BPG is committed to discovery and dissemination of knowledge
Review
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
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 norovirusE. cloacae, enteric microbiotaBacterial histo-blood group antigen analogs support persistent viral infection in the gut[8,9]
Influenza virusStaphylococcus aureusBacterial proteases activate viral hemagglutinin through cleavage[10]
RotavirusGut 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]
HIVMycobacterium tuberculosisMycobacterial components enhance HIV transcriptional activity[13]
Indirect interactions
AdenovirusS. pneumoniaeEnhances bacterial attachment to respiratory epithelial cells[14]
RhinovirusRespiratory pathogensIncreases the expression of host cell adhesion molecules for bacterial binding[15]
Measles virusMultiple opportunistic pathogensSystemic immune suppression facilitates secondary bacterial infections[16,17]
HerpesvirusesPorphyromonas gingivalis, Bacteroides forsythus (now Tannerella forsythia), Campylobacter rectusViral immunosuppression enables bacterial colonization of the oral mucosa[18]
Parainfluenza virusStreptococcus pneumoniae, Haemophilus influenzaeViral 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 generalizabilityLTC-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]9161 HIVMetagenomics sequencingFaecalibacterium prausnitzii, Subdoligranulum sp., Coprococcus comes, Prevotella copri, Prevotella stercorea, Bacteroides coprophilus, Bacteroides coprocola, Bacteroides intestinalis, Bacteroides salyersiaeBacteroidesSmall sample size; cross-sectional; single population (Chinese); ART regimen effects not fully differentiatedHIV 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]383HIV- (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 modelsViremic group: Peptoniphilus, Porphyromonas, Prevotella, MurdochiellaViremic group: Bacteroides, Brachyspira, Faecalibacterium, Helicobacter. All HIV+ groups: BacteroidesLack of dietary data; residual confounding possible; 16S limits species-level resolution; no data on time since ART initiationHIV 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-responders16S rDNA gene sequencingMegamonas, Desulfovibrionales (Proteobacteria)Clostridiales (esp. Clostridiaceae, Catenibacterium), Lachnospiraceae, ProteobacteriaSmall sample size; did not analyze fungal mycobiome (including probiotic colonization); cannot establish causality; short intervention periodSaccharomyces boulardii reduced pro-inflammatory Clostridiaceae; immunologic non-responders had higher baseline Lachnospiraceae and Proteobacteria
Dillon et al[48]3218 untreated HIV+16S rRNA sequencing (colonic mucosa and stool)Prevotella stercoreaButyrate-producing bacteria: Roseburia intestinalis, Faecalibacterium prausnitzii, Eubacterium rectale (in colonic mucosa)Small cross-sectional study; sexual preference unmatched; fecal butyrate not measuredHIV 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- controls21 (chronic HIV, suppressed VL)16S rRNA pyrosequencing (V3-V5 region)Proteobacteria, Gammaproteobacteria, Enterobacteriales, Enterobacteriaceae, Erysipelotrichi, Erysipelotrichales, Erysipelotrichaceae, BarnesiellaRikenellaceae, AlistipesSmall sample size; exploratory design; no correction for multiple comparisons; immune activation markers not assessedART-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: 2026 (all on suppressive ART)16S rRNA sequencing (V4 region, rectal swabs)Fusobacterium (phylum: Fusobacteria), Gallicola, BilophilaLactobacillales, CorynebacteriumSmall sample size; all male participants; dietary data not collected; rectal swabs may not fully reflect luminal microbiotaPoor 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+ mothers16S rRNA sequencing (V3-V4 region)HEU children: Faecalibacterium prausnitzii, Prevotella copri, Lachnospiraceae, Klebsiella, Lactococcus, Ruminococcaceae, EnterobacteriaceaeHEU children: Bacteroides uniformis, ParaprevotellaCross-sectional, small sample; breastfeeding differences between HEU and unexposed children; functional predictions inferred, not measuredHEU 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, MegamonasBacteroidetes, Bacteroides, Dialister, Clostridium XIVa, Clostridium XIVb, Barnesiella, CoprococcusCross-sectional; all male; sexual behavior not controlled; fecal samples may not reflect mucosa-associated microbiotaHIV-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, HowardellaFirmicutes, Lachnospiraceae, Oxalobacteraceae, Eggerthella, Barnesiella, Odoribacter, Oscillospira, Oxalobacter, Roseburia, AlistipesSmall, mostly male cohort; observational; prebiotic/probiotic use uncontrolled; genus-level data limits species resolutionHIV 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]168 HIV+ (HAART-naïve, low CD4 counts in 6/8)16S rRNA sequencing (V3-V4)Proteobacteria, Burkholderia (specifically Burkholderia fungorum), Bradyrhizobium (specifically Bradyrhizobium pachyrhizi), Ralstonia, FusobacteriumFirmicutes, LactobacillusSmall sample; case-control design limits causation; no HAART-treated comparison; clinical symptom correlations limitedHIV-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 (n)
Cirrhosis-related cases (n)
Methodology
Increased taxa
Decreased taxa
Limitations
Conclusion
Elsherbiny et al[62], 202580 (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, MuribaculaceaeSVR group: Actinobacteria. All CHC vs HCs: Reduced diversity; Bacteroides, Agathobacter, ParabacteroidesSingle-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], 2023120 subjects (180 samples)Control: 60 HCs. CHC (pre-DAA): 60 patients. SVR24 (post-DAA): 60 patients16S rRNA sequencing (V3-V4)Ruminococcaceae, Eubacterium, Agathobacter, Alistipes, Bifidobacterium, Klebsiella, Lactobacillus, Actinobacteria, Firmicutes, LactobacillusBacteroidetes, LachnoclostridiumRelatively small sample size; short follow-up (24 weeks); unmeasured confounders (diet, smoking); baseline differences in liver function between CHC and control groupsGut 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], 202570 CHB patients + 8 HCsFunctional cure: 18 (HBsAg-, HBV DNA-). Low-titer DNA: 40. High-titer DNA: 12. HC: 816S rRNA sequencing (V3-V4)Clostridium bartlettii, Butyricimonas, Coprococcus catus, Bifidobacterium breve, CampylobacterNot reportedSmall sample size; exploratory design; in vitro SCFA concentrations may not reflect physiological levels in the liverButyrate-producing bacteria are enriched in HBsAg-negative patients; sodium butyrate directly suppresses HBsAg production in infected hepatocytes, especially post-infection
Inoue et al[65], 2025272 (174 active HCV, 75 post-SVR, 23 HCs)CH-HCV: 95, LC/HCC-HCV: 79, CH-SVR: 29, LC/HCC-SVR: 46, healthy: 2316S rRNA sequencing; fecal BA profiling; RNA-seqBlautia, Fusicatenibacter, Roseburia, Faecalibacterium, Subdoligranulum, CollinsellaStreptococcus, Streptococcus salivarius, Eubacterium hallii group, Ruminococcus torques groupCohort separation for multi-omic analyses; limited SVR48 sample size; partially uses database-derived RNA-seq dataHCV eradication partially restores gut dysbiosis and BA profiles, with post-SVR Blautia enrichment correlating with improved liver fibrosis and function
Li et al[66], 202588AHE-elderly: 58, HCs-elderly: 30, self-healing: 46, non-self-healing: 1216S rRNA sequencingAHE-elderly vs HC: Firmicutes, Lactobacillales, Bacilli, Streptococcaceae. Non-self-healing vs self-healing: Bifidobacteriaceae, Bacteroidia, Bacteroides fragilisAHE-elderly vs HC: Proteobacteria, Bacteroidetes. Self-healing vs non-self-healing: Firmicutes, Bacillus, Lactobacillus, StreptococcusNo significant difference in alpha diversity; lack of longitudinal data; unclear causal relationship between microbiota changes and HEV infectionBacteroidetes distinguish AHE patients from controls, with Bacteroides fragilis enriched in non-self-healing cases and serving as a predictive biomarker
Li et al[67], 202579HBC: 46, HCs: 33, BA-N: 24, BA-H: 2216S rRNA sequencing (V4-V5)Streptococcus, Veillonella, LactobacillalesBacteroides, Akkermansia, ClostridialesCross-sectional design; small sample size; no BA composition or metabolomic analysis; potential confounders (diet, medication) not fully addressedHBC 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], 2025123HBV-LC: 83, HC: 40, MELD < 21: 68, MELD ≥ 21: 15, CTP-C: 2216S rRNA sequencing (V3-V4)Klebsiella, Streptococcus, Fusobacterium, EnterococcusAlistipes, Lachnospira, Agathobacter, Parabacteroides, RoseburiaSingle-center design; modest sample size; cross-sectional design limits causal inference; 16S rRNA does not capture full functional potentialHBV-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], 202142Pre-DAA: 14, EOT: 14, post-24: 14 (samples from the same 14 patients)16S rRNA sequencing (V3-V4)Faecalibacterium, BacillusBacteroides, FusobacteriumSmall sample size; lack of a HC group; intra-personal comparison only; cannot determine if pre-treatment microbiota differed from healthy individualsHCV 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], 202462OBI: 24, HBV carriers: 18, HCs: 2016S rRNA sequencing (V3-V4)Subdoligranulum, MegamonasFaecalibacteriumSmall sample size; all participants were male; potential unmeasured confounders (diet); cannot establish causalityOBI 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], 20239030 HCs + 30 HBV-LC + 30 HBV-HCC16S rRNA sequencing (V3-V4) + flow cytometryProteobacteria, Actinobacteriota, Campylobacterota, Streptococcaceae, Enterobacteriaceae, Klebsiella, StreptococcusBacteroidota, Firmicutes, Lachnospiraceae, Ruminococcaceae, Oscillospiraceae, Rikenellaceae, Barnesiella, Agathobacter, PrevotellaCross-sectional design; small sample size; dietary/age confounders; 16S limits functionHBV-CLD progression involves enrichment of pro-inflammatory taxa and depletion of butyrate producers, correlating with T-cell immunosuppression
Hsu et al[72], 2022126HCV patients: 42 (pre-Tx: 42, post-Tx: 42), HCs: 84Prospective cohort; 16S rRNA (V3-V4), DADA2, LEfSe; matched controlsCoriobacteriaceae, Staphylococcaceae, Peptostreptococcaceae, SuccinivibrionaceaeMorganellaceae, Pasteurellaceae, MoraxellaceaeShort-term follow-up (12 weeks post-DAA); modest sample size; exploratory differential taxa need validation; cirrhosis subgroup is smallGut 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], 202520Group M (minimal injury): 9, group S (significant injury): 11Human: Metagenomic sequencing. Mouse: 16S rRNA sequencing (V3-V4), FMTParabacteroides distasonis, Bacteroides dorei, Bacteroides finegoldii, Bacteroides ovatus, Bacteroides clarusEubacterium sp. CAG_180, Gemmiger formicilis, Oscillibacter sp. ER4, Subdoligranulum variabile, Faecalibacterium sp. CAG_74_58_120Small sample size (n = 20); FMT from a single cirrhotic donor; mechanistic pathways (BA crosstalk) require further validationGut 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], 2023950Viral hepatitis: 656 (HBV: 546, HCV: 86, HEV: 24), HC: 294Meta-analysis of 13 studies; 16S rRNA sequencing (multiple regions)Butyricimonas, Escherichia-Shigella, Lactobacillus, VeillonellaClostridia_UCG-014, Dorea, Monoglobus, RuminococcusLack of HAV/HDV data; variability in sequencing regions/platforms; cannot establish causalityViral 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], 2023118 subjects COVID-19: 61 (asymptomatic-moderate). Control: 57Metagenomics, metatranscriptomics, MAG reconstruction, VF/ARG prediction, virome analysisPrevotella 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 expressionCOVID-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], 2025121 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 fungiBlautia 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 casesMild 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], 2023187 recovered patients (84 symptomatic)16S rRNA sequencing; clinical surveys (SF-36, SAS, SDS); lab tests; pulmonary function; chest CTVeillonellaSCFA-producers: Eubacterium hallii group, Subdoligranulum, Ruminococcus, Dorea, Coprococcus, Eubacterium ventriosum group, AgathobacterSingle-center, cross-sectional; no longitudinal monitoring; diet/Lifestyle not fully controlled; mechanisms not clarifiedLong 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], 202456 total, PLWH-CoV: 12 (mild: 7; moderate/severe: 5), PLWH controls: 25, HCs: 1916S rRNA sequencingAcute: 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 analyzedSARS-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], 2024146 COVID-19 patients (92 hospitalized, 54 ambulatory) vs 110 HCsShotgun metagenomicsEnterococcus faecium, Bacteroides spp., Alistipes, EnterobacteriaceaeRoseburia, Faecalibacterium prausnitzii, Lachnospiraceae, Eubacterium rectale, Prevotella spp.High antibiotic use; heterogeneous sampling timing; phenotypic heterogeneity across patient groupsAcute 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], 202339 post-COVID-19 patients before and after 14-day rehabilitation vs 48 healthy volunteersClinical 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 acidLactobacillus spp., Bifidobacterium spp., Faecalibacterium prausnitzii, Phenylpropionic acidSmall cohort; no untreated controls; RT-PCR instead of sequencing; limited GI symptom assessment; diet confounded resultsPost-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], 2023178 post-COVID-19 patients: Asymptomatic (A, n = 48), non-infected contacts (N, n = 46), severe (S, n = 86)16S rRNA sequencingRF39 (order), Clostridia UCG-014, Oscillospirales UCG-010, Akkermansia, Prevotellaceae (family), Lactobacillus, Romboutsia, Ruminococcus gnavus, ErysipelatoclostridiumParasutterella, Flavonifractor, Ruminococcus gnavus, Subdoligranulum, Methanobrevibacter, Lachnospiraceae UCG-010, Lachnospiraceae NK4A136, Barnesiella, Eubacterium xylanophilum, Eubacterium siraeumCross-sectional (3 months only); no acute phase data; diet/medications not controlled; 16S lacks functional resolutionNo 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], 2025200 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 biosynthesisFaecalibacterium prausnitzii, Roseburia spp., Bifidobacterium longum, Dysosmobacter welbionis, Coprococcus comes (SCFA-producers)Treatment heterogeneity (antibiotics) in the French cohort; ML model not transferable; causality not establishedCOVID-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 393HIV-1 (pseudovirus)In vitro (TZM-bl cells)HIV-infected adultsExpresses CD4; binds HIV-1 pseudovirus; ↓ infection approximately 60%-70%[138]
Lactobacillus rhamnosus GR-1 and L. reuteri RC-14HIVClinical (24 adult women)HIV-infected womenResolved diarrhea; ↑/stable CD4 in 11/12 vs 3/12 control[139]
Lactobacillus acidophilus ATCC 4356 engineered with human CD4 (in yogurt)HIV-1In vitro & humanized BLT miceHIV-infected women (Nigeria)Surface CD4 binds HIV gp120; blocks viral entry[140]
Lactobacillus casei Shirota (in fermented milk)HIVClinical (children, n = 60)HIV-infected Vietnamese children↑ CD4+, Th2, Th17; ↓ Tregs, CD8+ activation; ↓ viral load[141]
Lactobacillus casei ShirotaEBV, 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 GGRotavirus, CryptosporidiumClinical (children)Children with gastroenteritisImproved intestinal barrier function (↓ L:M ratio); ↑ serum IgG (rotavirus); ↓ reinfection rates in rotavirus-positive children[143]
Lactobacillus casei ShirotaNorovirusClinical (elderly)Older adults in a care facilityShortened fever duration; modulated gut microbiota (↑ Lactobacillus, Bifidobacterium; ↓ Enterobacteriaceae); ↑ acetic acid[144]
Lactobacillus rhamnosus GGRhinovirusClinical (infants)Preterm infants (32-36 weeks)↓ RTI incidence, ↓ rhinovirus infections; no effect on severity or shedding[145]
Enterococcus faecium NCIMB 10415Swine 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 BB536InfluenzaClinical (elderly)Elderly (mean age 86.7 years)↓ Influenza and fever incidence, ↑ NK cell activity and neutrophil bactericidal activity; maintained innate immunity[147]
Bifidobacterium adolescentis SPM0212HBVIn 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 322426SARS-CoV-2Clinical (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 SBT2055RSVIn vivo and in vitroBALB/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]