Next Article in Journal
Metallic Shipwrecks and Bacteria: A Love-Hate Relationship
Previous Article in Journal
Deep-Sea Cold Seep Campylobacterota: Diversity, Growth, Metabolic Characteristics, and Nutrient Production
Previous Article in Special Issue
A Comparison of Three Automated Nucleic Acid Extraction Systems for Human Stool Samples
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Beneficial Microbiota in COVID-19: Insights from Key Bacterial Genera

by
Pabulo Henrique Rampelotto
1,*,
Clarissa Reginato Taufer
2 and
Juliana da Silva
2,3
1
Bioinformatics and Biostatistics Core Facility, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
2
Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
3
Graduate Program in Health and Human Development, Universidade La Salle, Canoas 92010-000, Brazil
*
Author to whom correspondence should be addressed.
Microorganisms 2025, 13(5), 1029; https://doi.org/10.3390/microorganisms13051029
Submission received: 7 April 2025 / Revised: 17 April 2025 / Accepted: 28 April 2025 / Published: 29 April 2025
(This article belongs to the Special Issue Advances in Human Microbiomes)

Abstract

:
The COVID-19 pandemic has highlighted the need for a comprehensive understanding of the factors influencing disease severity and progression. Emerging research indicates that the human microbiota, particularly beneficial bacteria, significantly impacts immune responses and health outcomes in COVID-19 patients. While existing studies provide general insights into the relationship between the microbiota and probiotics with COVID-19, they often lack a detailed exploration of how specific bacterial taxa might be used as adjunctive treatments. This review aims to address this gap by focusing on ten key genera of beneficial bacteria, discussing their roles in COVID-19 and evaluating their potential as probiotics for prevention and treatment. The review covers the impact of these microbes on human health, their population alterations in COVID-19 patients, and their interactions with other viral infections. Among these microbes, several exhibit distinct patterns of abundance in COVID-19 patients, influencing disease outcomes and highlighting their potential roles in infection dynamics. In COVID-19 patients, populations of Akkermansia, Ruminococcus, and Roseburia are consistently reduced, while those of Faecalibacterium show a significant decline in more severe cases. Bacteroides presents varying effects depending on the species involved. Alterations in the abundance of Blautia and Lachnospiraceae are associated with increased inflammation and disease severity. Likewise, the depletion of Lachnospira and Coprococcus populations, both linked to anti-inflammatory effects, may exacerbate symptom severity. Oscillospira, though less studied, is connected to overall health and could have implications for viral infections. This review synthesizes the current understanding of these beneficial microbes to highlight the importance of maintaining a healthy microbiota to alleviate the impact of COVID-19 and contribute to the development of novel therapeutic strategies involving microbiota modulation.

1. Introduction

The human microbiota plays a significant role in maintaining health and regulating immune responses [1]. Beneficial bacteria within these microbial communities are essential for various physiological processes, such as digestion, vitamin synthesis, and defense against pathogens [2]. Maintaining the delicate balance of the microbiota is the key to homeostasis, as disruptions can contribute to metabolic disorders, inflammatory diseases, and infections [3,4].
The COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, has brought unprecedented challenges to global health. While much attention has been focused on the direct impact of the virus on respiratory and systemic health, several studies demonstrate that the gut microbiota may also play a significant role in disease progression and severity. Alterations in the composition and function of the gut microbiota have been observed in COVID-19 patients, indicating a potential link between microbial dysbiosis and the clinical outcomes of the disease [5,6]. Clinical studies indicate that COVID-19 patients exhibit significant alterations in their gut microbiota, with a reduction in levels of beneficial bacteria and an increase in levels of opportunistic pathogens [6]. This dysbiosis is associated with disease severity and inflammatory markers, highlighting the importance of maintaining a healthy microbiota [7].
Probiotics, live microorganisms that provide health benefits to the host when administered in sufficient amounts, have been widely studied for their potential to modulate immune responses and support gut health during infections, including COVID-19. Probiotic supplementation could therefore be a valuable adjunctive therapy in managing COVID-19, helping to restore microbial balance and improve clinical outcomes (Mak et al., 2020) [8]. Numerous studies demonstrate that probiotics can enhance the gut barrier function, modulate systemic inflammation, and influence the immune system’s response to viral infections [9,10,11].
In COVID-19, probiotics may offer several potential benefits. They can help restore the balance of the gut microbiota disrupted by the viral infection and associated treatments, such as antibiotics [8]. Probiotics like Bifidobacterium and Lactobacillus enhance the production of antiviral cytokines and reduce the severity of respiratory infections [12,13]; however, it remains unclear to what extent diet and medications might influence the efficacy of probiotics in restoring the gut microbiota balance during COVID-19 [14]. In addition, some probiotic strains reduce the duration and severity of symptoms in viral infections [15], suggesting a potential role in alleviating COVID-19 symptoms. In previous reviews, we detailed the involvement of Bifidobacterium and Lactobacillus in COVID-19 [16,17,18]. However, the roles of other well-known probiotics have yet to be thoroughly reviewed, as the literature on these genera is scattered, making it difficult to achieve a clear understanding.
This review aims to explore the relationship between ten key genera of beneficial or probiotic bacteria and COVID-19. Each section discusses the influence of these genera on human health, their population alterations in COVID-19 patients, and their interactions with other viral infections, which can contribute to understanding the relationship between these genera and viral infections. This knowledge could facilitate the development of innovative therapeutic strategies aimed at restoring microbial balance and enhancing host defenses against SARS-CoV-2 and other viral pathogens.

2. Key Bacterial Genera

2.1. Akkermansia

Akkermansia, particularly the species Akkermansia muciniphila, is a beneficial bacterium that plays a significant role in maintaining gut barrier integrity and modulating immune responses. This mucin-degrading bacterium resides in the mucus layer of the gut, contributing to the maintenance of the gut lining and promoting overall mucosal health [19]. Akkermansia abundance is inversely associated with various metabolic disorders, including obesity, type 2 diabetes, and metabolic syndrome [20,21]. Its presence is linked to reduced inflammation and improved insulin sensitivity, indicating its potential as a therapeutic target for metabolic diseases [22].
In COVID-19, an increase in Akkermansia populations has been observed in both infected patients and those who have recovered [23,24,25,26,27]. This increase correlates with worsening disease severity and higher rates of SARS-CoV-2 viral replication. Specifically, patients exhibiting lower viral loads (Ct > 31) showed decreased Akkermansia levels compared to those with higher replication rates (Ct ≤ 31) [28]. These findings suggest a relationship between disease severity, viral load, and altered Akkermansia abundance, indicating that dysbiosis may persist even after recovery from COVID-19. In addition, the enrichment of Akkermansia muciniphila populations has been positively associated with levels of pro-inflammatory cytokines such as IL-1β, IL-6, and CXCL8 [7], which could imply potential protective effects during COVID-19. However, Akkermansia abundance was reduced in the oral microbiota of post-COVID-19 patients (1–8 months after infection) and in those who received antibiotics during treatment [29,30], as well as in children [31,32]. Furthermore, this genus was enriched in patients with other respiratory diseases compared to COVID-19 patients in intensive care units, emphasizing the complex relationship between Akkermansia and respiratory illnesses [33].
The involvement of Akkermansia in other viral infections has also been investigated. Its ability to strengthen the gut barrier and modulate immune responses suggests a protective role in viral infections affecting the gastrointestinal tract. An increase in Akkermansia abundance was reported in children living with HIV undergoing antiretroviral treatment, potentially linked to the beneficial effects of this treatment on inflammation [34]. In addition, Akkermansia muciniphila may protect hosts from Severe Fever with Thrombocytopenia Syndrome (SFTS) by attenuating systemic inflammation through the production of the β-carboline alkaloid harmaline, which suppresses NF-κB-mediated inflammation [35].
In animal models, an increase in Akkermansia abundance was observed following influenza A virus infection, particularly on day 7 post-infection [36]. Similarly, in Severe Fever with Thrombocytopenia Syndrome virus-infected mice, those that survived showed a significant increase in Akkermansia muciniphila abundance three days post-infection [35]. By contrast, H3N2-infected mice exhibited lower levels of Akkermansia [37]. The increase in Akkermansia muciniphila abundance was associated with rapidly spreading viral infections, and CD8 T cells were shown to modulate several dysbiotic taxa after lymphocytic choriomeningitis virus (LCMV) infection in mice, resulting in significant anorexic behavior [38]. Akkermansia muciniphila not only proliferated following LCMV infection but also during fasting conditions. Notably, experimental enrichment of Akkermansia muciniphila populations after LCMV infection suppressed the expression of Granzyme B and T-BET in virus-specific CD8 T cells, suggesting that natural enrichment may help mitigate initial immune responses.
This variability highlights the complexity of Akkermansia’s role in viral infections, with evidence suggesting that its increased abundance is associated with fast-spreading viruses and that it may have the potential to modulate immune responses and maintenance of the gut lining, which could be beneficial in mitigating the effects of viral infections, including in cases of COVID-19.

2.2. Bacteroides

Bacteroides is a dominant genus in the human gut microbiota, playing a crucial role in the digestion of complex carbohydrates and the production of short-chain fatty acids (SCFAs), which are essential for gut health [39]. As commensal bacteria, Bacteroides species contribute to the maintenance of gut homeostasis; however, imbalances in their populations have been associated with various diseases, including inflammatory bowel disease (IBD) and obesity [40,41]. The impact of Bacteroides on inflammation varies between species: some protect against excessive inflammation, while others can exacerbate inflammatory responses, highlighting the importance of species composition in shaping clinical outcomes. Additionally, certain species may act as opportunistic pathogens, causing infections outside the gut, such as bacteremia and abscesses [42,43].
In COVID-19, several studies have reported significant alterations in Bacteroides populations. Al Bataineh et al. (2020) observed a reduction in Bacteroides abundance among COVID-19 patients compared to healthy controls, suggesting that SARS-CoV-2 may disrupt the gut microbiota, leading to a loss of beneficial Bacteroides species [44]. On the other hand, Reinold et al. (2021) and Mizutani et al. (2020) found an increase in the relative abundance of Bacteroides in patients positive for SARS-CoV-2 [45,46]. Bucci et al. (2023) further identified Bacteroides as one of the 10 key variables predicting COVID-19 severity [47], and Galperine et al. (2023) recognized it as a signature genus in COVID-19 patients with diarrhea [48]. In addition, an increase in the abundance of the opportunistic pathogen Bacteroides nordii was observed in the feces of COVID-19 patients [6]. Worth noting, Bacteroides dorei, Bacteroides thetaiotaomicron, Bacteroides massiliensis, and Bacteroides ovatus populations were inversely correlated with SARS-CoV-2 load in fecal samples [6], with Bacteroides dorei downregulating ACE2 expression in the murine colon, a key receptor for SARS-CoV-2 entry [49]. These findings demonstrate the complexity of microbiota alterations in COVID-19 and suggest that the impact on Bacteroides may vary depending on the study population, disease severity, and specific species involved.
Bacteroides has also been studied in the context of other viral infections. The oropharyngeal microbiota of patients with influenza A virus had a reduced abundance of Bacteroides [50]. In addition, Bacteroides species have been implicated in modulating immune responses during viral infections. Bacteroides fragilis, for example, produces polysaccharide A, which can enhance the immune response and protect against viral encephalitis in mice [51]. Polysaccharide A also induces the expression of IFN-β, which regulates natural resistance to viral infections, including influenza virus A, in murine models and in vitro analyses [52] and protects against intestinal inflammation by inducing IL-10-producing CD4+ regulatory T cells [53]. In mice infected with H3N2, a lower abundance of Bacteroides was observed, along with reduced levels of acetate in the intestine and serum [37]. Bacteroides produce acetate, and this SCFA could enhance the host’s antiviral response [54], reduce inflammatory reactions, and prevent lung injuries [55]. Furthermore, pretreatment with acetate in H3N2-infected mice was able to partially restore the airway epithelial barrier function and reduce the levels of inflammatory cytokines TNF-α, IL-6, and IL-1β in the lungs [37]. Changes in Bacteroides populations have also been observed in HIV-infected individuals, with a shift toward a more inflammatory gut microbiota [56]. These findings suggest the role of Bacteroides in both maintaining immune homeostasis and responding to viral infections. Maintaining a healthy population of Bacteroides may be beneficial in mitigating the effects of viral infections and could be effective in reducing disease severity, including in cases of COVID-19.

2.3. Blautia

Blautia are known for their ability to produce SCFAs, particularly acetate and butyrate, which are beneficial for gut health and have anti-inflammatory [57]. Blautia species are associated with a healthy gut microbiota and have been inversely correlated with various diseases. Reduced levels of Blautia have been observed in patients with IBD, obesity, type 2 diabetes, and some types of cancer [57,58]. Consequently, a reduction in Blautia abundance has been implicated in increased systemic inflammation and disease severity.
In COVID-19, several studies have documented alterations in the abundance of Blautia. Al Bataineh et al. (2020) reported an increase in the abundance of this genus in COVID-19 patients [44], while Maeda et al. (2022) and Gaibani et al. (2021) identified Blautia as one of the characteristic bacteria in healthy controls [23,59]. Gaibani et al. (2021) also observed that Blautia was associated with COVID-19 patients who did not require ICU admission or develop bloodstream infections, suggesting a potential protective role for this taxon [23]. Wu et al. (2021) identified reduced Blautia abundance in the gut microbiota of COVID-19 patients and proposed it as a potential biomarker due to its decreased levels [60]. Similarly, Schult et al. (2022) reported a reduced abundance of Blautia in severe and fatal cases, indicating that lower levels of this beneficial bacterium may be linked to worse disease outcomes [61]. In nasopharyngeal samples, Romani et al. (2024) observed a reduction in Blautia populations in children with COVID-19, with the genus being negatively associated with the disease [32]. Collectively, these findings suggest that Blautia may play a protective role in COVID-19, and its reduced abundance could be associated with disease progression and severity.
In other viral infections, a reduction in the abundance of Blautia has been consistently observed and linked to reduced inflammation. Gu et al. (2020) found significant depletion of this genus in the gut microbiota of influenza A patients [5]. In children with rotavirus, combining conventional therapy with zinc supplementation showed a negative correlation between Blautia abundance and levels of inflammatory markers like IL-6, TNF-α, PCR, and D-lactate [62]. Animal studies demonstrated that Blautia faecis, isolated from post-influenza mice, reduced IL-8 production induced by TNF-α in intestinal epithelial cells [63]. In HIV-positive patients, Vesterbacka et al. (2017) reported reduced Blautia levels, with higher levels found in antiretroviral therapy-naïve patients compared to elite controllers, indicating a positive association with immune activation [64]. Mutlu et al. (2024) observed increased Blautia abundance in control groups [65], while Vázquez-Castellanos et al. (2015) identified it as a biomarker for these groups [66]. In co-infected HIV and hepatitis C virus (HCV) patients, direct-acting antiviral treatment increased Blautia abundance, suggesting microbiota restoration post-treatment [67].

2.4. Coprococcus

Coprococcus is also an SCFA-producing genus of Lachnospiraceae. The genus has been associated with various diseases, primarily due to its role in gut health and overall microbiome balance. Decreased levels of Coprococcus have been linked to obesity, with studies showing that its abundance is reduced in obese individuals, which indicates a potential role in metabolic regulation and energy homeostasis [68,69]. In addition, lower levels of Coprococcus have been observed in IBD, contributing to dysbiosis that exacerbates inflammation and gastrointestinal symptoms [70,71]. Furthermore, recent studies indicate an association between Coprococcus and neurodegenerative diseases, such as Parkinson’s disease, where gut microbiota imbalances may influence neuroinflammation and the gut–brain axis [72,73].
In COVID-19, several studies have reported alterations in Coprococcus populations. Albrich et al. (2022) found lower Coprococcus levels in high-risk COVID-19 groups compared to healthy controls [74], while Wu et al. (2021) identified its reduction as a potential biomarker for COVID-19 [60]. Gaibani et al. (2021) further noted that Coprococcus populations were enriched in COVID-19 patients who did not require ICU admission compared to both COVID-19 and non-COVID-19 patients in the ICU, suggesting a possible protective or modulatory role in the severity of the disease [23]. Similarly, Schult et al. (2022) observed a reduction in Coprococcus abundance in severe and fatal cases of COVID-19, implying that lower levels of this beneficial bacterium may be linked to worse outcomes [61]. In post-COVID-19 patients, Zhang et al. (2023a) reported a lower relative abundance of Coprococcus compared to healthy controls [75]. At the species level, Li et al. (2021) [76] reported a reduction in Coprococcus catus populations in COVID-19 patients compared to controls, with an inverse association observed between Coprococcus catus abundance and IL-8 levels in post-COVID-19 patients, suggesting that its reduction may contribute to increased inflammation that persists after recovery [77]. Altogether, these findings indicate that disruptions in Coprococcus populations may contribute to both the severity and the prolonged impact of COVID-19.
Although the role of Coprococcus in other viral infections has not been extensively studied, its known benefits in maintaining gut health and modulating immune responses suggest a potential protective role. The production of butyrate and propionate by Coprococcus is a key factor for enhancing gut barrier integrity and reducing systemic inflammation, both of which are essential during viral infections [78]. In HIV, dysbiosis and reduced Coprococcus levels have been linked to increased systemic inflammation and immune activation [79].

2.5. Faecalibacterium

Faecalibacterium, particularly Faecalibacterium prausnitzii, is a prominent and beneficial member of the human gut microbiota. It is a major producer of butyrate, an SCFA that serves as an energy source for colonic epithelial cells and exhibits anti-inflammatory properties [80]. Faecalibacterium prausnitzii abundance has been inversely associated with inflammatory conditions such as Crohn’s disease and ulcerative colitis, suggesting its protective role in gut health [81,82,83]. As one of the primary producers of butyrate, the reduction in Faecalibacterium prausnitzii populations, particularly in severe cases of COVID-19, suggests a breakdown of the intestinal barrier and an increase in systemic inflammation. Reduced levels of Faecalibacterium have also been observed in patients with obesity, type 2 diabetes, and IBD, further highlighting its importance in maintaining metabolic and gastrointestinal health [84,85,86].
In COVID-19, studies have shown varying effects on Faecalibacterium populations. While Al Bataineh et al. (2020) observed an increase in the abundance of this bacterium in COVID-19 patients compared to healthy controls, several other studies reported a decrease [44]. Bucci et al. (2023) identified Faecalibacterium as a biomarker for predicting disease severity [47], but Albrich et al. (2022) [74], Maeda et al. (2022) [59], Reinold et al. (2021) [45], Mizutani et al. (2020) [46], Schult et al. (2022) [61], Hazan et al. (2022) [87], and Yeoh et al. (2021) [7] all noted reduced levels in patients with severe or high-risk conditions. Specifically, the abundance of Faecalibacterium prausnitzii was inversely correlated with the severity of COVID-19, highlighting its potential protective role in severe COVID-19 cases [6]. These data suggest that lower levels of this beneficial bacterium may be linked to worse disease outcomes, while its presence could help fight infection. In addition, Faecalibacterium prausnitzii abundance was reduced in samples collected up to 30 days after disease resolution and for at least 6 months after recovery in severe patients, demonstrating the delay in restoring a healthy microbiota and its potential implications for post-COVID-19 health [7,59]. Tang et al. (2020) identified that Faecalibacterium prausnitzii abundance was negatively related to PRC in the critical COVID-19 group [88]. It is also important to highlight that the reduction in the diversity of Faecalibacterium observed in patients with bloodstream infections [89] was comparable to the significant decrease in diversity seen in severely ill mice intentionally infected with SARS-CoV-2 [90,91]. These findings indicate that Faecalibacterium levels may be disrupted in COVID-19, with potential implications for disease severity, progression, and inflammation.
Studies on other viral diseases have also reported a reduction in Faecalibacterium populations. In H1N1 patients, the abundance of Faecalibacterium was reduced compared to healthy controls [5], and in a Chinese cohort, Faecalibacterium abundance was negatively associated with the risk of H1N1 severity [92], indicating a protective role in the infection. Faecalibacterium abundance was also reduced in patients with H7N9, especially in those treated with antibiotics [93]. In addition, Faecalibacterium prausnitzii was linked to anti-inflammatory effects both in vitro and in vivo models of colitis [83]. Faecalibacterium populations were also consistently depleted among HIV+ individuals, which can contribute to inflammation and immune dysregulation, exacerbating the disease’s progression and affecting overall health [94].

2.6. Lachnospira

Lachnospira is also an SCFA-producing genus of Lachnospiraceae. The genus has been associated with various diseases, particularly due to its role in maintaining gut health and influencing systemic inflammation. Decreased levels of Lachnospira were linked to obesity and metabolic syndrome, with studies indicating that lower abundance of this genus correlates with increased body mass index and insulin resistance [95,96]. In IBD, reduced Lachnospira levels were observed, suggesting a potential role in gut barrier function and immune modulation, which may exacerbate inflammatory responses [70,71]. Furthermore, Lachnospira has been implicated in neurodegenerative conditions, including Alzheimer’s disease, where dysbiosis and altered gut microbiota composition may influence neuroinflammation and cognitive decline [97,98].
In COVID-19, some studies have reported changes in Lachnospira populations. Albrich et al. (2022) observed that Lachnospira levels were lower in high-risk COVID-19 groups compared to healthy controls [74]. Moreira-Rosário et al. (2021) also reported a significant reduction in Lachnospira populations in COVID-19 patients, particularly in those with severe disease [99]. Gaibani et al. (2021) identified the genus as one of the main discriminants of the microbiota in healthy controls [23]. Romani et al. (2022) identified the absence of Lachnospira in patients with moderate COVID-19 compared to patients with mild and asymptomatic cases, suggesting that the presence or absence of Lachnospira may be linked to the severity of the disease [31].
Some studies have also observed changes in Lachnospira abundance in other viral infections. In HIV, Lachnospira abundance presented significant correlations with the number of CD4 + T cells and HIV viral load [100,101], and a significant association between total fiber intake and Lachnospira abundance was observed [102]. Lower abundance of Lachnospira was observed in HCV-infected patients [103], which could suggest loss of protection and persistent inflammation since this genus is an SCFA-producing taxa. In addition, the lower abundance of Lachnospira observed in HCV-infected people was completely abolished after 3 months under sustained viral response, which could suggest increased production of butyrate and, therefore, a healthier gut. Interestingly, Lachnospira abundance was decreased in patients with neuroinfection when compared to controls [104].

2.7. Lachnospiraceae Unclassified

The family Lachnospiraceae is a morphologically and phylogenetically varied taxon, with a high number of species [105]. One of the main challenges in studying Lachnospiraceae is its diversity, as the family includes numerous members that are difficult to classify at the genus level using 16S rRNA gene sequencing. As a result, many microbiome studies report these bacteria as “Lachnospiraceae unclassified” or “Lachnospiraceae incertae sedis” (unclassified or uncertain placement) [106]. This classification challenge demonstrates the complexity and diversity within this family and highlights the need for more advanced techniques to accurately identify and characterize its members.
In humans, the family Lachnospiraceae has been linked to various diseases. Reduced levels of Lachnospiraceae have been consistently associated with obesity, type 2 diabetes, and metabolic syndrome, and a lower abundance of this family correlates with increased body mass index and insulin resistance, suggesting a role in metabolic regulation [96,107]. High abundance of Lachnospiraceae in the human gut microbiome is related to protection against different types of cancer [108,109,110]. In addition, Lachnospiraceae abundance is inversely correlated with cardiovascular disease risk factors [111,112] and there is emerging evidence that Lachnospiraceae may influence the development of allergic diseases [113,114]. As such, a balanced gut microbiota, including this family, may play an important role in immune system development and the prevention of allergies.
In COVID-19, several studies have reported changes in Lachnospiraceae populations. Albrich et al. (2022) demonstrated that Lachnospiraceae was associated with protective immune responses and its abundance was reduced in high-risk COVID-19 groups [74]. Maeda et al. (2020) reported that the abundance of Lachnospiraceae was lower in severe COVID-19 cases compared to healthy controls [59], while Galperine et al. (2023) identified a reduction in the abundance of members of Lachnospiraceae in ventilated COVID-19 patients compared to non-ventilated COVID-19 patients [48]. Schult et al. (2022) also observed a reduction in Lachnospiraceae abundance in severe and fatal COVID-19 cases [61]. Zuo et al. (2020a) identified higher abundance of Lachnospiraceae in stool samples with low or no infectivity of SARS-CoV-2, as well as other SCFA-producing bacteria, while fecal samples with high infectivity exhibited a higher abundance of opportunistic bacteria [115]. These findings suggest that Lachnospiraceae levels may be disrupted in COVID-19, potentially affecting disease severity and progression, and that higher levels may be associated with an increased ability to combat SARS-CoV-2 infection in the gut.
The involvement of Lachnospiraceae in other viral infections has been mainly studied in HIV. San-Juan-Vergara et al. (2018) reported, for the first time, a predominantly Lachnospiraceae-based signature in HIV-infected individuals [116]. Vujkovic-Cvijin et al. (2020) showed HIV-associated dysbiosis characterized by depletion of Lachnospiraceae, regardless of sexual preference [117]. Interestingly, although several previous studies have reported members of the Lachnospiraceae family associated with healthy individuals, the abundance of Lachnospiraceae UCG-004 was increased in a cohort of 13 HIV-positive patients [65,118]. In patients with H1N1, Lachnospiraceae populations were drastically reduced compared to healthy controls, and the reduction was negatively correlated with inflammatory cytokine levels in mouse models of COPD co-infected with H1N1 and Haemophilus influenzae, demonstrating its anti-inflammatory potential in viral infections [5,119].

2.8. Oscillospira

Oscillospira is a genus of anaerobic, Gram-positive bacteria within the family Ruminococcaceae. As SCFA-producers, this genus is important for maintaining gut homeostasis, and lower levels of Oscillospira are associated with obesity and metabolic syndrome, suggesting its role in regulating energy metabolism and gut health [120,121]. In addition, Oscillospira abundance was inversely correlated with Crohn’s disease [122], nonalcoholic fatty liver diseases [123], and gastric cancer [124].
In COVID-19, alterations in the abundance of Oscillospira have been documented in a few studies. Gaibani et al. (2021) identified a correlation between elevated levels of Oscillospira and non-ICU COVID-19 patients, suggesting that higher concentrations of this genus may be associated with milder disease manifestations [23]. On the other hand, Schult et al. (2022) reported a significant reduction in Oscillospira abundance in severe and fatal COVID-19 cases, indicating that lower levels of these beneficial bacteria may correlate with adverse clinical outcomes [61]. These findings indicate that the abundance of Oscillospira may be disrupted in COVID-19, with the observed association between increased Oscillospira abundance and less severe cases, alongside the decline noted in severe cases, suggesting a potential role for these bacteria in modulating the inflammatory response and influencing disease progression.
The contribution of Oscillospira in the context of other viral infections has presented mixed results. Wang et al. (2020) observed enrichment of this genus in HIV-infected individuals [125], while Chandiwana et al. (2023) observed a low abundance [126]. In elite controllers of HIV, Oscillospira abundance showed a significant positive correlation with the CD4/CD8 ratio among HIV-positive individuals, indicating an association with reduced systemic inflammation within this cohort [64]. Furthermore, Oscillospira was found to be abundant in these individuals, and their microbiota profile closely resembled that of HIV-negative controls.

2.9. Roseburia

Roseburia is an SCFA-producing genus of Lachnospiraceae and important for gut health and metabolic regulation, thereby playing a protective role against gastrointestinal disorders and conditions like IBD and ulcerative colitis [127,128,129]. Higher levels of Roseburia are associated with improved metabolic health, including lower body weight and enhanced insulin sensitivity, which may help to prevent obesity and type 2 diabetes [130,131]. Furthermore, Roseburia may influence cardiovascular health by improving lipid profiles and reducing systemic inflammation [132] and reducing the risk of acute myocardial infarction [133], as well as having potential effects on mental health through the gut-brain axis [134].
In COVID-19, several studies have consistently reported decreased levels of Roseburia in patients. Albrich et al. (2022) [74], Tao et al. (2020) [135], Gu et al. (2020) [5], Moreira-Rosário et al. (2020) [99], Schult et al. (2022) [61], and Reinold et al. (2021) [45] observed lower Roseburia abundance in severe or high-risk COVID-19 cases. Fan et al. (2023) found reduced Roseburia abundance in severe compared to moderate cases [136], while Galperine et al. (2023) reported lower levels in ventilated versus non-ventilated patients [48]. These data suggest that lower levels of this beneficial bacterium may be associated with worse disease outcomes.
A few studies have observed changes in Roseburia abundance in other viral infections. In HIV, dysbiosis and reduced levels of Roseburia were linked to increased systemic inflammation and immune activation [56]. In addition, butyric acid levels were positively associated with Roseburia abundance [137]. In H9N7 patients, Roseburia abundance decreased drastically in intestinal samples compared to healthy controls [93]. Interestingly, Roseburia abundance was decreased in patients with neuroinfection when compared to controls [104].

2.10. Ruminococcus

Ruminococcus, as an SCFA-producing genus of Lachnospiraceae, plays an important role in human health through its ability to ferment complex carbohydrates, produce beneficial SCFAs, maintain gut barrier integrity, and modulate immune responses. Its presence is associated with metabolic health and reduced inflammation, making it an important component of a healthy gut microbiome [138,139]. Changes in Ruminococcus populations have been observed in several diseases, including Crohn’s disease [140], IBD [141] and major depression [142], while its increased relative abundance is associated with reduced cardiovascular risk [143].
In COVID-19, changes in Ruminococcus populations were observed in several studies. Albrich et al. (2022) found that Ruminococcus levels were lower in high-risk COVID-19 groups compared to healthy controls [74]. Similarly, Schult et al. (2022) observed a reduction in Ruminococcus abundance in severe and fatal COVID-19 cases [61], suggesting that lower levels of these beneficial bacteria may be associated with worse disease outcomes. Gaibani et al. (2021) noted enrichment of Ruminococcus populations in COVID-19 patients who had not been admitted to the ICU [23], indicating that higher levels of this genus may be associated with less severe disease. Zuo et al. (2020b) reported that COVID-19 patients treated with antibiotics showed a reduction in Ruminococcus obeum abundance compared to patients not treated with antibiotics [6]. Zhang et al. (2023a) identified that Ruminococcus abundance was also reduced in post-COVID-19 patients, depleted in symptomatic recovered patients, and was negatively related to C-reactive protein and natural killer cells [75], suggesting that the reduction in Ruminococcus abundance may also persist in the post-recovery phase, potentially aiding in the recovery process. This decrease in abundance across various stages of the disease highlights the potential importance of Ruminococcus in modulating the immune response and overall gut health during and after COVID-19, as well as the consequences of antibiotic treatments.
In COVID-19, changes in Ruminococcus populations have been observed across several studies. Albrich et al. (2022) [74] and Schult et al. (2022) [61] found lower levels of Ruminococcus in high-risk and severe COVID-19 cases, suggesting an association with worse outcomes. On the other hand, Gaibani et al. (2021) reported higher Ruminococcus levels in patients who avoided ICU admission [23], indicating a potential link to less severe disease. Zuo et al. (2020b) noted reduced Ruminococcus obeum abundance in antibiotic-treated patients [6], while Zhang et al. (2023a) found decreased Ruminococcus abundance in post-COVID-19 patients [75], associated with lower C-reactive protein and natural killer cells. These findings suggest that Ruminococcus may play a role in immune response and gut health during and after COVID-19, with antibiotic treatment potentially impacting its levels.
In other viral infections, significant changes in Ruminococcus abundance were also observed in different studies. Gu et al. (2020) demonstrated that the reduction in Ruminococcus populations is a biomarker capable of differentiating COVID-19 patients from H1N1 patients [5]. In H7N9 patients, Ruminococcus abundance was reduced in intestinal samples compared to healthy controls [93]. In patients with cirrhotic hepatitis C, Ruminococcus abundance is also characteristically reduced [144]. Members of this genus express Histo-blood group antigens (HBGA)-like molecules, which are putative receptors for viruses. In stool samples from children with rotavirus, a binding interaction between Ruminococcus and rotavirus was identified and confirmed in vitro [145]. The presence of Ruminococcus was also associated with lower IgA titers against rotavirus in adults, which demonstrates how this taxon can negatively affect rotavirus infection (Rodríguez-Díaz et al., 2017) [146]. In HIV, dysbiosis and reduced levels of Ruminococcus have been linked to increased systemic inflammation and immune activation [56].

3. Probiotic Supplementation as Adjunctive Treatment for COVID-19

Probiotic supplementation has been widely studied for its potential health benefits, particularly in maintaining gut health, modulating immune responses, and preventing or alleviating various diseases. Consequently, probiotics and microbiota-targeted therapies hold significant potential as adjunctive treatments for COVID-19. By modulating immune responses, enhancing gut barrier integrity, and preventing secondary infections, these therapies could improve clinical outcomes and reduce disease severity. Most studies of COVID-19 with probiotic supplementation have used Bifidobacterium or Lactobacillus (or a combination of both), as explored in our previous reviews focusing on these two genera [16,17,18]. While these probiotics have shown promise in modulating immune responses and improving gut health, there is a need to expand our current knowledge to include other beneficial genera explored in this study to fully understand their potential in managing COVID-19. Here, we summarize the potential mechanisms and benefits of using these genera as adjunctive treatment for COVID-19.

3.1. Akkermansia Supplementation

Akkermansia supplementation can modulate the immune system by promoting the production of anti-inflammatory cytokines and reducing pro-inflammatory responses [21]. This immune modulation could help to alleviate the hyperinflammatory response observed in severe COVID-19 cases. In addition, pasteurized Akkermansia muciniphila supplementation improves insulin sensitivity and reduces blood marker levels of liver dysfunction and inflammation [147]. Furthermore, oral administration of Akkermansia has also been shown to enhance mouse defenses against the H7N9 virus, significantly reducing viral loads of IL-6 and IL-1β in the lungs and levels of IL-6 and TNF-α in the blood during H7N9 infection. Therefore, suppressing these cytokines should improve the outcomes of the viral infection [148]. A membrane protein in Akkermansia, Amuc_1100, interacts with Toll-like receptor 2 (TLR) to bolster intestinal barrier function and can activate TLR4, increasing IL-10 production [22,149]. Currently, the effects of pasteurized Akkermansia muciniphila in reducing symptoms related to irritable bowel syndrome, as well as its potential benefits for anxiety and depression, along with its safety and tolerability, are being evaluated in clinical trials (NCT05348642). These studies could establish Akkermansia muciniphila as a viable therapeutic option for managing gut health, improving host antiviral immune responses, and addressing associated mental health conditions like anxiety and depression.
In addition, live Akkermansia muciniphila has been shown to protect against lethal sepsis in a murine model. This species can produce a tripeptide Arg-Lys-His (RKH), which also protects against organ damage associated with sepsis, reduces pulmonary inflammation, and decreases the overproduction of pro-inflammatory factors [150]. This protective ability against sepsis is particularly important in the context of COVID-19, as SARS-CoV-2 can induce sepsis independent of secondary bacterial or fungal infections [151].
Akkermansia muciniphila can also protect against Clostridioides difficile infection in mice, where its oral administration prevented weight loss; reduced histological colon damage; significantly lowered levels of G-CSF, MCP-1, IL-17A, IL-1α, IL-6, and TNF-α; and increased the production of SCFAs [152]. In vitro, both live and dead bacteria, as well as their supernatant, alleviated inflammation, improved intestinal barrier function, and modulated the production of IL-1β, TNF-α, and IL-10 [153]. The ability to prevent infections, like secondary infections, is essential in the context of COVID-19, as it can help to avoid additional complications and the deterioration of the patient’s health.

3.2. Bacteroides Supplementation

While direct studies on Bacteroides supplementation in COVID-19 are limited, the existing research supports further exploration of its potential benefits. In mouse models infected with Influenza A, administration of Bacteroides dorei led to increased survival, improved lung pathology, and reduced weight loss, lung index, and colon length [154]. It also reduced the viral load in lung tissue and increased the expression of type 1 interferon at 3 days post-infection. At 7 days post-infection, it decreased lung and serum levels of interferon, IL-1β, IL-6, TNF-α, IL-10, MCP-1, and IP-10. Bacteroides dorei treatment also altered the gut microbiota, which may contribute to the restoration of a healthy microbiota and its beneficial effects. Bacteroides thetaiotaomicron, a next-generation probiotic, is currently being developed as a biotherapeutic for Crohn’s disease. It has demonstrated good tolerability and safety in clinical trials, including the first human safety study, which is relevant for expanding its potential applications (NCT02704728) [155]. These bacteria have the ability to digest dietary fibers and host glycans while producing short-chain and organic acids, and their cell-surface architecture facilitates both interactions with and evasion of the host immune system [156]. These features make Bacteroides a promising candidate for supplementation, particularly in modulating the gut microbiome and enhancing immune responses, which are important for the treatment of inflammatory and infectious diseases, such as COVID-19.

3.3. Blautia Supplementation

Blautia supplementation may also modulate the immune system, promoting anti-inflammatory production and lessening pro-inflammatory responses, which could help to address hyperinflammatory reactions in severe COVID-19 cases [63]. Increasing Blautia abundance through dietary interventions, such as high-fiber diets, has been associated with improved gut health and reduced inflammation [63]. The efficacy and safety of Blautix, a strain of Blautia hydrogenotrophica, has been tested for irritable bowel syndrome, showing improvements in intestinal pain and bowel habits [157]. It was demonstrated in a mouse model that the administration of Blautia faecis protected against secondary bacterial infections caused by Streptococcus pneumoniae. Mice infected with Influenza A and treated with Blautia faecis showed reduced bacterial load in the lungs, lower bacterial counts in the spleen, indicating less bacterial dissemination from the lungs, and improved survival. This was associated with less post-influenza weight loss and faster weight recovery. Additionally, Blautia administration also reduced the expression of genes encoding inflammatory cytokines, such as TNFα, IL-1β, OAS3, and ISG15 [65]. These findings highlight the potential of Blautia, which plays an important role in modulating the inflammatory response and protecting against secondary bacterial infections, as observed in COVID-19.

3.4. Coprococcus Supplementation

In mouse models of colitis, the administration of Coprococcus eutactus improved colitis by significantly reducing levels of pro-inflammatory cytokines TNF-α, IL-1β, and IL-6, while increasing those of IL-10, IL-4, and IL-5 [158]. It also restored tight junction integrity and epithelial barrier function by elevating levels of key proteins and mucin genes Coprococcus can reduce inflammation and improve epithelial barrier integrity, both of which are important for controlling COVID-19, as these clinical features are often disrupted in the disease. Additionally, Coprococcus has been linked to improved mental health outcomes by producing metabolites that influence the gut–brain axis [159]. In this context, patients with irritable bowel syndrome exhibit microbiota profiles like those with depression, both showing a decrease in Coprococcus abundance [160]. This relationship suggests that intestinal inflammation may interfere with communication with the central nervous system [161], which is particularly relevant for COVID-19 patients, who often experience neurological and psychological symptoms. Future clinical trials should evaluate Coprococcus-based probiotics in COVID-19 treatment, focusing on disease severity, recovery time, and overall health, including potential benefits for neurological and psychological symptoms.

3.5. Faecalibacterium Supplementation

Several studies have shown that Faecalibacterium prausnitzii supplementation can reduce inflammation and improve gut barrier function in animal models of colitis, with ongoing clinical trials evaluating its efficacy in human IBD patients [162]. In mice, Faecalibacterium duncaniae provided protection against influenza infection, restoring SCFA levels in infected animals, impairing viral replication in the lungs, and reducing pulmonary inflammation, in addition to demonstrating potential protection against secondary bacterial infections [163]. In chronic kidney disease models, Faecalibacterium prausnitzii improved intestinal permeability and inflammation, primarily through the beneficial effects of butyrate produced by Faecalibacterium prausnitzii [164]. To increase the presence of Faecalibacterium prausnitzii in Crohn’s disease, a study evaluated the use of fructooligosaccharides to increase the abundance of this taxon (NCT02539849). Another clinical trial is assessing the EXL01 strain of Faecalibacterium prausnitzii for maintaining clinical response/remission induced by corticosteroids and preventing recurrence of Clostridioides difficile infection in high-risk patients, as a low abundance of Faecalibacterium prausnitzii is predictive of Clostridioides difficile recurrence (NCT06306014), with completion expected by 2027.
These studies indicate that supplementation with Faecalibacterium or strategies to increase its abundance may help reduce intestinal inflammation, improve gut barrier integrity, and regulate the immune system, as well as prevent recurrences of infections such as those caused by Clostridioides difficile, which may be particularly relevant in the context of COVID-19.

3.6. Lachnospira and Lachnospiraceae Supplementation

Lachnospira and Lachnospiraceae species produce SCFAs and may help manage hyperinflammatory responses in severe COVID-19 cases. Furthermore, Lachnospira and Lachnospiraceae are linked to reduced inflammation and improved gut health, making them potential therapeutic targets for COVID-19 patients with comorbidities like IBD and metabolic disorders [165,166]. Increasing the abundance of these bacteria through high-fiber diets has been associated with improved gut health and reduced inflammation [167]. Furthermore, isolates of Lachnospiraceae can significantly reduced colonization by Clostridioides difficile in mouse models and lowered levels of Clostridioides difficile cytotoxins, resulting in less severe clinical and histopathological signs and reduced mortality [168]. These findings suggest that Lachnospira and Lachnospiraceae contribute to gut health and inflammation reduction and have significant therapeutic potential for the prevention and management of secondary bacterial infections, as seen in COVID-19.

3.7. Oscillospira Supplementation

Different studies have shown that Oscillospira can produce a variety of SCFAs, especially butyrate [120,169,170], which indicates that the potential immune modulation by these bacteria may help to alleviate hyperinflammatory responses in severe COVID-19 cases. In addition, Oscillospira is associated with leanness and overall metabolic health [171], making it a potential therapeutic target for COVID-19 patients with comorbidities like obesity and metabolic syndrome [172]. By potentially improving gut health and enhancing the immune response, Oscillospira supplementation could serve as a therapeutic target for these vulnerable populations. Its ability to modulate inflammation and promote metabolic health may help to reduce the risks associated with COVID-19 in patients suffering from these comorbidities [172]. As studies exploring new probiotics continue to unfold, targeting Oscillospira may provide a novel approach to improving outcomes for individuals with underlying health issues during viral infections.

3.8. Roseburia Supplementation

By producing butyrate, Roseburia enhances gut barrier integrity, preventing the translocation of pathogens and toxins into the bloodstream, a key function given the gut barrier dysfunction often observed in COVID-19 patients [7]. The administration of a butyrate-producing strain of Roseburia hominis in rat models prevented the development of visceral hypersensitivity. In addition, supplementation prevented the reduction in mRNA expression of occludin protein, which is essential for the formation of tight junctions, and increased butyrate levels [173]. Supplementation with Roseburia intestinalis in murine models has also been shown to improve impaired colon barrier function, restore gastrointestinal function, and recover the microbiota, as well as reduce serum levels of IL-6 and IL-17 [129]. Furthermore, Roseburia intestinalis has the ability to reverse memory impairment [174], which is an important approach, especially for post-COVID-19 patients [175].
Roseburia abundance is also inversely correlated with the development of atherosclerotic lesions [132]. In this context, COVID-19 and atherosclerosis exhibit a bidirectional relationship, where cardiovascular diseases increase susceptibility to viral infections, and SARS-CoV-2 infection can damage the endothelium and cardiomyocytes, exacerbating atherosclerosis [176,177]. Targeting the gut microbiota to enhance Roseburia levels may offer a novel therapeutic approach that could improve outcomes for individuals with underlying heart conditions during viral infections. This strategy could promote gut health, modulate the immune system, and even reverse memory impairment, making it a promising approach for at-risk groups, acute diseases, and post-COVID-19 recovery.

3.9. Ruminococcus Supplementation

Ruminococcus species degrade complex polysaccharides and produce SCFAs, including butyrate [178]. Ruminococcus bromii, for example, is specialized in breaking down resistant starch [179], which is prevalent in foods such as cereal grains, beans, lentils, and certain fruits like plantains and green bananas. Through this process, Ruminococcus bromii produces SCFAs, including acetate, propionate, and butyrate, which are vital for gut health. Similarly, bifidobacteria are recognized for their substantial repertoire of starch-degrading enzymes, enabling them to effectively utilize resistant starch as well [180]. As such, the combination of strains of these two genera could significantly enhance starch degradation and SCFA production, particularly in the context of COVID-19. Notably, Ruminococcus bromii and Bifidobacterium adolescentis are known as primary degraders of resistant starch in the human gut [179]. Therefore, the combination of Ruminococcus bromii and Bifidobacterium adolescentis could not only enhance resistant starch degradation but also contribute to a more balanced immune response and reduced inflammation, helping to alleviate the adverse effects of the virus on the body. This integrated approach could open new avenues for dietary and probiotic interventions in patients affected by COVID-19.
In mouse models of induced colitis, the administration of Ruminococcus intestinalis significantly reduced IL-17 expression and improved pathological signs of inflammation in the colon compared to mice that did not receive the treatment [181]. Furthermore, in hamsters infected with SARS-CoV-2, administration of a Ruminococcus strain resulted in no clinical symptoms of SARS-CoV-2 infection, improved survival rates and body weight, and no signs of pneumonia were observed. Gross examination of the lung specimens demonstrated protective effects against the infection [182]. Additionally, Ruminococcus torques is being evaluated in healthy overweight individuals for its potential metabolic effects, including metabolism, energy, glucose regulation, and inflammatory response (NCT05448274). These studies suggest that different species of Ruminococcus may have protective and therapeutic properties in various conditions, such as intestinal inflammation, with the potential to modulate the immune system, improve metabolic health, and even address viral infections like COVID-19.

3.10. Key Points on Probiotics Supplementation and COVID-19 Treatment

The studies presented in this section indicate that various probiotic bacterial genera may significantly improve clinical outcomes and decrease the severity of COVID-19. These beneficial bacteria could positively influence patient health by reducing disease severity and speeding up recovery. Table 1 summarizes the mechanisms of action, health benefits, clinical evidence, and ongoing research for the different probiotic genera that have shown promise in the context of COVID-19 treatment. However, to confirm these advantages, future clinical trials are necessary to assess the effectiveness of probiotics derived from these species in treating COVID-19. Studies should concentrate on key factors like disease severity, recovery time, and overall patient well-being. Conducting these trials is of primary relevance to determining whether probiotic administration could serve as an effective strategy for managing COVID-19, particularly regarding clinical management and the quality of life for those affected. Ongoing investigations into the role of these microorganisms in influencing immune responses and supporting gut health show promise, potentially leading to new complementary treatments that alleviate the virus’s negative effects. Therefore, continuous exploration of the relationship between gut microbiota and COVID-19 is essential for developing new efficient therapeutic approaches.

4. Concluding Remarks

This review has explored the relationship between nine key genera (Akkermansia, Bacteroides, Blautia, Coprococcus, Faecalibacterium, Lachnospira, Oscillospira, Roseburia, and Ruminococcus), as well as the family Lachnospiraceae, and their relevance to COVID-19. Each of these genera plays a major role in maintaining gut homeostasis, producing SCFAs, and modulating systemic inflammation. Alterations in their abundance have been observed in COVID-19 patients, with trends suggesting that disruptions in populations of these beneficial bacteria are associated with worse disease outcomes. Furthermore, the relationship between these genera and other viral infections, such as HIV and influenza, suggests that maintaining a healthy gut microbiota could be beneficial in mitigating the effects of viral diseases. The production of SCFAs like butyrate by these bacteria enhances gut barrier integrity and reduces systemic inflammation, which are critical during viral infections. Importantly, emerging evidence suggests that the dysbiosis observed in COVID-19 may also play a role in the development of long COVID, a condition characterized by prolonged symptoms following acute infection. Understanding how these gut microbiota alterations contribute to long COVID could provide valuable insights for therapeutic interventions. In summary, maintaining a healthy microbiota, particularly the beneficial genera discussed in this review, is essential for immune function and overall health. As we continue to combat the COVID-19 pandemic, integrating microbiota-focused approaches into treatment strategies could provide new opportunities for improving patient outcomes during acute infection but also offer potential pathways for managing long COVID and enhancing recovery.

Author Contributions

P.H.R.: Conceptualization, Writing—Original Draft; C.R.T.: Writing—Review & Editing; J.d.S.: Writing—Review & Editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior—CAPES (No. 88887.513461/2020-00 and No. 88887.798411/2022-00).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Lloyd-Price, J.; Abu-Ali, G.; Huttenhower, C. The healthy human microbiome. Genome Med. 2016, 8, 51. [Google Scholar] [CrossRef] [PubMed]
  2. Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef] [PubMed]
  3. Lin, L.; Zhang, J. Role of intestinal microbiota and metabolites on gut homeostasis and human diseases. BMC Immunol. 2017, 18, 2. [Google Scholar] [CrossRef] [PubMed]
  4. Tilg, H.; Zmora, N.; Adolph, T.E.; Elinav, E. The intestinal microbiota fuelling metabolic inflammation. Nat. Rev. Immunol. 2020, 20, 40–54. [Google Scholar] [CrossRef]
  5. 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]
  6. Zuo, T.; Zhang, F.; Lui, G.C.Y.; Yeoh, Y.K.; Li, A.Y.L.; Zhan, H.; Wan, Y.; Chung, A.C.K.; Cheung, C.P.; Chen, N.; et al. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology 2020, 159, 944–955.e8. [Google Scholar] [CrossRef]
  7. 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]
  8. Mak, J.W.Y.; Chan, F.K.L.; Ng, S.C. Probiotics and COVID-19: One size does not fit all. Lancet Gastroenterol. Hepatol. 2020, 5, 644–645. [Google Scholar] [CrossRef]
  9. George Kerry, R.; 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]
  10. 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. 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]
  11. Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Gil-Campos, M.; Gil, A. Mechanisms of Action of Probiotics. Adv. Nutr. 2019, 10, S49–S66. [Google Scholar] [CrossRef] [PubMed]
  12. Jespersen, L.; Tarnow, I.; Eskesen, D.; Morberg, C.M.; Michelsen, B.; Bügel, S.; Dragsted, L.O.; Rijkers, G.T.; Calder, P.C. Effect of Lactobacillus paracasei subsp. paracasei, L. casei 431 on immune response to influenza vaccination and upper respiratory tract infections in healthy adult volunteers: A randomized, double-blind, placebo-controlled, parallel-group study. Am. J. Clin. Nutr. 2015, 101, 1188–1196. [Google Scholar] [CrossRef] [PubMed]
  13. Luoto, R.; Ruuskanen, O.; Waris, M.; Kalliomäki, M.; Salminen, S.; Isolauri, E. Prebiotic and probiotic supplementation prevents rhinovirus infections in preterm infants: A randomized, placebo-controlled trial. J. Allergy Clin. Immunol. 2014, 133, 405–413. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, F.; Lau, R.I.; Liu, Q.; Su, Q.; Chan, F.K.L.; Ng, S.C. Gut microbiota in COVID-19: Key microbial changes, potential mechanisms and clinical applications. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 323–337. [Google Scholar] [CrossRef]
  15. 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]
  16. Taufer, C.R.; da Silva, J.; Rampelotto, P.H. The Influence of Probiotic Lactobacilli on COVID-19 and the Microbiota. Nutrients 2024, 16, 1350. [Google Scholar] [CrossRef] [PubMed]
  17. Taufer, C.R.; Rampelotto, P.H. Lactobacilli in COVID-19: A Systematic Review Based on Next-Generation Sequencing Studies. Microorganisms 2024, 12, 284. [Google Scholar] [CrossRef]
  18. Taufer, C.R.; Rampelotto, P.H. The Role of Bifidobacterium in COVID-19: A Systematic Review. Life 2023, 13, 1847. [Google Scholar] [CrossRef]
  19. Derrien, M.; Vaughan, E.E.; Plugge, C.M.; de Vos, W.M. Akkermansia municiphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol. 2004, 54, 1469–1476. [Google Scholar] [CrossRef]
  20. Dao, M.C.; Everard, A.; Aron-Wisnewsky, J.; Sokolovska, N.; Prifti, E.; Verger, E.O.; Kayser, B.D.; Levenez, F.; Chilloux, J.; Hoyles, L.; et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: Relationship with gut microbiome richness and ecology. Gut 2016, 65, 426–436. [Google Scholar] [CrossRef]
  21. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef]
  22. Plovier, H.; Everard, A.; Druart, C.; Depommier, C.; Van Hul, M.; Geurts, L.; Chilloux, J.; Ottman, N.; Duparc, T.; Lichtenstein, L.; et al. A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat. Med. 2017, 23, 107–113. [Google Scholar] [CrossRef] [PubMed]
  23. Gaibani, P.; D’Amico, F.; Bartoletti, M.; Lombardo, D.; Rampelli, S.; Fornaro, G.; Coladonato, S.; Siniscalchi, A.; Re, M.C.; Viale, P.; et al. The gut microbiota of critically Ill patients with COVID-19. Front. Cell. Infect. Microbiol. 2021, 11, 670424. [Google Scholar] [CrossRef]
  24. Galeeva, J.S.; Fedorov, D.E.; Starikova, E.V.; Manolov, A.I.; Pavlenko, A.V.; Selezneva, O.V.; Klimina, K.M.; Veselovsky, V.A.; Morozov, M.D.; Yanushevich, O.O.; et al. Microbial Signatures in COVID-19: Distinguishing Mild and Severe Disease via Gut Microbiota. Biomedicines 2024, 12, 996. [Google Scholar] [CrossRef] [PubMed]
  25. Leftwich, H.K.; Vargas-Robles, D.; Rojas-Correa, M.; Yap, Y.R.; Bhattarai, S.; Ward, D.V.; Fujimori, G.; Forconi, C.S.; Yeboah, T.; Carter, A.; et al. The microbiota of pregnant women with SARS-CoV-2 and their infants. Microbiome 2023, 11, 141. [Google Scholar] [CrossRef]
  26. Lin, R.; Xiao, M.; Cao, S.; Sun, Y.; Zhao, L.; Mao, X.; Chen, P.; Tong, X.; Ou, Z.; Zhu, H.; et al. Distinct gut microbiota and health outcomes in asymptomatic infection, viral nucleic acid test re-positive, and convalescent COVID-19 cases. mLife 2022, 1, 183–197. [Google Scholar] [CrossRef]
  27. Mańkowska-Wierzbicka, D.; Zuraszek, J.; Wierzbicka, A.; Gabryel, M.; Mahadea, D.; Baturo, A.; Zakerska-Banaszak, O.; Slomski, R.; Skrzypczak-Zielinska, M.; Dobrowolska, A. Alterations in Gut Microbiota Composition in Patients with COVID-19: A Pilot Study of Whole Hypervariable 16S rRNA Gene Sequencing. Biomedicines 2023, 11, 367. [Google Scholar] [CrossRef] [PubMed]
  28. Talaga-Ćwiertnia, K.; Sroka-Oleksiak, A.; Zapała, B.; Salamon, D.; Krawczyk, A.; Brzychczy-Włoch, M.; Gosiewski, T. New insights into diversity of the upper respiratory tract microbiota and its relationship with SARS-CoV-2 viral load in the nasopharyngeal epithelial cells in patients with COVID-19. Polish Arch. Intern. Med. 2023, 133, 16442. [Google Scholar] [CrossRef]
  29. Brzychczy-Sroka, B.; Talaga-Ćwiertnia, K.; Sroka-Oleksiak, A.; Gurgul, A.; Zarzecka-Francica, E.; Ostrowski, W.; Kąkol, J.; Zarzecka, J.; Brzychczy-Włoch, M. Oral microbiota study of the patients after hospitalisation for COVID-19, considering selected dental indices and antibiotic therapy using the next generation sequencing method (NGS). J. Oral Microbiol. 2023, 15, 2264591. [Google Scholar] [CrossRef]
  30. Ferreira-Junior, A.S.; Borgonovi, T.F.; De Salis, L.V.V.; Leite, A.Z.; Dantas, A.S.; De Salis, G.V.V.; Cruz, G.N.F.; De Oliveira, L.F.V.; Gomes, E.; Penna, A.L.B.; et al. Detection of Intestinal Dysbiosis in Post-COVID-19 Patients One to Eight Months after Acute Disease Resolution. Int. J. Environ. Res. Public Health 2022, 19, 189. [Google Scholar] [CrossRef]
  31. Romani, L.; Del Chierico, F.; Macari, G.; Pane, S.; Ristori, M.V.; Guarrasi, V.; Gardini, S.; Pascucci, G.R.; Cotugno, N.; Perno, C.F.; et al. The Relationship Between Pediatric Gut Microbiota and SARS-CoV-2 Infection. Front. Cell. Infect. Microbiol. 2022, 12, 908492. [Google Scholar] [CrossRef] [PubMed]
  32. Romani, L.; Del Chierico, F.; Pane, S.; Ristori, M.V.; Pirona, I.; Guarrasi, V.; Cotugno, N.; Bernardi, S.; Lancella, L.; Perno, C.F.; et al. Exploring nasopharyngeal microbiota profile in children affected by SARS-CoV-2 infection. Microbiol. Spectr. 2024, 12, e0300923. [Google Scholar] [CrossRef] [PubMed]
  33. Newsome, R.C.; Gharaibeh, R.Z.; Gauthier, J.; Hernandez, M.C.; Abraham, G.E.; Robinson, T.; Williams, H.; Sloan, M.; Owings, A.H.; Laird, H.; et al. The Gut Microbiome Of COVID-19 Recovered Patients Returns To Uninfected Status In A Minority-Dominated United States Cohort. Gastroenterology 2021, 160, S-371. [Google Scholar] [CrossRef]
  34. Abange, W.B.; Martin, C.; Nanfack, A.J.; Yatchou, L.G.; Nusbacher, N.; Nguedia, C.A.; Kamga, H.G.; Fokam, J.; Kennedy, S.P.; Ndjolo, A.; et al. Alteration of the gut fecal microbiome in children living with HIV on antiretroviral therapy in Yaounde, Cameroon. Sci. Rep. 2021, 11, 7666. [Google Scholar] [CrossRef] [PubMed]
  35. Xie, J.; Li, H.; Zhang, X.; Yang, T.; Yue, M.; Zhang, Y.; Chen, S.; Cui, N.; Yuan, C.; Li, J.; et al. Akkermansia muciniphila protects mice against an emerging tick-borne viral pathogen. Nat. Microbiol. 2023, 8, 91–106. [Google Scholar] [CrossRef]
  36. Heumel, S.; de Rezende Rodovalho, V.; Urien, C.; Specque, F.; Brito Rodrigues, P.; Robil, C.; Delval, L.; Sencio, V.; Descat, A.; Deruyter, L.; et al. Shotgun metagenomics and systemic targeted metabolomics highlight indole-3-propionic acid as a protective gut microbial metabolite against influenza infection. Gut Microbes 2024, 16, 2325067. [Google Scholar] [CrossRef]
  37. Hu, X.; Zhao, Y.; Yang, Y.; Gong, W.; Sun, X.; Yang, L.; Zhang, Q.; Jin, M. Akkermansia muciniphila Improves Host Defense Against Influenza Virus Infection. Front. Microbiol. 2021, 11, 586476. [Google Scholar] [CrossRef]
  38. Labarta-Bajo, L.; Gramalla-Schmitz, A.; Gerner, R.R.; Kazane, K.R.; Humphrey, G.; Schwartz, T.; Sanders, K.; Swafford, A.; Knight, R.; Raffatellu, M.; et al. CD8 T cells drive anorexia, dysbiosis, and blooms of a commensal with immunosuppressive potential after viral infection. Proc. Natl. Acad. Sci. USA 2020, 117, 24998–25007. [Google Scholar] [CrossRef]
  39. Portincasa, P.; Bonfrate, L.; Vacca, M.; De Angelis, M.; Farella, I.; Lanza, E.; Khalil, M.; Wang, D.Q.H.; Sperandio, M.; Di Ciaula, A. Gut Microbiota and Short Chain Fatty Acids: Implications in Glucose Homeostasis. Int. J. Mol. Sci. 2022, 23, 1105. [Google Scholar] [CrossRef]
  40. Bloom, S.M.; Bijanki, V.N.; Nava, G.M.; Sun, L.; Malvin, N.P.; Donermeyer, D.L.; Dunne, W.M.; Allen, P.M.; Stappenbeck, T.S. Commensal Bacteroides species induce colitis in host-genotype-specific fashion in a mouse model of inflammatory bowel disease. Cell Host Microbe 2011, 9, 390–403. [Google Scholar] [CrossRef]
  41. Zhou, Y.; Zhi, F. Lower Level of Bacteroides in the Gut Microbiota Is Associated with Inflammatory Bowel Disease: A Meta-Analysis. Biomed Res. Int. 2016, 2016, 5828959. [Google Scholar] [CrossRef] [PubMed]
  42. Brook, I. The role of anaerobic bacteria in bacteremia. Anaerobe 2010, 16, 183–189. [Google Scholar] [CrossRef] [PubMed]
  43. Majewska, A.; Kierzkowska, M.; Kawecki, D. What we actually know about the pathogenicity of Bacteroides pyogenes. Med. Microbiol. Immunol. 2021, 210, 157–163. [Google Scholar] [CrossRef] [PubMed]
  44. Al Bataineh, M.T.; Henschel, A.; Mousa, M.; Daou, M.; Waasia, F.; Kannout, H.; Khalili, M.; Kayasseh, M.A.; Alkhajeh, A.; Uddin, M.; et al. Gut Microbiota Interplay with COVID-19 Reveals Links to Host Lipid Metabolism Among Middle Eastern Populations. Front. Microbiol. 2021, 12, 761067. [Google Scholar] [CrossRef]
  45. Reinold, J.; Farahpour, F.; Fehring, C.; Dolff, S.; Konik, M.; Korth, J.; van Baal, L.; Hoffmann, D.; Buer, J.; Witzke, O.; et al. A Pro-Inflammatory Gut Microbiome Characterizes SARS-CoV-2 Infected Patients and a Reduction in the Connectivity of an Anti-Inflammatory Bacterial Network Associates with Severe COVID-19. Front. Cell. Infect. Microbiol. 2021, 11, 747816. [Google Scholar] [CrossRef]
  46. Mizutani, T.; Ishizaka, A.; Koga, M.; Ikeuchi, K.; Saito, M.; Adachi, E.; Yamayoshi, S.; Iwatsuki-Horimoto, K.; Yasuhara, A.; Kiyono, H.; et al. Correlation Analysis between Gut Microbiota Alterations and the Cytokine Response in Patients with Coronavirus Disease during Hospitalization. Microbiol. Spectr. 2022, 10, e0168921. [Google Scholar] [CrossRef]
  47. Bucci, V.; Ward, D.V.; Bhattarai, S.; Rojas-Correa, M.; Purkayastha, A.; Holler, D.; Qu, M.D.; Mitchell, W.G.; Yang, J.; Fountain, S.; et al. The intestinal microbiota predicts COVID-19 severity and fatality regardless of hospital feeding method. mSystems 2023, 8, 6–12. [Google Scholar] [CrossRef]
  48. Galperine, T.; Choi, Y.; Pagani, J.L.; Kritikos, A.; Papadimitriou-Olivgeris, M.; Méan, M.; Scherz, V.; Opota, O.; Greub, G.; Guery, B.; et al. Temporal changes in fecal microbiota of patients infected with COVID-19: A longitudinal cohort. BMC Infect. Dis. 2023, 23, 537. [Google Scholar] [CrossRef]
  49. Geva-Zatorsky, N.; Sefik, E.; Kua, L.; Pasman, L.; Tan, T.G.; Ortiz-Lopez, A.; Yanortsang, T.B.; Yang, L.; Jupp, R.; Mathis, D.; et al. Mining the human gut microbiota for immunomodulatory organisms. Cell 2017, 168, 928–943.e11. [Google Scholar] [CrossRef]
  50. Lu, H.F.; Li, A.; Zhang, T.; Ren, Z.G.; He, K.X.; Zhang, H.; Yang, J.Z.; Luo, Q.X.; Zhou, K.; Chen, C.L.; et al. Disordered oropharyngeal microbial communities in h7n9 patients with or without secondary bacterial lung infection. Emerg. Microbes Infect. 2017, 6, 1–11. [Google Scholar] [CrossRef] [PubMed]
  51. Ramakrishna, C.; Kujawski, M.; Chu, H.; Li, L.; Mazmanian, S.K.; Cantin, E.M. Bacteroides fragilis polysaccharide A induces IL-10 secreting B and T cells that prevent viral encephalitis. Nat. Commun. 2019, 10, 2153. [Google Scholar] [CrossRef] [PubMed]
  52. Stefan, K.L.; Kim, M.V.; Iwasaki, A.; Kasper, D.L. Commensal Microbiota Modulation of Natural Resistance to Virus Infection. Cell 2020, 183, 1312–1324.e10. [Google Scholar] [CrossRef] [PubMed]
  53. Mazmanian, S.K.; Round, J.L.; Kasper, D.L. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 2008, 453, 620–625. [Google Scholar] [CrossRef]
  54. Niu, J.; Cui, M.; Yang, X.; Li, J.; Yao, Y.; Guo, Q.; Lu, A.; Qi, X.; Zhou, D.; Zhang, C.; et al. Microbiota-derived acetate enhances host antiviral response via NLRP3. Nat. Commun. 2023, 14, 642. [Google Scholar] [CrossRef] [PubMed]
  55. 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]
  56. Vujkovic-Cvijin, I.; Dunham, R.M.; Iwai, S.; Maher, M.C.; Albright, R.G.; Broadhurst, M.J.; Hernandez, R.D.; Lederman, M.M.; Huang, Y.; Somsouk, M.; et al. Dysbiosis of the Gut Microbiota Is Associated with HIV Disease Progression and Tryptophan Catabolism. Sci. Transl. Med. 2013, 5, 557–562. [Google Scholar] [CrossRef]
  57. Liu, X.; Mao, B.; Gu, J.; Wu, J.; Cui, S.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Blautia—A new functional genus with potential probiotic properties? Gut Microbes 2021, 13, 1875796. [Google Scholar] [CrossRef] [PubMed]
  58. Baxter, N.T.; Ruffin, M.T.; Rogers, M.A.M.; Schloss, P.D. Microbiota-based model improves the sensitivity of fecal immunochemical test for detecting colonic lesions. Genome Med. 2016, 8, 37. [Google Scholar] [CrossRef]
  59. Maeda, Y.; Motooka, D.; Kawasaki, T.; Oki, H.; Noda, Y.; Adachi, Y.; Niitsu, T.; Okamoto, S.; Tanaka, K.; Fukushima, K.; et al. Longitudinal alterations of the gut mycobiota and microbiota on COVID-19 severity. BMC Infect. Dis. 2022, 22, 572. [Google Scholar] [CrossRef]
  60. Wu, Y.; Cheng, X.; Jiang, G.; Tang, H.; Ming, S.; Tang, L.; Lu, J.; Guo, C.; Shan, H.; Huang, X. Altered oral and gut microbiota and its association with SARS-CoV-2 viral load in COVID-19 patients during hospitalization. npj Biofilms Microbiomes 2021, 7, 61. [Google Scholar] [CrossRef]
  61. Schult, D.; Reitmeier, S.; Koyumdzhieva, P.; Lahmer, T.; Middelhoff, M.; Erber, J.; Schneider, J.; Kager, J.; Frolova, M.; Horstmann, J.; et al. Gut bacterial dysbiosis and instability is associated with the onset of complications and mortality in COVID-19. Gut Microbes 2022, 14, 2031840. [Google Scholar] [CrossRef]
  62. Xu, N.; Zhang, W.; Huo, J.; Tao, R.; Jin, T.; Zhang, Y.; Wang, Y.; Zhu, L.; Li, J.J.; Yao, Q.; et al. Characterization of changes in the intestinal microbiome following combination therapy with zinc preparation and conventional treatment for children with rotavirus enteritis. Front. Cell. Infect. Microbiol. 2023, 13, 1153701. [Google Scholar] [CrossRef]
  63. Verstraeten, S.; Sencio, V.; Raise, A.; Huillet, E.; Layec, S.; Deruyter, L.; Heumel, S.; Auger, S.; Robert, V.; Langella, P.; et al. Description of a Newly Isolated Blautia faecis Strain and Its Benefit in Mouse Models of Post-Influenza Secondary Enteric and Pulmonary Infections. Nutrients 2022, 14, 1478. [Google Scholar] [CrossRef]
  64. Vesterbacka, J.; Rivera, J.; Noyan, K.; Parera, M.; Neogi, U.; Calle, M.; Paredes, R.; Sönnerborg, A.; Noguera-Julian, M.; Nowak, P. Richer gut microbiota with distinct metabolic profile in HIV infected Elite Controllers. Sci. Rep. 2017, 7, 6269. [Google Scholar] [CrossRef]
  65. Mutlu, E.A.; Keshavarzian, A.; Losurdo, J.; Swanson, G.; Siewe, B.; Forsyth, C.; French, A.; DeMarais, P.; Sun, Y.; Koenig, L.; et al. A Compositional Look at the Human Gastrointestinal Microbiome and Immune Activation Parameters in HIV Infected Subjects. PLoS Pathog. 2014, 10, e1003829. [Google Scholar] [CrossRef]
  66. Vázquez-Castellanos, J.F.; Serrano-Villar, S.; Latorre, A.; Artacho, A.; Ferrús, M.L.; Madrid, N.; Vallejo, A.; Sainz, T.; Martínez-Botas, J.; Ferrando-Martínez, S.; et al. Altered metabolism of gut microbiota contributes to chronic immune activation in HIV-infected individuals. Mucosal Immunol. 2015, 8, 760–772. [Google Scholar] [CrossRef]
  67. Chuaypen, N.; Jinato, T.; Avihingsanon, A.; Nookaew, I.; Tanaka, Y.; Tangkijvanich, P. Long-term benefit of DAAs on gut dysbiosis and microbial translocation in HCV-infected patients with and without HIV coinfection. Sci. Rep. 2023, 13, 14413. [Google Scholar] [CrossRef]
  68. Naderpoor, N.; Mousa, A.; Gomez-Arango, L.F.; Barrett, H.L.; Nitert, M.D.; Courten, B.D. Faecal microbiota are related to insulin sensitivity and secretion in overweight or obese adults. J. Clin. Med. 2019, 8, 452. [Google Scholar] [CrossRef] [PubMed]
  69. Wang, W.; Wang, F.; Li, Y.; Shi, Y.; Wang, X.; Chen, X.; Zheng, W.; Hsing, J.C.; Lu, Y.; Wu, Y.S.; et al. Distinct Gut Microbiota Profiles in Normal Weight Obesity and Their Association with Cardiometabolic Diseases: Results from Two Independent Cohort Studies. J. Cachexia Sarcopenia Muscle 2025, 16, e13644. [Google Scholar] [CrossRef]
  70. LeGates, T.A.; Altimus, C.M.; Wang, H.; Lee, H.-K.; Yang, S.; Zhao, H.; Kirkwood, A.; Weber, E.T.; Hattar, S. Aberrant light directly impairs mood and learning through melanopsin-expressing neurons. Nature 2012, 491, 594–598. [Google Scholar] [CrossRef]
  71. Lepage, P.; Leclerc, M.C.; Joossens, M.; Mondot, S.; Blottière, H.M.; Raes, J.; Ehrlich, D.; Doré, J. A metagenomic insight into our gut’s microbiome. Gut 2013, 62, 146–158. [Google Scholar] [CrossRef] [PubMed]
  72. Malhotra, K.; Katsanos, A.H.; Lambadiari, V.; Goyal, N.; Palaiodimou, L.; Kosmidou, M.; Krogias, C.; Alexandrov, A.V.; Tsivgoulis, G. GLP-1 receptor agonists in diabetes for stroke prevention: A systematic review and meta-analysis. J. Neurol. 2020, 267, 2117–2122. [Google Scholar] [CrossRef] [PubMed]
  73. Li, Z.; Liang, H.; Hu, Y.; Lu, L.; Zheng, C.; Fan, Y.; Wu, B.; Zou, T.; Luo, X.; Zhang, X.; et al. Gut bacterial profiles in Parkinson’s disease: A systematic review. CNS Neurosci. Ther. 2023, 29, 140–157. [Google Scholar] [CrossRef] [PubMed]
  74. Albrich, W.C.; Ghosh, T.S.; Ahearn-Ford, S.; Mikaeloff, F.; Lunjani, N.; Forde, B.; Suh, N.; Kleger, G.-R.; Pietsch, U.; Frischknecht, M.; et al. A high-risk gut microbiota configuration associates with fatal hyperinflammatory immune and metabolic responses to SARS-CoV-2. Gut Microbes 2022, 14, 2073131. [Google Scholar] [CrossRef]
  75. Zhang, D.; Zhou, Y.; Ma, Y.; Chen, P.; Tang, J.; Yang, B.; Li, H.; Liang, M.; Xue, Y.; Liu, Y.; et al. Gut Microbiota Dysbiosis Correlates with Long COVID-19 at One-Year After Discharge. J. Korean Med. Sci. 2023, 38, e120. [Google Scholar] [CrossRef]
  76. Li, S.; Yang, S.; Zhou, Y.; Disoma, C.; Dong, Z.; Du, A.; Zhang, Y.; Chen, Y.; Huang, W.; Chen, J.; et al. Microbiome Profiling Using Shotgun Metagenomic Sequencing Identified Unique Microorganisms in COVID-19 Patients With Altered Gut Microbiota. Front. Microbiol. 2021, 12, 712081. [Google Scholar] [CrossRef]
  77. Cai, J.; Xu, J.; Tan, Y.; Xiang, Y.; Li, Z.; Zheng, J.; Li, Y. Gut microbiota alteration and its association with immune function in post-COVID-19 patients. Folia Microbiol. 2024, 69, 857–864. [Google Scholar] [CrossRef]
  78. Belkaid, Y.; Hand, T.W. Role of the Microbiota in Immunity and Inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef]
  79. Zhao, H.; Feng, A.; Luo, D.; Wu, H.; Zhang, G.; Zhang, L.; Yuan, J.; Lin, Y.-F.; Li, L.; Zou, H. Altered gut microbiota is associated with different immunologic responses to antiretroviral therapy in HIV-infected men who have sex with men. J. Med. Virol. 2023, 95, e28674. [Google Scholar] [CrossRef]
  80. Louis, P.; Flint, H.J. Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol. Lett. 2009, 294, 1–8. [Google Scholar] [CrossRef]
  81. Jia, W.; Whitehead, R.N.; Griffiths, L.; Dawson, C.; Waring, R.H.; Ramsden, D.B.; Hunter, J.O.; Cole, J.A. Is the abundance of Faecalibacterium prausnitzii relevant to Crohn’s disease? FEMS Microbiol. Lett. 2010, 310, 138–144. [Google Scholar] [CrossRef] [PubMed]
  82. Leylabadlo, H.E.; Ghotaslou, R.; Feizabadi, M.M.; Farajnia, S.; Moaddab, S.Y.; Ganbarov, K.; Khodadadi, E.; Tanomand, A.; Sheykhsaran, E.; Yousefi, B.; et al. The critical role of Faecalibacterium prausnitzii in human health: An overview. Microb. Pathog. 2020, 149, 104344. [Google Scholar] [CrossRef] [PubMed]
  83. Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermúdez-Humarán, L.G.; Gratadoux, J.-J.; Blugeon, S.; Bridonneau, C.; Furet, J.-P.; Corthier, G.; et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl. Acad. Sci. USA 2008, 105, 16731–16736. [Google Scholar] [CrossRef] [PubMed]
  84. Maioli, T.U.; Borras-Nogues, E.; Torres, L.; Barbosa, S.C.; Martins, V.D.; Langella, P.; Azevedo, V.A.; Chatel, J.M. Possible Benefits of Faecalibacterium prausnitzii for Obesity-Associated Gut Disorders. Front. Pharmacol. 2021, 12, 740636. [Google Scholar] [CrossRef]
  85. Xuan, W.; Ou, Y.; Chen, W.; Huang, L.; Wen, C.; Huang, G.; Tang, W.; Zeng, D.; Huang, S.; Xiao, L.; et al. Faecalibacterium prausnitzii Improves Lipid Metabolism Disorder and Insulin Resistance in Type 2 Diabetic Mice. Br. J. Biomed. Sci. 2023, 80, 10794. [Google Scholar] [CrossRef] [PubMed]
  86. Zhao, H.; Xu, H.; Chen, S.; He, J.; Zhou, Y.; Nie, Y. Systematic review and meta-analysis of the role of Faecalibacterium prausnitzii alteration in inflammatory bowel disease. J. Gastroenterol. Hepatol. 2021, 36, 320–328. [Google Scholar] [CrossRef]
  87. Hazan, S.; Stollman, N.; Bozkurt, H.S.; Dave, S.; Papoutsis, A.J.; Daniels, J.; Barrows, B.D.; Quigley, E.M.; Borody, T.J. Lost microbes of COVID-19: Bifidobacterium, Faecalibacterium depletion and decreased microbiome diversity associated with SARS-CoV-2 infection severity. BMJ Open Gastroenterol. 2022, 9, e000871. [Google Scholar] [CrossRef]
  88. Tang, L.; Gu, S.; Gong, Y.; Li, B.; Lu, H.; Li, Q.; Zhang, R.; Gao, X.; Wu, Z.; Zhang, J.; et al. Clinical Significance of the Correlation between Changes in the Major Intestinal Bacteria Species and COVID-19 Severity. Engineering 2020, 6, 1178–1184. [Google Scholar] [CrossRef]
  89. Bernard-Raichon, L.; Venzon, M.; Klein, J.; Axelrad, J.E.; Zhang, C.; Sullivan, A.P.; Hussey, G.A.; Casanovas-Massana, A.; Noval, M.G.; Valero-Jimenez, A.M.; et al. Gut microbiome dysbiosis in antibiotic-treated COVID-19 patients is associated with microbial translocation and bacteremia. Nat Commun. 2022, 13, 5926. [Google Scholar] [CrossRef]
  90. Seibert, B.; Cáceres, C.J.; Cardenas-Garcia, S.; Carnaccini, S.; Geiger, G.; Rajao, D.S.; Ottesen, E.; Perez, D.R. Mild and Severe SARS-CoV-2 Infection Induces Respiratory and Intestinal Microbiome Changes in the K18-hACE2 Transgenic Mouse Model. Microbiol Spectr. 2021, 9, e0053621. [Google Scholar] [CrossRef]
  91. Sencio, V.; Machelart, A.; Robil, C.; Benech, N.; Hoffmann, E.; Galbert, C.; Deryuter, L.; Heumel, S.; Hantute-Ghesquier, A.; Flourens, A.; et al. Alteration of the gut microbiota following SARS-CoV-2 infection correlates with disease severity in hamsters. Gut Microbes 2022, 14, 2018900. [Google Scholar] [CrossRef] [PubMed]
  92. Liao, Q.; Wang, F.; Zhou, W.; Liao, G.; Zhang, H.; Shu, Y.; Chen, Y. Identification of Causal Relationships between Gut Microbiota and Influenza a Virus Infection in Chinese by Mendelian Randomization. Microorganisms 2024, 12, 1170. [Google Scholar] [CrossRef] [PubMed]
  93. Qin, N.; Zheng, B.; Yao, J.; Guo, L.; Zuo, J.; Wu, L.; Zhou, J.; Liu, L.; Guo, J.; Ni, S.; et al. Influence of H7N9 virus infection and associated treatment on human gut microbiota. Sci. Rep. 2015, 5, 14771. [Google Scholar] [CrossRef] [PubMed]
  94. Xie, Y.; Sun, J.; Wei, L.; Jiang, H.; Hu, C.; Yang, J.; Huang, Y.; Ruan, B.; Zhu, B. Altered gut microbiota correlate with different immune responses to HAART in HIV-infected individuals. BMC Microbiol. 2021, 21, 11. [Google Scholar] [CrossRef]
  95. Tsai, C.-C.; Chiu, M.-H.; Kek, H.-P.; Yang, M.-C.; Su, Y.-T.; Liu, H.-K.; Wu, M.-S.; Yeh, Y.-T. The Reduced Gut Lachnospira Species Is Linked to Liver Enzyme Elevation and Insulin Resistance in Pediatric Fatty Liver Disease. Int. J. Mol. Sci. 2024, 25, 3640. [Google Scholar] [CrossRef]
  96. Zhao, L.; Zhang, F.; Ding, X.; Wu, G.; Lam, Y.Y.; Wang Xuejiao Fu, H.; Xue, X.; Lu, C.; Ma, J.; Yu, L.; et al. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science 2018, 80, 359, 1151–1156. [Google Scholar] [CrossRef]
  97. Ghezzi, L.; Cantoni, C.; Rotondo, E.; Galimberti, D. The Gut Microbiome–Brain Crosstalk in Neurodegenerative Diseases. Biomedicines 2022, 10, 1486. [Google Scholar] [CrossRef]
  98. Li, H.; Cui, X.; Lin, Y.; Huang, F.; Tian, A.; Zhang, R. Gut microbiota changes in patients with Alzheimer’s disease spectrum based on 16S rRNA sequencing: A systematic review and meta-analysis. Front. Aging Neurosci. 2024, 16, 1422350. [Google Scholar] [CrossRef]
  99. Moreira-Rosário, A.; Marques, C.; Pinheiro, H.; Araújo, J.R.; Ribeiro, P.; Rocha, R.; Mota, I.; Pestana, D.; Ribeiro, R.; Pereira, A.; et al. Gut Microbiota Diversity and C-Reactive Protein Are Predictors of Disease Severity in COVID-19 Patients. Front. Microbiol. 2021, 12, 705020. [Google Scholar] [CrossRef]
  100. Serrano-Villar, S.; Vázquez-Castellanos, J.F.; Vallejo, A.; Latorre, A.; Sainz, T.; Ferrando-Martínez, S.; Rojo, D.; Martínez-Botas, J.; Del Romero, J.; Madrid, N.; et al. The effects of prebiotics on microbial dysbiosis, butyrate production and immunity in HIV-infected subjects. Mucosal Immunol. 2017, 10, 1279–1293. [Google Scholar] [CrossRef]
  101. Zhou, J.; Zhang, Y.; Ye, L.; Cui, P.; Luo, L.; Chen, H.; Liang, B.; Ning, C.; Tian, L.; Zhong, X.; et al. Gut Microbiome Changes Associated with HIV Infection and Antiretroviral Therapy. SSRN Electron. J. 2020, 10, 434. [Google Scholar] [CrossRef]
  102. Manzano, M.; Talavera-Rodríguez, A.; Moreno, E.; Madrid, N.; Gosalbes, M.J.; Ron, R.; Dronda, F.; Pérez-Molina, J.A.; Lanza, V.F.; Díaz, J.; et al. Relationship of Diet to Gut Microbiota and Inflammatory Biomarkers in People with HIV. Nutrients 2022, 14, 1221. [Google Scholar] [CrossRef]
  103. Pérez-Matute, P.; Íñiguez, M.; Villanueva-Millán, M.J.; Recio-Fernández, E.; Vázquez, A.M.; Sánchez, S.C.; Morano, L.E.; Oteo, J.A. Short-term effects of direct-acting antiviral agents on inflammation and gut microbiota in hepatitis C-infected patients. Eur. J. Intern. Med. 2019, 67, 47–58. [Google Scholar] [CrossRef]
  104. Grochowska, M.; Laskus, T.; Paciorek, M.; Pollak, A.; Lechowicz, U.; Makowiecki, M.; Horban, A.; Radkowski, M.; Perlejewski, K. Patients with Infections of The Central Nervous System Have Lowered Gut Microbiota Alpha Diversity. Curr. Issues Mol. Biol. 2022, 44, 2903–2914. [Google Scholar] [CrossRef] [PubMed]
  105. Stackebrandt, E. The Family Lachnospiraceae. In The Prokaryotes: Firmicutes and Tenericutes; Rosenberg, E., DeLong, E.F., Lory, S., Stackebrandt, E., Thompson, F., Eds.; Springer: Berlin/Heidelberg, Germany, 2014; pp. 197–201. [Google Scholar] [CrossRef]
  106. Vacca, M.; Celano, G.; Calabrese, F.M.; Portincasa, P.; Gobbetti, M.; De Angelis, M. The controversial role of human gut lachnospiraceae. Microorganisms 2020, 8, 573. [Google Scholar] [CrossRef] [PubMed]
  107. Gradisteanu Pircalabioru, G.; Liaw, J.; Gundogdu, O.; Corcionivoschi, N.; Ilie, I.; Oprea, L.; Musat, M.; Chifiriuc, M.-C. Effects of the Lipid Profile, Type 2 Diabetes and Medication on the Metabolic Syndrome—Associated Gut Microbiome. Int. J. Mol. Sci. 2022, 23, 7509. [Google Scholar] [CrossRef]
  108. Hexun, Z.; Miyake, T.; Maekawa, T.; Mori, H.; Yasukawa, D.; Ohno, M.; Nishida, A.; Andoh, A.; Tani, M. High abundance of Lachnospiraceae in the human gut microbiome is related to high immunoscores in advanced colorectal cancer. Cancer Immunol. Immunother. 2023, 72, 315–326. [Google Scholar] [CrossRef]
  109. Liu, F.; Li, J.; Guan, Y.; Lou, Y.; Chen, H.; Xu, M.; Deng, D.; Chen, J.; Ni, B.; Zhao, L.; et al. Dysbiosis of the gut microbiome is associated with tumor biomarkers in lung cancer. Int. J. Biol. Sci. 2019, 15, 2381–2392. [Google Scholar] [CrossRef]
  110. Zhang, X.; Yu, D.; Wu, D.; Gao, X.; Shao, F.; Zhao, M.; Wang, J.; Ma, J.; Wang, W.; Qin, X.; et al. Tissue-resident Lachnospiraceae family bacteria protect against colorectal carcinogenesis by promoting tumor immune surveillance. Cell Host Microbe 2023, 31, 418–432.e8. [Google Scholar] [CrossRef]
  111. Ahrens, A.P.; Culpepper, T.; Saldivar, B.; Anton, S.; Stoll, S.; Handberg, E.M.; Xu, K.; Pepine, C.; Triplett, E.W.; Aggarwal, M. A six-day, lifestyle-based immersion program mitigates cardiovascular risk factors and induces shifts in gut microbiota, specifically lachnospiraceae, ruminococcaceae, faecalibacterium prausnitzii: A pilot study. Nutrients 2021, 13, 3459. [Google Scholar] [CrossRef]
  112. Tindall, A.M.; McLimans, C.J.; Petersen, K.S.; Kris-Etherton, P.M.; Lamendella, R. Walnuts and Vegetable Oils Containing Oleic Acid Differentially Affect the Gut Microbiota and Associations with Cardiovascular Risk Factors: Follow-up of a Randomized, Controlled, Feeding Trial in Adults at Risk for Cardiovascular Disease. J. Nutr. 2020, 150, 806–817. [Google Scholar] [CrossRef] [PubMed]
  113. Ćesić, D.; Lugović Mihić, L.; Ozretić, P.; Lojkić, I.; Buljan, M.; Šitum, M.; Zovak, M.; Vidović, D.; Mijić, A.; Galić, N.; et al. Association of Gut Lachnospiraceae and Chronic Spontaneous Urticaria. Life 2023, 13, 1280. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, M.; Cui, Y.; Liu, P.; Mo, R.; Wang, H.; Li, Y.; Wu, Y. Oat β-(1 → 3, 1 → 4)-D-glucan alleviates food allergy-induced colonic injury in mice by increasing Lachnospiraceae abundance and butyrate production. Carbohydr. Polym. 2024, 344, 122535. [Google Scholar] [CrossRef] [PubMed]
  115. Zuo, T.; Liu, Q.; Zhang, F.; Lui, G.C.-Y.; Tso, E.Y.K.; Yeoh, Y.K.; Chen, Z.; Boon, S.S.; Chan, F.K.; Chan, P.K.S.; et al. Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19. Gut 2020, 70, 276–284. [Google Scholar] [CrossRef]
  116. San-Juan-Vergara, H.; Zurek, E.; Ajami, N.J.; Mogollon, C.; Peña, M.; Portnoy, I.; Vélez, J.I.; Cadena-Cruz, C.; Diaz-Olmos, Y.; Hurtado-Gómez, L.; et al. A Lachnospiraceae-dominated bacterial signature in the fecal microbiota of HIV-infected individuals from Colombia, South America. Sci. Rep. 2018, 8, 4479. [Google Scholar] [CrossRef]
  117. Vujkovic-Cvijin, I.; Sortino, O.; Verheij, E.; Sklar, J.; Wit, F.W.; Kootstra, N.A.; Sellers, B.; Brenchley, J.M.; Ananworanich, J.; Loeff, M.S.; et al. HIV-associated gut dysbiosis is independent of sexual practice and correlates with noncommunicable diseases. Nat. Commun. 2020, 11, 2448. [Google Scholar] [CrossRef]
  118. Do Nascimento, W.M.; Machiavelli, A.; Ferreira, L.G.E.; Silveira, L.C.; de Azevedo, S.S.D.; Bello, G.; Smith, D.P.; Mezzari, M.P.; Petrosino, J.F.; Duarte, R.T.D.; et al. Gut microbiome profiles and associated metabolic pathways in HIV-infected treatment-naïve patients. Cells 2021, 10, 385. [Google Scholar] [CrossRef]
  119. Wu, X.; Li, R.F.; Lin, Z.S.; Xiao, C.; Liu, B.; Mai, K.L.; Zhou, H.X.; Zeng, D.Y.; Cheng, S.; Weng, Y.C.; et al. Coinfection with influenza virus and non-typeable Haemophilus influenzae aggregates inflammatory lung injury and alters gut microbiota in COPD mice. Front. Microbiol. 2023, 14, 1137369. [Google Scholar] [CrossRef]
  120. Konikoff, T.; Gophna, U. Oscillospira: A Central, Enigmatic Component of the Human Gut Microbiota. Trends Microbiol. 2016, 24, 523–524. [Google Scholar] [CrossRef]
  121. Murugesan, S.; Ulloa-Martínez, M.; Martínez-Rojano, H.; Galván-Rodríguez, F.M.; Miranda-Brito, C.; Romano, M.C.; Piña-Escobedo, A.; Pizano-Zárate, M.L.; Hoyo-Vadillo, C.; García-Mena, J. Study of the diversity and short-chain fatty acids production by the bacterial community in overweight and obese Mexican children. Eur. J. Clin. Microbiol. Infect. Dis. 2015, 34, 1337–1346. [Google Scholar] [CrossRef]
  122. Zhan, S.; Liu, C.; Meng, J.; Mao, R.; Tu, T.; Lin, J.; Chen, M.; Zeng, Z.; Zhuang, X. Mucosa-Associated Oscillospira sp. Is Related to Intestinal Stricture and Post-Operative Disease Course in Crohn’s Disease. Microorganisms 2023, 11, 794. [Google Scholar] [CrossRef] [PubMed]
  123. Kim, H.N.; Joo, E.J.; Cheong, H.S.; Kim, Y.; Kim, H.L.; Shin, H.; Chang, Y.; Ryu, S. Gut microbiota and risk of persistent nonalcoholic fatty liver diseases. J. Clin. Med. 2019, 8, 1089. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, S.; Dai, J.; Lan, X.; Fan, B.; Dong, T.; Zhang, Y.; Han, M. Intestinal bacteria are potential biomarkers and therapeutic targets for gastric cancer. Microb. Pathog. 2021, 151, 104747. [Google Scholar] [CrossRef] [PubMed]
  125. Wang, Z.; Usyk, M.; Sollecito, C.C.; Qiu, Y.; Williams-Nguyen, J.; Hua, S.; Gradissimo, A.; Wang, T.; Xue, X.; Kurland, I.J.; et al. Altered Gut Microbiota and Host Metabolite Profiles in Women with Human Immunodeficiency Virus. Clin. Infect. Dis. 2020, 71, 2345–2353. [Google Scholar] [CrossRef]
  126. Chandiwana, P.; Munjoma, P.T.; Mazhandu, A.J.; Li, J.; Baertschi, I.; Wyss, J.; Jordi, S.B.U.; Mazengera, L.R.; Yilmaz, B.; Misselwitz, B.; et al. Antenatal gut microbiome profiles and effect on pregnancy outcome in HIV infected and HIV uninfected women in a resource limited setting. BMC Microbiol. 2023, 23, 4. [Google Scholar] [CrossRef]
  127. Nie, K.; Ma, K.; Luo, W.; Shen, Z.; Yang, Z.; Xiao, M.; Tong, T.; Yang, Y.; Wang, X. Roseburia intestinalis: A Beneficial Gut Organism from the Discoveries in Genus and Species. Front. Cell. Infect. Microbiol. 2021, 11, 757718. [Google Scholar] [CrossRef]
  128. Tamanai-Shacoori, Z.; Smida, I.; Bousarghin, L.; Loreal, O.; Meuric, V.; Fong, S.B.; Bonnaure-Mallet, M.; Jolivet-Gougeon, A. Roseburia spp.: A marker of health? Future Microbiol. 2017, 12, 157–170. [Google Scholar] [CrossRef]
  129. Xu, F.; Cheng, Y.; Ruan, G.; Fan, L.; Tian, Y.; Xiao, Z.; Chen, D.; Wei, Y. New pathway ameliorating ulcerative colitis: Focus on Roseburia intestinalis and the gut–brain axis. Therap. Adv. Gastroenterol. 2021, 14, 175628482110044. [Google Scholar] [CrossRef]
  130. Haro, C.; Montes-Borrego, M.; Rangel-Zúñiga, O.A.; Alcalã-Diaz, J.F.; Gamez-Delgado, F.; Pérez-Martinez, P.; Delgado-Lista, J.; Quintana-Navarro, G.M.; Tinahones, F.J.; Landa, B.B.; et al. Two healthy diets modulate gut microbial community improving insulin sensitivity in a human obese population. J. Clin. Endocrinol. Metab. 2016, 101, 233–242. [Google Scholar] [CrossRef]
  131. Huang, W.; Zhu, W.; Lin, Y.; Chan, F.K.; Xu, Z.; Ng, S.C. Roseburia hominis improves host metabolism in obesity by production of nicotinamide riboside. In Basic Gastroenterology; BMJ Publishing Group Ltd. and British Society of Gastroenterology: London, UK, 2024; pp. A204.3–A205. [Google Scholar] [CrossRef]
  132. Kasahara, K.; Krautkramer, K.A.; Org, E.; Romano, K.A.; Kerby, R.L.; Vivas, E.I.; Mehrabian, M.; Denu, J.M.; Bäckhed, F.; Lusis, A.J.; et al. Interactions between Roseburia intestinalis and diet modulate atherogenesis in a murine model. Nat. Microbiol. 2018, 3, 1461–1471. [Google Scholar] [CrossRef]
  133. Liu, C.; Sun, Z.; Shali, S.; Mei, Z.; Chang, S.; Mo, H.; Xu, L.; Pu, Y.; Guan, H.; Chen, G.C.; et al. The gut microbiome and microbial metabolites in acute myocardial infarction. J. Genet. Genom. 2022, 49, 569–578. [Google Scholar] [CrossRef]
  134. Li, J.; Ma, Y.; Bao, Z.; Gui, X.; Li, A.N.; Yang, Z.; Li, M.D. Clostridiales are predominant microbes that mediate psychiatric disorders. J. Psychiatr. Res. 2020, 130, 48–56. [Google Scholar] [CrossRef] [PubMed]
  135. Tao, W.; Zhang, G.; Wang, X.; Guo, M.; Zeng, W.; Xu, Z.; Cao, D.; Pan, A.; Wang, Y.; Zhang, K.; et al. Analysis of the intestinal microbiota in COVID-19 patients and its correlation with the inflammatory factor IL-18. Med. Microecol. 2020, 5, 100023. [Google Scholar] [CrossRef] [PubMed]
  136. Fan, R.; Liu, S.; Sun, N.; Yang, Y.; Deng, X.; Hu, B.; Sun, C.; Wen, C.; Li, H.; Cheng, D.; et al. Gut microbiota composition is associated with disease severity and host immune responses in COVID-19. Front. Cell. Infect. Microbiol. 2023, 13, 1274690. [Google Scholar] [CrossRef]
  137. Qing, Y.; Xie, H.; Su, C.; Wang, Y.; Yu, Q.; Pang, Q.; Cui, F. Gut Microbiome, Short-Chain Fatty Acids, and Mucosa Injury in Young Adults with Human Immunodeficiency Virus Infection. Dig. Dis. Sci. 2019, 64, 1830–1843. [Google Scholar] [CrossRef]
  138. Crost, E.H.; Coletto, E.; Bell, A.; Juge, N. Ruminococcus gnavus: Friend or foe for human health. FEMS Microbiol. Rev. 2023, 47, fuad014. [Google Scholar] [CrossRef] [PubMed]
  139. Kandasamy, S.; Letchumanan, V.; Hong, K.W.; Chua, K.O.; Mutalib, N.S.A.; Ng, A.L.O.; Ming, L.C.; Lim, H.X.; Thurairajasingam, S.; Law, J.W.F.; et al. The Role of Human Gut Microbe Ruminococcus gnavus in Inflammatory Diseases. Prog. Microbes Mol. Biol. 2023, 6, 1. [Google Scholar] [CrossRef]
  140. Takahashi, K.; Nishida, A.; Fujimoto, T.; Fujii, M.; Shioya, M.; Imaeda, H.; Inatomi, O.; Bamba, S.; Andoh, A.; Sugimoto, M. Reduced Abundance of Butyrate-Producing Bacteria Species in the Fecal Microbial Community in Crohn’s Disease. Digestion 2016, 93, 59–65. [Google Scholar] [CrossRef]
  141. Hall, A.B.; Yassour, M.; Sauk, J.; Garner, A.; Jiang, X.; Arthur, T.; Lagoudas, G.K.; Vatanen, T.; Fornelos, N.; Wilson, R.; et al. A novel Ruminococcus gnavus clade enriched in inflammatory bowel disease patients. Genome Med. 2017, 9, 103. [Google Scholar] [CrossRef]
  142. Maes, M.; Vasupanrajit, A.; Jirakran, K.; Klomkliew, P.; Chanchaem, P.; Tunvirachaisakul, C.; Payungporn, S. Exploration of the Gut Microbiome in Thai Patients with Major Depressive Disorder Shows a Specific Bacterial Profile with Depletion of the Ruminococcus Genus as a Putative Biomarker. Cells 2023, 12, 1240. [Google Scholar] [CrossRef]
  143. Lakshmanan, A.P.; Al Zaidan, S.; Bangarusamy, D.K.; Al-Shamari, S.; Elhag, W.; Terranegra, A. Increased Relative Abundance of Ruminoccocus Is Associated with Reduced Cardiovascular Risk in an Obese Population. Front. Nutr. 2022, 9, 849005. [Google Scholar] [CrossRef] [PubMed]
  144. Wang, Y.; Pan, C.Q.; Xing, H. Advances in Gut Microbiota of Viral Hepatitis Cirrhosis. Biomed Res. Int. 2019, 2019, 9726786. [Google Scholar] [CrossRef] [PubMed]
  145. Gozalbo-Rovira, R.; Rubio-Del-campo, A.; Santiso-Bellón, C.; Vila-Vicent, S.; Buesa, J.; Delgado, S.; Molinero, N.; Margolles, A.; Yebra, M.J.; Collado, M.C.; et al. Interaction of intestinal bacteria with human rotavirus during infection in children. Int. J. Mol. Sci. 2021, 22, 1010. [Google Scholar] [CrossRef] [PubMed]
  146. Rodríguez-Díaz, J.; García-Mantrana, I.; Vila-Vicent, S.; Gozalbo-Rovira, R.; Buesa, J.; Monedero, V.; Collado, M.C. Relevance of secretor status genotype and microbiota composition in susceptibility to rotavirus and norovirus infections in humans. Sci. Rep. 2017, 7, 45559. [Google Scholar] [CrossRef]
  147. Depommier, C.; Everard, A.; Druart, C.; Plovier, H.; Van Hul, M.; Vieira-Silva, S.; Falony, G.; Raes, J.; Maiter, D.; Delzenne, N.M. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nat. Med. 2019, 25, 1096–1103. [Google Scholar] [CrossRef]
  148. Hu, L.; Sun, L.; Yang, C.; Zhang, D.W.; Wei, Y.Y.; Yang, M.M.; Wu, H.M.; Fei, G.H. Gut microbiota-derived acetate attenuates lung injury induced by influenza infection via protecting airway tight junctions. J. Transl. Med. 2024, 22, 570. [Google Scholar] [CrossRef]
  149. Ottman, N.; Reunanen, J.; Meijerink, M.; Pietila, T.E.; Kainulainen, V.; Klievink, J.; Huuskonen, L.; Aalvink, S.; Skurnik, M.; Boeren, S.; et al. Pili-like proteins of Akkermansia muciniphila modulate host immune responses and gut barrier function. PLoS ONE 2017, 12, e0173004. [Google Scholar] [CrossRef]
  150. Xie, S.; Li, J.; Lyu, F.; Xiong, Q.; Gu, P.; Chen Yuqi Chen, M.; Bao, J.; Zhang, X.; Wei, R.; Deng Youpeng Wang, H.; et al. Novel tripeptide RKH derived from Akkermansia muciniphila protects against lethal sepsis. Gut 2024, 73, 78–91. [Google Scholar] [CrossRef]
  151. Koçak Tufan, Z.; Kayaaslan, B.; Mer, M. COVID-19 and Sepsis. Turkish J. Med. Sci. 2021, 51, 3301–3311. [Google Scholar] [CrossRef]
  152. Wu, Z.; Xu, Q.; Gu, S.; Chen, Y.; Lv, L.; Zheng, B.; Wang, Q.; Wang, K.; Wang, S.; Xia, J.; et al. Akkermansia muciniphila Ameliorates Clostridioides difficile Infection in Mice by Modulating the Intestinal Microbiome and Metabolites. Front. Microbiol. 2022, 13, 841920. [Google Scholar] [CrossRef]
  153. Nasiri, G.; Azimirad, M.; Goudarzi, H.; Amirkamali, S.; Yadegar, A.; Ghalavand, Z.; Shahrokh, S.; Asadzadeh Aghdaei, H.; Zali, M.R. The inhibitory effects of live and UV-killed Akkermansia muciniphila and its derivatives on cytotoxicity and inflammatory response induced by Clostridioides difficile RT001 in vitro. Int. Microbiol. 2024, 27, 393–409. [Google Scholar] [CrossRef] [PubMed]
  154. Song, L.; Huang, Y.; Liu, G.; Li, X.; Xiao, Y.; Liu, C.; Zhang, Y.; Li, J.; Xu, J.; Lu, S.; et al. A Novel Immunobiotics Bacteroides dorei Ameliorates Influenza Virus Infection in Mice. Front. Immunol. 2022, 12, 828887. [Google Scholar] [CrossRef] [PubMed]
  155. Hansen, R.; Sanderson, I.R.; Muhammed, R.; Allen, S.; Tzivinikos, C.; Henderson, P.; Gervais, L.; Jeffery, I.B.; Mullins, D.P.; O’Herlihy, E.A.; et al. A Double-Blind, Placebo-Controlled Trial to Assess Safety and Tolerability of (Thetanix) Bacteroides thetaiotaomicron in Adolescent Crohn’s Disease. Clin. Transl. Gastroenterol. 2021, 12, e00287. [Google Scholar] [CrossRef] [PubMed]
  156. Porter, N.T.; Luis, A.S.; Martens, E.C. Bacteroides thetaiotaomicron. Trends Microbiol. 2018, 26, 966–967. [Google Scholar] [CrossRef]
  157. Quigley, E.M.M.; Markinson, L.; Stevenson, A.; Treasure, F.P.; Lacy, B.E. Randomised clinical trial: Efficacy and safety of the live biotherapeutic product MRx1234 in patients with irritable bowel syndrome. Aliment. Pharmacol. Ther. 2023, 57, 81–93. [Google Scholar] [CrossRef]
  158. Yang, R.; Shan, S.; Shi, J.; Li, H.; An, N.; Li, S.; Cui, K.; Guo, H.; Li, Z. Coprococcus eutactus, a Potent Probiotic, Alleviates Colitis via Acetate-Mediated IgA Response and Microbiota Restoration. J. Agric. Food Chem. 2022, 71, 3273–3284. [Google Scholar] [CrossRef]
  159. Valles-Colomer, M.; Falony, G.; Darzi, Y.; Tigchelaar, E.F.; Wang, J.; Tito, R.Y.; Schiweck, C.; Kurilshikov, A.; Joossens, M.; Wijmenga, C.; et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat. Microbiol. 2019, 4, 623–632. [Google Scholar] [CrossRef]
  160. Liu, Y.; Zhang, L.; Wang, X.; Wang, Z.; Zhang, J.; Jiang, R.; Wang, X.; Wang, K.; Liu, Z.; Xia, Z.; et al. Similar Fecal Microbiota Signatures in Patients with Diarrhea-Predominant Irritable Bowel Syndrome and Patients with Depression. Clin. Gastroenterol. Hepatol. 2016, 14, 1602–1611.e5. [Google Scholar] [CrossRef]
  161. Notting, F.; Pirovano, W.; Sybesma, W.; Kort, R. The butyrate-producing and spore-forming bacterial genus Coprococcus as a potential biomarker for neurological disorders. Gut Microbiome 2023, 4, e16. [Google Scholar] [CrossRef]
  162. Martín, R.; Miquel, S.; Langella, P.; Bermúdez-Humarán, L.G. The role of metagenomics in understanding the human microbiome in health and disease. Virulence 2014, 5, 413–423. [Google Scholar] [CrossRef]
  163. Chollet, L.; Heumel, S.; Deruyter, L.; Bouilloux, F.; Delval, L.; Robert, V.; Gevaert, M.H.; Pichavant, M.; Sencio, V.; Robil, C.; et al. Faecalibacterium duncaniae as a novel next generation probiotic against influenza. Front. Immunol. 2024, 15, 1347676. [Google Scholar] [CrossRef] [PubMed]
  164. Li, H.B.; Xu, M.L.; Xu, X.D.; Tang, Y.Y.; Jiang, H.L.; Li, L.; Xia, W.J.; Cui, N.; Bai, J.; Dai, Z.M.; et al. Faecalibacterium prausnitzii Attenuates CKD via Butyrate-Renal GPR43 Axis. Circ. Res. 2022, 131, E120–E134. [Google Scholar] [CrossRef]
  165. Gevers, D.; Kugathasan, S.; Denson, L.A.; Vázquez-Baeza, Y.; Van Treuren, W.; Ren, B.; Schwager, E.; Knights, D.; Song, S.J.; Yassour, M.; et al. The Treatment-Naive Microbiome in New-Onset Crohn’s Disease. Cell Host Microbe 2014, 15, 382–392. [Google Scholar] [CrossRef] [PubMed]
  166. Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D.; et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [Google Scholar] [CrossRef] [PubMed]
  167. De Filippis, F.; Vitaglione, P.; Cuomo, R.; Berni Canani, R.; Ercolini, D. Dietary Interventions to Modulate the Gut Microbiome-How Far Away Are We from Precision Medicine. Inflamm Bowel Dis. 2018, 24, 2142–2154. [Google Scholar] [CrossRef] [PubMed]
  168. Reeves, A.E.; Koenigsknecht, M.J.; Bergin, I.L.; Young, V.B. Suppression of Clostridium difficile in the gastrointestinal tracts of germfree mice inoculated with a murine isolate from the family Lachnospiraceae. Infect. Immun. 2012, 80, 3786–3794. [Google Scholar] [CrossRef]
  169. Gophna, U.; Konikoff, T.; Nielsen, H.B. Oscillospira and related bacteria—From metagenomic species to metabolic features. Environ. Microbiol. 2017, 19, 835–841. [Google Scholar] [CrossRef]
  170. Louis, P.; Hold, G.L.; Flint, H.J. The gut microbiota, bacterial metabolites and colorectal cancer. Nat. Rev. Microbiol. 2014, 12, 661–672. [Google Scholar] [CrossRef]
  171. Yang, J.; Li, Y.; Wen, Z.; Liu, W.; Meng, L.; Huang, H. Oscillospira—A candidate for the next-generation probiotics. Gut Microbes 2021, 13, 1987783. [Google Scholar] [CrossRef]
  172. Makhoul, E.; Aklinski, J.L.; Miller, J.; Leonard, C.; Backer, S.; Kahar, P.; Parmar, M.S.; Khanna, D. A Review of COVID-19 in Relation to Metabolic Syndrome: Obesity, Hypertension, Diabetes, and Dyslipidemia. Cureus 2022, 14, e27438. [Google Scholar] [CrossRef]
  173. Zhang, J.; Song, L.; Wang, Y.; Liu, C.; Zhang, L.; Zhu, S.; Liu, S.; Duan, L. Beneficial effect of butyrate-producing Lachnospiraceae on stress-induced visceral hypersensitivity in rats. J. Gastroenterol. Hepatol. 2019, 34, 1368–1376. [Google Scholar] [CrossRef]
  174. Sun, X.; Zhou, C.; Ju, M.; Feng, W.; Guo, Z.; Qi, C.; Yang, K.; Xiao, R. Roseburia intestinalis Supplementation Could Reverse the Learning and Memory Impairment and m6A Methylation Modification Decrease Caused by 27-Hydroxycholesterol in Mice. Nutrients 2024, 16, 1288. [Google Scholar] [CrossRef] [PubMed]
  175. Ahmed, M.; Roy, S.; Iktidar, M.A.; Chowdhury, S.; Akter, S.; Islam, A.M.K.; Hawlader, M.D.H. Post-COVID-19 memory complaints: Prevalence and associated factors. Neurologia 2022, 39, 651–657. [Google Scholar] [CrossRef] [PubMed]
  176. Makarova, Y.A.; Ryabkova, V.A.; Salukhov, V.V.; Sagun, B.V.; Korovin, A.E.; Churilov, L.P. Atherosclerosis, Cardiovascular Disorders and COVID-19: Comorbid Pathogenesis. Diagnostics 2023, 13, 478. [Google Scholar] [CrossRef] [PubMed]
  177. Vilaplana-Carnerero, C.; Giner-Soriano, M.; Dominguez, À.; Morros, R.; Pericas, C.; Álamo-Junquera, D.; Toledo, D.; Gallego, C.; Redondo, A.; Grau, M. Atherosclerosis, Cardiovascular Disease, and COVID-19: A Narrative Review. Biomedicines 2023, 11, 1206. [Google Scholar] [CrossRef]
  178. Flint, H.J.; Scott, K.P.; Duncan, S.H.; Louis, P.; Forano, E. Microbial degradation of complex carbohydrates in the gut. Gut Microbes 2012, 3, 289–306. [Google Scholar] [CrossRef]
  179. Ze, X.; Duncan, S.H.; Louis, P.; Flint, H.J. Ruminococcus bromii is a keystone species for the degradation of resistant starch in the human colon. ISME J. 2012, 6, 1535–1543. [Google Scholar] [CrossRef]
  180. Jung, D.H.; Park, C.S. Resistant starch utilization by Bifidobacterium, the beneficial human gut bacteria. Food Sci. Biotechnol. 2023, 32, 441–452. [Google Scholar] [CrossRef]
  181. Zhu, C.; Song, K.; Shen, Z.; Quan, Y.; Tan, B.; Luo, W.; Wu, S.; Tang, K.; Yang, Z.; Wang, X. Roseburia intestinalis inhibits interleukin-17 excretion and promotes regulatory T cells differentiation in colitis. Mol. Med. Rep. 2018, 17, 7567–7574. [Google Scholar] [CrossRef]
  182. Wang, M.; Lkhagva, E.; Kim, S.; Zhai, C.; Islam, M.M.; Kim, H.J.; Hong, S.T. The gut microbe pair of Oribacterium sp. GMB0313 and Ruminococcus sp. GMB0270 confers complete protection against SARS-CoV-2 infection by activating CD8+ T cell-mediated immunity. Gut Microbes 2024, 16, 2342497. [Google Scholar] [CrossRef]
Table 1. Comparison of probiotic genera discussed in this study for adjunctive treatment of COVID-19.
Table 1. Comparison of probiotic genera discussed in this study for adjunctive treatment of COVID-19.
GenusMechanisms of ActionHealth BenefitsClinical EvidenceOngoing Research/Trials
AkkermansiaModulates immune response, enhances gut barrier integrity, produces anti-inflammatory cytokinesImproves insulin sensitivity, reduces liver dysfunction, protects against viral infectionsReduces IL-6 and TNF-α levels in H7N9 virus models; protective in sepsis modelsTrials on irritable bowel syndrome and mental health conditions (NCT05348642)
BacteroidesAlters gut microbiota, digests dietary fibers, produces SCFAsIncreases survival in Influenza A models, improves gut healthIncreases type 1 interferon expression; reduces inflammatory cytokine levels in mouse modelsDevelopment as a biotherapeutic for Crohn’s disease (NCT02704728)
BlautiaPromotes anti-inflammatory cytokine production, modulates gut healthProtects against secondary bacterial infections, improves bowel habitsReduces bacterial load in Influenza A models; improves survivalEfficacy of Blautix for irritable bowel syndrome [157]
CoprococcusReduces pro-inflammatory cytokine levels, restores epithelial barrier functionImproves mental health outcomes, influences gut-brain axisLinked to reduced inflammation in colitis models; associated with mental healthFuture trials to evaluate neurological benefits in COVID-19 patients
FaecalibacteriumImproves gut barrier function, produces butyrateReduces intestinal inflammation, protects against infectionsProvides protection against influenza; improves gut health in chronic kidney disease modelsTrials on maintaining clinical response in Crohn’s disease (NCT02539849)
LachnospiraProduces SCFAs, reduces inflammationImproves gut health, may help manage hyperinflammatory responsesReduces Clostridioides difficile colonization in mouse modelsPotential therapeutic targets for COVID-19 patients with comorbidities
OscillospiraProduces SCFAs, associated with metabolic healthAlleviates hyperinflammatory responses, supports overall gut healthLinked to metabolic health; potential impact on obesity-related COVID-19 risksTargeting Oscillospira for therapeutic approaches in viral infections
RoseburiaProduces butyrate, enhances gut barrier integrityPrevents pathogen translocation, improves memory functionImproves colon barrier function and reduces inflammatory cytokines in murine modelsInvestigating role in cardiovascular health and COVID-19 recovery
RuminococcusDegrades polysaccharides, produces SCFAsModulates immune response, improves metabolic healthProtects against SARS-CoV-2 in hamster models; reduces inflammation in colitis modelsEvaluating metabolic effects in overweight individuals (NCT05448274)
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

Rampelotto, P.H.; Taufer, C.R.; da Silva, J. The Role of Beneficial Microbiota in COVID-19: Insights from Key Bacterial Genera. Microorganisms 2025, 13, 1029. https://doi.org/10.3390/microorganisms13051029

AMA Style

Rampelotto PH, Taufer CR, da Silva J. The Role of Beneficial Microbiota in COVID-19: Insights from Key Bacterial Genera. Microorganisms. 2025; 13(5):1029. https://doi.org/10.3390/microorganisms13051029

Chicago/Turabian Style

Rampelotto, Pabulo Henrique, Clarissa Reginato Taufer, and Juliana da Silva. 2025. "The Role of Beneficial Microbiota in COVID-19: Insights from Key Bacterial Genera" Microorganisms 13, no. 5: 1029. https://doi.org/10.3390/microorganisms13051029

APA Style

Rampelotto, P. H., Taufer, C. R., & da Silva, J. (2025). The Role of Beneficial Microbiota in COVID-19: Insights from Key Bacterial Genera. Microorganisms, 13(5), 1029. https://doi.org/10.3390/microorganisms13051029

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