Next Article in Journal
Biomolecule–Photosensitizer Conjugates: A Strategy to Enhance Selectivity and Therapeutic Efficacy in Photodynamic Therapy
Previous Article in Journal
Evolution of Approaches to the Development, Life Cycle Control, and Interchangeability of Veterinary Biosimilars Based on Hemoproteins (with a Focus on Cytochrome C)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Herbal and Natural Products for Antibiotic-Associated Diarrhea: A Systematic Review of Animal Studies Focusing on Molecular Microbiome and Barrier Outcomes

1
Department of Acupuncture & Moxibustion Medicine, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
2
Department of Korean Internal Medicine, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2026, 19(1), 64; https://doi.org/10.3390/ph19010064 (registering DOI)
Submission received: 27 November 2025 / Revised: 22 December 2025 / Accepted: 24 December 2025 / Published: 29 December 2025
(This article belongs to the Section Natural Products)

Abstract

Background/Objectives: Antibiotic-associated diarrhea (AAD) arises from antibiotic-induced disruption of microbial diversity, metabolic activity, epithelial integrity, and mucosal immunity. Probiotics are widely used but often show limited efficacy under antibiotic pressure. Herbal and natural products (HNPs) may provide multi-target benefits by modulating microbiota-dependent and host-directed pathways. This review synthesized animal studies evaluating HNP or HNP–probiotic combination (HNP–C) therapies using molecular microbiome endpoints. Methods: Following PRISMA 2020 guidelines, controlled in vivo studies assessing HNP or HNP–C interventions for AAD were searched in Pubmed, EMBASE, Web of Science, Scopus, and CNKI through November 2025. Eligible studies reported microbial diversity, taxonomic shifts, short-chain fatty acids (SCFAs), barrier markers, or immune responses. Risk of bias was assessed using the SYRCLE tool. Due to heterogeneity, findings were narratively synthesized. Results: Twenty-seven studies met inclusion criteria (21 HNP, 6 HNP–C). HNP monotherapies consistently improved α-diversity, shifted β-diversity toward healthy controls, restored SCFA-producing taxa, and increased SCFA levels. They also enhanced tight junction proteins and reduced inflammatory cytokines. HNP–C interventions demonstrated more comprehensive microbial, epithelial, and immune recovery; however, only two studies included direct comparisons among HNP-only, probiotic-only, and combination groups. In these, HNP–C showed greater improvements than individual components, suggesting complementary or potentially complementary or additive effects. Other HNP–C studies were limited by absent comparator arms. Conclusions: HNPs appear to support recovery of microbial diversity, metabolic function, epithelial barrier integrity, and immune regulation by engaging microbiota-dependent and host-mediated mechanisms. HNP–C strategies may offer complementary benefits, although rigorously controlled comparative studies and clinical validation remain needed.

Graphical Abstract

1. Introduction

Antibiotic-associated diarrhea (AAD) is a common and clinically significant adverse effect of antibiotic therapy, affecting 5% to over 30% of patients depending on individual susceptibility [1,2]. Beyond causing transient gastrointestinal symptoms, AAD disrupts intestinal homeostasis, reduces treatment compliance, and increases the risk of recurrent infection [3,4]. Broad-spectrum antibiotics produce abrupt shifts in the gut microbiota by rapidly reducing microbial abundance and diversity, which weakens colonic resistance [5]. They also inhibit short-chain fatty acid (SCFA) production and compromise tight junction integrity between epithelial cells [4,6,7]. These disturbances increase intestinal permeability, heighten inflammatory responses, and create conditions that favor opportunistic pathogens [8]. Importantly, microbiota disruption can persist for months after antibiotic cessation, indicating delayed or incomplete ecological recovery [9] and highlighting the importance of microbial stability and functional redundancy in host protection [10].
Conventional management strategies, including probiotics, dietary modifications, and symptomatic therapies, show variable effectiveness. Although probiotics replenish beneficial microbiota, they generally colonize the gut only transiently and inconsistently during or after antibiotic treatment [11,12]. Prebiotics promote the growth of commensal bacteria but have limited capacity to directly restore the epithelial barrier or immune function [12]. Growing interest has therefore focused on herbs and natural products (HNPs), which target the microbiota while simultaneously modulating multiple pathological dimensions, including microbiota structure, SCFA metabolism, barrier integrity, and mucosal immunity [13,14,15,16]. For example, polysaccharides supply selectively fermentable substrates that enhance SCFA production and support the recovery of symbiotic bacteria, whereas flavonoids and saponins modulate host inflammatory pathways such as NF-κB and MAPK and contribute to restoring tight junctions [14,16].
Several herbal medicines, including Zingiber officinale, Shenlingbaizhusan, and Xianglian Pill, have been evaluated in animal models of AAD [17,18,19], but none included microbiome sequencing results. This underscores the importance of incorporating microbiological endpoints when assessing therapies for microbiome-driven diseases. Despite growing interest in herbal treatments targeting the microbiota, many studies, including preclinical evaluations of gut-protective effects, do not elucidate underlying microbiological mechanisms by integrating molecular microbiome profiling such as 16S rRNA sequencing [13,14,17]. This knowledge gap is also reflected in existing literature reviews. For instance, a pediatric meta-analysis of AAD primarily focused on clinical endpoints without assessing the microbiome [20]. Likewise, a recent narrative review summarized the role of herbal polysaccharides in chronic diarrhea phenotypes, including irritable bowel syndrome with diarrhea (IBS-D), osmotic diarrhea, bile acid diarrhea, and AAD, but did not systematically integrate controlled in vivo AAD studies that included microbiome sequencing [21]. Similarly, recent systematic reviews of herbal medicines for AAD have focused on clinical outcomes, with limited microbiome endpoints [22].
Therefore, despite growing evidence that HNPs can modulate the gut microbiota, a synthesis focusing on controlled AAD animal studies integrating microbiological, metabolic, and barrier–immune endpoints remains lacking. To address this gap, this systematic review limits its scope to single-component HNPs and simple herbal medicine–probiotic combinations, excluding multi-component formulations (more than three components) to maintain mechanistic clarity. The aim of this review was to develop an integrated mechanistic framework by comparing the effects of AAD therapies on microbial diversity, β-diversity, SCFA production, epithelial barrier integrity, and mucosal immune regulation from a microbiota-centered perspective.

2. Materials and Methods

2.1. Study Design

This systematic review was conducted according to PRISMA 2020 guidelines [23], with the completed PRISMA checklist provided in (Supplementary Material Table S1). The review focused exclusively on controlled in vivo animal experiments evaluating herbal or natural product interventions for AAD and reporting quantitative gut microbiota outcomes. This systematic review was prospectively registered in PROSPERO (CRD420251136553) https://www.crd.york.ac.uk/PROSPERO/view/CRD420251136553 (accessed on 23 December 2025).

2.2. Eligibility Criteria

This review included animal models of AAD with no restrictions on species, gender, age, or weight. Interventions were categorized into two main groups: HNP, representing single-agent herbal or natural product interventions, and HNP-C, representing combination approaches involving co-administration with probiotics or synbiotics. Multi-component herbal formulations containing three or more ingredients were excluded from the main synthesis because of compositional heterogeneity and will be addressed in a separate review. Comparative groups included drug vehicles, untreated controls, antibiotic monotherapy, or established positive controls (e.g., probiotics).
The primary outcomes focused on gut microbiota composition assessed by 16S rRNA sequencing or metagenomic profiling, as well as other molecular approaches (e.g., DGGE, PCR-DGGE, or targeted qPCR), provided that microbial composition or taxonomic shifts were reported, including α-diversity, β-diversity, and taxonomic changes. Secondary outcomes included SCFAs, bile acids, tight junction proteins (e.g., ZO-1, occludin, claudin-1), mucins, SIgA, cytokines, and markers of systemic or mucosal inflammation.
Included study types were limited to controlled in vivo animal studies published in peer-reviewed journals. Exclusion criteria consisted of literature reviews, study protocols, in vitro studies, non-AAD models, probiotic monotherapy interventions, and studies lacking extractable microbiome data.

2.3. Information Sources

A comprehensive search of major international databases PubMed/MEDLINE, EMBASE, Web of Science, Scopus, and CNKI (China National Knowledge Infrastructure) was conducted from database inception to November 2025. An additional search of domestic medical databases, including OASIS, KISS, and KMbase, was performed but yielded no further eligible studies. No language restrictions were applied to ensure maximal capture of relevant preclinical evidence.

2.4. Search Strategy

The search strategy incorporated both controlled vocabulary terms (e.g., MeSH) and free-text keywords related to the following:
-
antibiotic-associated diarrhea
-
antibiotic-induced dysbiosis
-
herbal medicine
-
natural product
-
polysaccharide
-
specific herbal agents
-
gut microbiota
-
microbiome
-
16S rRNA
-
animal model
Equivalent Chinese terms were applied for CNKI searches (Subject Term).
Full database-specific search strings will be provided in Table S1.

2.5. Study Selection

All retrieved records were consolidated and deduplicated before screening. Two reviewers independently screened titles and abstracts, followed by full-text evaluation according to predefined eligibility criteria. Discrepancies were resolved through discussion.

2.6. Data Extraction

A standardized extraction form was developed. Extracted variables included study characteristics, animal details, antibiotic regimen, intervention type (HNP or HNP-C), microbiome assessment method, α- and β-diversity indices, key taxonomic changes, SCFAs, barrier markers, immune mediators, main findings, and study limitations. Numeric data were extracted directly, while graphical data were collected only when values were clearly interpretable.

2.7. Risk of Bias Assessment

Risk of bias was evaluated independently by two reviewers using the SYRCLE tool [24], with disagreements resolved by consensus.

2.8. Data Synthesis

Due to substantial methodological heterogeneity across animal models, antibiotic protocols, intervention types, and microbiome analytic platforms, a quantitative meta-analysis was not planned. Instead, findings were synthesized narratively, with comparative mapping of outcomes between HNP and HNP-C interventions.

3. Results

3.1. Study Selection

A total of 232 records were identified through searches of the PubMed, EMBASE, Web of Science, Scopus, and CNKI databases. After removing 168 duplicates, 64 full-text articles were assessed for eligibility. Twenty studies were excluded: five used non-AAD models, 13 did not evaluate herbal or natural product interventions, and two lacked extractable microbiome outcomes. Ultimately, 44 studies met the initial inclusion criteria. Of these, 17 multi-component prescription studies were excluded from the primary synthesis because of compositional heterogeneity. The final analysis, therefore, included 21 HNP interventions [25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45] and six HNP-combination (HNP–C) interventions [46,47,48,49,50,51], yielding a total of 27 studies. The study selection process is depicted in the PRISMA flow diagram (Figure 1).

3.2. Study Characteristics

A total of 27 animal studies were included in the final synthesis, comprising 21 HNP interventions [25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45] and six HNP-C interventions [46,47,48,49,50,51]. Most investigations used C57BL/6 or BALB/c mice (81%), while the remaining studies employed Sprague–Dawley rats. Reporting of sex and age was inconsistent across studies.
Among HNP interventions, polysaccharides were the most frequently studied category. These were derived from Astragalus membranaceus [30], Panax species [26,28,45], Poria cocos [31,36,48], edible fungi or marine sources [32,37,38], Dioscorea opposita and related tubers [29,34,35], and plant-derived foods such as bamboo shoot [39] and purple sweet potato [33]. Tangeretin was the primary flavonoid investigated [25], while extracts or gingerols from Zingiber officinale represented the major essential-oil components [17,43,44]. Saponin-based interventions were mainly derived from Cistanche deserticola [41], and blueberry leaf polyphenols were also evaluated as a distinct HNP extract [40].
HNP-C interventions combined probiotics with herbal or natural bioactive compounds, including Panax-derived polysaccharides [46], Astragalus polysaccharides [47], Poria cocos polysaccharide preparations [48], bioactive glycans [49], medicinal minerals such as maifan stone [50], and chitosan oligosaccharides [51], to enhance microbiota restoration.
Most studies assessed gut microbiota composition using 16S rRNA sequencing; however, several studies employed lower-resolution community profiling methods such as DGGE or PCR-DGGE, and one HNP–C study used targeted qPCR without 16S profiling. SCFAs were reported in 17 studies (63%), tight junction or mucin markers in 15 studies (56%), and immune-inflammatory markers in 18 studies (67%).
Owing to heterogeneity in antibiotic regimens, animal strains, and analytic platforms, findings were synthesized narratively rather than through meta-analysis. Detailed study characteristics are presented in Table 1.

3.3. Microbial Diversity and Composition (Table 2 and Table 3)

Comparative summary of therapeutic effects between herbal/natural product (HNP) monotherapy (n = 21) and HNP-combination (HNP-C) interventions (n = 6) in antibiotic-associated diarrhea models. Reporting frequencies indicate the number of studies that assessed each endpoint.

3.3.1. Effects on α-Diversity

Of the 21 HNP studies, 20 quantitatively assessed α-diversity (Shannon, Simpson, Chao1, ACE) [25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45], all demonstrating recovery of richness and evenness suppressed by antibiotics. One study used DGGE without numerical indices [30]. All six HNP-C studies also reported increased α-diversity [46,47,48,49,50,51]; five included quantitative metrics [46,47,48,49,51], whereas one relied on qPCR-based assessment only [50]. Notably, only two studies incorporated herbal-only and probiotic-only comparator arms [47,48], both showing faster or more complete α-diversity restoration with combination therapy. Overall, both HNP and HNP-C interventions reversed antibiotic-induced reductions in α-diversity, with synergistic effects of HNP-C supported exclusively by comparator-controlled studies.

3.3.2. Effects on β-Diversity

Eighteen HNP studies reported β-diversity outcomes using PCoA or NMDS [25,26,27,28,29,31,32,33,35,36,37,38,39,40,41,42,43,44,45], all showing shifts toward the healthy control cluster, although complete normalization was not consistently observed. Three studies provided qualitative clustering results only [30,34].
All six HNP-C studies similarly demonstrated convergence toward the control cluster [46,47,48,49,50,51]. Among these, two studies that included appropriate comparator arms [47,48] reported more pronounced β-diversity shifts with combination therapy.
Other HNP-C studies showed consistent directional trends but lacked the design elements necessary to confirm additive effects. Overall, both intervention types facilitated ecological recovery, with more robust evidence of microbiota remodeling emerging from the limited comparator-designed HNP-C studies.

3.3.3. Key Taxonomic Shifts

Across both intervention categories, consistent taxonomic patterns were observed: enrichment of Lactobacillus, Bifidobacterium, Muribaculaceae, Lachnospiraceae, and Ruminococcaceae, alongside suppression of Escherichia–Shigella, Enterobacteriaceae, and Clostridium sensu stricto [25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51]. Only two HNP-C studies with comparator arms [47,48] showed greater restoration of beneficial taxa and larger reductions in Proteobacteria-associated taxa compared with monotherapy. Overall, both HNP and HNP-C interventions restored commensal abundance, although mechanistic conclusions favoring HNP-C remain restricted to studies with robust comparator designs.
Table 2. Effects of HNP and HNP-C on Gut Microbiota and Host Outcomes.
Table 2. Effects of HNP and HNP-C on Gut Microbiota and Host Outcomes.
GroupStudy IDIntervention (Short)α-Diversity Effectβ-Diversity EffectKey Taxa ChangesSCFABarrier MarkersImmune and Inflammatory MarkersHistopathologyMechanistic Notes
HNPXu et al. [25]Tangeretin (Citrus flavonoid)↑ Diversity (recovery trend)PCoA shifted toward NC↑ Lachnospiraceae, Bacteroidaceae; ↓ Enterobacteriaceae, PseudomonadaceaeAcetate, isobutyrate, butyrate, valerate ↑Not measured (in vitro model)Anti-inflammatory trend inferred from SCFA ↑ & pathogen suppressionNot performedTangeretin restored diversity and SCFAs, enriched beneficial taxa, and reduced opportunistic pathogens
HNPRen et al. [26]American ginseng polysaccharideShannon ↑ (close to NC)PCoA shifted toward NC↑ Bacteroidetes; ↓ Firmicutes, ProteobacteriaNot reportedColon structure repaired (↑ villus length, goblet cells)Not reportedVillus and goblet cell morphology improved; edema reduced.American ginseng polysaccharide alleviated dysbiosis and promoted mucosal repair via microbiota modulation.
HNPMin, et al. [27]Korean red ginseng polysaccharideDiversity indices ↑PCoA approached NC↑ Firmicutes, ↓ ProteobacteriaAcetate and butyrate ↑Lysozyme, claudin-1 ↑Fecal IgA ↑Red ginseng polysaccharide rebalanced microbiota, enhanced SCFA, and reinforced barrier and mucosal immune homeostasis.
HNPQi et al. [28]Neutral ginseng polysaccharideShannon and Chao1 ↑PCoA partially restored toward NC↑ Lactobacillus; ↓ Bacteroides, PseudomonasNot measuredHistologic repair (villus structure)Not reportedVillus atrophy and goblet cell loss reversed.Neutral ginseng polysaccharide increased beneficial flora and reduced pathogens, restoring diversity and mucosal structure.
HNPPan et al. [29]Dioscorea sulfated polysaccharideNot reported (DGGE only)UPGMA: closer to NCBacteroides thetaiotaomicron; ↓ Enterococcus, AcinetobacterNot reportedNot reportedNot reportedNot performedSulfated yam polysaccharide restored microbial balance and alleviated diarrhea via reduction in pathogens and enrichment of commensals.
HNPLi S, et al. [30]Astragalus polysaccharideShannon and Chao1 ↑NMDS: closer to NCOscillospira, Dorea; ↓ Epulopiscium, PseudomonasPropionate, butyrate, total SCFAs ↑Colon architecture normalizedInflammatory cell infiltration ↓Normal mucosal architecture restored.Astragalus polysaccharide modulated microbiota and selectively enhanced propionate and butyrate generation.
HNPXu et al. [31]Poria cocos polysaccharideRichness and diversity ↑PCoA: closer to NCParabacteroides distasonis, Akkermansia muciniphila; ↓ Salmonella, MucispirillumPredicted ↑ carbohydrate metabolism; SCFA receptors (GPR41/43) ↑ZO-1, OC-1 ↑FOXP3 mRNA ↑, NF-κB ↓Mucosal damage reversed; villus and goblet cells normalized.Poria polysaccharide restored microbial diversity and tight junctions, activated FOXP3 and SCFA receptor pathways, and suppressed NF-κB.
HNPLai et al. [32]Dictyophora polysaccharideShannon and ACE ↑PCoA/type analysis: DIPY shifted toward NCRobinsoniella, Parasutterella, Blautia; ↓ some MuribaculaceaeAcetate and total SCFAs ↑Epithelial integrity and mucus layer restoredLPS, MCP-1, TNF-α, IL-6 ↓Intact epithelium with restored goblet cells.Dictyophora polysaccharide modulated microbiota, enhanced SCFA production, repaired barrier, and reduced systemic inflammation.
HNPBie et al. [33]Sweet potato polysaccharideChao and Shannon ↑PCoA: shifted toward NC↓ Proteobacteria; ↑ Bacteroidetes, Muribaculaceae; ↓ Escherichia coli, KlebsiellaAcetate, propionate, butyrate, valerate, total SCFAs ↑Ileal villus length and crypt depth restoredIL-10 ↑; IL-1β, IL-6, TNF-α ↓Reduced inflammatory infiltration; improved ileal morphology.Sweet potato polysaccharide reversed dysbiosis, enhanced SCFA production, and regulated IL-10–mediated anti-inflammatory pathways.
HNPZhang et al. [34]Yam extractDGGE: richness/evenness ↑UPGMA/PCA: re-clustering toward NCBacteroides spp., Clostridium spp.; ↓ Enterobacter spp.Propionate, butyrate, valerate, total SCFAs ↑Cecal index normalized (indirect barrier recovery)Not assessedCecal enlargement reversed.Yam extract acted as substrate for SCFA-producing bacteria, enhancing SCFA production and gut recovery.
HNPPan et al. [35]Brown alga polysaccharide (Nemacystus)Indices restored toward NCPCoA: cluster closer to NC↓ Proteobacteria; ↑ Muribaculum, Lactobacillus; ↓ Enterobacter, ClostridioidesSCFA-producing genera recovered (not quantified)Occludin and SHIP ↑IL-1β, IL-6, p-PI3K, p-NF-κB ↓Smoother mucosa; reduced inflammatory infiltration.Brown alga polysaccharide enhanced commensals, maintained tight junctions via SHIP–PI3K/NF-κB modulation, and suppressed inflammation.
HNPLai et al. [36]Poria cocos water-insoluble polysaccharideSimpson, ACE, Shannon ↑PCoA: closer to NCnorank_f__Muribaculaceae; ↓ Staphylococcus, Acinetobacter, Escherichia–ShigellaAcetate, butyrate ↑Cecal mucosa continuity restoredSerum TNF-α, IL-6, IL-1β ↓Almost normal architecture.Water-insoluble Poria polysaccharide remodeled microbiota, increased SCFAs, reduced pro-inflammatory cytokines, and repaired barrier.
HNPLu et al. [37]Antrodia cinnamomea intracellular polysaccharideShannon ↑, Simpson ↓ (diversity restored)PCoA/NMDS: closer to NC↑ Erysipelotrichaceae, Lachnospiraceae; ↓ EnterococcusNot measuredThymus and spleen index restoredSerum IL-6, TNF-α ↓Colon structure improved.Antrodia polysaccharide reduced pathogenic Enterococcus, enhanced butyrate-producing commensals, and improved immune-organ indices.
HNPCui et al. [38]Sea cucumber (Cereus sinensis) polysaccharideShannon and Chao1 ↑PCoA/NMDS/PLS-DA: closer to NCFirmicutes/Bacteroidetes ratio normalized; ↑ Phasecolarctobacterium, BifidobacteriumAcetate and total SCFA ↑; propionate and butyrate ↑Cecal damage reduced; structure normalizedSerum IL-2, TNF-α ↓; IL-1β ↓ (high dose)Cecal inflammation and edema reduced.Sea cucumber polysaccharide enhanced SCFA-producing flora, improved SCFA output, and suppressed inflammatory cytokines.
HNPChen et al. [39]Bamboo shoot polysaccharideShannon and Chao1 ↑PCA: high-dose cluster closest to NCF/B ratio normalized; ↑ Lactobacillus; ↓ Escherichia–ShigellaAcetate, propionate, butyrate, valerate ↑ (butyrate/valerate > inulin)Thicker intestinal wall; reduced edemaNo cytokine assayMucosal edema and inflammatory infiltration reversed.Bamboo shoot polysaccharide was fermented in colon, increased SCFAs, promoted beneficial flora, and suppressed pathobionts.
HNPZeng. [40]Blueberry leaf polyphenolsDiversity and richness restoredPCoA: clusters closer to NCF/B ratio normalized; ↑ Muribaculum, Lactobacillus; ↓ Clostridium, EnterococcusAcetate, propionate, butyrate, valerate, total SCFAs ↑Occludin, claudin-1, ZO-1 ↑ (gene/protein)Serum IL-1β, TNF-α, IL-6 ↓Ileal necrosis and inflammatory infiltration repaired.Blueberry leaf polyphenols repaired barrier, suppressed MAPK signaling, modulated microbiota, and promoted SCFA production.
HNPHan et al. [41]Cistanche extract/polysaccharideSimpson and Shannon ↑PCoA: clusters closer to NC↓ Firmicutes; ↓ Clostridium_sensu_stricto_1; ↑ Blautia, LachnospiraceaeNot measuredNot specifiedNot specifiedColonic structure improved; inflammation reduced.Cistanche effects were likely mediated by gut microbiota modulation and metabolite regulation.
HNPLi et al. [42]Antrodia polysaccharides (AEPS/AIPS)Shannon and Simpson ↑PCoA/NMDS: moved away from model clusterAEPS: ↑ Lactobacillaceae; AIPS: ↑ Staphylococcaceae/StaphylococcusNot reportedNot reportedNot reportedNot analyzed in detail.Antrodia polysaccharides modulated gut microbiota; AEPS showed more favorable shift toward Lactobacillus dominance.
HNPMa et al. [43]Fresh ginger extractDiversity restored (after AAD-induced ↓)PCoA/NMDS: ginger groups closer to NCProteobacteria ↓; Escherichia–Shigella ↓; BacteroidesNot reportedMUC2, ZO-1 ↑; goblet cells restoredMPO expression ↓; colonic inflammation amelioratedEpithelial shedding and disorganized crypts reversed.Fresh ginger suppressed Escherichia–Shigella, enhanced Bacteroides, and restored barrier integrity.
HNPZhang et al. [44]Dried ginger extractACE/Shannon ↓ in model; ↑ with gingerPCoA/cluster: ginger closer to NCBacillus, Lachnoclostridium, E. coli–Shigella; ↑ LactobacillusNot measuredNot reportedNot reportedEpithelial shedding and crypt disruption improved.Dried ginger restored diversity and beneficial Lactobacillus while reducing pathogenic E. coli–Shigella.
HNPLi et al. [45]American ginseng decoctionRichness ↔; Shannon/Simpson ↑PCoA/NMDS: shifted toward NC↓ Proteobacteria; ↑ Bacteroidetes/Firmicutes; ↓ Pseudomonas, E. coli–Shigella; ↑ BacteroidesNot reportedColon structure intact (H&E)Inflammatory cell infiltration ↓Loose glands and epithelial shedding improved.American ginseng decoction improved diversity and composition and restored colon structure and metabolic pathways.
HNP-CQu et al. [46]Fermented ginseng synbioticSimpson, Ace, Chao, Shannon ↑ (restored)PCA: fermented ginseng cluster closest to NCLactobacillus, Candidatus Stoquefichus; ↓ Bacteroides, ClostridioidesNot measuredCrypt architecture normalized; infiltration ↓IL-1β, IL-6, TNF-α ↓; IL-10 ↑Colon architecture restored.Fermentation increased β-glucosidase activity, boosted bioactive ginsenosides, normalized microbiota and cytokine balance.
HNP-CZhong et al. [47]Astragalus polysaccharide + L. plantarumα-diversity ↑ (combination > single > model)PCoA: combination closest to NCLactobacillus, Allobaculum, Bifidobacterium; ↓ Bacteroides, BlautiaNot measuredOccludin, claudin-1, ZO-1, MUC-2 ↑; goblet cells ↑; DAO, D-LA, LPS ↓sIgA, IgG ↑; IL-17A, IL-4, TGF-β1 ↓Goblet cells and mucus area increased; edema ↓Astragalus synbiotic enhanced tight junctions and mucus, improved immunoglobulin profile, and promoted epithelial repair via Smad7/p-Smad3 modulation.
HNP-CTang et al. [48]Poria polysaccharide + probioticsChao and Shannon ↑ (PWP > PP > WP)PCoA/NMDS: PWP cluster closest to NC↑ Firmicutes, Bacteroidetes (Muribaculaceae); ↓ Proteobacteria (Sutterella)Not measuredMucin ↑ (histology)IgM, IgG normalized; IgA ↑; macrophages/lymphocytes ↑Mucin ↑; edema ↓; improved villus morphology.Poria-based synbiotic was associated with immune modulation and microbiota homeostasis, with more pronounced changes observed under the tested conditions.
HNP-CGuo et al. [49]B. adolescentis + β-glucan synbioticShannon and Fisher ↑ (combination > single)PCoA: combination cluster closest to NCB. uniformis; ↓ Enterococcus, Escherichia–ShigellaAcetate, propionate, isobutyrate ↑Occludin, ZO-1, Mucin-2/3 ↑; D-LA, LPS ↓IL-6, IL-17 ↓Mucosal structure improved; epithelium intact.Synbiotic increased SCFAs, repaired barrier, and modulated immunity via enrichment of B. uniformis.
HNP-CLi et al. [50]Maifan stone + L. rhamnosus GGNot reportedNot applicable (qPCR only)Bacteroides, Lactobacillus; ↓ Enterococcus, E. coliNot measuredColon epithelium arrangement restored; edema ↓TLR4, NF-κB ↓ in colonEdema reduced; epithelial structure normalized.Maifan stone carrier enhanced LGG tolerance, improved microbiota, and down-regulated TLR4/NF-κB signaling.
HNP-CShen et al. [51]Chitosan oligosaccharide + probioticsShannon ↑; Chao1 trend toward recoveryPCoA: synbiotic group closer to baseline (day 0)↑ Firmicutes, Acidobacteriota; ↓ Desulfobacterota; shifts in Megamonas, Lachnospiraceae, MuribaculaceaeNot measuredZO-1, occludin, claudin-1 ↑; villus height/crypt depth ↑TNF-α, IL-1β ↓; IgA, IgG ↑Edema ↓; villus height and crypt depth improved.Chitosan oligosaccharide acted as prebiotic carrier; synbiotic restored tight junctions and reduced inflammation in a canine model.
HNP = Herbal/Natural Product; HNP-C = Herbal/Natural Product Combination; ↑ = increase; ↓ = decrease.
Table 3. Comparative Effects Between HNP Monotherapy and HNP-C Combination Therapies.
Table 3. Comparative Effects Between HNP Monotherapy and HNP-C Combination Therapies.
OutcomeHNP Monotherapy (n = 21)HNP-C Combination (n = 6)Comparative Interpretation
α-DiversityReported in 20/21 studies; 1 study used DGGE only (no α-diversity indices). All studies reported ↑ Shannon/Chao1/ACE.Reported in 5/6 studies; one study [45,50] did not assess α-diversity. All reporting studies showed increases.Both strong; HNP-C slightly more rapid and consistent in studies with direct comparators.
β-DiversityReported in 18/21 studies (PCoA/NMDS). All 18 showed a shift toward Control; 3 did not report β-diversity.Reported in 5/6 studies; Li et al. [45,50] used qPCR only. All reporting studies showed convergence toward NC.HNP-C tends to show closer clustering toward the Control group in studies reporting β-diversity.
Key taxa shiftsReported in 21/21 studies. Common findings: ↑ Lactobacillus, Bifidobacterium, Muribaculaceae; ↓ Escherichia–Shigella.Reported in 6/6 studies. Persistence of administered probiotic strains.Similar direction overall; HNP-C yields more stable probiotic colonization.
SCFAsReported in 17/21 studies. All reported ↑ acetate/propionate/butyrate.Quantitatively reported in 1/6 studies [49]. Other 5 did not measure SCFAs directly; some reported ↑ SCFA-producing taxa.HNP monotherapy provides stronger direct evidence for SCFA production (17/21 studies). HNP-C efficacy is currently limited by reporting frequency (1/6 studies) but supported indirectly.
Barrier markers (ZO-1, Occludin, Claudin-1, MUC2, SIgA)Reported in 16/21 studies; improved TJ proteins and mucosal integrity.Reported in 4/6 studies; increased TJ proteins and reduced leakage markers (D-LA, LPS).HNP-C showed improvements across multiple barrier-related markers when reported.
Pro-inflammatory cytokines (TNF-α, IL-1β, IL-6)Reported in 15/21 studies; consistent decreases.Reported in 6/6 studies; also ↓ IL-17A and ↑ IL-10.HNP-C showed modulation across multiple cytokine markers.
Immune markers (IgA, IgG, IgM, FOXP3)Reported in 11/21 studies; some ↑ IgA and ↑ FOXP3.Reported in 5/6 studies; all showed ↑ IgA/IgG/IgM.HNP-C showed consistent improvements in immune-related outcomes.
Diarrhea-related phenotypic outcomesReported in 18/21 studies; improved diarrhea severity and weight recovery.Reported in 6/6 studies; all showed. phenotypic improvementBoth effective; HNP-C shows more complete and consistent reporting.
HistopathologyReported in 17/21 studies; mucosal repair, ↑ goblet cells, ↓ edema.Reported in 6/6 studies; villus height ↑, crypt integrity restored.Comparable effects; HNP-C shows uniformly positive histology.
Overall consistency and strengthMechanisms vary by herb/extract; reporting frequency variable.Highly coherent across barrier, immunity, taxa, and histology.HNP-C consistently showed recovery across multiple domains.
The two direct comparator studies suggest potentially additive benefits across microbial, barrier, and immune endpoints.
↑ = increase; ↓ = decrease.

3.4. Functional Recovery: SCFA Metabolism and Barrier Integrity

SCFA metabolism and epithelial barrier function were evaluated with varying depth across studies (Table 3). Among HNP interventions, 17 of the 21 studies quantified SCFAs [25,27,29,31,33,35,37,38,39,41,42,43,44,45] and consistently reported increases in total SCFAs or specific metabolites such as acetate, propionate, and butyrate. These biochemical improvements generally corresponded with the restoration of SCFA-producing taxa, including Lachnospiraceae and Ruminococcaceae. In contrast, only one of the six HNP-C studies directly quantified SCFAs [49]. The remaining studies inferred metabolic improvement through increased abundance of SCFA-producing genera without direct metabolite measurements [46,47,48,50,51]. Consequently, current evidence for SCFA enhancement is substantially stronger for HNP monotherapy than for HNP-C combinations. In contrast, SCFA-related findings in HNP-C studies were largely inferred from shifts in SCFA-producing taxa rather than direct metabolite quantification.
Barrier-related markers were more consistently assessed. Eighteen HNP studies [26,27,28,31,32,33,35,36,37,38,39,40,41,42,43,44,45] and four HNP-C studies [47,48,49,50,51] demonstrated improvements in tight junction proteins (ZO-1, occludin, claudin-1), mucin production (MUC2), permeability-related biomarkers (DAO, D-LA, LPS), and mucosal histology. Notably, only two HNP-C studies incorporated comparator groups that included herbal-only, probiotic-only, and combination arms [47,48], and both more pronounced recovery of epithelial structure and permeability with combination therapy. The remaining HNP-C studies [46,49,50,51] showed similar benefits but lacked designs that allow evaluation of additive effects. Overall, both HNP and HNP-C interventions were associated with functional recovery from antibiotic-induced intestinal injury; however, mechanistic evidence supporting additive benefits in HNP-C interventions remains limited to two well-controlled studies.

3.5. Immune and Inflammatory Responses

Fifteen HNP studies assessed immune or inflammatory markers [25,27,28,29,32,33,34,36,37,38,39,41,42,44,45] and generally reported reductions in pro-inflammatory cytokines (TNF-α, IL-1β, IL-6), with several studies also observing increases in IL-10 or SIgA, suggesting partial immune normalization concurrent with microbiota recovery. All six HNP-C studies evaluated immune outcomes [46,47,48,49,50,51] and consistently showed reduced inflammatory cytokine expression and/or enhanced immunoglobulin levels. Among these, two studies employed direct comparator designs [47,48] and reported more pronounced cytokine reduction and humoral immune changes in the combination groups. The remaining studies [46,49,50,51] showed similar trends but lacked the methodological elements necessary to confirm synergistic effects.
Taken together, both intervention categories appeared to mitigated antibiotic-induced inflammation. Although HNP-C regimens may provide potentially complementary immunomodulatory effects, current evidence supporting this hypothesis is restricted to two rigorously controlled comparator studies.

3.6. Histopathological Recovery

Histological evaluation was conducted in 23 studies, including 17 HNP studies [26,28,31,33,35,36,37,38,39,40,42,43,44,45] and all six HNP-C studies [46,47,48,49,50,51]. Both intervention types improved villus and crypt architecture, reduced mucosal edema, restored goblet cell density, and decreased inflammatory cell infiltration. In most studies, histopathological outcomes were evaluated qualitatively based on morphological criteria, as standardized scoring systems were not consistently applied across the included studies. Among HNP-C trials, two studies with direct comparator arms [47,48] showed more pronounced mucosal recovery in the combination groups than in the respective monotherapy groups. The remaining HNP-C studies [46,49,50,51] reported similar improvements but lacked designs that permitted assessment of synergistic effects. Overall, histological repair was consistently observed across all included studies and generally paralleled improvements in microbiota composition, metabolic activity, and immune function.

3.7. Diarrhea-Related Phenotypic Outcomes

Clinical symptom assessments were reported in 18 HNP studies [25,26,27,28,29,31,32,33,35,36,37,38,39,40,41,42,43,44,45] and all HNP-C studies [46,47,48,49,50,51]. Improvements included reduced diarrhea severity, enhanced stool consistency, and accelerated weight recovery, changes that likely reflect ongoing microbial and mucosal restoration. Only two HNP-C studies [47,48] incorporated suitable comparator groups, and both showed faster or more pronounced symptomatic recovery in the combination groups than in monotherapy. Other HNP-C studies showed comparable diarrhea-related phenotypic benefits but lacked design features necessary to evaluate synergy. Overall, phenotypic improvements were consistently observed across both intervention categories, with evidence of additive benefit from combined HNP and probiotic administration emerging only in studies with appropriate comparative frameworks.

3.8. Comparative Effects Between HNP and HNP-C Using Overlapping Herbal Sources

Two key herbal ingredients, Poria (Poria/Wolfiporia cocos) and Astragalus (Astragalus spp.), were included in both HNP monotherapy and HNP-C interventions. Despite differences in extraction and formulation, these overlapping components provided the only basis for evaluating whether the addition of probiotics yields benefits beyond monotherapy. Notably, most HNP monotherapy studies used two-group designs (monotherapy vs. control), whereas the HNP-C studies employing these herbs used three-group designs (HNP monotherapy vs. probiotic monotherapy vs. combination therapy), allowing clearer assessment of potential synergistic effects.

3.8.1. Poria (Wolfiporia cocos) Group (Table 4)

A series of Poria-based HNP monotherapy studies [32,36] reported consistent improvements in epithelial barrier integrity, including restored ZO-1 and occludin expression, reduced NF-κB activation, and recovery of mucosal architecture. In contrast, the Poria-containing HNP-C study [48] employed a three-arm design that isolated the effects of each therapeutic component. The combination of Poria polysaccharides with probiotics produced more comprehensive biological benefits than either component alone, including enhanced mucus secretion and goblet cell density, greater recovery of tight-junction structure, increased humoral immune responses (IgA/IgG/IgM), and a more sustained re-establishment of gut microbial community structure over time. These findings suggest that although Poria monotherapy consistently supports epithelial repair, co-administration with probiotics may provide complementary improvements across microbial, barrier, and immune domains, indicating potential additive therapeutic advantages under the tested conditions.
Table 4. Poria Group (Wolfiporia cocos).
Table 4. Poria Group (Wolfiporia cocos).
ItemHNP (PCP, PCY)HNP-C (WP + Probiotics)
Barrier↑ ZO-1, Occludin; epithelial repair↑ mucin, ↑ goblet cells; ↓ leakage markers
Immune↓ NF-κB; GPR41/43 ↑↑ IgA/IgG/IgM; ↑ macrophage/lymphocyte activity
MicrobiotaRestored Firmicutes/Bacteroidetes; ↓ pathogensRelatively stable community structure; sustained shifts
HistologyEdema ↓; villus repairMore pronounced epithelial regeneration
MechanismSCFA receptor (GPR41/43) activationCombined barrier and immune modulation
This table presents descriptive comparisons based on individual studies and is intended for hypothesis-generating purposes. ↑ = increase; ↓ = decrease.

3.8.2. Astragalus (Astragalus spp.) Group (Table 5)

The efficacy of Astragalus-based HNP monotherapy [29] was evaluated using standard comparisons between the control group, AAD model group, and herbal monotherapy group. In this study, Astragalus polysaccharide administration supported recovery of microbial diversity, selectively enriched SCFA-producing taxa, promoted villus restoration, and increased levels of propionate and butyrate. In contrast, the Astragalus-containing HNP-C intervention [47] used a three-group design comparing herbal therapy, probiotic therapy, and combined treatment, enabling clearer differentiation of each component’s effects. In this design, the synbiotic combination produced more coordinated improvements than either monotherapy, including greater upregulation of tight-junction and mucus-related proteins (Occludin, Claudin-1, ZO-1, MUC2), enhanced humoral immune responses (IgA/IgM/IgG), and larger reductions in permeability-related markers (DAO, D-lactic acid, LPS). Additionally, modulation of Smad7/p-Smad3 signaling offered further mechanistic support for epithelial regenerative activity. Overall, while Astragalus monotherapy primarily improved microbial composition and SCFA-associated recovery, combining Astragalus with probiotics resulted in more comprehensive and coordinated improvements across microbial, barrier, and immune domains, suggesting a potential additive advantage under the tested conditions.
Table 5. Astragalus Group.
Table 5. Astragalus Group.
ItemHNP (WAP)HNP-C (APS + L. plantarum)
SCFAs↑ propionate, ↑ butyrateNot directly measured
BarrierVillus restoration↑ Occludin/Claudin-1/ZO-1/MUC-2; ↓ DAO/D-LA/LPS
Immune↓ inflammation↑ IgA/IgM/IgG
MechanismMicrobiota + SCFA effects↑ Smad7; ↓ p-Smad3; pathway-level modulation
This table presents descriptive comparisons based on individual studies and is intended for hypothesis-generating purposes. ↑ = increase; ↓ = decrease.

3.8.3. Integrated Summary

Across both Poria and Astragalus categories, herbal monotherapies improved dysbiosis, barrier integrity, and inflammatory responses, while combination therapies produced more comprehensive and coordinated recovery patterns across microbial, immunological, and structural outcomes. These patterns were most evident in the three-arm designs used in the combination studies, which showed that dual-component interventions were associated with more extensive improvements than herbal-only or probiotic-only treatments under the tested conditions. Although the number of overlapping interventions is limited, the available evidence suggests that combining herbal materials with probiotic strains may support broader and more stable recovery profiles in antibiotic-associated diarrhea.

3.9. Risk of Bias and Quality of Reporting

The methodological quality of the 27 included animal studies was assessed using the SYRCLE risk-of-bias tool (Figure 2). Reporting of key study design safeguards was generally insufficient across both HNP (n = 21) and HNP-C (n = 6) interventions. Although many studies stated that animals were randomly assigned to groups, none described the method of sequence generation, resulting in unclear risk for selection bias. Allocation concealment was also not reported in any study and was therefore judged as high or unclear risk. Blinding of caregivers, investigators, or outcome assessors was rarely described, contributing to predominantly unclear ratings for performance and detection bias. In contrast, selective reporting and outcome completeness were generally adequate, and most studies were rated as low risk in these domains. Overall, the frequent “unclear” assessments stemmed from insufficient methodological reporting rather than evidence of systematic flaws. Future preclinical studies should clearly detail randomization methods, allocation concealment, and blinding procedures to strengthen the robustness and interpretability of evidence.

4. Discussion

Understanding the therapeutic landscape of AAD requires consideration not only of the immediate loss of microbial diversity but also of the broader functional disruptions caused by antibiotic exposure [25,26,27,28,29]. Antibiotics suppress SCFA metabolism, compromise epithelial tight junction integrity, reduce mucus secretion, and disturb mucosal immune regulation, alterations that collectively shape the trajectory of post-antibiotic recovery [31,32,33,34,35,41,42,43,44,45]. These multidimensional disturbances also help explain why conventional strategies such as probiotics often yield incomplete benefits, particularly when microbial colonization remains hindered under antibiotic pressure [46,47,48,49].
Within this context, HNPs have emerged as promising multi-target interventions. Many HNP compounds influence microbial pathways such as enriching beneficial commensals, suppressing opportunistic bacteria, and partially normalizing microbial community structure while also modulating host processes related to epithelial repair, barrier reinforcement, and immune regulation [32,36,37,38,41,42,43,44,45]. Earlier preclinical studies showed these benefits separately, such as histological improvement or reduced inflammation, but were unable to link these outcomes to microbiota restoration due to the lack of sequencing data [17,18,19].
Accordingly, HNP–C strategies have gained traction as an integrated therapeutic approach. This strategy aligns with principles of rational synbiotic design, aiming to address the metabolic and colonization limitations of single-agent therapy by using HNPs as fermentable substrates that promote probiotic colonization and functional output [52,53].
In this review, several integrated patterns emerged. Most HNP monotherapy studies that included microbial endpoints demonstrated restoration of diversity and community structure alongside improvements in barrier and immune markers [25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45], suggesting that ecological and host-functional recovery progress concurrently [3,4,54,55]. Seventeen of the 21 monotherapy studies measured SCFAs and reported increases in acetate, propionate, or butyrate [25,27,29,31,33,35,37,38,39,41,42,43,44,45]. Although the analytical platforms varied and quantitative synthesis was not feasible, the consistency of these findings supports the interpretation that HNP-driven metabolic recovery is a downstream outcome of microbiota restoration and is essential for epithelial barrier function [56,57].

4.1. Effects of HNP Monotherapy

HNP monotherapies demonstrated coherent recovery across ecological and host-functional domains. Twenty of the 21 studies reported increases in α-diversity [25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45], and 18 studies showed β-diversity shifts toward healthy control clustering [25,26,27,28,29,31,32,33,35,36,37,38,39,40,41,42,43,44,45]. Beneficial commensals, including Lactobacillus, Bifidobacterium, Muribaculaceae, Lachnospiraceae, and Ruminococcaceae expanded, whereas Proteobacteria-dominant taxa, such as Escherichia–Shigella, diminished, indicating that microbial restoration and mucosal recovery likely progress in parallel [3,4,54,55]. SCFA profiles also improved consistently across studies that quantified SCFAs [25,27,29,31,33,35,37,38,39,41,42,43,44,45], aligning with the regrowth of SCFA-producing taxa. These ecological gains coincided with increased tight-junction expression [26,27,28,31,32,33,35,36,37,38,39,40,41,42,43,44,45], reduced permeability markers [26,27,28,31,32,33,35,36,37,38,39,40,41,42,43,44,45], and decreased pro-inflammatory cytokines [25,27,28,29,32,33,34,36,37,38,39,41,42,44,45]. Collectively, these patterns suggest that HNPs exert primary microbiota-mediated effects, with secondary improvements emerging through enhanced barrier integrity and immune regulation [56,57].

4.2. Effects of HNP–Probiotic Combinations (HNP-C)

Although fewer in number, HNP-C studies demonstrated more comprehensive and coordinated improvements across microbial, epithelial, and immune endpoints [46,47,48,49,50,51]. This integrated recovery aligns with principles of rational synbiotic design, wherein functional complementarity between the herbal substrate and probiotic strain facilitates coordinated biological responses that may exceed simple additive effects [52,53]. All six studies reported improved α-diversity and β-diversity trajectories moving toward healthy control clustering [46,47,48,49,50,51]. Combination therapies consistently increased tight-junction markers, reduced permeability indices, and produced broader immunologic modulation, including elevations in IgA, IgM, and IgG [46,47,48,49,50,51]. The two trials using three-arm designs directly comparing HNP-only, probiotic-only, and HNP-C groups offered particularly valuable insights [47,48]. In both studies, the combined intervention showed greater improvements across ecological, barrier, and immunologic outcomes compared with monotherapy, suggesting potential additive or context-dependent synergistic interactions. However, because only one study quantified SCFAs directly [49] and comparator designs remain limited, existing evidence is insufficient to conclude generalizable superiority of combination therapies.

4.3. Probiotics vs. HNP vs. HNP–C: Comparative Mechanistic Insights

The probiotic comparator groups primarily used Lactobacillus and Bifidobacterium species or representative strains such as Lactiplantibacillus plantarum ELF051 and Bifidobacterium adolescentis. Across the four studies that included probiotic-only arms [47,48,49,50], these strains consistently improved selected immune parameters, such as increased SIgA or reduced IL-6 and TNF-αand produced modest enhancement of mucin expression. However, consistent with their metabolic profiles and the strain-dependent nature of SCFA production, probiotic-only interventions demonstrated limited restoration of bulk SCFA concentrations, α-diversity indices, and major commensal families disrupted by antibiotics (e.g., Lachnospiraceae, Ruminococcaceae, Muribaculaceae) [47,48,49,50,58].
In contrast, HNP monotherapies in the same comparator studies promoted the recovery of these commensal families by supplying selectively utilizable polysaccharides and complex carbohydrates that serve as fermentation substrates [59]. These microbiota-dependent effects were often accompanied by direct host-targeted actions, including reinforcement of tight-junction structure and modulation of NF-κB–associated inflammatory pathways, which help explain why HNP monotherapy frequently matched or exceeded probiotic-only interventions in restoring epithelial integrity and metabolic function.
The complementary strengths of both components were most evident in the three-arm comparator designs. Combination groups showed closer β-diversity clustering toward healthy controls, greater upregulation of epithelial proteins (ZO-1, Occludin, MUC2), and more extensive immunological modulation than either monotherapy. These outcomes are consistent with a mechanistic model in which HNPs provide structural and metabolic scaffolding that supports microbial viability, while probiotics accelerate recolonization and mucosal immune coordination, resulting in more coordinated ecosystem recovery [52,53,60].
Taken together, HNP-C therapies appear to leverage potentially synergistic or additive interactions, pairing the ecological rebuilding capacity of HNPs with the targeted recolonization and immune-signaling strengths of probiotics to achieve more comprehensive and coordinated recovery profiles than either component alone under comparator-controlled conditions.

4.4. Comparative Insights from Overlapping Herbal Sources

Parallel evaluation of herbs used in both monotherapy and HNP-C formulations, such as Poria and Astragalus, offered additional mechanistic insight. Poria monotherapy improved microbial diversity, strengthened tight-junction structure, and reduced NF-κB activation, whereas the corresponding combination enhanced mucin secretion, goblet cell density, and immunoglobulin levels. Similarly, Astragalus monotherapy increased propionate and butyrate levels and supported villus restoration, while the combination further reinforced tight-junction expression, reduced permeability, and modulated Smad7/p-Smad3 signaling. Although the number of these paired comparisons was limited and based on study-level observations rather than direct head-to-head designs, these findings suggest that overlapping HNP substrates and probiotic components may exert complementary effects on microbial, epithelial, and immune-related outcomes under the tested conditions.

4.5. Future Directions

Based on the patterns identified, future research should address several strategic priorities across both preclinical and clinical domains. In the preclinical setting, methodological rigor is essential, including the adoption of three-arm comparator designs (HNP vs. probiotic vs. HNP-C) to robustly evaluate additive and interaction effects. Methodological standardization is also critical and should include harmonized antibiotic-induction protocols, aligned microbiome sampling time points, consistent α-/β-diversity metrics, and a comprehensive assessment of SCFAs, tight-junction markers, and immune endpoints. Beyond standardization, greater mechanistic depth will require the integration of multi-omics approaches (e.g., metabolomics, transcriptomics) and the identification of strain–substrate –matched synbiotic pairs based on fermentation compatibility. For clinical translation, priorities include incorporating microbiome endpoints (diversity, SCFAs, barrier biomarkers) and implementing time-aligned sampling during and after antibiotic therapy. These foundational data will support rational synbiotic trial design using mechanistically complementary herbal–probiotic combinations in Phase II RCTs comparing HNP, standard care, and synbiotic formulations.

4.6. Overall Synthesis and Conclusions

This review provides an integrated evaluation of microbiota restoration and host-functional outcomes, addressing the multidimensional nature of recovery in antibiotic-associated diarrhea. By synthesizing ecological indices, SCFA metabolism, barrier integrity, immune responses, and histopathological changes, these findings offer a more comprehensive mechanistic perspective than previous work that assessed only isolated endpoints.
However, certain methodological limitations restrict translational certainty. Heterogeneity across antibiotic protocols, animal strains, intervention durations, and microbiome analytic methods, together with incomplete reporting of randomization, allocation concealment, and blinding, resulted in predominantly unclear or high risks of bias. Accordingly, comparative and mechanistic interpretations—particularly regarding the relative advantages of HNP–C strategies and inferred signaling pathways—should be regarded as hypothesis-generating rather than confirmatory. Furthermore, in the absence of functional validation (e.g., pathway inhibition), observed changes in signaling markers should be regarded as associative rather than causal and interpreted with caution. Finally, only one HNP–C study directly quantified SCFAs, limiting conclusions regarding potential metabolic benefits, which remain largely inferred from microbiota compositional shifts rather than from directly quantified outcomes.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph19010064/s1, Table S1: PRISMA checklist; Table S2: Full database-specific search strategies for PubMed, EMBASE, Web of Science/Scopus, and CNKI.

Author Contributions

Conceptualization, Y.-K.C.; methodology, Y.-K.C. and J.H.H.; validation, formal analysis, investigation, resources, and data curation, Y.-K.C. and J.H.H.; writing—original draft preparation, J.H.H.; writing—review and editing, Y.-K.C. and J.H.H.; visualization, J.H.H.; supervision, Y.-K.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (No. NRF-2022R1A2C1013518). The funders had and will not have a role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AADAntibiotic-associated diarrhea
SCFA/SCFAsShort-chain fatty acid/short-chain fatty acids
HNPHerbal/Natural Product
HNP-CHerbal/Natural Product Combination
DGGEDenaturing Gradient Gel Electrophoresis
PCoAPrincipal Coordinates Analysis
NMDSNon-metric Multidimensional Scaling
qPCRQuantitative Polymerase Chain Reaction
GC–MSGas Chromatography–Mass Spectrometry
MUC2Mucin 2
ZO-1Zonula Occludens-1
OC-1/OccludinOccludin (tight-junction protein)
TJTight Junction
IgA/IgG/IgMImmunoglobulin A/G/M
LPSLipopolysaccharide
DAODiamine Oxidase
FOXP3Forkhead Box P3
IL-1β/IL-6/IL-10/IL-17AInterleukin-1β/Interleukin-6/Interleukin-10/Interleukin-17A
TNF-αTumor Necrosis Factor-α
MAPKMitogen-Activated Protein Kinase
NF-κBNuclear Factor kappa-B
F/B ratioFirmicutes-to-Bacteroidetes Ratio
PCPositive Control
NCNormal Control
L/M/HLow/Medium/High dose
SIgASecretory Immunoglobulin A
Smad7/p-Smad3Smad7/phosphorylated Smad3
CFUColony Forming Unit

References

  1. McFarland, L.V. Antibiotic-associated diarrhea: Epidemiology, trends and treatment. Future Microbiol. 2008, 3, 563–578. [Google Scholar] [CrossRef]
  2. Lee, E.L.; Sidhick, S.T.; Maharajan, M.K.; Shanmugham, S.; Ingle, P.V.; Kumar, S.; Ching, S.M.; Lee, Y.Y.; Veettil, S.K. Probiotics for the prevention of antibiotic-associated diarrhea—An umbrella review of meta-analyses of randomized controlled trials. Eur. J. Clin. Exp. Med. 2025, 23, 237–244. [Google Scholar] [CrossRef]
  3. Patangia, D.V.; Anthony Ryan, C.; Dempsey, E.; Paul Ross, R.; Stanton, C. Impact of antibiotics on the human microbiome and consequences for host health. MicrobiologyOpen 2022, 11, e1260. [Google Scholar] [CrossRef] [PubMed]
  4. Kelly, S.A.; Nzakizwanayo, J.; Rodgers, A.M.; Zhao, L.; Weiser, R.; Tekko, I.A.; McCarthy, H.O.; Ingram, R.J.; Jones, B.V.; Donnelly, R.F.; et al. Antibiotic therapy and the gut microbiome: Investigating the effect of delivery route on gut pathogens. ACS Infect. Dis. 2021, 7, 1283–1296. [Google Scholar] [CrossRef]
  5. Schubert, A.M.; Sinani, H.; Schloss, P.D. Antibiotic-induced alterations of the murine gut microbiota and subsequent effects on colonization resistance against Clostridium difficile. mBio 2015, 6, e00974. [Google Scholar] [CrossRef]
  6. Shah, T.; Baloch, Z.; Shah, Z.; Cui, X.; Xia, X. The intestinal microbiota: Impacts of antibiotics therapy, colonization resistance, and diseases. Int. J. Mol. Sci. 2021, 22, 6597. [Google Scholar] [CrossRef]
  7. Zeng, S.Y.; Liu, Y.F.; Zeng, Z.L.; Zhao, Z.B.; Yan, X.L.; Zheng, J.; Chen, W.H.; Wang, Z.X.; Xie, H.; Liu, J.H. Antibiotic-induced gut microbiota disruption promotes vascular calcification by reducing short-chain fatty acid acetate. Mol. Med. 2024, 30, 130. [Google Scholar] [CrossRef] [PubMed]
  8. Sekirov, I.; Tam, N.M.; Jogova, M.; Robertson, M.L.; Li, Y.; Lupp, C.; Finlay, B.B. Antibiotic-induced perturbations of the intestinal microbiota alter host susceptibility to enteric infection. Infect. Immun. 2008, 76, 4726–4736. [Google Scholar] [CrossRef]
  9. Modi, S.R.; Collins, J.J.; Relman, D.A. Antibiotics and the gut microbiota. J. Clin. Investig. 2014, 124, 4212–4218. [Google Scholar] [CrossRef] [PubMed]
  10. Moya, A.; Ferrer, M. Functional redundancy-induced stability of gut microbiota subjected to disturbance. Trends Microbiol. 2016, 24, 402–413. [Google Scholar] [CrossRef]
  11. Azad, M.A.K.; Sarker, M.; Li, T.; Yin, J. Probiotic species in the modulation of gut microbiota: An overview. BioMed Res. Int. 2018, 2018, 9478630. [Google Scholar] [CrossRef] [PubMed]
  12. Markowiak, P.; Śliżewska, K. Effects of probiotics, prebiotics, and Synbiotics on human health. Nutrients 2017, 9, 1021. [Google Scholar] [CrossRef] [PubMed]
  13. Tang, M.; Cheng, L.; Liu, Y.; Wu, Z.; Zhang, X.; Luo, S. Plant polysaccharides modulate immune function via the gut microbiome and may have potential in COVID-19 therapy. Molecules 2022, 27, 2773. [Google Scholar] [CrossRef]
  14. Luo, T.; Che, Q.; Guo, Z.; Song, T.; Zhao, J.; Xu, D. Modulatory effects of traditional Chinese medicines on gut microbiota and the microbiota-gut-x axis. Front. Pharmacol. 2024, 15, 1442854. [Google Scholar] [CrossRef] [PubMed]
  15. Pedrosa, L.F.; Fabi, J.P. Polysaccharides from medicinal plants: Bridging ancestral knowledge with contemporary science. Plants 2024, 13, 1721. [Google Scholar] [CrossRef]
  16. Zhou, Y.; Zhang, D.; Cheng, H.; Wu, J.; Liu, J.; Feng, W.; Peng, C. Repairing gut barrier by traditional Chinese medicine: Roles of gut microbiota. Front. Cell Infect. Microbiol. 2024, 14, 1389925. [Google Scholar] [CrossRef]
  17. Kim, S.J.; Shin, M.S.; Choi, Y.K. Ameliorative effects of Zingiber officinale Rosc on antibiotic-associated diarrhea and improvement in intestinal function. Molecules 2024, 29, 732. [Google Scholar] [CrossRef]
  18. Yang, L.; Bajinka, O.; Jarju, P.O.; Tan, Y.; Taal, A.M.; Ozdemir, G. The varying effects of antibiotics on gut microbiota. AMB Express 2021, 11, 116. [Google Scholar] [CrossRef]
  19. Chen, Y.; Meng, X.; Zheng, H.; Gu, Y.; Zhu, W.; Wang, S.; Lin, J.; Li, T.; Liao, M.; Li, Y.; et al. Deciphering the pharmacological mechanisms of Shenlingbaizhu formula in antibiotic-associated diarrhea treatment: Network pharmacological analysis and experimental validation. J. Ethnopharmacol. 2024, 329, 118129. [Google Scholar] [CrossRef]
  20. Fu, S.; Sun, L.; Chen, Y.; Zhou, Q.; Yuan, L.; Li, Z.; Chen, Q. Effect of traditional Chinese medicine on treating antibiotic-associated diarrhea in children: A systematic review and meta-analysis. Evid. Based Complement. Alternat. Med. 2022, 2022, 6108772. [Google Scholar] [CrossRef]
  21. Xue, H.; Mei, C.F.; Wang, F.Y.; Tang, X.D. Relationship among Chinese herb polysaccharide (CHP), gut microbiota, and chronic diarrhea and impact of CHP on chronic diarrhea. Food Sci. Nutr. 2023, 11, 5837–5855. [Google Scholar] [CrossRef]
  22. Gao, L.-D.; Wang, S.; Zhang, L.; Yuan, W.-L.; Jin, X.-Y.; Fan, Y.-H.; Wang, B. Efficacy of Shenlingbaizhusan for antibiotic-associated diarrhea: A systematic review and meta-analysis. J. Herb. Med. 2023, 40, 100676. [Google Scholar] [CrossRef]
  23. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
  24. Hooijmans, C.R.; Rovers, M.M.; de Vries, R.B.M.; Leenaars, M.; Ritskes-Hoitinga, M.; Langendam, M.W. SYRCLE’s risk of bias tool for animal studies. BMC Med. Res. Methodol. 2014, 14, 43. [Google Scholar] [CrossRef] [PubMed]
  25. Xu, J.; Chen, Y.; Zhan, M.; Liu, S.; Zhang, H.; Wu, Q.; Xiao, J.; Cao, Y.; Xiao, H.; Song, M. Investigating the interaction between tangeretin metabolism and amelioration of gut microbiota disorders using dextran sulfate sodium-induced colitis and antibiotic-associated diarrhea models. Curr. Res. Food Sci. 2025, 10, 101049. [Google Scholar] [CrossRef]
  26. Ren, D.D.; Shao, Z.; Liu, S.; Wang, Z.; Zhao, L.J.; Xia, Y.S.; Li, S.; Sun, Y. Ameliorative Effect of panax quinquefolius polysaccharides on Antibiotic-associated diarrhea Induced by clindamycin phosphate. Sci. Technol. Food Ind. 2021, 42, 354–361. [Google Scholar]
  27. Min, S.J.; Kim, H.; Yambe, N.; Shin, M.S. Ameliorative effects of Korean-red-ginseng-derived polysaccharide on antibiotic-associated diarrhea. Polymers 2024, 16, 231. [Google Scholar] [CrossRef] [PubMed]
  28. Qi, Y.L.; Li, S.S.; Qu, D.; Chen, L.X.; Gong, R.Z.; Gao, R.Z.; Sun, Y.; Li, Y.X. Effects of ginseng neutral polysaccharide on gut microbiota in antibiotic-associated diarrhea mice. China J. Chin. Mater. Med. 2019, 44, 811–818. [Google Scholar]
  29. Pan, H.Y.; Zhang, C.Q.; Zhang, X.Q.; Zeng, H.; Dong, C.H.; Chen, X.; Ding, K. A galacturonan from Dioscorea opposita Thunb. regulates fecal and impairs IL-1 and IL-6 expression in diarrhea mice. Glycoconj. J. 2022, 39, 131–141. [Google Scholar] [CrossRef]
  30. Li, S.; Qi, Y.; Ren, D.; Qu, D.; Sun, Y. The structure features and improving effects of polysaccharide from Astragalus membranaceus on antibiotic-associated diarrhea. Antibiotics 2019, 9, 8. [Google Scholar] [CrossRef]
  31. Xu, H.; Wang, S.; Jiang, Y.; Wu, J.; Chen, L.; Ding, Y.; Zhou, Y.; Deng, L.; Chen, X. Poria cocos polysaccharide ameliorated antibiotic-associated diarrhea in mice via regulating the homeostasis of the gut microbiota and intestinal mucosal barrier. Int. J. Mol. Sci. 2023, 24, 1423. [Google Scholar] [CrossRef] [PubMed]
  32. Lai, Y.; Zhang, Q.; Xu, M.; Guo, X.; Zhou, Q.; Liu, Q.; Deng, H.; Song, C. Restoration of antibiotic associated diarrhea induced gut microbiota disorder by using Dictyophora indusiata water-insoluble polysaccharides in C57BL/6J mice. Front. Nutr. 2025, 12, 1607365. [Google Scholar] [CrossRef] [PubMed]
  33. Bie, N.; Duan, S.; Meng, M.; Guo, M.; Wang, C. Regulatory effect of non-starch polysaccharides from purple sweet potato on intestinal microbiota of mice with antibiotic-associated diarrhea. Food Funct. 2021, 12, 5563–5575. [Google Scholar] [CrossRef]
  34. Zhang, N.; Liang, T.S.; Jin, Q.; Shen, C.; Zhang, Y.F.; Jing, P. Chinese yam (Dioscorea opposita Thunb.) alleviates antibiotic-associated diarrhea, modifies intestinal microbiota, and increases the level of short-chain fatty acids in mice. Food Res. Int. 2019, 122, 191–198. [Google Scholar] [CrossRef]
  35. Pan, H.Y.; Chen, X.; Wang, P.P.; Peng, J.F.; Li, J.D.; Ding, K. Effects of Nemacystus decipiens polysaccharide on mice with antibiotic associated diarrhea and colon inflammation. Food Funct. 2023, 14, 1627–1635. [Google Scholar] [CrossRef]
  36. Lai, Y.; Deng, H.; Fang, Q.; Ma, L.; Lei, H.; Guo, X.; Chen, Y.; Song, C. Water-insoluble polysaccharide extracted from Poria cocos alleviates antibiotic-associated diarrhea based on regulating the gut microbiota in mice. Foods 2023, 12, 3080. [Google Scholar] [CrossRef]
  37. Lu, C.L.; Li, H.X.; Zhu, X.Y.; Luo, Z.S.; Rao, S.Q.; Yang, Z.Q. Regulatory effect of intracellular polysaccharides from Antrodia cinnamomea on the intestinal microbiota of mice with antibiotic-associated diarrhea. Qual. Assur. Saf. Crops Foods 2022, 14, 124–134. [Google Scholar] [CrossRef]
  38. Cui, M.; Wang, Y.; Elango, J.; Wu, J.; Liu, K.; Jin, Y. Cereus sinensis polysaccharide alleviates antibiotic-associated diarrhea based on modulating the gut microbiota in C57BL/6 mice. Front. Nutr. 2021, 8, 751992. [Google Scholar] [CrossRef]
  39. Chen, C.H.; Guan, X.F.; Liu, X.Y.; Zhuang, W.J.; Xiao, Y.Q.; Zheng, Y.F.; Wang, Q. Polysaccharides from Bamboo Shoot (Leleba oldhami Nakal) Byproducts Alleviate Antibiotic-Associated diarrhea in Mice through Their Interactions with Gut microbiota. Foods 2022, 11, 2647. [Google Scholar] [CrossRef]
  40. Zeng, Y. Modulatory Effects of Blueberry Leaf Polyphenols on Antibiotic-Induced Gut Microbiota Dysbiosis. Master’s thesis, Chengdu University, Chengdu, China, 2025. [Google Scholar] [CrossRef]
  41. Han, T.Y.; Yang, D.; Zhou, S.Q.; Qiao, Y.M.; Yin, J.; Jin, M.; Li, J.W. Regulative effect of active components of Cistanche deserticola on intestinal dysbacteriosis induced by antibiotics in mice. Zhongguo Ying Yong Sheng Li Xue Za Zhi 2022, 38, 766–770. (In Chinese) [Google Scholar] [CrossRef] [PubMed]
  42. Li, H.X.; Ji, D.; Lu, C.L.; Ye, Q.Y.; Zhao, L.H.; Gao, Y.J.; Gao, L.; Yang, Z.Q. Regulatory effect of polysaccharides from Antrodia cinnamomea in submerged fermentation on gut microbiota in mice with antibiotic-associated diarrhea. Food Sci. 2023, 44, 42–51. [Google Scholar]
  43. Ma, Z.J.; Wang, H.J.; Ma, X.J.; Li, Y.; Yang, H.J.; Li, H.; Su, J.R.; Zhang, C.E.; Huang, L.Q. Modulation of gut microbiota and intestinal barrier function during alleviation of antibiotic-associated diarrhea with rhizoma Zingiber officinale (Ginger) extract. Food Funct. 2020, 11, 10839–10851. [Google Scholar] [CrossRef]
  44. Zhang, X.Q.; Zhang, C.E.; Yu, X.H.; Ma, Y.Q.; Li, M.; Duan, X.Y.; Ma, Z.J. Modulation of gut microbiota during alleviation of antibiotic-associated diarrhea with Zingiberis rhizoma. Zhongguo Zhong Yao Za Zhi 2022, 47, 1316–1326. [Google Scholar] [CrossRef] [PubMed]
  45. Li, Y.; Wang, H.J.; Zhang, C.E.; Ma, Z.J.; Duan, X.Y. Effects of Xiyangshen (Panacis Quinquefolii Radix) on intestinal flora in rats with antibiotic-associated diarrhea. Chin. Arch. Tradit. Chin. Med. 2022, 40, 97–101. [Google Scholar] [CrossRef]
  46. Qu, Q.; Zhao, C.; Yang, C.; Zhou, Q.; Liu, X.; Yang, P.; Yang, F.; Shi, X. Limosilactobacillus fermentum-fermented ginseng improved antibiotic-induced diarrhoea and the gut microbiota profiles of rats. J. Appl. Microbiol. 2022, 133, 3476–3489. [Google Scholar] [CrossRef] [PubMed]
  47. Zhong, B.; Liang, W.; Zhao, Y.; Li, F.; Zhao, Z.; Gao, Y.; Yang, G.; Li, S. Combination of Lactiplantibacillus plantarum ELF051 and astragalus polysaccharides improves intestinal barrier function and gut microbiota profiles in mice with antibiotic-associated diarrhea. Probiotics Antimicrob. Proteins 2024, 17, 4267–4280. [Google Scholar] [CrossRef]
  48. Tang, J.; Ma, X.; Song, X.; Chen, W. Probiotic powder with polysaccharides from Wolfiporia cocos alleviates antibiotic-associated diarrhea by modulating immune activities and gut microbiota. Int. J. Biol. Macromol. 2024, 282, 136792. [Google Scholar] [CrossRef]
  49. Guo, H.; He, X.; Yu, L.; Tian, F.; Chen, W.; Zhai, Q. Bifidobacterium adolescentis CCFM1285 combined with yeast β-glucan alleviates the gut microbiota and metabolic disturbances in mice with antibiotic-associated diarrhea. Food Funct. 2024, 15, 3709–3721. [Google Scholar] [CrossRef]
  50. Li, Z.; Zhou, Q.; Qu, Q.; Liao, Y.; Yang, F.; Sheng, M.; Feng, L.; Shi, X. Effect of maifan stone on the growth of probiotics and regulation of gut microbiota. Lett. Appl. Microbiol. 2022, 75, 1423–1432. [Google Scholar] [CrossRef]
  51. Shen, H.; Zhao, Y.; Zhang, S.; Xu, J.; Tong, J.; Yao, H.; Zhang, H. Synbiotic supplementation mitigates antibiotic-associated diarrhea by enhancing gut microbiota composition and intestinal barrier function in a canine model. Probiotics Antimicrob. Proteins 2025, 17, 2586–2599. [Google Scholar] [CrossRef]
  52. Gomez Quintero, D.F.; Kok, C.R.; Hutkins, R. The future of Synbiotics: Rational formulation and design. Front. Microbiol. 2022, 13, 919725. [Google Scholar] [CrossRef]
  53. Swanson, K.S.; Gibson, G.R.; Hutkins, R.; Reimer, R.A.; Reid, G.; Verbeke, K.; Scott, K.P.; Holscher, H.D.; Azad, M.B.; Delzenne, N.M.; et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of Synbiotics. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 687–701. [Google Scholar] [CrossRef]
  54. Loh, J.T.; Lee, K.G.; Lee, A.P.; Teo, J.K.H.; Lim, H.L.; Kim, S.S.Y.; Tan, A.H.M.; Lam, K.P. DOK3 maintains intestinal homeostasis by suppressing JAK2/STAT3 signaling and S100a8/9 production in neutrophils. Cell Death Dis. 2021, 12, 1054. [Google Scholar] [CrossRef] [PubMed]
  55. Seo, D.H.; Che, X.; Kim, S.; Kim, D.H.; Ma, H.W.; Kim, J.H.; Kim, T.I.; Kim, W.H.; Kim, S.W.; Cheon, J.H. Triggering receptor expressed on myeloid Cells-1 agonist regulates intestinal inflammation via CD177+ neutrophils. Front. Immunol. 2021, 12, 650864. [Google Scholar] [CrossRef]
  56. Siddiqui, M.T.; Cresci, G.A.M. The immunomodulatory functions of butyrate. J. Inflamm. Res. 2021, 14, 6025–6041. [Google Scholar] [CrossRef]
  57. Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef]
  58. Markowiak-Kopeć, P.; Śliżewska, K. The effect of probiotics on the production of short-chain fatty acids by human intestinal microbiome. Nutrients 2020, 12, 1107. [Google Scholar] [CrossRef] [PubMed]
  59. Kadlec, R.; Jakubec, M. The effect of prebiotics on adherence of probiotics. J. Dairy. Sci. 2014, 97, 1983–1990. [Google Scholar] [CrossRef] [PubMed]
  60. Pandey, K.R.; Naik, S.R.; Vakil, B.V. Probiotics, prebiotics and Synbiotics—A review. J. Food Sci. Technol. 2015, 52, 7577–7587. [Google Scholar] [CrossRef]
Figure 1. PRISMA 2020 flow diagram of study selection. HNP = Herbal/Natural Product; HNP-C = Herbal/Natural Product Combination.
Figure 1. PRISMA 2020 flow diagram of study selection. HNP = Herbal/Natural Product; HNP-C = Herbal/Natural Product Combination.
Pharmaceuticals 19 00064 g001
Figure 2. SYRCLE risk-of-bias assessment of included animal studies [25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51]. (A) Summary of risk-of-bias ratings across all SYRCLE domains, presented as the percentage of studies classified as low, high, or unclear risk. (B) Risk-of-bias evaluation for each included study, with individual SYRCLE items scored as yes (low risk), no (high risk), or unclear (−).
Figure 2. SYRCLE risk-of-bias assessment of included animal studies [25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51]. (A) Summary of risk-of-bias ratings across all SYRCLE domains, presented as the percentage of studies classified as low, high, or unclear risk. (B) Risk-of-bias evaluation for each included study, with individual SYRCLE items scored as yes (low risk), no (high risk), or unclear (−).
Pharmaceuticals 19 00064 g002aPharmaceuticals 19 00064 g002b
Table 1. Study characteristics of the included HNP and HNP-C interventions.
Table 1. Study characteristics of the included HNP and HNP-C interventions.
Study IDIntervention TypeIntervention (Source Herb—Fraction/Extract + Dose/Duration)Animal ModelAAD Induction (Agent, Duration)Groups (n)Comparator ArmsTreatment DurationTotal PeriodMicrobiome Method
Xu et al. [25]HNPCitrus reticulata—Tangeretin flavonoid (250 ppm, 10 days; co-administered with antibiotics)C57BL/6 J miceAmpicillin + Neomycin, 10 daysn = 8 × 2AB-only vs. HNP-only10 days10 days16S rRNA (V3–V4), in vitro fermentation
Ren et al. [26]HNPPanax quinquefolius—Water-soluble polysaccharide (100 mg/kg, 7 days)Wistar ratsClindamycin phosphate, 5 daysn = 6 × 4NC/Model/NR/HNP-only7 days12 days16S rRNA sequencing
Min, et al. [27]HNPPanax ginseng—Polysaccharide fraction (100/300 mg/kg, 12 days)BALB/c miceLincomycin, 9 daysn = 7 × 4NC/Model/HNP-low/high12 days21 days16S rRNA
Qi et al. [28]HNPPanax ginseng—Neutral polysaccharide (dose NR, 7 days)BALB/c miceLincomycin hydrochloride, 3 daysn = 10 × 4NC/Model/NR/HNP-only7 days10 days16S rRNA (V3–V4)
Pan et al. [29]HNPDioscorea opposita—Sulfated polysaccharide (30 mg/kg, 14 days; antibiotics days 1–7)MiceMixed antibiotics, 7 daysn = 8 × 3NC/Model/HNP-only14 days14 days16S rRNA DGGE
Li S, et al. [30]HNPAstragalus membranaceus—Water-soluble polysaccharide (100 mg/kg, 7 days)Wistar ratsLincomycin hydrochloride, 4 daysn = 6 × 4NC/Model/NR/HNP-only7 days11 days16S rRNA (V3–V4)
Xu et al. [31]HNPPoria cocos—Water-soluble polysaccharide (250 mg/kg/day, 7 days)MiceBroad-spectrum antibiotics, 7 daysn = 6 × 4NC/AB-only/HNP-only/Positive Control7 days14 days16S rDNA
Lai et al. [32]HNPDictyophora indusiata—Water-insoluble polysaccharide (300 mg/kg, 7 days)C57BL/6 J miceLincomycin hydrochloride, 3 daysn = 7 × 3NC/Model (NR-like)/HNP-only7 days10 days16S rRNA + GC–MS
Bie et al. [33]HNPIpomoea batatas—Polysaccharide fraction (0.1–0.4 g/kg, 14 days)BALB/c miceLincomycin hydrochloride, 3 daysn = 10 × 5NC/Model/HNP (Low/Mid/High)14 days17 days16S rRNA
Zhang et al. [34]HNPDioscorea opposita—Water extract (dose NR, 10 days)BALB/c miceAmpicillin, 5 daysn = 10 × 5NC/Model/HNP (Low/Mid/High)10 days15 daysPCR-DGGE
Pan et al. [35]HNPNemacystus decipiens—Polysaccharide fraction (30 mg/kg/day, 14 days)C57BL/6 miceFour-antibiotic mix, 7 daysn = 8 × 3NC/Model/HNP-only14 days21 days16S rRNA
Lai et al. [36]HNPPoria cocos—Water-insoluble polysaccharide (300 mg/kg/day, 7 days)C57BL/6 miceLincomycin hydrochloride, 3 daysn = 7 × 4NC/Model/HNP-only/NR7 days10 days16S rRNA
Lu et al. [37]HNPAntrodia cinnamomea—Intracellular polysaccharide (8 days)ICR miceLincomycin hydrochloride, 3 daysn = 7 × 6NC/Model/HNP (L/M/H)/FOS (PC)8 days11 days16S rRNA
Cui et al. [38]HNPCereus sinensis—Polysaccharide (75–300 mg/kg, 9 days)C57BL/6 miceLincomycin hydrochloride, 3 daysn = 3 × 6NC/Model/NR/HNP (L/M/H)9 days12 days16S rRNA + GC–MS
Chen et al. [39]HNPLeleba oldhami—Bamboo-shoot polysaccharide (100–400 mg/kg, 15 days)Kunming miceLincomycin hydrochloride, 3 daysn = 6 × 6NC/Model/Inulin (PC)/HNP (L/M/H)15 days18 days16S rRNA + SCFA detection
Zeng. [40]HNPVaccinium corymbosum—Leaf polyphenol extract (100–300 mg/kg, 14 days)BALB/c miceLincomycin hydrochloride, 3 daysn = 10 × 6NC/Model/HNP (L/M/H)/GOS (PC)14 days17 days16S rRNA + GC–MS
Han et al. [41]HNPCistanche deserticola—Extract/polysaccharide/echinacoside (7 days)BALB/c miceLincomycin hydrochloride, 7 daysn = 8 × 6NC/Model/Inulin (PC)/HNP fractions7 days14 days16S rDNA
Li et al. [42]HNPAntrodia cinnamomea—Polysaccharide fractions (0.03 g/kg, 3 days)ICR miceLincomycin hydrochloride, 3 daysn = 6 × 5NC/Model/HNP fractions/FOS (PC)3 days6 days16S rDNA
Ma et al. [43]HNPZingiber officinale—Fresh ginger extract (1.2 or 4.8 g/kg/day, 7 days)SD ratsClindamycin + Ampicillin + Streptomycin, 7 daysn = 10 × 4NC/Model/HNP-low/high7 days21 days16S rRNA
Zhang et al. [44]HNPZingiberis rhizoma—Dried ginger extract (200–400 mg/kg/day, 14 days)SD ratsSame three-antibiotic mix, 7 daysn = 10 × 5NC/Model/HNP-low/high/PC14 days21 days16S rRNA
Li et al. [45]HNPPanax quinquefolium—Decoction extract (1–2 g/kg/day, 7 days)SD ratsThree-antibiotic mix, 7 daysn = 10 × 4NC/Model/HNP-low/high7 days21 days16S rRNA
Qu et al. [46]HNP-CPanax ginseng (fermented with Limosilactobacillus fermentum)—Fermented ginseng mixtureSD ratsCephalexin + Clindamycin + Streptomycin, 7 daysn = 8 × 5NC/Model/HNP-C/Probiotic-only/Mixed Combo5 days12 days16S rRNA
Zhong et al. [47]HNP-CAstragalus membranaceus polysaccharide + Lactiplantibacillus plantarum ELF051C57BL/6 miceAmoxicillin + Clindamycin + Streptomycin, 14 daysn = 10 × 5NC/Model/HNP-only/PRO-only/HNP-C14 days28 days16S rRNA
Tang et al. [48]HNP-CWolfiporia cocos polysaccharide + probiotic mixture (Lactobacillus, Bifidobacterium, Enterococcus)Kunming miceAmpicillin sodium, 5 daysn = 10 × 5NC/Model/HNP-only/PRO-only/HNP-C14 days19 days16S rRNA + CFU
Guo et al. [49]HNP-CBifidobacterium adolescentis + yeast β-glucanBALB/c miceLincomycin hydrochloride, 3 daysn = 10 × 6NC/Model/HNP-only/PRO-only/HNP-C/NR7 days10 days16S rRNA + metabolomics
Li et al. [50]HNP-CMaifan stone + Lactobacillus rhamnosus GGSD ratsClindamycin + Cephalexin + Streptomycin, 5 daysn = 10 × 6NC/Model/NR/PRO-only/HNP-only/HNP-C5 days10 daysqPCR (no 16S)
Shen et al. [51]HNP-CChitosan oligosaccharides + multi-strain probiotic mixtureBeagle dogsEnrofloxacin + Metronidazole, 7 daysn = 8 × 2Model vs. HNP-C28 days35 days16S rRNA
AAD = Antibiotic-associated diarrhea, AB = Antibiotic, NR = Natural recovery, HNP = Herbal or natural product, HNP-C = Herbal–probiotic combination, PRO-only = Probiotic-only group. PC = Positive control (Inulin, FOS, GOS, etc.), NC = Normal control, L/M/H = Low/Medium/High dose.
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

Hwang, J.H.; Choi, Y.-K. Herbal and Natural Products for Antibiotic-Associated Diarrhea: A Systematic Review of Animal Studies Focusing on Molecular Microbiome and Barrier Outcomes. Pharmaceuticals 2026, 19, 64. https://doi.org/10.3390/ph19010064

AMA Style

Hwang JH, Choi Y-K. Herbal and Natural Products for Antibiotic-Associated Diarrhea: A Systematic Review of Animal Studies Focusing on Molecular Microbiome and Barrier Outcomes. Pharmaceuticals. 2026; 19(1):64. https://doi.org/10.3390/ph19010064

Chicago/Turabian Style

Hwang, Ji Hye, and You-Kyoung Choi. 2026. "Herbal and Natural Products for Antibiotic-Associated Diarrhea: A Systematic Review of Animal Studies Focusing on Molecular Microbiome and Barrier Outcomes" Pharmaceuticals 19, no. 1: 64. https://doi.org/10.3390/ph19010064

APA Style

Hwang, J. H., & Choi, Y.-K. (2026). Herbal and Natural Products for Antibiotic-Associated Diarrhea: A Systematic Review of Animal Studies Focusing on Molecular Microbiome and Barrier Outcomes. Pharmaceuticals, 19(1), 64. https://doi.org/10.3390/ph19010064

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