Abstract Metabolism and gut microbiota are essential for newborn health, influencing immune function, energy balance, and growth. Breast milk provides IgA, crucial for shaping gut microbiota in infants. In non-breastfed newborns, we observe the presence of IgM antibodies that can recognize various bacteria, influence bacterial clustering, and alter bacterial metabolism, such as carbon source utilization in vitro within small bacterial communities. Based on these findings, we developed a monoclonal IgM, M291, derived from a newborn-B cell, which mimics naturally occurring antibodies and could serve as a surrogate tool to modulate intestinal bacterial functions and metabolism. Oral administration of M291 alters the metabolome of germ-free mice colonized with a defined bacterial consortium or an infant gut microbiota, by modulating the bacterial transcriptome, while maintaining microbial abundance and diversity. This study establishes proof of concept for the design and application of newborn-derived antibodies to modulate microbial and host metabolism, including lipid metabolism and bile acid secretion, without significantly altering microbiota composition. Subject terms: Mucosal immunology, Microbiome, Metagenomics, Antibodies __________________________________________________________________ Here, the authors characterize a newborn-derived monoclonal IgM, showing it influences bacterial clustering, gene expression, and metabolic activity, thereby modulating gut bacterial functions and host metabolism without altering microbiota composition. Introduction The intestinal metabolome, consisting of various low-molecular-weight metabolites including sugars, organic acids, and amino acids, is shaped by the combined effects of host metabolism and gut microbiota activities^[38]1. This metabolomic profile enhances microbiota’s functionality and represents the intricate interactions between host physiology and microbial metabolism. Consequently, the intestinal metabolome acts as a comprehensive biomarker, mirroring the gut ecosystem’s dynamic state^[39]2,[40]3. The initial microbial colonization in the gut is crucial for the development and maturation of the intestinal mucosa and the immune system in neonates^[41]4. Gut colonization starts at birth, with aerobic and facultative anaerobic bacteria such as Enterobacteria and Streptococci playing a pivotal role during this initial phase^[42]5. The consumption of oxygen by these pioneer microbes creates a microenvironment conducive to the successive colonization of obligate anaerobes, including Bifidobacterium, Bacteroides, and Clostridium^[43]4,[44]6. The establishment of the gut microbiota and metabolome is influenced by multiple factors, including mode of delivery and feeding practices^[45]7–[46]9. Microbiota utilize nutrients to generate energy and a myriad of metabolites that are essential for physiological processes. Early seeders are essential for the maintenance of health, as dysbiosis is implicated in a spectrum of diseases such as obesity, inflammatory bowel disease, and asthma across both infant and adult populations^[47]10–[48]12. Recent studies have documented that gut bacteria and metabolites play pivotal roles in the maintenance of homeostasis by regulating the gut-systemic metabolic interplay, which may influence immune and metabolic development in early life^[49]10,[50]13. Indeed, an alteration can impact cognitive development^[51]14, asthma^[52]15, or allergies^[53]16. Thus, a balanced gut microbiota is essential for metabolic functions, nutrient absorption, and immune responses, highlighting its significance in early-life development and long-term health. Immunoglobulins (Igs) have been found to coat a significant portion of the gut microbiota^[54]17,[55]18. These Ig secreted in the intestinal lumen play multifaceted roles, including elimination of microorganisms and facilitation of communication between the host and microbiota, thereby promoting homeostasis. Thus, Ig functions can be categorized into elimination, neutralization, colonization, and sculpting of the microbiota^[56]19–[57]22. The interaction of Igs with the microbiota is capable of modulating bacterial gene expression. For example, a monoclonal antibody (mAb) that targets a capsular polysaccharide antigen has been demonstrated to downregulate the expression of specific epitopes, consequently impairing bacterial fitness^[58]23. Furthermore, IgA antibodies have been shown to influence bacterial fitness through multiple mechanisms, such as inhibiting bacterial motility or impeding the uptake of essential sugars, with the specific effects being dependent on the surface components recognized by the respective IgA. This indicates that the immune system possesses the capacity to concurrently impact the commensal gut bacteria through diverse mechanisms^[59]24. IgA is the predominant isotype present at mucosal surfaces in mammals, including the small intestine, where it is actively secreted^[60]25,[61]26. In addition to IgA, IgM, albeit to a lesser extent, is also detected in the gut lumen^[62]18. Both are transported into the gut lumen via the polymeric Ig receptor, with IgM forming pentamers and IgA forming dimers, to coat microbiota^[63]27. At birth, IgA is undetectable in the meconium and takes several weeks to be synthesized and appear in stools^[64]28,[65]29. Studies show that serum IgM levels in newborns are much higher than IgA^[66]30. In breastfed infants, intestinal IgM-positive plasma cells are present at birth^[67]31. IgA detection in stool initiates between 2 to 4 weeks of age in full-term infants, mediated through both T cell-dependent and T cell-independent pathways^[68]32. However, this detection depends on mucosal tissue^[69]33. This developmental trajectory reflects the gradual maturation of mucosal immunity after birth. We recently identified a perinatal-specific population of B cells that produce antibodies capable of recognizing a broad spectrum of microbiota species^[70]34. In vitro, these antibodies modulated carbon source utilization and influenced the dynamics of bacterial communities^[71]34. In the present study, we characterized intestinal antibodies during the first month of life by analyzing their microbial recognition and their impact on bacterial behavior both individually and within communities. Our findings show that neonatal IgM antibodies can modulate bacterial clusters and influence microbial abundance. We developed a newborn B-cell-derived monoclonal IgM pentameric antibody and investigated its interactions with microbial consortia in vitro as well as in vivo. Notably, we show that a single monoclonal IgM can alter the in vivo metabolome of germ-free mice colonized with neonatal microbiota—not by changing the overall microbial community structure, but by modulating bacterial functions. This study opens the scope for using mAbs as a novel therapeutic tool for shaping the intestinal milieu at the metabolic level. Results Characterization of newborn intestinal Ig during the first month of life In the intestinal lumen of formula-fed neonates during the first month of life, IgM was found to be the predominant Ig, whereas IgA levels increased over time (Fig. [72]1A, Supplementary Fig. [73]1A). There was a notable shift in the ratio of intestinal Ig isotypes, from IgM predominance to a more balanced IgA presence. Specifically, IgM accounted for about 80% of total intestinal Igs in the first week, dropping to 50% by the fourth week (Fig. [74]1B). As gut colonization begins shortly after birth, we examined the binding affinity of intestinal Igs to gut bacteria throughout the first month. In the first two weeks, a significantly higher proportion of bacteria from neonatal fecal samples were coated with IgM than IgA (Fig. [75]1C, Supplementary Fig. [76]1B), aligning with the early IgM prevalence. By month’s end, IgA-coated bacteria became more prevalent, mirroring increased IgA secretion (Fig. [77]1B, [78]1C). Analyzing Ig binding to peptidoglycan (PGN) of Gram-positive bacteria and lipopolysaccharide (LPS) of Gram-negative bacteria, IgM showed stronger PGN recognition, while IgA recognized both PGN and LPS equally (Fig. [79]1D). Fig. 1. Characterization of Intestinal Secreted Neonatal Immunoglobulins During the First Month of Life. [80]Fig. 1 [81]Open in a new tab A Intestinal secreted IgA (blue) and IgM (red) levels were quantified over the first month of life using ELISA. B Weekly proportions of secreted IgA and IgM in total immunoglobulins within fecal samples were assessed. w1 (Days 1 to 7, n = 10), w2 (Days 8 to 14, n = 9), w3 (Days 15 to 21, n = 7), and w4 (Days 22 to 28, n = 7). C The frequency of bacteria-bound intestinal IgA and IgM was measured weekly by FACS. D Total secreted intestinal immunoglobulins (normalized to 9 µg/ml) were tested by ELISA for reactivity against PGN and LPS. Recognition of six primary bacterial colonizers by intestinal secreted immunoglobulins: E. coli (E) E. faecalis (F) S. epidermidis (G) S. salivarius (H) B. bifidum (I) and B. thetaiotaomicron (J). K Cumulative bacterial recognition summary of E–J. L Average recognition of Gram-positive versus Gram-negative bacteria is represented as a heat map. All data are presented as cumulative area under the curve (CAUC). Each point represents an individual biological sample (n = 4–10). Box-and-whisker plots: center line = median; box = 25th–75th percentiles; whiskers = 5th–95th percentiles (C–L). Statistical analyses were conducted according to the respective panels: p values were calculated with the two-sided Mann-Whitney U test for panels B–J. Significance levels are indicated as follows: *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001; n.s, not significant. Intestinal Igs are known to coat bacteria^[82]18. We subsequently evaluated the binding specificity of neonatal IgA and IgM to early-colonizing bacterial species, including Gram-positive (Enterococcus faecalis, Staphylococcus epidermidis, Streptococcus salivarius, Bifidobacteria bifidum) and Gram-negative bacteria (Escherichia coli, Bacteroidetes thetaiotaomicron) (Fig. [83]1E–J). Despite the observed differences in PGN and LPS binding between IgM and IgA (Fig. [84]1D), both Igs demonstrated comparable binding to these bacteria. The recognition of all bacterial species tested increases significantly after the second week (Fig. [85]1E–J), as demonstrated by the cumulative area under the curve (CAUC) (Fig. [86]1K). Furthermore, an analysis of IgM binding across 20 bacterial species reveals a progressive increase in bacterial recognition from week 1 to week 4 (Fig. [87]S1). Notably, strong recognition is observed early for C. perfringens and E. cloacae, followed by S. aureus and S. epidermidis—three of the four Gram-positive bacteria tested. By week 3, additional species, such as L. crispatus and E. faecium, are recognized. Interestingly, S. salivarius, B. thetaiotaomicron, and B. dorei consistently show low recognition. By week 4, only B. thetaiotaomicron remains at low recognition levels (Supplementary Fig. [88]1C). The average CAUC of bacterial recognition indicated that Gram-positive bacteria are generally more recognized than Gram-negative bacteria (Fig. [89]1L), aligning with the higher binding to PGN compared to LPS (Fig. [90]1D). During the first month of life, we concurrently analyzed the gut microbiota using 16S rRNA sequencing (Supplementary Fig. [91]1D–F). Our findings revealed a notable increase in the abundance of Firmicutes relative to Actinobacteria, while the proportion of Proteobacteria remained relatively stable at approximately 20% throughout this period (Supplementary Fig. [92]1D). From the second week, which coincided with a decline in Bifidobacteria (Supplementary Fig. [93]1E). Notably, we observed a higher prevalence of Gram-positive bacteria compared to Gram-negative bacteria (Supplementary Fig. [94]1F), which aligns with the increased capacity of intestinal immunoglobulins to recognize PGN over LPS, thereby enhancing the recognition of Gram-positive bacteria (Fig. [95]1D–L). These results demonstrate that IgM are the primary antibodies in the neonatal gut shortly after birth, with a preferential PGN recognition over LPS. Both intestinal IgA and IgM broadly recognized bacterial species throughout the first month of life. Newborn intestinal Igs affect bacterial community metabolism We subsequently evaluated the impact of early intestinal antibody isotypes on the neonatal bacterial community. We isolated secreted IgA (Nint_pA) and IgM (Nint_pM) from samples collected at week 3, and the binding capacity to PGN and LPS was assessed (Supplementary Fig. [96]2A, B). Notably, Nint_pM exhibited stronger binding to PGN, while Nint_pA showed an increased recognition of both bacterial antigens relative to Nint_pM (Supplementary Fig. [97]2A, B), indicating similar reactivity patterns between bulk and purified Igs. Further analysis of bacterial recognition across six selected species—E. coli, E. faecalis, S. epidermidis, S. salivarius, B. bifidum and B. thetaiotaomicron—reveals binding by both Nint_pA and Nint_pM, with Nint_pA showing a higher binding capacity (Supplementary Fig. [98]2C). To ascertain the influence of neonatal intestinal Igs on bacterial growth, we cultured individual bacterial strains in the presence of either Nint_pM (Fig. [99]2A) or Nint_pA (Fig. [100]2B). In the presence of either Igs, E. coli, E. faecalis, S. epidermidis, S. salivarius, and B. thetaiotaomicron display significantly enhanced growth compared to controls (Fig. [101]2A, B). Fig. 2. Impact of newborn intestinal immunoglobulins on bacterial clustering and microbial consortia. [102]Fig. 2 [103]Open in a new tab Indicated bacterial strains were cultured individually in AF medium in the presence of neonatal intestinal IgM (Nint_pM) (A) or IgA (Nint_pA) (B) at a concentration of 5 mg/ml. Growth was assessed by OD600nm and expressed as a percentage relative to the average of control cultures without immunoglobulins (PBS). C The MIC6 microbial community was incubated with Nint_pA or Nint_pM, and immunoglobulin-bound bacterial clustering was analyzed by FACS. D Supernatants from MIC6 cultured in the presence of Nint_pA or Nint_pM were used to culture individual bacteria. Growth was assessed based on OD600nm and expressed as a percentage of the MIC6 supernatant without immunoglobulins, serving as a reference control. E Utilization of indicated carbon sources by the MIC6 consortium for growth was analyzed using Eco-plates in the absence (gray) and presence of 5 mg/ml Nint_pM (red) or Nint_pA (blue). Box-and-whisker plots: center line = median; box = 25th–75th percentiles; whiskers = 5th–95th percentiles (A, B, D. Box plots represent median and maximum and minimum of variation (n = 3) E. Statistical analyses were conducted according to the respective panels: p values were calculated with the two-sided Mann-Whitney U test for A, B, and D, or the two-sided unpaired Student’s t-test for E. Significance levels are indicated by ****, ***, **, and * for p values < 0.0001, <0.001, <0.01, and <0.05, respectively; n.s = not significant. We investigated the capacity of dimeric IgA and pentameric IgM in the gut to form diverse bacterial clusters through cross-species binding. Bacteria were labeled with fluorescent amino acids, and clustering was analyzed by FACS in the presence or absence of Igs (Supplementary Fig. [104]2D–F). Clustering occurred without Igs, but pentameric Nint_pM enhanced multi-species clustering more than dimeric Nint_pA, which mainly formed smaller clusters (Fig. [105]2C). Nint_pM facilitated the formation of 3- to 4-member clusters involving E. coli, E. faecalis, S. epidermidis, and S. salivarius, while Nint_pA predominantly bound single species, especially E. faecalis, S. salivarius, and B. bifidum. Nint_pA induced two-species clusters with E. faecalis associating with S. epidermidis or S. salivarius. Notably, both Nint_pA and Nint_pM reduced clustering with B. bifidum and B. thetaiotaomicron (Fig. [106]2C). Bacterial clustering can influence microbial interactions, affecting growth, gene expression, and metabolic functions^[107]35,[108]36. To further investigate the effects of Nint_pA and Nint_pM, we cultured a microbial consortium of 6 bacterial species (MIC6) overnight with each isotypic antibody preparation. Neither IgA nor IgM impacted bacterial composition of MIC6 culture (Supplementary Fig. [109]2G). We then used supernatants from MIC6 cultures treated with IgA or IgM preparations to individually culture the 6 bacterial strains. Comparisons with untreated supernatant (sMIC_PBS) revealed that while B. thetaiotaomicron and S. salivarius growth remained stable, S. epidermidis growth decreased with sMIC_pA, and B. bifidum growth increased with sMIC_pM (Fig. [110]2D). Additionally, sMIC_pM significantly enhanced the growth of E.coli, E.faecalis, S.epidermidis, and B.bifidum compared to sMIC_pA. Subsequently, we examined if metabolism of MIC6 modulated by IgA or IgM could influence bacterial functioning through differential nutrient utilization. MIC6 carbon source usage was analyzed with or without Nint_pA or Nint_pM (Fig. [111]2E). Nint_pM increased glycogen (GLG) usage and decreased glucose-3-phosphate (G3P), Polysorbate 80 (T80), and γ-hydroxybutyrate acid (GHB) uptake compared to controls. Nint_pA only decreased G3P uptake (Fig. [112]2E). In summary, neonatal intestinal IgM, particularly the pentameric form, preferentially binds to peptidoglycan (PGN) and induces bacterial aggregation. Although purified IgM isotypes affected growth of individual bacterial cultures, this effect is not seen when bacteria are grown as part of a complex microbial community. However, community metabolism was influenced, impacting nutrient usage and secretion, thereby affecting bacterial growth. These results indicate a subtle, indirect influence of secretory intestinal antibodies on microbial community dynamics. In summary, we found that IgA and IgM are able to modulate individual bacterial cultures and modulate differentially bacterial metabolism. However, IgM have a stronger impact on bacterial aggregation and metabolism compared to IgA. Designing a newborn mAb to modulate bacterial functions We explored the impact of neonatal B cell-derived microbiota-reactive monoclonal mAbs, specifically BRG291-derived monomeric IgG (G291)^[113]34 and pentameric IgM (M291) (Supplementary Fig. [114]3A), on bacterial communities, focusing on their binding to bacterial surface antigens PGN (Fig. [115]3A) and LPS (Fig. [116]3B). M291 demonstrated robust PGN binding, exceeding that of the polyreactive ED38 reference mAb (Fig. [117]3A). Neither M291 nor G291 bound to LPS (Fig. [118]3B). A subsequent analysis of 20 bacterial species, selected for their representation of gut microbiota diversity, revealed that M291 recognized bacteria more broadly and more strongly than G291 (Fig. [119]3C–E, Supplementary Fig. [120]3B). These findings suggest that the pentameric M291 has enhanced bacterial recognition, especially for PGN, akin to neonatal intestinal secreted IgM. Fig. 3. Microbial reactivity of neonatal G291 and M291 mAbs. [121]Fig. 3 [122]Open in a new tab Monoclonal antibodies derived from BRG291, produced as monomeric IgG [G291] and pentameric IgM [M291], were evaluated by ELISA for their reactivity toward PGN (A) and LPS B. Included as controls were the mGO53 mAb (negative control) and the polyreactive ED38 mAb (positive control). C, D The same antibodies, G291 and M291, were assessed for their reactivity with six primary bacterial colonizers in the MIC6 consortium: E. coli, E. faecalis, S. epidermidis, S. salivarius, B. bifidum, and B. thetaiotaomicron. Results are displayed as concentration-dependent binding curves (A, B) or cumulative area under the curve (CAUC) values (C, D). E Comparative recognition of 17 bacterial strains by G291 and M291 assessed by ELISA. Results are shown as AUC calculated on concentration-dependent binding curves. We assessed effects of G291 and M291 on individual bacterial cultures and the MIC6 consortium. No significant growth differences were observed with G291 on single bacterial cultures (Fig. [123]4A), whereas M291 significantly enhanced the growth of E. faecalis, S. salivarius, B. bifidum, and B. thetaiotaomicron (Fig. [124]4B). Before analyzing the impact on the MIC6 consortium, we first examined bacterial cluster formation within the MIC6 community using FACS (Supplementary Fig. [125]4A and [126]4B). M291 or G291 did not significantly affect the overall bacterial clustering, with clusters of four bacteria observed under both conditions. However, a greater diversity among clusters containing at least two bacteria was obtained in the presence of M291. Specifically, among the 12 clusters composed of two different bacteria, 8 were enhanced with M291 treatment, compared to only 5 with G291 (Fig. [127]4C). Overnight culture of MIC6 with M291 showed a reduction in the number of S. epidermidis and B. thetaiotaomicron compared to the control and G291 (Supplementary Fig. [128]4C). When supernatants from the mAb treated MIC6 cultures were applied to single bacterial cultures, M291 supernatant inhibited the growth of E. coli, E. faecalis, S. epidermidis, S. salivarius and B. thetaiotaomicron, whereas the G291 supernatant had no effect compared to the control (Fig. [129]4D). Further evaluation revealed that G291 and M291 impacted carbon source utilization differently. Both antibodies significantly affect the usage of β-Methyl-D-Glucoside (βMDG), T80, and lactose (Lac), with M291 enhancing βMDG and T80 utilization over G291, though lactose was unaffected. M291, but not G291, significantly impacted L-Arginine (L-Arg) and GHB utilization, while G291 uniquely enhanced N-acetylglucosamine (GlcNac) usage (Fig. [130]4E). Fig. 4. Comparative effect of the pentameric and monomeric forms of a neonatal monoclonal antibody on bacterial clustering and carbon source utilization in vitro. [131]Fig. 4 [132]Open in a new tab Indicated bacterial strains were cultured individually in AF medium with 5 mg/ml of G291 (A) or M291 (B). Growth was measured via OD600 nm and expressed as a percentage relative to controls without immunoglobulins. C MIC6 was incubated with G291 or M291 (5 mg/ml), and bacterial complexes were analyzed by FACS. Changes relative to MIC6 without antibodies are displayed in a heat map. D MIC6 culture supernatants from incubations with G291 or M291 were used to grow individual bacteria. Growth was calculated as a percentage of the control (supernatant without immunoglobulin; gray) based on OD600 nm. E Carbon source utilization by MIC6 was assessed with Eco-plates, in the absence (gray) and presence of G291 (green) and M291 (orange), done in triplicates. A, B and D Each point represents an individual technical replicates of one of 3 to 4 biological samples (n = 6-12). Box-and-whisker plots: center line = median; box = 25th–75th percentiles; whiskers = 5th–95th percentiles. E Box plots represent the median, as well as the minimum and maximum values (n = 3). Statistical analyses were conducted according to the respective panels: p values were calculated with the two-sided Mann-Whitney U test for A, B, and D, or the two-sided unpaired Student’s t-test for E. Significance levels are indicated by ****, ***, **, and * for p values < 0.0001, <0.001, <0.01, and <0.05, respectively; n.s = not significant. Given that complement components in the intestinal lumen can be mobilized with or without antibody action to target bacteria^[133]37,[134]38, we evaluated the combined effect of complement with M291 on the MIC6 bacterial community (Supplementary Fig. [135]4D and [136]4E). While bacterial CFU remained unchanged (Supplementary Fig. [137]4D), carbon source utilization was altered, with enhanced usage of α-cyclodextrin (α-CD), βMDG, L-Arg, GHB, and decreased GLG usage (Supplementary Fig. [138]4E). These findings indicate that a pentameric mAb, like M291, had a greater influence over bacterial community behavior, affecting growth, carbon source utilization, and metabolite production, compared to its monomeric counterpart. Influence of M291 treatment on MIC6 colonization and metabolome To investigate the in vivo effects of M291 on gut colonization, we colonized germ-free (GF) mice with the MIC6 bacterial consortium. Mice were orally inoculated with MIC6, with a booster dose given on day 3. Colonization was confirmed on days 7 and 10 using quantitative PCR (qPCR), and the mice were euthanized 11 days after the initial gavage (Fig. [139]5A). Experimental design included, one control group of GF mice without MIC6 (GF_UC), one group receiving the MIC6 consortium without treatment (GF_Co_UN), and one group treated with M291 via drinking water, both before and after MIC6 administration (GF_Co_M291) (Fig. [140]5A). By day 7 post-colonization, mice were predominantly colonized by B. bifidum and B. thetaiotaomicron, followed by S. epidermidis, E. faecalis, and S. salivarius, with E. coli remaining less prevalent (Fig. [141]5B). This bacterial hierarchy was maintained at day 10. Following M291 treatment, the overall hierarchical distribution among species was preserved at both day 7 and day 10 (Fig. [142]5B), with a stable relative abundance across bacterial communities at day 7 (Supplementary Fig. [143]5A). However, M291 treatment significantly reduced E.coli and B. thetaiotaomicron at day 7, and B. bifidum at day 10 (Fig. [144]5B). Altogether, these data suggest that the MIC6 colonization can be slightly modulated by M291 treatment. Fig. 5. Effect of M291 treatment on the metabolome of MIC6 colonized GF mice. [145]Fig. 5 [146]Open in a new tab A Schematic representation of the experimental design. GF mice were either left uncolonized (GF_UC) or colonized with the MIC6 bacterial consortium, comprising E. coli, E. faecalis, S. epidermidis, S. salivarius, B. bifidum, and B. thetaiotaomicron. Colonized mice were divided into two groups: untreated (GF_Co_UN) or treated with M291 administered via drinking water (GF_Co_M291). B Fecal samples were collected, and bacterial growth was assessed by qPCR at day 7 (left panel) and day 10 (right panel). Each point represents an individual technical replicates of one of 4 biological replicates (n = 12). Box-and-whisker plots: center line = median; box = 25th–75th percentiles; whiskers = 5th–95th percentiles. C Differential metabolite profiles among GF_UC (green), GF_Co_UN (blue), and GF_Co_M291 (red) were visualized using a circular heat map (left panel) and PCA (right panel). D–H Metabolite profile analysis of GF_Co_UN and GF_Co_M291 groups. D Volcano plot comparing metabolites highlighting upregulated metabolites in red and downregulated metabolites in green. E Heat map of the 50 most significantly altered metabolites. F Relative distribution of metabolite superclasses. G Ontology of significantly upregulated metabolites within the lipids and lipid-like molecules superclass in GF_Co_M291. H Dynamic Autocorrelation (DA) score of pathways related to secondary metabolites. Statistical analyses were calculated with the two-sided Mann-Whitney U test. Significance levels are indicated by **, and * for p values < 0.01, and <0.05, respectively. We next analyzed the metabolites induced by MIC6 colonization. We identified nearly 1, 000 metabolites, revealing distinct metabolite profiles that enabled clear differentiation among the three groups (Fig. [147]5C). Following bacterial colonization, 609 metabolites showed significant differences compared to GF mice (Supplementary Fig. [148]5B), with 308 upregulated and distributed across various classes, in particular lipids and organic acids (Supplementary Fig. [149]5C). MIC6 bacterial colonization primarily triggered metabolic processes at the cellular level and within the digestive system, indicating an overall boost in intestinal activity (Supplementary Fig. [150]5D). M291 treatment induced a significant response, resulting in 490 metabolites, with 307 exhibiting significant differences (Fig. [151]5D). Metabolite variety is further illustrated among the top 50 metabolites, showing greater ontological diversity for M291 treatment group compared to the untreated group (Fig. [152]5E). Metabolites are distributed across various classes, with two major super classes, organic acids (n = 148) and lipid-related molecules (n = 141) supporting cellular growth, energy production, tissue remodeling, and immune system maturation, making them vital for healthy growth (Fig. [153]5F). Among lipids, fatty acyls were heavily promoted as they are key in energy storage or in hormone synthesis. Notably, steroids and their derivatives were found to be predominantly induced upon M291 treatment, which are essential for healthy development and functioning (Fig. [154]5G). A detailed analysis of the secondary metabolic pathways revealed a general upregulation in response to M291 treatment, except pyruvate metabolism pathway (Fig. [155]5H). Interestingly, bile acids (BAs) secretion was induced by M291 treatment (Fig. [156]5H and Supplementary Fig. [157]5E). Collectively, these results demonstrate the distinct impact of M291 on the interplay between host and bacterial metabolisms within a well-defined bacterial consortium. Importantly, M291 treatment influenced the metabolome without disrupting the microbial community structure. Metabolites produced in M291-treated MIC6 colonized mice can affect bacterial functions To evaluate the overall impact of metabolomic changes on bacterial growth, we cultured individual bacterial strains with cecal content from GF mice. Growth rates were calculated relative to the GF_UC baseline. Application of the GF_Co_UN metabolome significantly enhanced the growth of B. bifidum, S. epidermidis, E. faecalis, and E. coli compared to GF_Co_M291 (Fig. [158]6A). These variations may reflect differences in metabolic pathways, where 27 pathways were activated with M291 treatment (Fig. [159]5H). This overall enrichment of pathways may indicate an elevated metabolic activity that limits nutrient or secondary metabolite availability for bacterial growth. The metabolic effects of cecal content were dependent on bacterial concentration and the quantity of cecal content. Increasing the quantity of cecal content used to culture bacteria significantly influenced the growth of B. bifidum, S. epidermidis, E. faecalis, E. coli, and B. thetaiotaomicron (Supplementary Fig. [160]6A), suggesting that metabolites at lower concentrations can impact B. thetaiotaomicron growth. Additionally, when reducing initial bacterial load, growth of E. coli and S. salivarius decreased significantly (Supplementary Fig. [161]6B). Interestingly, lower metabolite concentrations had no impact on E. coli growth but did reduce the growth potential of S. salivarius (Supplementary Fig. [162]6C). When testing metabolite effects in our MIC6 setup, we did not observe any significant changes in growth dynamics induced by the GF_Co_UN metabolic content compared to the one of GF_UC group. However, GF_Co_M291 significantly enhanced the growth of E. coli, E. faecalis, and B. thetaiotaomicron (Fig. [163]6B). Cecal metabolomic changes, particularly those induced by M291 treatment, significantly influenced the growth of specific gut bacterial species, highlighting the complex interplay between host metabolism and gut microbiota. These findings suggest that targeted metabolic interventions could be used to modulate gut microbial community functions, potentially offering benefits for various gastrointestinal conditions. Fig. 6. Impact of metabolome from mAb treated colonized GF mice on in vitro bacterial culture. [164]Fig. 6 [165]Open in a new tab Indicated bacterial strains were cultured individually (A) or as the MIC6 consortium (B) in 1 mL of cecal metabolite content derived from GF_Co_M291, GF_Co_UN, or GF_UC. Growth was assessed by measuring the OD at 600 nm and expressed as a percentage relative to the control (GF_UC), represented by the dashed line. A, B Each point represents an individual technical replicates of one of 4 biological replicates (n = 12). Box-and-whisker plots: center line = median; box = 25th–75th percentiles; whiskers = 5th–95th percentiles. Statistical analyses were conducted using the two-sided Mann-Whitney U test. Significance levels are indicated as follows: **** for p values < 0.0001, *** for p values < 0.001, ** for p values < 0.01, and * for p values < 0.05. Influence of M291 treatment on newborn FMT and associated metabolome Using microbiota from non-breastfed newborns, we performed a fecal microbiota transplant (FMT) in GF mice, which were then treated with either M291 (GF_FMT_M291) or Nint_pM (GF_FMT_NintpM) as previously described, and compared them to untreated control (GF_FMT_UN) and uncolonized (GF_UC) groups (Fig. [166]7A). We conducted an analysis of FMT sequencing and the gut microbiota composition of colonized GF mice (GF_FMT). Bacterial composition was severely modified at the phylum, family and genera levels upon colonization (Fig. [167]7B–D, Supplementary [168]7A, B) reflecting a heavy loss in diversity. Among all GF_FMT samples, seven species accounted for up to 99.9 % of the total detected bacteria, with E. faecalis, K. aerogenes, and C. butyricum emerging as the predominant species while C. paraputrificum, B. breve, E. fergusonii and K. pneumoniae were present at lower abundance (Fig. [169]7D, Supplementary Fig. [170]7A). Evenness was higher in GF_FMT_NintpM compared to GF_FMT_UN or GF_FMT_M291 at day 3 suggesting uniformity in species abundance, that no single species overwhelmingly dominates the community. This significant difference diminishes by day 7 and is no longer observed by day 10 (Supplementary Fig. [171]7B). The global analysis revealed a distinct separation among the three groups on day 3, which gradually converged as the colonization process progressed (Supplementary Fig. [172]7C). Fig. 7. Bacterial abundance and gene expression in GF mice following FMT colonization. [173]Fig. 7 [174]Open in a new tab A Experimental design procedure for GF mice, left uncolonized (GF_UC), or colonized through Fecal Matter Transfer (FMT) under three conditions: without treatment (GF_FMT_UN), under M291 treatment (GF_FMT_M291), and under Nint_pM treatment (GF_FMT_NintpM). B Relative abundance of phyla, C and genera of bacteria identified in the FMT input and in GF mice after receiving FMT samples. D Relative abundance of main bacterial species identified across all conditions post-colonization. Metatranscriptomic analysis performed at Day 10. Heat maps of taxonomic analysis (E) and differentially expressing genes (DEG) identified (F) of input (green), GF_FMT_UN (blue) and GF_FMT_M291 (red). G Pathways identified from differentially expressing bacterial genes in untreated vs. M291 treated mice represented as donuts corresponding to major KEGG pathway process (upper panel) and gene classes (lower panel) involved in Metabolism pathways. Monitoring of microbial colonization over the experimental period showed a clear dominance of C. butyricum, K. aerogenes, and E. faecalis, which together constituted 98–99% of the population by day 7. Meanwhile, lower-abundance species, including C. paraputrificum, B. breve, and E. fergusonii displayed increasing colonization over time. By day 10, E. fergusonii and K. pneumoniae showed a tendency to decline (Supplementary Fig. [175]7D). Following antibody treatment, colonization of C. butyricum, K. aerogenes, and E. faecalis remained unaffected. However, B. breve and K. pneumoniae exhibited trends opposite to those observed in GF_FMT_UN-colonized mice, with B. breve decreasing by day 10, while K. pneumoniae increased (Supplementary Fig. [176]7D). Nevertheless, no significant changes in species relative abundance were observed. As shown by bacterial gene sequencing analysis, the bacterial composition at the genus level in FMT samples differs from that in GF_FMT_UN. However, no significant differences were observed between GF_FMT_UN and GF_FMT_M291, as evidenced by the lack of clustering between these groups (Fig. [177]7E). Metatranscriptomic analysis revealed differential expression of 348 bacterial genes (Fig. [178]7F, Supplementary Fig. [179]7E). FMT samples were clearly distinct from GF_FMT_UN, indicating active bacterial metabolism. In contrast, GF_FMT_M291 samples exhibited a distinct pattern of gene regulation upon M291 treatment (Fig. [180]7F). Collectively, these data demonstrate that M291 treatment does not alter the overall bacterial species composition at the community level but significantly affects gene expression. Notably, of the 348 differentially expressed genes, 264 are involved in metabolic pathways, with 93 genes implicated in energy or carbohydrate metabolism (Fig. [181]7G). These findings highlight the modulation by M291 of bacterial functions, particularly those related to metabolic activity and carbohydrate utilization. We therefore performed metabolite analysis of cecal content, revealing distinct clustering patterns between the four different groups (Fig. [182]8A). We analyzed the effects of the metabolome from GF colonized mice on bacterial growth in vitro. MIC6 bacteria were cultured individually or as a consortium (Supplementary Fig. [183]8A–C). Cecal content from uncolonized GF mice served as a control for bacterial growth. Notably, E. faecalis growth was enhanced only in the GF_FMT_UN group, while treatments with GF_FMT_M291 and GF_FMT_NintpM did not significantly affect its growth. M291 treatment promoted the growth of B. thetaiotaomicron while inhibiting E. coli growth, whereas Nint_pM treatment specifically enhanced E. coli growth (Supplementary Fig. [184]8A). The impact of cecal content on bacterial growth was concentration-dependent. (Supplementary Fig. [185]8B). Furthermore, the effects of the metabolome were tested using the entire MIC6 community. Interestingly, S. epidermidis growth remained unaffected, whereas E. coli, E. faecalis, S. salivarius, B. bifidum, and B. thetaiotaomicron were all significantly impacted by GF_FMT_M291 and GF_FMT_NintpM treatments, which notably decreased their growth (Supplementary Fig. [186]8C). Additionally, M291 treatment associated metabolome altered the carbohydrate usage profile within the bacterial consortium, with GF_FMT_M291 exhibiting a more diversified carbohydrate utilization compared to GF_FMT_UN (Supplementary Fig. [187]8D). Taken together, our findings suggest that microbiota-targeting antibody treatments have a modest influence on bacterial composition in vivo. However, the associated evenness is linked to more substantial changes in the metabolome, as evidenced by its significant impact on the growth of bacterial species. Fig. 8. Differential impact of FMT and M291 on the metabolome of colonized germ-free mice. [188]Fig. 8 [189]Open in a new tab A Metabolomic analysis across GF_UC (light blue), GF_FMT_UN (green), GF_FMT_M291 (dark blue), and GF_FMT_NintpM (red) groups as defined in Fig. [190]7A, displayed as a circular HMP and a PCA plot. B Volcano plot comparing metabolites between GF_FMT_UN and GF_FMT_M291, with upregulated and downregulated metabolites. C Heat map highlighting the 50 most significant metabolites in the comparison of GF_FMT_UN and GF_FMT_M291. D Relative distribution of metabolite superclasses in GF_FMT_M291 and GF_FMT_UN. E Ontology of significantly upregulated metabolites within the lipids and lipid-like molecules superclass in GF_FMT_M291. F Pathway impact and statistical significance of the metabolic pathways identified through pathway enrichment analysis of GF_FMT_M291 and GF_FMT_U. Note that the y-axis shows the negative logarithm of p-values;,therefore, pathways with higher statistical significance are plotted higher in the graph. G Metabolites of linoleic acid metabolism pathway analysis LA (Linoleic Acid); γ-LA(gamma Linoleic Acid); AA (Arachidonic Acid). Each point represents a biological replicates (n = 4). Box-and-whisker plots: center line = median; box = 25th–75th percentiles; whiskers = 5th–95th percentiles. Statistical analyses were conducted using the two-sided Mann-Whitney U test. Significance levels are indicated as follows: ** and * for p values < 0.01, and <0.05, respectively; n.s.= not significant. ** for p values = 0.0079, and * for p values = 0.0159. Following FMT colonization, 1360 metabolites were identified in GF mice, of which 510 were differentially expressed, with 148 metabolites being upregulated as a result of the FMT colonization (Supplementary Fig. [191]8E). We analyzed the metabolome of the FMT colonized mice. Among these 510 metabolites induced in GF_FMT_UN, 182 were classified as lipids and lipid-like molecules and 83 organic acids and derivatives (Supplementary Fig. [192]8F). These metabolites primarily influence intestinal and cellular functions, with notable activation in the biosynthesis of secondary metabolites and amino acid metabolism, signaling an active intestinal microbiome (Supplementary Fig. [193]8G). Interestingly, GF mice colonized with distinct pioneer microbiota shared 349 metabolites, with lipids and lipid-like molecules constituting approximately one-third of the common metabolites (Supplementary Fig. [194]8H). Among this superclass, half were fatty acyls, and a quarter were steroids and derivatives (Supplementary Fig. [195]8H). Treatment with M291 led to the differential expression of 613 metabolites between the GF_FMT_UN and GF_FMT_M291 groups, with 202 metabolites upregulated in the presence of M291 (Fig. [196]8B). Among the top 50 differentially expressed compounds, metabolite diversity was notably greater in the absence of M291 (Fig. [197]8C). Analysis of these 613 metabolites indicated that lipids and lipid-like molecules, along with organoheterocyclic compounds, remained the dominant superclasses, with lignans, neolignans, and related compounds emerging as the third largest superclass (Fig. [198]8D). Ontology analysis within these superclasses highlighted a significant representation of prenol lipids and steroids among lipid and lipid-like molecules (Fig. [199]8E). Upon analyzing the secondary class pathways following M291 treatment, we observed the up-regulation of 17 pathways, with 2 remaining unchanged and 11 being down-regulated. Among the down-regulated pathways, we focused on the linoleic acid (LA) metabolism (Fig. [200]8F), which is significantly influenced by various bacterial genera within the gut, such as Bifidobacteria and Roseburia^[201]39,[202]40. LA metabolites significantly influence gut health, immunity, and host physiology. LA is a precursor to arachidonic acid (AA), which undergoes metabolism via three primary enzymatic routes: cyclooxygenase, lipoxygenase, and cytochrome P450 pathways. These pathways generate bioactive lipids—prostaglandins, leukotrienes, and epoxyeicosatrienoic acids—that are essential for cell differentiation, tissue development, organ function, and disease pathogenesis^[203]41. Upon FMT colonization, we observed a decrease in LA levels. This reduction in LA directly impacts the entire metabolic pathway, extending from LA to AA. Notably, the downstream metabolites derived from AA metabolism exhibit differential alterations. Specifically, while prostaglandins and leukotrienes remain unchanged, the levels of 14(15)-epoxyeicosatrienoic acid (14(15)EpETE) are decreased (Fig. [204]8G). Upon treatment with M291, there is a significant enhancement in the downregulation of the LA metabolic pathway, extending from LA to AA metabolites (Fig. [205]8G). Concurrently, while 14(15)EpETE levels remain unchanged, there is an upregulation of leukotriene production and a downregulation of prostaglandin synthesis (Fig. [206]8G). Collectively, the observation underscores the intricate relationship between LA metabolism and the modulation of gut health and immune responses following M291 treatment and FMT colonization. Metabolomic analysis indicates that M291 treatment significantly alters the metabolic profile, of the gut particularly enhancing lipid metabolism. The treatment also differentially impacts the growth of specific bacterial species within the MIC6 community, suggesting a complex interplay between bacterial growth and metabolic activity. These findings underscore the potential of targeted metabolic interventions in modulating gut microbiota functions, with implications for therapeutic strategies in gut-related disorders. Discussion This study provides novel insights into the role of neonatal IgM antibodies with broad microbial reactivities in modulating the interplay between gut microbiota and host metabolism. We developed the pentameric M291 mAb that exerts distinct effects on bacterial growth and functions, metabolic pathways, without significantly disrupting the microbial community structure. This suggests its therapeutic potential as a modulator of gut metabolism, with implications for early-life development and long-term health outcomes. Upon birth, neonates are exposed to microorganisms that initiate the colonization. The balance between pathogen defense and minimal inflammation is critical for establishing symbiotic relationships with commensal bacteria. We characterized gut-secreted antibodies, their binding capacity to various microbial species and their potential influence on gut colonization^[207]42. Secretory Igs production correlates with the proportion of coated bacteria in the neonatal gut and their ability to recognize bacterial antigens and species throughout the first month of life, with a clear preference for Gram-positive bacteria. Such coated bacteria form clusters that may prevent direct bacterial interaction with epithelial cells, thereby protecting the host; modulating bacterial gene expression in bacteria such as B. thetaiotaomicron; and potentially promoting microbial community homeostasis through effects on motility, bacterial fitness, or bacteriophage susceptibility^[208]43. IgM predominates in the gut immediately after birth, in agreement a late IgA production^[209]28,[210]29. Our previous work demonstrated that pools of IgG mAbs derived from newborn B cells can modulate the metabolic activities of synthetic microbial communities in in vitro bacterial cultures^[211]34. The binding properties of M291 to PGN and its enhanced bacterial recognition compared to its monomeric counterpart (G291) underscore the importance of polymeric antibody structures in mediating host-microbiota interactions. M291 promoted bacterial cluster formation and impacted bacterial growth within the MIC6 community, indicating that multivalency of IgM antibodies is well-suited to modulate complex microbial consortia. Nevertheless, the lack of a disease model in this study restricts our ability to extrapolate these findings to pathological contexts where such interventions might be most needed. Recently, a mouse monoclonal IgA (W27) has been demonstrated to modulate gut microbiota by selectively inhibiting the growth of certain bacteria, such as E.coli, without affecting beneficial species like L. casei. This indicates that IgA has a regulatory role in gut microbial balance and health^[212]44. Our study shows that M291 treatment influenced bacterial growth and metabolism. Cecal content from M291-treated mice, when added to cultures, especially affected growth of B. bifidum and B. thetaiotaomicron in relation with carbon source availability. For example, growth of B. thetaiotaomicron is inhibited by B. breve in the presence of glucose but not affected by B. longum^[213]45. B. fragilis supports B. longum survival and changes organic acid production in co-culture. Bifidobacterium can utilize complex carbohydrates like human milk oligosaccharides and plant-derived glycans through transport systems, degrading them into simpler sugars for fermentation. This metabolic strategy supports cross-feeding interactions among Bifidobacterium strains and other gut bacterial spp., processes modulated by host dietary inputs and critical for maintaining gut homeostasis^[214]46. In addition, Bifidobacteria can compete for nutrients, which can help reduce their bacterial growth and colonization^[215]47. This competition can also lead to changes in the composition of the gut microbiome, favoring the growth of beneficial bacteria like Bacteroidetes^[216]47. Our findings suggest that the metabolites produced in response to M291 treatment serve as modulatory factors, shaping bacterial physiology and interspecies interactions. Utilizing a GF mouse model, we demonstrated the impact of mAb treatment on the metabolome while bacterial composition remained similar between groups. With two distinct consortia, we observed a differential impact of mAb treatment on the metabolome, adjusted according to the bacterial species used as colonizers of GF mice. Using a well-defined community composed of E. coli, E. faecalis, S. salivarius, S. epidermidis, B. bifidum, and B. thetaiotaomicron, we identified a significant number of metabolites and pathways that are modulated. Lipid and protein metabolism were upregulated in the presence of mAb, indicating a stimulation of the intestinal tract involving both host and microbiota. We observed a substantial upregulation of the BAs secretion pathway, crucial for digestion and absorption of dietary fats and fat-soluble vitamins, as well as regulation of cholesterol and glucose metabolism, contributing to overall nutritional health^[217]48. BAs are crucial for digesting fats, absorbing vitamins, and shaping the gut microbiota, impacting long-term host health by regulating bacterial growth and gut health^[218]49,[219]50. Effect of M291 on lipid-related molecules like prenol lipids and steroids shows it can adjust the interaction between the host and microbes’ metabolisms. M291 also affected LA metabolism, which is important for gut health. It lowered LA levels and changed the AA pathway. AA is a key fatty acid that helps with gut health by producing substances like prostaglandins and leukotrienes, which control inflammation and immune reactions^[220]51. Gut bacteria can change AA into substances that affect gut health^[221]52. The modulation of lipid mediators, particularly by M291 treatment, could have a complex impact on gut homeostasis and immune responses. This suggests that LA levels can influence the stress response and metabolic activity of certain gut bacteria. A reduction in LA levels, has been shown to induce metabolic stress in intestinal bacteria like B. breve, affecting their metabolic capabilities and growth rate^[222]39. LA metabolites, found in fermentation pellets, indicate their incorporation into bacterial cells, implying that LA levels could alter the composition and functionality of the gut microbiota^[223]39. Decreased prostaglandin A1 in gut metabolism can significantly affect gastrointestinal protection, inflammation, and immune responses, while the upregulation of leukotriene D4 is associated with exacerbated effects. These findings indicate that fecal FMT and M291 treatment profoundly influence gut homeostasis and immune responses, highlighting the intricate interplay between dietary lipid metabolism and gut microbiota in modulating host health and disease progression. M291 treatment significantly alters the metabolic profile of the gut, enhancing lipid metabolism and differentially impacting the growth of specific bacterial species within the MIC6 community, underscoring the potential of targeted metabolic interventions in modulating gut microbiota functions with implications for therapeutic strategies in gut-related disorders. These metabolites are important for immune control and keeping tissues healthy, so effect of M291 on LA metabolism might be important for gut health and overall immunity^[224]53. The relevance of our findings is underscored by the analysis of metabolites generated from consortia comprising bacteria not typically associated with FMT. This is illustrated by the effect of metabolites from M291-treated GF mice on the utilization of carbon sources by the MIC6 community. This shows that M291 treatment can influence the metabolism of bacteria, directly or indirectly, through metabolite interactions. Metatranscriptomic analysis demonstrates a change in bacterial metabolism, due to a differential bacterial gene expression upon treatment. A capsular polysaccharide Ab has been shown to change the gene expression of S. pneumoniae during colonization, affecting bacterial fitness^[225]54. These adaptations could affect the microbial ecosystem’s dynamics, showing the complex relationship between metabolites, bacterial physiology, and community function. Our results show that antibodies can change how the gut microbiota works, as seen by increased activity in metabolic pathways, including those for lipids and proteins, when treated with mAb. The activation of BAs secretion pathways also shows the complex interaction between the host’s factors and microbial metabolism, which is important for nutrient absorption and health. Mechanisms involve microbiota-dependent changes in bile acid metabolism, affecting signaling through bile acid receptors such as FXR and TGR5^[226]55. These changes influence lipid metabolism by altering bile acid recycling and enterohepatic signaling pathways. Adaptive responses to dietary lipids are regulated by epithelial processes involved in both digestion and absorption^[227]56,[228]57. The mechanisms underlying these interactions appear to involve bacteria-derived small molecules that upregulate lipid absorption genes in small intestinal epithelial cells. L-lactate from L. paracasei promotes lipid storage by inhibiting beta-oxidation, while acetate from E. coli enhances lipid oxidation, highlighting their roles as key regulators of enterocyte lipid metabolism^[229]58. Duodenal bacterial isolates decrease lipid absorption in both cultured enterocytes and mice, providing a mechanism by which they may exacerbate environmental enteric dysfunction and stunting in children^[230]59. Together, these insights highlight the central role of regulating lipid metabolism and its broader implications for addressing overnutrition, undernutrition, and related disorders. Monoclonals have greatly developed highlighting their therapeutic applications across various diseases, including cancer and infectious diseases^[231]60,[232]61 and here we present a new path, a new field of use that influences metabolism. We demonstrate the potential of neonatal-derived antibodies as tools for therapeutic intervention in the gut. By modulating specific bacterial behaviors and metabolic pathways, mAb like M291 offer a targeted approach to restoring or enhancing gut homeostasis. This approach is particularly relevant in the context of microbiota gut colonization, where interventions to prevent dysbiosis could have lifelong health benefits. The ability of M291 to selectively influence metabolomic profiles without disrupting microbial diversity further underscores its potential for safe and effective modulation of the gut environment. However, because only a single monoclonal antibody (M291) was evaluated, we cannot yet generalize these observations to other antibody clones or isotypes, and systematic screens will be required to define structure–function relationships across the broader neonatal antibody repertoire. Therefore, antibodies targeting microbiota act as modulators of microbial metabolism, affecting bacterial dynamics and ecosystem balance. Future studies should explore the applicability of such interventions in models of neonates, dysbiosis, disease, as well as the potential synergistic effects of combining mAb treatments with dietary or probiotic strategies. Methods Inclusion and ethics statements This research follows the guidelines of inclusion and ethics by Nature Communications. All authors have fulfilled the authorship criteria. Preparation of newborn fecal samples Fecal samples for intestinal IgM/IgA analysis were obtained from formula-fed healthy newborns during the first month of life with the written consent of the parents under the approval No. 2022-IPID-01. Fecal samples were resuspended at 100 mg/ml in cold sterile PBS and homogenized by pipetting and vortexing. Fibers were spun down at 500 × g for 5 min. Supernatants were collected, filtered through 100 μm, and spun down at 10,000 × g or 10 min. Supernantants were collected, filtered at 0.22 μm, and directly frozen at −80 °C (fecal water). The bacterial pellets were washed twice with cold PBS and resuspended in 30% sterile glycerol at a concentration of 1 g/mL prior to storage at −80 °C and use for FMT germ-free mouse experiments. Newborn intestinal Ig purification Newborn fecal samples with an IgA:IgM ratio of 1:2 from three samples of Week 2 and Week 3 (days 12, 15 and 19) were pooled, and IgA were purified by affinity chromatography using Peptide M agarose (gel-pdm-5) stack into Poly-Prep® Chromatography Columns (7311550; Bio-Rad). After IgA purification, the flow-through from the Peptide M agarose column was used for IgM precipitation. An equal volume of saturated ammonium sulfate solution was added to the flow-through to achieve a final concentration of 50% ammonium sulfate. The mixture was incubated overnight at 4 °C with continuous stirring. The precipitate was collected by centrifugation at 10,000 × g for 30 min, and the resulting pellet was resuspended in PBS. The resuspended pellet was dialyzed against PBS using a dialysis bag with a molecular weight cut-off (MWCO) of 100 kDa overnight at 4 °C. The purity of IgA and IgM was assessed by cross-ELISA (IgM detection on purified IgA fraction; IgA detection on purified IgM fraction) reaching 99% and 93%, respectively. ELISA Bacterial recognition was measured by ELISA as previously described^[233]62. Briefly, high-binding 96-well ELISA plates were coated overnight with 250 ng/well of purified LPS (L2637-10MG; Sigma-Aldrich), PGN (69554-10MG; Sigma-Aldrich), or with PFA 0.2% fixed bacteria on precoated Poly-L-lysine (3ug/ml) plates. After blocking and washing steps with 0.001% Tween 20-PBS, mAbs or purified intestinal antibodies were adjusted and tested at 4 μg/ml and 3 consecutive 1:3 dilutions in PBS. ELISA plates were developed using goat HRP-conjugated anti-human IgG, IgM, or IgA antibodies. Results are given as calculated AUC from titration curves in the graphs. ED38 and mGO53 mAb were used as polyreactive and non-polyreactive controls, respectively. Results were calculated as the area under the curve (AUC) from titration curves. First, the curve was created by plotting the ERL data over time. Then, AUCs were divided into multiple small trapezoid areas, which are measured individually, using the trapezoid rule [area = ½ (base a + base b) x height], and added up to get the total AUC. Production of G291 and M291 monoclonal antibodies BRG291 sequences were selected from our previous study^[234]34 to produce monomeric IgG1 and pentameric IgM forms. Recombinant antibodies were produced on selected sequences cloned into human Igγ1- Igλ-expressing vectors as previously. Recombinant G291 (IgG1) was produced by transient co-transfection of adherent HEK 293 T using PEI- precipitation method as previously described^[235]34, and purified by affinity chromatography using Protein G Sepharose® 4 Fast Flow (Cytiva). Purified antibodies were dialyzed against PBS^[236]34. M291 was produced by co-transfection with J chain-containing plasmid of Freestyle™ 293-F suspension cells. M291 was purified using size-exclusion chromatography with multi-angle light scattering (SEC-MALS). The molecular weight was confirmed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and high-performance liquid chromatography (HPLC), achieving an 85.3% purity of the pentameric form of M291. Bacterial strains All bacterial strains were procured from the Chinese Human Gut Microbial Biobank (hGMB). E. coli, E. faecalis, S. epidermidis, S. salivarius, B. bifidum, and B. thetaiotaomicron were used^[237]34. Bacterial medium and growth conditions Bacterial strains were obtained from different sources and cultured in AF media adapted from^[238]63, including brain heart infusion 18 g/L, trypticase Soy Broth 15 g/L, yeast extract 5 g/L, KH2PO4 2.5 g/L, heamin 0.5 mg/L D-glucose 0.5 g/L) for synthetic consortia design culture in aerobic or anaerobic conditions. Anaerobic bacterial strains and synthetic consortia were culture in anaerobic conditions (7% H2, 10% CO2, 83% N2). Each bacterial strain individual culture condition was assessed to accurately define the starting CFU number and the growth culture duration required to analyze growth at exponential phase. For individual culture experiments, the effect of immunoglobulins was calculated as follows: [MATH: Growth(% )=bacteria growth(OD)*100averag eofbacterialgrowthinPBS(< mrow>OD) :MATH] Synthetic MIC6 bacterial consortia cultures Synthetic bacterial community was cultured as previously described^[239]63. MIC6 is composed of E. coli, E. faecalis, S. epidermidis, S. salivarius, Bifidobacterium bifidum, and B. thetaiotaomicron. Briefly, bacterial strain monocultures were prepared from an overnight culture and subculture and were diluted to OD[600] 0.1 in AF medium for overnight culture. Monoculture were were mixed after reaching the exponential phased in the following proportions: E. coli (OD 0.01), E. faecalis (OD 0.01), S. epidermidis (OD 0.1), S. salivarius (OD 0.01), B. bifidum (OD 0.01), and B. thetaiotaomicron (OD 0.1) before co-culturing in AF medium. After overnight culture the co-culture was diluted 10 times in AF medium for 2 consecutive days and 100 times for 2 consecutive days incubated at 37 °C without shaking under anaerobic conditions. Synthetic consortia species stability was assessed by qPCR and stored in glycerol at −80 °C for further experiments. In indicated experiments, bacterial consortia were incubated with 50 µl of mAbs (60 µg/ml) or with purified intestinal Ig (60 µg/ml) 30 min at 4 °C. After wash, bacteria were incubated either with 50 µL of guinea pig complement (Cedarlane) or anti-IgG1 crosslinking Abs (clone HP6069), then O.N. cultured. at 37 °C in 1 mL AF media. Microbial ecological cultures Community–level physiological profiling has been performed to monitor changes in MIC culture performed in an anaerobic chamber using the Biolog EcoPlate system, which contains 31 independent carbon sources that are useful for their metabolism. The metabolic fingerprint reaction patterns is monitored by following the respiration that rapidly reduces the tetrazolium dye included with the carbon source and was measured by OD at 590 nm. Fluorescent bacterial labeling Bacterial strains were grown individually until reaching the exponential phase and diluted to OD[600] 0.2 and stained with fluorescent D-amino Acids (FDAAs)^[240]64. Briefly, bacteria were cultured in AF media in the presence of 0.5 mM of either RADA (# 6649 TOCRIS), sBADA (# 7860 TOCRIS), YADA (# 6650 TOCRIS) or Rf470DL (# 7406 TOCRIS) with a final concentration of DMSO 1%. Bacteria were cultured for a few hours at 37 °C before washing in cold sterile PBS and fixed 30mn PFA 4%. Bacterial fluorescent labeling was assessed by FACS. FAADs bacteria were stored −80 °C in 30% glycerol and adjusted to OD1 for further analyzes. Analysis of bacterial coated antibodies Fluorescent bacteria were pooled at 2 µL for each strain. To measure IgM- and IgA-coated intestinal bacteria, 5 µL of microbial pellets were resuspended in PBS. The bacterial suspensions were passed through a 40 µm cell strainer to eliminate aggregates and ensure uniformity. Fluorescent bacterial mixes (E. coli + E. faecalis + S. epidermidis + S. salivarius and B. bifidum + B. thetaiotaomicron) were then treated with 1 µg of G291, M291, Int_pM, Int_pA, or PBS as a control at 4 °C for 30 min. Following treatment, bacteria were washed twice with sterile, cold PBS before proceeding to FACS cluster analysis. Staining included IgM PerCP-Cy5.5 (clone: MHM-88, 1:100), anti-human IgA Viogreen (clone: IS11-8E10, 1:100), or anti-human IgG BV605 (clone: G18-145, 1:100) for 30 min at 4 °C. The bacteria were washed again and directly analyzed using the Spectral Cytek Aurora. For the analysis of IgM- and IgA-coated intestinal bacteria, 5 µL of microbial pellets were resuspended in PBS, filtered through a 40 µm strainer, and incubated at 4 °C for 30 min with anti-human IgM PerCP-Cy5.5 (clone: MHM-88, 1:100) and anti-human IgA Viogreen (clone: IS11-8E10, 1:100) before FACS analysis. To investigate fluorescent bacterial clustering, we initially analyzed bacterial clusters in the absence of antibodies to establish a baseline of natural bacterial interactions. Subsequently, we applied the same gating strategy to bacterial mixtures in the presence of antibodies. By comparing the proportions of each cluster, we observed variations between the PBS control and antibody-treated samples. These differences reflect the potential of the antibodies to modulate bacterial interactions, either by enhancing or reducing them. Extraction of DNA from bacteria DNA from 1 ml of a single bacterial culture or from synthetic consortia was extracted using the E.Z.N.A bacterial kit (Omega, D3350) following the manufacturer’s instructions. Briefly, bacteria pellets were incubated with 100 µl of TE buffer and 10 µl of lysozyme for 10 min at 37 °C. For Gram-positive bacteria and synthetic consortia, this step was directly followed by a glass bead lysis 5 min at a maximum of speed. Supernatants were incubated with 100 µl of BTL and 20 µl protease K at 55 °C for 1 h to disrupt the cell wall, and 5 µl of RNase A was added, mix gently and incubate RT for 5 min. After spinning, supernatants were incubated 10 min at 60 °C with 220 µl of BDL buffer and transfer into the provided column t after adding 220 µl of ethanol 75%. After spinning, flow through were discard and 500 µl of HBC was added to the column before 2 steps of wash. DNA were then eluted using elution buffer pre-warmed at 65 °C and the DNA amount was measured at 260 and 280 nm. Real-time quantitative PCR Real-time qPCR was performed using an ABI Quantstudio Sequence Detection System apparatus. The qPCR reaction mixture (20 µl) was composed of 10 µM of each primer, 2x Power SYBR Green PCR Master Mix (AidLab, PC3302), and 2 µl of extracted DNA. The amplification program consisted of one cycle of 95 °C for 10 min, followed by 40 cycles of 95 °C for 1mn, 55 °C for 50 s, and 72 °C for 1 min followed by eluting curve program consisting on 50 °C for 15 s and 95 °C for 15 s. Primers used for MIC6 qPCR were as follows: E. coli-F (5’-CATTGACGTTACCCGCAGAAGAAGC-3’); E.coli-R(5’-CTCTACGAGACTCAAGCTTGC-3’)^[241]65; E. faecalis-F (5’-ATCAAGTACAG TTAGTCT-3’); E. faecalis-R (5’-ACGATTCAAAGCTAACTG-3’)^[242]66; S. epidermidis -F (5’-CATTGGATTACCTCTTTGTTCAGC-3’); S. epidermidis-R (5’-CAAGCGAAATCTGTT GGGG-3’)^[243]67; S. salivarius-F (5’-GTTCCAGCAGCTAAAGAGGAAG-3’); S. salivarius-R (5’-CCGGTGCTACTTTAGCTACTGG-3’)^[244]68; B. thetaiotaomicron-F(5’-GGARCATGT GGTTTAATTCGATGAT-3’); B. thetaiotaomicron -R (5’-AGCTGACGACAACCATGC AG-3’)^[245]69; B. bifidum-F(5’-AGGGTTCGATTCTGCTCAG B. bifidum -R(5’-CATCCG GCATTACCACCC-3’)^[246]70. Determination of copy number Bacterial suspensions were plated on AF agar plates ON 37 °C with serial dilutions from OD[600] 10^–3 to 10^–8 to count CFU/ml, and DNA from the same dilutions was extracted to do qPCR. Linear curves from OD and CT values were used to calculate the copy numbers for each strain/growth condition. Germ-free mouse colonization and treatment Animal experiments were approved by the ethical committee (Ethical Approval No. A2023025). 6 weeks old Germ free male C57Bl/6 mice were kept in cage setups with autoclaved food and water to minimize unwanted cross-contaminations in 12 light/12 dark cycles with 18-23 °C with 40-60% humidity. Mice and the environment were tested to confirm germ-free conditions. Mice were orally administered with 150 µl of MIC6 consortium (10^9 CFU of each bacterial strain composing MIC6 (6 × 10^9 CFU / mouse)) or FMT (equivalent to 30 µg fecal matter). M291 or Nint_pM was provided in the drinking water at 20 µg/ml 2 days before and all along the experiment^[247]71. Mice were euthanized 11 days after the first gavage. Preparation of cecal content for metabolome analysis and bacterial culture Caecum from mice were retrieved and weighted. Cecal content was homogenized in cold sterile PBS at 1 g/ml. Homogenate was spun down at 10,000 × g for 10 min at 4degres. Supernatant were collected and analyzed for metabolome content. Pellets and 0.22 µm filtered supernatants were aliquoted individually and immediately frozen at −80 °C for bacterial growth experiments. Metabolomics Supernatants were prepared from the caecum as described above. The sample (100 µL) was thoroughly mixed with 400 µL of cold methanol acetonitrile (v/v, 1:1) via vortexing. Then, the mixture was processed with sonication for 1 h in an ice bath. The mixture was then incubated at −20 °C for 1 h and centrifuged at 4 °C for 20 min at a speed of 14,000 g. The supernatants were then harvested and dried under vacuum for LC-MS analysis. UHPLC-MS/MS analysis. Metabolomics profiling was analyzed using a UPLC-ESI-Q-Orbitrap-MS system (UHPLC, Shimadzu Nexera X2 LC-30AD, Shimadzu, Japan) coupled with Q-Exactive Plus (Thermo Scientific, San Jose, USA). For liquid chromatography (LC) separation, samples were analyzed using the ACQUITY UPLC® HSS T3 column (2.1×100 mm, 1.8 μm) (Waters, Milford, MA, USA). The flow rate was 0.3 mL/min, and the mobile phase contained A: 0.1% FA in water and B: 100% acetonitrile (ACN). The gradient was 0% buffer B for 2 min and linearly increased to 48% in 4 min, then up to 100% in 4 min and maintained for 2 min, then decreased to 0% buffer B in 0.1 min, with a 3 min re-equilibration period employed. The electrospray ionization (ESI) with positive and negative modes was applied for MS data acquisition separately. The HESI source conditions were set as follows: Spray Voltage: 3.8 kv (positive) and 3.2 kv (negative); Capillary Temperature: 320 °C; Sheath Gas (nitrogen) Flow: 30 arb (arbitrary units); Aux Gas Flow: 5 arb; Probe Heater Temp: 350 °C; S-Lens RF Level: 50. The instrument was set to acquire over the m/z range 70-1050 Da for full MS. The full MS scans were acquired at a resolution of 70,000 at m/z 200, and 17,500 at m/z 200 for the MS/MS scan. The maximum injection time was set to 100 ms for MS and 50 ms for MS/MS. The isolation window for MS2 was set to 2 m/z, and the normalized collision energy (stepped) was set as 20, 30, and 40 for fragmentation. Data preprocessing and filtering. The raw MS data were processed using MS-DIAL for peak alignment, retention time correction, and peak area extraction. The metabolites were identified by accurate mass (mass tolerance <10 ppm) and MS/MS data (mass tolerance <0.02 Da), which were matched with HMDB, MassBank, and other public databases and our self-built metabolite standard library. In the extracted-ion features, only the variables having more than 50% of the nonzero measurement values in at least one group were kept. KEGG enrichment analysis. To identify the perturbed biological pathways, the differential metabolite data were subjected to KEGG pathway analysis using the KEGG database ([248]http://www.kegg.jp). KEGG enrichment analyses were conducted with Fisher’s exact test, and FDR correction for multiple tests was performed. Enriched KEGG pathways were nominally statistically significant at the p < 0.05 level. Sequencing of fecal samples Total microbial genomic DNA was extracted from mouse fecal samples using the FastPure Stool DNA Isolation Kit (MJYH, Shanghai, China) according to manufacturer’s instructions. The quality and concentration of DNA were determined by 1.0% agarose gel electrophoresis and a NanoDrop® ND-2000 spectrophotometer (Thermo Scientific Inc., USA) and kept at −80 °C prior to further use. The hypervariable region V3-V4 of the bacterial 16S rRNA gene were amplified with primer pairs 338 F (5’-ACTCCTACGGGAGGCAGCAG-3’) and 806 R (5’-GGACTACHVGGGTWTCTAA T-3’) by a T100 Thermal Cycler (BIO-RAD, USA). The PCR reaction mixture including 4 μL 5 × Fast Pfu buffer, 2 μL 2.5 mM dNTPs, 0.8 μL each primer (5 μM), 0.4 μL Fast Pfu polymerase, 10 ng of template DNA, and ddH2O to a final volume of 20 µL. PCR amplification cycling conditions were as follows: initial denaturation at 95 °C for 3 min, followed by 27 cycles of denaturing at 95 °C for 30 s, annealing at 55 °C for 30 s, and extension at 72 °C for 45 s, and a single extension at 72 °C for 10 min, and end at 4 °C. All samples were amplified in triplicate. The PCR product was extracted from 2% agarose gel and purified. Then quantified using Synergy HTX (Biotek, USA). Purified amplicons were pooled in equimolar amounts and paired-end sequenced on an Illumina NextSeq 2000 PE300 platform (Illumina, San Diego, USA) according to the standard protocols by Majorbio Bio-Pharm Technology Co. Ltd. (Shanghai, China). Sequencing for metatranscriptomics Total RNA was extracted from the tissue using TRIzol® Reagent according to the manufacturer’s instructions. Then RNA quality was determined by the 5300 Bioanalyser (Agilent) and quantified using the ND-2000 (NanoDrop Technologies). Only high-quality RNA sample (OD260/280 = 1.8 ~ 2.2, OD260/230 ≥ 2.0, RQN ≥ 6.5, 28S:18S ≥ 1.0, >1 μg) was used to construct sequencing library. RNA purification, reverse transcription, library construction, and sequencing were performed at Shanghai Majorbio Bio-pharm Biotechnology Co., Ltd. (Shanghai, China) according to the manufacturer’s instructions. The RNA-seq transcriptome library was prepared following Illumina® Stranded mRNA Prep, Ligation (San Diego, CA) using 1 μg of total RNA. Shortly, messenger RNA was isolated according to the polyA selection method by oligo (dT) beads, and then fragmented by the fragmentation buffer first. Secondly, double-stranded cDNA was synthesized using a SuperScript double-stranded cDNA synthesis kit (Invitrogen, CA) with random hexamer primers. Then the synthesized cDNA was subjected to end-repair, phosphorylation, and adapter addition according to library construction protocol. Libraries were size-selected for cDNA target fragments of 300 bp on 2% Low Range Ultra Agarose, followed by PCR amplification using Phusion DNA polymerase (NEB) for 15 PCR cycles. After quantification by Qubit 4.0, the sequencing library was performed on the NovaSeq X Plus platform (PE150) using the NovaSeq Reagent Kit. The raw paired-end reads were trimmed and quality controlled by fastp with default parameters. Then clean reads were separately aligned to the reference genome with orientation mode using HISAT2 software. The mapped reads of each sample were assembled by StringTie in a reference-based approach. To identify DEGs (differential expression genes) between two different samples, the expression level of each transcript was calculated according to the transcripts per million reads (TPM) method. RSEM was used to quantify gene abundances. Essentially, differential expression analysis was performed using the DESeq2 or DEGseq. DEGs with |log2FC | ≧1 and FDR <0.05(DESeq2) or FDR < 0.001(DEGseq) were considered to be significantly different expressed genes. In addition, functional-enrichment analysis, including GO and KEGG were performed to identify which DEGs were significantly enriched in GO terms and metabolic pathways at a Bonferroni-corrected P-value < 0.05 compared with the whole-transcriptome background. GO functional enrichment and KEGG pathway analysis were carried out by Goatools and Python scipy software, respectively. All the alternative splice events that occurred in our sample were identified by using the recently releases program rMATS. Only the isoforms that were similar to the reference or comprised novel splice junctions were considered, and the splicing differences were detected as exon inclusion, exclusion, alternative 5′, 3′, and intron retention events. Data and statistical analysis Analyses were performed using GraphPad Prism (v 8.3.0). AUC of bacterial recognition of mAbs was calculated using R function smplot2::sm_auc_all (v 0.1.0)82 8 9, and the background AUC, which was calculated as the mean of the three lowest values in each assay, was subtracted. R function sva::ComBat (v 3.48.0)83 was applied to remove the batch effect of experiments14. Heatmaps were plotted using R package 12 pheatmap (v 1.0.12) or using Prism GraphPad v9. The number of donors and experiments and methods of tests were indicated in each Fig. legend. Statistical significance of difference between groups were tested using Student’s t test or Mann-Whitney test according to test requirements. Error bars represent SD and P values < 0.05, **P < 0.01). Statistics and reproducibility Sample sizes were determined by the availability of specimens. All statistical analyses were conducted using GraphPad Prism 9.5.0. Sample sizes (n) are specified in each figure legend and represent biological replicates unless otherwise indicated. We applied a two-sided Mann-Whitney U test. Statistical significance was defined as follows: P < 0.05 (*), P < 0.01 (**), P < 0.001 (***), P < 0.0001 (****); ns denotes not significant. For data visualization, box-and-whisker plots display the median as a center line, with the box spanning the 25th to 75th percentiles and whiskers extending to the 5th and 95th percentiles. Heatmaps present z-scored values unless otherwise stated, and volcano plots illustrate log₂ fold-change against −log₁₀ P-value. Biological and technical replicates are explicitly distinguished in the figure legends and throughout the manuscript to ensure clarity and reproducibility. No statistical method was used to predetermine sample size. No data were excluded from the analyses. The experiments were not randomized. The Investigators were not blinded to allocation during experiments and outcome assessment. Reporting summary Further information on research design is available in the [249]Nature Portfolio Reporting Summary linked to this article. Supplementary information [250]Supplementary Information^ (1.7MB, pdf) [251]Reporting Summary^ (188.6KB, pdf) [252]Transparent Peer Review file^ (1.7MB, pdf) Acknowledgements