Current Topic in Lactic Acid Bacteria and Probiotics
Korean Society for Lactic Acid Bacteria and Probiotics
Review Article

Akkermansia muciniphila: A Promising Next Generation Probiotic

Muhammad Adeel Hasnain1https://orcid.org/0009-0003-5702-9621, Gi-Seong Moon1,2,*https://orcid.org/0000-0003-3033-5250
1Major in IT·Biohealth Convergence, Department of IT·Energy Convergence, Graduate School, Korea National University of Transportation, Chungju 27469, Republic of Korea.
2Department of Biotechnology, Korea National University of Transportation, Jeungpyeong 27909, Republic of Korea.
*Corresponding author : Gi-Seong Moon, Department of Biotechnology, Korea National University of Transportation, Jeungpyeong 27909, Republic of Korea, Tel: +82-43-820-5251 Fax: +82-43-820-5272 E-mail: gsmoon@ut.ac.kr

Copyright © Korean Society for Lactic Acid Bacteria and Probiotics. This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Received: May 10, 2025; Revised: May 23, 2025; Accepted: May 27, 2025

Published Online: Jun 30, 2025

Abstract

An important mucin-degrading member of the phylum Verrucomicrobia, Akkermansia muciniphila(AKK) has become a promising next-generation probiotic with important host health implications. AKK, which is mostly found in the intestinal mucus layer, regulates gut barrier integrity, immunological responses, and metabolic activities to help maintain gut homeostasis. Recent research has explored its potential to contribute to the improvement of various diseases conditions such as obesity, type 2 diabetes, nonalcoholic fatty liver disease, cardiovascular illnesses, and neurodegenerative diseases. Its extracellular vesicles and outer membrane proteins affect T-cell responses, systemic inflammation, and even neurotrophic signaling, which is involved in mood and cognitive function. Its action seems context-dependent, though, as both positive and negative effects have been noted in various neurological conditions including multiple sclerosis and Parkinson’s disease. Significant strain-level diversity is shown by genomic analysis, indicating functional heterogeneity that could be the cause of these disparate results. Current research on AKK’s diverse functions in health and illness is discussed in this review, highlighting the directions for further studies to further explore its probiotic potential.

Keywords: next generation probiotic; gut barrier integrity; immune modulation; therapeutic potential; extracellular vesicles

Introduction

Human microbiota,which is also called the hidden organ includes a variety of microorganisms dominated by bacterial community. Microbiota coexists and coevolves in various sites of human body mainly gut, skin and oral cavity. It contributes genetic information, surpassing 150 times that of the entire human genome (Hou et al., 2022). It plays an important role in host’s health status through physiological metabolic and immune modulations such as food fermentation, protection against the pathogens, vitamins production and immune response stimulation (Fujimura et al., 2010).

Gastrointestinal tract has a diverse microbial ecology where two bacterial phyla Bacteroidetes and Firmicutes predominate, while Actinobacteria, Proteobacteria, and Verrucomicrobia are typically present in lower abundance (Lozupone et al., 2012). Among the gut microbiota, Akkermansia is notable for its mucin-degrading capacity, involvement in metabolic homeostasis, and potential as a therapeutic agent in metabolic disorders.

Akkermansia muciniphila (referred as AKK hereafter) has been consistently associated with host health, and its reduced abundance is linked to a range of pathological conditions in both murine models and humans. Although the precise molecular signaling pathways through which this probiotic confers benefits to the host are still being explored actively, its preventive and therapeutic potential in conditions such as obesity, aging, diabetes and various other metabolic syndromes is well established (Hasnain et al., 2024). Importantly, AKK plays a central role in preserving gut barrier integrity and modulating immune responses, thereby limiting chronic low-grade inflammation-a key contributor to the pathogenesis of numerous diseases (Aja et al., 2025).

This mini-review aims to provide a concise overview of the current understanding of AKK as a next-generation probiotic, with a focus on its roles in metabolic regulation, immune modulation, and gut barrier function. We explore emerging evidence linking AKK to various health conditions, including metabolic disorders, inflammation, and neurodegeneration. Additionally, we highlight the underlying mechanisms by which this bacterium may exert its beneficial effects, discuss and identify key concerning areas that warrant further investigation.

Taxonomy, Genomics and Physiology

AKK is a Gram-negative, oval-shaped, non-motile oxygen-tolerant anaerobe which was first discovered in 2004 in human fecal samples. It belongs to phylum Verrucomicrobia and was the 1st cultivated member of Akkermansia genus (Aja et al., 2025). While it’s most abundant in colon, mucus layer of small intestine is also inhabited by AKK (Hasnain et al., 2024). First genome of AKK was sequenced in 2011. With a total size of 2.7 Mb, 11% of its large secretome (61 proteins) were involved in mucus degradation along with 43% other proteins with no known function which might also play role in mucin modification/processing (van Passel et al., 2011). In addition, its genome analysis revealed the presence of two CRISPR loci and many phage-derived sequences highlighting the vital role of viral infections in their evolution. Analysis of various other AKK isolates have demonstrated the possibility of horizonal gene transfer since the CRISPR genes are located close to predicted phage genes (Becken et al., 2021; van Passel et al., 2011). While a type-strain, MucT (ATCC BAA-835) remains most extensively studied, pangenome analysis of 234 isolates revealed the presence of significant genomic diversity in AKK strains based on which these can be categorized into separate clades and subspecies with diverse attributes (Mueller et al., 2024).

Since it colonizes intestinal mucosa, its energy harvesting and synthesis pathways are based on mucin degradation and processing. It thrives by degrading mucin, its main energy and nitrogen source. It metabolizes mucin-derived monosaccharides such as fucose, galactose and N-acetylgalactosamine, with enhanced growth observed when mucin is co-metabolized with these sugars (Ottman et al., 2017). Although it can ferment some non-mucin sugars like fructose and human milk oligosaccharides, it does not efficiently utilize sugars like maltose, melibiose or trehalose, despite genomic indications of potential metabolism (Aja et al., 2025; Ottman et al., 2017). Notably, threonine is the only essential amino acid AKK cannot produce, likely due to its abundance in mucin, reflecting its adaptation to the mucosal niche (Ottman et al., 2017; Schrager 1970).

Role in Gut Health

Intestinal tight junctions and mucus are important in keeping the damage from digestive secretions and pathogens. AKK is involved in improving the intestinal barrier integrity through i) enhancement of mucus-producing goblet cells and regulating their autophagy process, ii) intestinal stems cell stimulation and differentiation to intestinal epithelial cells and Paneth/ goblet cells, iii) upregulation of tight junction proteins e.g. Occludin, Claudin-3, ZO-1 directly or through its extra extracellular vesicles (EVs) mainly via inhibition of NF-κB pathway and yet-to-establish mechanisms (Mo et al., 2024). In addition, AKK improves the leaky gut situation through suppressing the production of inflammatory cytokines like TNF-α and IL-8. Short-chain fatty acids (SCFAs), and proteins like Amuc_1100 and P9 stimulate L cells to secrete glucagon-like peptide-1 (GLP-1) which improves barrier function and reduces inflammation (Mo et al., 2024; Yoon et al., 2021). AKK and its out-membrane protein i.e. Amuc_1100 have been shown to induce CD8+ cytotoxic T lymphocytes activity and Treg differentiation thereby inhibiting colitis-associated colorectal cancer (Wang et al., 2020). Two other membrane proteins from AKK, namely Amuc_2172 and Amuc_2173 further reprogram tumor immune microenvironments in intestinal tumors (Jiang et al., 2023; Mo et al, 2024). Further, through TLR2/NLRP3 signaling, AKK shifts macrophages toward M1 phenotype in anti-cancer immunity (Fan et al., 2021).

Implications in Metabolic Disorders and Cardiovascular Health

Recent research has found changes in the abundance of AKK to be associated with various metabolic disorders like obesity, type 2 diabetes mellitus (T2D), nonalcoholic fatty liver disease and cardiovascular diseases (Everard et al., 2013; Shi et al., 2021; Zhang et al., 2021).

Obesity and its associated disorders have become a serious health issue. The role of gut microbiota in metabolism and energy harvesting process is well established and gut dysbiosis has been linked to imbalance in energy intake and expenditure leading to obesity and related disorders. Recent research has suggested that AKK is a promising candidate for preventing or ameliorating obesity and associated diseases (Hasnain et al., 2024). Live and pasteurized AKK and its EVs plays a protective role against obesity primarily by enhancing gut barrier integrity and modulating host metabolism. In high-fat diet (HFD)-induced models, it reduces intestinal permeability, lowers plasma lipopolysaccharide (LPS) levels, and mitigates systemic inflammation (Ashrafian et al., 2021; Depommier et al., 2020). Mechanistically, AKK promotes thermogenesis, increases GLP-1 secretion, elevates endocannabinoid levels, and downregulates genes involved in adipogenesis and carbohydrate transport. These actions collectively reduce fat accumulation, improve insulin sensitivity, and decrease energy efficiency. While animal studies demonstrate consistent anti-obesity effects with live, pasteurized, or EVs derived from AKK, clinical evidence in humans remains limited and warrants further investigation (Zhao et al., 2024).

T2D is a heterogeneous metabolic disorder characterized by hyperglycemia, insulin resistance, obesity, and low-grade inflammation, with a rapidly increasing global prevalence. If left uncontrolled, T2D leads to severe complications such as diabetic nephropathy, cardiomyopathy, retinopathy, and cognitive impairment, significantly contributing to mortality (Gregg et al., 2014). AKK exerts multifaceted beneficial effects in T2D through several interconnected mechanisms. It modulates host glucose metabolism by enhancing GLP-1 secretion via microbial metabolites such as propionate and direct interaction of outer membrane protein P9 with intercellular adhesion molecule (ICAM)-2, which together stimulate insulin release and improve glycemic control (Li et al., 2023; Psichas et al., 2015; Yoon et al., 2021). The bacterium also activates the phosphatidylinositol 3-kinase (PI3K)-Akt pathway and promotes lipid oxidation through upregulation of the liver kinase B1 (LKB1)-AMPK signaling axis, thereby mitigating hepatic steatosis (Huang et al 2018; Jiafeng et al; 2022; Rao et al., 2021). Furthermore, it influences bile acid metabolism by modulating farnesoid X receptor signaling and fibroblast growth factor 15/19 (FGF15/19) pathways, contributing to improved lipid and glucose homeostasis (Li et al., 2023). AKK strengthens the gut barrier by increasing mucin (Muc2) production, tight junction protein expression (e.g., ZO-1, occludin), and stimulating Wnt signaling in intestinal stem cells, which collectively reduce metabolic endotoxemia by limiting LPS translocation (Guo et al., 2022; Li et al., 2023). Additionally, AKK contributes to microbiota homeostasis by enhancing microbial diversity and rebalancing the Firmicutes/Bacteroidetes ratio, which is often disrupted in T2D. Its beneficial effects are not limited to live bacteria; components such as pasteurized cells, outer membrane protein Amuc_1100, and extracellular vesicles also retain immunomodulatory and metabolic regulatory properties, supporting its potential as a next-generation probiotic for T2D.

AKK supports cardiovascular health by reducing endothelial inflammation through downregulation of TNF-α, MCP-1, and ICAM-1, thereby inhibiting macrophage adhesion and atherosclerotic plaque formation (Gofron et al., 2024; Li et al., 2016). It lowers systemic inflammatory markers such as CRP and TNF receptor II without altering lipid or glucose levels, reducing atherosclerosis severity (Gofron et al., 2024). The bacterium also produces propionate, which inhibits arterial calcification, and reduces TMAO synthesis, thereby protecting against atrial fibrillation and cardiomyocyte pyroptosis. These benefits are primarily observed with live bacterial supplementation. Additionally, AKK improves gut barrier function and lipid metabolism, contributing to weight reduction and blood pressure control, indirectly mitigating cardiovascular risk (Gofron et al., 2024; Luo et al., 2022; Yan et al., 2022). Table 1 enlists recent research regarding the ameliorative potential of AKK in different disease conditions.

Table 1. Application of AKK in various disease conditions
Type of disorder Type of model Main observation Reference
Depression CRS mouse model ↓ Depression-like behavior; ↑ BDNF, ↑ dopamine, regulation of gut microbiota and metabolic pathways Ding et al., 2021
Antibiotic-treated mouse model ↓ Depression-like behavior, ↑ BDNF and 5-HT, modulated gut-brain axis, normalized HPA axis Sun et al., 2023
Murine alcohol + LPS model ↓ Depression-like symptoms, ↑ occludin, BDNF, and 5-HT; ↓ LPS, TNF-α, IL-1α, and IL-6 Guo et al., 2022
Alzheimer’s disease (AD) APP/PS1 transgenic mouse model ↓ Aβ plaques and levels; Improved cognition and gut barrier integrity Ou et al., 2020
Cardiovascular disease Atherosclerosis (ApoE/ mice) ↓ Atherosclerotic lesions; Improved lipid profiles, and restored gut microbial balance Xiao et al., 2024
Abdominal aortic aneurysm (mice) ↓ Aneurysm development, restored microbiota diversity, ↓ systemic inflammation He et al., 2022
Metabolic disease HFD-fed rats Protected β-cells from apoptosis, promoted differentiation, ↑ gut barrier function Yan et al., 2024
Obesity & T2D Meta-analysis of animal studies ↓ Body weight by 10.4%, fasting glucose by 21.2%; ↑ insulin by 26.9%, improved glucose tolerance Liu et al., 2024
Colorectal cancer Mouse tumorigenesis model Amuc_2172 inhibits tumorigenesis by upregulating HSP70 and enhancing CD8+ T cell activity Jiang et al., 2023
Sepsis Mouse sepsis model Arg-Lys-His tripeptide from AKK reduces inflammation via TLR4 inhibition Xie and Prasad, 2020
Metabolic syndrome/gut barrier integrity Mouse gut permeability model AKK-EVs promote gut barrier integrity Chelakkot et al., 2018
Type 2 diabetes Clinical trial (probiotic formulation) ↓ Postprandial glucose levels; ↑ insulin sensitivity and metabolic markers Perraudeau et al., 2020
Age-related muscle decline Clinical trial (pasteurized AKK HB05) ↑ Muscle strength, function, and physical performance Kang et al., 2024
Chronic respiratory symptoms Clinical trial (ETB-F01, heat-killed AKK) Improved respiratory symptoms and lung function Lee et al., 2024
Obesity and diabetes HFD mouse model ↓ Body weight gain, fat mass, insulin resistance, lowered plasma leptin and triglycerides; ↑ glucose tolerance, gut barrier integrity Plovier et al., 2017
Obesity, insulin resistance Randomized, double-blind, placebo-controlled pilot study ↑ Insulin sensitivity; ↓ plasma total cholesterol, fat mass, plasma LPS, creatine kinase Depommier et al., 2019
Download Excel Table

AKK and Neurological Disorders

In neuropsychiatric conditions, AKK modulates host physiology through multifaceted mechanisms involving metabolic, immunological, and gut-brain signaling pathways. Its outer membrane protein Amuc_1100 and secreted protein P9 influence serotonin biosynthesis and brain-derived neurotrophic factor (BDNF) signaling, critical in depression and anxiety-like behaviors (Lei et al., 2023). Amuc_1100 acts via TLR2 to promote 5-HT production and upregulate BDNF, while P9 enhances GLP-1 secretion with downstream neuroprotective effects (Holst et al., 2011; Wang et al., 2021). In Parkinson’s disease (PD) and Alzheimer’s disease (AD), studies consistently report elevated levels of AKK in patient microbiota, but with mixed implications. While some studies suggest AKK may help reduce inflammation, others associate its increased abundance with damage to the gut lining and higher levels of endotoxins in the blood (Lei et al., 2023). Therefore, further research is necessary to clarify the dual role of AKK in PD and determine whether its increased abundance is protective or contributes to disease pathology. In PD and multiple sclerosis (MS), both human and mouse studies show elevated AKK abundance correlating with either beneficial modulation of T-cell responses or exacerbation of inflammation, depending on context and strain (Takewaki et al., 2020). The production of SCFAs, particularly butyrate, by AKK or its microbial consortia, is linked to anti-inflammatory effects, enhanced synaptic plasticity, and modulation of the enteric and central nervous systems (Lei et al., 2023; Yang et al., 2019). Hence, clinical and animal studies across conditions like depression, AD, and stroke reveal alterations in AKK levels, often tied to behavioral and cognitive outcomes. These findings suggest a complex, context-dependent role for AKK in neurological disorders, balancing mucosal health and immune regulation with potential pathogenic effects in dysregulated states (Lei et al., 2023).

Conclusions

AKK is gaining attention as a strong candidate for next-generation probiotics because of its important role in supporting gut and metabolic health. It helps maintain the gut barrier, supports the immune system, and produces useful compounds like SCFAs, which are linked to benefits in conditions such as obesity, type 2 diabetes, and inflammatory bowel disease. Different strains of AKK have shown differences in how they function, suggesting that their health effects may depend on the specific strain used. While animal studies have shown encouraging results, more research in humans is needed to confirm these findings. Future studies should focus on testing the safety of using live AKK, understanding how different strains work, and exploring how it interacts with diet and other gut microbes. There is also potential in combining AKK with other gut-based treatments to manage complex health problems. With more evidence, this microbe could become a valuable tool in future treatments for metabolic and gut-related diseases. Preclinical data substantially supports its therapeutic promise, however, there are still a limited number of human studies. Therefore, further research is needed into strain-specific effects, administration methods, and long-term safety.

Acknowledgements

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (RS-2023-00244482). This research was also supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (No. 2021R1A6A1A03046418).

References

1.

Aja E, Zeng A, Gray W, Connelley K, Chaganti A and Jacobs JP (2025) Health effects and therapeutic potential of the gut microbe Akkermansia muciniphila. Nutrients, 17, 562.

2.

Ashrafian F, Keshavarz Azizi Raftar S, Lari A, Shahryari A, Abdollahiyan S, Moradi HR, Masoumi M, Davari M, Khatami S, Omrani MD, Vaziri F, Masotti A and Siadat SD (2021) Extracellular vesicles and pasteurized cells derived from Akkermansia muciniphila protect against high-fat induced obesity in mice. Microbial Cell Factories, 20, 219.

3.

Becken B, Davey L, Middleton DR, Mueller KD, Sharma A, Holmes ZC, Dallow E, Remick B, Barton GM, David LA, McCann JR, Armstrong SC, Malkus P and Valdivia RH (2021) Genotypic and phenotypic diversity among human isolates of Akkermansia muciniphila. MBio, 12, e00478-21.

4.

Chelakkot C, Choi Y, Kim D-K, Park HT, Ghim J, Kwon Y, Jeon J, Kim M-S, Jee Y-K, Gho YS, Park H-S, Kim Y-K and Ryu SH (2018) Akkermansia muciniphila-derived extracellular vesicles influence gut permeability through the regulation of tight junctions. Experimental & Molecular Medicine, 50, e450.

5.

Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-Silva S, Falony G, Raes J, Maiter D, Delzenne NM, De Barsy M, Loumaye A, Hermans MP, Thissen J-P, De Vos WM and Cani PD (2019) Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nature Medicine, 25, 1096-1103.

6.

Depommier C, Van Hul M, Everard A, Delzenne NM, De Vos WM and Cani PD (2020) Pasteurized Akkermansia muciniphila increases whole-body energy expenditure and fecal energy excretion in diet-induced obese mice. Gut Microbes, 11, 1231-1245.

7.

Ding Y, Bu F, Chen T, Shi G, Yuan X, Feng Z, Duan Z, Wang R, Zhang S, Wang Q, Zhou J and Chen Y (2021) A next-generation probiotic: Akkermansia muciniphila ameliorates chronic stress–induced depressive-like behavior in mice by regulating gut microbiota and metabolites. Applied Microbiology and Biotechnology, 105, 8411-8426.

8.

Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Guiot Y, Derrien M, Muccioli GG, Delzenne NM, De Vos WM and Cani PD (2013) Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proceedings of the National Academy of Sciences, 110, 9066-9071.

9.

Fan L, Xu C, Ge Q, Lin Y, Wong CC, Qi Y, Ye B, Lian Q, Zhuo W, Si J, Chen S and Wang L (2021) A. muciniphila suppresses colorectal tumorigenesis by inducing TLR2/NLRP3-mediated M1-like TAMs. Cancer Immunology Research, 9, 1111-1124.

10.

Fujimura KE, Slusher NA, Cabana MD and Lynch SV (2010) Role of the gut microbiota in defining human health. Expert Review of Anti-Infective Therapy, 8, 435-454.

11.

Gofron K, Berezowski A, Gofron M, Borówka M, Dziedzic M, Kazimierczak W, Kwiatkowski M, Gofron M, Nowaczyk Z and Małgorzewicz S (2024) Akkermansia muciniphila–impact on the cardiovascular risk, the intestine inflammation and obesity. Acta Biochimica Polonica, 71.

12.

Gregg EW, Li Y, Wang J, Rios Burrows N, Ali MK, Rolka D, Williams DE and Geiss L (2014) Changes in diabetes-related complications in the United States, 1990-2010. New England Journal of Medicine, 370, 1514-1523.

13.

Guo D, Park C, Li Y, Li B, Yang Q, Deng Y, Gao NL, Li R, Wang X, Yi L and Liu Z (2022) Akkermansia muciniphila ameliorates depressive disorders in a murine alcohol-LPS (mALPS) model. Food & Function, 13, 12766-12776.

14.

Hasnain MA, Kang D and Moon G-S (2024) Research trends of next generation probiotics. Food Science and Biotechnology, 33, 2111-2121.

15.

He X, Bai Y, Zhou H and Wu K (2022) Akkermansia muciniphila alters gut microbiota and immune system to improve cardiovascular diseases in murine model. Frontiers in Microbiology, 13.

16.

Holst JJ, Burcelin R and Nathanson E (2011) Neuroprotective properties of GLP-1: Theoretical and practical applications. Current Medical Research and Opinion, 27, 547-558.

17.

Hou K, Wu ZX, Chen XY, Wang JQ, Zhang D, Xiao C, Zhu D, Koya JB, Wei L, Li J and Chen ZS (2022) Microbiota in health and diseases. Signal Transduction and Targeted Therapy, 7.

18.

Huang X, Liu G, Guo J and Su Z (2018) The PI3K/AKT pathway in obesity and type 2 diabetes. International Journal of Biological Sciences, 14, 1483-1496.

19.

Jiafeng X, Longxian L, Boqiang L, Shuting W, Sitong Z, Zhengjie W, Liya Y, Xiaoyuan B, Qiangqiang W, Kaicen W, Aoxiang Z, Shengjie L, Ren Y, Huiyong J, Kaijin X and Lanjuan L (2022) Akkermansia muciniphila ameliorates acetaminophen-induced liver injury by regulating gut microbial composition and metabolism. Microbiology Spectrum, 10, e01596-21.

20.

Jiang Y, Xu Y, Zheng C, Ye L, Jiang P, Malik S, Xu G, Zhou Q and Zhang M (2023) Acetyltransferase from Akkermansia muciniphila blunts colorectal tumourigenesis by reprogramming tumour microenvironment. Gut, 72, 1308-1318.

21.

Kang CH, Jung ES, Jung SJ, Han YH, Chae SW, Jeong DY, Kim BC, Lee SO and Yoon SJ (2024) Pasteurized Akkermansia muciniphila HB05 (HB05P) improves muscle strength and function: a 12-week, randomized, double-blind, placebo-controlled clinical trial. Nutrients, 16.

22.

Lee HW, Lee SN, Seo JG, Koo Y, Kang SY, Choi CW, Park SY, Lee SY, Kim SR, Kim JH and Choi HS (2024) Efficacy of ETB-F01, heat-killed Akkermansia muciniphila strain EB-AMDK19, in patients with respiratory symptoms: A multicenter clinical trial. Nutrients, 16.

23.

Lei W, Cheng Y, Gao J, Liu X, Shao L, Kong Q, Zheng N, Ling Z and Hu W (2023) Akkermansia muciniphila in neuropsychiatric disorders: friend or foe? Frontiers in Cellular and Infection Microbiology, 13, 1224155.

24.

Li J, Lin S, Vanhoutte PM, Woo CW and Xu A (2016) Akkermansia muciniphila protects against atherosclerosis by preventing metabolic endotoxemia-induced inflammation in Apoe−/− mice. Circulation, 133, 2434-2446.

25.

Li J, Yang G, Zhang Q, Liu Z, Jiang X and Xin Y (2023) Function of Akkermansia muciniphila in type 2 diabetes and related diseases. Frontiers in Microbiology, 14, 1172400.

26.

Liu E, Ji X and Zhou K (2024) Akkermansia muciniphila for the prevention of type 2 diabetes and obesity: A meta-analysis of animal studies. Nutrients, 16, 3440.

27.

Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK and Knight R (2012) Diversity, stability and resilience of the human gut microbiota. Nature, 489, 220-230.

28.

Luo Y, Zhang Y, Han X, Yuan Y, Zhou Y, Gao Y, Yu H, Zhang J, Shi Y, Duan Y, Zhao X, Yan S, Hao H, Dai C, Zhao S, Shi J, Li W, Zhang S, Xu W, Fang N, Gong Y and Li Y (2022) Akkermansia muciniphila prevents cold-related atrial fibrillation in rats by modulation of TMAO-induced cardiac pyroptosis. EBioMedicine, 82, 104087.

29.

Mo C, Lou X, Xue J, Shi Z, Zhao Y, Wang F and Chen G (2024) The influence of Akkermansia muciniphila on intestinal barrier function. Gut Pathogens, 16.

30.

Mueller KD, Panzetta ME, Davey L, McCann JR, Rawls JF, Flores GE and Valdivia RH (2024) Pangenomic analysis identifies correlations between Akkermansia species and subspecies and human health outcomes. Microbiome Research Reports, 3.

31.

Ottman N, Davids M, Suarez-Diez M, Boeren S, Schaap PJ, Martins dos Santos VAP, Smidt H, Belzer C and de Vos WM (2017) Genome-scale model and omics analysis of metabolic capacities of Akkermansia muciniphila reveal a preferential mucin-degrading lifestyle. Applied and Environmental Microbiology, 83, e01014-17.

32.

Ou Z, Deng L, Lu Z, Wu F, Liu W, Huang D and Peng Y (2020) Protective effects of Akkermansia muciniphila on cognitive deficits and amyloid pathology in a mouse model of Alzheimer’s disease. Nutrition & Diabetes. 10, 12.

33.

Perraudeau F, McMurdie P, Bullard J, Cheng A, Cutcliffe C, Deo A, Eid J, Gines J, Iyer M, Justice N, Loo WT, Nemchek M, Schicklberger M, Souza M, Stoneburner B, Tyagi S and Kolterman O (2020) Improvements to postprandial glucose control in subjects with type 2 diabetes: a multicenter, double blind, randomized placebo-controlled trial of a novel probiotic formulation. BMJ Open Diabetes Research and Care, 8, e001319.

34.

Plovier H, Everard A, Druart C, Depommier C, Van Hul M, Geurts L, Chilloux J, Ottman N, Duparc T, Lichtenstein L, Myridakis A, Delzenne NM, Klievink J, Bhattacharjee A, van der Ark KCH, Aalvink S, Martinez LO, Dumas M-E, Maiter D and Cani PD (2017) A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nature Medicine, 23, 107-113.

35.

Psichas A, Sleeth ML, Murphy KG, Brooks L, Bewick GA, Hanyaloglu AC, Ghatei MA, Bloom SR and Frost G (2015) The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. International Journal of Obesity, 39, 424-429.

36.

Rao Y, Kuang Z, Li C, Guo S, Xu Y, Zhao D, Hu Y, Song B, Jiang Z, Ge Z, Liu X, Li C, Chen S, Ye J, Huang Z, Lu Y (2021) Gut Akkermansia muciniphila ameliorates metabolic dysfunction-associated fatty liver disease by regulating the metabolism of L-aspartate via gut-liver axis. Gut Microbes, 13, e1927633.

37.

Schrager J (1970) The chemical composition and function of gastrointestinal mucus. Gut, 11, 450.

38.

Shi Z, Lei H, Chen G, Yuan P, Cao Z, Ser HL, Zhu X, Wu F, Liu C, Dong M, Song Y, Guo Y, Chen C, Hu K, Zhu Y, Zeng XA, Zhou J, Lu Y, Patterson AD, Zhang L. Impaired Intestinal Akkermansia muciniphila and aryl hydrocarbon receptor ligands contribute to nonalcoholic fatty liver disease in mice. mSystems6, e00985-20.

39.

Sun Y, Zhu H, Cheng R, Tang Z and Zhang M (2023) Outer membrane protein Amuc_1100 of Akkermansia muciniphila alleviates antibiotic-induced anxiety and depression-like behavior in mice. Physiology & Behavior, 258, 114023.

40.

Takewaki D, Suda W, Sato W, Takayasu L, Kumar N, Kimura K, Kaga N, Mizuno T, Miyake S, Hattori M and Yamamura T (2020) Alterations of the gut ecological and functional microenvironment in different stages of multiple sclerosis. Proceedings of the National Academy of Sciences, 117, 22402-22412.

41.

van Passel MWJ, Kant R, Zoetendal EG, Plugge CM, Derrien M, Malfatti SA, Chain PSG, Woyke T, Palva A, de Vos WM and Smidt H (2011) The genome of Akkermansia muciniphila, a dedicated intestinal mucin degrader, and its use in exploring intestinal metagenomes. PLoS ONE, 6, e16876.

42.

Wang J, Xu W, Wang R, Cheng R, Tang Z and Zhang M (2021) The outer membrane protein Amuc_1100 of Akkermansia muciniphila promotes intestinal 5-HT biosynthesis and extracellular availability through TLR2 signalling. Food & Function, 12, 3597-3610.

43.

Wang L, Tang L, Feng Y, Zhao S, Han M, Zhang C, Yuan G, Zhu J, Cao S, Wu Q, Li L and Zhang Z (2020) A purified membrane protein from Akkermansia muciniphila or the pasteurised bacterium blunts colitis associated tumourigenesis by modulation of CD8+ T cells in mice. Gut, 69, 1988.

44.

Xiao X, Wu Y, Jie Z, Lin L, Li Y, Hu W, Li Y and Zhong S (2024) Akkermansia muciniphila supplementation improves hyperlipidemia, cardiac function, and gut microbiota in high fat fed apolipoprotein E–deficient mice. Prostaglandins & Other Lipid Mediators, 175, 106906.

45.

Xie C and Prasad AA (2020) Probiotics treatment improves hippocampal dependent cognition in a rodent model of Parkinson’s disease. Microorganisms, 8, 1661.

46.

Yan J, Pan Y, Shao W, Wang C, Wang R, He Y, Zhang M, Wang Y, Li T, Wang Z, Liu W, Wang Z, Sun X and Dong S (2022) Beneficial effect of the short-chain fatty acid propionate on vascular calcification through intestinal microbiota remodelling. Microbiome, 10, 195.

47.

Yan S, Chen L, Li N, Wei X, Wang J, Dong W, Wang Y, Shi J, Ding X and Peng Y (2024) Effect of Akkermansia muciniphila on pancreatic islet β-cell function in rats with prediabetes mellitus induced by a high-fat diet. Bioresources and Bioprocessing, 11, 51.

48.

Yang Y, Zhong Z, Wang B, Xia X, Yao W, Huang L, Wang Y and Ding W (2019) Early-life high-fat diet-induced obesity programs hippocampal development and cognitive functions via regulation of gut commensal Akkermansia muciniphila. Neuropsychopharmacology, 44, 2054-2064.

49.

Yoon HS, Cho CH, Yun MS, Jang SJ, You HJ, Kim J, Han D, Cha KH, Moon SH, Lee K, Kim Y-J, Lee S-J, Nam T-W and Ko G (2021) Akkermansia muciniphila secretes a glucagon-like peptide-1-inducing protein that improves glucose homeostasis and ameliorates metabolic disease in mice. Nature Microbiology, 6, 563-573.

50.

Zhang J, Ni Y, Qian L, Fang Q, Zheng T, Zhang M, Gao Q, Zhang Y, Ni J, Hou X, Bao Y, Kovatcheva-Datchary P, Xu A, Li H, Panagiotou G and Jia W (2021) Decreased abundance of Akkermansia muciniphila leads to the impairment of insulin secretion and glucose homeostasis in lean type 2 diabetes. Advanced Science, 8, 2100536.

51.

Zhao Y, Yang H, Wu P, Yang S, Xue W, Xu B, Zhang S, Tang B and Xu D (2024) Akkermansia muciniphila: A promising probiotic against inflammation and metabolic disorders. Virulence, 15.