1
|
Mohsen E, Haffez H, Ahmed S, Hamed S, El-Mahdy TS. Multiple Sclerosis: A Story of the Interaction Between Gut Microbiome and Components of the Immune System. Mol Neurobiol 2025; 62:7762-7775. [PMID: 39934561 PMCID: PMC12078361 DOI: 10.1007/s12035-025-04728-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025]
Abstract
Multiple sclerosis (MS) is defined as an inflammatory disorder that chronically affects the central nervous system of young people mostly and is distributed globally. It is associated with degeneration and demyelination of the myelin sheath around the nerves, resulting in multiple neurological disability symptoms ranging from mild to severe cases that end with paralysis sometimes. MS is one of the rising diseases globally that is unfortunately associated with reduced quality of life and adding national economic burdens. The definite MS mechanism is not clearly defined; however, all the previous researches confirm the role of the immune system as the master contributor in the pathogenesis. Innate and adaptive immune cells are activated peripherally then attracted toward the central nervous system (CNS) due to the breakdown of the blood-brain barrier. Recently, the gut-brain axis was shown to depend on gut metabolites that are produced by different microorganisms in the colon. The difference in microbiota composition between individuals is responsible for diversity in secreted metabolites that affect immune responses locally in the gut or systemically when reach blood circulation to the brain. It may enhance or suppress immune responses in the central nervous system (CNS) (repeated short forms); consequently, it may exacerbate or ameliorate MS symptoms. Recent data showed that some metabolites can be used as adjuvant therapy in MS and other inflammatory diseases. This review sheds light on the nature of MS and the possible interaction between gut microbiota and immune system regulation through the gut-brain axis, hence contributing to MS pathogenesis.
Collapse
Affiliation(s)
- Esraa Mohsen
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, PO Box 11795, Cairo, Egypt
| | - Hesham Haffez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, PO Box 11795, Cairo, Egypt
- Center of Scientific Excellence "Helwan Structural Biology Research (HSBR), Helwan University, Cairo, 11795, Egypt
| | - Sandra Ahmed
- Department of Neurology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Selwan Hamed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, PO Box 11795, Cairo, Egypt.
| | - Taghrid S El-Mahdy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, PO Box 11795, Cairo, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| |
Collapse
|
2
|
Fields BD, Pascal DG, Rando OK, Huddleston ME, Ingram K, Hopton R, Grogg MW, Nelson MT, Voigt CA. Design of a Continuous GAA-Producing Probiotic as a Potential Mitigator of the Effects of Sleep Deprivation. ACS Synth Biol 2025. [PMID: 40378286 DOI: 10.1021/acssynbio.4c00690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Creatine is a popular athletic supplement that has also been shown to improve cognitive performance upon sleep deprivation. However, it is rapidly cleared from the gastrointestinal tract a few hours after consumption. Toward providing a persistent creatine dose, we engineered the human probiotic Escherichia coli Nissle (EcN) to produce guanidinoacetic acid (GAA), which is converted to creatine in the liver. We find GAA-producing enzymes present in the human microbiome and compare their activities to known enzymes. Three copies of arginine:glycine amidinotransferase (AGAT) from Actinokineospora terrae are expressed from the genome, and native gcvP, argR, and argA are edited or deleted to improve substrate availability without negatively impacting cell viability. A standard EcN dose (1012 cells) produces 41 ± 7 mg GAA per hour under laboratory conditions. This work demonstrates that a probiotic bacterium can be engineered to produce sustained GAA titers known to impact cognitive performance.
Collapse
Affiliation(s)
- Brandon D Fields
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Daniel G Pascal
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Olivia K Rando
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Mary E Huddleston
- Blue Halo Inc., 4401 Dayton-Xenia Rd, Dayton, Ohio 45432, United States
| | - Katherine Ingram
- Blue Halo Inc., 4401 Dayton-Xenia Rd, Dayton, Ohio 45432, United States
| | - Rachel Hopton
- Blue Halo Inc., 4401 Dayton-Xenia Rd, Dayton, Ohio 45432, United States
| | - Matthew W Grogg
- United States Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
| | - M Tyler Nelson
- United States Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
| | - Christopher A Voigt
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
3
|
Patton S, Silva DP, Fuques E, Klinges G, Muller EM, Thurber RLV. Antibiotic type and dose variably affect microbiomes of a disease-resistant Acropora cervicornis genotype. ENVIRONMENTAL MICROBIOME 2025; 20:46. [PMID: 40317056 PMCID: PMC12049008 DOI: 10.1186/s40793-025-00709-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 04/16/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND As coral diseases become more prevalent and frequent, the need for new intervention strategies also increases to counteract the rapid spread of disease. Recent advances in coral disease mitigation have resulted in increased use of antibiotics on reefs, as their application may halt disease lesion progression. Although efficacious, consequences of deliberate microbiome manipulation resulting from antibiotic administration are less well-understood- especially in non-diseased corals that appear visually healthy. Therefore, to understand how apparently healthy corals are affected by antibiotics, we investigated how three individual antibiotics, and a mixture of the three, impact the microbiome structure and diversity of a disease-resistant Caribbean staghorn coral (Acropora cervicornis) genotype. Over a 96-hour, aquarium-based antibiotic exposure experiment, we collected and processed coral tissue and water samples for 16S rRNA gene analysis. RESULTS We found that antibiotic type and dose distinctively impact microbiome alpha diversity, beta diversity, and community composition. In experimental controls, microbiome composition was dominated by an unclassified bacterial taxon from the order Campylobacterales, while each antibiotic treatment significantly reduced the relative abundance of this taxon. Those taxa that persisted following antibiotic treatment largely differed by antibiotic type and dose, thereby indicating that antibiotic treatment may result in varying potential for opportunist establishment. CONCLUSION Together, these data suggest that antibiotics induce microbiome dysbiosis- hallmarked by the loss of a dominant bacterium and the increase in taxa associated with coral stress responses. Understanding the off-target consequences of antibiotic administration is critical not only for informed, long-term coral restoration practices, but also for highlighting the importance of responsible antibiotic dissemination into natural environments.
Collapse
Affiliation(s)
- Sunni Patton
- Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, Santa Barbara, CA, 93106-9620, USA.
- Department of Microbiology, Oregon State University, 226 Nash Hall, Corvallis, OR, 97331, USA.
| | - Denise P Silva
- Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, Santa Barbara, CA, 93106-9620, USA
- Department of Microbiology, Oregon State University, 226 Nash Hall, Corvallis, OR, 97331, USA
| | - Eddie Fuques
- Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, Santa Barbara, CA, 93106-9620, USA
- Department of Microbiology, Oregon State University, 226 Nash Hall, Corvallis, OR, 97331, USA
| | - Grace Klinges
- Center for Global Discovery and Conservation Science, Arizona State University, Hilo, HI, 96720, USA
- Mote Marine Laboratory, 1600 Ken Thompson Pkwy, Sarasota, FL, 34236, USA
- Mote Marine Laboratory International Center for Coral Reef Research and Restoration, 24244 Overseas Hwy, Summerland Key, FL, 33042, USA
| | - Erinn M Muller
- Mote Marine Laboratory, 1600 Ken Thompson Pkwy, Sarasota, FL, 34236, USA
- Mote Marine Laboratory International Center for Coral Reef Research and Restoration, 24244 Overseas Hwy, Summerland Key, FL, 33042, USA
| | - Rebecca L Vega Thurber
- Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, Santa Barbara, CA, 93106-9620, USA
- Department of Microbiology, Oregon State University, 226 Nash Hall, Corvallis, OR, 97331, USA
| |
Collapse
|
4
|
Randolph NK, Salerno M, Klein H, Diaz-Campos D, van Balen JC, Winston JA. Preparation of Fecal Microbiota Transplantation Products for Companion Animals. PLoS One 2025; 20:e0319161. [PMID: 40203217 PMCID: PMC11981653 DOI: 10.1371/journal.pone.0319161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 01/28/2025] [Indexed: 04/11/2025] Open
Abstract
Fecal microbiota transplantation (FMT) is increasingly utilized in small animal medicine for the treatment of a variety of gastrointestinal and non-gastrointestinal disorders. Despite proven clinical efficacy, there is no detailed protocol available for the preparation and storage of FMT products for veterinarians in a variety of clinical settings. Herein, the effect of processing technique on the microbial community structure was assessed with amplicon sequence analysis. Microbial viability was assessed with standard culture techniques using selective media. Given the fastidious nature of many intestinal microbes, colony forming units are considered surrogate viable microbes, representing a portion of potentially viable microbes. FMT products from four screened canine fecal donors and six screened feline fecal donors were processed aerobically according to a double centrifugation protocol adapted from the human medical literature. Fresh feces from an additional three screened canine fecal donors were used to evaluate the effect of cryopreservative, centrifugation, and short-term storage on microbial community structure and in vitro surrogate bacterial viability. Finally, fresh feces from a third group of three screened canine and three screened feline fecal donors were used to evaluate the long-term in vitro surrogate bacterial viability of three frozen and lyophilized FMT products. Microbiota analysis revealed that each canine fecal donor has a unique microbial profile. Processing of canine and feline feces for FMT does not significantly alter the overall microbial community structure. The addition of cryopreservatives and lyopreservatives significantly improved long-term viability, up to 6 months, for frozen and lyophilized FMT products compared to unprocessed raw feces with no cryopreservative. These results prove the practicality of this approach for FMT preparation in veterinary medicine and provide a detailed protocol for researchers and companion animal practitioners. Future in vivo research is needed to evaluate how the preparation and microbial viability of FMT impacts the recipient's microbial community and clinical outcomes across multiple disease phenotypes.
Collapse
Affiliation(s)
- Nina K. Randolph
- Department of Veterinary Clinical Sciences. College of Veterinary Medicine, The Ohio State University. Columbus, Ohio, United States of America
- Comparative Hepatobiliary and Intestinal Research Program, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Matthew Salerno
- Department of Veterinary Clinical Sciences. College of Veterinary Medicine, The Ohio State University. Columbus, Ohio, United States of America
- Comparative Hepatobiliary and Intestinal Research Program, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Hannah Klein
- Department of Veterinary Clinical Sciences. College of Veterinary Medicine, The Ohio State University. Columbus, Ohio, United States of America
- Comparative Hepatobiliary and Intestinal Research Program, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Dubraska Diaz-Campos
- Department of Veterinary Clinical Sciences. College of Veterinary Medicine, The Ohio State University. Columbus, Ohio, United States of America
| | - Joany C. van Balen
- Department of Veterinary Clinical Sciences. College of Veterinary Medicine, The Ohio State University. Columbus, Ohio, United States of America
| | - Jenessa A. Winston
- Department of Veterinary Clinical Sciences. College of Veterinary Medicine, The Ohio State University. Columbus, Ohio, United States of America
- Comparative Hepatobiliary and Intestinal Research Program, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
5
|
Cao J, Wang S, Ding R, Liu Y, Yuan B. Comparative analyses of the gut microbiome of two sympatric rodent species, Myodes rufocanus and Apodemus peninsulae, in northeast China based on metagenome sequencing. PeerJ 2025; 13:e19260. [PMID: 40226542 PMCID: PMC11988107 DOI: 10.7717/peerj.19260] [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: 06/04/2024] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
The gut microbiota is integral to an animal's physiology, influencing nutritional metabolism, immune function, and environmental adaptation. Despite the significance of gut microbiota in wild rodents, the Korean field mouse (Apodemus peninsulae) and the gray red-backed vole (Myodes rufocanus) remain understudied. To address this, a metagenomic sequencing analysis of the gut microbiome of these sympatric rodents in northeast China's temperate forests was conducted. Intestinal contents were collected from A. peninsulae and M. rufocanus within the Mudanfeng National Nature Reserve. High-throughput sequencing elucidated the gut microbiome's composition, diversity, and functional pathways. Firmicutes, Bacteroidetes, and Proteobacteria were identified as the dominant phyla, with M. rufocanus showing greater microbiome diversity. Key findings indicated distinct gut bacterial communities between the species, with M. rufocanus having a higher abundance of Proteobacteria. The gut microbiota of A. peninsulae and M. rufocanus differed marginally in functional profiles, specifically in the breakdown of complex carbohydrates, which might reflect their distinct food preferences albeit both being herbivores with a substantial dietary overlap. The investigation further elucidated gut microbiota's contributions to energy metabolism and environmental adaptation mechanisms. This study aligns with information on rodent gut microbiota in literature and highlights the two understudied rodent species, providing comparative data for future studies investigating the role of gut microbiota in wildlife health and ecosystem functioning.
Collapse
Affiliation(s)
- Jing Cao
- College of Biology and Food, Shangqiu Normal University, Shangqiu, Henan, China
| | - Shengze Wang
- School of Life Science, Liaocheng University, Liaocheng, Shandong, China
| | - Ruobing Ding
- College of Biology and Food, Shangqiu Normal University, Shangqiu, Henan, China
| | - Yijia Liu
- College of Biology and Food, Shangqiu Normal University, Shangqiu, Henan, China
| | - Baodong Yuan
- School of Life Science, Liaocheng University, Liaocheng, Shandong, China
| |
Collapse
|
6
|
Zou Y, Zou X, Lin C, Han C, Zou Q. Inference of functional differentiation of intestinal microbes between two wild zokor species based on metagenomics. PEST MANAGEMENT SCIENCE 2025; 81:1860-1872. [PMID: 39628107 DOI: 10.1002/ps.8587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/28/2024] [Accepted: 11/21/2024] [Indexed: 03/15/2025]
Abstract
BACKGROUND Currently, there are fewer studies on the intestinal microbes of wild zokors, and it is unclear how zokors adapt to special underground environments by regulating their intestinal microbes. Here, we explored the function of intestinal microbes of Eospalax cansus and Eospalax rothschildi based on metagenomics. RESULTS Both zokor species have similar intestinal microbial composition, but E. cansus has a higher proportion of bacteria involved in carbohydrate degradation. Functional analysis based on KEGG and CAZy databases indicated that the intestinal microbes of E. cansus harboured stronger carbohydrate degradation ability, mainly in starch and sucrose metabolism, and further in cellulose degradation. Furthermore, the cellulase activity was significantly higher in E. cansus than that in E. rothschildi. Eospalax cansus has a stronger microbial fermentation ability due to an increase in fibre-degrading bacteria like unclassified_f_Lachnospiraceae, Ruminococcus, and Clostridium. In addition, the dominant bacteria isolated from zokor were Bacillus, some of which could degrade both cellulose and hemicellulose. CONCLUSION Metagenomic analysis and bacterial isolation experiments indicate that E. cansus has a stronger microbial cellulose-degrading capacity, possibly as an adaptation to its limited food resources underground. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yao Zou
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
- Key Laboratory of National Forestry and Grassland Administration on Management of Western Forest Bio-Disaster, Northwest Agriculture and Forestry University, Yangling, China
| | - Xuan Zou
- Key Laboratory of National Forestry and Grassland Administration on Management of Western Forest Bio-Disaster, Northwest Agriculture and Forestry University, Yangling, China
| | - Chen Lin
- School of Informatics, Xiamen University, Xiamen, China
| | - Chongxuan Han
- Key Laboratory of National Forestry and Grassland Administration on Management of Western Forest Bio-Disaster, Northwest Agriculture and Forestry University, Yangling, China
| | - Quan Zou
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
| |
Collapse
|
7
|
Zhang Y, Liu R, Chen Y, Cao Z, Liu C, Bao R, Wang Y, Huang S, Pan S, Qin L, Wang J, Ning G, Wang W. Akkermansia muciniphila supplementation in patients with overweight/obese type 2 diabetes: Efficacy depends on its baseline levels in the gut. Cell Metab 2025; 37:592-605.e6. [PMID: 39879980 DOI: 10.1016/j.cmet.2024.12.010] [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: 06/07/2024] [Revised: 10/23/2024] [Accepted: 12/17/2024] [Indexed: 01/31/2025]
Abstract
Akkermansia muciniphila is a promising target for managing obesity and type 2 diabetes (T2D), but human studies are limited. We conducted a 12-week randomized, double-blind, placebo-controlled trial involving 58 participants with overweight or obese T2D, who received A. muciniphila (AKK-WST01) or placebo, along with routine lifestyle guidance. Both groups showed decreases in body weight and glycated hemoglobin (HbA1c), without significant between-group differences. In participants with low baseline A. muciniphila, AKK-WST01 supplementation showed high colonization efficiency and significant reductions in body weight, fat mass, and HbA1c, which were not found in the placebo group. However, AKK-WST01 supplementation showed poor colonization and no significant clinical improvements in participants with high baseline A. muciniphila. These findings were verified in germ-free mice receiving feces with low or high A. muciniphila. Our study indicates that metabolic benefits of A. muciniphila supplementation could depend on its baseline intestinal levels, supporting the potential for gut microbiota-guided probiotic supplementation. (ClinicalTrials.gov number, NCT04797442).
Collapse
Affiliation(s)
- Yifei Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yufei Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhiwen Cao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Cong Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Riqiang Bao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yufan Wang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Shan Huang
- Department of Endocrinology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Shijia Pan
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Li Qin
- Department of Endocrinology, Chongming hospital affiliated to Shanghai University of Health & Medicine Sciences, Shanghai 202150, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
8
|
Ren X, Wang L, Yu C, An J, Fu S, Sun H, Zhao M, Te R, Bai X, Yuan J, Liu Y, He J. Impact of oat grain supplementation on growth performance, rumen microbiota, and fatty acid profiles in Hu sheep. Front Microbiol 2025; 16:1528298. [PMID: 40092034 PMCID: PMC11907649 DOI: 10.3389/fmicb.2025.1528298] [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: 11/15/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
The intestinal microbiota plays a vital role in animal growth and development. In this study, we explored the impact of oat grain dietary supplementation on growth performance, intestinal microbiota, short-chain fatty acids (SCFAs), and fatty acids (FAs) in Hu sheep. Thirty-two Hu lambs were randomly assigned to a control group (RC) or an oat grain-supplemented group (RO). After 90 days on their respective diets, rumen digesta were collected from six randomly selected Hu lambs per group to assess microbial diversity, SCFAs, and FAs. The RO diet significantly enhanced growth in Hu sheep (p < 0.01) and increased α-diversity, as indicated by Chao1 and Shannon indices. Core phyla in both groups were Firmicutes and Bacteroidota, with predominant genera including Prevotella, Rikenellaceae_RC9_gut_group, and F082. Oat grain supplementation led to significant shifts in microbial composition, increasing the abundance of Acidobacteriota, Proteobacteria, Chloroflexi, Actinobacteriota, and Subgroup_2, while decreasing Bacteroidota and Oscillospiraceae (p < 0.05). The RO group also exhibited lower levels of isobutyric and citraconic acids but higher levels of azelaic acid (p < 0.05). These results indicate that oat grain supplementation enhances beneficial rumen microbes and optimizes FAs and SCFAs composition, thereby promoting weight gain in Hu sheep.
Collapse
Affiliation(s)
- Xiaoqi Ren
- Research Institute of Biotechnology, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, China
| | - Liwei Wang
- Research Institute of Biotechnology, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
- College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Chuanzong Yu
- Research Institute of Biotechnology, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - Jianghong An
- Research Institute of Biotechnology, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - Shaoyin Fu
- Research Institute of Biotechnology, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - Hua Sun
- Research Institute of Biotechnology, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - Mengran Zhao
- Research Institute of Biotechnology, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - Rigele Te
- Research Institute of Biotechnology, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
| | - Xiaobo Bai
- Research Institute of Biotechnology, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
- College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Jingda Yuan
- Research Institute of Biotechnology, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, China
| | - Yongbin Liu
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, China
- College of Life Sciences, Inner Mongolia University, Hohhot, China
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Jiangfeng He
- Research Institute of Biotechnology, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, China
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, China
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
9
|
Dickey JR, Mercer NM, Kuijpers MCM, Props R, Jackrel SL. Biodiversity within phytoplankton-associated microbiomes regulates host physiology, host community ecology, and nutrient cycling. mSystems 2025; 10:e0146224. [PMID: 39873522 PMCID: PMC11834400 DOI: 10.1128/msystems.01462-24] [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: 10/30/2024] [Accepted: 01/03/2025] [Indexed: 01/30/2025] Open
Abstract
Biological diversity is declining across the tree of life, including among prokaryotes. With the increasing awareness of host-associated microbes as potential regulators of eukaryotic host physiology, behavior, and ecology, it is important to understand the implications of declining diversity within host microbiomes on host fitness, ecology, and ecosystem function. We used phytoplankton and their associated environmental microbiomes as model systems to test the independent and interactive effects of declining microbiome diversity with and without other stressors often caused by human activity-elevated temperature and altered nutrient availability. We found effects of low microbiome diversity on host physiology, phytoplankton community dynamics, and nutrient cycling. Low microbiome diversity caused greater host cellular stress, as indicated by elevated δ13C and δ15N. Microbiome diversity also significantly affected host cell morphological metrics, likely as a consequence of this effect on cell stress. Despite causing greater host cellular stress, the effects of low microbiome diversity on host community ecology included elevated phytoplankton community diversity and biomass. The diversity of these host-associated microbes also had cascading implications on ecosystem nutrient cycling, where lower microbiome diversity caused a depletion of total dissolved N and P in the environment. The magnitude of these effects, caused by microbiome diversity, was greatest among nutrient-depleted environments and at elevated temperatures. Our results emphasize the widespread implications of declining host-associated microbial diversity from host cellular physiology to ecosystem nutrient cycling. These demonstrated effects of declining microbiome diversity are likely to be amplified in ecosystems experiencing multiple stressors caused by anthropogenic activities. IMPORTANCE As evidence is emerging of the key roles that host-associated microbiomes often play in regulating the physiology, fitness, and ecology of their eukaryotic hosts, human activities are causing declines in biological diversity, including within the microbial world. Here, we use a multifactorial manipulative experiment to test the effects of declining diversity within host microbiomes both alone and in tandem with the effects of emerging global changes, including climate warming and shifts in nutrient bioavailability, which are inflicting increasing abiotic stress on host organisms. Using single-celled eukaryotic phytoplankton that harbor an external microbiome as a model system, we demonstrate that diversity within host-associated microbiomes impacts multiple tiers of biological organization, including host physiology, the host population and community ecology, and ecosystem nutrient cycling. Notably, these microbiome diversity-driven effects became magnified in abiotically stressful environments, suggesting that the importance of microbiome diversity may have increased over time during the Anthropocene.
Collapse
Affiliation(s)
- Jonathan R. Dickey
- Department of Ecology, Behavior and Evolution, University of California San Diego School of Biological Sciences, La Jolla, California, USA
| | - Nikki M. Mercer
- Department of Ecology, Behavior and Evolution, University of California San Diego School of Biological Sciences, La Jolla, California, USA
| | - Mirte C. M. Kuijpers
- Department of Ecology, Behavior and Evolution, University of California San Diego School of Biological Sciences, La Jolla, California, USA
| | | | - Sara L. Jackrel
- Department of Ecology, Behavior and Evolution, University of California San Diego School of Biological Sciences, La Jolla, California, USA
| |
Collapse
|
10
|
Eisen A, Kiernan MC. The Neonatal Microbiome: Implications for Amyotrophic Lateral Sclerosis and Other Neurodegenerations. Brain Sci 2025; 15:195. [PMID: 40002527 PMCID: PMC11852589 DOI: 10.3390/brainsci15020195] [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: 01/13/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Most brain development occurs in the "first 1000 days", a critical period from conception to a child's second birthday. Critical brain processes that occur during this time include synaptogenesis, myelination, neural pruning, and the formation of functioning neuronal circuits. Perturbations during the first 1000 days likely contribute to later-life neurodegenerative disease, including sporadic amyotrophic lateral sclerosis (ALS). Neurodevelopment is determined by many events, including the maturation and colonization of the infant microbiome and its metabolites, specifically neurotransmitters, immune modulators, vitamins, and short-chain fatty acids. Successful microbiome maturation and gut-brain axis function depend on maternal factors (stress and exposure to toxins during pregnancy), mode of delivery, quality of the postnatal environment, diet after weaning from breast milk, and nutritional deficiencies. While the neonatal microbiome is highly plastic, it remains prone to dysbiosis which, once established, may persist into adulthood, thereby inducing the development of chronic inflammation and abnormal excitatory/inhibitory balance, resulting in neural excitation. Both are recognized as key pathophysiological processes in the development of ALS.
Collapse
Affiliation(s)
- Andrew Eisen
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Matthew C. Kiernan
- Neuroscience Research Australia, University of New South Wales, Randwick, Sydney, NSW 2031, Australia;
| |
Collapse
|
11
|
Yu F, Zhu C, Wu W. Senile Osteoarthritis Regulated by the Gut Microbiota: From Mechanisms to Treatments. Int J Mol Sci 2025; 26:1505. [PMID: 40003971 PMCID: PMC11855920 DOI: 10.3390/ijms26041505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/19/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
Osteoarthritis (OA) is a chronic, progressive degenerative joint disease that affects the entire synovial joint, leading to the progressive degeneration of articular cartilage. It seriously affects the quality of life and global disability of patients. OA is affected by a variety of factors; the most significant risk factor for OA is age. As individuals age, the risk and severity of OA increase due to the exacerbation of cartilage degeneration and wear and tear. In recent years, research has indicated that the gut microbiota may play a significant role in the aging and OA processes. It is anticipated that regulating the gut microbiota may offer novel approaches to the treatment of OA. The objective of this paper is to examine the relationship between the gut microbiota and senile OA, to investigate the potential mechanisms involved. This review also summarizes the therapeutic strategies related to gut flora in OA management, such as prebiotics and probiotics, diet, exercise, traditional Chinese medicine (TCM) modification, and fecal microbiota transplantation (FMT), highlighting the potential clinical value of gut flora and elucidating the current challenges. The foundation for future research directions is established through the summarization of current research progress.
Collapse
Affiliation(s)
- Fan Yu
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (F.Y.); (C.Z.)
| | - Chenyu Zhu
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (F.Y.); (C.Z.)
| | - Wei Wu
- School of Athletic Performance, Shanghai University of Sports, Shanghai 200438, China
| |
Collapse
|
12
|
Larsson SC, Ericson U, Dekkers KF, Arage G, Rašo LM, Sayols-Baixeras S, Hammar U, Baldanzi G, Nguyen D, Nielsen HB, Holm JB, Risérus U, Michaëlsson K, Sundström J, Smith JG, Engström G, Ärnlöv J, Orho-Melander M, Fall T, Ahmad S. Meat intake in relation to composition and function of gut microbiota. Clin Nutr 2025; 45:124-133. [PMID: 39798223 DOI: 10.1016/j.clnu.2024.12.034] [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: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 01/15/2025]
Abstract
OBJECTIVE Meat intake is suggested to affect gut microbiome composition and the risk of chronic diseases. We aimed to identify meat-associated gut microbiome features and their association with host factors. DESIGN Gut microbiota species were profiled by deep shotgun metagenomics sequencing in 9669 individuals. Intake of white meat, unprocessed red meat, and processed red meat was assessed using a food frequency questionnaire. The associations of meat intake with alpha-diversity and relative abundance of gut microbiota species were tested using linear regression models with adjustment for dietary fiber intake, body mass index, and other potential confounders. Meat-associated species were further assessed for association with enrichment of microbial gene function, meat-associated plasma metabolites, and clinical biomarkers. RESULTS Higher intake of processed red meat was associated with reduced alpha microbial diversity. White meat, unprocessed, and processed red meat intakes were associated with 36, 14, and 322 microbiota species, respectively. Species associated with processed red meat were enriched for bacterial pathways like amino acid degradation, while those negatively linked were enriched for pathways like homoacetogenesis. Furthermore, species positively associated with processed red meat were to a large extent associated with reduced trimethylamine N-oxide and glutamine levels but increased creatine and carnitine metabolites, fasting insulin and glucose, C-reactive protein, apolipoprotein A1, and triglyceride levels and higher blood pressure. CONCLUSION This largest to date population-based study on meat and gut microbiota suggests that meat intake, particularly processed red meat, may modify the gut microbiota composition, functional capacity, and health-related biomarkers.
Collapse
Affiliation(s)
- Susanna C Larsson
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Sweden; Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ulrika Ericson
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Koen F Dekkers
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Getachew Arage
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Luka Marko Rašo
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Sergi Sayols-Baixeras
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Sweden; Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden; CIBER Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Ulf Hammar
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gabriel Baldanzi
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Diem Nguyen
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | - Ulf Risérus
- Clinical Nutrition and Metabolism, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Karl Michaëlsson
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Sweden
| | - Johan Sundström
- Clinical Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden; The George Institute for Global Health, Sydney, Australia
| | - J Gustav Smith
- The Wallenberg Laboratory/Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and the Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Cardiology, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden; Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Johan Ärnlöv
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Huddinge, Sweden; School of Health and Social Studies, Dalarna University, Falun, Sweden; Center for Clinical Research Dalarna, Falun, Uppsala University, Sweden
| | | | - Tove Fall
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Shafqat Ahmad
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden; Preventive Medicine Division, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States; School of Natural Sciences, Technology and Environmental Studies, Södertörn University Sweden, Sweden.
| |
Collapse
|
13
|
Bang S, Shin YH, Park SM, Deng L, Williamson RT, Graham DB, Xavier RJ, Clardy J. Unusual Phospholipids from Morganella morganii Linked to Depression. J Am Chem Soc 2025; 147:2998-3002. [PMID: 39818770 PMCID: PMC11783507 DOI: 10.1021/jacs.4c15158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/19/2025]
Abstract
A multifactorial association study detected a probable causal connection between the prevalence of Morganella morganii in the gut microbiome and the incidence of major depressive disorder (MDD) in the human host. A bioassay-guided fractionation approach identified bacterially produced metabolites that induced pro-inflammatory immune responses. The metabolites are unusual phospholipids that resemble conventional cardiolipins, in which diethanolamine (DEA) replaces the central glycerol. These molecular chimeras of endogenous metabolites from phospholipid biosynthetic pathways and the industrially produced micropollutant DEA activate TLR2/TLR1 receptors and induce the production of pro-inflammatory cytokines, especially IL-6. Their activity in conventional immunomodulatory assays largely parallels that of immunogenic cardiolipins with conventional structures. The molecular mechanism connecting these chimeric cardiolipins to MDD is supported by other studies and has implications for conditions other than MDD.
Collapse
Affiliation(s)
- Sunghee Bang
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| | - Yern-Hyerk Shin
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| | - Sung-Moo Park
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Lei Deng
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - R. Thomas Williamson
- Department
of Chemistry and Biochemistry, University
of North Carolina Wilmington, Wilmington, North Carolina 28409, United States
| | - Daniel B. Graham
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Ramnik J. Xavier
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Jon Clardy
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| |
Collapse
|
14
|
Liu AJ, Lynch JB. Bugs take the sting out. Cell Host Microbe 2025; 33:15-16. [PMID: 39788094 DOI: 10.1016/j.chom.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025]
Abstract
Jiang et al. investigate the role of the microbiota in postherpetic neuralgia (PHN), a chronic pain condition resulting from varicella-zoster virus reactivation. They identify microbiome alterations in PHN patients, linking microbes and pain sensitivity. The researchers identify butyrate-producing Roseburia intestinalis as a mediator of pain sensitivity along the "gut-brain axis."
Collapse
Affiliation(s)
- Arren J Liu
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jonathan B Lynch
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
15
|
White PS, Broe TY, Kuijpers MCM, Dickey JR, Jackrel SL. Host identity drives the assembly of phytoplankton microbiomes across a continental-scale environmental gradient. THE ISME JOURNAL 2025; 19:wraf083. [PMID: 40302044 PMCID: PMC12118458 DOI: 10.1093/ismejo/wraf083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/03/2025] [Accepted: 04/28/2025] [Indexed: 05/01/2025]
Abstract
Host-associated microbiomes often promote host health, yet the key drivers of microbiome assembly and its consequences for host fitness remain unclear. We aimed to determine the relative roles of host identity versus the environment in driving host-microbiome assembly and the consequences of this variation in assembly for host fitness, which may help predict the resilience of host-associated microbiomes and host health amidst fluctuating environmental conditions. Here, we tracked microbiome assembly in association with initially axenic phytoplankton when incubated in seawater originating from four nearshore locations along a continental-scale environmental gradient of North America. Microbiome assembly was highly deterministic. Unexpectedly, host species identity was the overwhelming driver of microbiome community assembly despite continental-scale variation in the environment. Although secondary to host identity, the environment was a significant driver of microbiome assembly for each species evaluated, which, in turn, conferred cascading effects on host fitness as shown by thermal tolerance growth assays. We also found that host-specific microbiomes had host-specific fitness effects, particularly under thermally stressful conditions. Overall, our results advance our understanding of microbiome assembly by empirically demonstrating that although variation among host microbiomes imparted by the local environment has significant implications for host health, the host species is the overwhelming driver of microbiome assembly regardless of wide-scale variation in the environment.
Collapse
Affiliation(s)
- Patricia Signe White
- Department of Ecology, Behavior, and Evolution, University of California San Diego, La Jolla, CA 92093-0116, United States
- Department of Integrative Biology, University of California Berkeley, Berkeley, CA 94720-2284, United States
| | - Taryn Y Broe
- Department of Ecology, Behavior, and Evolution, University of California San Diego, La Jolla, CA 92093-0116, United States
| | - Mirte C M Kuijpers
- Department of Ecology, Behavior, and Evolution, University of California San Diego, La Jolla, CA 92093-0116, United States
| | - Jonathan R Dickey
- Department of Ecology, Behavior, and Evolution, University of California San Diego, La Jolla, CA 92093-0116, United States
| | - Sara L Jackrel
- Department of Ecology, Behavior, and Evolution, University of California San Diego, La Jolla, CA 92093-0116, United States
| |
Collapse
|
16
|
Corl A, Charter M, Rozman G, Turjeman S, Toledo S, Kamath PL, Getz WM, Nathan R, Bowie RCK. Social, environmental, and developmental factors affect the microbiota of barn owls (Tyto alba) in a cross-fostering experiment. Anim Microbiome 2024; 6:77. [PMID: 39719636 DOI: 10.1186/s42523-024-00365-w] [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: 05/15/2024] [Accepted: 12/11/2024] [Indexed: 12/26/2024] Open
Abstract
BACKGROUND Species host diverse microbial communities that can impact their digestion and health, which has led to much interest in understanding the factors that influence their microbiota. We studied the developmental, environmental, and social factors that influence the microbiota of nestling barn owls (Tyto alba) through a partial cross-fostering experiment that manipulated the social and nest environment of the nestlings. We then examined the nestling microbiota before and three weeks after the exchange of nestlings between nests, along with the microbiota of the adults at the nest and nestlings in unmanipulated nests. RESULTS We found that nestlings had higher bacterial diversity and different bacterial communities than adults. The microbiota of nestlings was more like that of their mothers than their fathers, but the similarity to the father tended to increase with the amount of time the father was in close proximity to the nest, as measured from movement data. Cross-fostered offspring had higher bacterial diversity and greater changes in bacterial community composition over time than control offspring. Cross-fostering led the microbiota of the nestlings in the experiment to converge on similar bacterial communities. The microbiota of nestling owls therefore rapidly changed along with alterations to their social and nest environments. CONCLUSIONS These results highlight the dynamic nature of the microbiota during early development and that social interactions can shape microbial communities.
Collapse
Affiliation(s)
- Ammon Corl
- Museum of Vertebrate Zoology, University of California, Berkeley, 3101 Valley Life Sciences Building, Berkeley, CA, 94720-3160, USA.
- Department of Integrative Biology, University of California, Berkeley, 3060 Valley Life Sciences Building, Berkeley, CA, 94720, USA.
| | - Motti Charter
- The Shamir Research Institute, Department of Geography and Environmental Studies, University of Haifa, 199 Aba Hushi Boulevard, Mount Carmel, Haifa, 3498838, Israel
| | - Gabe Rozman
- Movement Ecology Lab, Department of Ecology, Evolution, and Behavior, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus at Givat Ram, Jerusalem, 91904, Israel
| | - Sondra Turjeman
- Movement Ecology Lab, Department of Ecology, Evolution, and Behavior, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus at Givat Ram, Jerusalem, 91904, Israel
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, 1311502, Israel
| | - Sivan Toledo
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Pauline L Kamath
- School of Food and Agriculture, University of Maine, 5735 Hitchner Hall, Orono, ME, 04469, USA
- Maine Center for Genetics in the Environment, University of Maine, 5703 Alumni Hall, Orono, ME, 04469, USA
| | - Wayne M Getz
- Environmental Science, Policy, and Management, University of California, Berkeley, VLSB 5048-B, Berkeley, CA, 94720, USA
- School of Mathematical Sciences, University of KwaZulu, Natal, South Africa
| | - Ran Nathan
- Movement Ecology Lab, Department of Ecology, Evolution, and Behavior, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus at Givat Ram, Jerusalem, 91904, Israel
| | - Rauri C K Bowie
- Museum of Vertebrate Zoology, University of California, Berkeley, 3101 Valley Life Sciences Building, Berkeley, CA, 94720-3160, USA.
- Department of Integrative Biology, University of California, Berkeley, 3060 Valley Life Sciences Building, Berkeley, CA, 94720, USA.
| |
Collapse
|
17
|
Zhang B, Yang H, Cai G, Nie Q, Sun Y. The interactions between the host immunity and intestinal microorganisms in fish. Appl Microbiol Biotechnol 2024; 108:30. [PMID: 38170313 DOI: 10.1007/s00253-023-12934-1] [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: 08/03/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 01/05/2024]
Abstract
There is a huge quantity of microorganisms in the gut of fish, which exert pivotal roles in maintaining host intestinal and general health. The fish immunity can sense and shape the intestinal microbiota and maintain the intestinal homeostasis. In the meantime, the intestinal commensal microbes regulate the fish immunity, control the extravagant proliferation of pathogenic microorganisms, and ensure the intestinal health of the host. This review summarizes developments and progress on the known interactions between host immunity and intestinal microorganisms in fish, focusing on the recent advances in zebrafish (Danio rerio) showing the host immunity senses and shapes intestinal microbiota, and intestinal microorganisms tune host immunity. This review will offer theoretical references for the development, application, and commercialization of intestinal functional microorganisms in fish. KEY POINTS: • The interactions between the intestinal microorganisms and host immunity in zebrafish • Fish immunity senses and shapes the microbiota • Intestinal microbes tune host immunity in fish.
Collapse
Affiliation(s)
- Biyun Zhang
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, Fujian, China
| | - Hongling Yang
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, Fujian, China
| | - Guohe Cai
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, Fujian, China
| | - Qingjie Nie
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, Fujian, China
| | - Yunzhang Sun
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, Fujian, China.
| |
Collapse
|
18
|
Pepke ML, Hansen SB, Limborg MT. Unraveling host regulation of gut microbiota through the epigenome-microbiome axis. Trends Microbiol 2024; 32:1229-1240. [PMID: 38839511 DOI: 10.1016/j.tim.2024.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 06/07/2024]
Abstract
Recent studies of dynamic interactions between epigenetic modifications of a host organism and the composition or activity of its associated gut microbiota suggest an opportunity for the host to shape its microbiome through epigenetic alterations that lead to changes in gene expression and noncoding RNA activity. We use insights from microbiota-induced epigenetic changes to review the potential of the host to epigenetically regulate its gut microbiome, from which a bidirectional 'epigenome-microbiome axis' emerges. This axis embeds environmentally induced variation, which may influence the adaptive evolution of host-microbe interactions. We furthermore present our perspective on how the epigenome-microbiome axis can be understood and investigated within a holo-omic framework with potential applications in the applied health and food sciences.
Collapse
Affiliation(s)
- Michael L Pepke
- Center for Evolutionary Hologenomics, Globe Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, DK-1353 Copenhagen, Denmark.
| | - Søren B Hansen
- Center for Evolutionary Hologenomics, Globe Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, DK-1353 Copenhagen, Denmark
| | - Morten T Limborg
- Center for Evolutionary Hologenomics, Globe Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, DK-1353 Copenhagen, Denmark.
| |
Collapse
|
19
|
Jiang S, Guo X, Qian X, Ning X, Zhang C, Yin S, Zhang K. Sex-bias of core intestinal microbiota in different stocks of Chinese mitten crabs (Eriocheir sinensis). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101281. [PMID: 38935994 DOI: 10.1016/j.cbd.2024.101281] [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: 04/09/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
The differences in intestinal microbiota composition are synergistically shaped by internal and external factors of the host. The core microbiota plays a vital role in maintaining intestinal homeostasis. In this study, we conducted 16S rRNA sequencing analysis to investigate the stability of intestinal microbiota and sex-bias of six stocks of Chinese mitten crabs (105 females; and 110 males). The dominant phyla in all six stocks were Proteobacteria, Tenericutes, Bacteroidetes and Firmicutes; however, their relative abundance differed significantly. Twenty-seven core operational taxonomic units (OTUs), corresponding to 18 genera, were screened. Correlation analysis revealed that OTUs of four stocks in the Yangtze River system play important roles in maintaining the stability of intestinal microbiota. Additionally, the core intestinal microbiota was significantly sex-biased, and the top three genera in terms of relative abundance (Acinetobacter, Vibrio, and Candidatus_Hepatoplasma) were significantly dominant in female crabs. Network structure analysis also confirmed gender differences in the association pattern of intestinal microbiota. The intestinal microbiota of male crabs has a higher degree of functional enrichment. This study provided a theoretical basis for further investigating exploring the shaping effect of gender and geographical factors on the intestinal microbiota of Chinese mitten crabs.
Collapse
Affiliation(s)
- Su Jiang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Xinping Guo
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Xiaobin Qian
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Xianhui Ning
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang 222005, China
| | - Cong Zhang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang 222005, China
| | - Shaowu Yin
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang 222005, China.
| | - Kai Zhang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang 222005, China.
| |
Collapse
|
20
|
Ferreira SCM, Jarquín-Díaz VH, Planillo A, Ďureje Ľ, Martincová I, Kramer-Schadt S, Forslund-Startceva SK, Heitlinger E. Eco-evolutionary dynamics of host-microbiome interactions in a natural population of closely related mouse subspecies and their hybrids. Proc Biol Sci 2024; 291:20241970. [PMID: 39689880 DOI: 10.1098/rspb.2024.1970] [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: 06/19/2024] [Revised: 11/01/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024] Open
Abstract
Closely related host species share similar symbionts, but the effects of host genetic admixture and environmental conditions on these communities remain largely unknown. We investigated the influence of host genetic admixture and environmental factors on the intestinal prokaryotic and eukaryotic communities (fungi, parasites) of two house mouse subspecies (Mus musculus domesticus and M. m. musculus) and their hybrids in two settings: (i) wild-caught mice from the European hybrid zone and (ii) wild-derived inbred mice in a controlled laboratory environment before and during a community perturbation (infection). In wild-caught mice, environmental factors strongly predicted the overall microbiome composition. Subspecies' genetic distance significantly influenced the overall microbiome composition, and each component (bacteria, parasites and fungi). While hybridization had a weak effect, it significantly impacted fungal composition. We observed similar patterns in wild-derived mice, where genetic distances and hybridization influenced microbiome composition, with fungi being more stable to infection-induced perturbations than other microbiome components. Subspecies' genetic distance has a stronger and consistent effect across microbiome components than differences in expected heterozygosity among hybrids, suggesting that host divergence and host filtering play a key role in microbiome divergence, influenced by environmental factors. Our findings offer new insights into the eco-evolutionary processes shaping host-microbiome interactions.
Collapse
Affiliation(s)
- Susana C M Ferreira
- Division of Computational Systems Biology, Center for Microbiology and Ecological Systems Science, University of Vienna, Djerassipl. 1, Vienna 1030, Austria
- Department of Molecular Parasitology, Institute for Biology, Humboldt University Berlin (HU). Philippstr. 13 Haus 14, Berlin 10115, Germany
- Department of Interdisciplinary Life Sciences, Research Institute of Wildlife Ecology, University of Veterinary Medicine Vienna, Savoyenstraße 1, Vienna A-1160, Austria
| | - Víctor Hugo Jarquín-Díaz
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Lindenberger Weg 80, Berlin 13125, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC). Robert-Rössle-Str. 10, Berlin 13125, Germany
- Research Group Ecology and Evolution of Molecular Parasite-Host Interactions, Leibniz Institute for Zoo and Wildlife Research (IZW). Alfred-Kowalke-Straße 17, Berlin 10315, Germany
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Aimara Planillo
- Department of Ecological Dynamics, Leibniz Institute for Zoo and Wildlife Research (IZW). Alfred-Kowalke-Straße 17, Berlin 10315, Germany
| | - Ľudovít Ďureje
- Research Facility Studenec, Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic
| | - Iva Martincová
- Research Facility Studenec, Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic
| | - Stephanie Kramer-Schadt
- Department of Ecological Dynamics, Leibniz Institute for Zoo and Wildlife Research (IZW). Alfred-Kowalke-Straße 17, Berlin 10315, Germany
- Institute of Ecology, Chair of Planning-related Animal Ecology, Technische Universität Berlin (TUB), Rothenburgstr. 12, Berlin 12165, Germany
| | - Sofia K Forslund-Startceva
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Lindenberger Weg 80, Berlin 13125, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC). Robert-Rössle-Str. 10, Berlin 13125, Germany
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Emanuel Heitlinger
- Department of Molecular Parasitology, Institute for Biology, Humboldt University Berlin (HU). Philippstr. 13 Haus 14, Berlin 10115, Germany
- Research Group Ecology and Evolution of Molecular Parasite-Host Interactions, Leibniz Institute for Zoo and Wildlife Research (IZW). Alfred-Kowalke-Straße 17, Berlin 10315, Germany
| |
Collapse
|
21
|
Hu Q, Cheng L, Cao X, Shi F, Ma Y, Mo L, Li J, Zhu S, Liu Z. Comparative analysis of gut microbiota of Chinese Kunming dog, German Shepherd dog, and Belgian Malinois dog. J Vet Sci 2024; 25:e85. [PMID: 39608779 PMCID: PMC11611487 DOI: 10.4142/jvs.24181] [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: 06/20/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 11/30/2024] Open
Abstract
IMPORTANCE The composition of the gut microbiota is essential for a dog's health and its adaptation to the environment. Different bacteria can produce the same essential metabolites beneficial to health owing to bacterial functional redundancy in microbial communities. OBJECTIVE This study examined the gut bacterial communities of dogs from different breeds, all kept under identical domestication conditions. METHODS Noninvasive sampling and 16S rRNA high-throughput sequencing were used to compare the composition and function of the gut microbiota of three dog breeds: the Chinese Kunming dog (CKD), German Shepherd dog (GSD), and Belgian Malinois dog (BMD). RESULTS The gut microbiota of the three dog breeds consisted of 257 species across 146 genera, 60 families, 35 orders, 15 classes, and 10 phyla. The dominant bacterial phyla across the three breeds were Firmicutes (57.44%), Fusobacteriota (28.86%), and Bacteroidota (7.63%), while the dominant bacterial genera across the three breeds were Peptostreptococcus (21.08%), Fusobacterium (18.50%), Lactobacillus (12.37%), and Cetobacter (10.29%). Further analysis revealed significant differences in the intestinal flora of the three breeds at the phylum and genus levels. The intestinal flora of BMD was significantly richer than that of CKD and GSD. The functional prediction and Kyoto Encyclopedia of Genes and Genomes analysis showed that the primary functions of the gut microbiota in these breeds were similar, with significant enrichment in various metabolic pathways, including carbohydrate and amino acid metabolism, secondary metabolite biosynthesis, and microbial metabolism in different environments. The intestinal flora of these breeds also played a crucial role in genetic information processing, including transcription, translation, replication, and material transport. CONCLUSIONS AND RELEVANCE These results provide novel insights into the intestinal flora of intervention dogs and suggest novel methods to improve their health status, which help increase microbial diversity and normalize metabolite production in diseased dogs.
Collapse
Affiliation(s)
- Qingmei Hu
- School of Agriculture & Life Sciences, Kunming University, Kunming 650214, China
| | - Luguang Cheng
- Kunming Police Dog Base, Ministry of Public Security, Kunming 650204, China
| | - Xueting Cao
- School of Agriculture & Life Sciences, Kunming University, Kunming 650214, China
| | - Feng Shi
- School of Agriculture & Life Sciences, Kunming University, Kunming 650214, China
| | - Yunjie Ma
- School of Agriculture & Life Sciences, Kunming University, Kunming 650214, China
| | - Liling Mo
- School of Agriculture & Life Sciences, Kunming University, Kunming 650214, China
| | - Junyu Li
- Department of Ultrasonography, People's Hospital of Fengdu County, Chongqing City, Fengdu 408200, China
| | - Siyi Zhu
- School of Agriculture & Life Sciences, Kunming University, Kunming 650214, China
| | - Zichao Liu
- School of Agriculture & Life Sciences, Kunming University, Kunming 650214, China.
| |
Collapse
|
22
|
Yang S, Huo M, Su Z, Wang F, Zhang Y, Zhong C, Shi Y. The impact of dietary supplementation of Quercetagetin on growth, antioxidant capacity, and gut microbiota of diquat-challenged broilers. Front Microbiol 2024; 15:1453145. [PMID: 39539705 PMCID: PMC11557422 DOI: 10.3389/fmicb.2024.1453145] [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: 06/22/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024] Open
Abstract
This experiment aimed to investigate the effects of Quercetagetin (QG) on the growth performance, antioxidant capacity, and cecal microbiota of broilers. Two hundred and forty 21-day-old WOD168 broilers with similar body weights were randomly divided into five groups of six replicates each with eight chickens. The control group was fed a basal diet composed of corn and soybean meal, while the experimental groups received basal diets supplemented with 0, 10, 20, and 40 mg/kg QG, along with intraperitoneal injection of 20 mg/kg body weight Diquat (DQ). The experiment lasted for 21 days. The results showed that: (1) QG significantly alleviated the decrease in average daily feed intake and average daily gain induced by Diquat, reduced the elevation of serum ACTH content, and significantly increased GH content (P < 0.05); (2) QG supplementation significantly mitigated the decrease in serum CAT activity and duodenal GSH-Px activity induced by Diquat (P < 0.05), as well as the increase in MDA content (P < 0.05); additionally, QG significantly increased the gene expression levels of GSH-Px, Nrf2, and Keap1 (P < 0.05); (3) Alpha and Beta diversity analysis revealed that QG supplementation significantly increased the cecal microbial OTUs and Chao1 index of broilers (P < 0.05). At the phylum level, compared with the Diquat group, the LQG group significantly decreased the relative abundance of Firmicutes (P < 0.05) and significantly increased the relative abundance of Bacteroidota (P < 0.05). At the genus level, compared with the CON group, the Diquat group significantly decreased the abundance of Lactobacillus and Alistipes (P < 0.05), while QG supplementation significantly alleviated the decrease in the abundance of Lactobacillus and Alistipes (P < 0.05). In conclusion, the addition of an appropriate amount (20 mg/kg) of QG to the diet can promote the growth of broilers, enhance antioxidant capacity, and improve intestinal health.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuxiang Shi
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan, China
| |
Collapse
|
23
|
Li IC, Lee YL, Li TJ, Tsai YS, Chen YL, Chen CC. Whole-Genome Sequencing of Three Lactiplantibacillus plantarum Strains Reveals Potential Metabolites for Boosting Host Immunity Safely. J Microbiol Biotechnol 2024; 34:2079-2090. [PMID: 39263794 PMCID: PMC11540610 DOI: 10.4014/jmb.2402.02013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/03/2024] [Accepted: 07/24/2024] [Indexed: 09/13/2024]
Abstract
In response to the growing demand for immune-related products, this study evaluated the safety and immune-modulating potential of three newly discovered Lactiplantibacillus plantarum strains (GKM3, GKK1, and GKD7) through toxicity tests and whole-genome sequencing. Safety evaluations, including the analysis of antimicrobial resistance genes, virulence factors, plasmids, and prophages, classified these strains as safe for human consumption. Acute oral toxicity tests further supported their safety. To evaluate their immune-modulating potential, dendritic cells were exposed to these strains, and the secretion of key cytokines (IFN-β and IL-12) was measured. Among the strains, GKK1 exhibited the highest enhancement of IFN-β and IL-12 production, suggesting its potential as an immune-stimulating probiotic. Bioinformatics analysis revealed potential metabolic pathways and secondary metabolites, including predicted bacteriocins, associated with immune modulation. The presence of a nitrate reductase region in the GKK1 strain indicated its ability to produce nitric oxide, a critical molecule involved in immune regulation and host defense. The presence of glucorhamnan-related gene clusters in GKK1 also suggested immune-enhancing effects. Nitrate reductase expression was confirmed using qPCR, with the highest levels detected in GKK1. Moreover, this study is the first to show an anti-inflammatory effect of plantaricin A, linked to its presence in strain GKM3 and its potential therapeutic applications due to sequence similarity to known anti-inflammatory peptides. Overall, these three L. plantarum strains demonstrated a safe profile and GKK1 showed potential as an immunity-enhancing probiotic. However, additional investigation is required to confirm the involvement of specific metabolic pathways, secondary metabolites, and bacteriocins in immune responses.
Collapse
Affiliation(s)
- I-Chen Li
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 110, Taiwan
| | - Tsung-Ju Li
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
| | - You-Shan Tsai
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
| | - Yen-Lien Chen
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
| | - Chin-Chu Chen
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
- Department of Food Science, Nutrition, and Nutraceutical Biotechnology, Shih Chien University, Taipei City 104, Taiwan
- Institute of Food Science and Technology, National Taiwan University, Taipei City 106, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan City 320, Taiwan
| |
Collapse
|
24
|
Cao Y, Yang N, Gu J, Zhang X, Ye J, Chen J, Li H. Distinct biogeographic patterns for bacteria and fungi in association with Bursaphelenchus xylophilus nematodes and infested pinewood. Microbiol Spectr 2024; 12:e0077824. [PMID: 39162557 PMCID: PMC11448397 DOI: 10.1128/spectrum.00778-24] [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: 03/25/2024] [Accepted: 07/15/2024] [Indexed: 08/21/2024] Open
Abstract
Pinewood nematodes (PWN, Bursaphelenchus xylophilus) are destructive plant parasitic nematodes that cause pine wilt disease (PWD) by attacking the vascular systems of pine trees, resulting in widespread tree mortality. Research has shown that there are connections between nematode-associated microbes and PWD. Yet the variations in microbial communities across different geographic regions are not well-understood. In this study, we examined the bacterial and fungal communities associated with nematodes and infested wood collected from 34 sites across three vegetation zones in China, as well as samples from the United States, using 16S rRNA and internal transcribed spacer (ITS) gene amplicon sequencing. The predominant genera Pseudomonas and Rhodococcus were found in nematodes, and Acinetobacter was present in the wood of PWD-infected pine trees across China. Network analysis revealed that core bacterial taxa belonged to the Pseudomonadota and Actinomycetota phyla for the nematodes, whereas the Pseudomonadota and Bacteroidota phyla were dominant in the infested wood. Identification of enriched key microbial taxa in nematodes and infested wood across vegetation zones indicates distinct biogeographic microbial community structures and key bacterial species. Although the nematode-associated bacterial community showed consistency across geographic distances, the similarity of the PWD pine trees' bacterial community decreased with distance, suggesting a spatial correlation with environmental variables. Our findings enhance our understanding of the microbiota associated with pinewood nematode (PWN) and offer valuable insights into PWD management. IMPORTANCE Our research uncovered specific bacteria and fungi linked to pinewood nematode (PWN) and infested wood in three different vegetation zones in China, as well as samples from the United States. This sheds light on the critical roles of certain microbial groups, such as Pseudomonas, Acinetobacter, and Stenotrophomonas, in influencing PWN fitness. Understanding these patterns provides valuable insights into the dynamics of PWN-associated microbiomes, offering potential strategies for managing pine wilt disease (PWD). We found significant correlations between geographic distance and similarity in bacterial communities in the infested wood, indicating a spatial influence on wood-associated microbial communities due to limited dispersal and localized environmental conditions. Further investigations of these spatial patterns and driving forces are crucial for understanding the ecological processes that shape microbial communities in complex ecosystems and, ultimately, for mitigating the transmission of PWN in forests.
Collapse
Affiliation(s)
- Yuyu Cao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of Ministry of Agriculture and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang, China
| | - Nan Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of Ministry of Agriculture and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang, China
| | - Jianfeng Gu
- Ningbo Key Laboratory of Port Biological and Food Safety Testing (Technical Centre of Ningbo Customs/Ningbo Inspection and Quarantine Science Technology Academy), No. 8, Huikang Road, Ningbo, Zhejiang 315100, China
| | - Xingyao Zhang
- Key Laboratory of Forest Protection of National Forestry and Grassland Administration, Ecology and Nature Conservation Institute, Chinese Academy of Forestry, Beijing 100091, China
| | - Jianren Ye
- Co-Innovation Centre for Sustainable Forestry in Southern China, Forestry and Grassland, College of Soil and Water Conservation, Nanjing Forestry University, Nanjing 210037, China
| | - Jianping Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of Ministry of Agriculture and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang, China
| | - Hongjie Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of Ministry of Agriculture and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
25
|
Sonnega S, Sheriff MJ. Harnessing the gut microbiome: a potential biomarker for wild animal welfare. Front Vet Sci 2024; 11:1474028. [PMID: 39415953 PMCID: PMC11479891 DOI: 10.3389/fvets.2024.1474028] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
The welfare of wild animal populations is critically important to conservation, with profound implications for ecosystem health, biodiversity, and zoonotic disease transmission. Animal welfare is typically defined as the accumulated affective mental state of an animal over a particular time period. However, the assessment of animal welfare in the wild poses unique challenges, primarily due to the lack of universally applicable biomarkers. This perspective explores the potential role of the gut microbiome, a dynamic and non-invasive biomarker, as a novel avenue for evaluating animal welfare in wild animals. The gut microbiome, through interactions with the host's physiology, behavior, and cognition, offers a promising opportunity to gain insights into the well-being of animals. In this synthesis, we discuss the distinction between fitness and welfare, the complexities of assessing welfare in wild populations, and the linkages between the gut microbiome and aspects of animal welfare such as behavior and cognition. We lastly elucidate how the gut microbiome could serve as a valuable tool for wildlife managers, with the potential to serve as a non-invasive yet informative window into the welfare of wild animals. As this nascent field evolves, it presents unique opportunities to enhance our understanding of the well-being of wild animals and to contribute to the preservation of ecosystems, biodiversity, and human health.
Collapse
Affiliation(s)
- Sam Sonnega
- Department of Biology, UMass Dartmouth, Dartmouth, MA, United States
| | | |
Collapse
|
26
|
Schneider KM, Kummen M, Trivedi PJ, Hov JR. Role of microbiome in autoimmune liver diseases. Hepatology 2024; 80:965-987. [PMID: 37369002 PMCID: PMC11407779 DOI: 10.1097/hep.0000000000000506] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/25/2023] [Indexed: 06/29/2023]
Abstract
The microbiome plays a crucial role in integrating environmental influences into host physiology, potentially linking it to autoimmune liver diseases, such as autoimmune hepatitis, primary biliary cholangitis, and primary sclerosing cholangitis. All autoimmune liver diseases are associated with reduced diversity of the gut microbiome and altered abundance of certain bacteria. However, the relationship between the microbiome and liver diseases is bidirectional and varies over the course of the disease. This makes it challenging to dissect whether such changes in the microbiome are initiating or driving factors in autoimmune liver diseases, secondary consequences of disease and/or pharmacological intervention, or alterations that modify the clinical course that patients experience. Potential mechanisms include the presence of pathobionts, disease-modifying microbial metabolites, and more nonspecific reduced gut barrier function, and it is highly likely that the effect of these change during the progression of the disease. Recurrent disease after liver transplantation is a major clinical challenge and a common denominator in these conditions, which could also represent a window to disease mechanisms of the gut-liver axis. Herein, we propose future research priorities, which should involve clinical trials, extensive molecular phenotyping at high resolution, and experimental studies in model systems. Overall, autoimmune liver diseases are characterized by an altered microbiome, and interventions targeting these changes hold promise for improving clinical care based on the emerging field of microbiota medicine.
Collapse
Affiliation(s)
| | - Martin Kummen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Palak J. Trivedi
- National Institute for Health and Care Research Birmingham Biomedical Research Centre, Centre for Liver and Gastroenterology Research, University of Birmingham, UK
- Liver Unit, University Hospitals Birmingham Queen Elizabeth, Birmingham, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, UK
- Institute of Applied Health Research, University of Birmingham, UK
| | - Johannes R. Hov
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
27
|
Li Y, Han S. Metabolomic Applications in Gut Microbiota-Host Interactions in Human Diseases. Gastroenterol Clin North Am 2024; 53:383-397. [PMID: 39068001 DOI: 10.1016/j.gtc.2023.12.008] [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: 07/30/2024]
Abstract
The human gut microbiota, consisting of trillions of microorganisms, encodes diverse metabolic pathways that impact numerous aspects of host physiology. One key way in which gut bacteria interact with the host is through the production of small metabolites. Several of these microbiota-dependent metabolites, such as short-chain fatty acids, have been shown to modulate host diseases. In this review, we examine how disease-associated metabolic signatures are identified using metabolomic platforms, and where metabolomics is applied in gut microbiota-disease interactions. We further explore how integration of metagenomic and metabolomic data in human studies can facilitate biomarkers discoveries in precision medicine.
Collapse
Affiliation(s)
- Yuxin Li
- Biochemistry Graduate Program, Duke University School of Medicine, Durham, NC 27710, USA
| | - Shuo Han
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA; Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, NC 27710, USA.
| |
Collapse
|
28
|
Eterovick PC, Schmidt R, Sabino-Pinto J, Yang C, Künzel S, Ruthsatz K. The microbiome at the interface between environmental stress and animal health: an example from the most threatened vertebrate group. Proc Biol Sci 2024; 291:20240917. [PMID: 39291456 PMCID: PMC11409201 DOI: 10.1098/rspb.2024.0917] [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: 02/18/2024] [Revised: 06/05/2024] [Accepted: 07/25/2024] [Indexed: 09/19/2024] Open
Abstract
Nitrate pollution and global warming are ubiquitous stressors likely to interact and affect the health and survival of wildlife, particularly aquatic ectotherms. Animal health is largely influenced by its microbiome (commensal/symbiotic microorganisms), which responds to such stressors. We used a crossed experimental design including three nitrate levels and five temperature regimes to investigate their interactive and individual effects on an aquatic ectotherm, the European common frog. We associated health biomarkers in larvae with changes in gut bacteria diversity and composition. Larvae experienced higher stress levels and lower body condition under high temperatures and nitrate exposure. Developmental rate increased with temperature but decreased with nitrate pollution. Alterations in bacteria composition but not diversity are likely to correlate with the observed outcomes in larvae health. Leucine degradation decreased at higher temperatures corroborating accelerated development, nitrate degradation increased with nitrate level corroborating reduced body condition and an increase in lysine biosynthesis may have helped larvae deal with the combined effects of both stressors. These results reinforce the importance of associating traditional health biomarkers with underlying microbiome changes. Therefore, we urge studies to investigate the effects of environmental stressors on microbiome composition and consequences for host health in a world threatened by biodiversity loss.
Collapse
Affiliation(s)
- Paula Cabral Eterovick
- Zoological Institute, Technische Universität Braunschweig, Mendelssohnstraße 4, 38106, Braunschweig, Germany
| | - Robin Schmidt
- Zoological Institute, Technische Universität Braunschweig, Mendelssohnstraße 4, 38106, Braunschweig, Germany
| | - Joana Sabino-Pinto
- GELIFES—Groningen Institute for Evolutionary Life Sciences, University of Groningen, 9747, AG Groningen, The Netherlands
| | - Chen Yang
- Department of Biostatistics, Southern Medical University, 510515, Guangzhou, People’s Republic of China
| | - Sven Künzel
- Max-Planck-Institut für Evolutionsbiologie, 24306, Plön, Germany
| | - Katharina Ruthsatz
- Zoological Institute, Technische Universität Braunschweig, Mendelssohnstraße 4, 38106, Braunschweig, Germany
| |
Collapse
|
29
|
Rowaiye A, Ibeanu GC, Bur D, Nnadi S, Mgbeke OE, Morikwe U. Gut microbiota alteration - Cancer relationships and synbiotic roles in cancer therapies. THE MICROBE 2024; 4:100096. [DOI: 10.1016/j.microb.2024.100096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
30
|
Koellsch C, Poulin R, Salloum PM. Microbial artists: the role of parasite microbiomes in explaining colour polymorphism among amphipods and potential link to host manipulation. J Evol Biol 2024; 37:1009-1022. [PMID: 38989853 DOI: 10.1093/jeb/voae085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/16/2024] [Accepted: 07/10/2024] [Indexed: 07/12/2024]
Abstract
Parasite infections are increasingly reported to change the microbiome of the parasitized hosts, while parasites bring their own microbes to what can be a multi-dimensional interaction. For instance, a recent hypothesis suggests that the microbial communities harboured by parasites may play a role in the well-documented ability of many parasites to manipulate host phenotype, and explain why the degree to which host phenotype is altered varies among conspecific parasites. Here, we explored whether the microbiomes of both hosts and parasites are associated with variation in host manipulation by parasites. Using colour quantification methods applied to digital images, we investigated colour variation among uninfected Transorchestia serrulata amphipods, as well as amphipods infected with Plagiorhynchus allisonae acanthocephalans and with a dilepidid cestode. We then characterized the bacteriota of amphipod hosts and of their parasites, looking for correlations between host phenotype and the bacterial taxa associated with hosts and parasites. We found large variation in amphipod colours, and weak support for a direct impact of parasites on the colour of their hosts. Conversely, and most interestingly, the parasite's bacteriota was more strongly correlated with colour variation among their amphipod hosts, with potential impact of amphipod-associated bacteria as well. Some bacterial taxa found associated with amphipods and parasites may have the ability to synthesize pigments, and we propose they may interact with colour determination in the amphipods. This study provides correlational support for an association between the parasite's microbiome and the evolution of host manipulation by parasites and host-parasite interactions more generally.
Collapse
Affiliation(s)
- Célia Koellsch
- Department of Zoology, University of Otago, Dunedin, New Zealand
| | - Robert Poulin
- Department of Zoology, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
31
|
Wang Z, Li S, Zhang S, Zhang T, Wu Y, Liu A, Wang K, Ji X, Cao H, Zhang Y, Tan EK, Wang Y, Wang Y, Liu W. Hosts manipulate lifestyle switch and pathogenicity heterogeneity of opportunistic pathogens in the single-cell resolution. eLife 2024; 13:RP96789. [PMID: 39190452 PMCID: PMC11349298 DOI: 10.7554/elife.96789] [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] [Indexed: 08/28/2024] Open
Abstract
Host-microbe interactions are virtually bidirectional, but how the host affects their microbiome is poorly understood. Here, we report that the host is a critical modulator to regulate the lifestyle switch and pathogenicity heterogeneity of the opportunistic pathogens Serratia marcescens utilizing the Drosophila and bacterium model system. First, we find that Drosophila larvae efficiently outcompete S. marcescens and typically drive a bacterial switch from pathogenicity to commensalism toward the fly. Furthermore, Drosophila larvae reshape the transcriptomic and metabolic profiles of S. marcescens characterized by a lifestyle switch. More importantly, the host alters pathogenicity and heterogeneity of S. marcescens in the single-cell resolution. Finally, we find that larvae-derived AMPs are required to recapitulate the response of S. marcescens to larvae. Altogether, our findings provide an insight into the pivotal roles of the host in harnessing the life history and heterogeneity of symbiotic bacterial cells, advancing knowledge of the reciprocal relationships between the host and pathogen.
Collapse
Affiliation(s)
- Ziguang Wang
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
- College of Life Sciences, Nankai UniversityTianjinChina
- First Clinical Medical College, Mudanjiang Medical CollegeMudanjiangChina
| | - Shuai Li
- Bioinformatics Center, College of Biology, Hunan UniversityChangshaChina
| | - Sheng Zhang
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Tianyu Zhang
- Liangzhu Laboratory, Zhejiang UniversityHangzhouChina
| | - Yujie Wu
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Anqi Liu
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Kui Wang
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Xiaowen Ji
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Haiqun Cao
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Yinglao Zhang
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital CampusSingaporeSingapore
| | | | - Yirong Wang
- Bioinformatics Center, College of Biology, Hunan UniversityChangshaChina
| | - Wei Liu
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| |
Collapse
|
32
|
Ji G, Zhao J, Si X, Song W. Targeting bacterial metabolites in tumor for cancer therapy: An alternative approach for targeting tumor-associated bacteria. Adv Drug Deliv Rev 2024; 211:115345. [PMID: 38834140 DOI: 10.1016/j.addr.2024.115345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 05/11/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
Emerging evidence reveal that tumor-associated bacteria (TAB) can facilitate the initiation and progression of multiple types of cancer. Recent work has emphasized the significant role of intestinal microbiota, particularly bacteria, plays in affecting responses to chemo- and immuno-therapies. Hence, it seems feasible to improve cancer treatment outcomes by targeting intestinal bacteria. While considering variable richness of the intestinal microbiota and diverse components among individuals, direct manipulating the gut microbiota is complicated in clinic. Tumor initiation and progression requires the gut microbiota-derived metabolites to contact and reprogram neoplastic cells. Hence, directly targeting tumor-associated bacteria metabolites may have the potential to provide alternative and innovative strategies to bypass the gut microbiota for cancer therapy. As such, there are great opportunities to explore holistic approaches that incorporates TAB-derived metabolites and related metabolic signals modulation for cancer therapy. In this review, we will focus on key opportunistic areas by targeting TAB-derived metabolites and related metabolic signals, but not bacteria itself, for cancer treatment, and elucidate future challenges that need to be addressed in this emerging field.
Collapse
Affiliation(s)
- Guofeng Ji
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jingjing Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China
| | - Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China.
| |
Collapse
|
33
|
Stothart MR, McLoughlin PD, Medill SA, Greuel RJ, Wilson AJ, Poissant J. Methanogenic patterns in the gut microbiome are associated with survival in a population of feral horses. Nat Commun 2024; 15:6012. [PMID: 39039075 PMCID: PMC11263349 DOI: 10.1038/s41467-024-49963-x] [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: 11/03/2023] [Accepted: 06/21/2024] [Indexed: 07/24/2024] Open
Abstract
Gut microbiomes are widely hypothesised to influence host fitness and have been experimentally shown to affect host health and phenotypes under laboratory conditions. However, the extent to which they do so in free-living animal populations and the proximate mechanisms involved remain open questions. In this study, using long-term, individual-based life history and shallow shotgun metagenomic sequencing data (2394 fecal samples from 794 individuals collected between 2013-2019), we quantify relationships between gut microbiome variation and survival in a feral population of horses under natural food limitation (Sable Island, Canada), and test metagenome-derived predictions using short-chain fatty acid data. We report detailed evidence that variation in the gut microbiome is associated with a host fitness proxy in nature and outline hypotheses of pathogenesis and methanogenesis as key causal mechanisms which may underlie such patterns in feral horses, and perhaps, wild herbivores more generally.
Collapse
Affiliation(s)
- Mason R Stothart
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Biology, University of Oxford, Oxford, United Kingdom.
| | - Philip D McLoughlin
- Department of Biology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Sarah A Medill
- Department of Biology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Ruth J Greuel
- Department of Biology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Alastair J Wilson
- Centre for Ecology and Conservation, University of Exeter, Penryn, United Kingdom
| | - Jocelyn Poissant
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
34
|
Zhu L, Ma S, He C, Bai L, Tu W, Wu X. Microbial and Metabolic Profiling of Obese and Lean Luchuan Pigs: Implications for Phenotypic Divergence. Animals (Basel) 2024; 14:2111. [PMID: 39061573 PMCID: PMC11273426 DOI: 10.3390/ani14142111] [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: 06/19/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Luchuan (LC) pigs are a Chinese breed renowned for their distinctive black and white coloring, superior meat quality and rapid reproduction, but their growth rate is slow. Over the course of approximately two decades of controlled breeding, the LC pigs maintained at the Shanghai Academy of Agricultural Sciences (Shanghai, China) have diverged into two phenotypes: one characterized by obesity (FLC) and the other by leanness (LLC). Recent studies indicate a correlation between microorganisms and the differentiation of host phenotypes. In this study, we examined the fecal microbiota profiles and serum metabolites of FLC and LLC pigs. The body weight, chest circumference, and alanine aminotransferase and aspartate aminotransferase enzyme activities were increased in the FLC pigs compared to the LLC pigs. Conversely, the levels of the Fusobacterium and Streptococcus genera were lower in the FLC pigs, while the number of Firmicutes, Lactobacillus, Phascolartobacterium, and Rikenellaceae_RC9_gut_group members were higher. A total of 52 metabolites were altered between the two groups, with many playing crucial roles in prolactin signaling, oocyte meiosis, and aldosterone-regulated sodium reabsorption pathways. The correlation analyses demonstrated a significant association between the modified microbiota and metabolites and the phenotypic variations observed in the LC pigs. Specifically, Jeotgalicoccus was positively correlated with the body weight and chest circumference, but was negatively correlated with metabolites such as 2-mercaptobenzothiazole and N1-pyrazin-2-yl-4-chlorobenzamide, which were positively associated with Bacteroides. These results provide compelling evidence for a novel relationship between the gut microbiome and metabolome in the phenotypic differentiation of LC pigs.
Collapse
Affiliation(s)
- Lihui Zhu
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China;
| | - Shengwei Ma
- Key Laboratory of Agricultural Genetics and Breeding, Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (S.M.); (C.H.); (L.B.)
| | - Chuan He
- Key Laboratory of Agricultural Genetics and Breeding, Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (S.M.); (C.H.); (L.B.)
| | - Lan Bai
- Key Laboratory of Agricultural Genetics and Breeding, Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (S.M.); (C.H.); (L.B.)
| | - Weilong Tu
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China;
| | - Xiao Wu
- Key Laboratory of Agricultural Genetics and Breeding, Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (S.M.); (C.H.); (L.B.)
| |
Collapse
|
35
|
Han S, Guiberson ER, Li Y, Sonnenburg JL. High-throughput identification of gut microbiome-dependent metabolites. Nat Protoc 2024; 19:2180-2205. [PMID: 38740909 DOI: 10.1038/s41596-024-00980-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/18/2024] [Indexed: 05/16/2024]
Abstract
A significant hurdle that has limited progress in microbiome science has been identifying and studying the diverse set of metabolites produced by gut microbes. Gut microbial metabolism produces thousands of difficult-to-identify metabolites, which present a challenge to study their roles in host biology. In recent years, mass spectrometry-based metabolomics has become one of the core technologies for identifying small metabolites. However, metabolomics expertise, ranging from sample preparation to instrument use and data analysis, is often lacking in academic labs. Most targeted metabolomics methods provide high levels of sensitivity and quantification, while they are limited to a panel of predefined molecules that may not be informative to microbiome-focused studies. Here we have developed a gut microbe-focused and wide-spectrum metabolomic protocol using liquid chromatography-mass spectrometry and bioinformatic analysis. This protocol enables users to carry out experiments from sample collection to data analysis, only requiring access to a liquid chromatography-mass spectrometry instrument, which is often available at local core facilities. By applying this protocol to samples containing human gut microbial metabolites, spanning from culture supernatant to human biospecimens, our approach enables high-confidence identification of >800 metabolites that can serve as candidate mediators of microbe-host interactions. We expect this protocol will lower the barrier to tracking gut bacterial metabolism in vitro and in mammalian hosts, propelling hypothesis-driven mechanistic studies and accelerating our understanding of the gut microbiome at the chemical level.
Collapse
Affiliation(s)
- Shuo Han
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA.
- Duke Microbiome Center, Duke University School of Medicine, Durham, NC, USA.
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.
| | - Emma R Guiberson
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuxin Li
- Biochemistry Graduate Program, Duke University School of Medicine, Durham, NC, USA
| | - Justin L Sonnenburg
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
- Chan-Zuckerberg Biohub, San Francisco, CA, USA.
- Center for Human Microbiome Studies, Stanford, CA, USA.
| |
Collapse
|
36
|
Seong H, Han SH, Kim G, Han NS. Viability and probiotic activity of Lactiplantibacillus plantarum PMO08 in human gastrointestinal tract analyzed by in vitro gut model. Food Sci Biotechnol 2024; 33:2223-2231. [PMID: 39130653 PMCID: PMC11315860 DOI: 10.1007/s10068-024-01622-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: 04/16/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 08/13/2024] Open
Abstract
This study aimed to evaluate the survivability of Lactiplantibacillus plantarum PMO08 in the human gastrointestinal tract and its adaptability in the colon using in vitro models. After exposure to gastric and small intestinal conditions, the majority (92.70 ± 1.14%) of PMO08 was found to be damaged, as determined by confocal microscopy and flow cytometry. During in vitro colonic fermentation, PMO08 not only increased abundance up to 0.47 ± 0.04% compared with the control sample (0.00 ± 0.00%) at 24 h but also facilitated the growth of beneficial or commensal bacteria, thereby increasing the α-diversity indices. Additionally, PMO08 significantly elevated the levels of short-chain fatty acids (SCFAs) and various organic acids. Our results demonstrate that PMO08 possesses moderate viability under gastrointestinal conditions but exhibits superior probiotic activity in the colon.
Collapse
Affiliation(s)
- Hyunbin Seong
- Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk Republic of Korea
| | - Seung Hee Han
- Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk Republic of Korea
| | - Geonhee Kim
- Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk Republic of Korea
| | - Nam Soo Han
- Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk Republic of Korea
| |
Collapse
|
37
|
Zhang K, He C, Wang L, Suo L, Guo M, Guo J, Zhang T, Xu Y, Lei Y, Liu G, Qian Q, Mao Y, Kalds P, Wu Y, Cuoji A, Yang Y, Brugger D, Gan S, Wang M, Wang X, Zhao F, Chen Y. Compendium of 5810 genomes of sheep and goat gut microbiomes provides new insights into the glycan and mucin utilization. MICROBIOME 2024; 12:104. [PMID: 38845047 PMCID: PMC11155115 DOI: 10.1186/s40168-024-01806-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 04/03/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Ruminant gut microbiota are critical in ecological adaptation, evolution, and nutrition utilization because it regulates energy metabolism, promotes nutrient absorption, and improves immune function. To study the functional roles of key gut microbiota in sheep and goats, it is essential to construct reference microbial gene catalogs and high-quality microbial genomes database. RESULTS A total of 320 fecal samples were collected from 21 different sheep and goat breeds, originating from 32 distinct farms. Metagenomic deep sequencing and binning assembly were utilized to construct a comprehensive microbial genome information database for the gut microbiota. We successfully generated the largest reference gene catalogs for gut microbiota in sheep and goats, containing over 162 million and 82 million nonredundant predicted genes, respectively, with 49 million shared nonredundant predicted genes and 1138 shared species. We found that the rearing environment has a greater impact on microbial composition and function than the host's species effect. Through subsequent assembly, we obtained 5810 medium- and high-quality metagenome-assembled genomes (MAGs), out of which 2661 were yet unidentified species. Among these MAGs, we identified 91 bacterial taxa that specifically colonize the sheep gut, which encode polysaccharide utilization loci for glycan and mucin degradation. CONCLUSIONS By shedding light on the co-symbiotic microbial communities in the gut of small ruminants, our study significantly enhances the understanding of their nutrient degradation and disease susceptibility. Our findings emphasize the vast potential of untapped resources in functional bacterial species within ruminants, further expanding our knowledge of how the ruminant gut microbiota recognizes and processes glycan and mucins. Video Abstract.
Collapse
Affiliation(s)
- Ke Zhang
- International Joint Agriculture Research Center for Animal Bio-Breeding, Ministry of Agriculture and Rural Affairs/Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Chong He
- College of Information Engineering, Northwest A&F University, Yangling, 712100, China
| | - Lei Wang
- Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai Province, Key Laboratory of Animal Genetics and Breeding On Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Qinghai Academy of Animal and Veterinary Medicine, Qinghai University, Xining, 810016, China
| | - Langda Suo
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
- Key Laboratory of Animal Genetics and Breeding On Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, 850009, China
| | - Mengmeng Guo
- College of Animal Engineering, Yangling Vocational and Technical College, Yangling, 712100, China
| | - Jiazhong Guo
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611100, China
| | - Ting Zhang
- International Joint Agriculture Research Center for Animal Bio-Breeding, Ministry of Agriculture and Rural Affairs/Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yangbin Xu
- International Joint Agriculture Research Center for Animal Bio-Breeding, Ministry of Agriculture and Rural Affairs/Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yu Lei
- International Joint Agriculture Research Center for Animal Bio-Breeding, Ministry of Agriculture and Rural Affairs/Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Gongwei Liu
- International Joint Agriculture Research Center for Animal Bio-Breeding, Ministry of Agriculture and Rural Affairs/Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Quan Qian
- International Joint Agriculture Research Center for Animal Bio-Breeding, Ministry of Agriculture and Rural Affairs/Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yunrui Mao
- International Joint Agriculture Research Center for Animal Bio-Breeding, Ministry of Agriculture and Rural Affairs/Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Peter Kalds
- International Joint Agriculture Research Center for Animal Bio-Breeding, Ministry of Agriculture and Rural Affairs/Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yujiang Wu
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
- Key Laboratory of Animal Genetics and Breeding On Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, 850009, China
| | - Awang Cuoji
- Institute of Animal Sciences, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850009, China
- Key Laboratory of Animal Genetics and Breeding On Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, 850009, China
| | - Yuxin Yang
- International Joint Agriculture Research Center for Animal Bio-Breeding, Ministry of Agriculture and Rural Affairs/Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Daniel Brugger
- Institute of Animal Nutrition and Dietetics, Vetsuisse-Faculty, University of Zurich, 8057, Zurich, Switzerland
| | - Shangquan Gan
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Meili Wang
- College of Information Engineering, Northwest A&F University, Yangling, 712100, China
| | - Xiaolong Wang
- International Joint Agriculture Research Center for Animal Bio-Breeding, Ministry of Agriculture and Rural Affairs/Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, China.
- School of Future Technology On Bio-Breeding, Northwest A&F University, Yangling, 712100, China.
| | - Fangqing Zhao
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 102206, China.
| | - Yulin Chen
- International Joint Agriculture Research Center for Animal Bio-Breeding, Ministry of Agriculture and Rural Affairs/Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, China.
- School of Future Technology On Bio-Breeding, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
38
|
Bennett GM, Kwak Y, Maynard R. Endosymbioses Have Shaped the Evolution of Biological Diversity and Complexity Time and Time Again. Genome Biol Evol 2024; 16:evae112. [PMID: 38813885 PMCID: PMC11154151 DOI: 10.1093/gbe/evae112] [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: 04/23/2024] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 05/31/2024] Open
Abstract
Life on Earth comprises prokaryotes and a broad assemblage of endosymbioses. The pages of Molecular Biology and Evolution and Genome Biology and Evolution have provided an essential window into how these endosymbiotic interactions have evolved and shaped biological diversity. Here, we provide a current perspective on this knowledge by drawing on decades of revelatory research published in Molecular Biology and Evolution and Genome Biology and Evolution, and insights from the field at large. The accumulated work illustrates how endosymbioses provide hosts with novel phenotypes that allow them to transition between adaptive landscapes to access environmental resources. Such endosymbiotic relationships have shaped and reshaped life on Earth. The early serial establishment of mitochondria and chloroplasts through endosymbioses permitted massive upscaling of cellular energetics, multicellularity, and terrestrial planetary greening. These endosymbioses are also the foundation upon which all later ones are built, including everything from land-plant endosymbioses with fungi and bacteria to nutritional endosymbioses found in invertebrate animals. Common evolutionary mechanisms have shaped this broad range of interactions. Endosymbionts generally experience adaptive and stochastic genome streamlining, the extent of which depends on several key factors (e.g. mode of transmission). Hosts, in contrast, adapt complex mechanisms of resource exchange, cellular integration and regulation, and genetic support mechanisms to prop up degraded symbionts. However, there are significant differences between endosymbiotic interactions not only in how partners have evolved with each other but also in the scope of their influence on biological diversity. These differences are important considerations for predicting how endosymbioses will persist and adapt to a changing planet.
Collapse
Affiliation(s)
- Gordon M Bennett
- Department of Life and Environmental Sciences, University of California, Merced, CA, USA
- National Science Foundation Biological Integration Institute—INSITE, University of California, Merced, CA, USA
| | - Younghwan Kwak
- Department of Life and Environmental Sciences, University of California, Merced, CA, USA
- National Science Foundation Biological Integration Institute—INSITE, University of California, Merced, CA, USA
| | - Reo Maynard
- Department of Life and Environmental Sciences, University of California, Merced, CA, USA
| |
Collapse
|
39
|
Härer A, Frazier CJ, Rennison DJ. Host ecotype and rearing environment are the main drivers of threespine stickleback gut microbiota diversity in a naturalistic experiment. ROYAL SOCIETY OPEN SCIENCE 2024; 11:240649. [PMID: 39100190 PMCID: PMC11296155 DOI: 10.1098/rsos.240649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/01/2024] [Indexed: 08/06/2024]
Abstract
Host-microbiota interactions play a critical role in the hosts' biology, and thus, it is crucial to elucidate the mechanisms that shape gut microbial communities. We leveraged threespine stickleback fish (Gasterosteus aculeatus) as a model system to investigate the contribution of host and environmental factors to gut microbiota variation. These fish offer a unique opportunity for experiments in naturalistic conditions; we reared benthic and limnetic ecotypes from three different lakes in experimental ponds, allowing us to assess the relative effects of shared environment (pond), geographic origin (lake-of-origin), trophic ecology and genetics (ecotype) and biological sex on gut microbiota α- and β-diversity. Host ecotype had the strongest influence on α-diversity, with benthic fish exhibiting higher diversity than limnetic fish, followed by the rearing environment. β-diversity was primarily shaped by rearing environment, followed by host ecotype, indicating that environmental factors play a crucial role in determining gut microbiota composition. Furthermore, numerous bacterial orders were differentially abundant across ponds, underlining the substantial contribution of environmental factors to gut microbiota variation. Our study illustrates the complex interplay between environmental and host ecological or genetic factors in shaping the stickleback gut microbiota and highlights the value of experiments conducted under naturalistic conditions for understanding gut microbiota dynamics.
Collapse
Affiliation(s)
- Andreas Härer
- School of Biological Sciences, Department of Ecology, Behavior & Evolution, University of California San Diego, La Jolla, CA, USA
| | - Christine J. Frazier
- School of Biological Sciences, Department of Ecology, Behavior & Evolution, University of California San Diego, La Jolla, CA, USA
| | - Diana J. Rennison
- School of Biological Sciences, Department of Ecology, Behavior & Evolution, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
40
|
Wu Y, Wang Q, Yang W, Zhang S, Mao CX, He N, Zhou S, Zhou C, Liu W. The cluster digging behavior of larvae confers trophic benefits to fitness in insects. INSECT SCIENCE 2024; 31:870-884. [PMID: 38161191 DOI: 10.1111/1744-7917.13307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/20/2023] [Accepted: 10/30/2023] [Indexed: 01/03/2024]
Abstract
Collective behaviors efficiently impart benefits to a diversity of species ranging from bacteria to humans. Fly larvae tend to cluster and form coordinated digging groups under crowded conditions, yet understanding the rules governing this behavior is in its infancy. We primarily took advantage of the Drosophila model to investigate cooperative foraging behavior. Here, we report that Drosophila-related species and the black soldier fly have evolved a conserved strategy of cluster digging in food foraging. Subsequently, we investigated relative factors, including larval stage, population density, and food stiffness and quality, that affect the cluster digging behavior. Remarkably, oxygen supply through the posterior breathing spiracles is necessary for the organization of digging clusters. More importantly, we theoretically devise a mathematical model to accurately calculate how the cluster digging behavior expands food resources by diving depth, cross-section area, and food volume. We found that cluster digging behavior approximately increases 2.2 fold depth, 1.7-fold cross-section area, and 1.9 fold volume than control groups, respectively. Amplification of food sources significantly facilitates survival, larval development, and reproductive success of Drosophila challenged with competition for limited food resources, thereby conferring trophic benefits to fitness in insects. Overall, our findings highlight that the cluster digging behavior is a pivotal behavior for their adaptation to food scarcity, advancing a better understanding of how this cooperative behavior confers fitness benefits in the animal kingdom.
Collapse
Affiliation(s)
- Yujie Wu
- School of Plant Protection, Anhui Agricultural University; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, Hefei, China
- College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Qiang Wang
- School of Teacher Education, Nanjing Xiaozhuang University, Nanjing, China
| | - Weikang Yang
- School of Plant Protection, Anhui Agricultural University; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, Hefei, China
| | - Sheng Zhang
- School of Plant Protection, Anhui Agricultural University; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, Hefei, China
| | - Chuan-Xi Mao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Science, Hubei University, Wuhan, China
| | - Nana He
- School of Plant Protection, Anhui Agricultural University; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, Hefei, China
| | - Shaojie Zhou
- School of Plant Protection, Anhui Agricultural University; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, Hefei, China
| | - Chuanming Zhou
- School of Plant Protection, Anhui Agricultural University; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, Hefei, China
| | - Wei Liu
- School of Plant Protection, Anhui Agricultural University; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Province Engineering Laboratory for Green Pesticide Development and Application, Hefei, China
| |
Collapse
|
41
|
Qiu Y, Hou Y, Gohel D, Zhou Y, Xu J, Bykova M, Yang Y, Leverenz JB, Pieper AA, Nussinov R, Caldwell JZK, Brown JM, Cheng F. Systematic characterization of multi-omics landscape between gut microbial metabolites and GPCRome in Alzheimer's disease. Cell Rep 2024; 43:114128. [PMID: 38652661 PMCID: PMC11968202 DOI: 10.1016/j.celrep.2024.114128] [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: 12/22/2023] [Revised: 03/06/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024] Open
Abstract
Shifts in the magnitude and nature of gut microbial metabolites have been implicated in Alzheimer's disease (AD), but the host receptors that sense and respond to these metabolites are largely unknown. Here, we develop a systems biology framework that integrates machine learning and multi-omics to identify molecular relationships of gut microbial metabolites with non-olfactory G-protein-coupled receptors (termed the "GPCRome"). We evaluate 1.09 million metabolite-protein pairs connecting 408 human GPCRs and 335 gut microbial metabolites. Using genetics-derived Mendelian randomization and integrative analyses of human brain transcriptomic and proteomic profiles, we identify orphan GPCRs (i.e., GPR84) as potential drug targets in AD and that triacanthine experimentally activates GPR84. We demonstrate that phenethylamine and agmatine significantly reduce tau hyperphosphorylation (p-tau181 and p-tau205) in AD patient induced pluripotent stem cell-derived neurons. This study demonstrates a systems biology framework to uncover the GPCR targets of human gut microbiota in AD and other complex diseases if broadly applied.
Collapse
Affiliation(s)
- Yunguang Qiu
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yuan Hou
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dhruv Gohel
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yadi Zhou
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jielin Xu
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Marina Bykova
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yuxin Yang
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Andrew A Pieper
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Jessica Z K Caldwell
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA; Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Las Vegas, NV 89106, USA
| | - J Mark Brown
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA; Department of Cancer Biology, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44195, USA; Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA; Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
42
|
Dong JH, Xu X, Ren ZX, Zhao YH, Zhang Y, Chen L, Wu Y, Chen G, Cao R, Wu Q, Wang H. The adaptation of bumblebees to extremely high elevation associated with their gut microbiota. mSystems 2024; 9:e0121923. [PMID: 38329353 PMCID: PMC10949452 DOI: 10.1128/msystems.01219-23] [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: 11/15/2023] [Accepted: 01/09/2024] [Indexed: 02/09/2024] Open
Abstract
Bumblebees are among the most abundant and important pollinators for sub-alpine and alpine flowering plant species in the Northern Hemisphere, but little is known about their adaptations to high elevations. In this article, we focused on two bumblebee species, Bombus friseanus and Bombus prshewalskyi, and their respective gut microbiota. The two species, distributed through the Hengduan Mountains of southwestern China, show species replacement at different elevations. We performed genome sequencing based on 20 worker bee samples of each species. Applying evolutionary population genetics and metagenomic approaches, we detected genes under selection and analyzed functional pathways between bumblebees and their gut microbes. We found clear genetic differentiation between the two host species and significant differences in their microbiota. Species replacement occurred in both hosts and their bacteria (Snodgrassella) with an increase in elevation. These extremely high-elevation bumblebees show evidence of positive selection related to diverse biological processes. Positively selected genes involved in host immune systems probably contributed to gut microbiota changes, while the butyrate generated by gut microbiota may influence both host energy metabolism and immune systems. This suggests a close association between the genomes of the host species and their microbiomes based on some degree of natural selection.IMPORTANCETwo closely related and dominant bumblebee species, distributed at different elevations through the Hengduan Mountains of southwestern China, showed a clear genomic signature of adaptation to elevation at the molecular level and significant differences in their respective microbiota. Species replacement occurred in both hosts and their bacteria (Snodgrassella) with an increase in elevation. Bumblebees' adaptations to higher elevations are closely associated with their gut microbiota through two biological processes: energy metabolism and immune response. Information allowing us to understand the adaptive mechanisms of species to extreme conditions is implicit if we are to conserve them as their environments change.
Collapse
Affiliation(s)
- Jiu-Hong Dong
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xin Xu
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Zong-Xin Ren
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Yan-Hui Zhao
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Yaran Zhang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Li Chen
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - You Wu
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Guotao Chen
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ruiqing Cao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qi Wu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Hong Wang
- CAS Key Laboratory for Plant Diversity and Biogeography of East Asia, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
43
|
Suárez J. Scrutinizing microbiome determinism: why deterministic hypotheses about the microbiome are conceptually ungrounded. HISTORY AND PHILOSOPHY OF THE LIFE SCIENCES 2024; 46:12. [PMID: 38347271 PMCID: PMC10861753 DOI: 10.1007/s40656-024-00610-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 01/16/2024] [Indexed: 02/15/2024]
Abstract
This paper addresses the topic of determinism in contemporary microbiome research. I distinguish two types of deterministic claims about the microbiome, and I show evidence that both types of claims are present in the contemporary literature. First, the idea that the host genetics determines the composition of the microbiome which I call "host-microbiome determinism". Second, the idea that the genetics of the holobiont (the individual unit composed by a host plus its microbiome) determines the expression of certain phenotypic traits, which I call "microbiome-phenotype determinism". Drawing on the stability of traits conception of individuality (Suárez in Hist Philos Life Sci 42:11, 2020) I argue that none of these deterministic hypotheses is grounded on our current knowledge of how the holobiont is transgenerationally assembled, nor how it expresses its phenotypic traits.
Collapse
Affiliation(s)
- Javier Suárez
- BIOETHICS Research Group - Department of Philosophy, University of Oviedo, Oviedo, Spain.
- Institute of Philosophy, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
44
|
Bringhurst B, Greenwold M, Kellner K, Seal JN. Symbiosis, dysbiosis and the impact of horizontal exchange on bacterial microbiomes in higher fungus-gardening ants. Sci Rep 2024; 14:3231. [PMID: 38332146 PMCID: PMC10853281 DOI: 10.1038/s41598-024-53218-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/30/2024] [Indexed: 02/10/2024] Open
Abstract
Advances in our understanding of symbiotic stability have demonstrated that microorganisms are key to understanding the homeostasis of obligate symbioses. Fungus-gardening ants are excellent model systems for exploring how microorganisms may be involved in symbiotic homeostasis as the host and symbionts are macroscopic and can be easily experimentally manipulated. Their coevolutionary history has been well-studied; examinations of which have depicted broad clade-to-clade specificity between the ants and fungus. Few studies hitherto have addressed the roles of microbiomes in stabilizing these associations. Here, we quantified changes in microbiome structure as a result of experimentally induced horizontal exchange of symbionts. This was done by performing cross-fostering experiments forcing ants to grow novel fungi and comparing known temporally unstable (undergoing dysbiosis) and stable combinations. We found that fungus-gardening ants alter their unstable, novel garden microbiomes into configurations like those found in native gardens. Patterns of dysbiosis/symbiosis appear to be predictable in that two related species with similar specificity patterns also show similar patterns of microbial change, whereas a species with more relaxed specificity does not show such microbiome change or restructuring when growing different fungi. It appears that clade-to-clade specificity patterns are the outcomes of community-level interactions that promote stability or cause symbiotic collapse.
Collapse
Affiliation(s)
- Blake Bringhurst
- Department of Biology, University of Texas at Tyler, 3900 University Blvd, Tyler, TX, 757998, USA
- Department of Evolution, Ecology and Organismal Biology, The Ohio State University, 1315 Kinnear Rd, Columbus, OH, 43212, USA
| | - Matthew Greenwold
- Department of Biology, University of Texas at Tyler, 3900 University Blvd, Tyler, TX, 757998, USA
| | - Katrin Kellner
- Department of Biology, University of Texas at Tyler, 3900 University Blvd, Tyler, TX, 757998, USA
| | - Jon N Seal
- Department of Biology, University of Texas at Tyler, 3900 University Blvd, Tyler, TX, 757998, USA.
| |
Collapse
|
45
|
Yu Z, Cantet JM, Paz HA, Kaufman JD, Orellano MS, Ipharraguerre IR, Ríus AG. Heat stress-associated changes in the intestinal barrier, inflammatory signals, and microbiome communities in dairy calves. J Dairy Sci 2024; 107:1175-1196. [PMID: 37730180 DOI: 10.3168/jds.2023-23873] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023]
Abstract
Recent studies indicate that heat stress pathophysiology is associated with intestinal barrier dysfunction, local and systemic inflammation, and gut dysbiosis. However, inconclusive results and a poor description of tissue-specific changes must be addressed to identify potential intervention targets against heat stress illness in growing calves. Therefore, the objective of this study was to evaluate components of the intestinal barrier, pro- and anti-inflammatory signals, and microbiota community composition in Holstein bull calves exposed to heat stress. Animals (mean age = 12 wk old; mean body weight = 122 kg) penned individually in temperature-controlled rooms were assigned to (1) thermoneutral conditions (constant room temperature at 19.5°C) and restricted offer of feed (TNR, n = 8), or (2) heat stress conditions (cycles of room temperatures ranging from 20 to 37.8°C) along with ad libitum offer of feed (HS, n = 8) for 7 d. Upon treatment completion, sections of the jejunum, ileum, and colon were collected and snap-frozen immediately to evaluate gene and protein expression, cytokine concentrations, and myeloperoxidase activity. Digesta aliquots of the ileum, colon, and rectum were collected to assess bacterial communities. Plasma was harvested on d 2, 5, and 7 to determine cytokine concentrations. Overall, results showed a section-specific effect of HS on intestinal integrity. Jejunal mRNA expression of TJP1 was decreased by 70.9% in HS relative to TNR calves. In agreement, jejunal expression of heat shock transcription factor-1 protein, a known tight junction protein expression regulator, decreased by 48% in HS calves. Jejunal analyses showed that HS decreased concentrations of IL-1α by 36.6% and tended to decrease the concentration of IL-17A. Conversely, HS elicited a 3.5-fold increase in jejunal concentration of anti-inflammatory IL-36 receptor antagonist. Plasma analysis of pro-inflammatory cytokines showed that IL-6 decreased by 51% in HS relative to TNR calves. Heat stress alteration of the large intestine bacterial communities was characterized by increased genus Butyrivibrio_3, a known butyrate-producing organism, and changes in bacteria metabolism of energy and AA. A strong positive correlation between the rectal temperature and pro-inflammatory Eggerthii spp. was detected in HS calves. In conclusion, this work indicates that HS impairs the intestinal barrier function of jejunum. The pro- and anti-inflammatory signal changes may be part of a broader response to restore intestinal homeostasis in jejunum. The changes in large intestine bacterial communities favoring butyrate-producing organisms (e.g., Butyrivibrio spp.) may be part of a successful response to maintain the integrity of the colonic mucosa of HS calves. The alteration of intestinal homeostasis should be the target for heat stress therapies to restore biological functions, and, thus highlights the relevance of this work.
Collapse
Affiliation(s)
- Z Yu
- Department of Animal Science, University of Tennessee Institute of Agriculture, Knoxville, TN 37996
| | - J M Cantet
- Department of Animal Science, University of Tennessee Institute of Agriculture, Knoxville, TN 37996
| | - H A Paz
- Department of Pediatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205; Arkansas Children's Nutrition Center, Little Rock, AR 72202
| | - J D Kaufman
- Department of Animal Science, University of Tennessee Institute of Agriculture, Knoxville, TN 37996
| | - M S Orellano
- Centro de Investigaciones y Transferencia de Villa María, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Villa María, Villa María, Córdoba 5900, Argentina
| | - I R Ipharraguerre
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel 24118, Germany
| | - A G Ríus
- Department of Animal Science, University of Tennessee Institute of Agriculture, Knoxville, TN 37996.
| |
Collapse
|
46
|
Hammer TJ. Why do hosts malfunction without microbes? Missing benefits versus evolutionary addiction. Trends Microbiol 2024; 32:132-141. [PMID: 37652785 DOI: 10.1016/j.tim.2023.07.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 09/02/2023]
Abstract
Microbes are widely recognized to be vital to host health. This new consensus rests, in part, on experiments showing how hosts malfunction when microbes are removed. More and more microbial dependencies are being discovered, even in fundamental processes such as development, immunity, physiology, and behavior. But why do they exist? The default explanation is that microbes are beneficial; when hosts lose microbes, they also lose benefits. Here I call attention to evolutionary addiction, whereby a host trait evolves a need for microbes without having been improved by them. Evolutionary addiction should be considered when interpreting microbe-removal experiments, as it is a distinct and potentially common process. Further, it may have unique implications for the evolution and stability of host-microbe interactions.
Collapse
Affiliation(s)
- Tobin J Hammer
- Department of Ecology and Evolutionary Biology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
47
|
Li J, Li Y, Xiao H, Li W, Ye F, Wang L, Li Y, Wang C, Wu Y, Xuan R, Huang Y, Huang J. The intestinal microflora diversity of aboriginal chickens in Jiangxi province, China. Poult Sci 2024; 103:103198. [PMID: 38016408 PMCID: PMC10696398 DOI: 10.1016/j.psj.2023.103198] [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: 07/21/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 11/30/2023] Open
Abstract
Intestinal microbiota can coevolve with host to form symbiotic relationship and be participated in the regulation of host physiological function. At present, there is no clear explanation on the effect of intestinal microflora in Jiangxi aboriginal chickens. Here, we investigated the association between gut microbiota and host genome of Jiangxi local chickens using 16S rRNA sequencing and genome-wide association studies (GWAS). The results showed that the breeds and genders had important effects on the intestinal microbiota of chickens. A total of 28 SNPs in 14 regions of the chicken genome were related to the relative abundance of microorganisms in 5 genera: Clostridium_sensu_stricto_1, Enterococcus, Gallibacterium, Turicibacter, and Rikenellaceae_RC9_gut_group. A total of 17 candidate genes were identified composition of chicken microbiome and show an association between the host genome and the chicken intestinal microbiota, which also unveiled the diversity of intestinal microbes in Jiangxi chickens. Given the correlation between chicken genome and intestinal microbe found in the present study, a new idea for the protection of aboriginal chicken genetic resources in China could be provided.
Collapse
Affiliation(s)
- Jiawei Li
- College of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Yuping Li
- College of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Huiyuan Xiao
- College of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Wen Li
- Changsheng Town People's Government of Ningdu County, Ganzhou, China
| | - Fengchun Ye
- Jiangxi Yifeng County Qiaoxi Veterinary Station, Yichun, China
| | - Liping Wang
- College of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Yuhang Li
- College of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Cong Wang
- College of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Yan Wu
- College of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Rui Xuan
- College of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Yuxuan Huang
- College of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Jianhua Huang
- College of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China.
| |
Collapse
|
48
|
Parks M, Lee JS, Camua K, Hollender E. Turtle species and ecology drive carapace microbiome diversity in three seasonally interconnected wetland habitats. Access Microbiol 2024; 6:000682.v3. [PMID: 38361649 PMCID: PMC10866032 DOI: 10.1099/acmi.0.000682.v3] [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: 08/01/2023] [Accepted: 12/22/2023] [Indexed: 02/17/2024] Open
Abstract
Different species of freshwater turtles exhibit primary behaviours ranging from aerial basking to benthic bottom-walking, cycle between wet and dry conditions at different time intervals, and undertake short-distance overland movements between aquatic habitats. These behaviours in turn may impact the accumulation of microbes on external shell surfaces of turtles and provide novel niches for differentiation of microbial communities. We assessed microbial diversity using 16S and 18S rRNA metabarcoding on carapace surfaces of six species of freshwater turtles residing in three adjacent and seasonally interconnected wetland habitats in southeast Oklahoma (United States). Communities were highly diverse, with nearly 4200 prokaryotic and 500 micro-eukaryotic amplicon sequence variants recovered, and included taxa previously reported as common or differentially abundant on turtle shells. The 16S rRNA alpha diversity tended to be highest for two species of benthic turtles, while 18S rRNA alpha diversity was highest for two basking and one shallow-water benthic species. Beta diversity of communities was more strongly differentiated by turtle species than by collection site, and ordination patterns were largely reflective of turtle species' primary habits (i.e. benthic, basking, or benthic-basking). Our data support that freshwater turtles could play a role in microbial ecology and evolution in freshwater habitats and warrant additional exploration including in areas with high native turtle diversity and inter-habitat turtle movements.
Collapse
Affiliation(s)
- Matthew Parks
- Department of Biology, University of Central Oklahoma, 100 N University Drive, Edmond, Oklahoma 73034, USA
| | - Jun Sheng Lee
- Department of Biology, University of Central Oklahoma, 100 N University Drive, Edmond, Oklahoma 73034, USA
- DNA Reference Lab, 5282 Medical Dr. Suite 312, San Antonio, Texas 78229, USA
| | - Kassandra Camua
- Department of Biology, University of Central Oklahoma, 100 N University Drive, Edmond, Oklahoma 73034, USA
| | - Ethan Hollender
- Department of Biological Sciences, 601 Science Engineering Hall, University of Arkansas, Fayetteville, Arkansas 72701, USA
| |
Collapse
|
49
|
Zhang B, Jiang X, Yu Y, Cui Y, Wang W, Luo H, Stergiadis S, Wang B. Rumen microbiome-driven insight into bile acid metabolism and host metabolic regulation. THE ISME JOURNAL 2024; 18:wrae098. [PMID: 38836500 PMCID: PMC11193847 DOI: 10.1093/ismejo/wrae098] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/20/2024] [Accepted: 06/04/2024] [Indexed: 06/06/2024]
Abstract
Gut microbes play a crucial role in transforming primary bile acids (BAs) into secondary forms, which influence systemic metabolic processes. The rumen, a distinctive and critical microbial habitat in ruminants, boasts a diverse array of microbial species with multifaceted metabolic capabilities. There remains a gap in our understanding of BA metabolism within this ecosystem. Herein, through the analysis of 9371 metagenome-assembled genomes and 329 cultured organisms from the rumen, we identified two enzymes integral to BA metabolism: 3-dehydro-bile acid delta4,6-reductase (baiN) and the bile acid:Na + symporter family (BASS). Both in vitro and in vivo experiments were employed by introducing exogenous BAs. We revealed a transformation of BAs in rumen and found an enzyme cluster, including L-ribulose-5-phosphate 3-epimerase and dihydroorotate dehydrogenase. This cluster, distinct from the previously known BA-inducible operon responsible for 7α-dehydroxylation, suggests a previously unrecognized pathway potentially converting primary BAs into secondary BAs. Moreover, our in vivo experiments indicated that microbial BA administration in the rumen can modulate amino acid and lipid metabolism, with systemic impacts underscored by core secondary BAs and their metabolites. Our study provides insights into the rumen microbiome's role in BA metabolism, revealing a complex microbial pathway for BA biotransformation and its subsequent effect on host metabolic pathways, including those for glucose, amino acids, and lipids. This research not only advances our understanding of microbial BA metabolism but also underscores its wider implications for metabolic regulation, offering opportunities for improving animal and potentially human health.
Collapse
Affiliation(s)
- Boyan Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - Xianzhe Jiang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - Yue Yu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - Yimeng Cui
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - Wei Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - Hailing Luo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - Sokratis Stergiadis
- Department of Animal Sciences, School of Agriculture Policy and Development, University of Reading, Reading RG6 6EU, United Kingdom
| | - Bing Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| |
Collapse
|
50
|
Ahn JS, Koo BC, Choi YJ, Jung WW, Kim HS, Lee SJ, Hong ST, Chung HJ. Identification of Muscle Strength-Related Gut Microbes through Human Fecal Microbiome Transplantation. Int J Mol Sci 2024; 25:662. [PMID: 38203833 PMCID: PMC10779158 DOI: 10.3390/ijms25010662] [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: 12/01/2023] [Revised: 12/28/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024] Open
Abstract
The gut microbiome is well known for its influence on human physiology and aging. Therefore, we speculate that the gut microbiome may affect muscle strength in the same way as the host's own genes. To demonstrate candidates for gut microbes affecting muscle strength, we remodeled the original gut microbiome of mice into human intestinal microbiome through fecal microbiome transplantation (FMT), using human feces and compared the changes in muscle strength in the same mice before and three months after FMT. After comparing before and after FMT, the mice were divided into three groups based on the observed changes in muscle strength: positive, none, and negative changes in muscle strength. As a result of analyzing the α-diversity, β-diversity, and co-occurrence network of the intestinal microbial community before and after FMT, it was observed that a more diverse intestinal microbial community was established after FMT in all groups. In particular, the group with increased muscle strength had more gut microbiome species and communities than the other groups. Fold-change comparison showed that Eisenbergiella massiliensis and Anaeroplasma abactoclasticum from the gut microbiome had positive contributions to muscle strength, while Ileibacterium valens and Ethanoligenens harbinense had negative effects. This study identifies candidates for the gut microbiome that contribute positively and those that contribute negatively to muscle strength.
Collapse
Affiliation(s)
- Ji-Seon Ahn
- Gwangju Center, Korea Basic Science Institute, Gwangju 61751, Jeolla, Republic of Korea; (J.-S.A.); (Y.-J.C.)
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Jeonbuk, Republic of Korea
| | - Bon-Chul Koo
- Division of Bioconvergence Analysis, Korea Basic Science Institute, Ochang 28119, Chungbuk, Republic of Korea;
- Department of Biomedical Laboratory Science, Cheongju University, Cheongju 28503, Chungbuk, Republic of Korea; (W.-W.J.); (H.-S.K.); (S.-J.L.)
| | - Yu-Jin Choi
- Gwangju Center, Korea Basic Science Institute, Gwangju 61751, Jeolla, Republic of Korea; (J.-S.A.); (Y.-J.C.)
| | - Woon-Won Jung
- Department of Biomedical Laboratory Science, Cheongju University, Cheongju 28503, Chungbuk, Republic of Korea; (W.-W.J.); (H.-S.K.); (S.-J.L.)
| | - Hyun-Sook Kim
- Department of Biomedical Laboratory Science, Cheongju University, Cheongju 28503, Chungbuk, Republic of Korea; (W.-W.J.); (H.-S.K.); (S.-J.L.)
| | - Suk-Jun Lee
- Department of Biomedical Laboratory Science, Cheongju University, Cheongju 28503, Chungbuk, Republic of Korea; (W.-W.J.); (H.-S.K.); (S.-J.L.)
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Jeonbuk, Republic of Korea
| | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju 61751, Jeolla, Republic of Korea; (J.-S.A.); (Y.-J.C.)
| |
Collapse
|