for rotavirus (RT), influenza virus, and human respiratory syncytial virus (RSV). Rotavirus (RV), a leading cause of severe diarrhea in children under five globally, alters the gut microbiome’s composition, shifting from Bacteroidetes to Firmicutes, which decreases bacterial diversity and increases the presence of harmful bacteria like Shigella [71]. The gastrointestinal (GI) tract, containing nearly 70% of the body’s immune cells, forms a critical link between the immune system and gut microorganisms, with notable interactions between the immune system and GI viruses. A recent study demonstrated that a combination of Bifidobacterium longum and Chlorella sorokiniana could enhance the cellular antiviral immune response [72]. Furthermore, Lactobacillus and Bifidobacterium have shown immunomodulatory effects against RV, both independently and in combination with RV vaccines, acting as immunostimulants [73,74,75,76]. Probiotics have been identified as beneficial adjuncts to RV vaccinations. Beyond GI viruses, there is growing evidence of probiotics and the gut microbiota exerting antiviral effects against respiratory viruses like the influenza virus [76,77]. Studies suggest that probiotics administered nasally or orally can bolster resistance to respiratory viral infections by stimulating the release of protective cytokines from alveolar macrophages and NK cells, crucial in defense mechanisms [78,79,80]. RSV, known for causing severe respiratory illness in infants and children, has a unique relationship with gut microbiota and probiotics [81,82,83]. Oral administration of specific Lactobacillus strains has been shown to significantly reduce RSV levels in the lungs by modulating the innate immune response in the respiratory system [84,85,86,87,88].

2.3. Probiotics and Their Role in COVID-19 Disease

Evidence suggests that post-COVID-19 recovery can be accompanied by intestinal microbial dysbiosis, a disruption in gut microbial balance lasting up to six months [89]. This dysbiosis, marked by a decrease in beneficial bacteria like Lactobacillus and Bifidobacterium, could hinder the recovery process in COVID-19 patients [90]. Reestablishing a balanced relationship between the lungs and the gut microbiota is thought to offer therapeutic advantages in combating COVID-19. Comprehending how probiotics or their metabolites exert their influence is essential for utilizing their potential in stabilizing the gut microbiota and potentially preventing or alleviating SARS-CoV-2 infection. Computational analyses, including drug discovery through computer modeling and machine learning predictive models, are crucial in the field of probiotics research. These tools help analyze extensive datasets and explore various mechanisms related to the microbiome and protein structures [90]. SARS-CoV-2 primarily targets the respiratory tract by binding to the ACE2 receptor, found in various organs, particularly type II alveolar and airway epithelial cells [90]. The virus also affects the gastrointestinal tract, causing symptoms like nausea, diarrhea, and vomiting, with its RNA detectable in fecal samples of infected individuals [91,92]. Compared to healthy counterparts, COVID-19 patients show a reduction in gut microbiota diversity, with a decrease in immunomodulatory commensals like Eubacterium rectale and Bifidobacterium. In contrast, genera such as Collinsella, Streptococcus, and Morganella are more abundant in these patients, along with species like Coprobacillus, Clostridium ramosum, and Clostridium hathewayi, particularly in those with a higher potential for SARS-CoV-2 transmission [93]. The gut–lung axis (GLA), which describes the bidirectional interaction between respiratory mucosa and gut microbiota, is pivotal for effective COVID-19 treatment by modulating the immune response [92]. The presence of specific microbial genera and species in the gut is associated with varying levels of SARS-CoV-2 infectivity, indicating the role of gut microbiota in immune enhancement through short-chain fatty acid production [94]. Healthy lungs also host a unique microbiota, including species of Prevotella, Streptococcus, Veillonella, Fusobacterium, and Haemophilus [95,96]. While the exact role of the microbiome in disease progression remains to be fully understood, there is a notable association between gut bacteria imbalances and increased susceptibility to pulmonary diseases [97]. This link is exemplified in conditions like inflammatory bowel disease (IBD), which are known to increase the risk of respiratory tract infections, highlighting the interplay between the lungs and the gut microbiota [98,99,100,101,102].

3. Probiotics as Adjuvant Treatment in COVID-19 Disease

Countries globally are grappling with the surge of infectious diseases, notably the COVID-19 pandemic, which has impacted millions. While therapeutic and preventive measures have shown success, the advent of new viral strains remains a concern. This situation highlights the necessity for innovative approaches to tackle viral infections that cause significant damage to organs such as the respiratory tract, liver, and colon, including the emerging challenge of long COVID [103,104].
In managing COVID-19, strategies like antiviral and anti-infective therapies are crucial but so is maintaining proper acid–base balance and restoring the microecological equilibrium [105]. A well-functioning intestinal ecosystem is key to defending against infections, and disturbances in nutritional and microecological balance can hinder the recovery of both intestinal health and lung function [84,85,86]. Patients with COVID-19 often exhibit microbial dysbiosis, characterized by a decline in Lactobacillus and Bifidobacterium levels in the gut [105]. Consequently, probiotics are being explored as a potential strategy against COVID-19 (Table 1) [106].
Research is increasingly focusing on probiotics with antiviral properties against the disease. Clinical trials have indicated that probiotics, including Lactobacillus acidophilus and Bifidobacterium infantis, may enhance immune function and reduce secondary infections in severe cases [107]. A study highlighted the effectiveness of a probiotic formula containing Lactiplantibacillus plantarum strains in improving COVID-19 outcomes in a controlled setting [108]. Additionally, diets rich in fermented vegetables have been linked to lower COVID-19 mortality rates [109,110]. Strains such as Lactobacillus, Bifidobacterium spp., Leuconostoc mesenteroides, and Pediococcus pentosaceus have shown promise in reducing the severity of COVID-19 [111], prompting the U.S. Food and Drug Administration to approve a microbiota-specific formula for early COVID-19 management in patients with obesity or type 2 diabetes [112].
The gut–lung axis is instrumental in understanding how probiotics might serve as adjunctive treatments for COVID-19, given their role in regulating both gut and lung environments. Meta-analyses have demonstrated that probiotics can prevent respiratory infections and reduce the incidence of acute respiratory infections without adverse effects [113,114], highlighting the interplay between the gastrointestinal tract’s immune and inflammatory state and other systems like the lungs [115,116]. Probiotic therapy in COVID-19 management focuses on restoring the gut microbiota’s diversity, composition, function, and metagenomic potential to healthy levels. Probiotics support the recovery of a healthy gut microbiota, bolster the intestinal barrier, and inhibit pathogen colonization [117,118,119,120,121]. They have been shown to modulate both innate and adaptive immune responses, enhancing the host’s immune response and improving gut flora [120,122,123,124,125]. Changes in immune cell levels, including naïve T helper cells and NK cells, have been observed in COVID-19 patients [126]. Clinical trials reveal that Lactobacillus acidophilus and L. plantarum can modulate cytokine release, providing immunomodulatory effects [124,127]. Omics techniques have identified probiotic-derived immunomodulatory genes and pathways, underscoring their role in cytokine regulation [128,129,130]. However, more research is essential to fully comprehend the extent of probiotics’ impact on maintaining gut immunological homeostasis in COVID-19 patients. A summary of studies on probiotics and their effects in COVD-19 infections is illustrated in Table 2.

4. Conclusions

Understanding the therapeutic roles of probiotics as adjunctive treatments, especially for emerging diseases like COVID-19 and long COVID, is becoming increasingly crucial. Detailed analysis and extensive research are vital in enhancing our comprehension of probiotics’ therapeutic effects. Furthermore, maintaining stringent production standards is essential for ensuring the quality and genetic stability of probiotic products. The application of probiotics in adjunctive COVID-19 and long COVID treatment has broadened their potential uses, enriching our knowledge of their function in the gut ecosystem and their interaction with the respiratory system. Longitudinal studies are necessary to illuminate the role of microbiota and the gut–lung axis in respiratory diseases, potentially leading to the use of specific probiotic strains in COVID-19 and long COVID treatment strategies. Over the past decade, significant advances have been made in understanding how the microbiota influences host immunity. Integrating technologies like artificial intelligence, machine learning, and computational studies with genomics- and omics-based analyses offers an efficient approach for the scientific community to further explore the host–microbiome relationship. Utilizing advancements in bioinformatics and computational research could unravel the molecular mechanisms through which probiotics affect SARS-CoV-2 and long COVID, bridging the divide between fundamental scientific discoveries and clinical practices, especially in modifying the microbiome to treat inflammatory diseases. Despite the availability of vaccines and widespread vaccination campaigns, the daily count of COVID-19 and long COVID cases remains high, continuing to impact human health and economies globally. Given the potential antiviral properties of probiotics and their byproducts, incorporating probiotics as an additional preventive measure alongside vaccines could represent a promising strategy to combat COVID-19 and long COVID.

Author Contributions

Conceptualization, M.G. (Marta Giovanetti) and G.G.; methodology, M.G. (Marta Giovanetti) and G.G.; formal analysis, M.G. (Marta Giovanetti), G.P., A.A., G.R., M.G. (Michele Guarino), M.C., E.R. and G.G; investigation, M.G. (Marta Giovanetti), G.P., A.A., G.R., M.G. (Michele Guarino), M.C., E.R. and G.G; data curation, M.G. (Marta Giovanetti); writing—original draft preparation, M.G. (Marta Giovanetti) and G.G.; writing—review and editing, M.G. (Marta Giovanetti), G.P., A.A., G.R., M.G. (Michele Guarino), M.C., E.R. and G.G.; visualization, M.G. (Marta Giovanetti), G.P. and G.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by PON “Ricerca e Innovazione” 2014–2020 and by the CRP-ICGEB RESEARCH GRANT 2020 Project CRP/BRA20-03, Contract CRP/20/03.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Tosta, S.; Moreno, K.; Schuab, G.; Fonseca, V.; Segovia, F.M.C.; Kashima, S.; Elias, M.C.; Sampaio, S.C.; Ciccozzi, M.; Alcantara, L.C.J.; et al. Global SARS-CoV-2 genomic surveillance: What we have learned (so far). Infect. Genet. Evol. 2023, 108, 105405. [Google Scholar] [CrossRef] [PubMed]
  2. Giovanetti, M.; Slavov, S.N.; Fonseca, V.; Wilkinson, E.; Tegally, H.; Patané, J.S.L.; Viala, V.L.; San, E.J.; Rodrigues, E.S.; Santos, E.V.; et al. Genomic epidemiology of the SARS-CoV-2 epidemic in Brazil. Nat. Microbiol. 2022, 7, 1490–1500. [Google Scholar] [CrossRef]
  3. Xavier, J.S.; Moir, M.; Tegally, H.; Sitharam, N.; Karim, W.A.; San, J.E.; Linhares, J.; Wilkinson, E.; Ascher, D.B.; Baxter, C.; et al. SARS-CoV-2 Africa dashboard for real-time COVID-19 information. Nat. Microbiol. 2022, 8, 1–4. [Google Scholar] [CrossRef] [PubMed]
  4. Hu, B.; Guo, H.; Zhou, P.; Shi, Z.-L. Characteristics of SARS-CoV-2 and COVID-19. Nat. Rev. Microbiol. 2021, 19, 141–154. [Google Scholar] [CrossRef] [PubMed]
  5. Giovanetti, M.; Branda, F.; Cella, E.; Scarpa, F.; Bazzani, L.; Ciccozzi, A.; Slavov, S.N.; Benvenuto, D.; Sanna, D.; Casu, M.; et al. Epidemic history and evolution of an emerging threat of international concern, the severe acute respiratory syndrome coronavirus 2. J. Med. Virol. 2023, 95, e29012. [Google Scholar] [CrossRef] [PubMed]
  6. Rocchi, G.; Giovanetti, M.; Benedetti, F.; Borsetti, A.; Ceccarelli, G.; Zella, D.; Altomare, A.; Ciccozzi, M.; Guarino, M.P.L. Gut Microbiota and COVID-19: Potential Implications for Disease Severity. Pathogens 2022, 11, 1050. [Google Scholar] [CrossRef]
  7. Taufer, C.R.; Rampelotto, P.H. The Role of Bifidobacterium in COVID-19: A Systematic Review. Life 2023, 13, 1847. [Google Scholar] [CrossRef]
  8. Beyerstedt, S.; Casaro, E.B.; Rangel, É.B. COVID-19: Angiotensin-converting enzyme 2 (ACE2) expression and tissue susceptibility to SARS-CoV-2 infection. Eur. J. Clin. Microbiol. Infect. Dis. 2021, 40, 905–919. [Google Scholar] [CrossRef]
  9. V’Kovski, P.; Kratzel, A.; Steiner, S.; Stalder, H.; Thiel, V. Coronavirus biology and replication: Implications for SARS-CoV-2. Nat. Rev. Microbiol. 2021, 19, 155–170. [Google Scholar] [CrossRef] [PubMed]
  10. Mokhtari, T.; Hassani, F.; Ghaffari, N.; Ebrahimi, B.; Yarahmadi, A.; Hassanzadeh, G. COVID-19 and multiorgan failure: A narrative review on potential mechanisms. Histochem. J. 2020, 51, 613–628. [Google Scholar] [CrossRef] [PubMed]
  11. Merad, M.; Blish, C.A.; Sallusto, F.; Iwasaki, A. The immunology and immunopathology of COVID-19. Science 2022, 375, 1122–1127. [Google Scholar] [CrossRef]
  12. Azkur, A.K.; Akdis, M.; Azkur, D.; Sokolowska, M.; van de Veen, W.; Brüggen, M.; O’mahony, L.; Gao, Y.; Nadeau, K.; Akdis, C.A. Immune response to SARS-CoV-2 and mechanisms of immunopathological changes in COVID-19. Allergy 2020, 75, 1564–1581. [Google Scholar] [CrossRef]
  13. Diamond, M.S.; Kanneganti, T.-D. Innate immunity: The first line of defense against SARS-CoV-2. Nat. Immunol. 2022, 23, 165–176. [Google Scholar] [CrossRef]
  14. Celardo, I.; Pace, L.; Cifaldi, L.; Gaudio, C.; Barnaba, V. The immune system view of the coronavirus SARS-CoV-2. Biol. Direct 2020, 15, 30. [Google Scholar] [CrossRef]
  15. Montazersaheb, S.; Khatibi, S.M.H.; Hejazi, M.S.; Tarhriz, V.; Farjami, A.; Sorbeni, F.G.; Farahzadi, R.; Ghasemnejad, T. COVID-19 infection: An overview on cytokine storm and related interventions. Virol. J. 2022, 19, 92. [Google Scholar] [CrossRef]
  16. Barbeta, E.; Motos, A.; Torres, A.; Ceccato, A.; Ferrer, M.; Cilloniz, C.; Bueno, L.; Badia, J.R.; Castro, P.; Ferrando, C.; et al. SARS-CoV-2–induced Acute Respiratory Distress Syndrome: Pulmonary Mechanics and Gas-Exchange Abnormalities. Ann. Am. Thorac. Soc. 2020, 17, 1164–1168. [Google Scholar] [CrossRef] [PubMed]
  17. Sekirov, I.; Russell, S.L.; Antunes, L.C.M.; Finlay, B.B.; Galla, S.; Chakraborty, S.; Cheng, X.; Yeo, J.; Mell, B.; Zhang, H.; et al. Gut Microbiota in Health and Disease. Physiol. Rev. 2010, 90, 859–904. [Google Scholar] [CrossRef] [PubMed]
  18. Hou, K.; Wu, Z.-X.; Chen, X.-Y.; Wang, J.-Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in health and diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar] [CrossRef]
  19. Guinane, C.M.; Cotter, P.D. Role of the gut microbiota in health and chronic gastrointestinal disease: Understanding a hidden metabolic organ. Ther. Adv. Gastroenterol. 2013, 6, 295–308. [Google Scholar] [CrossRef]
  20. Iizuka, M.; Saito, H.; Yukawa, M.; Itou, H.; Shirasaka, T.; Chiba, M.; Fukushima, T.; Watanabe, S. No evidence of persistent mumps virus infection in inflammatory bowel disease. Gut 2001, 48, 637–641. [Google Scholar] [CrossRef] [PubMed]
  21. Hrncir, T. Gut Microbiota Dysbiosis: Triggers, Consequences, Diagnostic and Therapeutic Options. Microorganisms 2022, 10, 578. [Google Scholar] [CrossRef]
  22. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. 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]
  23. Floch, M.H. Probiotics and Prebiotics. Gastroenterol. Hepatol. (N. Y.) 2014, 10, 680–681. [Google Scholar] [PubMed]
  24. Gasbarrini, G.; Bonvicini, F.; Gramenzi, A. Probiotics History. J. Clin. Gastroenterol. 2016, 50, S116–S119. [Google Scholar] [CrossRef] [PubMed]
  25. Vallianou, N.; Stratigou, T.; Christodoulatos, G.S.; Tsigalou, C.; Dalamaga, M. Probiotics, Prebiotics, Synbiotics, Postbiotics, and Obesity: Current Evidence, Controversies, and Perspectives. Curr. Obes. Rep. 2020, 9, 179–192. [Google Scholar] [CrossRef] [PubMed]
  26. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [PubMed]
  27. Burokas, A.; Arboleya, S.; Moloney, R.D.; Peterson, V.L.; Murphy, K.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Targeting the Microbiota-Gut-Brain Axis: Prebiotics Have Anxiolytic and Antidepressant-like Effects and Reverse the Impact of Chronic Stress in Mice. Biol. Psychiatry 2017, 82, 472–487. [Google Scholar] [CrossRef] [PubMed]
  28. Barathikannan, K.; Chelliah, R.; Rubab, M.; Daliri, E.B.-M.; Elahi, F.; Kim, D.-H.; Agastian, P.; Oh, S.-Y.; Oh, D.H. Gut Microbiome Modulation Based on Probiotic Application for Anti-Obesity: A Review on Efficacy and Validation. Microorganisms 2019, 7, 456. [Google Scholar] [CrossRef]
  29. Krumbeck, J.A.; Rasmussen, H.E.; Hutkins, R.W.; Clarke, J.; Shawron, K.; Keshavarzian, A.; Walter, J. Probiotic Bifidobacterium strains and galactooligosaccharides improve intestinal barrier function in obese adults but show no synergism when used together as synbiotics. Microbiome 2018, 6, 121. [Google Scholar] [CrossRef] [PubMed]
  30. Kazemi, A.; Noorbala, A.A.; Azam, K.; Eskandari, M.H.; Djafarian, K. Effect of probiotic and prebiotic vs placebo on psychological outcomes in patients with major depressive disorder: A randomized clinical trial. Clin. Nutr. 2019, 38, 522–528. [Google Scholar] [CrossRef]
  31. Aguilar-Toalá, J.E.; Garcia-Varela, R.; Garcia, H.S.; Mata-Haro, V.; González-Córdova, A.F.; Vallejo-Cordoba, B.; Hernández-Mendoza, A. Postbiotics: An evolving term within the functional foods field. Trends Food Sci. Technol. 2018, 75, 105–114. [Google Scholar] [CrossRef]
  32. Oberg, T.S.; Steele, J.L.; Ingham, S.C.; Smeianov, V.V.; Briczinski, E.P.; Abdalla, A.; Broadbent, J.R. Correction to: Intrinsic and inducible resistance to hydrogen peroxide in Bifidobacterium species. J. Ind. Microbiol. Biotechnol. 2018, 45, 765. [Google Scholar] [CrossRef]
  33. Cai, Z.; Hussain, M.D.; Yan, L.-J. Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer’s disease. Int. J. Neurosci. 2014, 124, 307–321. [Google Scholar] [CrossRef] [PubMed]
  34. Lin, H.V.; Frassetto, A.; Kowalik, E.J., Jr.; Nawrocki, A.R.; Lu, M.M.; Kosinski, J.R.; Hubert, J.A.; Szeto, D.; Yao, X.; Forrest, G.; et al. Butyrate and Propionate Protect against Diet-Induced Obesity and Regulate Gut Hormones via Free Fatty Acid Receptor 3-Independent Mechanisms. PLoS ONE 2012, 7, e35240. [Google Scholar] [CrossRef]
  35. Cuevas-González, P.; Liceaga, A.; Aguilar-Toalá, J. Postbiotics and paraprobiotics: From concepts to applications. Food Res. Int. 2020, 136, 109502. [Google Scholar] [CrossRef] [PubMed]
  36. Clemente, J.C.; Manasson, J.; Scher, J.U. The role of the gut microbiome in systemic inflammatory disease. BMJ 2018, 360, j5145. [Google Scholar] [CrossRef]
  37. Sorboni, S.G.; Moghaddam, H.S.; Jafarzadeh-Esfehani, R.; Soleimanpour, S. A Comprehensive Review on the Role of the Gut Microbiome in Human Neurological Disorders. Clin. Microbiol. Rev. 2022, 35, e0033820. [Google Scholar] [CrossRef]
  38. Latif, A.; Shehzad, A.; Niazi, S.; Zahid, A.; Ashraf, W.; Iqbal, M.W.; Rehman, A.; Riaz, T.; Aadil, R.M.; Khan, I.M.; et al. Probiotics: Mechanism of action, health benefits and their application in food industries. Front. Microbiol. 2023, 14, 1216674. [Google Scholar] [CrossRef]
  39. Roager, H.M.; Licht, T.R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef]
  40. Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Gil-Campos, M.; Gil, A. Mechanisms of Action of Probiotics. Adv. Nutr. Int. Rev. J. 2019, 10, S49–S66. [Google Scholar] [CrossRef]
  41. Fusco, A.; Savio, V.; Cimini, D.; D’ambrosio, S.; Chiaromonte, A.; Schiraldi, C.; Donnarumma, G. In Vitro Evaluation of the Most Active Probiotic Strains Able to Improve the Intestinal Barrier Functions and to Prevent Inflammatory Diseases of the Gastrointestinal System. Biomedicines 2023, 11, 865. [Google Scholar] [CrossRef] [PubMed]
  42. Kerry, R.G.; Patra, J.K.; Gouda, S.; Park, Y.; Shin, H.-S.; Das, G. Benefaction of probiotics for human health: A review. J. Food Drug Anal. 2018, 26, 927–939. [Google Scholar] [CrossRef] [PubMed]
  43. Jang, W.J.; Lee, J.M.; Hasan, T.; Lee, B.-J.; Lim, S.G.; Kong, I.-S. Effects of probiotic supplementation of a plant-based protein diet on intestinal microbial diversity, digestive enzyme activity, intestinal structure, and immunity in olive flounder (Paralichthys olivaceus). Fish Shellfish. Immunol. 2019, 92, 719–727. [Google Scholar] [CrossRef] [PubMed]
  44. Stratiki, Z.; Costalos, C.; Sevastiadou, S.; Kastanidou, O.; Skouroliakou, M.; Giakoumatou, A.; Petrohilou, V. The effect of a bifidobacter supplemented bovine milk on intestinal permeability of preterm infants. Early Hum. Dev. 2007, 83, 575–579. [Google Scholar] [CrossRef]
  45. Shehata, A.A.; Yalçın, S.; Latorre, J.D.; Basiouni, S.; Attia, Y.A.; El-Wahab, A.A.; Visscher, C.; El-Seedi, H.R.; Huber, C.; Hafez, H.M.; et al. Probiotics, Prebiotics, and Phytogenic Substances for Optimizing Gut Health in Poultry. Microorganisms 2022, 10, 395. [Google Scholar] [CrossRef]
  46. Ahmadizar, F.; Vijverberg, S.J.H.; Arets, H.G.M.; de Boer, A.; Lang, J.E.; Garssen, J.; Kraneveld, A.; Maitland-van Der Zee, A.H. Early-life antibiotic exposure increases the risk of developing allergic symptoms later in life: A meta-analysis. Allergy 2018, 73, 971–986. [Google Scholar] [CrossRef]
  47. Cox, L.M.; Yamanishi, S.; Sohn, J.; Alekseyenko, A.V.; Leung, J.M.; Cho, I.; Kim, S.G.; Li, H.; Gao, Z.; Mahana, D.; et al. Altering the Intestinal Microbiota during a Critical Developmental Window Has Lasting Metabolic Consequences. Cell 2014, 158, 705–721. [Google Scholar] [CrossRef]
  48. Hornef, M.W.; Torow, N. ‘Layered immunity’ and the ‘neonatal window of opportunity’—Timed succession of non-redundant phases to establish mucosal host–microbial homeostasis after birth. Immunology 2020, 159, 15–25. [Google Scholar] [CrossRef]
  49. Frei, R.; Akdis, M.; O’mahony, L. Prebiotics, probiotics, synbiotics, and the immune system. Curr. Opin. Gastroenterol. 2015, 31, 153–158. [Google Scholar] [CrossRef]
  50. Abrahamsson, T.R.; Wu, R.Y.; Jenmalm, M.C. Gut microbiota and allergy: The importance of the pregnancy period. Pediatr. Res. 2015, 77, 214–219. [Google Scholar] [CrossRef]
  51. Romano-Keeler, J.; Weitkamp, J.-H. Maternal influences on fetal microbial colonization and immune development. Pediatr. Res. 2015, 77, 189–195. [Google Scholar] [CrossRef] [PubMed]
  52. Scott, N.A.; Mann, E.R. Regulation of mononuclear phagocyte function by the microbiota at mucosal sites. Immunology 2020, 159, 26–38. [Google Scholar] [CrossRef]
  53. Ganal-Vonarburg, S.C.; Duerr, C.U. The interaction of intestinal microbiota and innate lymphoid cells in health and disease throughout life. Immunology 2020, 159, 39–51. [Google Scholar] [CrossRef]
  54. Bunker, J.J.; Bendelac, A. IgA Responses to Microbiota. Immunity 2018, 49, 211–224. [Google Scholar] [CrossRef] [PubMed]
  55. Hoces, D.; Arnoldini, M.; Diard, M.; Loverdo, C.; Slack, E. Growing, evolving and sticking in a flowing environment: Understanding IgA interactions with bacteria in the gut. Immunology 2020, 159, 52–62. [Google Scholar] [CrossRef] [PubMed]
  56. Nibbering, B.; Ubags, N.D.J. Microbial interactions in the atopic march. Clin. Exp. Immunol. 2020, 199, 12–23. [Google Scholar] [CrossRef] [PubMed]
  57. Petruzziello, C.; Saviano, A.; Ojetti, V. Probiotics, the Immune Response and Acute Appendicitis: A Review. Vaccines 2023, 11, 1170. [Google Scholar] [CrossRef]
  58. Srivastav, S.; Neupane, S.; Bhurtel, S.; Katila, N.; Maharjan, S.; Choi, H.; Hong, J.T.; Choi, D.-Y. Probiotics mixture increases butyrate, and subsequently rescues the nigral dopaminergic neurons from MPTP and rotenone-induced neurotoxicity. J. Nutr. Biochem. 2019, 69, 73–86. [Google Scholar] [CrossRef]
  59. Sajedi, D.; Shabani, R.; Elmieh, A. Changes in leptin, serotonin, and cortisol after eight weeks of aerobic exercise with probiotic intake in a cuprizone-induced demyelination mouse model of multiple sclerosis. Cytokine 2021, 144, 155590. [Google Scholar] [CrossRef]
  60. Gangaraju, D.; Raghu, A.V.; Gurudutt, P.S. Green synthesis of γ-aminobutyric acid using permeabilized probiotic Enterococcus faecium for biocatalytic application. Nano Sel. 2022, 3, 1436–1447. [Google Scholar] [CrossRef]
  61. Roobab, U.; Batool, Z.; Manzoor, M.F.; Shabbir, M.A.; Khan, M.R.; Aadil, R.M. Sources, formulations, advanced delivery and health benefits of probiotics. Curr. Opin. Food Sci. 2020, 32, 17–28. [Google Scholar] [CrossRef]
  62. Harata, G.; He, F.; Takahashi, K.; Hosono, A.; Miyazawa, K.; Yoda, K.; Hiramatsu, M.; Kaminogawa, S. Human Lactobacillus Strains from the Intestine can Suppress IgE-Mediated Degranulation of Rat Basophilic Leukaemia (RBL-2H3) Cells. Microorganisms 2016, 4, 40. [Google Scholar] [CrossRef] [PubMed]
  63. Fiocchi, A.; Pawankar, R.; Cuello-Garcia, C.; Ahn, K.; Al-Hammadi, S.; Agarwal, A.; Beyer, K.; Burks, W.; Canonica, G.W.; Ebisawa, M.; et al. World Allergy Organization-McMaster University Guidelines for Allergic Disease Prevention (GLAD-P): Probiotics. World Allergy Organ. J. 2015, 8, 4. [Google Scholar] [CrossRef] [PubMed]
  64. Liang, H.; Zhang, Y.; Miao, Z.; Cheng, R.; Jiang, F.; Ze, X.; Shen, X.; He, F. Anti-allergic effects of two potential probiotic strains isolated from infant feces in China. J. Funct. Foods 2022, 92, 105070. [Google Scholar] [CrossRef]
  65. Di Costanzo, M.; Amoroso, A.; Canani, R.B. Gut microbiota as a target for food allergy. J. Pediatr. Gastroenterol. Nutr. 2016, 63, S11–S13. [Google Scholar] [CrossRef]
  66. Galli, S.J.; Metz, M.; Starkl, P.; Marichal, T.; Tsai, M. Mast cells and IgE in defense against lethality of venoms: Possible “benefit” of allergy. Allergo J. 2020, 29, 46–62. [Google Scholar] [CrossRef] [PubMed]
  67. Villena, J.; Shimosato, T.; Vizoso-Pinto, M.G.; Kitazawa, H. Editorial: Nutrition, Immunity and Viral Infections. Front. Nutr. 2020, 7, 125. [Google Scholar] [CrossRef]
  68. Boutayeb, A. The Burden of Communicable and Non-Communicable Diseases in Developing Countries. In Handbook of Disease Burdens and Quality of Life Measures; Springer: Berlin/Heidelberg, Germany, 2010; Volume 100, pp. 532–545. [Google Scholar]
  69. Wang, B.; Yao, M.; Lv, L.; Ling, Z.; Li, L. The Human Microbiota in Health and Disease. Engineering 2017, 3, 71–82. [Google Scholar] [CrossRef]
  70. Wu, J.-Y.; Huang, P.-Y.; Liu, T.-H.; Kuo, C.-Y.; Tsai, Y.-W.; Tang, H.-J.; Lai, C.-C. Clinical efficacy of probiotics in the treatment of patients with COVID-19: A systematic review and meta-analysis of randomized controlled trials. Expert Rev. Anti-Infect. Ther. 2023, 21, 667–674. [Google Scholar] [CrossRef]
  71. Ma, C.; Wu, X.; Nawaz, M.; Li, J.; Yu, P.; Moore, J.E.; Xu, J. Molecular Characterization of Fecal Microbiota in Patients with Viral Diarrhea. Curr. Microbiol. 2011, 63, 259–266. [Google Scholar] [CrossRef]
  72. Rajput, S.; Paliwal, D.; Naithani, M.; Kothari, A.; Meena, K.; Rana, S. COVID-19 and Gut Microbiota: A Potential Connection. Indian J. Clin. Biochem. 2021, 36, 266–277. [Google Scholar] [CrossRef]
  73. Romero-Arguelles, R.; Tamez-Guerra, P.; González-Ochoa, G.; Romo-Sáenz, C.I.; Gomez-Flores, R.; Flores-Mendoza, L.; Aros-Uzarraga, E. Bifidobacterium longum and Chlorella sorokiniana Improve the IFN Type I-Mediated Antiviral Response in Rotavirus-Infected Cells. Microorganisms 2023, 11, 1237. [Google Scholar] [CrossRef] [PubMed]
  74. Rodriguez, W.J.; Kim, H.W.; Brandt, C.D.; Bise, B.; Kapikian, A.Z.; Chanock, R.M.; Curlin, G.; Parrott, R.H. Rotavirus Gastroenteritis in the Washington, DC, Area: Incidence of cases resulting in admission to the hospital. Am. J. Dis. Child. 1980, 134, 777–779. [Google Scholar] [CrossRef] [PubMed]
  75. Tate, J.E.; Burton, A.H.; Boschi-Pinto, C.; Parashar, U.D.; World Health Organization. Coordinated Global Rotavirus Surveillance Network Global, Regional, and National Estimates of Rotavirus Mortality in Children <5 Years of Age, 2000–2013. Clin. Infect. Dis. 2016, 62 (Suppl. 2), S96–S105. [Google Scholar] [CrossRef] [PubMed]
  76. Zhang, W.; Azevedo, M.S.; Wen, K.; Gonzalez, A.; Saif, L.J.; Li, G.; Yousef, A.E.; Yuan, L. Probiotic Lactobacillus acidophilus enhances the immunogenicity of an oral rotavirus vaccine in gnotobiotic pigs. Vaccine 2008, 26, 3655–3661. [Google Scholar] [CrossRef]
  77. Vlasova, A.N.; Chattha, K.S.; Kandasamy, S.; Liu, Z.; Esseili, M.; Shao, L.; Rajashekara, G.; Saif, L.J. Lactobacilli and bifidobacteria promote immune homeostasis by modulating innate immune responses to human rotavirus in neonatal gnotobiotic pigs. PLoS ONE 2013, 8, e76962. [Google Scholar] [CrossRef]
  78. Lehtoranta, L.; Pitkäranta, A.; Korpela, R. Probiotics in respiratory virus infections. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 1289–1302. [Google Scholar] [CrossRef]
  79. 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.; et al. 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, 17360. [Google Scholar] [CrossRef]
  80. Smith, A.M.; Smith, A.P. A Critical, Nonlinear Threshold Dictates Bacterial Invasion and Initial Kinetics During Influenza. Sci. Rep. 2016, 6, 38703. [Google Scholar] [CrossRef]
  81. Tate, M.D.; Schilter, H.C.; Brooks, A.G.; Reading, P.C. Responses of Mouse Airway Epithelial Cells and Alveolar Macrophages to Virulent and Avirulent Strains of Influenza A Virus. Viral Immunol. 2011, 24, 77–88. [Google Scholar] [CrossRef]
  82. 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]
  83. Groves, H.T.; Higham, S.L.; Moffatt, M.F.; Cox, M.J.; Tregoning, J.S. Respiratory Viral Infection Alters the Gut Microbiota by Inducing Inappetence. mBio 2020, 11, e03236-19. [Google Scholar] [CrossRef]
  84. Harding, J.N.; Siefker, D.; Vu, L.; You, D.; DeVincenzo, J.; Pierre, J.; Cormier, S.A. Altered gut microbiota in infants is associated with respiratory syncytial virus disease severity. BMC Microbiol. 2020, 20, 140. [Google Scholar] [CrossRef]
  85. Collins, P.L.; Melero, J.A. Progress in understanding and controlling respiratory syncytial virus: Still crazy after all these years. Virus Res. 2011, 162, 80–99. [Google Scholar] [CrossRef]
  86. Eguchi, K.; Fujitani, N.; Nakagawa, H.; Miyazaki, T. Prevention of respiratory syncytial virus infection with probiotic lactic acid bacterium Lactobacillus gasseri SBT2055. Sci. Rep. 2019, 9, 4812. [Google Scholar] [CrossRef]
  87. Chiba, E.; Tomosada, Y.; Vizoso-Pinto, M.G.; Salva, S.; Takahashi, T.; Tsukida, K.; Kitazawa, H.; Alvarez, S.; Villena, J. Immunobiotic Lactobacillus rhamnosus improves resistance of infant mice against respiratory syncytial virus infection. Int. Immunopharmacol. 2013, 17, 373–382. [Google Scholar] [CrossRef]
  88. Tomosada, Y.; Chiba, E.; Zelaya, H.; Takahashi, T.; Tsukida, K.; Kitazawa, H.; Alvarez, S.; Villena, J. Nasally administered Lactobacillus rhamnosus strains differentially modulate respiratory antiviral immune responses and induce protection against respiratory syncytial virus infection. BMC Immunol. 2013, 14, 40. [Google Scholar] [CrossRef]
  89. Chen, Y.; Gu, S.; Chen, Y.; Lu, H.; Shi, D.; Guo, J.; Wu, W.-R.; Yang, Y.; Li, Y.; Xu, K.-J.; et al. Six-month follow-up of gut microbiota richness in patients with COVID-19. Gut 2021, 71, 222–225. [Google Scholar] [CrossRef] [PubMed]
  90. Ni, W.; Yang, X.; Yang, D.; Bao, J.; Li, R.; Xiao, Y.; Hou, C.; Wang, H.; Liu, J.; Yang, D.; et al. Role of angiotensin-converting enzyme 2 (ACE2) in COVID-19. Crit. Care 2020, 24, 422. [Google Scholar] [CrossRef] [PubMed]
  91. Smyk, W.; Janik, M.K.; Portincasa, P.; Milkiewicz, P.; Lammert, F.; Krawczyk, M. COVID-19: Focus on the lungs but do not forget the gastrointestinal tract. Eur. J. Clin. Investig. 2020, 50, e13276. [Google Scholar] [CrossRef] [PubMed]
  92. Jones, D.L.; Baluja, M.Q.; Graham, D.W.; Corbishley, A.; McDonald, J.E.; Malham, S.K.; Hillary, L.S.; Connor, T.R.; Gaze, W.H.; Moura, I.B.; et al. Shedding of SARS-CoV-2 in feces and urine and its potential role in person-to-person transmission and the environment-based spread of COVID-19. Sci. Total Environ. 2020, 749, 141364. [Google Scholar] [CrossRef] [PubMed]
  93. Dang, A.T.; Marsland, B.J. Microbes, metabolites, and the gut–lung axis. Mucosal Immunol. 2019, 12, 843–850. [Google Scholar] [CrossRef] [PubMed]
  94. Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef]
  95. Gu, S.; Chen, Y.; Wu, Z.; Chen, Y.; Gao, H.; Lv, L.; Guo, F.; Zhang, X.; Luo, R.; Huang, C.; et al. Alterations of the Gut Microbiota in Patients with Coronavirus Disease 2019 or H1N1 Influenza. Clin. Infect. Dis. 2020, 71, 2669–2678. [Google Scholar] [CrossRef] [PubMed]
  96. Yeoh, Y.K.; Zuo, T.; Lui, G.C.-Y.; Zhang, F.; Liu, Q.; Li, A.Y.; Chung, A.C.; Cheung, C.P.; Tso, E.Y.; Fung, K.S.; et al. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut 2021, 70, 698–706. [Google Scholar] [CrossRef] [PubMed]
  97. Zuo, T.; Liu, Q.; Zhang, F.; Lui, G.; Tso, E.; Yeoh, Y.K.; Chen, Z.; Boon, S.; Chan, F.K.L.; Chan, P.; et al. Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19. Gut 2021, 70, 276–284. [Google Scholar] [CrossRef] [PubMed]
  98. Yamamoto, S.; Saito, M.; Tamura, A.; Prawisuda, D.; Mizutani, T.; Yotsuyanagi, H. The human microbiome and COVID-19: A systematic review. PLoS ONE 2021, 16, e0253293. [Google Scholar] [CrossRef] [PubMed]
  99. Fanos, V.; Pintus, M.C.; Pintus, R.; Marcialis, M. Lung microbiota in the acute respiratory disease: From coronavirus to metabolomics. J. Pediatr. Neonatal Individ. Med. 2020, 9, e090139. [Google Scholar] [CrossRef]
  100. Wypych, T.P.; Wickramasinghe, L.C.; Marsland, B.J. The influence of the microbiome on respiratory health. Nat. Immunol. 2019, 20, 1279–1290. [Google Scholar] [CrossRef]
  101. Carding, S.; Verbeke, K.; Vipond, D.T.; Corfe, B.M.; Owen, L.J. Dysbiosis of the gut microbiota in disease. Microb. Ecol. Health Dis. 2015, 26, 26191. [Google Scholar] [CrossRef]
  102. Raftery, A.L.; Tsantikos, E.; Harris, N.L.; Hibbs, M.L. Links Between Inflammatory Bowel Disease and Chronic Obstructive Pulmonary Disease. Front. Immunol. 2020, 11, 2144. [Google Scholar] [CrossRef] [PubMed]
  103. Rubin, R. COVID-19 Vaccines vs Variants—Determining How Much Immunity Is Enough. JAMA 2021, 325, 1241–1243. [Google Scholar] [CrossRef] [PubMed]
  104. Lopez Bernal, J.; Andrews, N.; Gower, C.; Gallagher, E.; Simmons, R.; Thelwall, S.; Stowe, J.; Tessier, E.; Groves, N.; Dabrera, G.; et al. Effectiveness of COVID-19 Vaccines against the B.1.617.2 (Delta) Variant. N. Engl. J. Med. 2021, 385, 585–594. [Google Scholar] [CrossRef]
  105. Viana, S.N.A.; Pereira, T.D.R.S.; Alves, J.d.C.; de Castro, C.T.; da Silva, L.S.C.; Pinheiro, L.H.S.; Roselino, M.N. Benefits of probiotic use on COVID-19: A systematic review and meta-analysis. Crit. Rev. Food Sci. Nutr. 2023, 21, 667–674. [Google Scholar] [CrossRef]
  106. Lahti, L.; Salojärvi, J.; Salonen, A.; Scheffer, M.; de Vos, W.M. Tipping elements in the human intestinal ecosystem. Nat. Commun. 2014, 5, 4344. [Google Scholar] [CrossRef]
  107. Dickson, R.P.; Singer, B.H.; Newstead, M.W.; Falkowski, N.R.; Erb-Downward, J.R.; Standiford, T.J.; Huffnagle, G.B. Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome. Nat. Microbiol. 2016, 1, 16113. [Google Scholar] [CrossRef]
  108. Zhang, Q.; Ran, X.; He, Y.; Ai, Q.; Shi, Y. Acetate Downregulates the Activation of NLRP3 Inflammasomes and Attenuates Lung Injury in Neonatal Mice with Bronchopulmonary Dysplasia. Front. Pediatr. 2021, 8, 595157. [Google Scholar] [CrossRef] [PubMed]
  109. Li, Q.; Cheng, F.; Xu, Q.; Su, Y.; Cai, X.; Zeng, F.; Zhang, Y. The role of probiotics in coronavirus disease-19 infection in Wuhan: A retrospective study of 311 severe patients. Int. Immunopharmacol. 2021, 95, 107531. [Google Scholar] [CrossRef]
  110. Gutiérrez-Castrellón, P.; Gandara-Martí, T.; Abreu, A.T.A.Y.; Nieto-Rufino, C.D.; López-Orduña, E.; Jiménez-Escobar, I.; Jiménez-Gutiérrez, C.; López-Velazquez, G.; Espadaler-Mazo, J. Probiotic improves symptomatic and viral clearance in COVID-19 outpatients: A randomized, quadruple-blinded, placebo-controlled trial. Gut Microbes 2022, 14, 2018899. [Google Scholar] [CrossRef]
  111. Bousquet, J.; Anto, J.M.; Czarlewski, W.; Haahtela, T.; Fonseca, S.C.; Iaccarino, G.; Blain, H.; Vidal, A.; Sheikh, A.; Akdis, C.A.; et al. Cabbage and fermented vegetables: From death rate heterogeneity in countries to candidates for mitigation strategies of severe COVID-19. Allergy 2020, 76, 735–750. [Google Scholar] [CrossRef]
  112. Zhao, L. Rutgers Researcher Invents Microbiota Formula to Help High Risk Patients Fight COVID-19; Rutgers the State University of New Jersey: New Brunswick, NJ, USA, 2021. [Google Scholar]
  113. Wang, Y.; Li, X.; Ge, T.; Xiao, Y.; Liao, Y.; Cui, Y.; Zhang, Y.; Ho, W.; Yu, G.; Zhang, T. Probiotics for prevention and treatment of respiratory tract infections in children: A systematic review and meta-analysis of randomized controlled trials. Medicine 2016, 95, e4509. [Google Scholar] [CrossRef] [PubMed]
  114. D’Ettorre, G.; Ceccarelli, G.; Marazzato, M.; Campagna, G.; Pinacchio, C.; Alessandri, F.; Ruberto, F.; Rossi, G.; Celani, L.; Scagnolari, C.; et al. Challenges in the Management of SARS-CoV-2 Infection: The Role of Oral Bacteriotherapy as Complementary Therapeutic Strategy to Avoid the Progression of COVID-19. Front. Med. 2020, 7, 389. [Google Scholar] [CrossRef] [PubMed]
  115. Hao, Q.; Lu, Z.; Dong, B.R.; Huang, C.Q.; Wu, T. Probiotics for preventing acute upper respiratory tract infections. Cochrane Database Syst. Rev. 2015, 21, CD006895. [Google Scholar] [CrossRef] [PubMed]
  116. Stavropoulou, E.; Bezirtzoglou, E. Probiotics as a Weapon in the Fight Against COVID-19. Front. Nutr. 2020, 7, 614986. [Google Scholar] [CrossRef]
  117. Marsland, B.J.; Trompette, A.; Gollwitzer, E.S. The Gut-Lung Axis in Respiratory Disease. Ann. Am. Thorac. Soc. 2015, 12 (Suppl. 2), S150–S156. [Google Scholar] [CrossRef]
  118. Ding, R.-X.; Goh, W.-R.; Wu, R.-N.; Yue, X.-Q.; Luo, X.; Khine, W.W.T.; Wu, J.-R.; Lee, Y.-K. Revisit gut microbiota and its impact on human health and disease. J. Food Drug Anal. 2019, 27, 623–631. [Google Scholar] [CrossRef]
  119. Ferreira, C.; Viana, S.D.; Reis, F. Gut Microbiota Dysbiosis–Immune Hyperresponse–Inflammation Triad in Coronavirus Disease 2019 (COVID-19): Impact of Pharmacological and Nutraceutical Approaches. Microorganisms 2020, 8, 1514. [Google Scholar] [CrossRef]
  120. Hiippala, K.; Jouhten, H.; Ronkainen, A.; Hartikainen, A.; Kainulainen, V.; Jalanka, J.; Satokari, R. The Potential of Gut Commensals in Reinforcing Intestinal Barrier Function and Alleviating Inflammation. Nutrients 2018, 10, 988. [Google Scholar] [CrossRef]
  121. Din, A.U.; Mazhar, M.; Waseem, M.; Ahmad, W.; Bibi, A.; Hassan, A.; Ali, N.; Gang, W.; Qian, G.; Ullah, R.; et al. SARS-CoV-2 microbiome dysbiosis linked disorders and possible probiotics role. Biomed. Pharmacother. 2020, 133, 110947. [Google Scholar] [CrossRef]
  122. Smythies, L.E.; Sellers, M.; Clements, R.H.; Mosteller-Barnum, M.; Meng, G.; Benjamin, W.H.; Orenstein, J.M.; Smith, P.D. Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J. Clin. Investig. 2005, 115, 66–75. [Google Scholar] [CrossRef]
  123. Batista, K.S.; de Albuquerque, J.G.; de Vasconcelos, M.H.A.; Bezerra, M.L.R.; Barbalho, M.B.d.S.; Pinheiro, R.O.; Aquino, J.d.S. Probiotics and prebiotics: Potential prevention and therapeutic target for nutritional management of COVID-19? Nutr. Res. Rev. 2021, 36, 181–198. [Google Scholar] [CrossRef]
  124. Sundararaman, A.; Ray, M.; Ravindra, P.V.; Halami, P.M. Role of probiotics to combat viral infections with emphasis on COVID-19. Appl. Microbiol. Biotechnol. 2020, 104, 8089–8104. [Google Scholar] [CrossRef] [PubMed]
  125. Zhao, W.; Peng, C.; Sakandar, H.A.; Kwok, L.-Y.; Zhang, W. Meta-Analysis: Randomized Trials of Lactobacillus plantarum on Immune Regulation Over the Last Decades. Front. Immunol. 2021, 12, 643420. [Google Scholar] [CrossRef]
  126. Grayson, M.H.; Camarda, L.E.; Hussain, S.-R.A.; Zemple, S.J.; Hayward, M.; Lam, V.; Hunter, D.A.; Santoro, J.L.; Rohlfing, M.; Cheung, D.S.; et al. Intestinal Microbiota Disruption Reduces Regulatory T Cells and Increases Respiratory Viral Infection Mortality Through Increased IFNγ Production. Front. Immunol. 2018, 9, 1587. [Google Scholar] [CrossRef]
  127. Zhao, W.; Liu, Y.; Kwok, L.-Y.; Cai, T.; Zhang, W. The immune regulatory role of Lactobacillus acidophilus: An updated meta-analysis of randomized controlled trials. Food Biosci. 2020, 36, 100656. [Google Scholar] [CrossRef]
  128. Yan, F.; Polk, D.B. Probiotics and immune health. Curr. Opin. Gastroenterol. 2011, 27, 496–501. [Google Scholar] [CrossRef] [PubMed]
  129. Anwar, F.; Altayb, H.N.; Al-Abbasi, F.A.; Al-Malki, A.L.; Kamal, M.A.; Kumar, V. Antiviral effects of probiotic metabolites on COVID-19. J. Biomol. Struct. Dyn. 2020, 39, 4175–4184. [Google Scholar] [CrossRef]
  130. Balmeh, N.; Mahmoudi, S.; Fard, N.A. Manipulated bio antimicrobial peptides from probiotic bacteria as proposed drugs for COVID-19 disease. Informatics Med. Unlocked 2021, 23, 100515. [Google Scholar] [CrossRef]
  131. Manna, S.; Chowdhury, T.; Chakraborty, R.; Mandal, S.M. Probiotics-Derived Peptides and Their Immunomodulatory Molecules Can Play a Preventive Role Against Viral Diseases Including COVID-19. Probiotics Antimicrob. Proteins 2020, 13, 611–623. [Google Scholar] [CrossRef]
  132. Hegazy, M.A.-E.; Ashoush, O.A.; Hegazy, M.T.; Wahba, M.; Lithy, R.M.; Abdel-Hamid, H.M.; Elshafy, S.A.A.; Abdelfatah, D.; Ibrahim, M.H.E.-D.; Abdelghani, A. Beyond probiotic legend: ESSAP gut microbiota health score to delineate SARS-CoV-2 infection severity. Br. J. Nutr. 2021, 127, 1180–1189. [Google Scholar] [CrossRef]
  133. Rodriguez-Blanque, R.; Sánchez-García, J.C.; Cobos-Vargas, Á.; Quesada, A.A.; Maldonado-Lobón, J.A.; Olivares, M.; Blanco-Rojo, R. Evaluation of the effect of Loigolactobacillus coryniformis K8 CECT 5711 consumption in health care workers exposed to COVID-19. Front. Nutr. 2022, 9, 962566. [Google Scholar] [CrossRef] [PubMed]
  134. Sohail, A.; Cheema, H.A.; Mithani, M.S.; Shahid, A.; Nawaz, A.; Hermis, A.H.; Chinnam, S.; Nashwan, A.J.; Cherrez-Ojeda, I.; Awan, R.U.; et al. Probiotics for the prevention and treatment of COVID-19: A rapid systematic review and meta-analysis. Front. Nutr. 2023, 10, 1274122. [Google Scholar] [CrossRef] [PubMed]
Figure 1. SARS-CoV-2 life cycle.
Figure 1. SARS-CoV-2 life cycle.
Viruses 16 00370 g001
Figure 2. The role of probiotics in gut homeostasis.
Figure 2. The role of probiotics in gut homeostasis.
Viruses 16 00370 g002
Figure 3. The role of probiotics in modulating viral infections.
Figure 3. The role of probiotics in modulating viral infections.
Viruses 16 00370 g003
Table 1. Prebiotics and main prebiotic candidates.
Table 1. Prebiotics and main prebiotic candidates.
CategorySub-Category Food SourcesReferences
Fiber prebiotics Non-digestible carbohydrates with physiological effect Inulin and Inulin-type fructans (ITF)Inulin (DP 2–60)
Oligofructose (DP < 10)
Short-chain fructooligosaccharides (scFOS) (DP 2–4)
Asparagus, leeks, garlic, chicory root, onion, Jerusalem artichoke, wheat, banana, agave
Synthetized from sucrose
[26,27]
Galagto-oligosaccharides (GOS)Alpha-GOS
Beta-GOS
Milk,
produced enzymatically from lactose
[26,27,28,29,30]
Non-fiber prebiotics Lactulose Synthetic disaccharide from isomerization of lactose[26]
Fiber candidate as prebiotics
Non-digestible carbohydrates with physiological effect
Resistant starch (RS2) Corn, potato, tapioca[26]
Polydextrose Synthetic fiber[26]
Isomalto-oligosaccharides (IMOSs)Isomaltose, panose, isomaltriose, isomaltotetraose, and isomaltopentoseEnzymatically produced from maltose and maltooligosaccharides[26]
Xylo-oligosaccharides (XOS) and Arabinoxylane (AXOS) Cereals[26]
Human milk oligosaccharides (HMOs) Neutral HMOs (e.g., 2′-Fucosyllactose (2′-FL) and Lactodifucopentaose
Neutral N-containing HMOs (e.g., lacto-N-tetraose)
Acids (HMOs) (e.g., 2′-sialyllactose)
Human milk[26]
Non-fibers candidate as prebiotics Polyphenol Fruits and vegetables[26]
Polyunsaturated fatty acids Crop seeds and vegetable oils, fish and fish oil[26]
Resistant proteins (RP) Plant-based foods (e.g., soybean, buckwheat, rice, and potato),
eggshell membrane
[26]
Table 2. Summary of studies on the effects of probiotics in COVID-19 and long COVID infection.
Table 2. Summary of studies on the effects of probiotics in COVID-19 and long COVID infection.
Type of Study/Method FeaturesProbiotic TestedMain ResultsRef.
Computational study, molecular dynamics simulationPlantaricin compounds, resulting from the metabolism of Lacticaseibacillus plantarum.
  • Plantaricin compounds—BN, JLA-9, W, D—showed antiviral activity, blocking the entry of SARS-CoV-2 by binding to RNA-dependent RNA polymerase (RdRp), the receptor-binding domain of SARS-CoV-2 (RBD) and angiotensin-converting enzyme 2 (ACE2).
[129]
Molecular docking analyses and molecular dynamics simulationMetabolic compounds resulting from probiotic strains activity.
  • Glycocin F and Lactococcine G, derived from Lactococcus lactis and Lactobacillus plantarum, respectively, had high affinity for binding to viral proteins and could be administered as therapy to inhibit SARS-CoV-2 infection.
[130]
Molecular docking analyses and in silico experiments Four probiotic-derived polypeptides: subtilisin, Curvacin A, Sakacin P, Lactococcin Gb.
  • Peptides derived from probiotic strains’ activity—Subtilisin (Bacillus amyloliquefaciens), Curvacin A (Lactobacillus curvatus), Sakacin P (Lactobacillus sakei), Lactococcin Gb (Lactococcus lactis)—showed a higher affinity to bind and block S-protein or RBD of S1 subunit of SARS-CoV-2 and human ACE2 receptor molecule.
[131].
Longitudinal cohort study
[NCT04447144]
Commercial probiotic yogurt (PY) 1·4 × 109 CFU of Bifidum bacteria
  • The evaluation of commercial PY intake in 170 patients with mild or moderate COVID-19 infection showed a significant negative correlation between PY intake and SARS-COV-2 infection severity.
  • GI symptoms, such as diarrhea, were more frequent in COVID-19 patients who never ate PY than in those who consumed variable amounts.
[132]
Double-blind, RCT
[NCT04366180]
Lactobacillus coryniformis K8 CECT 5711 (3 × 109 CFU/day) vs. placebo (maltodextrin).
  • The administration of one capsule of L. coryniformis K8 per day helps to extend the immune protection generated by the COVID-19 vaccine over time.
[133]
Single-center, quadruple-blinded, RCT in adult symptomatic COVID-19 outpatients
[NCT04517422]
Placebo vs. probiotic formula, Lactiplantibacillus plantarum KABP022, KABP023, KAPB033, Pedicoccus acidilactici KABP021 (2 × 109 CFU), for 30 days.
  • Probiotic supplementation was well tolerated and reduced nasopharyngeal viral load, pulmonary infiltrates, and duration of digestive and nondigestive symptoms compared with placebo.
  • No significant changes in fecal microbiota composition were detected between the probiotic and placebo.
  • Supplementation with the probiotic significantly increased specific IgM and IgG against SARS-CoV-2.
[110,134]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Giovanetti, M.; Pannella, G.; Altomare, A.; Rocchi, G.; Guarino, M.; Ciccozzi, M.; Riva, E.; Gherardi, G. Exploring the Interplay between COVID-19 and Gut Health: The Potential Role of Prebiotics and Probiotics in Immune Support. Viruses 2024, 16, 370. https://doi.org/10.3390/v16030370

AMA Style

Giovanetti M, Pannella G, Altomare A, Rocchi G, Guarino M, Ciccozzi M, Riva E, Gherardi G. Exploring the Interplay between COVID-19 and Gut Health: The Potential Role of Prebiotics and Probiotics in Immune Support. Viruses. 2024; 16(3):370. https://doi.org/10.3390/v16030370

Chicago/Turabian Style

Giovanetti, Marta, Gianfranco Pannella, Annamaria Altomare, Giulia Rocchi, Michele Guarino, Massimo Ciccozzi, Elisabetta Riva, and Giovanni Gherardi. 2024. "Exploring the Interplay between COVID-19 and Gut Health: The Potential Role of Prebiotics and Probiotics in Immune Support" Viruses 16, no. 3: 370. https://doi.org/10.3390/v16030370

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