1
|
Zhao Y, Yang H, Wu P, Yang S, Xue W, Xu B, Zhang S, Tang B, Xu D. Akkermansia muciniphila: A promising probiotic against inflammation and metabolic disorders. Virulence 2024; 15:2375555. [PMID: 39192579 PMCID: PMC11364076 DOI: 10.1080/21505594.2024.2375555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/13/2024] [Accepted: 06/28/2024] [Indexed: 08/29/2024] Open
Abstract
Metabolic disease is a worldwide epidemic that has become a public health problem. Gut microbiota is considered to be one of the important factors that maintain human health by regulating host metabolism. As an abundant bacterium in the host gut, A. muciniphila regulates metabolic and immune functions, and protects gut health. Multiple studies have indicated that alterations in the abundance of A. muciniphila are associated with various diseases, including intestinal inflammatory diseases, obesity, type 2 diabetes mellitus, and even parasitic diseases. Beneficial effects were observed not only in live A. muciniphila, but also in pasteurized A. muciniphila, A. muciniphila-derived extracellular vesicles, outer membrane, and secreted proteins. Although numerous studies have only proven the simple correlation between multiple diseases and A. muciniphila, an increasing number of studies in animal models and preclinical models have demonstrated that the beneficial impacts shifted from correlations to in-depth mechanisms. In this review, we provide a comprehensive view of the beneficial effects of A. muciniphila on different diseases and summarize the potential mechanisms of action of A. muciniphila in the treatment of diseases. We provide a comprehensive understanding of A. muciniphila for improving host health and discuss the perspectives of A. muciniphila in the future studies.
Collapse
Affiliation(s)
- Yanqing Zhao
- Department of Human Parasitology, School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Huijun Yang
- The First School of Clinical Medicine, Hubei University of Medicine, Shiyan, Hubei, China
| | - Peng Wu
- Department of Human Parasitology, School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shuguo Yang
- Department of Human Parasitology, School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Wenkun Xue
- Department of Human Parasitology, School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Biao Xu
- Department of Human Parasitology, School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Sirui Zhang
- Department of Human Parasitology, School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Bin Tang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Daoxiu Xu
- Department of Human Parasitology, School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
2
|
Ta LP, Corrigan S, Horniblow RD. Novel pectin-carboxymethylcellulose-based double-layered mucin/chitosan microcomposites successfully protect the next-generation probiotic Akkermansia muciniphila through simulated gastrointestinal transit and alter microbial communities within colonic ex vivo bioreactors. Int J Pharm 2024; 665:124670. [PMID: 39244071 DOI: 10.1016/j.ijpharm.2024.124670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
The rapid acceleration of microbiome research has identified many potential Next Generation Probiotics (NGPs). Conventional formulation processing methods are non-compatible, leading to reduced viability and unconfirmed incorporation into intestinal microbial communities; consequently, demand for more bespoke formulation strategies of such NGPs is apparent. In this study, Akkermansia muciniphila (A.muciniphila) as a candidate NGP was investigated for its growth and metabolism properties, based on which a novel microcomposite-based oral formulation was formed. Initially, a chitosan-based microcomposite was coated with mucin to establish a surface culture of A.muciniphila. This was followed by 'double encapsulation' with pectin (PEC) using a novel Entrapment Deposition by Prilling method to create core-shell double-encapsulated microcapsules. The formulation of A.muciniphila was verified to require no oxygen-restriction properties, and additionally, biopolymers were selected, including carboxymethylcellulose (CMC), that support and enhance its growth; consequently, a high viability (6 log CFU/g) of A.muciniphila microencapsulated in PEC-CMC double-encapsulates was obtained. Subsequently, the high stability of the PEC-CMC double-encapsulates was verified in simulated gastric fluid, successfully protecting and then releasing the A.muciniphila under intestinal conditions. Finally, employing a model of gastrointestinal transit and faecal-inoculated colonic bioreactors, significant alterations in microbial communities following administration and successful establishment of A.muciniphila were demonstrated.
Collapse
Affiliation(s)
- Linh Phuong Ta
- Department of Biomedical Sciences, School of Infection, Inflammation, and Immunology, College of Medicine and Health, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Sarah Corrigan
- Department of Biomedical Sciences, School of Infection, Inflammation, and Immunology, College of Medicine and Health, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Richard D Horniblow
- Department of Biomedical Sciences, School of Infection, Inflammation, and Immunology, College of Medicine and Health, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
3
|
Xu X, Liu X, Liu L, Chen J, Guan J, Luo D. Metagenomic and transcriptomic profiling of the hypoglycemic and hypotriglyceridemic actions of Tremella fuciformis-derived polysaccharides in high-fat-diet- and streptozotocin-treated mice. Food Funct 2024. [PMID: 39432083 DOI: 10.1039/d4fo01870b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Mushroom polysaccharides have great anti-diabetes potential. The fruiting body of Tremella fuciformis is rich in polysaccharides. However, few studies have been performed to date on T. fuciformis-derived polysaccharides (TPs) in terms of anti-diabetes potential. Our previous studies showed that novel TPs with medium molecular weights exhibited the highest anti-skin aging activities among the tested samples in D-galactose-treated mice. In the present study, the effects of these novel TPs, named TP, on high-fat-diet- and streptozotocin-treated mice were assessed, and their potential biological mechanisms were explored by metagenomic and transcriptomic analyses. Oral administration of TP markedly reduced blood glucose and TG levels, alleviated emaciation, improved anti-oxidant capacity, and protected the functions of β-cells at a dose of 100 mg kg-1 in diabetic mice. Meanwhile, the taxonomic compositions and functional properties of fecal microbiota were altered considerably by TP, as evidenced by partial restoration of the imbalanced gut microbiota and the higher abundances of Bacteroides, Phocaeicola, Bifidobacterium, and Alistipes compared to the model mice, corresponding to the upregulation of four enriched KEGG pathways of microbial communities such as the digestive system, cardiovascular disease, parasitic infectious disease, and cell growth and death. Further transcriptomic analysis of liver tissues identified 35 enriched KEGG pathways associated with metabolism and cellular signaling processes in response to TP. These results demonstrated the biological mechanisms underlying the hypoglycemic and hypotriglyceridemic activities of TP. The findings expanded our understanding of the anti-diabetic mechanisms for mushroom polysaccharides and provided new clues for future studies.
Collapse
Affiliation(s)
- Xiaofei Xu
- College of Food Science and Engineering, Guangdong Ocean University, 1# Luoqin Road, Yangjiang 529500, China.
| | - Xiaofei Liu
- College of Food Science and Engineering, Guangdong Ocean University, 1# Luoqin Road, Yangjiang 529500, China.
| | - Liyan Liu
- College of Food Science and Engineering, Guangdong Ocean University, 1# Luoqin Road, Yangjiang 529500, China.
| | - Jin Chen
- College of Food Science and Engineering, Guangdong Ocean University, 1# Luoqin Road, Yangjiang 529500, China.
| | - Jingjing Guan
- College of Food Science and Engineering, Guangdong Ocean University, 1# Luoqin Road, Yangjiang 529500, China.
| | - Donghui Luo
- College of Food Science and Engineering, Guangdong Ocean University, 1# Luoqin Road, Yangjiang 529500, China.
| |
Collapse
|
4
|
Keshavarz Aziziraftar S, Bahrami R, Hashemi D, Shahryari A, Ramezani A, Ashrafan F, Siadat SD. The beneficial effects of Akkermansia muciniphila and its derivatives on pulmonary fibrosis. Biomed Pharmacother 2024; 180:117571. [PMID: 39418965 DOI: 10.1016/j.biopha.2024.117571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Pulmonary fibrosis (PF) is a progressive and debilitating respiratory condition characterized by excessive deposition of extracellular matrix proteins and scarring within the lung parenchyma. Despite extensive research, the pathogenesis of PF remains incompletely understood, and effective therapeutic options are limited. Emerging evidence suggests a potential link between gut microbiota dysbiosis and the development of PF, highlighting the gut-lung axis as a promising therapeutic target. Akkermansia muciniphila (A. muciniphila), a mucin-degrading bacterium residing in the gut mucosal layer, has garnered considerable interest due to its immunomodulatory and anti-inflammatory properties. This study investigates the therapeutic potential of live and pasteurized A. muciniphila, as well as its extracellular vesicles (EVs), in mitigating inflammation and fibrosis in a murine model of carbon tetrachloride (CCl4)-induced PF exacerbated by a high-fat diet (HFD). Male C57BL/6 mice were divided into groups receiving either a normal diet or an HFD, with or without CCl4 administration. The mice were then treated with live or pasteurized A. muciniphila, or its EVs. Lung tissue was analyzed for the expression of inflammatory markers and fibrosis markers using real-time PCR and ELISA. Administration of live and pasteurized A. muciniphila, as well as its EVs, significantly downregulated the expression of inflammatory and fibrosis markers in the lung tissue of CCl4-induced PF mice. Furthermore, these treatments ameliorated the increased production of IL-6 and reduced IL-10 levels observed in the HFD and CCl4-treated groups. These findings suggest that A. muciniphila and its derivatives exert protective effects against pulmonary inflammation and fibrosis, potentially through modulation of the gut-lung axis. The study highlights the therapeutic potential of A. muciniphila and its derivatives as novel interventions for the management of PF, warranting further preclinical and clinical investigations.
Collapse
Affiliation(s)
- Shahrbanoo Keshavarz Aziziraftar
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran; Department of Pathology, University of California San Francisco, San Francisco, US.
| | - Romina Bahrami
- B.S, Department of Microbiology and Microbial Biotech, Shahid Beheshti University, Tehran, Iran.
| | - Danial Hashemi
- B.S, Department of Animal Science and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Arefeh Shahryari
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran; Centennial College School of Engineering Technology and Applied Science Biotechnology Program Toronto, Ontario, Canada.
| | - Amitis Ramezani
- Clinical Research Department, Pasteur Institute of Iran, Tehran, Iran.
| | - Fatemeh Ashrafan
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran; Clinical Research Department, Pasteur Institute of Iran, Tehran, Iran.
| | - Seyed Davar Siadat
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran; Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
5
|
Ratnayani, Hegar B, Sunardi D, Fadilah F, Gunardi H, Fahmida U, Vidiawati D. Association of Gut Microbiota Composition with Stunting Incidence in Children under Five in Jakarta Slums. Nutrients 2024; 16:3444. [PMID: 39458441 PMCID: PMC11510009 DOI: 10.3390/nu16203444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Stunting can be linked to various factors, one of which is dysbiosis. This study aims to analyze the microbiota composition and related contributing factors of stunted and non-stunted children in the slum areas of Jakarta. METHODS The subjects in this study included 21 stunted (HAZ ≤ -2SD) and 21 non-stunted children (-2SD ≤ HAZ ≤ 3SD) aged 2-5 years. Microbiota analysis was performed by extracting DNA from the subjects' feces and then via 16S rRNA sequencing using next-generation sequencing (NGS). RESULTS The results of this study showed that in stunted children, the abundance of Mitsuokella (24,469 OTUs), Alloprevotella (23,952 OTUs), and Providencia alcalifaciens (861 OTUs) was higher, while in non-stunted children, that of Blautia (29,755 OTUs), Lachnospiraceae (6134 OTUs), Bilophila (12,417 OTUs), Monoglobus (484 OTUs), Akkermansia muciniphila (1116 OTUs), Odoribacter splanchnicus (42,993 OTUs), and Bacteroides clarus (8900 OTUs) was higher. Differences in microbiota composition in the two groups were influenced by nutrient intake, birth history, breastfeeding history, handwashing habits before eating, drinking water sources, and water sources for other activities. CONCLUSIONS This study highlights that stunted children have a significantly different gut microbiota composition compared to non-stunted children, with higher levels of pathogenic bacteria and lower levels of beneficial bacteria. Future research should focus on interventions that can improve the gut microbiota composition to prevent stunting in children.
Collapse
Affiliation(s)
- Ratnayani
- Department of Nutrition, Faculty of Medicine, Universitas Indonesia—Dr. Cipto Mangunkusumo General Hospital, Jakarta 10430, Indonesia; (R.); (D.S.); (U.F.)
- Nutrition Study Program, Faculty of Health Sciences and Technology, Binawan University, Jakarta 13630, Indonesia
| | - Badriul Hegar
- Department of Child Health, Faculty of Medicine, Universitas Indonesia—Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia;
| | - Diana Sunardi
- Department of Nutrition, Faculty of Medicine, Universitas Indonesia—Dr. Cipto Mangunkusumo General Hospital, Jakarta 10430, Indonesia; (R.); (D.S.); (U.F.)
| | - Fadilah Fadilah
- Department of Chemistry, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
- Bioinformatics Core Facilities, Institute of Medical Education and Research Indonesia (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Hartono Gunardi
- Department of Child Health, Faculty of Medicine, Universitas Indonesia—Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia;
| | - Umi Fahmida
- Department of Nutrition, Faculty of Medicine, Universitas Indonesia—Dr. Cipto Mangunkusumo General Hospital, Jakarta 10430, Indonesia; (R.); (D.S.); (U.F.)
- Southeast Asian Ministers of Education Organization Regional Centre for Food and Nutrition (SEAMEO RECFON), Jakarta 13120, Indonesia
| | - Dhanasari Vidiawati
- Department of Community Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| |
Collapse
|
6
|
Liu E, Ji X, Zhou K. Akkermansia muciniphila for the Prevention of Type 2 Diabetes and Obesity: A Meta-Analysis of Animal Studies. Nutrients 2024; 16:3440. [PMID: 39458436 PMCID: PMC11510203 DOI: 10.3390/nu16203440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND More than half of the states in the U.S. report that over 30% of adults are obese. Obesity increases the risk of many chronic diseases, including type 2 diabetes, hypertension, and cardiovascular disease, and can even reduce one's lifespan. Similarly, the prevalence of type 2 diabetes follows a comparable trend. As a result, researchers are striving to find solutions to reduce obesity rates, with a particular focus on gut health, which has been previously linked to both obesity and type 2 diabetes. Recent studies suggest that Akkermansia muciniphila (Akk) may have a positive probiotic effect on preventing the onset of type 2 diabetes and obesity. METHODS We conducted a quantitative meta-analysis of 15 qualified animal studies investigating the effects of Akk administration as a probiotic. RESULTS The statistical analyses showed that Akk administration significantly reduced body weight gain by 10.4% and fasting blood glucose by 21.2%, while also significantly improving glucose tolerance by 22.1% and increasing blood insulin levels by 26.9%. However, our analysis revealed substantial heterogeneity between the control and experimental groups across all subgroups. CONCLUSIONS Overall, Akk appears to be effective at reducing the onset of type 2 diabetes and diet-induced obesity. Long-term studies with larger sample sizes are needed to confirm these beneficial effects, as the current animal studies were of short duration (less than 20 weeks).
Collapse
Affiliation(s)
- Ethan Liu
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA;
| | - Xiangming Ji
- Department of Nutritional Sciences, The College of Health and Human Development, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kequan Zhou
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA;
| |
Collapse
|
7
|
Anderson MH, Ait-Aissa K, Sahyoun AM, Abidi AH, Kassan M. Akkermansia muciniphila as a Potential Guardian against Oral Health Diseases: A Narrative Review. Nutrients 2024; 16:3075. [PMID: 39339675 PMCID: PMC11434887 DOI: 10.3390/nu16183075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
The oral microbiome is a diverse ecosystem containing a community of symbiotic, commensal, and pathogenic microorganisms. One key microorganism linked to periodontal disease (PD) is Porphyromonas gingivalis (P. gingivalis), a Gram-negative anaerobic bacterium known to have several virulence factors that trigger inflammation and immune evasion. On the other hand, Akkermansia muciniphila (A. muciniphila), a symbiotic bacterium, has been recently shown to play an important role in mitigating inflammation and reducing periodontal damage. In vivo and in vitro studies have shown that A. muciniphila decreases inflammatory mediators and improves immune responses, suggesting its role in mitigating PD and related inflammatory systemic conditions such as diabetes, hypertension, and obesity. This review discusses the anti-inflammatory effects of A. muciniphila, its impact on periodontal health, and its potential role in managing systemic diseases. The overall aim is to elucidate how this bacterium might help reduce inflammation, improve oral health, and influence broader health outcomes.
Collapse
Affiliation(s)
- Molly H Anderson
- College of Dental Medicine, Lincoln Memorial University, LMU Tower, 1705 St. Mary Street, Knoxville, TN 37917, USA
| | - Karima Ait-Aissa
- College of Dental Medicine, Lincoln Memorial University, LMU Tower, 1705 St. Mary Street, Knoxville, TN 37917, USA
| | - Amal M Sahyoun
- College of Dental Medicine, Lincoln Memorial University, LMU Tower, 1705 St. Mary Street, Knoxville, TN 37917, USA
| | - Ammaar H Abidi
- College of Dental Medicine, Lincoln Memorial University, LMU Tower, 1705 St. Mary Street, Knoxville, TN 37917, USA
| | - Modar Kassan
- College of Dental Medicine, Lincoln Memorial University, LMU Tower, 1705 St. Mary Street, Knoxville, TN 37917, USA
| |
Collapse
|
8
|
Khalili L, Park G, Nagpal R, Salazar G. The Role of Akkermansia muciniphila on Improving Gut and Metabolic Health Modulation: A Meta-Analysis of Preclinical Mouse Model Studies. Microorganisms 2024; 12:1627. [PMID: 39203469 PMCID: PMC11356609 DOI: 10.3390/microorganisms12081627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
Akkermansia muciniphila (A. muciniphila) and its derivatives, including extracellular vesicles (EVs) and outer membrane proteins, are recognized for enhancing intestinal balance and metabolic health. However, the mechanisms of Akkermansia muciniphila's action and its effects on the microbiome are not well understood. In this study, we examined the influence of A. muciniphila and its derivatives on gastrointestinal (GI) and metabolic disorders through a meta-analysis of studies conducted on mouse models. A total of 39 eligible studies were identified through targeted searches on PubMed, Web of Science, Science Direct, and Embase until May 2024. A. muciniphila (alive or heat-killed) and its derivatives positively affected systemic and gut inflammation, liver enzyme level, glycemic response, and lipid profiles. The intervention increased the expression of tight-junction proteins in the gut, improving gut permeability in mouse models of GI and metabolic disorders. Regarding body weight, A. muciniphila and its derivatives prevented weight loss in animals with GI disorders while reducing body weight in mice with metabolic disorders. Sub-group analysis indicated that live bacteria had a more substantial effect on most analyzed biomarkers. Gut microbiome analysis using live A. muciniphila identified a co-occurrence cluster, including Desulfovibrio, Family XIII AD3011 group, and Candidatus Saccharimonas. Thus, enhancing the intestinal abundance of A. muciniphila and its gut microbial clusters may provide more robust health benefits for cardiometabolic, and age-related diseases compared with A. muciniphila alone. The mechanistic insight elucidated here will pave the way for further exploration and potential translational applications in human health.
Collapse
Affiliation(s)
- Leila Khalili
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
| | - Gwoncheol Park
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
| | - Ravinder Nagpal
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
| | - Gloria Salazar
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
- Center for Advancing Exercise and Nutrition Research on Aging (CAENRA), Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
9
|
Mo C, Lou X, Xue J, Shi Z, Zhao Y, Wang F, Chen G. The influence of Akkermansia muciniphila on intestinal barrier function. Gut Pathog 2024; 16:41. [PMID: 39097746 PMCID: PMC11297771 DOI: 10.1186/s13099-024-00635-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 07/20/2024] [Indexed: 08/05/2024] Open
Abstract
Intestinal barriers play a crucial role in human physiology, both in homeostatic and pathological conditions. Disruption of the intestinal barrier is a significant factor in the pathogenesis of gastrointestinal inflammatory diseases, such as inflammatory bowel disease. The profound influence of the gut microbiota on intestinal diseases has sparked considerable interest in manipulating it through dietary interventions, probiotics, and fecal microbiota transplantation as potential approaches to enhance the integrity of the intestinal barrier. Numerous studies have underscored the protective effects of specific microbiota and their associated metabolites. In recent years, an increasing body of research has demonstrated that Akkermansia muciniphila (A. muciniphila, Am) plays a beneficial role in various diseases, including diabetes, obesity, aging, cancer, and metabolic syndrome. It is gaining popularity as a regulator that influences the intestinal flora and intestinal barrier and is recognized as a 'new generation of probiotics'. Consequently, it may represent a potential target and promising therapy option for intestinal diseases. This article systematically summarizes the role of Am in the gut. Specifically, we carefully discuss key scientific issues that need resolution in the future regarding beneficial bacteria represented by Am, which may provide insights for the application of drugs targeting Am in clinical treatment.
Collapse
Affiliation(s)
- Chunyan Mo
- Medical School, Kunming University of Science and Technology, 727 Jingming South Road, Chenggong District, Kunming, 650500, China
| | - Xiran Lou
- Medical School, Kunming University of Science and Technology, 727 Jingming South Road, Chenggong District, Kunming, 650500, China
| | - Jinfang Xue
- Medical School, Kunming University of Science and Technology, 727 Jingming South Road, Chenggong District, Kunming, 650500, China
| | - Zhuange Shi
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan District, Kunming, 650034, China
| | - Yifang Zhao
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan District, Kunming, 650034, China
| | - Fuping Wang
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan District, Kunming, 650034, China
| | - Guobing Chen
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan District, Kunming, 650034, China.
| |
Collapse
|
10
|
Mruk-Mazurkiewicz H, Kulaszyńska M, Czarnecka W, Podkówka A, Ekstedt N, Zawodny P, Wierzbicka-Woś A, Marlicz W, Skupin B, Stachowska E, Łoniewski I, Skonieczna-Żydecka K. Insights into the Mechanisms of Action of Akkermansia muciniphila in the Treatment of Non-Communicable Diseases. Nutrients 2024; 16:1695. [PMID: 38892628 PMCID: PMC11174979 DOI: 10.3390/nu16111695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
This comprehensive review delineates the extensive roles of Akkermansia muciniphila in various health domains, spanning from metabolic and inflammatory diseases to neurodegenerative disorders. A. muciniphila, known for its ability to reside in the mucous layer of the intestine, plays a pivotal role in maintaining gut integrity and interacting with host metabolic processes. Its influence extends to modulating immune responses and potentially easing symptoms across several non-communicable diseases, including obesity, diabetes, inflammatory bowel disease, and cancer. Recent studies highlight its capacity to interact with the gut-brain axis, suggesting a possible impact on neuropsychiatric conditions. Despite the promising therapeutic potential of A. muciniphila highlighted in animal and preliminary human studies, challenges remain in its practical application due to stability and cultivation issues. However, the development of pasteurized forms and synthetic mediums offers new avenues for its use in clinical settings, as recognized by regulatory bodies like the European Food Safety Authority. This narrative review serves as a crucial resource for understanding the broad implications of A. muciniphila across different health conditions and its potential integration into therapeutic strategies.
Collapse
Affiliation(s)
- Honorata Mruk-Mazurkiewicz
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Monika Kulaszyńska
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Wiktoria Czarnecka
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Albert Podkówka
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Natalia Ekstedt
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Piotr Zawodny
- Medical Center Zawodny Clinic, Ku Słońcu 58, 71-047 Szczecin, Poland;
| | | | - Wojciech Marlicz
- Department of Gastroenterology, Pomeranian Medical University in Szczecin, Unii Lubelskiej, 71-252 Szczecin, Poland
| | - Błażej Skupin
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland
| | - Igor Łoniewski
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| | - Karolina Skonieczna-Żydecka
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland (N.E.); (I.Ł.)
| |
Collapse
|
11
|
Sundaram K, Teng Y, Mu J, Xu Q, Xu F, Sriwastva MK, Zhang L, Park JW, Zhang X, Yan J, Zhang SQ, Merchant ML, Chen SY, McClain CJ, Dryden GW, Zhang HG. Outer Membrane Vesicles Released from Garlic Exosome-like Nanoparticles (GaELNs) Train Gut Bacteria that Reverses Type 2 Diabetes via the Gut-Brain Axis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308680. [PMID: 38225709 PMCID: PMC11102339 DOI: 10.1002/smll.202308680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/19/2023] [Indexed: 01/17/2024]
Abstract
Gut microbiota function has numerous effects on humans and the diet humans consume has emerged as a pivotal determinant of gut microbiota function. Here, a new concept that gut microbiota can be trained by diet-derived exosome-like nanoparticles (ELNs) to release healthy outer membrane vesicles (OMVs) is introduced. Specifically, OMVs released from garlic ELN (GaELNs) trained human gut Akkermansia muciniphila (A. muciniphila) can reverse high-fat diet-induced type 2 diabetes (T2DM) in mice. Oral administration of OMVs released from GaELNs trained A. muciniphila can traffick to the brain where they are taken up by microglial cells, resulting in inhibition of high-fat diet-induced brain inflammation. GaELNs treatment increases the levels of OMV Amuc-1100, P9, and phosphatidylcholines. Increasing the levels of Amuc-1100 and P9 leads to increasing the GLP-1 plasma level. Increasing the levels of phosphatidylcholines is required for inhibition of cGas and STING-mediated inflammation and GLP-1R crosstalk with the insulin pathway that leads to increasing expression of Insulin Receptor Substrate (IRS1 and IRS2) on OMV targeted cells. These findings reveal a molecular mechanism whereby OMVs from plant nanoparticle-trained gut bacteria regulate genes expressed in the brain, and have implications for the treatment of brain dysfunction caused by a metabolic syndrome.
Collapse
Affiliation(s)
- Kumaran Sundaram
- Brown Cancer Center, University of Louisville, Louisville, KY40202, USA
| | - Yun Teng
- Brown Cancer Center, University of Louisville, Louisville, KY40202, USA
| | - Jingyao Mu
- Brown Cancer Center, University of Louisville, Louisville, KY40202, USA
| | - Qingbo Xu
- Department of Microbiology & Immunology, University of Louisville, Louisville, KY40202, USA
| | - Fangyi Xu
- Brown Cancer Center, University of Louisville, Louisville, KY40202, USA
| | | | - Lifeng Zhang
- Brown Cancer Center, University of Louisville, Louisville, KY40202, USA
| | - Juw Won Park
- Department of Computer Science and Engineering, University of Louisville, Louisville, KY40202, USA
- Kentucky IDeA Network of Biomedical Research Excellence Bioinformatics Core, University of Louisville, Louisville, KY 40202, USA
| | - Xiang Zhang
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - Jun Yan
- Brown Cancer Center, University of Louisville, Louisville, KY40202, USA
| | - Shuang Qin Zhang
- Section of Hematology/Oncology, University of Chicago, Chicago, IL 60637
| | - Michael L. Merchant
- Kidney Disease Program and Clinical Proteomics Center, University of Louisville, Louisville, KY, USA
| | - Shao-yu Chen
- Brown Cancer Center, University of Louisville, Louisville, KY40202, USA
| | - Craig J McClain
- Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - Gerald W Dryden
- Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - Huang-Ge Zhang
- Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA
- Brown Cancer Center, University of Louisville, Louisville, KY40202, USA
- Department of Microbiology & Immunology, University of Louisville, Louisville, KY40202, USA
| |
Collapse
|
12
|
Zhong S, Yang J, Huang H. Efficacy Assessment of the Co-Administration of Vancomycin and Metronidazole in Clostridioides difficile-Infected Mice Based on Changes in Intestinal Ecology. J Microbiol Biotechnol 2024; 34:828-837. [PMID: 38668685 DOI: 10.4014/jmb.2312.12034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 05/16/2024]
Abstract
Vancomycin (VAN) and metronidazole (MTR) remain the current drugs of choice for the treatment of non-severe Clostridioides difficile infection (CDI); however, while their co-administration has appeared in clinical treatment, the efficacy varies greatly and the mechanism is unknown. In this study, a CDI mouse model was constructed to evaluate the therapeutic effects of VAN and MTR alone or in combination. For a perspective on the intestinal ecology, 16S rRNA amplicon sequencing and non-targeted metabolomics techniques were used to investigate changes in the fecal microbiota and metabolome of mice under the co-administration treatment. As a result, the survival rate of mice under co-administration was not dramatically different compared to that of single antibiotics, and the former caused intestinal tissue hyperplasia and edema. Co-administration also significantly enhanced the activity of amino acid metabolic pathways represented by phenylalanine, arginine, proline, and histidine, decreased the level of deoxycholic acid (DCA), and downregulated the abundance of beneficial microbes, such as Bifidobacterium and Akkermansia. VAN plays a dominant role in microbiota regulation in co-administration. In addition, co-administration reduced or increased the relative abundance of antibiotic-sensitive bacteria, including beneficial and harmful microbes, without a difference. Taken together, there are some risks associated with the co-administration of VAN and MTR, and this combination mode should be used with caution in CDI treatment.
Collapse
Affiliation(s)
- Saiwei Zhong
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, P.R. China
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, P.R. China
| | - Jingpeng Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, P.R. China
| | - He Huang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, P.R. China
| |
Collapse
|
13
|
Sall I, Foxall R, Felth L, Maret S, Rosa Z, Gaur A, Calawa J, Pavlik N, Whistler JL, Whistler CA. Gut dysbiosis was inevitable, but tolerance was not: temporal responses of the murine microbiota that maintain its capacity for butyrate production correlate with sustained antinociception to chronic voluntary morphine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589671. [PMID: 38659831 PMCID: PMC11042308 DOI: 10.1101/2024.04.15.589671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The therapeutic benefits of opioids are compromised by the development of analgesic tolerance, which necessitates higher dosing for pain management thereby increasing the liability for dependence and addiction. Rodent models indicate opposing roles of the gut microbiota in tolerance: morphine-induced gut dysbiosis exacerbates tolerance, whereas probiotics ameliorate tolerance. Not all individuals develop tolerance which could be influenced by differences in microbiota, and yet no study has capitalized upon this natural variation to identify specific features linked to tolerance. We leveraged this natural variation in a murine model of voluntary oral morphine self-administration to elucidate the mechanisms by which microbiota influences tolerance. Although all mice shared similar and predictive morphine-driven microbiota changes that largely masked informative associations with variability in tolerance, our high-resolution temporal analyses revealed a divergence in the progression of dysbiosis that best explained differences in the development in tolerance. Mice that did not develop tolerance also maintained a higher abundance of taxa capable of producing the short-chain fatty acid (SCFA) butyrate, known to bolster intestinal barriers, suppress inflammation, and promote neuronal homeostasis. Furthermore, dietary butyrate supplementation significantly reduced the development of tolerance. These findings could inform immediate therapies to extend the analgesic efficacy of opioids.
Collapse
Affiliation(s)
- Izabella Sall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Graduate program in Molecular and Evolutionary Systems Biology, University of New Hampshire, Durham, NH, USA
| | - Randi Foxall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Lindsey Felth
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Soren Maret
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Zachary Rosa
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Anirudh Gaur
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Jennifer Calawa
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Microbiology Graduate Program, University of New Hampshire, Durham, NH, USA
| | - Nadia Pavlik
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Jennifer L. Whistler
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
- Department of Physiology and Membrane Biology, UC Davis School of Medicine, Davis, CA, USA
| | - Cheryl A. Whistler
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
14
|
Li H, Pu X, Lin Y, Yu X, Li J, Bo L, Wang H, Xu Y, Li X, Zheng D. Sijunzi decoction alleviates inflammation and intestinal epithelial barrier damage and modulates the gut microbiota in ulcerative colitis mice. Front Pharmacol 2024; 15:1360972. [PMID: 38650625 PMCID: PMC11033371 DOI: 10.3389/fphar.2024.1360972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Ethnopharmacological relevance As a representative classical prescription, Sijunzi decoction has powerful therapeutic effects on spleen-stomach qi insufficiency. Ulcerative colitis (UC) is a chronic, diffuse, and non-specifically inflammatory disorder, the etiology of which still remains unclear. In the traditional Chinese medicine (TCM) perspective, splenic asthenia is the primary cause of UC. Based on this, Sijunzi decoction has been extensively used in TCM clinical practice to alleviate UC in recent years. However, the pharmacological mechanism of Sijunzi decoction in modern medicine is still not completely clear, which limits its clinical application. Aim of the study The purpose of this study was to investigate the Sijunzi decoction's curative effect on acute UC mice and probe into its potential pharmacological mechanism. Materials and methods The UC mouse model was set up by freely ingesting a 3% dextran sulfate sodium (DSS) solution. The relieving role of Sijunzi decoction on UC in mice was analyzed by evaluating the changes in clinical parameters, colon morphology, histopathology, inflammatory factor content, intestinal epithelial barrier protein expression level, and gut microbiota balance state. Finally, multivariate statistical analysis was conducted to elucidate the relationship between inflammatory factors, intestinal epithelial barrier proteins, and gut microbiota. Results First, the research findings revealed that Sijunzi decoction could visibly ease the clinical manifestation of UC, lower the DAI score, and attenuate colonic damage. Moreover, Sijunzi decoction could also significantly inhibit IL-6, IL-1β, and TNF-α while increasing occludin and ZO-1 expression levels. Subsequently, further studies showed that Sijunzi decoction could remodel gut microbiota homeostasis. Sijunzi decoction was beneficial in regulating the levels of Alistipes, Akkermansia, Lachnospiraceae_NK4A136_group, and other bacteria. Finally, multivariate statistical analysis demonstrated that key gut microbes were closely associated with inflammatory factors and intestinal epithelial barrier proteins. Conclusion Sijunzi decoction can significantly prevent and treat UC. Its mechanism is strongly associated with the improvement of inflammation and intestinal epithelial barrier damage by regulating the gut microbiota.
Collapse
Affiliation(s)
- Hailun Li
- Department of Nephrology, Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Xing Pu
- Department of Nephrology, Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Yongtao Lin
- School of Nursing and Midwifery, Jiangsu College of Nursing, Huai’an, Jiangsu, China
| | - Xinxin Yu
- Department of Nephrology, Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Jing Li
- Department of Nephrology, Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Lin Bo
- Department of Nephrology, Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Hongwu Wang
- Department of Nephrology, Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Yong Xu
- Department of Nephrology, Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Xiang Li
- Department of Nephrology, Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Donghui Zheng
- Department of Nephrology, Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, China
| |
Collapse
|
15
|
Dedon LR, Yuan H, Chi J, Gu H, Arias AJ, Covault JM, Zhou Y. Baseline gut microbiome and metabolites are correlated with alcohol consumption in a zonisamide clinical trial of heavy drinking alcoholic civilians. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.02.24305199. [PMID: 38633809 PMCID: PMC11023652 DOI: 10.1101/2024.04.02.24305199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Development and severity of alcohol use disorder (AUD) has been linked to variations in gut microbiota and their associated metabolites in both animal and human studies. However, the involvement of the gut microbiome in alcohol consumption of individuals with AUD undergoing treatment remains unclear. To address this, stool samples (n=48) were collected at screening (baseline) and trial completion from a single site of a multi-site double-blind, placebo-controlled trial of Zonisamide in individuals with AUD. Alcohol consumption, gamma-glutamyl transferase (GGT), and phosphatidylethanol (PEth)levels were measured both at baseline and endpoint of 16-week trial period. Fecal microbiome was analyzed via 16S rRNA sequencing and metabolome via untargeted LC-MS. Both sex (p = 0.003) and psychotropic medication usage (p = 0.025) are associated with baseline microbiome composition. The relative abundance of 12 genera at baseline was correlated with percent drinking reduction, baseline and endpoint alcohol consumption, and changes in GGT and PeTH over the course of treatment (p.adj < 0.05). Overall microbiome community structure at baseline differed between high and low responders (67-100% and 0-33% drinking reduction, respectively; p = 0.03). A positive relationship between baseline fecal GABA levels and percent drinking reduction (R=0.43, p < 0.05) was identified by microbiome function prediction and confirmed by ELISA and metabolomics. Predicted microbiome function and metabolomics analysis have found that tryptophan metabolic pathways are over-represented in low responders. These findings highlight importance of baseline microbiome and metabolites in alcohol consumption in AUD patients undergoing zonisamide treatment.
Collapse
|
16
|
Faghfuri E, Gholizadeh P. The role of Akkermansia muciniphila in colorectal cancer: A double-edged sword of treatment or disease progression? Biomed Pharmacother 2024; 173:116416. [PMID: 38471272 DOI: 10.1016/j.biopha.2024.116416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/14/2024] Open
Abstract
Colorectal cancer (CRC) is the second most cancer-related death worldwide. In recent years, probiotics have been used to reduce the potential risks of CRC and tumors with various mechanisms. Different bacteria have been suggested to play different roles in the progression, prevention, or treatment of CRC. Akkermansia muciniphila is considered a next-generation probiotic for preventing and treating some diseases. Therefore, in this review article, we aimed to describe and discuss different mechanisms of A. muciniphila as an intestinal microbiota or probiotic in CRC. Some studies suggested that the abundance of A. muciniphila was higher or increased in CRC patients compared to healthy individuals. However, the decreased abundance of A. muciniphila was associated with severe symptoms of CRC, indicating that A. muciniphila did not play a role in the development of CRC. In addition, A. muciniphila administration elevates gene expression of proliferation-associated molecules such as S100A9, Dbf4, and Snrpd1, or markers for cell proliferation. Some other studies suggested that inflammation and tumorigenesis in the intestine might promoted by A. muciniphila. Overall, the role of A. muciniphila in CRC development or inhibition is still unclear and controversial. Various methods of bacterial supplementation, such as viability, bacterial number, and abundance, could all influence the colonization effect of A. muciniphila administration and CRC progression. Overall, A. mucinipila has been revealed to modulate the therapeutic potential of immune checkpoint inhibitors. Preliminary human data propose that oral consumption of A. muciniphila is safe, but its efficacy needs to be confirmed in more human clinical studies.
Collapse
Affiliation(s)
- Elnaz Faghfuri
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Pourya Gholizadeh
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
17
|
Midya V, Nagdeo K, Lane J, Torres-Olascoaga L, Martínez G, Horton M, Gennings C, Téllez-Rojo M, Wright R, Arora M, Eggers S. Akkermansia muciniphila modifies the association between metal exposure during pregnancy and depressive symptoms in late childhood. RESEARCH SQUARE 2024:rs.3.rs-3922286. [PMID: 38410473 PMCID: PMC10896378 DOI: 10.21203/rs.3.rs-3922286/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Emerging research suggests that exposures to metals during pregnancy and gut microbiome (GM) disruptions are associated with depressive disorders in childhood. Akkermansia muciniphila, a GM bacteria, has been studied for its potential antidepressant effects. However, its role in the influence of prenatal metal exposures on depressive symptoms during childhood is unknown. Leveraging a well-characterized pediatric longitudinal birth cohort and its microbiome substudy (n=112) and using a state-of-the-art machine-learning model, we investigated whether the presence of A.muciniphila in GM of 9-11-year-olds modifies the associations between exposure to a specific group of metals (or metal-clique) during pregnancy and concurrent childhood depressive symptoms. Among children with no A.muciniphila, a metal-clique of Zinc-Chromium-Cobalt was strongly associated with increased depression score (P<0.0001), whereas, for children with A.muciniphila, this same metal-clique was weakly associated with decreased depression score(P<0.4). Our analysis provides the first exploratory evidence hypothesizing A. muciniphila as a probiotic intervention attenuating the effect of prenatal metal-exposures-associated depressive disorders in late childhood.
Collapse
Affiliation(s)
| | | | | | | | - Gabriela Martínez
- Center for Research on Nutrition and Health, National Institute of Public Health
| | | | | | - Martha Téllez-Rojo
- Center for Research on Nutrition and Health, National Institute of Public Health
| | | | | | | |
Collapse
|
18
|
Bu L, Wang C, Bai J, Song J, Zhang Y, Chen H, Suo H. Gut microbiome-based therapies for alleviating cognitive impairment: state of the field, limitations, and future perspectives. Food Funct 2024; 15:1116-1134. [PMID: 38224464 DOI: 10.1039/d3fo02307a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Cognitive impairment (CI) is a multifaceted neurological condition that can trigger negative emotions and a range of concurrent symptoms, imposing significant public health and economic burdens on society. Therefore, it is imperative to discover a remedy for CI. Nevertheless, the mechanisms behind the onset of this disease are multifactorial, which makes the search for effective amelioration difficult and complex, hindering the search for effective measures. Intriguingly, preclinical research indicates that gut microbiota by influencing brain function, plays an important role in the progression of CI. Furthermore, numerous preclinical studies have highlighted the potential of probiotics, prebiotics, fecal microbiota transplantation (FMT), and diet in modulating the gut microbiota, thereby ameliorating CI symptoms. This review provides a comprehensive evaluation of CI pathogenesis, emphasizing the contribution of gut microbiota disorders to CI development. It also summarizes and discusses current strategies and mechanisms centered on the synergistic role of gut microbiota modulation in the microbiota-gut-brain axis in CI development. Finally, problems with existing approaches are contemplated and the development of microbial modulation strategies as therapeutic approaches to promote and restore brain cognition is discussed. Further research considerations and directions are highlighted to provide ideas for future CI prevention and treatment strategies.
Collapse
Affiliation(s)
- Linli Bu
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Junying Bai
- Citrus Research Institute, Southwest University, Chongqing 400715, China
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Yuhong Zhang
- Institute of Food Sciences and Technology, Tibet Academy of Agricultural and Animal Husbandry Sciences, Xizang 850000, China
| | - Hongyu Chen
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, China.
- Modern "Chuan Cai Yu Wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| |
Collapse
|
19
|
Verdegaal AA, Goodman AL. Integrating the gut microbiome and pharmacology. Sci Transl Med 2024; 16:eadg8357. [PMID: 38295186 DOI: 10.1126/scitranslmed.adg8357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
The gut microbiome harbors trillions of organisms that contribute to human health and disease. These bacteria can also affect the properties of medical drugs used to treat these diseases, and drugs, in turn, can reshape the microbiome. Research addressing interdependent microbiome-host-drug interactions thus has broad impact. In this Review, we discuss these interactions from the perspective of drug bioavailability, absorption, metabolism, excretion, toxicity, and drug-mediated microbiome modulation. We survey approaches that aim to uncover the mechanisms underlying these effects and opportunities to translate this knowledge into new strategies to improve the development, administration, and monitoring of medical drugs.
Collapse
Affiliation(s)
- Andrew A Verdegaal
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Andrew L Goodman
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT 06536, USA
| |
Collapse
|
20
|
Krigul KL, Feeney RH, Wongkuna S, Aasmets O, Holmberg SM, Andreson R, Puértolas-Balint F, Pantiukh K, Sootak L, Org T, Tenson T, Org E, Schroeder BO. A history of repeated antibiotic usage leads to microbiota-dependent mucus defects. Gut Microbes 2024; 16:2377570. [PMID: 39034613 PMCID: PMC11529412 DOI: 10.1080/19490976.2024.2377570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024] Open
Abstract
Recent evidence indicates that repeated antibiotic usage lowers microbial diversity and ultimately changes the gut microbiota community. However, the physiological effects of repeated - but not recent - antibiotic usage on microbiota-mediated mucosal barrier function are largely unknown. By selecting human individuals from the deeply phenotyped Estonian Microbiome Cohort (EstMB), we here utilized human-to-mouse fecal microbiota transplantation to explore long-term impacts of repeated antibiotic use on intestinal mucus function. While a healthy mucus layer protects the intestinal epithelium against infection and inflammation, using ex vivo mucus function analyses of viable colonic tissue explants, we show that microbiota from humans with a history of repeated antibiotic use causes reduced mucus growth rate and increased mucus penetrability compared to healthy controls in the transplanted mice. Moreover, shotgun metagenomic sequencing identified a significantly altered microbiota composition in the antibiotic-shaped microbial community, with known mucus-utilizing bacteria, including Akkermansia muciniphila and Bacteroides fragilis, dominating in the gut. The altered microbiota composition was further characterized by a distinct metabolite profile, which may be caused by differential mucus degradation capacity. Consequently, our proof-of-concept study suggests that long-term antibiotic use in humans can result in an altered microbial community that has reduced capacity to maintain proper mucus function in the gut.
Collapse
Affiliation(s)
- Kertu Liis Krigul
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Rachel H. Feeney
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Supapit Wongkuna
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Oliver Aasmets
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Sandra M. Holmberg
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Reidar Andreson
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Fabiola Puértolas-Balint
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Kateryna Pantiukh
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Linda Sootak
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Tõnis Org
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Tanel Tenson
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Elin Org
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Bjoern O. Schroeder
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| |
Collapse
|
21
|
Banerjee G, Papri SR, Satapathy SK, Banerjee P. Akkermansia muciniphila - A Potential Next-generation Probiotic for Non-alcoholic Fatty Liver Disease. Curr Pharm Biotechnol 2024; 25:426-433. [PMID: 37724669 DOI: 10.2174/1389201025666230915103052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/20/2023] [Accepted: 08/11/2023] [Indexed: 09/21/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a spectrum of liver conditions, and its growing prevalence is a serious concern worldwide, especially in Western countries. Researchers have pointed out several genetic mutations associated with NAFLD; however, the imbalance of the gut microbial community also plays a critical role in the progression of NAFLD. Due to the lack of approved medicine, probiotics gain special attention in controlling metabolic disorders like NAFLD. Among these probiotics, Akkermansia muciniphila (a member of natural gut microflora) is considered one of the most efficient and important bacterium in maintaining gut health, energy homeostasis, and lipid metabolism. In this perspective, we discussed the probable molecular mechanism of A. muciniphila in controlling the progression of NAFLD and restoring liver health. The therapeutic potential of A. muciniphila in NAFLD has been tested primarily on animal models, and thus, more randomized human trials should be conducted to prove its efficacy.
Collapse
Affiliation(s)
- Goutam Banerjee
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Suraya R Papri
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| | - Sanjaya K Satapathy
- 2Department of Medicine, Northwell Health Center for Liver Disease & Transplantation, North Shore, University Hospital/Northwell Health, 400 Community Drive, Manhasset, NY 11030, USA
| | - Pratik Banerjee
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61801, USA
| |
Collapse
|
22
|
Zhou L, Zhang CL, Jiang K, Cheng HY, Xiong WW, Zhu JX. Therapeutic Potential of Danyankang Capsule in High-Fat Diet-Induced Cholelithiasis and Its Impact on Liver FXR Signaling and Gut Microbiota. Biol Pharm Bull 2024; 47:680-691. [PMID: 38522942 DOI: 10.1248/bpb.b24-00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
Cholelithiasis, commonly known as gallstones, represents a prevalent hepatobiliary disorder. This study aimed to elucidate the therapeutic role and mechanism of Danyankang capsulein treating cholelithiasis induced by a high-fat diet in C57BL/6 mice. The therapeutical potential of Danyankang was assessed through biochemical analyses, histopathological examinations, protein detection, and 16S rDNA sequencing. A high-fat diet resulted in cholelithiasis manifestation in mice, with discernable abnormal serum biochemical indices and disrupted biliary cholesterol homeostasis. Danyankang treatment notably ameliorated liver inflammation symptoms and rectified serum and liver biochemical abnormalities. Concurrently, it addressed biliary imbalances. Elevated expressions of toll-like receptor 4 (TLR4), nuclear factor-kappaB (NF-κB)/pNF-κB, HMGCR, CYP7A1, and CYP8B1 observed at the inception of cholelithiasis, were notably reduced upon Danyankang administration. Furthermore, 16S rDNA analysis revealed a decline in species number and diversity of the intestinal flora in cholelithiasis-treated mice, while the decline was reversed with Danyankang treatment. Danyankang capsules reduced the abundance of Verrucomicrobiota and increased the abundance of Actinobacteriota and Proteobacteria. In conclusion, the present study demonstrates that Danyankang exerts potent therapeutic efficacy against high-fat diet-induced cholelithiasis. This beneficial outcome is potentially linked to the inhibition of the TLR4/pNF-κB and SHP/CYP7A1/CYP8B1 signaling pathways, as well as the enhancement of intestinal flora species abundance.
Collapse
Affiliation(s)
- Lin Zhou
- Research Center of Traditional Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine
| | - Chu-Ling Zhang
- Research Center of Traditional Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine
| | - Kun Jiang
- Guizhou Bailing Enterprise Group Pharmaceutical Co., Ltd
| | - Hong-Yu Cheng
- College of Humanities, Jiangxi University of Chinese Medicine
| | - Wen-Wen Xiong
- Medical Clinic, Jiangxi University of Chinese Medicine
| | - Ji-Xiao Zhu
- Research Center of Traditional Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine
| |
Collapse
|
23
|
Mitchell SB, Thorn TL, Lee MT, Kim Y, Comrie JMC, Bai ZS, Johnson EL, Aydemir TB. Metal transporter SLC39A14/ZIP14 modulates regulation between the gut microbiome and host metabolism. Am J Physiol Gastrointest Liver Physiol 2023; 325:G593-G607. [PMID: 37873588 PMCID: PMC10887856 DOI: 10.1152/ajpgi.00091.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023]
Abstract
Metal transporter SLC39A14/ZIP14 is localized on the basolateral side of the intestine, functioning to transport metals from blood to intestine epithelial cells. Deletion of Slc39a14/Zip14 causes spontaneous intestinal permeability with low-grade chronic inflammation, mild hyperinsulinemia, and greater body fat with insulin resistance in adipose. Importantly, antibiotic treatment reverses the adipocyte phenotype of Slc39a14/Zip14 knockout (KO), suggesting a potential gut microbial role in the metabolic alterations in the Slc39a14/Zip14 KO mice. Here, we investigated the hypothesis that increased intestinal permeability and subsequent metabolic alterations in the absence of Zip14 could be in part due to alterations in gut microbial composition. Dietary metals have been shown to be involved in the regulation of gut microbial diversity and composition. However, studies linking the action of intestinal metal transporters to gut microbial regulation are lacking. We showed the influence of deletion of metal transporter Slc39a14/Zip14 on gut microbiome composition and how ZIP14-linked changes to gut microbiome community composition are correlated with changes in host metabolism. Deletion of Slc39a14/Zip14 generated Zn-deficient epithelial cells and luminal content in the entire intestinal tract, a shift in gut microbial composition that partially overlapped with changes previously associated with obesity and inflammatory bowel disease (IBD), increased the fungi/bacteria ratio in the gut microbiome, altered the host metabolome, and shifted host energy metabolism toward glucose utilization. Collectively, our data suggest a potential predisease microbial susceptibility state dependent on host gene Slc39a14/Zip14 that contributes to intestinal permeability, a common trait of IBD, and metabolic disorders such as obesity and type 2 diabetes.NEW & NOTEWORTHY Metal dyshomeostasis, intestinal permeability, and gut dysbiosis are emerging signatures of chronic disorders, including inflammatory bowel diseases, type-2 diabetes, and obesity. Studies in reciprocal regulations between host intestinal metal transporters genes and gut microbiome are scarce. Our research revealed a potential predisease microbial susceptibility state dependent on the host metal transporter gene, Slc39a14/Zip14, that contributes to intestinal permeability providing new insight into understanding host metal transporter gene-microbiome interactions in developing chronic disease.
Collapse
Affiliation(s)
- Samuel B Mitchell
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Trista L Thorn
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Min-Ting Lee
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Yongeun Kim
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Janine M C Comrie
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Zi Shang Bai
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Elizabeth L Johnson
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Tolunay B Aydemir
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| |
Collapse
|
24
|
Ayala-García JC, García-Vera AM, Lagunas-Martínez A, Orbe-Orihuela YC, Castañeda-Márquez AC, Díaz-Benítez CE, Bermúdez-Morales VH, Cruz M, Bahena-Román M, Burguete-García AI. Interaction between Akkermansia muciniphila and Diet Is Associated with Proinflammatory Index in School-Aged Children. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1799. [PMID: 38002890 PMCID: PMC10670599 DOI: 10.3390/children10111799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND Imbalance in the intestinal microbiota can lead to chronic low-grade inflammation. Diet may influence this association. In this study, we aimed to evaluate the interaction between Akkermansia muciniphila (A. muciniphila) and dietary patterns using a proinflammatory index. METHODS We conducted a cross-sectional study with school-aged children. We quantified the relative abundance (RA) of A. muciniphila in feces using a polymerase chain reaction. We collected dietary information through employing a food frequency questionnaire and generated dietary patterns using principal component analysis. We generated a proinflammatory index from serum levels of interleukin-6, interleukin-10, tumor necrosis factor alpha, and adiponectin validated by receptor operating characteristic curves. We evaluated the association between A. muciniphila and the proinflammatory index using logistic regression, including an interaction term with dietary patterns. RESULTS We found that children with a low RA of A. muciniphila and a high intake of simple carbohydrates and saturated fats had increased odds of being high on the proinflammatory index. However, when the consumption of this dietary pattern is low, children with a low RA of A. muciniphila had decreased odds of being high on the proinflammatory index. CONCLUSIONS Our results suggest that the simultaneous presence of A. muciniphila and diet have a more significant impact on the presence of being high on the proinflammatory index compared to both factors separately.
Collapse
Affiliation(s)
- Juan Carlos Ayala-García
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (J.C.A.-G.); (A.L.-M.); (Y.C.O.-O.); (C.E.D.-B.); (V.H.B.-M.); (M.B.-R.)
| | - Alba Mariel García-Vera
- Escuela de Salud Pública de México, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico;
| | - Alfredo Lagunas-Martínez
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (J.C.A.-G.); (A.L.-M.); (Y.C.O.-O.); (C.E.D.-B.); (V.H.B.-M.); (M.B.-R.)
| | - Yaneth Citlalli Orbe-Orihuela
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (J.C.A.-G.); (A.L.-M.); (Y.C.O.-O.); (C.E.D.-B.); (V.H.B.-M.); (M.B.-R.)
| | | | - Cinthya Estefhany Díaz-Benítez
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (J.C.A.-G.); (A.L.-M.); (Y.C.O.-O.); (C.E.D.-B.); (V.H.B.-M.); (M.B.-R.)
| | - Víctor Hugo Bermúdez-Morales
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (J.C.A.-G.); (A.L.-M.); (Y.C.O.-O.); (C.E.D.-B.); (V.H.B.-M.); (M.B.-R.)
| | - Miguel Cruz
- Unidad de Investigación Médica en Bioquímica, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico;
| | - Margarita Bahena-Román
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (J.C.A.-G.); (A.L.-M.); (Y.C.O.-O.); (C.E.D.-B.); (V.H.B.-M.); (M.B.-R.)
| | - Ana Isabel Burguete-García
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (J.C.A.-G.); (A.L.-M.); (Y.C.O.-O.); (C.E.D.-B.); (V.H.B.-M.); (M.B.-R.)
| |
Collapse
|
25
|
Kumbhare SV, Pedroso I, Ugalde JA, Márquez-Miranda V, Sinha R, Almonacid DE. Drug and gut microbe relationships: Moving beyond antibiotics. Drug Discov Today 2023; 28:103797. [PMID: 37806386 DOI: 10.1016/j.drudis.2023.103797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/23/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023]
Abstract
Our understanding of drug-microbe relationships has evolved from viewing microbes as mere drug producers to a dynamic, modifiable system where they can serve as drugs or targets of precision pharmacology. This review highlights recent findings on the gut microbiome, particularly focusing on four aspects of research: (i) drugs for bugs, covering recent strategies for targeting gut pathogens; (ii) bugs as drugs, including probiotics; (iii) drugs from bugs, including postbiotics; and (iv) bugs and drugs, discussing additional types of drug-microbe interactions. This review provides a perspective on future translational research, including efficient companion diagnostics in pharmaceutical interventions.
Collapse
Affiliation(s)
| | | | - Juan A Ugalde
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Valeria Márquez-Miranda
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | | | | |
Collapse
|
26
|
Fan L, Xia Y, Wang Y, Han D, Liu Y, Li J, Fu J, Wang L, Gan Z, Liu B, Fu J, Zhu C, Wu Z, Zhao J, Han H, Wu H, He Y, Tang Y, Zhang Q, Wang Y, Zhang F, Zong X, Yin J, Zhou X, Yang X, Wang J, Yin Y, Ren W. Gut microbiota bridges dietary nutrients and host immunity. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2466-2514. [PMID: 37286860 PMCID: PMC10247344 DOI: 10.1007/s11427-023-2346-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 04/05/2023] [Indexed: 06/09/2023]
Abstract
Dietary nutrients and the gut microbiota are increasingly recognized to cross-regulate and entrain each other, and thus affect host health and immune-mediated diseases. Here, we systematically review the current understanding linking dietary nutrients to gut microbiota-host immune interactions, emphasizing how this axis might influence host immunity in health and diseases. Of relevance, we highlight that the implications of gut microbiota-targeted dietary intervention could be harnessed in orchestrating a spectrum of immune-associated diseases.
Collapse
Affiliation(s)
- Lijuan Fan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yaoyao Xia
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Youxia Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Jiahuan Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jie Fu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Leli Wang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Zhending Gan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Bingnan Liu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jian Fu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Congrui Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenhua Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Hui Han
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Hao Wu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yiwen He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yulong Tang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Qingzhuo Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yibin Wang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Fan Zhang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Xin Zong
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China.
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Wenkai Ren
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
27
|
Lai C, Chen L, Zhong X, Tang Z, Zhang B, Luo Y, Li C, Jin M, Chen X, Li J, Shi Y, Sun Y, Guo L. Long-term effects on liver metabolism induced by ceftriaxone sodium pretreatment. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 335:122238. [PMID: 37506808 DOI: 10.1016/j.envpol.2023.122238] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/16/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Ceftriaxone is an emerging contaminant due to its potential harm, while its effects on liver are still need to be clarified. In this study, we first pretreated the 8-week-old C57BL/6J mice with high dose ceftriaxone sodium (Cef, 400 mg/mL, 0.2 mL per dose) for 8 days to prepare a gut dysbiosis model, then treated with normal feed for a two-month recovery period, and applied non-targeted metabolomics (including lipidomics) to investigate the variations of fecal and liver metabolome, and coupled with targeted determination of fecal short-chain fatty acids (SCFAs) and bile acids (BAs). Lastly, the correlations and mediation analysis between the liver metabolism and gut metabolism/microbes were carried, and the potential mechanisms of the mal-effects on gut-liver axis induced by Cef pretreatment were accordingly discussed. Compared to the control group, Cef pretreatment reduced the rate of weight gain and hepatosomatic index, induced bile duct epithelial cells proliferated around the central vein and appearance of binucleated hepatocytes, decreased the ratio of total branching chains amino acids (BCAAs) to total aromatic amino acids (AAAs) in liver metabolome. In fecal metabolome, the total fecal SCFAs and BAs did not change significantly while butyric acid decreased and the primary BAs increased after Cef pretreatment. Correlation and mediation analysis revealed one potential mechanism that Cef may first change the intestinal microbiota (such as destroying its normal structure, reducing its abundance and the stability of the microbial network or certain microbe abundance like Alistipes), and then change the intestinal metabolism (such as acetate, caproate, propionate), leading to liver metabolic disorder (such as spermidine, inosine, cinnamaldehyde). This study proved the possibility of Cef-induced liver damage, displayed the overall metabolic profile of the liver following Cef pretreatment and provided a theoretical framework for further research into the mechanism of Cef-induced liver damage.
Collapse
Affiliation(s)
- Chengze Lai
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Linkang Chen
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Xiaoting Zhong
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Zeli Tang
- Department of Pathology, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Bin Zhang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Yu Luo
- Guangzhou Liwan District Center for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Chengji Li
- Yunfu Disease Control and Prevention Center, Guang Dong Province, China
| | - Mengcheng Jin
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Xu Chen
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Jinglin Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Yinying Shi
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Yanqin Sun
- Department of Pathology, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Lianxian Guo
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
28
|
Choi KJ, Yoon MY, Kim JE, Yoon SS. Gut commensal Kineothrix alysoides mitigates liver dysfunction by restoring lipid metabolism and gut microbial balance. Sci Rep 2023; 13:14668. [PMID: 37674003 PMCID: PMC10482948 DOI: 10.1038/s41598-023-41160-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as Non-Alcoholic Fatty Liver Disease, is a widespread liver condition characterized by excessive fat buildup in hepatocytes without significant alcohol consumption. Manipulation of the gut microbiome has been considered to prevent and improve the occurrence and progression of MASLD, particularly through the gut-liver axis. This study aimed to investigate the correlation between the gut microbiome and liver function and determine whether the gut microbiome can ameliorate MASLD. We comparatively analyzed the gut microbiome composition between mice fed normal chow and those fed a high-fat diet and observed that the abundance of Kineothrix alysoides decreased in the high-fat group. Further analysis showed that treatment with K. alysoides in the high-fat diet group led to decreased weight loss, and MASLD attenuation. Importantly, K. alysoides treatment attenuated MASLD in mice fed a high-fat, high-fructose diet (HFHF), which can cause advanced liver damage. Furthermore, administration of K. alysoides altered the gut microbial composition in the HFHF diet group and improved MASLD. Overall, these findings demonstrate the potential of K. alysoides in restoring gut health and facilitating lipid metabolism to prevent and treat MASLD.
Collapse
Affiliation(s)
- Kyoung Jin Choi
- Department of Microbiology and Immunology, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
- Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - Mi Young Yoon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji-Eun Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea.
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea.
| | - Sang Sun Yoon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea.
- Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea.
- BioMe Inc., Seoul, South Korea.
| |
Collapse
|
29
|
Rosel-Pech C, Pinto-Cardoso S, Chávez-Torres M, Montufar N, Osuna-Padilla I, Ávila-Ríos S, Reyes-Terán G, Aguirre-Alvarado C, Matías Juan NA, Pérez-Lorenzana H, Vázquez-Rosales JG, Bekker-Méndez VC. Distinct fecal microbial signatures are linked to sex and chronic immune activation in pediatric HIV infection. Front Immunol 2023; 14:1244473. [PMID: 37711620 PMCID: PMC10497879 DOI: 10.3389/fimmu.2023.1244473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/09/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Our understanding of HIV-associated gut microbial dysbiosis in children perinatally-infected with HIV (CLWH) lags behind that of adults living with HIV. Childhood represents a critical window for the gut microbiota. Any disturbances, including prolonged exposure to HIV, antiretroviral drugs, and antibiotics are likely to have a significant impact on long-term health, resulting in a less resilient gut microbiome. The objective of our study was to characterize the gut microbiota in CLWH, and compare it with HIV-unexposed and -uninfected children. Methods We enrolled 31 children aged 3 to 15 years; 15 were CLWH and 16 were HUU. We assessed dietary patterns and quality; quantified soluble and cellular markers of HIV disease progression by flow cytometry, enzyme-linked immunosorbent and multiplex-bead assays, and profiled the gut microbiota by 16S rRNA sequencing. We explored relationships between the gut microbiota, antibiotic exposure, dietary habits, soluble and cellular markers and host metadata. Results Children had a Western-type diet, their median health eating index score was 67.06 (interquartile range 58.76-74.66). We found no discernable impact of HIV on the gut microbiota. Alpha diversity metrics did not differ between CLWH and HUU. Sex impacted the gut microbiota (R-squared= 0.052, PERMANOVA p=0.024). Male children had higher microbial richness compared with female children. Two taxa were found to discriminate female from male children independently from HIV status: Firmicutes for males, and Bacteroides for females. Markers of HIV disease progression were comparable between CLWH and HUU, except for the frequency of exhausted CD4+ T cells (PD-1+) which was increased in CLWH (p=0.0024 after adjusting for confounders). Both the frequency of exhausted CD4+ and activated CD4+ T cells (CD38+ HLADR+) correlated positively with the relative abundance of Proteobacteria (rho=0.568. false discovery rate (FDR)-adjusted p= 0.029, and rho=0.62, FDR-adjusted p=0.0126, respectively). Conclusion The gut microbiota of CLWH appears similar to that of HUU, and most markers of HIV disease progression are normalized with long-term ART, suggesting a beneficial effect of the latter on the gut microbial ecology. The relationship between exhausted and activated CD4+ T cells and Proteobacteria suggests a connection between the gut microbiome, and premature aging in CLWH.
Collapse
Affiliation(s)
- Cecilia Rosel-Pech
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
- Unidad de Investigación Médica en Inmunología e Infectología, Hospital de Infectología “Dr. Daniel Méndez Hernández”, Centro Médico Nacional “La Raza”, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, Mexico
| | - Sandra Pinto-Cardoso
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, Mexico
| | - Monserrat Chávez-Torres
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, Mexico
| | - Nadia Montufar
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, Mexico
| | - Iván Osuna-Padilla
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, Mexico
| | - Santiago Ávila-Ríos
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, Mexico
| | - Gustavo Reyes-Terán
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Charmina Aguirre-Alvarado
- Unidad de Investigación Médica en Inmunología e Infectología, Hospital de Infectología “Dr. Daniel Méndez Hernández”, Centro Médico Nacional “La Raza”, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, Mexico
| | - Norma Angelica Matías Juan
- Hospital de Infectología “Dr. Daniel Méndez Hernández”, Centro Médico Nacional “La Raza”, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, Mexico
| | - Héctor Pérez-Lorenzana
- UMAE Hospital General Dr. Gaudencio González Garza, Centro Médico Nacional La Raza, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, Mexico
| | - José Guillermo Vázquez-Rosales
- Hospital de Pediatría “Doctor Silvestre Frenk Freund”, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, México, Mexico
| | - Vilma Carolina Bekker-Méndez
- Unidad de Investigación Médica en Inmunología e Infectología, Hospital de Infectología “Dr. Daniel Méndez Hernández”, Centro Médico Nacional “La Raza”, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, Mexico
| |
Collapse
|
30
|
Sivamani RK, Maloh J, Nong Y. Correlating the Gut Microbiota and Circulating Hormones with Acne Lesion Counts and Skin Biophysical Features. Microorganisms 2023; 11:2049. [PMID: 37630609 PMCID: PMC10459794 DOI: 10.3390/microorganisms11082049] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/31/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Acne vulgaris is a common inflammatory condition that is multi-factorial and impacted by both intrinsic and extrinsic features. Several previous studies have assessed for correlations between factors such as circulating hormones, stress, or the microbiome. However, there have not been any correlations specifically against lesion counts or differentiating correlations between inflammatory and non-inflammatory lesion counts. Here, we correlate several factors against acne lesions. Twenty men and women with mild to moderate acne were recruited, and their hormonal levels and their gut microbiome were collected and correlated against their inflammatory and non-inflammatory lesions of acne. Facial non-inflammatory lesions were weakly correlated to sebum excretion rate and weakly inversely correlated to forehead and cheek hydration. We examined stress through the use of a normalized peak-to-trough ratio (higher numbers indicated less stress), which correlated with skin hydration and inversely correlated with sebum excretion rate. Sebum excretion rate was weakly correlated to testosterone levels, and facial hydration correlated with estradiol levels. Correlations with the gut microbiome showed differential correlations with inflammatory and non-inflammatory lesions, with Clostridium sp AF 23-8 correlating to inflammatory lesion counts, while Actinomyces naeslundii str Howell 279 correlated to non-inflammatory lesions. Overall, measures of stress and circulating hormones correlate to skin biophysical properties and acne lesion counts. Also, different gut bacteria correlate with either inflammatory or non-inflammatory lesion counts. We hope that our findings stimulate further work on the gut-mind-stress-skin axis within acne.
Collapse
Affiliation(s)
- Raja K. Sivamani
- Integrative Skin Science and Research, Sacramento, CA 95815, USA
- Department of Dermatology, University of California-Davis, Sacramento, CA 95616, USA
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
- Pacific Skin Institute, Sacramento, CA 95815, USA
| | - Jessica Maloh
- Integrative Skin Science and Research, Sacramento, CA 95815, USA
| | - Yvonne Nong
- Integrative Skin Science and Research, Sacramento, CA 95815, USA
| |
Collapse
|
31
|
Lei W, Cheng Y, Gao J, Liu X, Shao L, Kong Q, Zheng N, Ling Z, Hu W. Akkermansia muciniphila in neuropsychiatric disorders: friend or foe? Front Cell Infect Microbiol 2023; 13:1224155. [PMID: 37492530 PMCID: PMC10363720 DOI: 10.3389/fcimb.2023.1224155] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/26/2023] [Indexed: 07/27/2023] Open
Abstract
An accumulating body of evidence suggests that the bacterium Akkermansia muciniphila exhibits positive systemic effects on host health, mainly by improving immunological and metabolic functions, and it is therefore regarded as a promising potential probiotic. Recent clinical and preclinical studies have shown that A. muciniphila plays a vital role in a variety of neuropsychiatric disorders by influencing the host brain through the microbiota-gut-brain axis (MGBA). Numerous studies observed that A. muciniphila and its metabolic substances can effectively improve the symptoms of neuropsychiatric disorders by restoring the gut microbiota, reestablishing the integrity of the gut mucosal barrier, regulating host immunity, and modulating gut and neuroinflammation. However, A. muciniphila was also reported to participate in the development of neuropsychiatric disorders by aggravating inflammation and influencing mucus production. Therefore, the exact mechanism of action of A. muciniphila remains much controversial. This review summarizes the proposed roles and mechanisms of A. muciniphila in various neurological and psychiatric disorders such as depression, anxiety, Parkinson's disease, Alzheimer's disease, multiple sclerosis, strokes, and autism spectrum disorders, and provides insights into the potential therapeutic application of A. muciniphila for the treatment of these conditions.
Collapse
Affiliation(s)
- Wenhui Lei
- Jinan Microecological Biomedicine Shandong Laboratory, Shandong First Medical University, Jinan, Shandong, China
| | - Yiwen Cheng
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jie Gao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Li Shao
- School of Clinical Medicine, Institute of Hepatology and Metabolic Diseases, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Qingming Kong
- School of Biological Engineering, Hangzhou Medical College, Institute of Parasitic Diseases, Hangzhou, Zhejiang, China
| | - Nengneng Zheng
- Department of Obstetrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zongxin Ling
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weiming Hu
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, Zhejiang, China
| |
Collapse
|
32
|
Sanada TJ, Hosomi K, Park J, Naito A, Sakao S, Tanabe N, Kunisawa J, Tatsumi K, Suzuki T. Partially hydrolyzed guar gum suppresses the progression of pulmonary arterial hypertension in a SU5416/hypoxia rat model. Pulm Circ 2023; 13:e12266. [PMID: 37448440 PMCID: PMC10336776 DOI: 10.1002/pul2.12266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
This study investigated the effects of partially hydrolyzed guar gum (PHGG) on the development of pulmonary arterial hypertension using a SU5416/hypoxia rat model. Our results demonstrated that PHGG treatment suppressed the development of pulmonary hypertension and vascular remodeling with an altered gut microbiota composition.
Collapse
Affiliation(s)
- Takayuki J. Sanada
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN)National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN)OsakaJapan
- Department of Respirology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Koji Hosomi
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN)National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN)OsakaJapan
| | - Jonguk Park
- Artificial Intelligence Center for Health and Biomedical ResearchNational Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN)OsakaJapan
| | - Akira Naito
- Department of Respirology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Nobuhiro Tanabe
- Department of Respirology, Graduate School of MedicineChiba UniversityChibaJapan
- Department of RespirologyChibaken Saiseikai Narashino HospitalNarashinoJapan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN)National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN)OsakaJapan
- Artificial Intelligence Center for Health and Biomedical ResearchNational Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN)OsakaJapan
- Graduate School of Pharmaceutical SciencesOsaka UniversityOsakaJapan
- Graduate School of MedicineOsaka UniversityOsakaJapan
- Graduate School of DentistryOsaka UniversityOsakaJapan
- Graduate School of ScienceOsaka UniversityOsakaJapan
- Department of Microbiology and Immunology, Graduate School of MedicineKobe UniversityHyogoJapan
- International Vaccine Design Center, Institute of Medical ScienceUniversity of TokyoTokyoJapan
- Research Organization for Nano and Life InnovationWaseda UniversityTokyoJapan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of MedicineChiba UniversityChibaJapan
| | - Takuji Suzuki
- Department of Respirology, Graduate School of MedicineChiba UniversityChibaJapan
| |
Collapse
|
33
|
Mishra G, Singh P, Molla M, Yimer YS, Dinda SC, Chandra P, Singh BK, Dagnew SB, Assefa AN, Ewunetie A. Harnessing the potential of probiotics in the treatment of alcoholic liver disorders. Front Pharmacol 2023; 14:1212742. [PMID: 37361234 PMCID: PMC10287977 DOI: 10.3389/fphar.2023.1212742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
In the current scenario, prolonged consumption of alcohol across the globe is upsurging an appreciable number of patients with the risk of alcohol-associated liver diseases. According to the recent report, the gut-liver axis is crucial in the progression of alcohol-induced liver diseases, including steatosis, steatohepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma. Despite several factors associated with alcoholic liver diseases, the complexity of the gut microflora and its great interaction with the liver have become a fascinating area for researchers due to the high exposure of the liver to free radicals, bacterial endotoxins, lipopolysaccharides, inflammatory markers, etc. Undoubtedly, alcohol-induced gut microbiota imbalance stimulates dysbiosis, disrupts the intestinal barrier function, and trigger immune as well as inflammatory responses which further aggravate hepatic injury. Since currently available drugs to mitigate liver disorders have significant side effects, hence, probiotics have been widely researched to alleviate alcohol-associated liver diseases and to improve liver health. A broad range of probiotic bacteria like Lactobacillus, Bifidobacteria, Escherichia coli, Sacchromyces, and Lactococcus are used to reduce or halt the progression of alcohol-associated liver diseases. Several underlying mechanisms, including alteration of the gut microbiome, modulation of intestinal barrier function and immune response, reduction in the level of endotoxins, and bacterial translocation, have been implicated through which probiotics can effectively suppress the occurrence of alcohol-induced liver disorders. This review addresses the therapeutic applications of probiotics in the treatment of alcohol-associated liver diseases. Novel insights into the mechanisms by which probiotics prevent alcohol-associated liver diseases have also been elaborated.
Collapse
Affiliation(s)
- Garima Mishra
- Pharmaceutical Chemistry Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Pradeep Singh
- Pharmaceutical Chemistry Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Mulugeta Molla
- Pharmacology and Toxicology Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Yohannes Shumet Yimer
- Social Pharmacy Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | | | - Phool Chandra
- Department of Pharmacology, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, India
| | | | - Samuel Berihun Dagnew
- Clinical Pharmacy Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Abraham Nigussie Assefa
- Social Pharmacy Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Amien Ewunetie
- Pharmacology and Toxicology Unit, Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| |
Collapse
|
34
|
Lu F, MacPherson CW, Tremblay J, Iskandar MM, Kubow S. Anthocyanin-rich blue potato meals protect against polychlorinated biphenyl-mediated disruption of short-chain fatty acid production and gut microbiota profiles in a simulated human digestion model. Front Nutr 2023; 10:1130841. [PMID: 37324735 PMCID: PMC10266533 DOI: 10.3389/fnut.2023.1130841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/09/2023] [Indexed: 06/17/2023] Open
Abstract
Background Polychlorinated biphenyls (PCBs) are ubiquitous environmental pollutants associated with a wide variety of adverse human health outcomes. PCB 126 and PCB 153 are among the most prevalent congeners associated with human exposure. Emerging studies have suggested that PCB exposure leads to lower gut microbial diversity although their effects on microbial production of health promoting short-chain fatty acids (SCFAs) has been scarcely studied. Blue potatoes are rich in anthocyanins (ACNs), which is a class of polyphenols that promote the growth of beneficial intestinal bacteria such as Bifidobacterium and Lactobacillus and increase the generation of SCFAs. A batch-culture, pH-controlled, stirred system containing human fecal microbial communities was utilized to assess whether human gut microbiota composition and SCFA production are affected by: (a) PCB 126 and PCB 153 exposure; and (b) ACN-rich digests in the presence and absence of the PCB congeners. Methods Anthocyanin-rich blue potato meals (11.03 g) were digested over 12 h with and without PCB 126 (0.5 mM) and PCB 153 (0.5 mM) using an in vitro simulated gut digestion model involving upper gastrointestinal digestion followed by metabolism by human fecal microbiota. Fecal digests were collected for analysis of gut microbial and SCFA profiles. Results Polychlorinated biphenyl-exposed fecal samples showed a significant (p < 0.05) decrease in species richness and a significantly (p < 0.05) different microbial community structure. PCB treatment was associated with an increased (p < 0.05) relative abundance of Akkermansia, Eggerthella, and Bifidobacterium and a decreased (p < 0.05) relative abundance of Veillonella, Streptococcus, and Holdemanella. ACN digests counteracted the altered abundances of Akkermansia and Bifidobacterium seen with the PCB treatment. PCB exposure was associated with a significant (p < 0.05) decrease in total SCFA and acetate concentrations. ACN digests were associated with significantly (p < 0.05) higher SCFA and acetate concentrations in the presence and absence of PCBs. Conclusion Human fecal matter exposed to PCB 126 and PCB 153 led to decreased abundance and altered gut microbiota profiles as well as lowered SCFA and acetate levels. Importantly, this study showed that prebiotic ACN-rich potatoes counteract PCB-mediated disruptions in human gut microbiota profiles and SCFA production.
Collapse
Affiliation(s)
- Fang Lu
- School of Human Nutrition, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| | | | - Julien Tremblay
- Energy, Mining and Environment, National Research Council Canada, Montreal, QC, Canada
| | - Michèle M. Iskandar
- School of Human Nutrition, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| | - Stan Kubow
- School of Human Nutrition, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| |
Collapse
|
35
|
Senchukova MA. Genetic heterogeneity of colorectal cancer and the microbiome. World J Gastrointest Oncol 2023; 15:443-463. [PMID: 37009315 PMCID: PMC10052667 DOI: 10.4251/wjgo.v15.i3.443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/06/2023] [Accepted: 02/22/2023] [Indexed: 03/14/2023] Open
Abstract
In 2020, the International Agency for Research on Cancer and the World Health Organization's GLOBOCAN database ranked colorectal cancer (CRC) as the third most common cancer in the world. Most cases of CRC (> 95%) are sporadic and develop from colorectal polyps that can progress to intramucosal carcinoma and CRC. Increasing evidence is accumulating that the gut microbiota can play a key role in the initiation and progression of CRC, as well as in the treatment of CRC, acting as an important metabolic and immunological regulator. Factors that may determine the microbiota role in CRC carcinogenesis include inflammation, changes in intestinal stem cell function, impact of bacterial metabolites on gut mucosa, accumulation of genetic mutations and other factors. In this review, I discuss the major mechanisms of the development of sporadic CRC, provide detailed characteristics of the bacteria that are most often associated with CRC, and analyze the role of the microbiome and microbial metabolites in inflammation initiation, activation of proliferative activity in intestinal epithelial and stem cells, and the development of genetic and epigenetic changes in CRC. I consider long-term studies in this direction to be very important, as they open up new opportunities for the treatment and prevention of CRC.
Collapse
Affiliation(s)
- Marina A Senchukova
- Department of Oncology, Orenburg State Medical University, Orenburg 460000, Russia
| |
Collapse
|
36
|
Krzyżek P, Marinacci B, Vitale I, Grande R. Extracellular Vesicles of Probiotics: Shedding Light on the Biological Activity and Future Applications. Pharmaceutics 2023; 15:522. [PMID: 36839844 PMCID: PMC9967243 DOI: 10.3390/pharmaceutics15020522] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
For many decades, the proper functioning of the human body has become a leading scientific topic. In the course of numerous experiments, a striking impact of probiotics on the human body has been documented, including maintaining the physiological balance of endogenous microorganisms, regulating the functioning of the immune system, enhancing the digestive properties of the host, and preventing or alleviating the course of many diseases. Recent research, especially from the last decade, shows that this health-benefiting activity of probiotics is largely conditioned by the production of extracellular vesicles. Although the importance of extracellular vesicles in the virulence of many live-threatening pathogens is widely described in the literature, much less is known with respect to the health-promoting effect of extracellular vesicles secreted by non-pathogenic microorganisms, including probiotics. Based on this, in the current review article, we decided to collect the latest literature data on the health-inducing properties of extracellular vesicles secreted by probiotics. The characteristics of probiotics' extracellular vesicles will be extended by the description of their physicochemical properties and the proteome in connection with the biological activities exhibited by these structures.
Collapse
Affiliation(s)
- Paweł Krzyżek
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Beatrice Marinacci
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, University “Gabriele d’Annunzio”, Chieti-Pescara, 66100 Chieti, Italy
| | - Irene Vitale
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
| | - Rossella Grande
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
| |
Collapse
|
37
|
Kulecka M, Fraczek B, Balabas A, Czarnowski P, Zeber-Lubecka N, Zapala B, Baginska K, Glowienka M, Szot M, Skorko M, Kluska A, Piatkowska M, Mikula M, Ostrowski J. Characteristics of the gut microbiome in esports players compared with those in physical education students and professional athletes. Front Nutr 2023; 9:1092846. [PMID: 36726816 PMCID: PMC9884692 DOI: 10.3389/fnut.2022.1092846] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction Esports is a category of competitive video games that, in many aspects, may be similar to traditional sports; however, the gut microbiota composition of players has not been yet studied. Materials and methods Here, we investigated the composition and function of the gut microbiota, as well as short chain fatty acids (SCFAs), and amino acids, in a group of 109 well-characterized Polish male esports players. The results were compared with two reference groups: 25 endurance athletes and 36 healthy students of physical education. DNA and metabolites isolated from fecal samples were analyzed using shotgun metagenomic sequencing and mass spectrometry, respectively. Physical activity and nutritional measures were evaluated by questionnaire. Results Although anthropometric, physical activity and nutritional measures differentiated esports players from students, there were no differences in bacterial diversity, the Bacteroidetes/Firmicutes ratio, the composition of enterotype clusters, metagenome functional content, or SCFA concentrations. However, there were significant differences between esports players and students with respect to nine bacterial species and nine amino acids. By contrast, all of the above-mentioned measures differentiated professional athletes from esports players and students, with 45 bacteria differentiating professional athletes from the former and 31 from the latter. The only species differentiating all three experimental groups was Parabacteroides distasonis, showing the lowest and highest abundance in esports players and athletes, respectively. Conclusion Our study confirms the marked impact of intense exercise training on gut microbial structure and function. Differences in lifestyle and dietary habits between esports players and physical education students appear to not have a major effect on the gut microbiota.
Collapse
Affiliation(s)
- Maria Kulecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Barbara Fraczek
- Department of Sports Medicine and Human Nutrition, Institute of Biomedical Sciences, University of Physical Education, Krakow, Poland
| | - Aneta Balabas
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Paweł Czarnowski
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Department of Biochemistry, Radioimmunology and Experimental Medicine, The Children's Memorial Health Institute, Warsaw, Poland
| | - Natalia Zeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Barbara Zapala
- Department of Clinical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Katarzyna Baginska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Maria Glowienka
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Monika Szot
- Department of Sports Dietetics, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Maciek Skorko
- Institute of Psychology, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Kluska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Magdalena Piatkowska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Michał Mikula
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Jerzy Ostrowski
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
38
|
Wastyk HC, Perelman D, Topf M, Fragiadakis GK, Robinson JL, Sonnenburg JL, Gardner CD, Sonnenburg ED. Randomized controlled trial demonstrates response to a probiotic intervention for metabolic syndrome that may correspond to diet. Gut Microbes 2023; 15:2178794. [PMID: 36803658 PMCID: PMC9980610 DOI: 10.1080/19490976.2023.2178794] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
An individual's immune and metabolic status is coupled to their microbiome. Probiotics offer a promising, safe route to influence host health, possibly via the microbiome. Here, we report an 18-week, randomized prospective study that explores the effects of a probiotic vs. placebo supplement on 39 adults with elevated parameters of metabolic syndrome. We performed longitudinal sampling of stool and blood to profile the human microbiome and immune system. While we did not see changes in metabolic syndrome markers in response to the probiotic across the entire cohort, there were significant improvements in triglycerides and diastolic blood pressure in a subset of probiotic arm participants. Conversely, the non-responders had increased blood glucose and insulin levels over time. The responders had a distinct microbiome profile at the end of the intervention relative to the non-responders and placebo arm. Importantly, diet was a key differentiating factor between responders and non-responders. Our results show participant-specific effects of a probiotic supplement on improving parameters of metabolic syndrome and suggest that dietary factors may enhance stability and efficacy of the supplement.
Collapse
Affiliation(s)
- Hannah C. Wastyk
- Department of Bioengineering, Stanford School of Medicine, Stanford, CA, USA
| | - Dalia Perelman
- Stanford Prevention Research Center, Department of Medicine, Stanford School of 4Medicine, Stanford, CA, USA
| | - Madeline Topf
- Microbiology & Immunology, Stanford School of Medicine, Stanford, CA, USA
| | | | - Jennifer L. Robinson
- Stanford Prevention Research Center, Department of Medicine, Stanford School of 4Medicine, Stanford, CA, USA
| | - Justin L. Sonnenburg
- Microbiology & Immunology, Stanford School of Medicine, Stanford, CA, USA,Center for Human Microbiome Studies, Stanford School of Medicine, Stanford University, Stanford, CA, USA,Chan Zuckerberg Biohub, San Francisco, CA, USA,CONTACT Justin L. Sonnenburg Microbiology & Immunology, Stanford School of Medicine, Stanford, CA94305, USA
| | - Christopher D. Gardner
- Stanford Prevention Research Center, Department of Medicine, Stanford School of 4Medicine, Stanford, CA, USA,Christopher D. Gardner Stanford Prevention Research Center, Department of Medicine, Stanford School of Medicine, Stanford, CA94305, USA
| | - Erica D. Sonnenburg
- Microbiology & Immunology, Stanford School of Medicine, Stanford, CA, USA,Center for Human Microbiome Studies, Stanford School of Medicine, Stanford University, Stanford, CA, USA,Erica D. Sonnenburg Microbiology & Immunology, Stanford School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
39
|
Xu Y, Huang Z, Zhang B, Yu C, Li L, Li X, Li Y. Intestinal bacterial community composition of juvenile Chinese mitten crab Eriocheir sinensis under different feeding times in lab conditions. Sci Rep 2022; 12:22206. [PMID: 36564429 PMCID: PMC9789113 DOI: 10.1038/s41598-022-26785-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Feeding time is an important factor affecting the physiological activity and feeding rhythm of crustaceans. However, little is known about the factors and mechanisms contributing to variations in feeding time in aquatic species or their impacts. Moreover, the gut microbiome largely affects host physiology and is associated with diet. To investigate the effects of different feeding times on the composition of intestinal bacterial communities, high-throughput 16S rRNA sequencing was used to monitor the gut bacteria of the Chinese mitten crab Eriocheir sinensis over a 10-day period under different feeding times: 06:00 h, 12:00 h, 18:00 h, and 24:00 h. Weight gain of the day-fed groups was significantly higher than that of the night-fed groups. Two probiotics, Akkermansia muciniphila and Faecalibacterium prausnitzii, were detected in the intestines of crabs in the 12:00 group. In addition, the diversity and richness of the flora in the 12:00 group were slightly higher than those in the other treatment groups. These results collectively indicate that different feeding times change the intestinal flora composition of Chinese mitten crabs, and further identified specific feeding times associated with a more significant weight gain effect. Our findings provide important insights into improving farming strategies for Chinese mitten crabs.
Collapse
Affiliation(s)
- Yingkai Xu
- grid.412557.00000 0000 9886 8131Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Dongling Road 120, Shenyang, 110866 China
| | - Ziwei Huang
- grid.412557.00000 0000 9886 8131Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Dongling Road 120, Shenyang, 110866 China
| | - Baoli Zhang
- grid.412557.00000 0000 9886 8131Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Dongling Road 120, Shenyang, 110866 China
| | - Changyue Yu
- grid.412557.00000 0000 9886 8131Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Dongling Road 120, Shenyang, 110866 China
| | - Lisong Li
- grid.412557.00000 0000 9886 8131Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Dongling Road 120, Shenyang, 110866 China
| | - Xiaodong Li
- grid.412557.00000 0000 9886 8131Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Dongling Road 120, Shenyang, 110866 China
| | - Yingdong Li
- grid.412557.00000 0000 9886 8131Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Dongling Road 120, Shenyang, 110866 China
| |
Collapse
|
40
|
Heat-Killed Bifidobacterium bifidum B1628 May Alleviate Dextran Sulfate Sodium-Induced Colitis in Mice, and the Anti-Inflammatory Effect Is Associated with Gut Microbiota Modulation. Nutrients 2022; 14:nu14245233. [PMID: 36558391 PMCID: PMC9784753 DOI: 10.3390/nu14245233] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease associated with gut dysbiosis. This study aimed to investigate the effects of heat-killed Bifidobacterium bifidum B1628 (HB1628) in dextran sulfate sodium (DSS)-induced colitis in mice. The following three mouse groups were included (n = eight per group): NC (normal control), DSS (colitis), and HB1628 (colitis and postbiotic). The mice in the DSS group showed significant weight loss and histological damage, developed bloody diarrhea, scored high in the disease activity index (DAI), and exhibited increases in pro-inflammatory cytokines (interleukin [IL]-1β, IL-6, and tumor necrosis factor [TNF]-α) and decreases in an anti-inflammatory cytokine (IL-13) in the serum. These changes were accompanied by gut microbiota modulation in colitis mice (decreases in Rikenellaceae and Eubacterium; increases in Peptostreptococcaceae, Bacteroides vulgatus, and Parasutterella excrementihominis). The HB1628 group had lower DAIs, histology scores, and serum levels of pro-inflammatory cytokines (IL-1β and TNF-α), but higher levels of an anti-inflammatory cytokine (IL-13), compared with the DSS group, suggesting a less severe inflammatory state after the HB1628 intervention. Additionally, HB1628 improved DSS-induced gut dysbiosis, which is evidenced by increases in intestinal beneficial bacteria, such as Lactobacillus, and decreases in known unfavorable taxa in IBD, e.g., Porphyromonadaceae, Subdoligranulum, Lachnospiraceae bacterium 3_1_46FAA, and Alistipes indistinctus. Functional metagenomics revealed three significantly enriched metabolic pathways in the HB1628 group (namely, the aerobic respiration I [cytochrome c] pathway and the superpathways of L-phenylalanine biosynthesis and L-tryptophan biosynthesis, respectively). In conclusion, our results showed that HB1628 effectively improved the inflammation state and tissue damage in DSS-induced colitis mice, and the symptom relief effect was accompanied by obvious gut microbiota remodulation.
Collapse
|
41
|
Akkermansia muciniphila and Faecalibacterium prausnitzii in Immune-Related Diseases. Microorganisms 2022; 10:microorganisms10122382. [PMID: 36557635 PMCID: PMC9782003 DOI: 10.3390/microorganisms10122382] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
Probiotics and synbiotics are used to treat chronic illnesses due to their roles in immune system modulation and anti-inflammatory response. They have been shown to reduce inflammation in a number of immune-related disorders, including systemic lupus erythematosus (SLE), human immunodeficiency virus (HIV), and chronic inflammatory skin conditions such as psoriasis and atopic dermatitis (AD). Akkermansia muciniphila (A. muciniphila) and Faecalibacterium prausnitzii (F. prausnitzii) are two different types of bacteria that play a significant part in this function. It has been established that Akkermansia and Faecalibacterium are abundant in normal populations and have protective benefits on digestive health while also enhancing the immune system, metabolism, and gut barrier of the host. They have the potential to be a therapeutic target in diseases connected to the microbiota, such as immunological disorders and cancer immunotherapy. There has not been a review of the anti-inflammatory effects of Akkermansia and Faecalibacterium, particularly in immunological diseases. In this review, we highlight the most recent scientific findings regarding A. muciniphila and F. prausnitzii as two significant gut microbiota for microbiome alterations and seek to provide cutting-edge insight in terms of microbiome-targeted therapies as promising preventive and therapeutic tools in immune-related diseases and cancer immunotherapy.
Collapse
|
42
|
Han Y, Li L, Wang B. Role of Akkermansia muciniphila in the development of nonalcoholic fatty liver disease: current knowledge and perspectives. Front Med 2022; 16:667-685. [DOI: 10.1007/s11684-022-0960-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/06/2022] [Indexed: 11/19/2022]
|
43
|
Lakshmanan AP, Murugesan S, Al Khodor S, Terranegra A. The potential impact of a probiotic: Akkermansia muciniphila in the regulation of blood pressure—the current facts and evidence. Lab Invest 2022; 20:430. [PMID: 36153618 PMCID: PMC9509630 DOI: 10.1186/s12967-022-03631-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022]
Abstract
Akkermansia muciniphila (A. muciniphila) is present in the human gut microbiota from infancy and gradually increases in adulthood. The potential impact of the abundance of A. muciniphila has been studied in major cardiovascular diseases including elevated blood pressure or hypertension (HTN). HTN is a major factor in premature death worldwide, and approximately 1.28 billion adults aged 30–79 years have hypertension. A. muciniphila is being considered a next-generation probiotic and though numerous studies had highlighted the positive role of A. muciniphila in lowering/controlling the HTN, however, few studies had highlighted the negative impact of increased abundance of A. muciniphila in the management of HTN. Thus, in the review, we aimed to discuss the current facts, evidence, and controversy about the role of A. muciniphila in the pathophysiology of HTN and its potential effect on HTN management/regulation, which could be beneficial in identifying the drug target for the management of HTN.
Collapse
|