1
|
Jin Y, Li W, Rasool HMH, Ning X, Ba X, Gao Y, Guo X, Ran T, Zhou J. Effectiveness of newly isolated bacteriophages targeting multidrug-resistant Extraintestinal Pathogenic Escherichia coli strain (TZ1_3) in food preservation and mice health modulation. Food Chem 2025; 472:142833. [PMID: 39827555 DOI: 10.1016/j.foodchem.2025.142833] [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: 09/26/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Bacteriophages are promising alternatives for combating multidrug-resistant bacterial infections. Two lytic bacteriophages, named P1 and P3, targeting pathogenic Escherichia coli (ExPEC; strain TZ1_3) were isolated and evaluated for their potential ability to control pathogenic numbers either in ExPEC-contaminated food or ExPEC-infected mice. Results showed that phages significantly reduced ExPEC numbers within 6 and 12 h in contaminated water, milk, beef, and chicken when applied at 106 plaque-forming units (PFU). Notably, phage therapy administered via intraperitoneal injection (1012 PFU) effectively reduced ExPEC numbers in the heart, liver, spleen and kidney, restored α-diversity of gut microbes, and increased levels of acetic (13.98 %-37.58 %) and valeric acid (10.27 %-31.51 %) in ExPEC-infected mice. Additionally, phage injections caused no detrimental effects on body weight (which increased by 6.49 %-8.11 %), and on gut microbes in healthy mice. Overall, this study highlights the potential of phages in controlling foodborne microorganisms.
Collapse
Affiliation(s)
- Youshun Jin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Wei Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Hafiz Muhammad Hamza Rasool
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Xuan Ning
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Xuli Ba
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Yidan Gao
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China
| | - Xinyu Guo
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China
| | - Tao Ran
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China
| | - Jizhang Zhou
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China.
| |
Collapse
|
2
|
Salvatore MM, Maione A, Buonanno A, Guida M, Andolfi A, Salvatore F, Galdiero E. Biological activities, biosynthetic capacity and metabolic interactions of lactic acid bacteria and yeast strains from traditional home-made kefir. Food Chem 2025; 470:142657. [PMID: 39756085 DOI: 10.1016/j.foodchem.2024.142657] [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: 10/09/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
Given the widespread industrial and domestic use of probiotic blends based on combinations of lactic acid bacteria (LAB) and yeasts to produce fermented foods or beverages that are supposed to provide health benefits, this study aimed to generate knowledge and concepts on biologically relevant activities, metabolism and metabolic interactions in yeast/LAB communities. For this, the postbiotic capabilities of three probiotic candidates, including two lactic acid bacteria (i.e., Lactococcus lactis subsp. hordniae and Lactococcus lactis subsp. lactis) and the yeast Pichia kudriavzevii, isolated from a traditional home-made kefir, were explored combining an assortment of bioassays with a GC-MS footprint metabolomic strategy. Cell-free supernatants from cultures showed antimicrobial/antioxidant activity and inhibited biofilm formation by Salmonella sp. Several bioactive secondary metabolites (including tyrosol, phenylethyl alcohol, 2,3-butanediol, erythritol, tryptophol, putrescine, cadaverine, 3-phenyllactate, 2-hydroxyisocaproate) were detected which may contribute to the odor and flavour of the fermented products and their effects on human body.
Collapse
Affiliation(s)
- Maria Michela Salvatore
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy.
| | - Angela Maione
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy.
| | - Annalisa Buonanno
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Marco Guida
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Portici, (NA), Italy
| | - Anna Andolfi
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Portici, (NA), Italy
| | - Francesco Salvatore
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Emilia Galdiero
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| |
Collapse
|
3
|
He GS, Xia JK, Li QH, Zheng Y, Shi CR, Li R, Hong Q, Chen XM. Specnuezhenide: Comprehensive review of pharmacology, pharmacokinetics and ethnomedicinal uses. Fitoterapia 2025; 181:106389. [PMID: 39805507 DOI: 10.1016/j.fitote.2025.106389] [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: 10/14/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
BACKGROUND Specnuezhenide (SPN) is a bioactive iridoid terpenoid compound mainly found in Ligustri Lucidi Fructus (LLF), that has a broad spectrum of pharmacological effects, including anti-neoplastic, hepatoprotective, anti-aging, anti-inflammatory, immune-modulatory properties. PURPOSE The present review provides a comprehensive summary of natural medicinal plants, traditional Chinese medicine compounds containing SPN, and their corresponding pharmacological mechanisms. METHODS Using several globally recognized databases such as Web of Science, Google Scholar, PubMed, ScienceDirect, Wiley, ACS, Springer, and CNKI until December 2024, A comprehensive literature search and analysis was carried out with the keywords "Specnuezhenide", " Pharmacology ", "Pharmacokinetics" and " Chinese herbal compound". RESULTS The results indicated that SPN is present in a diverse range of plants, including LLF, Osmanthus fragrans seeds and Naked barley. SPN plays an anti-inflammatory role by regulating the NF-κB and MAPK signaling pathways, down-regulating the expression of TNF-α, IL-1β, IL-6 and other cytokines. Furthermore, many Chinese herbal compounds have been found to contain SPN, such as treatment of spleen and kidney deficiency of compound Shenhua tablet, treatment of liver-kidney Yin deficiency of Er Zhi Wan, treatment of pulmonray abscess of Qidongning and treatment of stagnation of QI due to depression of the liver of Shuganzhi Tablet. SPN is primarily distributed in the stomach, intestine, and liver. However, due to its limited absorption in the gastrointestinal tract and low blood concentration, its bioavailability is significantly reduced. CONCLUSIONS Thereby, SPN holds immense potential in the prevention and treatment of liver, lung and kidney complications. This review intends to provide a novel insight for further development of SPN, hoping to reveal the potential of SPN and necessity of further studies in this field.
Collapse
Affiliation(s)
- Guo-Sen He
- The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China
| | - Ji-Kai Xia
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China; School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qi-Hu Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China; School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Zheng
- The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China
| | - Chun-Ru Shi
- The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China
| | - Run Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China
| | - Quan Hong
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China.
| | - Xiang-Mei Chen
- The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China.
| |
Collapse
|
4
|
Rodriguez VR, Essex M, Poddubnyy D. The gut microbiota in spondyloarthritis: an update. Curr Opin Rheumatol 2025:00002281-990000000-00161. [PMID: 39968641 DOI: 10.1097/bor.0000000000001079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
PURPOSE OF REVIEW This review provides an updated overview of the gut microbiota's involvement in spondyloarthritis (SpA) from a clinical perspective. It explores mechanisms by which the gut microbiota may influence SpA pathogenesis and considers the therapeutic implications of targeting the microbiome in SpA treatment. RECENT FINDINGS The pathogenesis of SpA is multifactorial, involving genetic predisposition, external factors and dysregulation of the immune system. Recent studies have identified alterations in the gut microbiome of patients with SpA, including changes in microbial diversity and specific taxa linked to disease activity. HLA-B27 status seems to influence gut microbiota composition, potentially impacting disease progression. In HLA-B27 transgenic rats, the association between gut microbiota and SpA development has been confirmed, supporting findings from human studies. A compromised gut barrier, influenced by proteins like zonulin, may allow microbial antigens to translocate, triggering immune responses associated with SpA. SUMMARY These findings highlight the potential for microbiota-modulating therapies, such as probiotics, prebiotics, diet and exercise, in managing SpA. However, methodological variability in human studies exposes the need for more rigorous research to better understand these associations. This may offer the opportunity to refine treatment strategies, offering a personalized approach to managing the disease.
Collapse
Affiliation(s)
- Valeria Rios Rodriguez
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Morgan Essex
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Denis Poddubnyy
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Division of Rheumatology, University of Toronto and University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Yoshimura Y, Wakabayashi H, Nagano F, Matsumoto A, Shimazu S, Shiraishi A, Kido Y, Bise T, Hamada T, Yoneda K, Maeda K. Systemic inflammation is associated with gut microbiota diversity in post-stroke patients. Eur Geriatr Med 2025:10.1007/s41999-025-01159-2. [PMID: 39934474 DOI: 10.1007/s41999-025-01159-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/21/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND There is growing interest in gut microbiota and health outcomes. However, the relationship between systemic inflammation and gut microbiota diversity in hospitalized patients remains unclear. This study aimed to investigate the association in post-stroke rehabilitation patients. METHODS A cross-sectional study was conducted on post-stroke patients admitted to a rehabilitation hospital. Systemic inflammation was assessed using the modified Glasgow Prognostic Score (mGPS). Gut microbiota diversity was evaluated using three indices: Shannon index, Operational Taxonomic Unit (OTU) richness, and Faith's Phylogenetic Diversity (PD). Multiple linear regression analyses were performed to examine the relationship between mGPS and gut microbiota diversity indices, adjusting for potential confounders. RESULTS A total of 156 patients (mean age 78.4 years; 55.7% men) were analyzed. The median mGPS was 0 (interquartile range: 0-1), with GPS distribution: 61.8% scored 0, 25.7% scored 1, and 12.5% scored 2. After adjusting for confounders, mGPS was significantly and negatively associated with the Shannon index (B = -0.143, 95% CI -0.288 to -0.002, β = -0.177) and OTU richness (B = -17.832, 95% CI -24.349 to -3.951, β = -0.208). However, no significant association was observed between mGPS and Faith's PD (B = -1.155, 95% CI -2.464 to 0.189, β = -0.155). CONCLUSION This study demonstrates a significant negative association between systemic inflammation and both quantitative and qualitative gut microbiota diversity in post-stroke patients.
Collapse
Affiliation(s)
- Yoshihiro Yoshimura
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, 760 Magate, Kikuyo, Kikuchi, Kumamoto, 869-1106, Japan.
| | - Hidetaka Wakabayashi
- Department of Rehabilitation Medicine, Tokyo Women's Medical University Hospital, Shinjuku, Tokyo, Japan
| | - Fumihiko Nagano
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, 760 Magate, Kikuyo, Kikuchi, Kumamoto, 869-1106, Japan
| | - Ayaka Matsumoto
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, 760 Magate, Kikuyo, Kikuchi, Kumamoto, 869-1106, Japan
| | - Sayuri Shimazu
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, 760 Magate, Kikuyo, Kikuchi, Kumamoto, 869-1106, Japan
| | - Ai Shiraishi
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, 760 Magate, Kikuyo, Kikuchi, Kumamoto, 869-1106, Japan
| | - Yoshifumi Kido
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, 760 Magate, Kikuyo, Kikuchi, Kumamoto, 869-1106, Japan
| | - Takahiro Bise
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, 760 Magate, Kikuyo, Kikuchi, Kumamoto, 869-1106, Japan
| | - Takenori Hamada
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, 760 Magate, Kikuyo, Kikuchi, Kumamoto, 869-1106, Japan
| | - Kouki Yoneda
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, 760 Magate, Kikuyo, Kikuchi, Kumamoto, 869-1106, Japan
| | - Keisuke Maeda
- Nutrition Therapy Support Center, Aichi Medical University Hospital, Nagakute, Aichi, Japan
- Department of Geriatric Medicine, Hospital, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| |
Collapse
|
6
|
Bijnens K, Thijs S, Alfano R, McAmmond B, Van Hamme J, Artois T, Plusquin M, Vangronsveld J, Smeets K. Impact of host physiology and external stressors on the bacterial community of Schmidtea mediterranea. Sci Rep 2025; 15:4398. [PMID: 39910204 PMCID: PMC11799148 DOI: 10.1038/s41598-025-86920-0] [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: 07/25/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
To fully comprehend host-microorganism interactions, it is crucial to understand the composition and diversity of the microbiome, as well as the factors that shape these characteristics. We investigated microbiome variation using the freshwater planarian Schmidtea mediterranea, an invertebrate model in regeneration biology and (eco-)toxicology, by exposing the organisms to various controlled conditions. The microbiome composition exhibited high variability, with most of the bacteria belonging to the Betaproteobacteria. Among the diverse microbial communities, a few genera, such as Curvibacter, were consistently present, but exhibited significant alterations in response to changing conditions. The relative abundance of Curvibacter fluctuated during the regeneration process, initially increasing before returning to a composition similar to the beginning situation. After applying external stress, the relative abundance of Curvibacter and other genera decreased. Variation over time, between different origin laboratories and between individuals, showed that additional, yet to-be-identified, factors of variation are present. Taking all results together, our study provides a solid basis for future research focusing on bacterial functionality in planarians and other invertebrates.
Collapse
Affiliation(s)
- Karolien Bijnens
- Centre for Environmental Sciences, Zoology, Biodiversity and Toxicology, Hasselt University, Diepenbeek, Belgium
| | - Sofie Thijs
- Centre for Environmental Sciences, Environmental Biology, Hasselt University, Diepenbeek, Belgium
| | - Rossella Alfano
- Centre for Environmental Sciences, Epidemiology, Hasselt University, Diepenbeek, Belgium
| | - Breanne McAmmond
- Department of Biological Sciences, Thompson Rivers University, Kamloops, BC, Canada
| | - Jonathan Van Hamme
- Department of Biological Sciences, Thompson Rivers University, Kamloops, BC, Canada
| | - Tom Artois
- Centre for Environmental Sciences, Zoology, Biodiversity and Toxicology, Hasselt University, Diepenbeek, Belgium
| | - Michelle Plusquin
- Centre for Environmental Sciences, Epidemiology, Hasselt University, Diepenbeek, Belgium
| | - Jaco Vangronsveld
- Centre for Environmental Sciences, Environmental Biology, Hasselt University, Diepenbeek, Belgium
- Department of Plant Physiology and Biophysics, Institute of Biology and Biotechnology, Maria Skłodowska-Curie University, Lublin, Poland
| | - Karen Smeets
- Centre for Environmental Sciences, Zoology, Biodiversity and Toxicology, Hasselt University, Diepenbeek, Belgium.
| |
Collapse
|
7
|
Sasaki Y, Matsuo A, Hashiguchi M, Fujimura K, Koshino H, Tanaka K, Ito Y, Kitahara K, Ishiwata A, Fujita K. Structural analysis of gum arabic side chains from Acacia seyal released by bifidobacterial β-arabino-oligosaccharide 3-O-β-l-arabinopyranosyl-α-l-arabinofuranosidase. Carbohydr Polym 2025; 349:122965. [PMID: 39643419 DOI: 10.1016/j.carbpol.2024.122965] [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: 08/30/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 12/09/2024]
Abstract
Gum arabic is widely used in the food and beverage industries for its emulsifying, stabilizing, and prebiotic effects, which promote Bifidobacterium growth. The two commercially approved varieties of gum arabic, namely, Acacia senegal gum and A. seyal gum, predominantly consist of arabinogalactan protein (AGP), albeit with different side chain modifications. We previously characterized two enzymes belonging to glycoside hydrolase (GH) family 39 in bifidobacteria involved in the release of α-d-Gal-(1→3)-α-l-Ara and β-l-Arap-(1→3)-α-l-Ara from the side chains of A. senegal gum. Although the carbohydrate structure of A. senegal gum is being increasingly explored, limited information is available on A. seyal gum. In this study, we discovered a novel GH39 β-arabino-oligosaccharide 3-O-β-l-arabinopyranosyl-α-l-arabinofuranosidase from Bifidobacterium catenulatum and revealed the accurate structure of β-l-arabino-oligosaccharides released from A. seyal gum as [β-l-Araf-(1→2)-]n-β-l-Arap-(1→3)-α-l-Araf-(1→) (n = 0-3). Growth assays and intracellular enzyme activity assessments using B. catenulatum revealed that β-l-arabino-oligosaccharides were degraded to l-arabinose by GH127 β-l-arabinofuranosidase and GH36 β-l-arabinopyranosidase. This study provides new insights into the diversity of AGP structures and the utilization mechanisms of A. seyal gum in bifidobacteria.
Collapse
Affiliation(s)
- Yuki Sasaki
- Faculty of Agriculture, Kagoshima University, Kagoshima, Kagoshima, Japan; Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Ayako Matsuo
- Faculty of Agriculture, Kagoshima University, Kagoshima, Kagoshima, Japan
| | - Mimika Hashiguchi
- Faculty of Agriculture, Kagoshima University, Kagoshima, Kagoshima, Japan
| | - Kanoko Fujimura
- Faculty of Agriculture, Kagoshima University, Kagoshima, Kagoshima, Japan
| | - Hiroyuki Koshino
- RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Katsunori Tanaka
- RIKEN Cluster for Pioneering Research, Wako, Saitama, Japan; Department of Chemical Science and Engineering, Tokyo Institute of Technology, Meguro, Tokyo, Japan
| | - Yukishige Ito
- RIKEN Cluster for Pioneering Research, Wako, Saitama, Japan; Graduate School of Science, Osaka University, Toyonaka, Osaka, Japan
| | - Kanefumi Kitahara
- Faculty of Agriculture, Kagoshima University, Kagoshima, Kagoshima, Japan
| | | | - Kiyotaka Fujita
- Faculty of Agriculture, Kagoshima University, Kagoshima, Kagoshima, Japan.
| |
Collapse
|
8
|
Baker JS, Qu E, Mancuso CP, Tripp AD, Conwill A, Lieberman TD. Previously hidden intraspecies dynamics underlie the apparent stability of two important skin microbiome species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.01.10.575018. [PMID: 38260404 PMCID: PMC10802602 DOI: 10.1101/2024.01.10.575018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Adult human facial skin microbiomes are remarkably similar at the species-level, dominated by Cutibacterium acnes and Staphylococcus epidermidis, yet each person harbors a unique community of strains. Understanding how person-specific communities assemble is critical for designing microbiome-based therapies. Here, using 4,055 isolate genomes and 360 metagenomes, we reconstruct on-person evolutionary history to reveal on and between-person strain dynamics. We find that multiple cells are typically involved in transmission, indicating ample opportunity for migration. Despite this accessibility, family members share only some of their strains. S. epidermidis communities are dynamic, with each strain persisting for an average of only 2 years. C. acnes strains are more stable and have a higher colonization rate during the transition to an adult facial skin microbiome, suggesting this window could facilitate engraftment of therapeutic strains. These previously undetectable dynamics may influence the design of microbiome therapeutics and motivate the study of their effects on hosts.
Collapse
|
9
|
Almousa S, Kim S, Kumar A, Su Y, Singh S, Mishra S, Fonseca MM, Rather HA, Romero-Sandoval EA, Hsu FC, Singh R, Yadav H, Mishra S, Deep G. Bacterial Nanovesicles as Interkingdom Signaling Moieties Mediating Pain Hypersensitivity. ACS NANO 2025; 19:3210-3225. [PMID: 39818729 DOI: 10.1021/acsnano.4c10529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Gut dysbiosis contributes to multiple pathologies, yet the mechanisms of the gut microbiota-mediated influence on systemic and distant responses remain largely elusive. This study aimed to identify the role of nanosized bacterial extracellular vesicles (bEVs) in mediating allodynia, i.e., pain hypersensitivity, in a diet-induced obesity (DIO) gut dysbiosis model. bEVs were enriched from the feces of lean (bEVLean) and DIO (bEVDIO) mice by an approach combining ultracentrifugation and immunoprecipitation and then extensively analyzed for purity and bacterial characteristics. Next, bEVs were injected, either intraplantarly or intravenously, in mice to assess pain sensitivity. Fluorescence-labeled bEVs were injected in mice by enema to assess biodistribution. The effect of bEV on immune cells and inflammation was analyzed by array, immunophenotyping, microscopy, NF-κB activation, and cellular uptake assays. Results showed that bEVDIO administration in wild-type mice replicated the allodynia phenotype observed in DIO mice for both mechanical and thermal stimuli. Importantly, this effect was compromised in TRPA1/TRPV1 double-knockout mice. Biodistribution analyses showed bEV entry into systemic circulation with subsequent localization at distant sites. Multiple analyses revealed that bEVDIO exposure incited systemic inflammation, primarily through modulating the innate immune system. This inflammatory mechanism involved LPS on the bEV surface, activating TLR2- and TLR4-related pathways, as confirmed using TLR2 and TLR4 inhibitors and shaving bEV surface proteins. Interestingly, the enhanced cellular uptake of bEVDIO was contingent on interactions involving LPS and proteins on bEVs and TLR2/TLR4 on monocytes. These findings illuminate the hitherto unexplored role of bEV as pivotal mediators of allodynia and inflammation linked to gut dysbiosis.
Collapse
Affiliation(s)
- Sameh Almousa
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Susy Kim
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Ashish Kumar
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Yixin Su
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Sangeeta Singh
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Shalini Mishra
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Miriam M Fonseca
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Hilal A Rather
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - E Alfonso Romero-Sandoval
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Rakesh Singh
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, Florida 33612, United States
| | - Santosh Mishra
- Comparative Pain Research and Education Center, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina 27607, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Gagan Deep
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| |
Collapse
|
10
|
Xu XT, Jiang MJ, Fu YL, Xie F, Li JJ, Meng QH. Gut microbiome composition in patients with liver cirrhosis with and without hepatic encephalopathy: A systematic review and meta-analysis. World J Hepatol 2025; 17:100377. [PMID: 39871903 PMCID: PMC11736471 DOI: 10.4254/wjh.v17.i1.100377] [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: 08/15/2024] [Revised: 10/26/2024] [Accepted: 11/19/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND The gut microbiome is associated with hepatic encephalopathy (HE), but research results on the gut microbiome characteristics of patients with liver cirrhosis with and without HE are inconsistent. AIM To study the gut microbiota characteristics of patients with liver cirrhosis with and without HE. METHODS We searched the PubMed, Web of Science, EMBASE, and Cochrane databases using two keywords, HE, and gut microbiome. According to the inclusion and exclusion criteria, suitable literature was screened to extract data on the diversity and composition of the fecal microbiota in patients with liver cirrhosis with and without HE. The data were analyzed using RevMan and STATA. RESULTS Seventeen studies were included: (1) A meta-analysis of 7 studies revealed that the Shannon index in liver cirrhosis patients with HE was significantly lower than that in patients without HE [-0.20, 95% confidence interval (CI): -0.28 to -0.13, I2 = 20%]; (2) The relative abundances of Lachnospiraceae (-2.73, 95%CI: -4.58 to -0.87, I2 = 38%) and Ruminococcaceae (-2.93, 95%CI: -4.29 to -1.56, I2 = 0%) in liver cirrhosis patients with HE was significantly lower than those in patients without HE; (3) In patients with HE, Enterococcus, Proteobacteria, Enterococcaceae, and Enterobacteriaceae proportions increased, but Ruminococcaceae, Lachnospiraceae, Prevotellaceae, and Bacteroidetes proportions decreased; (4) Differences in the fecal metabolome between liver cirrhosis patients with and without HE were detected; and (5) Differential gut microbiomes may serve as diagnostic and prognostic tools. CONCLUSION The gut microbiomes of patients with liver cirrhosis with and without HE differ. Some gut microbiomes may distinguish liver cirrhosis patients with or without HE and determine patient prognosis.
Collapse
Affiliation(s)
- Xiao-Tong Xu
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing 100069, China
- Interventional Therapy Center for Oncology, Beijing YouAn Hospital, Capital Medical University, Beijing 100069, China
| | - Min-Jie Jiang
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250000, Shandong Province, China
| | - Yun-Lai Fu
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing 100069, China
- Interventional Therapy Center for Oncology, Beijing YouAn Hospital, Capital Medical University, Beijing 100069, China
| | - Fang Xie
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing 100069, China
| | - Jian-Jun Li
- Interventional Therapy Center for Oncology, Beijing YouAn Hospital, Capital Medical University, Beijing 100069, China
| | - Qing-Hua Meng
- Interventional Therapy Center for Oncology, Beijing YouAn Hospital, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
11
|
Cubillejo I, Theis KR, Panzer J, Luo X, Banerjee S, Thummel R, Withey JH. Vibrio cholerae Gut Colonization of Zebrafish Larvae Induces a Dampened Sensorimotor Response. Biomedicines 2025; 13:226. [PMID: 39857809 PMCID: PMC11761238 DOI: 10.3390/biomedicines13010226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Cholera is a diarrheal disease prevalent in populations without access to clean water. Cholera is caused by Vibrio cholerae, which colonizes the upper small intestine in humans once ingested. A growing number of studies suggest that the gut microbiome composition modulates animal behavior. Zebrafish are an established cholera model that can maintain a complex, mature gut microbiome during infection. Larval zebrafish, which have immature gut microbiomes, provide the advantage of high-throughput analyses for established behavioral models. Methods: We identified the effects of V. cholerae O1 El Tor C6706 colonization at 5 days post-fertilization (dpf) on larval zebrafish behavior by tracking startle responses at 10 dpf. We also characterized the larval gut microbiome using 16S rRNA sequencing. V. cholerae-infected or uninfected control groups were exposed to either an alternating light/dark stimuli or a single-tap stimulus, and average distance and velocity were tracked. Results: While there was no significant difference in the light/dark trial, we report a significant decrease in distance moved for C6706-colonized larvae during the single-tap trial. Conclusion: This suggests that early V. cholerae colonization of the larval gut microbiome has a dampening effect on sensorimotor function, supporting the idea of a link between the gut microbiome and behavior.
Collapse
Affiliation(s)
- Isabella Cubillejo
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University, Detroit, MI 48201, USA; (I.C.)
| | - Kevin R. Theis
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University, Detroit, MI 48201, USA; (I.C.)
| | - Jonathan Panzer
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University, Detroit, MI 48201, USA; (I.C.)
| | - Xixia Luo
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Shreya Banerjee
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Ryan Thummel
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jeffrey H. Withey
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University, Detroit, MI 48201, USA; (I.C.)
| |
Collapse
|
12
|
Fanijavadi S, Jensen LH. Dysbiosis-NK Cell Crosstalk in Pancreatic Cancer: Toward a Unified Biomarker Signature for Improved Clinical Outcomes. Int J Mol Sci 2025; 26:730. [PMID: 39859442 PMCID: PMC11765696 DOI: 10.3390/ijms26020730] [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: 12/18/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with poor prognosis, primarily due to its immunosuppressive tumor microenvironment (TME), which contributes to treatment resistance. Recent research shows that the microbiome, including microbial communities in the oral cavity, gut, bile duct, and intratumoral environments, plays a key role in PDAC development, with microbial imbalances (dysbiosis) promoting inflammation, cancer progression, therapy resistance, and treatment side effects. Microbial metabolites can also affect immune cells, especially natural killer (NK) cells, which are vital for tumor surveillance, therapy response and treatment-related side effects. Dysbiosis can affect NK cell function, leading to resistance and side effects. We propose that a combined biomarker approach, integrating microbiome composition and NK cell profiles, can help predict treatment resistance and side effects, enabling more personalized therapies. This review examines how dysbiosis contributes to NK cell dysfunction in PDAC and discusses strategies (e.g., antibiotics, probiotics, vaccines) to modulate the microbiome and enhance NK cell function. Targeting dysbiosis could modulate NK cell activity, improve the effectiveness of PDAC treatments, and reduce side effects. However, further research is needed to develop unified NK cell-microbiome interaction-based biomarkers for more precise and effective patient outcomes.
Collapse
Affiliation(s)
- Sara Fanijavadi
- Cancer Polyclinic, Levanger Hospital, 7601 Levanger, Trøndelag, Norway
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
| | - Lars Henrik Jensen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
- Department of Oncology, Institute of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
| |
Collapse
|
13
|
Sharma SA, Oladejo SO, Kuang Z. Chemical interplay between gut microbiota and epigenetics: Implications in circadian biology. Cell Chem Biol 2025; 32:61-82. [PMID: 38776923 PMCID: PMC11569273 DOI: 10.1016/j.chembiol.2024.04.016] [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: 12/15/2023] [Revised: 03/22/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Circadian rhythms are intrinsic molecular mechanisms that synchronize biological functions with the day/night cycle. The mammalian gut is colonized by a myriad of microbes, collectively named the gut microbiota. The microbiota impacts host physiology via metabolites and structural components. A key mechanism is the modulation of host epigenetic pathways, especially histone modifications. An increasing number of studies indicate the role of the microbiota in regulating host circadian rhythms. However, the mechanisms remain largely unknown. Here, we summarize studies on microbial regulation of host circadian rhythms and epigenetic pathways, highlight recent findings on how the microbiota employs host epigenetic machinery to regulate circadian rhythms, and discuss its impacts on host physiology, particularly immune and metabolic functions. We further describe current challenges and resources that could facilitate research on microbiota-epigenetic-circadian rhythm interactions to advance our knowledge of circadian disorders and possible therapeutic avenues.
Collapse
Affiliation(s)
- Samskrathi Aravinda Sharma
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Sarah Olanrewaju Oladejo
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Zheng Kuang
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA.
| |
Collapse
|
14
|
Astigarraga M, Sánchez-Ruiz A, Diop-Aw A, Quintero R, Colmenarejo G. How Do Microbial Metabolites Interact with Their Protein Targets? J Chem Inf Model 2025; 65:201-213. [PMID: 39743764 DOI: 10.1021/acs.jcim.4c01875] [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: 01/04/2025]
Abstract
The design of drugs and nutraceutics that mimic microbial metabolites is an emerging drug modality in medicinal chemistry that attempts to modulate the myriad of interactions that these molecules establish with host and microbial proteins. Understanding how microbial metabolites interact with their target proteins is key to perform a rational design of metabolite mimetic molecules for therapeutic usage. In the present work, we address this question by analyzing the functional groups of these molecules and the interactions they display in a set of more than 71K protein-metabolite interactions from the PDB. Significant differences in the functional group distributions, their chemical features, and their co-occurrences are observed for distinct subsets of these molecules. The same is true for the distributions of interaction types. By correlating both data sets, we are able to explain the observed interaction patterns in terms of observed functional group patterns. These results will shed light on the rational design of novel metabolite mimetic molecules for therapeutic purposes.
Collapse
Affiliation(s)
- Mario Astigarraga
- Biostatistics and Bioinformatics Unit, IMDEA Food CEI UAM+CSIC, Madrid E28049, Spain
| | - Andrés Sánchez-Ruiz
- Biostatistics and Bioinformatics Unit, IMDEA Food CEI UAM+CSIC, Madrid E28049, Spain
| | - Aminata Diop-Aw
- Biostatistics and Bioinformatics Unit, IMDEA Food CEI UAM+CSIC, Madrid E28049, Spain
| | - Raquel Quintero
- Biostatistics and Bioinformatics Unit, IMDEA Food CEI UAM+CSIC, Madrid E28049, Spain
| | - Gonzalo Colmenarejo
- Biostatistics and Bioinformatics Unit, IMDEA Food CEI UAM+CSIC, Madrid E28049, Spain
| |
Collapse
|
15
|
Nishijima S, Stankevic E, Aasmets O, Schmidt TSB, Nagata N, Keller MI, Ferretti P, Juel HB, Fullam A, Robbani SM, Schudoma C, Hansen JK, Holm LA, Israelsen M, Schierwagen R, Torp N, Telzerow A, Hercog R, Kandels S, Hazenbrink DHM, Arumugam M, Bendtsen F, Brøns C, Fonvig CE, Holm JC, Nielsen T, Pedersen JS, Thiele MS, Trebicka J, Org E, Krag A, Hansen T, Kuhn M, Bork P. Fecal microbial load is a major determinant of gut microbiome variation and a confounder for disease associations. Cell 2025; 188:222-236.e15. [PMID: 39541968 DOI: 10.1016/j.cell.2024.10.022] [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: 03/08/2024] [Revised: 07/12/2024] [Accepted: 10/14/2024] [Indexed: 11/17/2024]
Abstract
The microbiota in individual habitats differ in both relative composition and absolute abundance. While sequencing approaches determine the relative abundances of taxa and genes, they do not provide information on their absolute abundances. Here, we developed a machine-learning approach to predict fecal microbial loads (microbial cells per gram) solely from relative abundance data. Applying our prediction model to a large-scale metagenomic dataset (n = 34,539), we demonstrated that microbial load is the major determinant of gut microbiome variation and is associated with numerous host factors, including age, diet, and medication. We further found that for several diseases, changes in microbial load, rather than the disease condition itself, more strongly explained alterations in patients' gut microbiome. Adjusting for this effect substantially reduced the statistical significance of the majority of disease-associated species. Our analysis reveals that the fecal microbial load is a major confounder in microbiome studies, highlighting its importance for understanding microbiome variation in health and disease.
Collapse
Affiliation(s)
- Suguru Nishijima
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Evelina Stankevic
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Oliver Aasmets
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Thomas S B Schmidt
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Naoyoshi Nagata
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan
| | - Marisa Isabell Keller
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Pamela Ferretti
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Helene Bæk Juel
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Anthony Fullam
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Christian Schudoma
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johanne Kragh Hansen
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Louise Aas Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
| | - Mads Israelsen
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Robert Schierwagen
- Department of Internal Medicine B, University of Münster, Münster, Germany
| | - Nikolaj Torp
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Anja Telzerow
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Rajna Hercog
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Stefanie Kandels
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Diënty H M Hazenbrink
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Manimozhiyan Arumugam
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Bendtsen
- Gastrounit, Medical Division, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Charlotte Brøns
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Cilius Esmann Fonvig
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens-Christian Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Trine Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Medical department, University Hospital Zeeland, Køge, Denmark
| | - Julie Steen Pedersen
- Gastrounit, Medical Division, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Maja Sofie Thiele
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Jonel Trebicka
- Department of Internal Medicine B, University of Münster, Münster, Germany; European Foundation for the Study of Chronic Liver Failure, EFCLIF, Barcelona, Spain
| | - Elin Org
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Aleksander Krag
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Michael Kuhn
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Peer Bork
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany; Max Delbrück Centre for Molecular Medicine, Berlin, Germany; Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
16
|
Lim MY, Hong S, Nam YD. Understanding the role of the gut microbiome in solid tumor responses to immune checkpoint inhibitors for personalized therapeutic strategies: a review. Front Immunol 2025; 15:1512683. [PMID: 39840031 PMCID: PMC11747443 DOI: 10.3389/fimmu.2024.1512683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Immunotherapy, especially immune checkpoint inhibitor (ICI) therapy, has yielded remarkable outcomes for some patients with solid cancers, but others do not respond to these treatments. Recent research has identified the gut microbiota as a key modulator of immune responses, suggesting that its composition is closely linked to responses to ICI therapy in cancer treatment. As a result, the gut microbiome is gaining attention as a potential biomarker for predicting individual responses to ICI therapy and as a target for enhancing treatment efficacy. In this review, we discuss key findings from human observational studies assessing the effect of antibiotic use prior to ICI therapy on outcomes and identifying specific gut bacteria associated with favorable and unfavorable responses. Moreover, we review studies investigating the possibility of patient outcome prediction using machine learning models based on gut microbiome data before starting ICI therapy and clinical trials exploring whether gut microbiota modulation, for example via fecal microbiota transplantation or live biotherapeutic products, can improve results of ICI therapy in patients with cancer. We also briefly discuss the mechanisms through which the gut microbial-derived products influence immunotherapy effectiveness. Further research is necessary to fully understand the complex interactions between the host, gut microbiota, and immunotherapy and to develop personalized strategies that optimize responses to ICI therapy.
Collapse
Affiliation(s)
- Mi Young Lim
- Personalized Diet Research Group, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Seungpyo Hong
- Department of Molecular Biology, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
| | - Young-Do Nam
- Personalized Diet Research Group, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
| |
Collapse
|
17
|
Caty SN, Alvarez-Buylla A, Vasek C, Tapia EE, Martin NA, McLaughlin T, Golde CL, Weber PK, Mayali X, Coloma LA, Morris MM, O'Connell LA. Alkaloids are associated with increased microbial diversity and metabolic function in poison frogs. Curr Biol 2025; 35:187-197.e8. [PMID: 39637856 DOI: 10.1016/j.cub.2024.10.069] [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: 01/11/2024] [Revised: 09/15/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024]
Abstract
Shifts in host-associated microbiomes can impact both host and microbes.1,2,3,4,5,6 It is of interest to understand how perturbations, like the introduction of exogenous chemicals,7,8,9,10,11,12,13 impact microbiomes. In poison frogs (family Dendrobatidae), the skin microbiome is exposed to alkaloids that the frogs sequester for defense.14,15,16,17,18,19 These alkaloids are antimicrobial20,21,22; however, their effect on the frogs' skin microbiome is unknown. To test this, we characterized microbial communities from field-collected dendrobatid frogs. Then, we conducted a laboratory experiment to monitor the effect of the alkaloid decahydroquinoline (DHQ) on the microbiome of two frog species with contrasting alkaloid loads in nature. In both datasets, we found that alkaloid-exposed microbiomes were more phylogenetically diverse, with an increase in diversity among rare taxa. To better understand the isolate-specific response to alkaloids, we cultured microbial isolates from poison frog skin and found that many isolates exhibited enhanced growth or were not impacted by the addition of DHQ. To further explore the microbial response to alkaloids, we sequenced the metagenomes from high- and low-alkaloid frogs and observed a greater diversity of genes associated with nitrogen and carbon metabolism in high-alkaloid frogs. From these data, we hypothesized that some strains may metabolize the alkaloids. We used stable isotope tracing coupled to nanoSIMS (nanoscale secondary ion mass spectrometry), which supported the idea that some of these isolates are able to metabolize DHQ. Together, these data suggest that poison frog alkaloids open new niches for skin-associated microbes with specific adaptations, such as alkaloid metabolism, that enable survival in this environment.
Collapse
Affiliation(s)
- Stephanie N Caty
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| | | | - Cooper Vasek
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Elicio E Tapia
- Leibniz Institute for the Analysis of Biodiversity Change, Martin-Luther-King-Platz 3, Hamburg 20146, Germany
| | - Nora A Martin
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Theresa McLaughlin
- Stanford University Mass Spectrometry, Stanford University, Stanford, CA 94305, USA
| | - Chloe L Golde
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Peter K Weber
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Xavier Mayali
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Luis A Coloma
- Centro Jambatu de Investigación y Conservación de Anfibios, Fundación Jambatu, San Rafael, Quito 170810, Ecuador
| | - Megan M Morris
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Lauren A O'Connell
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
18
|
Misera A, Kaczmarczyk M, Łoniewski I, Liśkiewicz P, Podsiadło K, Misiak B, Skonieczna-Żydecka K, Samochowiec J. Comparative analysis of gut microbiota in major depressive disorder and schizophrenia during hospitalisation - the case-control, post hoc study. Psychoneuroendocrinology 2025; 171:107208. [PMID: 39426041 DOI: 10.1016/j.psyneuen.2024.107208] [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: 05/06/2024] [Revised: 08/14/2024] [Accepted: 10/01/2024] [Indexed: 10/21/2024]
Abstract
The aim of this study was to investigate the relationship between gut microbiota and major depressive disorder (MDD) and schizophrenia (SCZ) by comparing 36 inpatients with these conditions to 29 healthy controls (HC) matched for age, sex, and body mass index (BMI). Individuals with SCZ exhibited greater microbiota richness compared to HC (FDR P(Q)=0.028). Taxonomically, while no significant differences were observed between the microbiota of MDD and SCZ patients in a head-to-head comparison, both patient groups differed significantly when compared to HC. Interestingly, besides common patterns (such as a higher abundance of Erysipelotrichaceae UCG-003 and Streptococcus, and a lower abundance of Lachnospiraceae ND3007 group), unique patterns were exhibited only in MDD (with a higher abundance of Anaerostipes, Q=0.004) or SCZ (with a higher abundance of Sutterella, Q=0.001, and a lower abundance of Clostridium sensu stricto 1, Q=0.002). The Random Forest algorithm identified Ruminococcus torques group, Lachnospiraceae UCG-001, and Erysipelotrichaceae UCG-003 as highly discriminative features for both SCZ and MDD, while Suturella and Holdemania were unique features for SZC, and Lachnospiraceae genus CAG-56 and Anaerostipes for MDD. Additionally, between 50 % and 60 % of the differentially abundant taxa were found among the top 10 influential features in the RF models. In conclusion, while no significant differences were found between the microbiota of MDD and SCZ patients, distinct microbial patterns were found in each group when compared to HC. The study did not confirm universal microbial biomarkers reported in other studies but showed that the observed differences concern the bacteria associated with inflammation, the production of short chain fatty acids (SCFA), and the synthesis of metabolites linked to mental health (lactic acid, gamma-aminobutyric acid - GABA). The application of machine learning holds promise for further understanding the complex relationship between microbiota and these psychiatric disorders. The observed results should be treated with caution due to the limitations of this study (mainly sample size), therefore further researches under standardized environmental conditions with consistent analytical and bioinformatics approaches are warranted.
Collapse
Affiliation(s)
- Agata Misera
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Mariusz Kaczmarczyk
- Sanprobi sp. z o. o. sp. k, Szczecin, Poland; Department of Biochemical Science, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Igor Łoniewski
- Sanprobi sp. z o. o. sp. k, Szczecin, Poland; Department of Biochemical Science, Pomeranian Medical University in Szczecin, Szczecin, Poland.
| | - Paweł Liśkiewicz
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | | | - Błażej Misiak
- Department of Psychiatry, Wrocław Medical University, Wrocław, Poland
| | | | - Jerzy Samochowiec
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
19
|
Wang J, Zhou T. Unveiling gut microbiota's role: Bidirectional regulation of drug transport for improved safety. Med Res Rev 2025; 45:311-343. [PMID: 39180410 DOI: 10.1002/med.22077] [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: 05/09/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/26/2024]
Abstract
Drug safety is a paramount concern in the field of drug development, with researchers increasingly focusing on the bidirectional regulation of gut microbiota in this context. The gut microbiota plays a crucial role in maintaining drug safety. It can influence drug transport processes in the body through various mechanisms, thereby modulating their efficacy and toxicity. The main mechanisms include: (1) The gut microbiota directly interacts with drugs, altering their chemical structure to reduce toxicity and enhance efficacy, thereby impacting drug transport mechanisms, drugs can also change the structure and abundance of gut bacteria; (2) bidirectional regulation of intestinal barrier permeability by gut microbiota, promoting the absorption of nontoxic drugs and inhibiting the absorption of toxic components; (3) bidirectional regulation of the expression and activity of transport proteins by gut microbiota, selectively promoting the absorption of effective components or inhibiting the absorption of toxic components. This bidirectional regulatory role enables the gut microbiota to play a key role in maintaining drug balance in the body and reducing adverse reactions. Understanding these regulatory mechanisms sheds light on novel approaches to minimize toxic side effects, enhance drug efficacy, and ultimately improve drug safety. This review systematically examines the bidirectional regulation of gut microbiota in drug transportation from the aforementioned aspects, emphasizing their significance in ensuring drug safety. Furthermore, it offers a prospective outlook from the standpoint of enhancing therapeutic efficacy and reducing drug toxicity, underscoring the importance of further exploration in this research domain. It aims to provide more effective strategies for drug development and treatment.
Collapse
Affiliation(s)
- Jinyi Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
20
|
Rodriguez J, Cordaillat-Simmons M, Badalato N, Berger B, Breton H, de Lahondès R, Deschasaux-Tanguy M, Desvignes C, D'Humières C, Kampshoff S, Lavelle A, Metwaly A, Quijada NM, Seegers JFML, Udocor A, Zwart H, Maguin E, Doré J, Druart C. Microbiome testing in Europe: navigating analytical, ethical and regulatory challenges. MICROBIOME 2024; 12:258. [PMID: 39695869 DOI: 10.1186/s40168-024-01991-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND In recent years, human microbiome research has flourished and has drawn attention from both healthcare professionals and general consumers as the human microbiome is now recognized as having a significant influence on human health. This has led to the emergence of companies offering microbiome testing services. Some of these services are sold directly to the consumer via companies' websites or via medical laboratory websites. METHODOLOGY In order to provide an overview of the consumer experience proposed by these microbiome testing services, one single faecal sample was sent to six different companies (five based in Europe and one based in the USA). Two out of the six testing kits were commercialized by medical laboratories, but without any requirement for a medical prescription. The analyses and reports received were discussed with a panel of experts (21 experts from 8 countries) during an online workshop. RESULTS This workshop led to the identification of several limitations and challenges related to these kits, including over-promising messages from the companies, a lack of transparency in the methodology used for the analysis and a lack of reliability of the results. The experts considered the interpretations and recommendations provided in the different reports to be premature due to the lack of robust scientific evidence and the analyses associated with the reports to be of limited clinical utility. The experts also discussed the grey areas surrounding the regulatory status of these test kits, including their positioning in the European market. The experts recommended a distinction between regulatory requirements based on the intended use or purpose of the kit: on the one hand, test kits developed to satisfy consumer curiosity, with a clear mention of this objective, and no mention of any disease or risk of disease, and on the other hand, in vitro diagnostic (IVD) CE-marked test kits, which could go deeper into the analysis and interpretation of samples, as such a report would be intended for trained healthcare professionals. CONCLUSIONS Recommendations or actions, specific to the context of use of microbiome testing kits, are listed to improve the quality and the robustness of these test kits to meet expectations of end users (consumers, patients and healthcare professionals). The need for standardization, robust scientific evidence, qualification of microbiome-based biomarkers and a clear regulatory status in Europe are the main issues that will require attention in the near future to align laboratory development with societal needs and thus foster translation into daily health practice.
Collapse
Affiliation(s)
| | | | | | - Bernard Berger
- Nestlé Research, Nestlé Institute of Health Sciences, Route du Jorat 57, 1000, Lausanne 26, Switzerland
| | | | | | - Mélanie Deschasaux-Tanguy
- Nutritional Epidemiology Research Team (EREN), Center for Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, CNAM, Université Sorbonne Paris Nord and Université Paris Cité, 93017, Bobigny, France
| | - Clara Desvignes
- Voisin Consulting Life Sciences, Boulogne-Billancourt, France
| | | | | | - Aonghus Lavelle
- Department of Anatomy & Neuroscience and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Amira Metwaly
- Chair of Nutrition and Immunology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Narciso M Quijada
- Department of Microbiology and Genetics, Institute for Agribiotechnology Research (CIALE), University of Salamanca (USAL), Salamanca, Spain
| | | | | | - Hub Zwart
- Erasmus School of Philosophy, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Emmanuelle Maguin
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Joël Doré
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
- MGP MetaGenoPolis, INRAE, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Céline Druart
- Pharmabiotic Research Institute, 11100, Narbonne, France
| |
Collapse
|
21
|
Dias NW, Poole R, Soffa DR, Brown KJH. Dynamic principles of the microbiome and the bovine vagina: a review. Front Microbiol 2024; 15:1434498. [PMID: 39703711 PMCID: PMC11655496 DOI: 10.3389/fmicb.2024.1434498] [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: 05/17/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024] Open
Abstract
The role of microbes inhabiting various body sites in supporting host physiology and health is substantial, and recent advancements in DNA sequencing technology have facilitated a more in-depth understanding of these microbial contributions. The influence of microbiota within a given organ can be broadly categorized as having two main functions: (1) promoting organ homeostasis and (2) creating conditions that inhibit the growth of pathogenic microorganisms, thereby protecting the host from diseases. In livestock production, numerous phenotypes critical to industry outcomes are affected by the microbiome, which has sparked considerable academic interest in recent years. This review aims to analyze the extensive data available on the microbiomes of humans and other mammalian species, examining microbiome ecology to elucidate principles that may assist in interpreting data on livestock microbiomes. Additionally, the review will discuss techniques available for investigating various microbiome aspects and will examine existing data on the reproductive microbiome, with a particular focus on the bovine vaginal microbiome.
Collapse
Affiliation(s)
- Nicholas Wege Dias
- Department of Animal Sciences & Industry, Kansas State University, Manhattan, KS, United States
| | - Rebecca Poole
- Department of Animal Science, Texas A and M University, College Station, TX, United States
| | - Dallas R. Soffa
- Department of Animal Science, Texas A and M University, College Station, TX, United States
| | | |
Collapse
|
22
|
Zhao W, Li S, Li Q, Li Q, Zheng Y, Lu H. Mendelian randomization reveals predictive, preventive, and personalized insights into inflammatory bowel disease: the role of gut microbiome and circulating inflammatory proteins. EPMA J 2024; 15:693-709. [PMID: 39635016 PMCID: PMC11612091 DOI: 10.1007/s13167-024-00384-2] [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: 09/09/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024]
Abstract
Background A chronic illness with increasing global frequency, inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD), profoundly affects patients' quality of life and healthcare systems. IBD pathogenesis consists of changes in gut microbiota, immune system dysregulation, and genetic predisposition. Although emerging data suggests that gut microbiota and circulating inflammatory proteins play critical roles in IBD, their utility as biomarkers for predictive, preventive, and personalized medicine (PPPM) remains incompletely understood. Working hypothesis and methods We hypothesized that specific gut microbiota and inflammatory proteins causally influence IBD risk and mediate pathways between gut microbiota and IBD development. We employed Mendelian randomization (MR) using genome-wide association studies (GWAS) to explore these causal relationships, including further analyses on UC and CD subtypes. Results We identified eight gut microbiota species linked to IBD, with four protective and four increasing risk. Nine inflammatory proteins were also associated, six increasing risk and three protective. MMP-10 and IL-10Rα mediated the effects of Clostridiaceae1 on IBD risk. For UC, five microbiota species were protective, five were risk factors, and two proteins increased risk while three were protective. IL-10Rα mediated the effects of Clostridiaceae1 on UC risk. For CD, eight microbiota species were protective, four increased risk, and nine proteins were implicated. However, no mediation pathways were supported by multivariable MR. Conclusions This study highlights specific gut microbiota and inflammatory proteins that may serve as therapeutic targets for PPPM in IBD, UC, and CD. These findings offer new insights into IBD pathogenesis and suggest potential avenues for improved prevention, early detection, and personalized treatment strategies. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00384-2.
Collapse
Affiliation(s)
- Wuqing Zhao
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Shixiao Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Qianqian Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Qiang Li
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ya Zheng
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Hong Lu
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
23
|
Comerlato CB, Zhang X, Walker K, Mayne J, Figeys D, Brandelli A. The Influence of Protein Secretomes of Enterococcus durans on ex vivo Human Gut Microbiome. Probiotics Antimicrob Proteins 2024; 16:1954-1965. [PMID: 37589783 DOI: 10.1007/s12602-023-10136-9] [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] [Accepted: 08/07/2023] [Indexed: 08/18/2023]
Abstract
The gut microbiome plays a critical role to all animals and humans health. Methods based on ex vivo cultures are time and cost-effective solutions for rapid evaluation of probiotic effects on microbiomes. In this study, we assessed whether the protein secretome from the potential probiotic Enterococcus durans LAB18S grown on fructoligosaccharides (FOS) and galactoligosaccharides (GOS) had specific effects on ex vivo cultured intestinal microbiome obtained from a healthy individual. Metaproteomics was used to evaluate changes in microbial communities of the human intestinal microbiome. Hierarchical clustering analysis revealed 654 differentially abundant proteins from the metaproteome samples, showing that gut microbial protein expression varied on the presence of different E. durans secretomes. Increased amount of Bacteroidetes phylum was observed in treatments with secretomes from E. durans cultures on FOS, GOS and albumin, resulting in a decrease of the Firmicutes to Bacteroidetes (F/B) ratio. The most functionally abundant bacterial taxa were Roseburia, Bacteroides, Alistipes and Faecalibacterium. The results suggest that the secretome of E. durans may have favorable effects on the intestinal microbial composition, stimulating growth and different protein expression of beneficial bacteria. These findings suggest that proteins secreted by E. durans growing on FOS and GOS have different effects on the modulation of gut microbiota functional activities during cultivation.
Collapse
Affiliation(s)
- Carolina Baldisserotto Comerlato
- Laboratório de Bioquímica e Microbiologia Aplicada, Instituto de Ciência e Tecnologia de Alimentos, Universidade Federal do Rio Grande do Sul, 91510-970, Porto Alegre, Brazil
| | - Xu Zhang
- School of Pharmaceutical Sciences, Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Krystal Walker
- School of Pharmaceutical Sciences, Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Janice Mayne
- School of Pharmaceutical Sciences, Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Daniel Figeys
- School of Pharmaceutical Sciences, Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| | - Adriano Brandelli
- Laboratório de Bioquímica e Microbiologia Aplicada, Instituto de Ciência e Tecnologia de Alimentos, Universidade Federal do Rio Grande do Sul, 91510-970, Porto Alegre, Brazil.
| |
Collapse
|
24
|
Gan G, Zhang R, Zeng Y, Lu B, Luo Y, Chen S, Lei H, Cai Z, Huang X. Fecal microbiota transplantation validates the importance of gut microbiota in an ApoE -/- mouse model of chronic apical periodontitis-induced atherosclerosis. BMC Oral Health 2024; 24:1455. [PMID: 39614243 DOI: 10.1186/s12903-024-05230-5] [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: 05/16/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Chronic apical periodontitis (CAP) has been linked to the development of atherosclerosis, although the underlying mechanisms remain unclear. This study aimed to investigate the role of gut microbiota disruption in CAP-induced atherosclerosis development, focusing on trimethylamine N-oxide (TMAO)-related metabolites. METHODS The study utilized fecal microbiota transplantation (FMT) to transfer gut microbiota from mice with CAP to healthy mice. Atherosclerosis development was assessed by analyzing lesions in the aortic arch and aortic root. Serum lipid and inflammatory factor levels were measured. Composition and diversity of gut microbiota were analyzed using targeted metabolomics, with a focus on the ratio of Firmicutes to Bacteroidetes. The expression of hepatic flavin-containing monooxygenase 3 (FMO3) and serum TMAO levels were also evaluated. RESULTS Mice receiving gut microbiota from CAP mice showed increased atherosclerotic lesions compared to controls, without significant differences in serum lipid or inflammatory factor levels. Alterations in gut microbiota composition were observed, characterized by an increase in the Firmicutes to Bacteroidetes ratio. Peptostreptococcaceae abundance positively correlated with atherosclerosis severity, while Odoribacteraceae showed a negative correlation. No significant differences were found in hepatic FMO3 expression or serum TMAO levels. CONCLUSIONS The study confirms the role of gut microbiota disruption in CAP-mediated atherosclerosis development, independent of serum lipid or TMAO levels. Alterations in gut microbiota composition, particularly increased Firmicutes to Bacteroidetes ratio and specific bacterial families, were associated with atherosclerosis severity. These findings highlight the intricate interplay between gut microbiota and cardiovascular health in the context of CAP.
Collapse
Affiliation(s)
- Guowu Gan
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Ren Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yu Zeng
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Beibei Lu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yufang Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shuai Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Huaxiang Lei
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Zhiyu Cai
- Department of Stomatology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
25
|
Li D, Li M, Gao H, Hu K, Xie R, Fan J, Huang M, Liao C, Han C, Guo Z, Chen X, Li M. Integrative multiomics analysis reveals association of gut microbiota and its metabolites with susceptibility to keloids. Front Microbiol 2024; 15:1475984. [PMID: 39669776 PMCID: PMC11636970 DOI: 10.3389/fmicb.2024.1475984] [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: 08/06/2024] [Accepted: 11/04/2024] [Indexed: 12/14/2024] Open
Abstract
Keloid scarring is a fibroproliferative disease of the skin, which can significantly impact one's quality of life through cosmetic concerns, physical discomfort (itchy; painful), restricted movement, and psychological distress. Owing to the poorly understood pathogenesis of keloids and their high recurrence rate, the efficacy of keloid treatment remains unsatisfactory, particularly in patients susceptible to multiple keloids. We conducted fecal metagenomic analyzes and both untargeted and targeted plasma metabolomics in patients with multiple keloids (MK, n = 56) and controls with normal scars (NS, n = 60); tissue-untargeted metabolomics (MK, n = 35; NS, n = 32), tissue-targeted metabolomics (MK, n = 41; NS, n = 36), and single-cell sequencing analyzes (GSE163973). Differences in the gut microbiota composition, plasma metabolites, and tissue metabolites were observed between the MK and NS groups; the core gut microbiota, Oxalobacter formigenes, Bacteroides plebeius, and Parabacteroides distasonis, were identified via the gut microbiome co-occurrence network. Single-cell data helped clarify the specific cells affected by plasma metabolites. An area under the curve analysis using a random forest model based on fecal metagenomics, plasma metabolomics, and tissue metabolomics revealed that gut bacteria, plasma, and tissue metabolites were effective in distinguishing between MK and NS groups. Decreased Bacteroides plebeius could lower uracil levels, altering systemic lipid metabolism, which may change the metabolic phenotype of secretory reticular fibroblasts in wounds, potentially leading to MK. These findings may open new avenues for understanding the multifactorial nature of keloid formation from the gut-skin axis and highlight the potential for novel therapeutic strategies targeting keloid lesions and the underlying systemic imbalances affected by the gut microbiome.
Collapse
Affiliation(s)
- Dang Li
- Nursing Department of Fujian Medical University Union Hospital, Fuzhou, China
| | - Minghao Li
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Hangqi Gao
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Kailun Hu
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Rongrong Xie
- Department of Plastic Surgery, The Second Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jing Fan
- Department of Gynecology, Fuzhou Children’s Hospital of Fujian Medical University, Fuzhou, China
| | - Mingquan Huang
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Chengxin Liao
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Chang Han
- Shanghai Majorbio Bio-Pharm Technology Co., Ltd., Shanghai, China
| | - Zhihui Guo
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Xiaosong Chen
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Ming Li
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| |
Collapse
|
26
|
Yang X, He M, Cao J, Tang Q, Yang B, Li T, Sun M. Acupuncture and Moxibustion for Inflammatory Bowel Disease: Regulatory Mechanisms Revealed by Microbiome and Metabolomic Analysis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1891-1923. [PMID: 39581856 DOI: 10.1142/s0192415x24500745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Acupuncture and moxibustion are widely acknowledged as effective complementary therapies for managing inflammatory bowel disease (IBD) in traditional Chinese medicine. However, the regulatory mechanisms by which these two therapies exert their therapeutic effects in IBD are yet to be fully elucidated. The objective of this study was to investigate the mechanisms of action underlying acupuncture and moxibustion and the regulative differences between them as therapeutic interventions for IBD. Using a dextran sodium sulfate-induced IBD mice model, the effects of the two treatments were evaluated by examination of body weight, stool samples, colon morphology, inflammatory factors, gut microbiota, and metabolites. The results indicated that both acupuncture and moxibustion mitigated body weight reduction; improved the structural characteristics of intestinal tissues; increased levels of anti-inflammatory cytokines including interleukin (IL)-10; and decreased levels of pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-[Formula: see text]), nuclear factor kappa B (NF-[Formula: see text]B), IL-6, IL-1[Formula: see text], and IL-17. Acupuncture and moxibustion had distinct effects on the regulation of the intestinal microbiota and metabolic pathways in IBD mice. Moxibustion regulated a greater number of metabolic pathways than acupuncture, the majority of which were associated with amino acid metabolism, brain signal transmission, energy metabolism, and anti-inflammatory pathways. These findings provide a scientific basis for the differential applications of acupuncture and moxibustion in clinical practice.
Collapse
Affiliation(s)
- Xinyue Yang
- School of Medicine, Lishui University, Lishui 323000, Zhejiang Province, P. R. China
- Changchun University of Chinese Medicine, Changchun 130117, Jilin Province, P. R. China
| | - Min He
- Changchun University of Chinese Medicine, Changchun 130117, Jilin Province, P. R. China
| | - Jiazhen Cao
- Changchun University of Chinese Medicine, Changchun 130117, Jilin Province, P. R. China
| | - Qingqing Tang
- Changchun University of Chinese Medicine, Changchun 130117, Jilin Province, P. R. China
| | - Bo Yang
- Changchun University of Chinese Medicine, Changchun 130117, Jilin Province, P. R. China
| | - Tie Li
- Changchun University of Chinese Medicine, Changchun 130117, Jilin Province, P. R. China
| | - Mengmeng Sun
- Changchun University of Chinese Medicine, Changchun 130117, Jilin Province, P. R. China
| |
Collapse
|
27
|
Horvath A, Habisch H, Prietl B, Pfeifer V, Balazs I, Kovacs G, Foris V, John N, Kleinschek D, Feldbacher N, Grønbæk H, Møller HJ, Žukauskaitė K, Madl T, Stadlbauer V. Alteration of the Gut-Lung Axis After Severe COVID-19 Infection and Modulation Through Probiotics: A Randomized, Controlled Pilot Study. Nutrients 2024; 16:3840. [PMID: 39599626 PMCID: PMC11597208 DOI: 10.3390/nu16223840] [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: 10/07/2024] [Revised: 10/30/2024] [Accepted: 11/02/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The gut-lung axis could be a potential therapeutic target for improving post-acute COVID-19 symptoms, and probiotics have been proposed as possible modulators. AIM We conducted a pilot study to understand alterations in the gut-lung axis and to explore the effects of a probiotic in post-acute COVID-19 disease. METHODS We included patients after severe COVID-19 disease (sCOV, n = 21) in a randomized, placebo-controlled trial to test the effect of a probiotic (Pro-Vi 5, Institute Allergosan, Graz, Austria) in a six-month intervention and used patients after mild disease (mCOV, n = 10) as controls, to compare the intestinal microbiome, metabolome, and patient-reported outcomes and biomarkers along the gut-lung axis at baseline and throughout probiotic intervention. RESULTS Compared to mCOV patients, sCOV patients showed lower microbial richness, which was significantly improved by probiotic intervention. A reorganization of Ruminococcaceae and Lachnospiraceae taxa was observed in sCOV patients but remained unaffected by the intervention. Serum metabolome showed a dysregulation of lipoproteins in accordance with higher BMI and comorbidities in sCOV patients. HDL and LDL fractions/components were temporarily decreased in the probiotic group. Stool metabolome was altered at baseline in sCOV patients and an increase in L-DOPA after 3 months and butyrate after 6 months of intervention could be observed. Probiotics partially improved reduced quality of life and modulated altered immune responses in sCOV patients. Increased intestinal permeability at baseline remained unaffected. CONCLUSION The study provides evidence of long-term alterations of the gut-lung axis after severe COVID-19 infection and suggests that probiotics can modulate the biomarkers of the gut-lung axis.
Collapse
Affiliation(s)
- Angela Horvath
- Center for Biomarker Research in Medicine (CBmed), Division of Translational Precision Medicine, Division of Precision Medicine Technologies, 8010 Graz, Austria; (A.H.); (B.P.); (V.P.); (I.B.); (N.F.)
- Division for Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria;
| | - Hansjörg Habisch
- Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, 8010 Graz, Austria; (H.H.); (T.M.)
- BioTechMed-Graz, 8010 Graz, Austria
| | - Barbara Prietl
- Center for Biomarker Research in Medicine (CBmed), Division of Translational Precision Medicine, Division of Precision Medicine Technologies, 8010 Graz, Austria; (A.H.); (B.P.); (V.P.); (I.B.); (N.F.)
- Division of Endocrinology and Diabetes, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Verena Pfeifer
- Center for Biomarker Research in Medicine (CBmed), Division of Translational Precision Medicine, Division of Precision Medicine Technologies, 8010 Graz, Austria; (A.H.); (B.P.); (V.P.); (I.B.); (N.F.)
- Division of Endocrinology and Diabetes, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Irina Balazs
- Center for Biomarker Research in Medicine (CBmed), Division of Translational Precision Medicine, Division of Precision Medicine Technologies, 8010 Graz, Austria; (A.H.); (B.P.); (V.P.); (I.B.); (N.F.)
- Division for Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria;
| | - Gabor Kovacs
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria; (G.K.); (V.F.); (N.J.)
| | - Vasile Foris
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria; (G.K.); (V.F.); (N.J.)
| | - Nikolaus John
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria; (G.K.); (V.F.); (N.J.)
| | - Daniela Kleinschek
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
| | - Nicole Feldbacher
- Center for Biomarker Research in Medicine (CBmed), Division of Translational Precision Medicine, Division of Precision Medicine Technologies, 8010 Graz, Austria; (A.H.); (B.P.); (V.P.); (I.B.); (N.F.)
- Division for Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria;
| | - Henning Grønbæk
- Departments of Hepatology and Gastroenterology, Aarhus University Hospital, 8200 Aarhus, Denmark;
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark;
| | - Holger Jon Møller
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark;
- Department of Clinical Biochemistry, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Kristina Žukauskaitė
- Division for Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria;
- Institute of Biosciences, Life Sciences Center, Vilnius University, 01513 Vilnius, Lithuania
| | - Tobias Madl
- Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, 8010 Graz, Austria; (H.H.); (T.M.)
- BioTechMed-Graz, 8010 Graz, Austria
| | - Vanessa Stadlbauer
- Center for Biomarker Research in Medicine (CBmed), Division of Translational Precision Medicine, Division of Precision Medicine Technologies, 8010 Graz, Austria; (A.H.); (B.P.); (V.P.); (I.B.); (N.F.)
- Division for Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria;
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
28
|
Ji X, Yu H, Wang L, Bao X, Si T, Li X, Wang H, Borjigidai A, Kusuma Aji G, Bai L, Fu M. Gut microbiota and metabolomics unveil the mechanisms of Lomatogonium rotatum in ameliorating visceral fat and serum lipids in high-fat diet-induced obese mice. Front Pharmacol 2024; 15:1418063. [PMID: 39559734 PMCID: PMC11570273 DOI: 10.3389/fphar.2024.1418063] [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: 04/16/2024] [Accepted: 10/21/2024] [Indexed: 11/20/2024] Open
Abstract
Lomatogonium rotatum (LR) is a folk medicinal herb traditionally used as a lipid-lowering and anti-obesity agent; but its pharmacological mechanism is unclear. In this study, we assessed the alterations of LR on gut microbes and serum metabolites in obese mice and their associated mechanisms of modulation on visceral fat and serum lipid by integrating gut microbiota and metabolomics analyses. Mice were fed a high-fat diet (HFD) to generate obesity and were then given LR and Orlistat orally at different doses (0.18, 0.9, 1.8 g/kg for LR and 0.048 g/kg for Orlistat) for a duration of 9 weeks. The impact of LR on weight loss was assessed through the examination of fat deposition, serum lipid indices, liver indices, and HE pathohistology. The effects of LR on gut microbiota and serum metabolites in obese mice were then investigated by 16S rRNA sequencing technology and untargeted metabolomics, and correlation analysis was performed. LR significantly reduced body weight, feed intake, Lee's index, visceral fat accumulation, serum TG, TC, AST and ALT, and elevated serum HDL levels in obese mice. In addition, 16S rRNA sequencing results indicated that the LR intervention remodeled microbial diversity and composition, increased the relative abundance of gut microbes Bacteroidetes and Porphyromonadaceae in HFD-induced obese mice, and decreased the Deferribacteres, Firmicutes and the Firmicutes/Bacteroidetes ratio. Correlation analyses showed that LR regulation of L-tyrosine and hesperetin metabolism, as well as alterations in the metabolic pathways of Phenylalanine, tyrosine and tryptophan biosynthesis, were associated with the changes in abundance of Bacteroidetes, Firmicutes, Porphyromonadaceae and Deferribacteres. Our study demonstrated that LR has lipid lowering and visceral fat reduction effects and its function may be closely related to the improvement of the gut microbiota and its associated metabolites.
Collapse
Affiliation(s)
- Xiaoping Ji
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
- College of Traditional Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Hongzhen Yu
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Lianqian Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Xuemei Bao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, China
| | - Tegele Si
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, China
| | - Xiaoman Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Hugejiletu Wang
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, China
| | - Almaz Borjigidai
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Galih Kusuma Aji
- Research Center for Agroindustry, National Research and Innovation Agency, Jakarta Pusat, Indonesia
| | - Laxinamujila Bai
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, China
| | - Minghai Fu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, China
| |
Collapse
|
29
|
Matsuki T, Nakamura S, Nishiyama M, Narimatsu H. Holistic Evaluation of the Gut Microbiota through Data Envelopment Analysis: A Cross-Sectional Study. Curr Dev Nutr 2024; 8:104469. [PMID: 39524216 PMCID: PMC11550754 DOI: 10.1016/j.cdnut.2024.104469] [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: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024] Open
Abstract
Background The gut microbiome plays a crucial role in human health, but maintaining a healthy gut microbiome remains challenging. Current approaches often focus on individual components rather than providing a holistic assessment. Objectives To introduce and evaluate a novel approach using data envelopment analysis (DEA) for assessing gut microbiota efficiency and identifying potential targets for personalized interventions. Methods We conducted a cross-sectional analysis of 577 participants from the Kanagawa "ME-BYO" Prospective Cohort Study. Lifestyle factors and gut microbiota composition were assessed. DEA was employed to calculate an efficiency score for each participant, incorporating multiple inputs (lifestyle factors) and outputs (gut microbiotas). This score represents how efficiently an individual's lifestyle factors contribute to their gut microbiota composition. Tobit regression analysis was used to assess associations between efficiency scores and demographic and health-related factors. Results The mean efficiency score was 0.86, with 14.2% of participants classified as efficient. Efficiency scores showed positive correlations with alcohol intake and Faith's phylogenetic diversity. Tobit regression analysis revealed significant associations between efficiency scores and sex, fat intake, and yogurt consumption. DEA identified specific targets for improving gut microbiota composition in inefficient individuals. Conclusions This study demonstrates the potential of DEA as a tool for evaluating gut microbiota efficiency and providing personalized recommendations for microbiota optimization. This approach could lead to more effective strategies for optimizing gut health across diverse populations.
Collapse
Affiliation(s)
- Taizo Matsuki
- Graduate School of Health Innovation, Kanagawa University of Human Services, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Kanagawa, Japan
| | - Sho Nakamura
- Graduate School of Health Innovation, Kanagawa University of Human Services, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Kanagawa, Japan
- Department of Medical Genetics, Kanagawa Cancer Center, Nakao, Asahi-ku, Yokohama, Kanagawa, Japan
| | - Minami Nishiyama
- Graduate School of Health Innovation, Kanagawa University of Human Services, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Kanagawa, Japan
| | - Hiroto Narimatsu
- Graduate School of Health Innovation, Kanagawa University of Human Services, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Kanagawa, Japan
- Department of Medical Genetics, Kanagawa Cancer Center, Nakao, Asahi-ku, Yokohama, Kanagawa, Japan
| |
Collapse
|
30
|
Ovaska M, Tamminen M, Lahdenperä M, Vahtera J, Rautava S, Gonzales-Inca C, Heiskanen MA, Lagström H. The role of early life factors and green living environment in the development of gut microbiota in infancy: Population-based cohort study. ENVIRONMENT INTERNATIONAL 2024; 193:109093. [PMID: 39490300 DOI: 10.1016/j.envint.2024.109093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/26/2024] [Accepted: 10/19/2024] [Indexed: 11/05/2024]
Abstract
OBJECTIVE Early life microbial exposure influences the composition of gut microbiota. We investigated how early life factors, and the green living environment around infants' homes, influence the development of gut microbiota during infancy by utilizing data from the Steps to Healthy Development follow-up study (the STEPS study). METHODS The gut microbiota was analyzed at early (∼3 months, n = 959), and late infancy (∼13 months, n = 984) using 16S rRNA amplicon sequencing, and combined with residential green environment, measured as (1) Normalized Difference Vegetation Index, (2) Vegetation Cover Diversity, and (3) Naturalness Index within a 750 m radius. We compared gut microbiota diversity and composition between early and late infancy, identified significant individual and family level early life factors influencing gut microbiota, and determined the role of the residential green environment measures on gut microbiota development. RESULTS Alpha diversity (t-test, p < 0.001) and beta diversity (PERMANOVA, R2 = 0.095, p < 0.001) differed between early and late infancy. Birth mode was the strongest contributor to the gut microbiota community composition in early infancy (PERMANOVA, R2 = 0.005, p < 0.01) and the presence of siblings in late infancy (PERMANOVA, R2 = 0.007, p < 0.01). Residential green environment showed no association with community composition, whereas time spend outdoors did (PERMANOVA, R2 = 0.002, p < 0.05). Measures of greenness displayed a statistically significant association with alpha diversity during early infancy, not during late infancy (glm, p < 0.05). In adjusted analysis, the associations remained only with the Naturalness Index, where higher human impact on living environment was associated with decreased species richness (glm, Observed richness, p < 0.05). CONCLUSIONS The role of the residential green environment to the infant gut microbiota is especially important in early infancy, however, other early life factors, such as birth mode and presence of sibling, had a more significant effect on the overall community composition.
Collapse
Affiliation(s)
- Minka Ovaska
- Department of Public Health, University of Turku and Turku University Hospital, Turku, Finland; Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland.
| | - Manu Tamminen
- Department of Biology, University of Turku, Turku, Finland
| | | | - Jussi Vahtera
- Department of Public Health, University of Turku and Turku University Hospital, Turku, Finland; Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Samuli Rautava
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland; Department of Pediatrics, University of Helsinki and New Children's Hospital, Helsinki University Hospital, Helsinki, Finland
| | | | - Marja A Heiskanen
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland; Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Hanna Lagström
- Department of Public Health, University of Turku and Turku University Hospital, Turku, Finland; Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland; Nutrition and Food Research Center, Faculty of Medicine, University of Turku, Turku, Finland
| |
Collapse
|
31
|
Huang Q, Xing J, Tang F, Ren J, Wang C, Xue F. Recombinant Lactiplantibacilllus plantarum modulate gut microbial diversity and function. BMC Microbiol 2024; 24:423. [PMID: 39438791 PMCID: PMC11494753 DOI: 10.1186/s12866-024-03570-4] [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: 10/17/2023] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Gut microbes are important regulators of host health and can also function as disease indicators. Lactiplantibacilllus plantarum(L. plantarum)used as express and delivery vaccines for mucosal immunity have been shown to activate specific immune responses in numerous studies. RESULTS The interaction between recombinant L. plantarum and the gut microbiota was investigated in this study. The results indicated a change in the amount of gut OTU by recombinant L. plantarum. Recombinant L. plantarum dramatically boosted the species diversity of gut bacteria based on the Shannon-Wiener index. Beta diversity analysis showed that microbial structure was changed by recombinant L. plantarum. Furthermore, recombinant NC8 L. plantarum expressing a fusion between the P14.5 protein of the African swine fever virus and IL-33 enhanced the functions of gut bacteria in metabolism and immune regulation. Increased levels of IgG and IgG1 in serum and sIgA in feces, as well as enrichment of CD4+ T cells and IgA+ B cells, indicated that the gut microbiota exerted an immunomodulatory role when mediated by recombinant L. plantarum. CONCLUSIONS These results revealed that recombinant L. plantarum exerted its potential role in the gut microbiota and gut immunity.These fndings contribute to a broader understanding and utilization of L. plantarum bacteria in various therapeutic applications.
Collapse
Affiliation(s)
- Quntao Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Junhong Xing
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Fang Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jianluan Ren
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Chunfeng Wang
- Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China.
| | - Feng Xue
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
32
|
Chen W, Zou H, Xu H, Cao R, Zhang H, Zhang Y, Zhao J. The potential influence and intervention measures of gut microbiota on sperm: it is time to focus on testis-gut microbiota axis. Front Microbiol 2024; 15:1478082. [PMID: 39439945 PMCID: PMC11493703 DOI: 10.3389/fmicb.2024.1478082] [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: 08/09/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
As the global male infertility rate continues to rise, there is an urgent imperative to investigate the underlying causes of sustained deterioration in sperm quality. The gut microbiota emerges as a pivotal factor in host health regulation, with mounting evidence highlighting its dual influence on semen. This review underscores the interplay between the Testis-Gut microbiota axis and its consequential effects on sperm. Potential mechanisms driving the dual impact of gut microbiota on sperm encompass immune modulation, inflammatory responses mediated by endotoxins, oxidative stress, antioxidant defenses, gut microbiota-derived metabolites, epigenetic modifications, regulatory sex hormone signaling. Interventions such as probiotics, prebiotics, synbiotics, fecal microbiota transplantation, and Traditional natural herbal extracts are hypothesized to rectify dysbiosis, offering avenues to modulate gut microbiota and enhance Spermatogenesis and motility. Future investigations should delve into elucidating the mechanisms and foundational principles governing the interaction between gut microbiota and sperm within the Testis-Gut microbiota Axis. Understanding and modulating the Testis-Gut microbiota Axis may yield novel therapeutic strategies to enhance male fertility and combat the global decline in sperm quality.
Collapse
Affiliation(s)
- Wenkang Chen
- Graduate School of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hede Zou
- Graduate School of China Academy of Chinese Medical Sciences, Beijing, China
| | - Haoran Xu
- Graduate School of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Rui Cao
- Graduate School of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hekun Zhang
- Graduate School of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yapeng Zhang
- Graduate School of China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiayou Zhao
- Graduate School of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
33
|
Soe Thu M, Sawaswong V, Chanchaem P, Klomkliew P, Campbell BJ, Hirankarn N, Fothergill JL, Payungporn S. Optimization of a DNA extraction protocol for improving bacterial and fungal classification based on Nanopore sequencing. Access Microbiol 2024; 6:000754.v3. [PMID: 39376590 PMCID: PMC11457918 DOI: 10.1099/acmi.0.000754.v3] [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: 11/21/2023] [Accepted: 06/03/2024] [Indexed: 10/09/2024] Open
Abstract
Ribosomal RNA gene amplicon sequencing is commonly used to evaluate microbiome profiles in health and disease and document the impact of interventional treatments. Nanopore sequencing is attractive since it can provide greater classification at the species level. However, optimized protocols to target marker genes for bacterial and fungal profiling are needed. To achieve an increased taxonomic resolution, we developed extraction and full-length amplicon PCR-based approaches using Nanopore sequencing. Three lysis conditions were applied to a mock microbial community, including known bacterial and fungal species: ZymoBIOMICS lysis buffer (ML) alone, incorporating bead-beating (MLB) or bead-beating plus MetaPolyzyme enzymatic treatment (MLBE). In profiling of bacteria in comparison to reference data, MLB had more statistically different bacterial phyla and genera than the other two conditions. In fungal profiling, MLB had a significant increase of Ascomycota and a decline of Basidiomycota, subsequently failing to detect Malassezia and Cryptococcus. Also, a principal coordinates analysis plot by the Bray-Curtis metric showed a significant difference among groups for bacterial (P=0.033) and fungal (P=0.012) profiles, highlighting the importance of understanding the biases present in pretreatment. Overall, microbial profiling and diversity analysis revealed that ML and MLBE are more similar than MLB for both bacteria and fungi; therefore, using this specific pipeline, bead-beating is not recommended for whole gene amplicon sequencing. However, ML alone was suggested as an optimal approach considering DNA yield, taxonomic classification, reagent cost and hands-on time. This could be an initial proof-of-concept study for simultaneous human bacterial and fungal microbiome studies.
Collapse
Affiliation(s)
- May Soe Thu
- Joint Chulalongkorn University–University of Liverpool Doctoral Program in Biomedical Sciences and Biotechnology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 3GE, UK
- Center of Excellence in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Vorthon Sawaswong
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | - Prangwalai Chanchaem
- Center of Excellence in Systems Microbiology, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pavit Klomkliew
- Center of Excellence in Systems Microbiology, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Barry J. Campbell
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 3GE, UK
| | - Nattiya Hirankarn
- Center of Excellence in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Joanne L. Fothergill
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, L69 3GE, UK
| | - Sunchai Payungporn
- Center of Excellence in Systems Microbiology, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
34
|
Toh KY, Toh TS, Chua KP, Rajakumar P, Lee JWJ, Chong CW. Identification of age-associated microbial changes via long-read 16S sequencing. Gut Pathog 2024; 16:56. [PMID: 39369250 PMCID: PMC11456230 DOI: 10.1186/s13099-024-00650-8] [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: 03/05/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Age-related gut microbial changes have been widely investigated over the past decade. Most of the previous age-related microbiome studies were conducted on the Western population, and the short-read sequencing (e.g., 16S V4 or V3-V4 region) was the most common microbiota profiling method. We evaluated the gut compositional differences using the long-read sequencing approach (i.e., PacBio sequencing targeting the full-length V1-V9 regions) to enable a deeper taxonomic resolution and better characterize the gut microbiome of Singaporeans from different age groups. RESULTS A total of 83 research participants were included in this study. Although no significant differences were detected in alpha and beta diversity, our study demonstrated several bacterial taxa with abundances that were significantly different across age groups. With young individuals as the reference group, Eggerthella lenta and Bacteroides uniformis were found to be significantly altered in the middle-aged group, while Catenibacterium mitsuokai and Bacteroides plebeius were significantly altered in the elderly group. These age-related differences in the gut microbiome were associated with aberrations in several predicted functional pathways, including dysregulations of pathways related to lipopolysaccharide and tricarboxylic acid cycle in older adults. CONCLUSIONS The utilization of long-read sequencing facilitated the identification of species- and strain-level differences across age groups, which was challenging with the partial 16S rRNA sequencing approach. Nevertheless, replication studies are warranted to confirm our findings, and if confirmed, further in vitro and in vivo studies are crucial to better understand the impact of the altered levels of age-related bacterial taxa. Additionally, the modest performance of strain-level taxonomic classification using 16S-ITS-23S gene sequences, likely due to the limited depth of currently available alignment databases, highlights the need for optimization and refinement in curating these databases for the long-read sequencing approach.
Collapse
Affiliation(s)
- Kai Yee Toh
- AMILI Pte Ltd, 89 Science Park Drive #03-09, The Rutherford, Lobby C, Singapore Science Park 1, Singapore, 118261, Singapore.
| | - Tzi Shin Toh
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Khi Pin Chua
- Pacific Biosciences of California, Menlo Park, CA, USA
| | - Priscilla Rajakumar
- AMILI Pte Ltd, 89 Science Park Drive #03-09, The Rutherford, Lobby C, Singapore Science Park 1, Singapore, 118261, Singapore
| | - Jonathan Wei Jie Lee
- AMILI Pte Ltd, 89 Science Park Drive #03-09, The Rutherford, Lobby C, Singapore Science Park 1, Singapore, 118261, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Gastroenterology and Hepatology, Department of Medicine , National University Health System, Singapore, 119228, Singapore
- iHealthtech, National University of Singapore, Singapore, 117599, Singapore
- SynCTI, National University of Singapore, Singapore, 117456, Singapore
| | - Chun Wie Chong
- School of Pharmacy, Monash University Malaysia, Selangor, Malaysia
- MUM Microbiome Research Centre, Monash University Malaysia, Selangor, Malaysia
| |
Collapse
|
35
|
Carlino N, Blanco-Míguez A, Punčochář M, Mengoni C, Pinto F, Tatti A, Manghi P, Armanini F, Avagliano M, Barcenilla C, Breselge S, Cabrera-Rubio R, Calvete-Torre I, Coakley M, Cobo-Díaz JF, De Filippis F, Dey H, Leech J, Klaassens ES, Knobloch S, O'Neil D, Quijada NM, Sabater C, Skírnisdóttir S, Valentino V, Walsh L, Alvarez-Ordóñez A, Asnicar F, Fackelmann G, Heidrich V, Margolles A, Marteinsson VT, Rota Stabelli O, Wagner M, Ercolini D, Cotter PD, Segata N, Pasolli E. Unexplored microbial diversity from 2,500 food metagenomes and links with the human microbiome. Cell 2024; 187:5775-5795.e15. [PMID: 39214080 DOI: 10.1016/j.cell.2024.07.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/17/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Complex microbiomes are part of the food we eat and influence our own microbiome, but their diversity remains largely unexplored. Here, we generated the open access curatedFoodMetagenomicData (cFMD) resource by integrating 1,950 newly sequenced and 583 public food metagenomes. We produced 10,899 metagenome-assembled genomes spanning 1,036 prokaryotic and 108 eukaryotic species-level genome bins (SGBs), including 320 previously undescribed taxa. Food SGBs displayed significant microbial diversity within and between food categories. Extension to >20,000 human metagenomes revealed that food SGBs accounted on average for 3% of the adult gut microbiome. Strain-level analysis highlighted potential instances of food-to-gut transmission and intestinal colonization (e.g., Lacticaseibacillus paracasei) as well as SGBs with divergent genomic structures in food and humans (e.g., Streptococcus gallolyticus and Limosilactobabillus mucosae). The cFMD expands our knowledge on food microbiomes, their role in shaping the human microbiome, and supports future uses of metagenomics for food quality, safety, and authentication.
Collapse
Affiliation(s)
- Niccolò Carlino
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Aitor Blanco-Míguez
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Michal Punčochář
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Claudia Mengoni
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Federica Pinto
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Alessia Tatti
- Scuola Universitaria Superiore IUSS Pavia, Pavia, Italy; Centre for Agriculture Food Environment, University of Trento, Trento, Italy; Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'Adige, Italy
| | - Paolo Manghi
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Federica Armanini
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Michele Avagliano
- Department of Agricultural Sciences, Division of Microbiology, University of Naples Federico II, Portici, Italy
| | - Coral Barcenilla
- Department of Food Hygiene and Technology, Universidad de León, León, Spain
| | - Samuel Breselge
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Raul Cabrera-Rubio
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; Department of Biotechnology, Institute of Agrochemistry and Food Technology - National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Inés Calvete-Torre
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias - Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Spain; Microhealth Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Mairéad Coakley
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | - José F Cobo-Díaz
- Department of Food Hygiene and Technology, Universidad de León, León, Spain
| | - Francesca De Filippis
- Department of Agricultural Sciences, Division of Microbiology, University of Naples Federico II, Portici, Italy; Task Force on Microbiome Studies, University of Naples Federico II, Portici, Italy
| | - Hrituraj Dey
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - John Leech
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | | | | | | | - Narciso M Quijada
- Austrian Competence Centre for Feed and Food Quality, Safety, and Innovation, FFoQSI GmbH, Tulln an der Donau, Austria; Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria; Institute for Agribiotechnology Research (CIALE), Department of Microbiology and Genetics, University of Salamanca, Salamanca, Spain
| | - Carlos Sabater
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias - Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Spain; Microhealth Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | | | - Vincenzo Valentino
- Department of Agricultural Sciences, Division of Microbiology, University of Naples Federico II, Portici, Italy
| | - Liam Walsh
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland; School of Microbiology, University College Cork, Cork, Ireland
| | | | - Francesco Asnicar
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Gloria Fackelmann
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Vitor Heidrich
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Abelardo Margolles
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias - Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Spain; Microhealth Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Viggó Thór Marteinsson
- Microbiology Research Group, Matís, Reykjavík, Iceland; University of Iceland, Faculty of Food Science and Nutrition, Reykjavík, Iceland
| | - Omar Rota Stabelli
- Centre for Agriculture Food Environment, University of Trento, Trento, Italy; Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'Adige, Italy
| | - Martin Wagner
- Austrian Competence Centre for Feed and Food Quality, Safety, and Innovation, FFoQSI GmbH, Tulln an der Donau, Austria; Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Danilo Ercolini
- Department of Agricultural Sciences, Division of Microbiology, University of Naples Federico II, Portici, Italy; Task Force on Microbiome Studies, University of Naples Federico II, Portici, Italy
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland; VistaMilk SFI Research Centre, Teagasc, Moorepark, Fermoy, Co. Cork, Ireland
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy; IEO, Istituto Europeo di Oncologia IRCSS, Milan, Italy; Department of Twins Research and Genetic Epidemiology, King's College London, London, UK.
| | - Edoardo Pasolli
- Department of Agricultural Sciences, Division of Microbiology, University of Naples Federico II, Portici, Italy; Task Force on Microbiome Studies, University of Naples Federico II, Portici, Italy
| |
Collapse
|
36
|
Talarico F, Tilocca B, Spagnuolo R, Abenavoli L, Luzza F, Roncada P. The effects of stress on gut virome: Implications on infectious disease and systemic disorders. Microbiologyopen 2024; 13:e1434. [PMID: 39311537 PMCID: PMC11418023 DOI: 10.1002/mbo3.1434] [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: 04/30/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 09/26/2024] Open
Abstract
The role of gut microbiota in health and disease is being thoroughly examined in various contexts, with a specific focus on the bacterial fraction due to its significant abundance. However, despite their lower abundance, viruses within the gut microbiota are gaining recognition for their crucial role in shaping the structure and function of the intestinal microbiota, with significant effects on the host as a whole, particularly the immune system. Similarly, environmental factors such as stress are key in modulating the host immune system, which in turn influences the composition of the gut virome and neurological functions through the bidirectional communication of the gut-brain axis. In this context, alterations in the host immune system due to stress and/or dysbiosis of the gut virome are critical factors in the development of both infectious and noninfectious diseases. The molecular mechanisms and correlation patterns between microbial species are not yet fully understood. This literature review seeks to explore the interconnected relationship between stress and the gut virome, with a focus on how this interaction is influenced by the host's immune system. We also discuss how disturbances in this finely balanced system can lead to the onset and/or progression of diseases.
Collapse
Affiliation(s)
| | - Bruno Tilocca
- Department of Health SciencesUniversity “Magna Graecia”CatanzaroItaly
| | - Rocco Spagnuolo
- Department of Health SciencesUniversity “Magna Graecia”CatanzaroItaly
| | | | - Francesco Luzza
- Department of Health SciencesUniversity “Magna Graecia”CatanzaroItaly
| | - Paola Roncada
- Department of Health SciencesUniversity “Magna Graecia”CatanzaroItaly
| |
Collapse
|
37
|
Ioannou M, Borkent J, Andreu-Sánchez S, Wu J, Fu J, Sommer IEC, Haarman BCM. Reproducible gut microbial signatures in bipolar and schizophrenia spectrum disorders: A metagenome-wide study. Brain Behav Immun 2024; 121:165-175. [PMID: 39032544 DOI: 10.1016/j.bbi.2024.07.009] [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: 01/12/2024] [Revised: 05/30/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Numerous studies report gut microbiome variations in bipolar disorder (BD) and schizophrenia spectrum disorders (SSD) compared to healthy individuals, though, there is limited consensus on which specific bacteria are associated with these disorders. METHODS In this study, we performed a comprehensive metagenomic shotgun sequencing analysis in 103 Dutch patients with BD/SSD and 128 healthy controls matched for age, sex, body mass index and income, while accounting for diet quality, transit time and technical confounders. To assess the replicability of the findings, we used two validation cohorts (total n = 203), including participants from a distinct population with a different metagenomic isolation protocol. RESULTS The gut microbiome of the patients had a significantly different β-diversity, but not α-diversity nor neuroactive potential compared to healthy controls. Initially, twenty-six bacterial taxa were identified as differentially abundant in patients. Among these, the previously reported genera Lachnoclostridium and Eggerthella were replicated in the validation cohorts. Employing the CoDaCoRe learning algorithm, we identified two bacterial balances specific to BD/SSD, which demonstrated an area under the receiver operating characteristic curve (AUC) of 0.77 in the test dataset. These balances were replicated in the validation cohorts and showed a positive association with the severity of psychiatric symptoms and antipsychotic use. Last, we showed a positive association between the relative abundance of Klebsiella and Klebsiella pneumoniae with antipsychotic use and between the Anaeromassilibacillus and lithium use. CONCLUSIONS Our findings suggest that microbial balances could be a reproducible method for identifying BD/SSD-specific microbial signatures, with potential diagnostic and prognostic applications. Notably, Lachnoclostridium and Eggerthella emerge as frequently occurring bacteria in BD/SSD. Last, our study reaffirms the previously established link between Klebsiella and antipsychotic medication use and identifies a novel association between Anaeromassilibacillus and lithium use.
Collapse
Affiliation(s)
- Magdalini Ioannou
- University of Groningen and University Medical Center Groningen, Department of Psychiatry, Groningen, the Netherlands; University of Groningen and University Medical Center Groningen, Department of Biomedical Sciences, Groningen, the Netherlands.
| | - Jenny Borkent
- University of Groningen and University Medical Center Groningen, Department of Biomedical Sciences, Groningen, the Netherlands
| | - Sergio Andreu-Sánchez
- University of Groningen and University Medical Center Groningen, Department of Genetics, Groningen, the Netherlands; University of Groningen and University Medical Center Groningen, Department of Pediatrics, Groningen, the Netherlands
| | - Jiafei Wu
- University of Groningen and University Medical Center Groningen, Department of Genetics, Groningen, the Netherlands
| | - Jingyuan Fu
- University of Groningen and University Medical Center Groningen, Department of Genetics, Groningen, the Netherlands; University of Groningen and University Medical Center Groningen, Department of Pediatrics, Groningen, the Netherlands
| | - Iris E C Sommer
- University of Groningen and University Medical Center Groningen, Department of Biomedical Sciences, Groningen, the Netherlands
| | - Bartholomeus C M Haarman
- University of Groningen and University Medical Center Groningen, Department of Psychiatry, Groningen, the Netherlands
| |
Collapse
|
38
|
Porreca A, Ibrahimi E, Maturo F, Marcos Zambrano LJ, Meto M, Lopes MB. Robust prediction of colorectal cancer via gut microbiome 16S rRNA sequencing data. J Med Microbiol 2024; 73. [PMID: 39377779 DOI: 10.1099/jmm.0.001903] [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: 10/09/2024] Open
Abstract
Introduction. The study addresses the challenge of utilizing human gut microbiome data for the early detection of colorectal cancer (CRC). The research emphasizes the potential of using machine learning techniques to analyze complex microbiome datasets, providing a non-invasive approach to identifying CRC-related microbial markers.Hypothesis/Gap Statement. The primary hypothesis is that a robust machine learning-based analysis of 16S rRNA microbiome data can identify specific microbial features that serve as effective biomarkers for CRC detection, overcoming the limitations of classical statistical models in high-dimensional settings.Aim. The primary objective of this study is to explore and validate the potential of the human microbiome, specifically in the colon, as a valuable source of biomarkers for colorectal cancer (CRC) detection and progression. The focus is on developing a classifier that effectively predicts the presence of CRC and normal samples based on the analysis of three previously published faecal 16S rRNA sequencing datasets.Methodology. To achieve the aim, various machine learning techniques are employed, including random forest (RF), recursive feature elimination (RFE) and a robust correlation-based technique known as the fuzzy forest (FF). The study utilizes these methods to analyse the three datasets, comparing their performance in predicting CRC and normal samples. The emphasis is on identifying the most relevant microbial features (taxa) associated with CRC development via partial dependence plots, i.e. a machine learning tool focused on explainability, visualizing how a feature influences the predicted outcome.Results. The analysis of the three faecal 16S rRNA sequencing datasets reveals the consistent and superior predictive performance of the FF compared to the RF and RFE. Notably, FF proves effective in addressing the correlation problem when assessing the importance of microbial taxa in explaining the development of CRC. The results highlight the potential of the human microbiome as a non-invasive means to detect CRC and underscore the significance of employing FF for improved predictive accuracy.Conclusion. In conclusion, this study underscores the limitations of classical statistical techniques in handling high-dimensional information such as human microbiome data. The research demonstrates the potential of the human microbiome, specifically in the colon, as a valuable source of biomarkers for CRC detection. Applying machine learning techniques, particularly the FF, is a promising approach for building a classifier to predict CRC and normal samples. The findings advocate for integrating FF to overcome the challenges associated with correlation when identifying crucial microbial features linked to CRC development.
Collapse
Affiliation(s)
- Annamaria Porreca
- Department of Economics, Statistics and Business, Faculty of Economics and Law, Universitas Mercatorum, Rome, Italy
| | - Eliana Ibrahimi
- Department of Biology, University of Tirana, Tirana, Albania
| | - Fabrizio Maturo
- Department of Economics, Statistics and Business, Faculty of Technological and Innovation Sciences, Universitas Mercatorum, Rome, Italy
| | - Laura Judith Marcos Zambrano
- Computational Biology Group, Precision Nutrition and Cancer Research Program, IMDEA Food Institute, Madrid, Spain
| | - Melisa Meto
- Department of Biology, University of Tirana, Tirana, Albania
| | - Marta B Lopes
- Center for Mathematics and Applications (NOVA Math), NOVA School of Science and Technology, Caparica, Portugal
- UNIDEMI, Research and Development Unit for Mechanical and Industrial Engineering, NOVA School of Science and Technology, Caparica, Portugal
| |
Collapse
|
39
|
Wirbel J, Essex M, Forslund SK, Zeller G. A realistic benchmark for differential abundance testing and confounder adjustment in human microbiome studies. Genome Biol 2024; 25:247. [PMID: 39322959 PMCID: PMC11423519 DOI: 10.1186/s13059-024-03390-9] [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: 12/30/2023] [Accepted: 09/06/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND In microbiome disease association studies, it is a fundamental task to test which microbes differ in their abundance between groups. Yet, consensus on suitable or optimal statistical methods for differential abundance testing is lacking, and it remains unexplored how these cope with confounding. Previous differential abundance benchmarks relying on simulated datasets did not quantitatively evaluate the similarity to real data, which undermines their recommendations. RESULTS Our simulation framework implants calibrated signals into real taxonomic profiles, including signals mimicking confounders. Using several whole meta-genome and 16S rRNA gene amplicon datasets, we validate that our simulated data resembles real data from disease association studies much more than in previous benchmarks. With extensively parametrized simulations, we benchmark the performance of nineteen differential abundance methods and further evaluate the best ones on confounded simulations. Only classic statistical methods (linear models, the Wilcoxon test, t-test), limma, and fastANCOM properly control false discoveries at relatively high sensitivity. When additionally considering confounders, these issues are exacerbated, but we find that adjusted differential abundance testing can effectively mitigate them. In a large cardiometabolic disease dataset, we showcase that failure to account for covariates such as medication causes spurious association in real-world applications. CONCLUSIONS Tight error control is critical for microbiome association studies. The unsatisfactory performance of many differential abundance methods and the persistent danger of unchecked confounding suggest these contribute to a lack of reproducibility among such studies. We have open-sourced our simulation and benchmarking software to foster a much-needed consolidation of statistical methodology for microbiome research.
Collapse
Affiliation(s)
- Jakob Wirbel
- Structural and Computational Biology Unit (SCB), European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Morgan Essex
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max-Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin (a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin), Berlin, Germany
| | - Sofia Kirke Forslund
- Structural and Computational Biology Unit (SCB), European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max-Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany.
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- Charité-Universitätsmedizin Berlin (a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin), Berlin, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany.
| | - Georg Zeller
- Structural and Computational Biology Unit (SCB), European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
- Center for Infectious Diseases (LUCID), Leiden University, Leiden University Medical Center (LUMC), Leiden, Netherlands.
- Center for Microbiome Analyses and Therapeutics (CMAT), Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
40
|
Maaskant A, Voermans B, Levin E, de Goffau MC, Plomp N, Schuren F, Remarque EJ, Smits A, Langermans JAM, Bakker J, Montijn R. Microbiome signature suggestive of lactose-intolerance in rhesus macaques (Macaca mulatta) with intermittent chronic diarrhea. Anim Microbiome 2024; 6:53. [PMID: 39313845 PMCID: PMC11421201 DOI: 10.1186/s42523-024-00338-z] [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: 04/02/2024] [Accepted: 09/06/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND Chronic diarrhea is a common cause of mortality and morbidity in captive rhesus macaques (Macaca mulatta). The exact etiology of chronic diarrhea in macaques remains unidentified. The occurrence of diarrhea is frequently linked to dysbiosis within the gut microbiome. Research into microbiome signatures correlated with diarrhea in macaques have predominantly been conducted with single sample collections. Our analysis was based on the metagenomic composition of longitudinally acquired fecal samples from rhesus macaques with chronic diarrhea and clinically healthy rhesus macaques that were obtained over the course of two years. We aimed to investigate potential relationships between the macaque gut microbiome, the presence of diarrhea and diet interventions with a selection of commercially available monkey diets. RESULTS The microbiome signature of macaques with intermittent chronic diarrhea showed a significant increase in lactate producing bacteria e.g. lactobacilli, and an increase in fermenters of lactate and succinate. Strikingly, two lactose free diets were associated with a lower incidence of diarrhea. CONCLUSION A lactose intolerance mechanism is suggested in these animals by the bloom of Lactobacillus in the presence of lactose resulting in an overproduction of intermediate fermentation products likely led to osmotically induced diarrhea. This study provides new insights into suspected microbiome-lactose intolerance relationship in rhesus macaques with intermittent chronic diarrhea. The integration of machine learning with metagenomic data analysis holds potential for developing targeted dietary interventions and therapeutic strategies and therefore ensuring a healthier and more resilient primate population.
Collapse
Affiliation(s)
- Annemiek Maaskant
- Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands.
- Department Population Health Sciences, Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, Heidelberglaan 8, 3584 CM, Utrecht, The Netherlands.
| | - Bas Voermans
- HORAIZON Technology BV, Marshallaan 2, 2625 GZ, Delft, The Netherlands.
- Department of Vascular Medicine, Amsterdam UMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Evgeni Levin
- HORAIZON Technology BV, Marshallaan 2, 2625 GZ, Delft, The Netherlands
| | - Marcus C de Goffau
- HORAIZON Technology BV, Marshallaan 2, 2625 GZ, Delft, The Netherlands
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Nicole Plomp
- Department of Microbiology and Systems Biology, Organization for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE, Leiden, The Netherlands
| | - Frank Schuren
- Department of Microbiology and Systems Biology, Organization for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE, Leiden, The Netherlands
| | - Edmond J Remarque
- Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands
| | - Antoine Smits
- Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands
| | - Jan A M Langermans
- Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands
- Department Population Health Sciences, Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, Heidelberglaan 8, 3584 CM, Utrecht, The Netherlands
| | - Jaco Bakker
- Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands
| | - Roy Montijn
- Department of Microbiology and Systems Biology, Organization for Applied Scientific Research (TNO), Sylviusweg 71, 2333 BE, Leiden, The Netherlands
| |
Collapse
|
41
|
Ma Z, Zuo T, Frey N, Rangrez AY. A systematic framework for understanding the microbiome in human health and disease: from basic principles to clinical translation. Signal Transduct Target Ther 2024; 9:237. [PMID: 39307902 PMCID: PMC11418828 DOI: 10.1038/s41392-024-01946-6] [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: 03/05/2024] [Revised: 07/03/2024] [Accepted: 08/01/2024] [Indexed: 09/26/2024] Open
Abstract
The human microbiome is a complex and dynamic system that plays important roles in human health and disease. However, there remain limitations and theoretical gaps in our current understanding of the intricate relationship between microbes and humans. In this narrative review, we integrate the knowledge and insights from various fields, including anatomy, physiology, immunology, histology, genetics, and evolution, to propose a systematic framework. It introduces key concepts such as the 'innate and adaptive genomes', which enhance genetic and evolutionary comprehension of the human genome. The 'germ-free syndrome' challenges the traditional 'microbes as pathogens' view, advocating for the necessity of microbes for health. The 'slave tissue' concept underscores the symbiotic intricacies between human tissues and their microbial counterparts, highlighting the dynamic health implications of microbial interactions. 'Acquired microbial immunity' positions the microbiome as an adjunct to human immune systems, providing a rationale for probiotic therapies and prudent antibiotic use. The 'homeostatic reprogramming hypothesis' integrates the microbiome into the internal environment theory, potentially explaining the change in homeostatic indicators post-industrialization. The 'cell-microbe co-ecology model' elucidates the symbiotic regulation affecting cellular balance, while the 'meta-host model' broadens the host definition to include symbiotic microbes. The 'health-illness conversion model' encapsulates the innate and adaptive genomes' interplay and dysbiosis patterns. The aim here is to provide a more focused and coherent understanding of microbiome and highlight future research avenues that could lead to a more effective and efficient healthcare system.
Collapse
Affiliation(s)
- Ziqi Ma
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Tao Zuo
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
42
|
Kurdi M, Bajwa SJS, Sharma R, Choudhary R. Gut Microbiota and Probiotics in Perioperative Management: A Narrative Review. Cureus 2024; 16:e68404. [PMID: 39360063 PMCID: PMC11445195 DOI: 10.7759/cureus.68404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2024] [Indexed: 10/04/2024] Open
Abstract
The human gut is the abode of several complex and diverse microbes. It is a fact that the human brain is interconnected with the spinal cord and sense organs; however, there is also a possibility of a connection between the brain and the gut microbiome. The human gut can be altered in various ways, the principal method being the intake of prebiotics, probiotics and synbiotics. Can this alteration in the gut microbiome be clinically utilised in the perioperative period? We conducted a literature search related to this topic using databases and search engines (Medical Literature Analysis and Retrieval System Online {MEDLINE}, Embase, Scopus, PubMed and Google Scholar). The search revealed some preclinical and clinical studies in animals and humans that demonstrate the alteration of the gut microbiome with the use of anxiolysis, probiotics/prebiotics and other perioperative factors including opioids, anaesthetics and perioperative stress. The significant effects of this alteration have been seen on preoperative anxiety and postoperative delirium/cognitive dysfunction/pain. These effects are described in this narrative review, which opens up newer vistas for high-quality research related to the gut microbiome, gut-brain axis, the related signaling pathways and their clinical application in the perioperative period.
Collapse
Affiliation(s)
- Madhuri Kurdi
- Department of Anaesthesiology, Karnataka Medical College and Research Institute, Hubballi, IND
| | - Sukhminder J S Bajwa
- Department of Anaesthesiology, Gian Sagar Medical College and Hospital, Patiala, IND
| | - Ridhima Sharma
- Department of Anaesthesiology, All India Institute of Medical Sciences, Nagpur, IND
| | - Ripon Choudhary
- Department of Anaesthesiology, Datta Meghe Medical College and Research Institute, Nagpur, IND
| |
Collapse
|
43
|
Hong J, Fu Y, Chen X, Zhang Y, Li X, Li T, Liu Y, Fan M, Lin R. Gut microbiome changes associated with chronic pancreatitis and pancreatic cancer: a systematic review and meta-analysis. Int J Surg 2024; 110:5781-5794. [PMID: 38847785 PMCID: PMC11392207 DOI: 10.1097/js9.0000000000001724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/19/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND The study of changes in the microbiome in chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC) holds significant potential for developing noninvasive diagnostic tools as well as innovative interventions to alter the progression of diseases. This systematic review and meta-analysis aimed to analyze in detail the taxonomic and functional characteristics of the gut microbiome in patients with CP and PDAC. METHODS Two researchers conducted a systematic search across public databases to gather all published research up to June 2023. Diversity and gut microbiota composition are the main outcomes the authors focus on. RESULTS This meta-analysis included 14 studies, involving a total of 1511 individuals in the PDAC ( n =285), CP ( n =342), and control ( n =649) groups. Our results show a significant difference in the composition of gut microbiota between PDAC/CP patients compared to healthy controls (HC), as evidenced by a slight decrease in α-diversity, including Shannon (SMD=-0.33; P =0.002 and SMD=-0.59; P <0.001, respectively) and a statistically significant β-diversity ( P <0.05). The pooled results showed that at the phylum level, the proportion of Firmicutes was lower in PDAC and CP patients than in HC patients. At the genus level, more than two studies demonstrated that four genera were significantly increased in PDAC patients compared to HC (e.g. Escherichia-Shigella and Veillonella ). CP patients had an increase in four genera (e.g. Escherichia-Shigella and Klebsiella ) and a decrease in eight genera (e.g. Coprococcus and Bifidobacterium ) compared to HC. Functional/metabolomics results from various studies also showed differences between PDAC/CP patients and HC. In addition, this study found no significant differences in gut microbiota between PDAC and CP patients. CONCLUSIONS Current evidence suggests changes in gut microbiota is associated with PDAC/CP, commonly reflected by a reduction in beneficial species and an increase in the pathogenic species. Further studies are needed to confirm these findings and explore therapeutic possibilities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mengke Fan
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
44
|
Li T, Yin D, Shi R. Gut-muscle axis mechanism of exercise prevention of sarcopenia. Front Nutr 2024; 11:1418778. [PMID: 39221163 PMCID: PMC11362084 DOI: 10.3389/fnut.2024.1418778] [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: 04/17/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Sarcopenia refers to an age-related systemic skeletal muscle disorder, which is characterized by loss of muscle mass and weakening of muscle strength. Gut microbiota can affect skeletal muscle through a variety of mechanisms. Gut microbiota present distinct features among elderly people and sarcopenia patients, including a decrease in microbial diversity, which might be associated with the quality and function of the skeletal muscle. There might be a gut-muscle axis; where gut microbiota and skeletal muscle may affect each other bi-directionally. Skeletal muscle can affect the biodiversity of the gut microbiota, and the latter can, in turn, affect the anabolism of skeletal muscle. This review examines recent studies exploring the relationship between gut microbiota and skeletal muscle, summarizes the effects of exercise on gut microbiota, and discusses the possible mechanisms of the gut-muscle axis.
Collapse
Affiliation(s)
| | | | - Rengfei Shi
- School of Health and Exercise, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
45
|
Jiang J, Czuchry D, Ru Y, Peng H, Shen J, Wang T, Zhao W, Chen W, Sui SF, Li Y, Li N. Activity-based metaproteomics driven discovery and enzymological characterization of potential α-galactosidases in the mouse gut microbiome. Commun Chem 2024; 7:184. [PMID: 39152233 PMCID: PMC11329505 DOI: 10.1038/s42004-024-01273-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024] Open
Abstract
The gut microbiota offers an extensive resource of enzymes, but many remain uncharacterized. To distinguish the activities of similar annotated proteins and mine the potentially applicable ones in the microbiome, we applied an effective Activity-Based Metaproteomics (ABMP) strategy using a specific activity-based probe (ABP) to screen the entire gut microbiome for directly discovering active enzymes and their potential applications, not for exploring host-microbiome interactions. By using an activity-based cyclophellitol aziridine probe specific to α-galactosidases (AGAL), we successfully identified and characterized several gut microbiota enzymes possessing AGAL activities. Cryo-electron microscopy analysis of a newly characterized enzyme (AGLA5) revealed the covalent binding conformations between the AGAL5 active site and the cyclophellitol aziridine ABP, which could provide insights into the enzyme's catalytic mechanism. The four newly characterized AGALs have diverse potential activities, including raffinose family oligosaccharides (RFOs) hydrolysis and enzymatic blood group transformation. Collectively, we present a ABMP platform that facilitates gut microbiota AGALs discovery, biochemical activity annotations and potential industrial or biopharmaceutical applications.
Collapse
Affiliation(s)
- Jianbing Jiang
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Diana Czuchry
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yanxia Ru
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Huipai Peng
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Junfeng Shen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Teng Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Wenjuan Zhao
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Weihua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Sen-Fang Sui
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Cryo-EM Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yaowang Li
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
| | - Nan Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Shenzhen Key Laboratory of Genome Manipulation and Biosynthesis, Shenzhen, China.
| |
Collapse
|
46
|
Zhang CY, Jiang SJ, Cao JJ, Xu Y, Wang XY, Li R, Miao ZW. Investigating the causal relationship between gut microbiota and gastroenteropancreatic neuroendocrine neoplasms: a bidirectional Mendelian randomization study. Front Microbiol 2024; 15:1420167. [PMID: 39193433 PMCID: PMC11347282 DOI: 10.3389/fmicb.2024.1420167] [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: 05/03/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024] Open
Abstract
Background The interaction between the intestinal flora and gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) remains poorly understood, despite the known effect of the gut microbiota on gastrointestinal adenocarcinomas. Hence, the present research aimed to determine the potential causal correlation between the intestinal flora and GEP-NENs by conducting a bidirectional Mendelian randomization (MR) analysis. Methods Two-sample MR analysis was conducted using the summary statistics of the gut microbiota from the MiBioGen consortium and those of GEP-NENs from the FinnGen research project. The inverse-variance weighted approach was utilized as the primary analytical method. To enhance the robustness of our findings, multiple sensitivity tests were performed, including Cochran's Q test for evaluating heterogeneity, the MR-Egger intercept test to detect horizontal pleiotropy, and the MR-PRESSO test to identify outliers and assess pleiotropy bias. Additionally, a leave-one-out analysis was performed to validate the consistency of our findings. The MR-Steiger test was also utilized to determine the causal direction in the correlation between the gut microbiota and GEP-NENs. Finally, a reverse MR analysis was performed to assess reverse causality between the intestinal flora and GEP-NENs. Results We identified 42 taxa of the gut microbiota that were potentially causally associated with GEP-NENs; of these taxa, 7, 8, 11, and 16 taxa were causally associated with pancreatic NENs, colorectal NENs, small intestinal NENs, and gastric NENs, respectively. After adjusting for false discovery rate (FDR) correction, we found significant causal links of Euryarchaeota with small intestinal NENs and Family XIII UCG-001 with gastric NENs. The sensitivity analyses confirmed the stability of these correlations. In the reverse MR analysis, colorectal NENs and small intestinal NENs were found to be associated with variations in 8 and 6 different taxa of the gut microbiota, respectively. After adjusting for FDR correction, no significant causal links were detected between GEP-NENs and the intestinal flora. Conclusion The present study reveals a potential causal association between certain taxa of the intestinal flora and GEP-NENs, thus providing new perspectives regarding the role of the intestinal flora in the development of these tumors. These insights could provide innovative approaches to screen and prevent these diseases.
Collapse
Affiliation(s)
- Chun-yu Zhang
- Zhangjiagang Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | | | - Jing-jing Cao
- Zhangjiagang Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Xu
- Zhangjiagang Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiao-yu Wang
- Zhangjiagang Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Rui Li
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhi-wei Miao
- Zhangjiagang Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
47
|
You M, Chen N, Yang Y, Cheng L, He H, Cai Y, Liu Y, Liu H, Hong G. The gut microbiota-brain axis in neurological disorders. MedComm (Beijing) 2024; 5:e656. [PMID: 39036341 PMCID: PMC11260174 DOI: 10.1002/mco2.656] [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: 07/24/2023] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 07/23/2024] Open
Abstract
Previous studies have shown a bidirectional communication between human gut microbiota and the brain, known as the microbiota-gut-brain axis (MGBA). The MGBA influences the host's nervous system development, emotional regulation, and cognitive function through neurotransmitters, immune modulation, and metabolic pathways. Factors like diet, lifestyle, genetics, and environment shape the gut microbiota composition together. Most research have explored how gut microbiota regulates host physiology and its potential in preventing and treating neurological disorders. However, the individual heterogeneity of gut microbiota, strains playing a dominant role in neurological diseases, and the interactions of these microbial metabolites with the central/peripheral nervous systems still need exploration. This review summarizes the potential role of gut microbiota in driving neurodevelopmental disorders (autism spectrum disorder and attention deficit/hyperactivity disorder), neurodegenerative diseases (Alzheimer's and Parkinson's disease), and mood disorders (anxiety and depression) in recent years and discusses the current clinical and preclinical gut microbe-based interventions, including dietary intervention, probiotics, prebiotics, and fecal microbiota transplantation. It also puts forward the current insufficient research on gut microbiota in neurological disorders and provides a framework for further research on neurological disorders.
Collapse
Affiliation(s)
- Mingming You
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Nan Chen
- Master of Public HealthSchool of Public HealthXiamen UniversityXiamenChina
| | - Yuanyuan Yang
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Lingjun Cheng
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Hongzhang He
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Yanhua Cai
- Master of Public HealthSchool of Public HealthXiamen UniversityXiamenChina
| | - Yating Liu
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Haiyue Liu
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Guolin Hong
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| |
Collapse
|
48
|
Liu X, Li S, Wang L, Ma K. Microecological regulation in HCC therapy: Gut microbiome enhances ICI treatment. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167230. [PMID: 38734322 DOI: 10.1016/j.bbadis.2024.167230] [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: 10/24/2023] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024]
Abstract
The exploration of the complex mechanisms of cancer immunotherapy is rapidly evolving worldwide, and our focus is on the interaction of hepatocellular carcinoma (HCC) with immune checkpoint inhibitors (ICIs), particularly as it relates to the regulatory role of the gut microbiome. An important basis for the induction of immune responses in HCC is the presence of specific anti-tumor cells that can be activated and reinforced by ICIs, which is why the application of ICIs results in sustained tumor response rates in the majority of HCC patients. However, mechanisms of acquired resistance to immunotherapy in unresectable HCC result in no long-term benefit for some patients. The significant heterogeneity of inter-individual differences in the gut microbiome in response to treatment with ICIs makes it possible to target modulation of specific gut microbes to assist in augmenting checkpoint blockade therapies in HCC. This review focuses on the complex relationship between the gut microbiome, host immunity, and HCC, and emphasizes that manipulating the gut microbiome to improve response rates to cancer ICI therapy is a clinical strategy with unlimited potential.
Collapse
Affiliation(s)
- Xuliang Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shiyao Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Liming Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China; Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China; Engineering Technology Research Center for Translational Medicine, The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China.
| | - Kexin Ma
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
49
|
Nicze M, Dec A, Borówka M, Krzyżak D, Bołdys A, Bułdak Ł, Okopień B. Molecular Mechanisms behind Obesity and Their Potential Exploitation in Current and Future Therapy. Int J Mol Sci 2024; 25:8202. [PMID: 39125772 PMCID: PMC11311839 DOI: 10.3390/ijms25158202] [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: 06/28/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Obesity is a chronic disease caused primarily by the imbalance between the amount of calories supplied to the body and energy expenditure. Not only does it deteriorate the quality of life, but most importantly it increases the risk of cardiovascular diseases and the development of type 2 diabetes mellitus, leading to reduced life expectancy. In this review, we would like to present the molecular pathomechanisms underlying obesity, which constitute the target points for the action of anti-obesity medications. These include the central nervous system, brain-gut-microbiome axis, gastrointestinal motility, and energy expenditure. A significant part of this article is dedicated to incretin-based drugs such as GLP-1 receptor agonists (e.g., liraglutide and semaglutide), as well as the brand new dual GLP-1 and GIP receptor agonist tirzepatide, all of which have become "block-buster" drugs due to their effectiveness in reducing body weight and beneficial effects on the patient's metabolic profile. Finally, this review article highlights newly designed molecules with the potential for future obesity management that are the subject of ongoing clinical trials.
Collapse
Affiliation(s)
- Michał Nicze
- Department of Internal Medicine and Clinical Pharmacology, Faculty of Medical Sciences, Medical University of Silesia in Katowice, Medyków 18, 40-752 Katowice, Poland (A.B.); (B.O.)
| | | | | | | | | | - Łukasz Bułdak
- Department of Internal Medicine and Clinical Pharmacology, Faculty of Medical Sciences, Medical University of Silesia in Katowice, Medyków 18, 40-752 Katowice, Poland (A.B.); (B.O.)
| | | |
Collapse
|
50
|
Gómez-García M, Moreno-Jimenez E, Morgado N, García-Sánchez A, Gil-Melcón M, Pérez-Pazos J, Estravís M, Isidoro-García M, Dávila I, Sanz C. The Role of the Gut and Airway Microbiota in Chronic Rhinosinusitis with Nasal Polyps: A Systematic Review. Int J Mol Sci 2024; 25:8223. [PMID: 39125792 PMCID: PMC11311313 DOI: 10.3390/ijms25158223] [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: 07/02/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
In recent years, there has been growing interest in understanding the potential role of microbiota dysbiosis or alterations in the composition and function of human microbiota in the development of chronic rhinosinusitis with nasal polyposis (CRSwNP). This systematic review evaluated the literature on CRSwNP and host microbiota for the last ten years, including mainly nasal bacteria, viruses, and fungi, following the PRISMA guidelines and using the major scientific publication databases. Seventy original papers, mainly from Asia and Europe, met the inclusion criteria, providing a comprehensive overview of the microbiota composition in CRSwNP patients and its implications for inflammatory processes in nasal polyps. This review also explores the potential impact of microbiota-modulating therapies for the CRSwNP treatment. Despite variability in study populations and methodologies, findings suggest that fluctuations in specific taxa abundance and reduced bacterial diversity can be accepted as critical factors influencing the onset or severity of CRSwNP. These microbiota alterations appear to be implicated in triggering cell-mediated immune responses, cytokine cascade changes, and defects in the epithelial barrier. Although further human studies are required, microbiota-modulating strategies could become integral to future combined CRSwNP treatments, complementing current therapies that mainly target inflammatory mediators and potentially improving patient outcomes.
Collapse
Affiliation(s)
- Manuel Gómez-García
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.G.-G.); (E.M.-J.); (N.M.); (A.G.-S.); (M.G.-M.); (J.P.-P.); (M.I.-G.); (I.D.); (C.S.)
- Pharmacogenetics and Precision Medicine Unit, Clinical Biochemistry Department, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Emma Moreno-Jimenez
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.G.-G.); (E.M.-J.); (N.M.); (A.G.-S.); (M.G.-M.); (J.P.-P.); (M.I.-G.); (I.D.); (C.S.)
- Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain
| | - Natalia Morgado
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.G.-G.); (E.M.-J.); (N.M.); (A.G.-S.); (M.G.-M.); (J.P.-P.); (M.I.-G.); (I.D.); (C.S.)
- Biomedical and Diagnostics Sciences Department, University of Salamanca, 37007 Salamanca, Spain
| | - Asunción García-Sánchez
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.G.-G.); (E.M.-J.); (N.M.); (A.G.-S.); (M.G.-M.); (J.P.-P.); (M.I.-G.); (I.D.); (C.S.)
- Biomedical and Diagnostics Sciences Department, University of Salamanca, 37007 Salamanca, Spain
- Results-Oriented Cooperative Research Networks in Health—Red de Enfermedades Inflamatorias, Carlos III Health Institute, 28220 Madrid, Spain
| | - María Gil-Melcón
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.G.-G.); (E.M.-J.); (N.M.); (A.G.-S.); (M.G.-M.); (J.P.-P.); (M.I.-G.); (I.D.); (C.S.)
- Otorhinolaryngology and Head and Neck Surgery Department, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Jacqueline Pérez-Pazos
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.G.-G.); (E.M.-J.); (N.M.); (A.G.-S.); (M.G.-M.); (J.P.-P.); (M.I.-G.); (I.D.); (C.S.)
- Pharmacogenetics and Precision Medicine Unit, Clinical Biochemistry Department, University Hospital of Salamanca, 37007 Salamanca, Spain
- Centre for Networked Biomedical Research in Cardiovascular Diseases (CIBERCV), Carlos III Health Institute, 28220 Madrid, Spain
| | - Miguel Estravís
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.G.-G.); (E.M.-J.); (N.M.); (A.G.-S.); (M.G.-M.); (J.P.-P.); (M.I.-G.); (I.D.); (C.S.)
- Results-Oriented Cooperative Research Networks in Health—Red de Enfermedades Inflamatorias, Carlos III Health Institute, 28220 Madrid, Spain
| | - María Isidoro-García
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.G.-G.); (E.M.-J.); (N.M.); (A.G.-S.); (M.G.-M.); (J.P.-P.); (M.I.-G.); (I.D.); (C.S.)
- Pharmacogenetics and Precision Medicine Unit, Clinical Biochemistry Department, University Hospital of Salamanca, 37007 Salamanca, Spain
- Results-Oriented Cooperative Research Networks in Health—Red de Enfermedades Inflamatorias, Carlos III Health Institute, 28220 Madrid, Spain
- Medicine Department, University of Salamanca, 37007 Salamanca, Spain
| | - Ignacio Dávila
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.G.-G.); (E.M.-J.); (N.M.); (A.G.-S.); (M.G.-M.); (J.P.-P.); (M.I.-G.); (I.D.); (C.S.)
- Biomedical and Diagnostics Sciences Department, University of Salamanca, 37007 Salamanca, Spain
- Results-Oriented Cooperative Research Networks in Health—Red de Enfermedades Inflamatorias, Carlos III Health Institute, 28220 Madrid, Spain
- Department of Allergy, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Catalina Sanz
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.G.-G.); (E.M.-J.); (N.M.); (A.G.-S.); (M.G.-M.); (J.P.-P.); (M.I.-G.); (I.D.); (C.S.)
- Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain
- Results-Oriented Cooperative Research Networks in Health—Red de Enfermedades Inflamatorias, Carlos III Health Institute, 28220 Madrid, Spain
| |
Collapse
|