Faecalibacterium Prausnitzii Probiotic for Gut Inflammation Control
Faecalibacterium prausnitzii represents one of the most abundant commensal bacteria in the healthy human gut, constituting 5-15% of the total bacterial population. Studies reveal its significant depletion in inflammatory bowel conditions, with reductions of 30-50% observed in Crohn's disease patients compared to healthy controls. F. prausnitzii produces butyrate at concentrations of 8-25 mM in the colon, which provides approximately 60-70% of colonocytes' energy requirements while simultaneously mediating anti-inflammatory effects through multiple signaling pathways.
The clinical challenge lies in maintaining viable F. prausnitzii populations within the intestinal environment while optimizing their metabolic output during inflammatory disease states.
This page brings together solutions from recent research—including heat-stable antimicrobial compounds with sub-10 kDa molecular weights, specific F. prausnitzii strains such as CNCM I-4573 and KBL1027, human milk oligosaccharide formulations that selectively promote F. prausnitzii growth, and co-administration strategies with mucin-degrading bacteria for enhanced engraftment. These and other approaches offer practical interventions for clinicians seeking to leverage F. prausnitzii's anti-inflammatory properties in treating intestinal disorders.
1. Composition of Human Milk Oligosaccharides with Constituent Sugars for Selective Bacterial Growth Regulation in Intestinal Microbiota
MEIJI CO LTD, 2025
Composition for controlling growth of beneficial bacteria in the intestinal microbiota using human milk oligosaccharides (HMOs) and their constituent sugars. The composition can be used as a prebiotic or synbiotic to selectively promote growth of bacteria like Faecalibacterium, Akkermansia, Blautia, Subdoligranulum, Bilophila, and Sutterella. This can have applications in treating conditions like irritable bowel syndrome, inflammatory bowel disease, Parkinson's, cancer, cognitive decline, and atopic dermatitis by modulating the intestinal bacterial community. The HMOs can contain sugars like fucose, lactose, sialic acid, etc.
2. Co-Administration of Mucin-Degrading and Butyrate-Producing Bacteria for Enhanced Gut Bacterial Engraftment
PENDULUM THERAPEUTICS INC, 2025
Enhancing engraftment of beneficial gut bacteria in order to improve their therapeutic effectiveness. The technique involves co-administering a mucin-degrading bacterium along with a butyrate-producing bacterium. The mucin-degrading bacterium primes the gut environment for the butyrate-producing bacterium to engraft more efficiently. This synergistic engraftment enhancement allows better colonization of the butyrate-producing bacterium for improved therapeutic benefits. The technique can be used in microbiome therapies for conditions like metabolic disorders, skin disorders, neurological disorders, dysbiosis, inflammation, etc.
3. Intestinal Bacteria Strains and Vesicles with ER Fragments Exhibiting Anti-Inflammatory and Antibacterial Properties
BIOBANKHEALING INC, 2025
Novel intestinal bacteria strains, vesicles derived from them, and endoplasmic reticulum (ER) fragments for use in treating inflammatory and bacterial diseases. The bacteria strains, isolated from the human gut, have anti-inflammatory and antibacterial properties. The vesicles and ER fragments from these strains also exhibit similar activities. The strains and derived products could be used to prevent, improve, or treat conditions like inflammatory bowel syndrome (IBS), irritable bowel disease (IBD), Clostridioides difficile infection (CDI), and bacterial infections.
4. Heat-Stable Antimicrobial Compounds from Faecalibacterium prausnitzii with Sub-10 kDa Molecular Weights
UNIV CORNELL, 2024
Antimicrobial compounds isolated from Faecalibacterium prausnitzii supernatants, comprising heat-stable secreted compounds with molecular weights less than 10 kDa, for inhibiting bacterial growth, preventing bacterial infection, and treating diseases such as irritable bowel disease, Crohn's disease, and ulcerative colitis.
5. Compositions of Viable Mediterraneibacter faecis Strains Isolated from Human Digestive Tract
MICROBA IP PTY LTD, 2024
Therapeutic compositions comprising bacterial strains isolated from the human digestive tract for treating inflammatory and autoimmune disorders. The compositions comprise viable cells of the Mediterraneibacter faecis species, which enhance gut barrier function and reduce inflammation. The strains can be administered alone or in combination with anti-inflammatory agents or nutritional supplements to treat conditions such as inflammatory bowel disease.
6. Microbial Consortium with Metabolite-Producing Microorganisms for Modulating Host Physiological Processes
BIOMICA LTD, 2024
Microbial consortium comprising two or more microorganisms capable of modulating physiological processes in the host, including gut-brain axis, enteric nervous system, and metabolic pathways. The microorganisms produce specific metabolites, such as GABA, that modulate host physiological processes, including serotonin levels, gut barrier function, and inflammatory response. The consortium can be used to treat functional gastrointestinal disorders by enhancing gut health, reducing inflammation, and modulating neurotransmitter activity.
7. Probiotic Composition with Faecalibacterium prausnitzii Strain CNCM I-4573
INSTITUT NATIONAL DE RECHERCHE POUR LAGRICULTURE LALIMENTATION ET LENVIRONNEMENT, 2024
A probiotic composition comprising a specific strain of Faecalibacterium prausnitzii (CNCM I-4573) for preventing and treating Clostridioides difficile (C. difficile) infections in individuals. The strain is administered orally or rectally to delay intestinal colonization by C. difficile, eliminate the bacteria from the intestine, and reduce symptoms associated with C. difficile infection. The composition can be administered simultaneously with or after antibiotic treatment and proton pump inhibitors (PPIs) to prevent C. difficile infection recurrence.
8. Faecalibacterium prausnitzii induces an anti-inflammatory response and a metabolic reprogramming in human monocytes
Camille Danne, Laura Creusot, Rodrigo de Oliveira Formiga - Cold Spring Harbor Laboratory, 2024
Background and aims. Faecalibacterium prausnitzii, a highly abundant bacterium in the human gut microbiota, has been linked to overall health and is decreased in several pathological conditions, such as Inflammatory Bowel Disease (IBD). F. prausnitzii has shown anti-inflammatory properties in human and mouse models, notably through the induction of IL-10 signaling. Here, we investigated which cell types from human blood and intestinal tissue are responsible for producing IL-10 induced by F. prausnitzii, and providing the first mechanistic insights. Methods. Immune cells isolated from human blood and intestinal lamina propria of patients with IBD and non-inflamed controls, were stimulated them with F. prausnitzii EXL01 strain or Escherichia coli lipopolysaccharide (LPS) and analysed by Legendplex, ELISA, flow cytometry, RNA-sequencing (RNAseq), and Seahorse technology. Results. F. prausnitzii EXL01 strain induced the direct and dose-dependent production of IL-10 in CD14+ monocytes from the systemic circulation and intestinal tissue of IBD patients and non-inflamed controls, without in... Read More
9. The gut microbiome regulates the clinical efficacy of sulfasalazine therapy for IBD-associated spondyloarthritis
Svetlana Lima, Sílvia Pires, Amanda Rupert - Elsevier BV, 2024
Sulfasalazine is a prodrug known to be effective for the treatment of inflammatory bowel disease (IBD)-associated peripheral spondyloarthritis (pSpA), but the mechanistic role for the gut microbiome in regulating its clinical efficacy is not well understood. Here, treatment of 22 IBD-pSpA subjects with sulfasalazine identifies clinical responders with a gut microbiome enriched in Faecalibacterium prausnitzii and the capacity for butyrate production. Sulfapyridine promotes butyrate production and transcription of the butyrate synthesis gene but in F. prausnitzii in vitro, which is suppressed by excess folate. Sulfasalazine therapy enhances fecal butyrate production and limits colitis in wild-type and gnotobiotic mice colonized with responder, but not non-responder, microbiomes. F. prausnitzii is sufficient to restore sulfasalazine protection from colitis in gnotobiotic mice colonized with non-responder microbiomes. These findings reveal a mechanistic link between the efficacy of sulfasalazine therapy and the gut microbiome with the potential to guide diagnostic and therapeutic approac... Read More
10. IDDF2024-ABS-0130 Faecalibacterium prausnitzii antagonizes helicobacter pylori colonization and inhibits gastric carcinogenesis
Lanping Jiang, Kaili Fu, Chi Chun Wong - BMJ Publishing Group Ltd and British Society of Gastroenterology, 2024
<h3>Background</h3> Probiotics could protect against cancers, however, the roles of probiotics in inhibiting GC and affecting <i>H. pylori</i> infection are largely unclear. We first identified <i>Faecalibacterium prausnitzii</i> is depleted in GC. We aimed to evaluate the antitumorigenic function and molecular mechanism of <i>F. prausnitzii</i> in GC and its interplay with <i>H. pylori</i>. <h3>Methods</h3> <i>F. prausnitzii</i> abundance was determined in 1343 human subjects consisting of 48 healthy controls (HC), 192 superficial gastritis (SG), 130 atrophy gastritis (AG), 123 intestinal metaplasia (IM), 80 gastric epithelial dysplasia (GED), 456 cancer adjacent normal (CAN) and 407 GC. The effect of <i>F. prausnitzii</i> on GC was evaluated in GC cells, patient-derived organoids (G9T, POA145), human GC xenograft, YTN16 allografts and N -methyl- N -nitrosourea (MNU) induced GC tumorigenesis mouse models. <i>F. prausnitzii</i> attachment to GC cells and its impact on <i>H. pylori</i> colonization was determined by scanning electron microscopy (SEM) and transmission electron microsco... Read More
11. Faecalibacterium prausnitzii regulates carbohydrate metabolic functions of the gut microbiome in C57BL/6 mice
Peiling Geng, Ni Zhao, Yufan Zhou - Cold Spring Harbor Laboratory, 2024
The probiotic impact of microbes on host metabolism and health depends on both host genetics and bacterial genomic variation. Faecalibacterium prausnitzii is the predominant human gut commensal emerging as a next-generation probiotic. Although this bacterium exhibits substantial intraspecies diversity, it is unclear whether genetically distinct F. prausnitzii strains might lead to functional differences in the gut microbiome. Here, we isolated and characterized a novel F. prausnitzii strain (UT1) that belongs to the most prevalent but underappreciated phylogenetic clade in the global human population. Genome analysis showed that this butyrate-producing isolate carries multiple putative mobile genetic elements, a clade-specific defense system, and a range of carbohydrate catabolic enzymes. Multiomic approaches were used to profile the impact of UT1 on the gut microbiome and associated metabolic activity of C57BL/6 mice at homeostasis. Both 16S rRNA and metagenomic sequencing demonstrated that oral administration of UT1 resulted in profound microbial compositional changes including a s... Read More
12. 138P Faecalibacterium prausnitzii enhances tumor response to perioperative nivolumab and chemotherapy
Alejandro Rodríguez-Festa, Roberto Serna‐Blasco, Ernest Nadal - Elsevier BV, 2024
Faecalibacterium prausnitzii is one of the most prevalent commensals within the gut microbiota, being one of the gut's major butyrate-producing bacteria. Recent studies suggest a potential role for F. prausnitzii in exerting anti-tumorigenic effects. In the context of the phase II NADIM II clinical trial, a total of 81 baseline stool from locally advanced non-small cell lung cancer (NSCLC) patients randomized into the experimental arm (neoadjuvant nivolumab plus chemotherapy followed by adjuvant nivolumab, n=55) or the control arm (chemotherapy alone, n=26) were collected. DNA was extracted using the QIAamp PowerFecal DNA Kit (Qiagen) and analyzed by QPCR using specific primers for F. prausnitzii strain ATCC 27768. In the experimental arm, the presence of F.prautnizzi was significantly associated with improved overall survival (HR: 0.24; 95CI: 0.08-0.76; P=0.015), and a trend was noted toward progression-free survival (HR: 0.46; 95IC: 0.18-1.19; P=0.1). Specifically, patients with detected F. prausnitzii in their fecal sample (n=46) exhibited a 88% probability of being alive and 68% ... Read More
13. The Role of Akkermansia muciniphila and Faecalibacterium prausnitzii in the Pathogenesis of Ulcerative Colitis and Crohn's Disease
Enes Ali Kurt, Mehmet Demırci, Doğukan Özbey - Clinical Laboratory Publications, 2024
Crohn's disease (CD) and ulcerative colitis (UC) are inflammatory bowel diseases with uncertain etiology. We aimed to determine the amounts of Akkermansia muciniphila and Faecalibacterium prausnitzii in the intestinal microbiota of these patients and to correlate their amounts with blood IL-8, IL-10, and IL-12 cytokine levels.
14. Composition Comprising Faecalibacterium cancerinhibens for Inflammatory Disease Analysis
CJ BIOSCIENCE INC, 2023
A composition for diagnosing and treating inflammatory diseases, such as inflammatory bowel disease (IBD), comprising a Faecalibacterium spp. microorganism. The composition can be used to predict the risk of developing IBD, diagnose IBD, or evaluate the effectiveness of IBD treatment. The microorganism, specifically Faecalibacterium cancerinhibens, has anti-inflammatory properties and can be administered orally to prevent or treat IBD.
15. Parabacteroides Distasonis Strain CNCM I-5828 with Visceral Pain Modulating Properties
GREENTECH, 2023
A Parabacteroides distasonis strain deposited under accession number CNCM I-5828, isolated from human feces, exhibits strong efficacy in preventing or treating visceral pain associated with Inflammatory Bowel Disease (IBD) and Irritable Bowel Syndrome (IBS). The strain can be used in nutritional supplements or as a medicament, particularly for visceral pain management.
16. Faecalibacterium Prausnitzii Strain KBL1027 with Unique Genetic Profile
KOBIOLABS INC, 2023
A novel strain of Faecalibacterium prausnitzii, designated KBL1027, having accession number KCTC14231BP, with anti-inflammatory properties and the ability to induce production of anti-inflammatory cytokines. The strain can prevent, improve, or treat inflammatory diseases, including inflammatory bowel disease, atopic dermatitis, and allergic diseases, by modulating the gut microbiota and enhancing the intestinal barrier function.
17. Faecalibacterium Prausnitzii Strain EB-FPDK9 with Characterized 16S rRNA Sequence
ENTEROBIOME INC, 2023
A novel strain of Faecalibacterium prausnitzii, designated EB-FPDK9, exhibiting anti-inflammatory and lipid accumulation inhibitory effects. The strain is isolated from human intestinal mucus and characterized by its 16S rRNA sequence. EB-FPDK9 is effective in preventing or treating inflammatory diseases, liver disease, and metabolic disorders, and can be administered as a pharmaceutical composition or incorporated into food products.
18. Synergy and oxygen adaptation for development of next-generation probiotics
Muhammad Tanweer Khan, Chinmay Dwibedi, Daniel Sundh - Springer Science and Business Media LLC, 2023
The human gut microbiota has gained interest as an environmental factor that may contribute to health or disease1. The development of next-generation probiotics is a promising strategy to modulate the gut microbiota and improve human health; however, several key candidate next-generation probiotics are strictly anaerobic2 and may require synergy with other bacteria for optimal growth. Faecalibacterium prausnitzii is a highly prevalent and abundant human gut bacterium associated with human health, but it has not yet been developed into probiotic formulations2. Here we describe the co-isolation of F. prausnitzii and Desulfovibrio piger, a sulfate-reducing bacterium, and their cross-feeding for growth and butyrate production. To produce a next-generation probiotic formulation, we adapted F. prausnitzii to tolerate oxygen exposure, and, in proof-of-concept studies, we demonstrate that the symbiotic product is tolerated by mice and humans (ClinicalTrials.gov identifier: NCT03728868 ) and is detected in the human gut in a subset of study participants. Our study describes a technology for t... Read More
19. <i>Faecalibacterium</i>: a bacterial genus with promising human health applications
Rebeca Martín, David Ríos-Covián, Eugénie Huillet - Oxford University Press (OUP), 2023
Abstract In humans, many diseases are associated with alterations in gut microbiota, namely increases or decreases in the abundance of specific bacterial groups. One example is the genus Faecalibacterium. Numerous studies have underscored that low levels of Faecalibacterium are correlated with inflammatory conditions, with inflammatory bowel disease (IBD) in the forefront. Its representation is also diminished in the case of several diseases, including colorectal cancer (CRC), dermatitis, and depression. Additionally, the relative presence of this genus is considered to reflect, at least in part, intestinal health status because Faecalibacterium is frequently present at reduced levels in individuals with gastrointestinal diseases or disorders. In this review, we first thoroughly describe updates to the taxonomy of Faecalibacterium, which has transformed a single-species taxon to a multispecies taxon over the last decade. We then explore the links discovered between Faecalibacterium abundance and various diseases since the first IBD-focused studies were published. Next, we examine cur... Read More
20. Microbial Consortia with Active and Supportive Microbes for Substrate Degradation in Gastrointestinal Tract
FEDERATION BIO INC, 2023
Microbial consortia for treating diseases by degrading disease-associated metabolic substrates in the gastrointestinal tract. The consortia comprise a plurality of active microbes that metabolize the substrate, along with a supportive community of microbes that metabolize byproducts produced by the active microbes. The consortia exhibit enhanced characteristics, including improved gastrointestinal engraftment, increased biomass, and enhanced substrate metabolism, compared to administering the active microbes alone.
Get Full Report
Access our comprehensive collection of 101 documents related to this technology