Copyright
©The Author(s) 2026.
World J Gastroenterol. Jan 21, 2026; 32(3): 112437
Published online Jan 21, 2026. doi: 10.3748/wjg.v32.i3.112437
Published online Jan 21, 2026. doi: 10.3748/wjg.v32.i3.112437
Table 1 Human viruses interact with bacteria either directly (via bacterial products that facilitate infection) or indirectly (through host environment changes that favor bacterial colonization)
| Viruses | Bacterial partner(s) | Mechanistic basis | Ref. |
| Direct interactions | |||
| Murine norovirus | E. cloacae, enteric microbiota | Bacterial histo-blood group antigen analogs support persistent viral infection in the gut | [8,9] |
| Influenza virus | Staphylococcus aureus | Bacterial proteases activate viral hemagglutinin through cleavage | [10] |
| Rotavirus | Gut microbiota (e.g., Escherichia coli, Bacteroides thetaiotaomicron) | Bacterial factors promote viral infectivity while reducing antibody neutralization | [11] |
| Reovirus T3SA+ | Enteric bacteria (Escherichia coli, etc.) | Bacterial lipopolysaccharide mediates enhanced viral binding and replication efficiency | [12] |
| HIV | Mycobacterium tuberculosis | Mycobacterial components enhance HIV transcriptional activity | [13] |
| Indirect interactions | |||
| Adenovirus | S. pneumoniae | Enhances bacterial attachment to respiratory epithelial cells | [14] |
| Rhinovirus | Respiratory pathogens | Increases the expression of host cell adhesion molecules for bacterial binding | [15] |
| Measles virus | Multiple opportunistic pathogens | Systemic immune suppression facilitates secondary bacterial infections | [16,17] |
| Herpesviruses | Porphyromonas gingivalis, Bacteroides forsythus (now Tannerella forsythia), Campylobacter rectus | Viral immunosuppression enables bacterial colonization of the oral mucosa | [18] |
| Parainfluenza virus | Streptococcus pneumoniae, Haemophilus influenzae | Viral infection increases bacterial adhesion to the respiratory epithelium | [19,20] |
Table 2 Dysbiosis of gut microbiota in individuals with human immunodeficiency virus infection
| Ref. | Total cases (n) | HIV-related cases (n) | Methodology | Increased taxa | Decreased taxa | Limitations | Conclusion |
| Rhoades et al[44] | 105 (≥ 55 years) | 58 LTC-HIV+ | 16S rRNA sequencing (V4) | Fusobacterium, Lactobacillus, Bifidobacteriales, Prevotella (in low CD4+) | Clostridia (SCFA producers), Oxalobacter, Streptococcus (vs HIV-) | Cross-sectional design; cannot establish causality; majority Caucasian male cohort limits generalizability | LTC-HIV+ individuals show gut dysbiosis - oral taxa enriched, butyrate producers depleted - with Prevotella inversely correlating with CD4+ counts, linking microbial shifts to immune dysfunction and chronic inflammation |
| Lu et al[45] | 91 | 61 HIV | Metagenomics sequencing | Faecalibacterium prausnitzii, Subdoligranulum sp., Coprococcus comes, Prevotella copri, Prevotella stercorea, Bacteroides coprophilus, Bacteroides coprocola, Bacteroides intestinalis, Bacteroides salyersiae | Bacteroides | Small sample size; cross-sectional; single population (Chinese); ART regimen effects not fully differentiated | HIV infection causes gut dysbiosis; increased butyrate producers (F. prausnitzii, Subdoligranulum sp., C. comes) link to poor CD4+ T-cell recovery and activation; microbiota modulation may aid immune reconstitution |
| Cook et al[46] | 383 | HIV- (200) HIV+ undetectable (< 20 c/mL, n = 66) HIV+ suppressed (20-200 c/mL, n = 72) HIV+ viremic (> 200 c/mL, n = 45) | 16S rRNA sequencing (V4); IPTW-adjusted models | Viremic group: Peptoniphilus, Porphyromonas, Prevotella, Murdochiella | Viremic group: Bacteroides, Brachyspira, Faecalibacterium, Helicobacter. All HIV+ groups: Bacteroides | Lack of dietary data; residual confounding possible; 16S limits species-level resolution; no data on time since ART initiation | HIV viremia correlates with rectal dysbiosis in a dose-dependent manner; viremic individuals show a pro-inflammatory microbiota (Prevotella, Porphyromonas), while suppressed/undetectable viremia associates with milder dysbiosis |
| Villar-García et al[47] | 44 (HIV+, on HAART) | 22 immunologic responders, 22 immunologic non-responders | 16S rDNA gene sequencing | Megamonas, Desulfovibrionales (Proteobacteria) | Clostridiales (esp. Clostridiaceae, Catenibacterium), Lachnospiraceae, Proteobacteria | Small sample size; did not analyze fungal mycobiome (including probiotic colonization); cannot establish causality; short intervention period | Saccharomyces boulardii reduced pro-inflammatory Clostridiaceae; immunologic non-responders had higher baseline Lachnospiraceae and Proteobacteria |
| Dillon et al[48] | 32 | 18 untreated HIV+ | 16S rRNA sequencing (colonic mucosa and stool) | Prevotella stercorea | Butyrate-producing bacteria: Roseburia intestinalis, Faecalibacterium prausnitzii, Eubacterium rectale (in colonic mucosa) | Small cross-sectional study; sexual preference unmatched; fecal butyrate not measured | HIV reduces colonic butyrate-producing bacteria (e.g., Roseburia intestinalis); low abundance links to microbial translocation and immune activation; butyrate may protect against T cell activation and HIV replication |
| Dinh et al[49] | 21 HIV+ on ART, 16 HIV- controls | 21 (chronic HIV, suppressed VL) | 16S rRNA pyrosequencing (V3-V5 region) | Proteobacteria, Gammaproteobacteria, Enterobacteriales, Enterobacteriaceae, Erysipelotrichi, Erysipelotrichales, Erysipelotrichaceae, Barnesiella | Rikenellaceae, Alistipes | Small sample size; exploratory design; no correction for multiple comparisons; immune activation markers not assessed | ART-treated, virologically suppressed HIV+ individuals show gut dysbiosis with increased Enterobacteriaceae and decreased Alistipes, correlating with elevated sCD14 and inflammatory cytokines (IL-1β, TNF-α) |
| Lee et al[50] | HIV+ on ART: 26 (16 oIR, 10 sIR) HIV- controls: 20 | 26 (all on suppressive ART) | 16S rRNA sequencing (V4 region, rectal swabs) | Fusobacterium (phylum: Fusobacteria), Gallicola, Bilophila | Lactobacillales, Corynebacterium | Small sample size; all male participants; dietary data not collected; rectal swabs may not fully reflect luminal microbiota | Poor CD4+ recovery on ART associates with increased Fusobacterium, elevated T-cell activation/Tregs, and reduced naïve CD4+ cells |
| Machiavelli et al[51] | 38 (19 mother-child pairs) | 12 HEU children; 12 HIV+ mothers | 16S rRNA sequencing (V3-V4 region) | HEU children: Faecalibacterium prausnitzii, Prevotella copri, Lachnospiraceae, Klebsiella, Lactococcus, Ruminococcaceae, Enterobacteriaceae | HEU children: Bacteroides uniformis, Paraprevotella | Cross-sectional, small sample; breastfeeding differences between HEU and unexposed children; functional predictions inferred, not measured | HEU children of HIV+ mothers show altered gut taxa and predicted metagenome without changes in diversity, growth, or inflammation markers |
| Ling et al[52] | 83 (Chinese population) | 67 HIV+ (35 HAART-naïve, 32 HAART-treated) | 16S rRNA pyrosequencing (V1-V3 regions) | Firmicutes, Prevotella, Faecalibacterium, Phascolarctobacterium, Butyricicoccus, Erysipelotrichaceae incertae sedis, Catenibacterium, Dorea, Enterobacter, Enterococcus, Megamonas | Bacteroidetes, Bacteroides, Dialister, Clostridium XIVa, Clostridium XIVb, Barnesiella, Coprococcus | Cross-sectional; all male; sexual behavior not controlled; fecal samples may not reflect mucosa-associated microbiota | HIV-1 infection causes gut dysbiosis (↑ Firmicutes/Bacteroidetes, pro-inflammatory genera); key taxa correlate with inflammation; short-term HAART only partially restores the microbiota |
| Liu et al[53] | 36 (35 male, 1 female) | 14 HIV+ (all ART-treated) | 16S rRNA sequencing (V3-V4 region) | Fusobacteria, Proteobacteria, Leptotrichiaceae, Desulfovibrionaceae, Enterobacter, Paraprevotella, Allisonella, Anaerovibrio, Howardella | Firmicutes, Lachnospiraceae, Oxalobacteraceae, Eggerthella, Barnesiella, Odoribacter, Oscillospira, Oxalobacter, Roseburia, Alistipes | Small, mostly male cohort; observational; prebiotic/probiotic use uncontrolled; genus-level data limits species resolution | HIV and aging interact to alter the stool microbiome; age-related taxa changes and associations with SCFAs, diet, and inflammation differ by HIV status |
| Yang et al[54] | 16 | 8 HIV+ (HAART-naïve, low CD4 counts in 6/8) | 16S rRNA sequencing (V3-V4) | Proteobacteria, Burkholderia (specifically Burkholderia fungorum), Bradyrhizobium (specifically Bradyrhizobium pachyrhizi), Ralstonia, Fusobacterium | Firmicutes, Lactobacillus | Small sample; case-control design limits causation; no HAART-treated comparison; clinical symptom correlations limited | HIV-induced immunosuppression reduces gut colonization resistance, enabling environmental bacteria (B. fungorum, B. pachyrhizi) invasion and loss of beneficial commensals, causing dysbiosis linked to immune dysfunction |
Table 3 Gut microbiota composition in hepatitis B virus/hepatitis C virus cirrhotic patients
| Ref. | Total cases | Cirrhosis-related cases (n) | Methodology | Increased taxa | Decreased taxa | Limitations | Conclusion |
| Elsherbiny et al[62], 2025 | 80 (60 CHC patients + 20 HCs) | Non-cirrhotic CHC (n = 60) | 16S rRNA sequencing (V3-V4) | SVR group: Elusimicrobium, Christensenellaceae R-7 group, Catenibacterium, Oceanobacillus, Candidatus Melainabacteria. Relapsed group: Prevotella, Bifidobacterium, Lactobacillus, Megasphaera, Mitsuokella. Non-treated group: Faecalibacterium, Asteroeplasma, Eubacterium coprostanoligenes, Lachnospiraceae, Akkermansia, Muribaculaceae | SVR group: Actinobacteria. All CHC vs HCs: Reduced diversity; Bacteroides, Agathobacter, Parabacteroides | Single-center design; modest sample size; unmeasured confounders (diet, lifestyle) | DAA therapy markedly modulates gut microbiota; SVR restores microbial diversity and composition toward that of HCs, whereas relapse is characterized by persistent dysbiosis |
| Huang et al[63], 2023 | 120 subjects (180 samples) | Control: 60 HCs. CHC (pre-DAA): 60 patients. SVR24 (post-DAA): 60 patients | 16S rRNA sequencing (V3-V4) | Ruminococcaceae, Eubacterium, Agathobacter, Alistipes, Bifidobacterium, Klebsiella, Lactobacillus, Actinobacteria, Firmicutes, Lactobacillus | Bacteroidetes, Lachnoclostridium | Relatively small sample size; short follow-up (24 weeks); unmeasured confounders (diet, smoking); baseline differences in liver function between CHC and control groups | Gut microbiota diversity and composition remained unchanged 6 months after DAA therapy; minor differences in CHC vs controls were unaffected by SVR |
| Honda et al[64], 2025 | 70 CHB patients + 8 HCs | Functional cure: 18 (HBsAg-, HBV | 16S rRNA sequencing (V3-V4) | Clostridium bartlettii, Butyricimonas, Coprococcus catus, Bifidobacterium breve, Campylobacter | Not reported | Small sample size; exploratory design; in vitro SCFA concentrations may not reflect physiological levels in the liver | Butyrate-producing bacteria are enriched in HBsAg-negative patients; sodium butyrate directly suppresses HBsAg production in infected hepatocytes, especially post-infection |
| Inoue et al[65], 2025 | 272 (174 active HCV, 75 post-SVR, 23 HCs) | CH-HCV: 95, LC/HCC-HCV: 79, CH-SVR: 29, LC/HCC-SVR: 46, healthy: 23 | 16S rRNA sequencing; fecal BA profiling; RNA-seq | Blautia, Fusicatenibacter, Roseburia, Faecalibacterium, Subdoligranulum, Collinsella | Streptococcus, Streptococcus salivarius, Eubacterium hallii group, Ruminococcus torques group | Cohort separation for multi-omic analyses; limited SVR48 sample size; partially uses database-derived RNA-seq data | HCV eradication partially restores gut dysbiosis and BA profiles, with post-SVR Blautia enrichment correlating with improved liver fibrosis and function |
| Li et al[66], 2025 | 88 | AHE-elderly: 58, HCs-elderly: 30, self-healing: 46, non-self-healing: 12 | 16S rRNA sequencing | AHE-elderly vs HC: Firmicutes, Lactobacillales, Bacilli, Streptococcaceae. Non-self-healing vs self-healing: Bifidobacteriaceae, Bacteroidia, Bacteroides fragilis | AHE-elderly vs HC: Proteobacteria, Bacteroidetes. Self-healing vs non-self-healing: Firmicutes, Bacillus, Lactobacillus, Streptococcus | No significant difference in alpha diversity; lack of longitudinal data; unclear causal relationship between microbiota changes and HEV infection | Bacteroidetes distinguish AHE patients from controls, with Bacteroides fragilis enriched in non-self-healing cases and serving as a predictive biomarker |
| Li et al[67], 2025 | 79 | HBC: 46, HCs: 33, BA-N: 24, BA-H: 22 | 16S rRNA sequencing (V4-V5) | Streptococcus, Veillonella, Lactobacillales | Bacteroides, Akkermansia, Clostridiales | Cross-sectional design; small sample size; no BA composition or metabolomic analysis; potential confounders (diet, medication) not fully addressed | HBC dysbiosis features reduced beneficial taxa and increased opportunistic pathogens; Akkermansiaceae decline and Lactobacillales rise with elevated BAs, which correlate with higher Child-Pugh scores, suggesting gut microbiota–BA crosstalk drives HBC progression |
| Shi et al[68], 2025 | 123 | HBV-LC: 83, HC: 40, MELD < 21: 68, MELD ≥ 21: 15, CTP-C: 22 | 16S rRNA sequencing (V3-V4) | Klebsiella, Streptococcus, Fusobacterium, Enterococcus | Alistipes, Lachnospira, Agathobacter, Parabacteroides, Roseburia | Single-center design; modest sample size; cross-sectional design limits causal inference; 16S rRNA does not capture full functional potential | HBV-LC patients show gut dysbiosis marked by enrichment of pathobionts (Klebsiella, Streptococcus) and loss of SCFA-producers (Alistipes, Lachnospira), with metabolite alterations (tocopherols, 21-hydroxypregnenolone) linked to specific microbial shifts and disease severity |
| Honda et al[69], 2021 | 42 | Pre-DAA: 14, EOT: 14, post-24: 14 (samples from the same 14 patients) | 16S rRNA sequencing (V3-V4) | Faecalibacterium, Bacillus | Bacteroides, Fusobacterium | Small sample size; lack of a HC group; intra-personal comparison only; cannot determine if pre-treatment microbiota differed from healthy individuals | HCV eradication did not significantly alter overall gut microbiota diversity but increased beneficial taxa such as Faecalibacterium and Bacillus at 24 weeks post-treatment, indicating a positive compositional shift despite stable global diversity |
| Liu et al[70], 2024 | 62 | OBI: 24, HBV carriers: 18, HCs: 20 | 16S rRNA sequencing (V3-V4) | Subdoligranulum, Megamonas | Faecalibacterium | Small sample size; all participants were male; potential unmeasured confounders (diet); cannot establish causality | OBI is characterized by enrichment of Subdoligranulum, potentially driving IFN-γ/IL-17A–mediated immune activation that suppresses HBV replication, alongside depletion of beneficial Faecalibacterium |
| Yan et al[71], 2023 | 90 | 30 HCs + 30 HBV-LC + 30 HBV-HCC | 16S rRNA sequencing (V3-V4) + flow cytometry | Proteobacteria, Actinobacteriota, Campylobacterota, Streptococcaceae, Enterobacteriaceae, Klebsiella, Streptococcus | Bacteroidota, Firmicutes, Lachnospiraceae, Ruminococcaceae, Oscillospiraceae, Rikenellaceae, Barnesiella, Agathobacter, Prevotella | Cross-sectional design; small sample size; dietary/age confounders; 16S limits function | HBV-CLD progression involves enrichment of pro-inflammatory taxa and depletion of butyrate producers, correlating with T-cell immunosuppression |
| Hsu et al[72], 2022 | 126 | HCV patients: 42 (pre-Tx: 42, post-Tx: 42), HCs: 84 | Prospective cohort; 16S rRNA (V3-V4), DADA2, LEfSe; matched controls | Coriobacteriaceae, Staphylococcaceae, Peptostreptococcaceae, Succinivibrionaceae | Morganellaceae, Pasteurellaceae, Moraxellaceae | Short-term follow-up (12 weeks post-DAA); modest sample size; exploratory differential taxa need validation; cirrhosis subgroup is small | Gut microbiota differs between HCV patients and HCs, but DAA-induced viral eradication does not significantly alter diversity or composition, suggesting viremia is not the main driver of dysbiosis |
| Wang et al[73], 2025 | 20 | Group M (minimal injury): 9, group S (significant injury): 11 | Human: Metagenomic sequencing. Mouse: 16S rRNA sequencing (V3-V4), FMT | Parabacteroides distasonis, Bacteroides dorei, Bacteroides finegoldii, Bacteroides ovatus, Bacteroides clarus | Eubacterium sp. CAG_180, Gemmiger formicilis, Oscillibacter sp. ER4, Subdoligranulum variabile, Faecalibacterium sp. CAG_74_58_120 | Small sample size | Gut dysbiosis contributes to histological liver damage in early CHB. FMT from an HBV-cirrhosis donor aggravated fibrosis in mice, implicating BA-microbiota crosstalk in disease progression |
| Yang et al[74], 2023 | 950 | Viral hepatitis: 656 (HBV: 546, HCV: 86, HEV: 24), HC: 294 | Meta-analysis of 13 studies; 16S rRNA sequencing (multiple regions) | Butyricimonas, Escherichia-Shigella, Lactobacillus, Veillonella | Clostridia_UCG-014, Dorea, Monoglobus, Ruminococcus | Lack of HAV/HDV data; variability in sequencing regions/platforms; cannot establish causality | Viral hepatitis reduces gut microbial diversity, with specific taxa and functions (tryptophan metabolism, LPS biosynthesis) serving as potential biomarkers and contributing to disease pathogenesis |
Table 4 Studies investigating microbiota and coronavirus disease 2019
| Ref. | COVID-related cases (n) | Methodology | Increased taxa | Decreased taxa | Limitations | Conclusion |
| de Nies et al[119], 2023 | 118 subjects COVID-19: 61 (asymptomatic-moderate). Control: 57 | Metagenomics, metatranscriptomics, MAG reconstruction, VF/ARG prediction, virome analysis | Prevotella stercorea, Prevotella spp. CAG 520, Roseburia spp. CAG 471, Firmicutes (AM10); Betaherpesvirus; Rotavirus C; VFs & AMR genes (Acidaminococcaceae, Erysipelatoclostridiaceae) | Turicibacter sanguinis, Roseburia faecis, Firmicutes (CAG 145) | Limited to asymptomatic-moderate cases; single-region cohort (Luxembourg); unclear mechanisms linking SARS-CoV-2 to VF/ARG expression | COVID-19 increases the virulence and AMR potential of commensals despite minimal taxonomic shifts, suggesting enhanced pathogenic potential of the gut microbiota |
| Li et al[120], 2025 | 121 participants total, NC: 53 (no COVID-19), C3M: 27 (3 months post-recovery), C6M: 41 (6 months post-recovery) | Metagenomic shotgun sequencing; ITS sequencing for fungi | Blautia massiliensis, Kluyveromyces spp., Bacteroides xylanisolvens, Phocaeicola vulgatus, Weissella confusa, Streptococcus thermophilus, Asterotremella spp., Gibberella spp. | Blautia wexlerae, Bifidobacterium pseudocatenulatum, Bifidobacterium longum, Eubacterium rectale, Anaerobutyricum hallii, Pyrenochaeta spp. | Cross-sectional design; excluded long COVID; smaller 3-month group; not generalizable to severe or unvaccinated cases | Mild COVID-19 induces long-term gut bacterial and fungal alterations lasting ≥ 6 months, with partial recovery and persistence of some pathogens |
| Zhang et al[121], 2023 | 187 recovered patients (84 symptomatic) | 16S rRNA sequencing; clinical surveys (SF-36, SAS, SDS); lab tests; pulmonary function; chest CT | Veillonella | SCFA-producers: Eubacterium hallii group, Subdoligranulum, Ruminococcus, Dorea, Coprococcus, Eubacterium ventriosum group, Agathobacter | Single-center, cross-sectional; no longitudinal monitoring; diet/Lifestyle not fully controlled; mechanisms not clarified | Long COVID (approximately 45% at 1 year) is linked to persistent dysbiosis marked by depletion of SCFA-producing commensals, correlating with impaired quality of life, anxiety/depression, and immune dysregulation, supporting gut-lung and gut-brain axis involvement |
| Ishizaka et al[122], 2024 | 56 total, PLWH-CoV: 12 (mild: 7; moderate/severe: 5), PLWH controls: 25, HCs: 19 | 16S rRNA sequencing | Acute: Enterococcus faecium. Recovery (1-3 months): Roseburia, Lachnospiraceae_unclassified, Faecalibacterium prausnitzii, Eubacterium rectale. Recovered vs long COVID: Prevotella spp. | Acute vs HC: Roseburia, Lachnospiraceae_unclassified. Long COVID vs recovered: Prevotella spp. | Small sample size (n = 12 PLWH-CoV, only 2 PASC); no pre-infection baseline; diet, ART regimens, and variant effects not analyzed | SARS-CoV-2 in PLWH causes persistent dysbiosis, marked by loss of SCFA-producers and enrichment of pathogens, with severity-linked delays in microbiome recovery and risk of PASC |
| Brīvība et al[123], 2024 | 146 COVID-19 patients (92 hospitalized, 54 ambulatory) vs 110 HCs | Shotgun metagenomics | Enterococcus faecium, Bacteroides spp., Alistipes, Enterobacteriaceae | Roseburia, Faecalibacterium prausnitzii, Lachnospiraceae, Eubacterium rectale, Prevotella spp. | High antibiotic use; heterogeneous sampling timing; phenotypic heterogeneity across patient groups | Acute COVID-19 shows reduced diversity with loss of butyrate producers; recovery involves their restoration, while Prevotella may protect against long COVID |
| Sorokina et al[124], 2023 | 39 post-COVID-19 patients before and after 14-day rehabilitation vs 48 healthy volunteers | Clinical questionnaires, CT; CBC, coagulation, biochemistry; serum IL-6, NSE (ECL); metabolites (GC-MS); microbiota (RT-PCR, colonoflor-16 kit) | Bacteroides spp., Escherichia coli, Enterobacter spp., Staphylococcus aureus, IL-6, succinic acid, fumaric acid, 4-hydroxybenzoic acid | Lactobacillus spp., Bifidobacterium spp., Faecalibacterium prausnitzii, Phenylpropionic acid | Small cohort; no untreated controls; RT-PCR instead of sequencing; limited GI symptom assessment; diet confounded results | Post-COVID-19 is marked by persistent dysbiosis (loss of SCFA-producers, enrichment of pathobionts) and sustained inflammatory/metabolic disturbances not resolved by standard rehabilitation, highlighting the need for personalized microbiome-targeted interventions |
| Tkacheva et al[125], 2023 | 178 post-COVID-19 patients: Asymptomatic (A, n = 48), non-infected contacts (N, n = 46), severe (S, n = 86) | 16S rRNA sequencing | RF39 (order), Clostridia UCG-014, Oscillospirales UCG-010, Akkermansia, Prevotellaceae (family), Lactobacillus, Romboutsia, Ruminococcus gnavus, Erysipelatoclostridium | Parasutterella, Flavonifractor, Ruminococcus gnavus, Subdoligranulum, Methanobrevibacter, Lachnospiraceae UCG-010, Lachnospiraceae NK4A136, Barnesiella, Eubacterium xylanophilum, Eubacterium siraeum | Cross-sectional (3 months only); no acute phase data; diet/medications not controlled; 16S lacks functional resolution | No major post-COVID microbiome differences by infection/severity at 3 months, but taxa correlated with immune, cardiovascular, and metabolic parameters, highlighting systemic associations beyond direct viral effects |
| Bredon et al[126], 2025 | 200 COVID-19 patients vs 102 HCs (Morocco & France cohorts) | Shotgun metagenomic sequencing, machine learning, metabolomics (tryptophan) | Ruminococcus gnavus, Klebsiella pneumoniae, K. variicola, Bacteroides ovatus, Enterococcus; ↑ L-tryptophan biosynthesis | Faecalibacterium prausnitzii, Roseburia spp., Bifidobacterium longum, Dysosmobacter welbionis, Coprococcus comes (SCFA-producers) | Treatment heterogeneity (antibiotics) in the French cohort; ML model not transferable; causality not established | COVID-19 induces gut dysbiosis with depletion of SCFA-producers and enrichment of pathobionts, alongside altered tryptophan metabolism. Dysbiosis correlates with disease severity; the ML model predicted severity in the Moroccan cohort |
Table 5 Therapeutic effects of probiotic supplementation on infectious disease outcomes
| Probiotic/strain | Virus | Model | Individuality | Mechanism against viral infection | Ref. |
| Lactobacillus casei 393 | HIV-1 (pseudovirus) | In vitro (TZM-bl cells) | HIV-infected adults | Expresses CD4; binds HIV-1 pseudovirus; ↓ infection approximately 60%-70% | [138] |
| Lactobacillus rhamnosus GR-1 and L. reuteri RC-14 | HIV | Clinical (24 adult women) | HIV-infected women | Resolved diarrhea; ↑/stable CD4 in 11/12 vs 3/12 control | [139] |
| Lactobacillus acidophilus ATCC 4356 engineered with human CD4 (in yogurt) | HIV-1 | In vitro & humanized BLT mice | HIV-infected women (Nigeria) | Surface CD4 binds HIV gp120; blocks viral entry | [140] |
| Lactobacillus casei Shirota (in fermented milk) | HIV | Clinical (children, | HIV-infected Vietnamese children | ↑ CD4+, Th2, Th17; ↓ Tregs, CD8+ activation; ↓ viral load | [141] |
| Lactobacillus casei Shirota | EBV, CMV (herpesviruses) | Clinical (athletes) | Healthy athletes | ↓ Plasma EBV and CMV IgG titres in seropositive individuals suggest improved immune control of viral reactivation | [142] |
| Lactobacillus rhamnosus GG | Rotavirus, Cryptosporidium | Clinical (children) | Children with gastroenteritis | Improved intestinal barrier function (↓ L:M ratio); ↑ serum IgG (rotavirus); ↓ reinfection rates in rotavirus-positive children | [143] |
| Lactobacillus casei Shirota | Norovirus | Clinical (elderly) | Older adults in a care facility | Shortened fever duration; modulated gut microbiota (↑ Lactobacillus, Bifidobacterium; ↓ Enterobacteriaceae); ↑ acetic acid | [144] |
| Lactobacillus rhamnosus GG | Rhinovirus | Clinical (infants) | Preterm infants (32-36 weeks) | ↓ RTI incidence, ↓ rhinovirus infections; no effect on severity or shedding | [145] |
| Enterococcus faecium NCIMB 10415 | Swine influenza A (H1N1, H3N2) | In vitro (cell culture) | Porcine macrophage and epithelial cells | ↓ Virus titers (up to 4-log); ↑ cell viability; ↑ NO release; ↓ TNF-α, IL-6, TLR3; ↑ IL-10, IFN-α; direct virus trapping by bacteria | [146] |
| Bifidobacterium longum BB536 | Influenza | Clinical (elderly) | Elderly (mean age 86.7 years) | ↓ Influenza and fever incidence, ↑ NK cell activity and neutrophil bactericidal activity; maintained innate immunity | [147] |
| Bifidobacterium adolescentis SPM0212 | HBV | In vitro (HepG2.2.15 cells) | Human hepatocyte-derived cell line | ↓ HBsAg and extracellular HBV DNA; ↑ MxA, STAT1 expression via IFN pathway; active antiviral components < 30 kDa | [148] |
| Bifidobacterium lactis DSM 32246, Bifidobacterium lactis DSM 32247, Lactobacillus acidophilus DSM 32241, Lactobacillus brevis DSM 27961, Lactobacillus helveticus DSM 322426 | SARS-CoV-2 | Clinical (hospitalized) | Hospitalized COVID-19 patients (Italy, n = 70) | ↓ Progression to respiratory failure; ↑ symptom resolution (fever, diarrhea, dyspnea); ↓ ICU admission and mortality; modulation of gut-lung axis; potential ↑ Nrf2/HO-1 antiviral pathways | [149] |
| Lactobacillus gasseri SBT2055 | RSV | In vivo and in vitro | BALB/c mice and HEp-2 cells | ↓ RSV titer; ↓ IL-6, TNF-α, IL-1β, CCL2; ↑ IFN-β, IFN-γ, OAS1a, ISG15; ↓ SRCAP expression linked to RSV replication | [150] |
- Citation: Velikova T, Ali H, Batselova H, Chervenkov L, Miteva D, Peruhova M, Gulinac M, Tomov L, Mitova-Mineva Y, Velev V. Interplay between viral infections and gut microbiota dysbiosis: Mechanisms and therapeutic potential. World J Gastroenterol 2026; 32(3): 112437
- URL: https://www.wjgnet.com/1007-9327/full/v32/i3/112437.htm
- DOI: https://dx.doi.org/10.3748/wjg.v32.i3.112437
