Graphical abstract graphic file with name fx1.jpg [53]Open in a new tab Highlights * • Probiotics promote HBV clearance by producing spermidine * • Probiotics and spermidine enhance IFN-γ^+CD4^+ T cell function through autophagy * • Probiotics induce the decline of serum HBsAg in patients with low HBsAg levels __________________________________________________________________ Commensal microbes are promising in the functional cure of HBV infection. Wang et al. demonstrate that probiotics and their metabolite spermidine effectively promote the decline of HBsAg and inhibit HBV replication via autophagy-enhanced IFN-γ^+CD4^+ T cell immunity, highlighting the therapeutic potential of probiotics and spermidine for the functional cure of HBV patients. Introduction Globally, approximately 296 million people are chronically infected with hepatitis B virus (HBV), which poses a high risk of liver cirrhosis and hepatocellular carcinoma, making it as one of the most serious common infectious agents.[54]^1 In 2016, the World Health Organization called for the goal of eliminating viral hepatitis, defined as 95% reduction in HBV incidence and 65% reduction in mortality by 2030.[55]^2^,[56]^3 The ideal goal of antiviral treatment for HBV patients is to achieve a functional cure, which is characterized by the persistent suppression of HBV DNA and the loss of serum hepatitis B surface antigen (HBsAg).[57]^4^,[58]^5 Unfortunately, achieving the functional cure for chronic HBV infection is difficult. The natural history has a success rate of less than 1%, and current antiviral drugs yield no more than 5% success rate even after long-term treatment.[59]^6^,[60]^7 Dysfunctional or exhausted T cells have been demonstrated to restrict the strength of HBV clearance, and recovery of T cell immunity is believed to be the most promising strategy for functional cure.[61]^8 Stimulating T cell co-stimulatory molecule OX40 and blockade of programmed cell death ligand 1 (PD-L1) synergistically promote the functional restoration of HBV-specific CD4^+ T cells, leading to the production of interferon (IFN)-γ and interleukin (IL)-21, which are essential for HBV clearance.[62]^9 Hoogeveen et al. also showed that functional cure is associated with robust responses by HBV-specific CD4^+ T cells.[63]^10 Therefore, it is important to develop exact and effective treatment strategies for restoring dysfunctional T cell immunity and achieve functional cure of HBV infection. The liver is the unique target organ that orchestrates HBV-mediated immune response and is coordinated by gut microbe-derived ligands or metabolites through the gut-liver axis.[64]^11 Altered and flexible profile of gut microbiota in HBV infection has been reported,[65]^12^,[66]^13^,[67]^14 and the establishment of gut microbiota determines the age-related HBV clearance, in which T cell response plays pivotal roles.[68]^15^,[69]^16 In the limited data of HBV patients, Ren et al. reported that fecal microbiota transplantation (FMT) presented the ability to induce the decline of HBV e-antigen (HBeAg) expression in persistent HBeAg-positive patients even after long-term antiviral treatment.[70]^17 Mechanistically, commensal microbes and their metabolites regulate the production of IL-22, IL-17A, and IL-21 to promote intestinal immune homeostasis.[71]^18^,[72]^19 However, multidrug-resistant organisms colonized in healthy donors’ feces during donation activities would pose a risk for FMT.[73]^20 Probiotics are widely used as a food supplement to assist in the treatment of many health or disease conditions.[74]^21 Currently, the molecular and immune mechanism by which probiotics promote HBV clearance remains unclear. Consequently, the key and specific microbes or metabolites that facilitate HBV clearance have yet to be identified. Here, we reported that probiotics (B. longum, L. acidophilus, and E. faecalis, BLE), widely used in clinical practice, promote HBV clearance in both specific-pathogen-free (SPF) mice and germ-free (GF) mice. Specifically, spermidine (SPD) derived from accumulated probiotics in the gut could transport to the liver and exhibit a similar anti-HBV effect to probiotics. Mechanistically, depletion of CD4^+ T cells or blockage of IL-21R or IFN-γ almost completely dampened probiotics-induced HBV inhibition, and SPD administration activated the CD4^+ T cell immunity through autophagy. In HBV patients undergoing antiviral treatment, probiotics showed the potential to induce the decline of serum HBsAg. Results Administration of probiotics BLE promotes HBV clearance and improves intestinal commensal homeostasis and barrier function To evaluate the efficiency of probiotics (BLE) in HBV clearance, HBV-carrier mouse models were prepared by hydrodynamical injection (HDI) of adeno-associated virus/HBV1.2 (AAV/HBV1.2) plasmids in 5-week-old C57BL/6 mice.[75]^22 BLE were administered by gavage daily starting from either 2 weeks prior to HDI (BLE Pre group) or after 3 days post injection (BLE group) ([76]Figure 1A). As shown in [77]Figures 1B–1E, both BLE and BLE Pre significantly inhibited HBV replication, displayed as decreased levels of serum HBsAg, HBV DNA, as well as hepatic pregenomic RNA (pgRNA) and HBV RNA. Consistently, BLE administration significantly decreased the percentage of HBsAg-positive mice within 6 weeks post injection (wpi) ([78]Figure 1F). In addition, no liver injury was detected in mice treated with BLE ([79]Figures S1A and S1B). Since BLE and BLE Pre showed similar anti-HBV effects, we used orally daily administration of BLE as the routine probiotic treatment in the following experiments. Figure 1. [80]Figure 1 [81]Open in a new tab Supplementation with BLE promoted HBV clearance in mice (A) Study design: male five-week-old C57BL/6J mice were hydrodynamically injected with 6 μg of AAV/HBV1.2 plasmid and gavage fed with probiotics (BLE, PBS as control) daily starting two weeks before (Pre) or at 3 dpi of HDI (control, n = 10; BLE Pre, n = 10; BLE, n = 10). (B–E) Levels of serum HBsAg (B), HBV DNA (C), hepatic pgRNA (D), and HBV RNA (E) were detected by ELISA, qPCR, and northern blot, respectively. (F) The percentages of HBsAg-positive mice (HBsAg > 1 ng/mL) at the indicated time points were calculated. Data were expressed as mean ± SEM. The statistical tests were performed by two-way analysis of variance (ANOVA) with Dunnett’s multiple comparison test (B and C), t test (D), or log rank (F). ∗0.01 < p value < 0.05; ∗∗0.001 < p value < 0.01; ∗∗∗∗p value < 0.0001. Probiotics show well-defined improvement in gut microbiota. We then investigated the effect of BLE administration on microbiota profile in HBV-carrier mice. Principal coordinate analysis (PCoA) revealed significant shifts (p = 0.004) in intestinal microbiome composition between control and BLE mice ([82]Figure 2A). Taxonomic alpha diversity (Chao diversity) analysis showed a marked increase in species richness in BLE groups ([83]Figure 2B). Moreover, linear discriminant analysis effect size (LEfSe) analysis displayed that the microbiome of BLE mice was significantly enriched with families Prevotellaceae, Clostridiaceae, Akkermansia, and Ruminococcaceae ([84]Figure 2C), which have been suggested to contribute to the restoration of exhausted CD8^+ T cell mediated by anti-PD-1 or CTLA-4 antibodies.[85]^23^,[86]^24^,[87]^25 Further, qPCR analysis using bacterium-specific primers showed that the abundance of BLE in the feces significantly increased after probiotics supplementation, suggesting the accumulation of probiotics bacteria ([88]Figure 2D). Figure 2. [89]Figure 2 [90]Open in a new tab Accumulation of BLE contributed to gut microbiota homeostasis and HBV clearance (A–C) 16S rRNA sequencing was performed with feces collected at 2 wpi. Beta diversity comparison via PCoA analysis (A) and alpha diversity comparison via Chao index analysis (B) showed a difference between the two groups. Bray-Curtis distance algorithm was used for PCoA analysis. (C) LEfSe (linear discriminant analysis effect size) predictions for bacterial families found in fecal pellets of control (green) and BLE (red) mice were shown, with cutoffs of LDA < −2 or LDA > 2. (D) Fecal bacterial genomic DNA was extracted from control and BLE mice. The expression of B. longum, L. acidophilus, and E. faecalis was determined by qPCR. (E) Study design: male three-week-old C57BL/6J mice were treated with antibiotics water for two weeks and then hydrodynamically injected with 6 μg of pAAV/HBV1.2 plasmid and fed with fecal microbiota from control and BLE mice daily starting at 3 dpi of HDI (FMT-control, n = 10; FMT-BLE, n = 10). (F–H) Levels of serum HBsAg (F), HBV DNA (G), and hepatic pgRNA (H) were detected by ELISA and qPCR. (I) The percentages of HBsAg-positive mice (HBsAg > 1 ng/mL) at the indicated time points were calculated. (J) Study design: male six-week-old C57BL/6J GF mice were hydrodynamically injected with 6 μg of pAAV/HBV1.2 plasmid and gavage fed with BLE daily starting at 3 dpi of HDI. (K) The expression of B. longum, L. acidophilus, and E. faecalis was determined by qPCR. (L) Levels of serum HBsAg, HBV DNA, and hepatic pgRNA were detected by ELISA and qPCR. Data were expressed as mean ± SEM. The statistical tests were performed by analysis of similarities (ANOSIM) (A), t test (B, D, H, K, and L), parametric factorial Kruskal-Wallis (KW) sum-rank test (C), two-way ANOVA (F and G), or log rank (I). ∗0.01 < p value < 0.05; ∗∗0.001 < p value < 0.01; ∗∗∗ 0.0001 < p value < 0.001; ∗∗∗∗p value < 0.0001. Intestinal microbiota plays a critical role in maintaining gut barrier and permeability, which is strongly associated with the development of chronic liver diseases.[91]^26 In accordance, we detected lower serum fluorescein isothiocyanate (FITC)-dextran concentration in BLE mice than in control mice ([92]Figure S1C), indicating improved intestinal barrier integrity in BLE mice. Consistently, the serum level of intestinal fatty acid binding protein (iFABP), a biomarker of enterocyte damage,[93]^27 significantly decreased in BLE mice ([94]Figure S1D). In contrast, there was higher expression of tight junction proteins, including Claudin 1, Occludin, and Tjp1, in small intestine (SI) tissues from BLE mice than those from control mice ([95]Figure S1E). It has been well documented that microbiota induces the production of IL-17A, IL-21, and IL-22 to improve gut integrity and homeostasis.[96]^18^,[97]^19 As expected, significantly higher expression of IL-17A, IL-21, and IL-22 at both the mRNA and protein levels was detected in SI tissues from BLE mice ([98]Figures S1F and S1G). Flow cytometric (FCM) analysis verified the marked increase in the frequency of T cells producing IL-17A, IL-21, and IL-22 in lymphocytes of small intestinal lamina propria, mesenteric lymph nodes (mLNs) and Peyer’s patches (PPs) from BLE mice than control mice ([99]Figures S1H–S1K). Collectively, these data suggest that accumulation of BLE improves intestinal commensal homeostasis and barrier functions in HBV-carrier mice. Accumulation of BLE contributes to HBV clearance in mice To assess whether gut microbiota is involved in BLE-mediated HBV suppression, feces were collected from PBS- or BLE-treated HBV-carrier mice (donor mice). Then, FMT experiments were performed with recipient mice, which were gut-sterilized using a 14-day antibiotic mixture (ABX) treatment followed by HDI of HBV at the age of 5 weeks ([100]Figure 2E). As shown in [101]Figures 2F–2H, transplantation of fecal microbiota from BLE mice (FMT-BLE) greatly promoted HBV clearance in recipient mice, displaying obviously decreased levels of serum HBsAg and HBV DNA and reduced hepatic pgRNA expression when compared to mice that received control fecal microbiota (FMT-Control). Similarly, the percentage of HBsAg-positive mice in FMT-BLE mice was significantly lower than that in FMT-Control mice ([102]Figure 2I). To further validate the direct role of BLE in HBV clearance, we included GF mice to establish a HBV model by HDI, followed by probiotics administration ([103]Figure 2J). Consisting with results in SPF mice, supplementation with BLE largely increased the numbers of BLE in the feces from GF mice ([104]Figure 2K) and decreased levels of serum HBsAg, HBV DNA, and hepatic pgRNA ([105]Figure 2L). Taken together, these data suggest that the accumulation of BLE contributes to HBV clearance in mice. IFN-γ and IL-21 produced by CD4^+ T cells are indispensable for BLE-induced HBV clearance To further evaluate the role of individual immune subsets in probiotics-mediated HBV clearance, AsGM1 and neutralized antibodies against CD4, CD8, and TCRγ/δ were used in probiotics-treated HBV-carrier mice. As shown in [106]Figures 3A–3C, depletion of CD8^+ T cells, γδT cells, or natural killer (NK) cells partially restored the reduced serum levels of HBsAg in BLE-treated mice. Notably, depletion of CD4^+ T cells almost completely abolished BLE-induced HBV clearance, suggesting that BLE promotes HBV clearance in a CD4^+ T cell-dependent manner. In line with the critical role of CD4^+ T cells, probiotics treatment led to augmentation of CD44^+CD62L^− effector CD4^+ T cells and CD44^+CD62L^+ central memory CD4^+ T cells in the spleen and liver ([107]Figure S2A). Figure 3. [108]Figure 3 [109]Open in a new tab Depletion of CD4^+ T cells and inactivation of IL-21 or IFN-γ abrogated the probiotics-initiated anti-HBV effects (A–C) Mice were treated i.p. with 100 mg neutralization antibody or isotype every 3 days from 0 to 6 wpi (control, n = 7; BLE, n = 7; BLE+αCD4, n = 9; BLE+αCD8, n = 5; BLE+αTCRγ/δ, n = 7; BLE+AsGM1, n = 5). Serum levels of HBsAg (A), HBV DNA (B), and liver pgRNA (C) were detected by ELISA and qPCR. HBV-carrier mice were treated with BLE or control PBS. (D and E) RT-qPCR analysis of transcription factors (D) and cytokines (E) in liver tissues. (F) FCM analysis of percentages of hepatic T-bet^+ Th1 cells, PD-1^+CXCR5^+ Tfh cells, and RORγt^+ Th17 cells among CD4^+ T cells. (G) IL-21 and IFN-γ in liver homogenates were analyzed by ELISA. (H and I) Percentages and MFI of IL-21 (H) and IFN-γ (I) in hepatic T-bet^+Th1 cells. (J–L) Antibody-mediated blockade of IL-21R and IFN-γ abolished the effect of probiotics-induced HBV clearance. Mice were treated i.p. with 100 mg neutralization antibody (anti-IL-21R and anti-IFN-γ) or isotype every 3 days from 0 to 6 wpi (control, n = 7; BLE, n = 7; BLE+αIL-21R, n = 4; BLE+αIFN-γ, n = 7). Serum levels of HBsAg (J), HBV DNA (K), and liver pgRNA (L) were detected by ELISA and qPCR. Data were expressed as mean ± SEM. The statistical tests were performed using two-way ANOVA with Dunnett’s multiple comparison test (A, B, J, and K) or t test. ns, no significance; ∗0.01 < p value < 0.05; ∗∗0.001 < p value < 0.01; ∗∗∗0.0001 < p value < 0.001; ∗∗∗∗p value < 0.0001. Growing evidence has demonstrated the involvement of different CD4^+ T subsets in anti-HBV immunity.[110]^28^,[111]^29 To gain deeper insight into the role of CD4^+ T subsets in BLE-mediated HBV clearance, we assessed hepatic mRNA expression of signature transcription factors and cytokines of Th1, Th17, follicular helper T (Tfh), and regulatory T (Treg) cells. Quantitative reverse-transcription PCR (RT-qPCR) revealed a significant increase in the expression of Eomes, Tbx21, Il-21, and Il12, while expression of Ifng but not Bcl-6, Rorc, Foxp3, Il17-a, and Tgfb showed no significant change in BLE mice ([112]Figures 3D and 3E). Consistently, FCM showed a significant increase of T-box expressed in T cell (T-bet)^+ Th1 cells, but not CXCR5^+PD-1^+ Tfh cells and ROR-γt^+ Th17 cells, in the livers from BLE-treated mice ([113]Figure 3F). In accordance, BLE treatment led to the accumulation of IL-21^+CD4^+ T cells and IFN-γ^+CD4^+ T cells in the liver and spleen ([114]Figures S2B–S2E). In addition, ELISA revealed higher levels of IL-21 and IFN-γ in liver homogenate in BLE mice ([115]Figure 3G). Tfh cells have been known as one of the main sources of IL-21 and IFN-γ.[116]^30 Further analysis revealed that the expression of IL-21 and IFN-γ significantly increased in Th1 cells, but not in Tfh cells, after BLE treatment ([117]Figures 3H, 3I, [118]S2F, and S2G). It has been reported that IL-21 signaling enhances cytotoxicity and IFN-γ production in NK and CD8^+ T cells.[119]^31 Consistently, BLE treatment increased the production of IFN-γ in splenic CD8^+ T cells ([120]Figures S2H and S2I), indicating the potential contribution of CD8^+ T cells to BLE-induced HBV clearance. In addition, BLE treatment also led to the augmentation of hepatic and splenic IFN-γ^+CD4^+ T cells in GF mice ([121]Figures S2J and S2K). Considering the critical role of IFN-γ and IL-21 in determining HBV persistence,[122]^16^,[123]^32 we therefore determined whether the clearance of HBV in probiotics-treated mice is mediated by the up-regulation of IL-21 or IFN-γ. As shown in [124]Figures 3J–3L, the treatment of neutralized antibodies against either IL-21R or IFN-γ completely abolished the anti-HBV effect mediated by probiotics treatment. In summary, these results illustrate that the probiotics administration enables the host to clear HBV by enhancing the production of IL-21 and IFN-γ in CD4^+ Th1 cells. SPD produced by BLE promotes HBV clearance The microbiota-derived metabolites serve as important signals contributing to gut homeostasis and host immunity. We then investigated whether probiotics affect the microbial metabolism of the host. Phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt) analysis of 16S rRNA sequencing data indicated the enrichment of metabolism-related pathways in BLE-treated mice ([125]Figure S3A; [126]Table S1). In accordance, the gas chromatograph coupled with a Pegasus HT time-of-flight mass spectrometer (GC-TOF-MS) detected the alteration of metabolites in feces from BLE-treated mice. Pathway enrichment analysis using MetaboAnalyst 5.0 ([127]https://www.metaboanalyst.ca) showed that multiple metabolic pathways were significantly enriched in BLE mice, and 15 of them were consistent with the results of PICRUSt analysis ([128]Figures S3A and S3B; the same pathways marked in red). Totally, untargeted metabolomic analysis showed that 38 metabolites were significantly different between control and BLE mice ([129]Figures 4A and [130]S3C). Among them, oligosaccharides (1-kestose, raffinose, maltotriose), amino acids (N-Acetyl-L-leucine, glutamic acid), and polyamine (SPD) were the most varied enriched metabolites. Recently, studies showed that SPD modulates CD4^+ T cell differentiation and enhances vaccine-induced T cell function.[131]^33^,[132]^34 Further targeted metabolomics analysis of polyamines (including SPD, spermine, and putrescine) demonstrated that SPD and putrescine rather than spermine were accumulated in the livers of BLE mice, and the concentration of SPD was higher than that of putrescine ([133]Figure S3D). More importantly, the concentration of SPD was accumulated in the feces, peripheral serum, and livers from BLE-treated GF mice ([134]Figure 4B). These results indicate that BLE-produced SPD may translocate from the intestine to the liver through the blood circulation. Figure 4. [135]Figure 4 [136]Open in a new tab BLE-derived spermidine promoted HBV clearance (A) Untargeted metabolomic analysis of feces from control and BLE mice by gas chromatograph system coupled with a Pegasus HT time-of-flight mass spectrometer (GC-TOF-MS). Heatmap of differentially expressed metabolites in mice fed with BLE were shown (control, n = 9; BLE, n = 9). (B) Targeted metabolomics analysis of SPD in feces, peripheral serum, and livers from GF mice. (C) Extracellular SPD concentrations in cultures of separate E. faecalis and mixed BLE in gifu anaerobe medium (GAM) medium supplemented with 2 mM arginine under different pH values. (D–F) HBV-carrier mice were fed with drinking water (control, n = 8) or 3 mM SPD-containing water (SPD, n = 8). Levels of serum HBsAg (D), HBV DNA (E), and liver pgRNA (F) were detected by ELISA or qPCR. (G and H) FCM analysis of percentages and MFI of hepatic/splenic IL-21^+CD4^+ T cells (G) and IFN-γ^+CD4^+ T cells (H). (I) IL-21 and IFN-γ in liver homogenates were analyzed by ELISA. Data were expressed as mean ± SEM. The statistical tests were performed using t test and two-way ANOVA (D and E). ∗0.01 < p value < 0.05; ∗∗0.001 < p value < 0.01; ∗∗∗∗p value < 0.0001. It has been recently reported that bioactive polyamine can be produced by a hybrid system involving the collaboration of two bacterial groups and triggered by environmental acidification. In this system, E. faecalis, one of the three probiotic strains in BLE, functions as agmatine deiminase system to promote the production of polyamine by the collaboration of bacteria that have an acid resistance system composed of arginine decarboxylase (AdiA) and arginine-agmatine antiporter (AdiC), such as E. coli and B. longum.[137]^35 We thus hypothesized whether the augmented SPD in the liver of BLE mice was attributed to the accumulation of BLE. To address this, we first used traditional bacterial culture methods to obtain culture supernatant and detect SPD concentration. We found that cultured BLE produced significantly higher level of SPD in the arginine-containing medium at pH 5.0 than at pH 7.0 ([138]Figure 4C). Moreover, administration of BLE produced markedly higher levels of SPD in the feces, liver tissues, and serum of antibiotics (ABX)-treated mice, while single E. faecalis was not effective to produce SPD both in vitro and in vivo ([139]Figures 4C and [140]S4A–S4C). Accordingly, in comparison to control and single E. faecalis-administrated mice, BLE administration significantly decreased the serum levels of HBsAg, HBV DNA, and hepatic pgRNA ([141]Figures S4D–S4F). Similar results were observed in FCM analysis of IFN-γ^+CD4^+ T cells in mice livers ([142]Figure S4G). Thus, BLE effectively produces SPD and enhances anti-HBV immunity, with E. faecalis being able to produce SPD with the cooperation of B. longum and L. acidophilus. We then further investigated the role of SPD in HBV clearance. Expectedly, SPD administration alone significantly decreased the levels of serum HBsAg, HBV DNA, and hepatic pgRNA in HBV-carrier mice ([143]Figures 4D–4F). In addition, SPD administration did not cause liver injury ([144]Figures S5A and S5B). Taken together, these results imply that BLE-derived SPD accumulated from the gut to the liver by peripheral circulation, which contributes to HBV clearance in mice. SPD improves intestinal homeostasis and enhances Th1 cell immunity through autophagy To further explore the mechanism underlying SPD-mediated HBV clearance, gut integrity and T cell immunity were evaluated. As shown in [145]Figures S5C–S5E, mice fed with SPD displayed a notable elevation in intestinal Claudin1 expression, accompanied by a clear reduction in serum levels of FITC-dextran and iFABP. This finding aligns with a previous report showing that SPD regulates the intestinal epithelial barrier function.[146]^36 Additionally, SPD feeding led to a great increase in the expression of intestinal IL-17A, IL-21, and IL-22 and the rise in the percentages and MFI of intestinal IL-17A^+CD4^+ T cells, IL-21^+CD4^+ T cells, and IL-22^+CD4^+ T cells ([147]Figures S5F–S5J). Furthermore, consistent with the results in BLE-treated mice, we detected a marked increase in the frequency of IL-21^+CD4^+ T cells and IFN-γ^+CD4^+ T cells in livers and spleens from SPD-treated mice ([148]Figures 4G and 4H). ELISA confirmed the significantly increased levels of IL-21 and IFN-γ in liver homogenate from SPD mice ([149]Figure 4I), suggesting that SPD enhances CD4^+ T cell immunity in HBV-carrier mice. It has been shown that SPD, a natural autophagy-promoting polyamine, exerts multiple functions, including extending lifespan and immune cell activation, in an autophagy-dependent manner.[150]^37 Therefore, we investigated whether SPD enhances CD4^+ T cell function via regulating the autophagy pathway. To address this issue, in vitro splenic cells isolated from OT-II mice were stimulated with ovabumin (OVA[323-339]) peptide with or without SPD. FCM confirmed that SPD enhanced the production of IFN-γ in OVA-stimulated OT-II CD4^+ T cells in a dose-dependent manner ([151]Figure 5A). Concurrently, SPD dose-dependently promoted the accumulation of autophagosomes in OT-II CD4^+ T cells ([152]Figure 5B), while the autophagy inhibitor 3-methyladenine (3-MA, 1 mM) further enhanced the SPD-induced accumulation of autophagosomes (SPD+3-MA, [153]Figure 5C), suggesting the activation of autophagy in SPD-treated CD4^+ T cells. Notably, 3-MA markedly decreased the SPD-induced augmentation of IFN-γ in CD4^+ T cells ([154]Figure 5D). In line with the notion that SPD acts as the only known substrate for the hypusination of eukaryotic translation initiation factor 5A (eIF5A), which is essential for the synthesis of the autophagy transcription factor EB (TFEB),[155]^38 treatment with GC7 (SPD+GC7), a specific inhibitor of eIF5A hypusination, decreased the accumulation of SPD-induced autophagosomes ([156]Figure 5C) and completely damaged SPD-induced accumulation of IFN-γ in CD4^+ T cells ([157]Figure 5D). Consistently, SPD induced autophagosome accumulation in hepatic CD4^+ T cells from HBV-carrier mice ([158]Figure 5E). More importantly, SPD significantly promoted the accumulation of autophagosomes and IFN-γ in CD4^+ T cells derived from peripheral blood of HBV patients, while treatment of GC7 largely damaged this induction ([159]Figures 5F and 5G). Taken together, these data imply that SPD enhances CD4^+ T cell function in an autophagy-dependent manner. Figure 5. [160]Figure 5 [161]Open in a new tab SPD enhanced CD4^+ T cell function through autophagy in vivo and in vitro (A–D) OT-II CD4^+ T cells were prepared by culturing splenic mononuclear cells isolated from OT-II mice with OVA[323-339] peptide for 3 days. OT-II CD4^+ T cells were then treated for 24 h with increasing concentrations of SPD (A and B). 3-MA (1 mM) and GC7 (10 μM) were used to inhibit autophagy (C and D). IFN-γ levels (A and D) and autophagosome levels (B and C) in CD4 ^+ T cells were measured by FCM. (E) Hepatic mononuclear cells were isolated from HBV mice, and levels of autophagosome in CD4 ^+ T cells were measured by FCM. (F and G) PBMCs isolated from HBV patients were stimulated with SPD and/or GC7 for 24 h. Autophagosome levels (F) and IFN-γ levels (G) in CD4^+ T cells were measured by FCM. Data were expressed as mean ± SEM. The statistical tests were performed using t test. ns, no significance; ∗0.01 < p value < 0.05; ∗∗0.001 < p value < 0.01; ∗∗∗ 0.0001 < p value < 0.001; ∗∗∗∗p value < 0.0001. BLE accelerates the decline of serum HBsAg in human flora-associated mice and HBV patients with antiviral treatment To address whether the probiotics would cause similar alterations in a more natural microbiome and evaluate the pre-clinical therapeutic potential of BLE and SPD in HBV clearance, we constructed the human flora-associated (HFA) model by reconstituting the microbiota of GF mice with fecal samples from HBV patients. Three weeks later, the HFA mice were injected with AAV/HBV1.2 by HDI and treated with either BLE or SPD, PBS as control ([162]Figure 6A). As shown in [163]Figures 6B–6D, both BLE and SPD showed significantly higher capacity of HBV clearance in HFA mice. Compared with PBS-treated HFA mice, BLE/SPD-treated HFA mice showed remarkably decreased levels of serum HBsAg, HBV DNA, and hepatic pgRNA. Both BLE and SPD significantly promoted the accumulation of autophagosomes in hepatic CD4^+ T cells ([164]Figure 6E) and enhanced the production of IFN-γ in hepatic and splenic CD4^+ T cells ([165]Figures 6F and [166]S6A). More importantly, both BLE and SPD significantly enhanced the production of IFN-γ in hepatic and splenic HBV-specific CD4^+ T cells, indicating the potential contribution of BLE and SPD to HBV-specific immunity ([167]Figures 6G, [168]S6B, and S6C). Figure 6. [169]Figure 6 [170]Open in a new tab Probiotics accelerated HBsAg clearance in HFA mice and HBV patients (A) Study design: male three-week-old C57BL/6J GF mice were fed with fecal microbiota from HBV patients daily for 3 weeks and then hydrodynamically injected with AAV/HBV1.2 plasmid and fed with BLE or SPD for 6 weeks (control, n = 4; BLE, n = 4; SPD, n = 4). (B–D) Levels of serum HBsAg (B), HBV DNA (C), and hepatic pgRNA (D) were detected by ELISA and qPCR. (E and F) Levels of autophagosome (E) and IFN-γ (F) in CD4^+ T cells were measured by FCM. (G) MHC II-HBcAg[128-140]-Tetramer was used to evaluate HBV-specific CD4^+ T cells. Percentages of IFN-γ in hepatic HBV-specific CD4^+ T cells were measured by FCM. (H) A total of 20 eligible patients who remained persistently positive for HBsAg following >6 months of ongoing nucleos(t)ide analogs- or PegIFNα2b-based antiviral therapy were enrolled; among them, 10 patients received BLE therapy (control, n = 10; BLE, n = 10). (I) HBsAg levels in each patient before and after BLE treatment were analyzed and compared with HBV patients without BLE treatment. (J and K) Levels of autophagosome (J) and IFN-γ (K) in CD4 ^+ T cells from PBMCs were measured by FCM. Data were expressed as mean ± SEM. The statistical tests were performed using two-way ANOVA with Dunnett’s multiple comparison test (B and C) or t test. ∗0.01 < p value < 0.05; ∗∗0.001 < p value < 0.01; ∗∗∗ 0.0001 < p value < 0.001. We therefore conducted the preliminary observation for clinical effects of BLE combined with antiviral treatment in HBV patients with persistent low level of serum HBsAg (less than 2,000 IU/mL), which were usually considered as advantaged population trend for functional cure[171]^39 ([172]Tables S2 and [173]S3; [174]Figure 6H). The dynamic profiles of serum HBsAg levels before and after BLE treatment have been illustrated ([175]Figure S7A). Compared with control patients, patients with BLE treatment for 6 months showed an obvious decline of serum HBsAg and HBeAg ([176]Figures 6I and [177]S7B). In detail, all 10 patients presented a declined trend of serum HBsAg level, and 3 of 10 patients even obtained the successful loss of serum HBsAg after the treatment (patients B5, B6, B7). Notably, patient B5 and B6 maintained the state of HBsAg negative for 6 months ([178]Figure S7A). Expectedly, BLE supplement significantly promoted the accumulation of autophagosomes and IFN-γ expression in CD4^+ T cells from peripheral blood mononuclear cells (PBMCs) ([179]Figures 6J and 6K). Although this study was a preliminary observation with small number of patients, these results clearly suggest the therapeutic potential of BLE toward the functional cure of HBV infection. SPD synergically enhances the anti-HBV effect of entecavir in mice Based on the aforementioned data proving the role of SPD in T cell activation and HBV clearance, we next determined whether co-administration of SPD could enhance the therapeutic effect of entecavir (ETV), the most widely used medication that efficiently inhibits HBV DNA replication but not HBsAg production.[180]^40 As shown in [181]Figures 7A–7D, compared to ETV monotherapy, which only suppressed HBV DNA level and showed minimal effects on HBsAg and pgRNA, the combination therapy (ETV+SPD) group demonstrated a more pronounced reduction in HBsAg and pgRNA levels, while displaying comparable inhibitory effects on HBV DNA. In accordance with the promotion of Th1 immunity in BLE-treated mice, RT-qPCR detected significantly increased expression of Il21, Ifng, Il12, Tbx21, and Eomes in mice with co-administration of ETV with SPD but not in mice with ETV monotherapy ([182]Figure 7E). Furthermore, compared with ETV monotherapy, combination therapy significantly increased the production of IFN-γ in splenic and hepatic CD4^+ T cells ([183]Figures 7F and 7G). Taken together, these data demonstrate that SPD greatly enhances the anti-HBV therapeutic effect of ETV by promoting antiviral immunity and suggest the therapeutic potential of SPD toward the functional cure of HBV infection. Figure 7. [184]Figure 7 [185]Open in a new tab Combination of SPD promoted the HBV inhibition in HBV-carrier mice (A) Male five-week-old HBV-carrier mice were administered with entecavir (ETV) or co-administered with 3 mM SPD water (ETV+SPD) combined with ETV (control, n = 5; SPD, n = 5; ETV, n = 5; ETV+SPD, n = 5). (B–D) Levels of serum HBsAg (B), HBV DNA (C), and liver pgRNA (D) were detected by ELISA or qPCR. (E) RT-qPCR analysis of transcription factors and cytokines in liver tissues. (F and G) FCM analysis of percentages and MFI of splenic (F) and hepatic (G) IFN-γ^+CD4^+ T cells. Data were expressed as mean ± SEM. The statistical tests were performed using t test. ns, no significance; ∗0.01 < p value < 0.05; ∗∗0.001 < p value < 0.01; ∗∗∗ 0.0001 < p value < 0.001. Discussion Functional cure of HBV infection is considered an achievable therapeutic goal, given that it is almost universally observed after adult infection. However, the current anti-HBV drugs, namely nucleos(t)ide analogs as well as peginterferon-α (PegIFNα), cured fewer than 1% in the natural history of chronic HBV infection.[186]^41 The high burden of viral antigens that promote T cell exhaustion with T cell dysfunction is the key barrier to functional cure.[187]^8 In this study, we demonstrate the critical role of probiotics in enhancing CD4^+ T cell function and promoting the clearance of HBV. Furthermore, probiotics showed the potential to accelerate the decline of serum HBsAg levels in HBV patients with common anti-HBV drugs. Probiotics are widely used as food supplement to assist in the treatment of many health conditions,[188]^21 and no liver injury was detected in mice treated with BLE in our studies. Our data here provide a safe and effective combination intervention strategy for chronic HBV infection. Scientific evidence supports the important roles of probiotics in alleviating the symptoms of several digestive system diseases.[189]^21 A large number of studies also have demonstrated the disturbed gut microbiota in HBV patients.[190]^42 In this study, using 16S rRNA sequencing, we showed that probiotics administration alters microbiome compositions and enriches commensal diversity in HBV-carrier mice. Further, qPCR analysis using bacterium-specific primers showed the accumulation of BLE in probiotics-treated mice. Then, FMT experiments and GF mice were used to verify that accumulation of BLE contributes to HBV clearance. Therapeutic strategies to cure persistent HBV infection almost invariably include approaches to enhance antiviral T cell immunity.[191]^43 Our data suggest that BLE-induced HBV clearance is achieved by provoking CD4^+ T cell function. Depletion of CD4^+ T cells, as well as blocking IL-21 and IFN-γ, the well-known effector molecules of CD4^+ Th1 cells, almost abolished BLE-initiated HBV clearance. Further expression analysis of signature transcription factors of different CD4^+ T cell subsets demonstrated the augmented expression of Tbx21 and Eomes but not Bcl-6, Rorc, and Foxp3 in probiotics-treated mice, suggesting the involvement of Th1 cells in probiotics-mediated HBV clearance. Although T-bet was also involved in Tfh cell development and germinal center generation, and expression of IL-21 and IFN-γ is detected in Tfh cells,[192]^44 FCM analysis showed no alteration in either IL-21^+ Tfh or IFN-γ^+ Tfh cells in BLE-treated mice. Meanwhile, BLE treatment did not influence the production of HBsAb ([193]Table S4). These results suggest that Th1 but not Tfh cells were the main effector cells contributing to BLE-induced HBV clearance. Another finding of our study is that we have demonstrated that SPD is the key metabolite responsible for HBV clearance and is produced by BLE. Firstly, PICRUSt analysis of 16S rRNA sequencing data and untargeted metabolomic analysis showed that the metabolism-related pathways, especially amino acid metabolism, were significantly enriched in mice with probiotics treatment. In particular, D-glutamine and D-glutamate metabolism and glutamic acid were the most abundant altered amino acid-related pathways and metabolites. It has been reported that glutamine metabolism promotes effector T cell generation and function,[194]^45 and glutamic acid is a highly effective regulator in initiating T cell-mediated immune responses.[195]^46 Moreover, beta-alanine metabolism, arginine and proline metabolism, and glutathione metabolism pathway contain the common metabolite SPD, which also enhanced the IFN-γ production of CD8^+ T cells.[196]^33 Secondly, targeted metabolomics analysis identified the enrichment of SPD in feces and livers from BLE-treated SPF mice. Moreover, single administration of SPD led to similar effects as probiotics in inhibiting HBV replication as well as improving intestinal homeostasis and Th1 cell function. Importantly, BLE supplementation increased SPD content in feces, peripheral serum, and livers and promoted HBV clearance in GF mice. Previous studies reported that SPD can be produced by many other microorganisms, such as Prevotellaceae, Clostridiaceae, and Ruminococcaceae.[197]^47 It needs to be further investigated whether BLE-induced alteration of other SPD-producing strains also participates in HBV clearance in SPF mice. Accumulated data demonstrated the effect of SPD on immunity, while its immunomodulatory effect was context dependent.[198]^34^,[199]^37^,[200]^38 SPD shifts Th17 cell polarization toward Tregs, while it did not affect the production of IFN-γ of naive CD4^+ T cells under Th1-polarizing conditions.[201]^34 Matsumoto et al. reported increased IFN-γ and augmented fecal polyamine in probiotics-treated patients with autoimmunity.[202]^48 Our studies confirmed that SPD administration enhanced CD4^+ T cell function and promoted HBV clearance in vivo. SPD, a natural autophagy-promoting polyamine, exerts multiple functions, mainly through autophagy. It acts as the only known substrate for the hypusination eIF5A, which is essential for the synthesis of the autophagy transcription factor TFEB.[203]^38 In vitro studies showed that both autophagy inhibitor 3-MA and eIF5A-hypusine inhibitor GC7 significantly decrease SPD-induced IFN-γ accumulation in CD4^+ T cells. Therefore, our work revealed that the SPD-autophagy pathway enhanced IFN-γ^+CD4^+ T cell immunity. However, other metabolites, especially oligosaccharides (1-kestose, raffinose, maltotriose), which are used as prebiotics to regulate the immune system,[204]^49^,[205]^50 were also up-regulated after probiotics treatment. Therefore, more work is required to determine the key metabolism-related pathways and master metabolites contributing to HBV clearance. The seroclearance or seroconversion of HBsAg has been identified as the key marker for HBV cure in clinics. However, the spontaneous conversion rate of HBsAg is as low as less than 1% in the natural population[206]^7 and as high as no more than 30% even in patients with HBsAg levels <1,000 IU/mL.[207]^51 Notably, our preliminary observational study reported that probiotics accelerate the decline of serum HBsAg levels in patients receiving anti-HBV drugs. Importantly, 3 of 10 patients obtained the successful loss of serum HBsAg after the treatment with PegIFNα2b and tenofovir in combination with probiotics. Although the sample size is small, the exciting data still showed the great potential of probiotics in the loss of HBsAg. A combination of probiotics or SPD and antiviral treatment might be the potential therapeutic strategy for HBV functional cure. In conclusion, SPD derived from BLE effectively promotes HBV clearance, which is likely to associate with more effective IFN-γ^+CD4^+ Th1 cells. Our study has offered valuable insight into the immune mechanism of microbiota-mediated HBV clearance. Furthermore, it provides possible cues to use probiotics and SPD combination therapy with anti-HBV drugs in the clinical treatment of chronic HBV infection. Limitations of the study Our study has some limitations. We used mice with hydrodynamic injection of AAV/HBV1.2 as the mouse model to perform HBV clearance studies. Although this is a commonly used HBV model, hydrodynamic injection induces the release of damage-associated molecular patterns upon hepatocellular death. This process is in sharp contrast to the stealthy nature of HBV, which typically evades from the innate immune detection machinery during natural infections.[208]^52 Besides, although the exciting data have been obtained in human beings, perspective and randomized controlled trial is required to confirm the therapeutic potential of BLE as well as the identified metabolite in the context of HBV inhibition. Moreover, although both BLE and SPD contributed to the function of HBV-specific CD4^+ T cells, whether they could suppress other pathogens by modulating host immunity needs to be further verified. Resource availability Lead contact Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Chunhong Ma (machunhong@sdu.edu.cn). Materials availability All unique/stable reagents generated in this study are available from the [209]lead contact upon request and upon completion of Materials Transfer Agreement. Data and code availability * • 16S rRNA sequencing data in this study have been deposited in the National Center for Biotechnology Information (NCBI) database under BioProject accession number SRA: PRJNA700493. Metabolomics data in this study have been deposited in the National Metabolomics Data Repository (NMDR) under Study ID ST003536. * • This paper does not report original code. * • Any additional information required to reanalyze the data reported in this paper is available from the [210]lead contact upon request. Acknowledgments