1
|
Zheng XQ, Wang DB, Jiang YR, Song CL. Gut microbiota and microbial metabolites for osteoporosis. Gut Microbes 2025; 17:2437247. [PMID: 39690861 DOI: 10.1080/19490976.2024.2437247] [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: 07/11/2024] [Revised: 11/13/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Osteoporosis is an age-related bone metabolic disease. As an essential endocrine organ, the skeletal system is intricately connected with extraosseous organs. The crosstalk between bones and other organs supports this view. In recent years, the link between the gut microecology and bone metabolism has become an important research topic, both in preclinical studies and in clinical trials. Many studies have shown that skeletal changes are accompanied by changes in the composition and structure of the gut microbiota (GM). At the same time, natural or artificial interventions targeting the GM can subsequently affect bone metabolism. Moreover, microbiome-related metabolites may have important effects on bone metabolism. We aim to review the relationships among the GM, microbial metabolites, and bone metabolism and to summarize the potential mechanisms involved and the theory of the gut‒bone axis. We also describe existing bottlenecks in laboratory studies, as well as existing challenges in clinical settings, and propose possible future research directions.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Ding-Ben Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Yi-Rong Jiang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| |
Collapse
|
2
|
Gillingham MAF, Prüter H, Montero BK, Kempenaers B. The costs and benefits of a dynamic host microbiome. Trends Ecol Evol 2025; 40:255-272. [PMID: 39690056 DOI: 10.1016/j.tree.2024.11.008] [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] [Received: 06/03/2024] [Revised: 10/31/2024] [Accepted: 11/11/2024] [Indexed: 12/19/2024]
Abstract
All species host a rich community of microbes. This microbiome is dynamic, and displays seasonal, daily, and even hourly changes, but also needs to be resilient to fulfill important roles for the host. In evolutionary ecology, the focus of microbiome dynamism has been on how it can facilitate host adaptation to novel environments. However, an hitherto largely overlooked issue is that the host needs to keep its microbiome in check, which is costly and leads to trade-offs with investing in other fitness-related traits. Investigating these trade-offs in natural vertebrate systems by collecting longitudinal data will lead to deeper insight into the evolutionary mechanisms that shape host-microbiome interactions.
Collapse
Affiliation(s)
- Mark A F Gillingham
- Department of Ornithology, Max Planck Institute for Biological Intelligence, Eberhard Gwinner Straße, 82319 Seewiesen, Germany.
| | - Hanna Prüter
- Department of Ornithology, Max Planck Institute for Biological Intelligence, Eberhard Gwinner Straße, 82319 Seewiesen, Germany
| | - B Karina Montero
- Biodiversity Research Institute, Consejo Superior de Investigaciones Científicas (CSIC) and Oviedo University-Principality of Asturias, University of Oviedo, Campus of Mieres, Mieres E-33600, Spain
| | - Bart Kempenaers
- Department of Ornithology, Max Planck Institute for Biological Intelligence, Eberhard Gwinner Straße, 82319 Seewiesen, Germany
| |
Collapse
|
3
|
Wang Z, Wang L, Guo C, Wang Z, Lun X, Ji H, Shang M, Wang X, Liu Q. Effects of Different Levels of Flea Infestation on Gut Microbiota of Brandt's Voles ( Lasiopodomys brandtii) in China. Animals (Basel) 2025; 15:669. [PMID: 40075951 PMCID: PMC11899220 DOI: 10.3390/ani15050669] [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: 01/29/2025] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Brandt's vole is a common small rodent, and its gut microbiota is critical to host health and immune function. The parasitic fleas commonly found in Brandt's voles cause an immune response, but their impact on the gut microbiota remains unclear. According to the level of flea infestation, Brandt's voles were divided into the control group, low-infestation group, and high-infestation group. The changes in the microbial community composition, abundance, and diversity of the gut microbiota were evaluated using 16S rRNA sequencing. Flea infestation significantly affected body weight, food intake, and gut microbiota structure. The low-infestation group exhibited the most pronounced changes in weight and food intake, while the high-infestation group showed the least. In the 4th week, 16S rRNA sequencing revealed an increase in alpha diversity and alterations in microbial composition. Beta-diversity analysis indicated significant differences in the intestinal microbiota between the experimental groups and the control group. By the 8th week, these differences had diminished, suggesting that the microbiota had stabilized or recovered over time. Overall, parasitic flea infestation significantly alters the diversity, structure, and characteristic microbial enrichment of the gut microbiota in Brandt's voles, potentially impacting host metabolism, immunity, and growth. While this study lasted 8 weeks, the long-term health effects of flea infestation may persist. Future research should elucidate the interaction mechanisms between parasites and hosts, define the time frames and mechanisms of these long-term impacts, and provide theoretical support for animal health management and disease control.
Collapse
Affiliation(s)
- Zhenxu Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (Z.W.); (X.L.)
| | - Lu Wang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (L.W.); (H.J.); (M.S.); (X.W.)
| | - Chenran Guo
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (C.G.); (Z.W.)
| | - Zihao Wang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (C.G.); (Z.W.)
| | - Xinchang Lun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (Z.W.); (X.L.)
| | - Haoqiang Ji
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (L.W.); (H.J.); (M.S.); (X.W.)
| | - Meng Shang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (L.W.); (H.J.); (M.S.); (X.W.)
| | - Xiaoxu Wang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (L.W.); (H.J.); (M.S.); (X.W.)
| | - Qiyong Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (Z.W.); (X.L.)
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (L.W.); (H.J.); (M.S.); (X.W.)
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (C.G.); (Z.W.)
| |
Collapse
|
4
|
Koppula S, Wankhede N, Kyada A, Ballal S, Arya R, Singh AK, Gulati M, Sute A, Sarode S, Polshettiwar S, Marde V, Taksande B, Upaganlawar A, Fareed M, Umekar M, Kopalli SR, Kale M. The gut-brain axis: Unveiling the impact of xenobiotics on neurological health and disorders. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111237. [PMID: 39732317 DOI: 10.1016/j.pnpbp.2024.111237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 11/12/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
The Gut-Brain Axis (GBA) is a crucial link between the gut microbiota and the central nervous system. Xenobiotics, originating from diverse sources, play a significant role in shaping this interaction. This review examines how these compounds influence neurotransmitter dynamics within the GBA. Environmental pollutants can disrupt microbial populations, impacting neurotransmitter synthesis-especially serotonin, gamma-aminobutyric acid (GABA), and dopamine pathways. Such disruptions affect mood regulation, cognition, and overall neurological function. Xenobiotics also contribute to the pathophysiology of neurological disorders, with changes in serotonin levels linked to mood disorders and imbalances in GABA and dopamine associated with anxiety, stress, and reward pathway disorders. These alterations extend beyond the GBA, leading to complications in neurological health, including increased risk of neurodegenerative diseases due to neuroinflammation triggered by neurotransmitter imbalances. This review provides a comprehensive overview of how xenobiotics influence the GBA and their implications for neurological well-being.
Collapse
Affiliation(s)
- Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea
| | - Nitu Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, -360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | | | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Astha Sute
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Sanskruti Sarode
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Shruti Polshettiwar
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Vaibhav Marde
- Indian Institute of Technology (IIT), Hyderabad, Telangana 502284, India
| | - Brijesh Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Aman Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 11597, Saudi Arabia
| | - Milind Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Mayur Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| |
Collapse
|
5
|
Rollins RL, Griffin CD, Cowie RH. Snail coprophagy: the encounter filter, food preferences, and rat lungworm (Angiostrongylus cantonensis) prevalence. Parasite 2024; 31:76. [PMID: 39715443 DOI: 10.1051/parasite/2024075] [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: 09/09/2024] [Accepted: 12/05/2024] [Indexed: 12/25/2024] Open
Abstract
Understanding the factors driving infection prevalence among host species is crucial for effective disease mitigation. Angiostrongylus cantonensis, the rat lungworm, causes neuroangiostrongyliasis and serves as an excellent model for studying infection dynamics across hosts. This study investigates the relative impact of encounter rates on A. cantonensis prevalence in snail hosts by assessing their coprophagic tendencies. Multiple-choice feeding assays were conducted with four snail species (Parmarion martensi, Laevicaulis alte, Lissachatina fulica, and Veronicella cubensis) differing in A. cantonensis prevalence. The snails were offered romaine lettuce, hibiscus flowers, papaya, and rat feces. The relative intake ratios (RIR) were calculated and used to evaluate 1) feces preference among the snail species, and 2) correlation between feces preference and A. cantonensis prevalence. We also compared preferences for feces from rats fed high-fat and balanced diets; no significant difference was observed. Feces made up the highest proportion of the diet of P. martensi (11.6%), followed by V. cubensis (7.8%), L. fulica (5.9%), and L. alte (5.1%). Additionally, P. martensi showed a significantly higher preference (RIR) than all other species. The correlation between feces preference and A. cantonensis prevalence among species was weakly positive. These findings suggest that the level of coprophagy influences encounter rates with A. cantonensis, contributing to variation in infection prevalence among snail species. However, other factors may also play a role, as preference and prevalence were only weakly correlated. Understanding these dynamics can inform strategies for managing the spread of A. cantonensis and mitigating its health impacts.
Collapse
Affiliation(s)
- Randi L Rollins
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Chasen D Griffin
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA - School of Life Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Robert H Cowie
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| |
Collapse
|
6
|
Gul S, Shi Y, Hu J, Song S. The Influence of Microbiota on Wild Birds' Parental Coprophagy Behavior: Current Advances and Future Research Directions. Microorganisms 2024; 12:2468. [PMID: 39770671 PMCID: PMC11677090 DOI: 10.3390/microorganisms12122468] [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: 10/22/2024] [Revised: 11/20/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
This comprehensive review provides an in-depth exploration of the intriguing phenomenon of parental coprophagy in wild birds and its profound implications on the influence of adult avian parents' health. This review investigates the composition and dynamics of avian feces' microbiota, casting light on the various dietary, environmental, and genetic factors that influence its diversity. Furthermore, it emphasizes parental coprophagy, a behavior observed in numerous bird species, particularly among herbivorous and passerine birds. The review investigates multiple hypotheses proposed to explain the occurrence of coprophagy. It delves into its function as a potential mechanism for transmitting microorganisms, particularly feces bacteria, from nestlings to their parents. This microbial transfer may affect the health and well-being of adult avian parents. In addition, the review highlights the current research deficits and debates surrounding coprophagy. These gaps include crucial aspects such as the onset of coprophagy, its long-term effects on both parents and offspring, the nutritional implications of consuming nestling feces, the potential risks of pathogen transmission, and the ecological and evolutionary factors that drive this behavior. As the review synthesizes existing knowledge and identifies areas requiring additional research, it emphasizes the significance of future studies that comprehensively address these gaps. By doing so, we can understand coprophagy's ecological and evolutionary significance in wild birds, advancing our knowledge on avian biology. This information can improve conservation efforts to protect migratory bird populations and their complex ecosystems.
Collapse
Affiliation(s)
- Saba Gul
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (S.G.); (Y.S.); (J.H.)
| | - Yurou Shi
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (S.G.); (Y.S.); (J.H.)
| | - Jie Hu
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (S.G.); (Y.S.); (J.H.)
- Institute of Environmental Sciences, Leiden University, 2333CC Leiden, The Netherlands
| | - Sen Song
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (S.G.); (Y.S.); (J.H.)
| |
Collapse
|
7
|
Wen J, Li W, Bo T, Ding B, Zhang X, Wang D. Involvement of the gut microbiota in the metabolic phenotypes of two sympatric gerbils. Comp Biochem Physiol A Mol Integr Physiol 2024; 297:111710. [PMID: 39067809 DOI: 10.1016/j.cbpa.2024.111710] [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: 06/03/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Temporal niche partitioning is a crucial strategy for sympatric species to avoid predation and competition for habitat space and food resources. This study investigated the effect of the gut microbiota on the metabolic rhythms of two sympatric gerbil species (Meriones unguiculatus and Meriones meridianus) to test the hypothesis that the oscillatory patterns of microbiota may not fully mirror those of the host's metabolism. Experiment 1 compared the circadian metabolic and gut microbiota rhythms of M. unguiculatus (n = 12) and M. meridianus (n = 12) and measured the subjects' body temperatures and environmental temperature preferences. In Experiment 2.1, six M. meridianus gerbils were treated with antibiotics, and in Experiment 2.2, 21 M. unguiculatus gerbils (seven per treatment) were randomly gavaged with saline or a gut microbiota suspension from either M. unguiculatus or M. meridianus; their metabolic rhythms were subsequently measured. The results showed that the two gerbils had different metabolic phenotypes that determined activity heterogeneity and contributed to their coexistence. The relative abundances of Bacteroidetes, Actinobacteria, and Cyanobacteria in M. meridianus varied rhythmically in parallel with the daily metabolic rate, which was significantly higher at night than during the day. The rhythm of the metabolic rate was not noticeable in M. unguiculatus. However, in M.unguiculatus, the relative abundances of Firmicutes, Bacteroidetes, Proteobacteria, and Verrucomicrobia were significantly higher during the day than at night, while Cyanobacteria exhibited the opposite pattern. Antibiotic treatment significantly weakened the metabolic rhythms of M. meridianus, and the circadian rhythms slowly recovered after stopping antibiotic gavage. However, after transplanting M. meridianus' gut microbiota into M. unguiculatus, the metabolic rate of M. unguiculatus was not significantly different from that of the control groups. Our hypothesis was partly supported: the microbiota was only partially involved in regulating the metabolic rhythms of gerbils, and other factors could compensate for the effect of the gut microbiota on host metabolic rhythms. This finding underscores the complexity of host-microbiota interactions and highlights the need for further exploration into the multifaceted mechanisms governing host metabolic regulation.
Collapse
Affiliation(s)
- Jing Wen
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China; State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory for Water Environment and Marine Biological Resources Protection in Zhejiang Province, Wenzhou 325035, China
| | - Wenting Li
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China; Key Laboratory for Water Environment and Marine Biological Resources Protection in Zhejiang Province, Wenzhou 325035, China
| | - Tingbei Bo
- School of Grassland Science, Beijing Forestry University, Beijing 100083, China
| | - Boyang Ding
- Physical Education College, Hebei Normal University, Shijiazhuang 050010, China
| | - Xueying Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Dehua Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; School of Life Sciences, Shandong University, Qingdao 266237, China.
| |
Collapse
|
8
|
Walrath T, Najarro KM, Giesy LE, Khair S, Orlicky DJ, McMahan RH, Kovacs EJ. Reducing the excessive inflammation after burn injury in aged mice by maintaining a healthier intestinal microbiome. FASEB J 2024; 38:e70065. [PMID: 39305117 PMCID: PMC11465428 DOI: 10.1096/fj.202401020r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/09/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024]
Abstract
One in six people are projected to be 65 years or older by 2050. As the population ages, better treatments for injuries that disproportionately impact the aged population will be needed. Clinical studies show that people aged 65 and older experience higher rates of morbidity and mortality after burn injury, including a greater incidence of pulmonary complications when compared to younger burn injured adults, which we and others believe is mediated, in part, by inflammation originating in the intestines. Herein, we use our clinically relevant model of scald burn injury in young and aged mice to determine whether cohousing aged mice with young mice or giving aged mice oral gavage of fecal material from young mice is sufficient to alter the microbiome of the aged mice and protect them from inflammation in the ileum and the lungs. Aged burn injured mice have less DNA expression of Bacteroidetes in the feces and an unhealthy Firmicutes/Bacteroidetes ratio. Both Bacteroidetes and the ratio of these two phyla are restored in aged burn injured by prior cohousing for a month with younger mice but not fecal transfer from young mice. This shift in the microbiome coincides with heightened expression of danger-associated molecular patterns (DAMP), and pro-inflammatory cytokine interleukin-6 (il6) in the ileum and lung of aged, burn injured mice, and heightened antimicrobial peptide camp in the lung. Cohousing reverses DAMP expression in the ileum and lung, and cathelicidin-related antimicrobial peptide protein (camp) in the lung, while fecal transfer heightened DAMPs while reducing camp in the lung, and also increased IL-6 protein in the lungs. These results highlight the importance of the intestinal microbiome in mediating inflammation within the gut-lung axis, giving insights into potential future treatments in the clinic.
Collapse
Affiliation(s)
- Travis Walrath
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kevin M. Najarro
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lauren E. Giesy
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Shanawaj Khair
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David J. Orlicky
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rachel H. McMahan
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Elizabeth J. Kovacs
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Veterans Health Administration, Eastern Colorado Health Care System, Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, Colorado, USA
| |
Collapse
|
9
|
Lipowska MM, Sadowska ET, Kohl KD, Koteja P. Experimental Evolution of a Mammalian Holobiont? Genetic and Maternal Effects on the Cecal Microbiome in Bank Voles Selectively Bred for Herbivorous Capability. ECOLOGICAL AND EVOLUTIONARY PHYSIOLOGY 2024; 97:274-291. [PMID: 39680902 DOI: 10.1086/732781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
AbstractMammalian herbivory represents a complex adaptation requiring evolutionary changes across all levels of biological organization, from molecules to morphology to behavior. Explaining the evolution of such complex traits represents a major challenge in biology, as it is simultaneously muddled and enlightened by a growing awareness of the crucial role of symbiotic associations in shaping organismal adaptations. The concept of hologenomic evolution includes the partnered unit of the holobiont, the host with its microbiome, as a selection unit that may undergo adaptation. Here, we test some of the assumptions underlying the concept of hologenomic evolution using a unique experimental evolution model: lines of the bank vole (Myodes [=Clethrionomys] glareolus) selected for increased ability to cope with a low-quality herbivorous diet and unselected control lines. Results from a complex nature-nurture design, in which we combined cross-fostering between the selected and control lines with dietary treatment, showed that the herbivorous voles harbored a cecal microbiome with altered membership and structure and changed abundances of several phyla and genera regardless of the origin of their foster mothers. Although the differences were small, they were statistically significant and partially robust to changes in diet and housing conditions. Microbial characteristics also correlated with selection-related traits at the level of individual variation. Thus, the results support the hypothesis that selection on a host performance trait leads to genetic changes in the host that promote the maintenance of a beneficial microbiome. Such a result is consistent with some of the assumptions underlying the concept of hologenomic evolution.
Collapse
|
10
|
Pieczyńska-Zając JM, Malinowska AM, Pruszyńska-Oszmałek E, Kołodziejski PA, Drzymała-Czyż S, Bajerska J. Effect of a high-fat high-fructose diet on the composition of the intestinal microbiota and its association with metabolic and anthropometric parameters in a letrozole-induced mouse model of polycystic ovary syndrome. Nutrition 2024; 124:112450. [PMID: 38669829 DOI: 10.1016/j.nut.2024.112450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/28/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024]
Abstract
OBJECTIVE It has been suggested that dysbiosis of the gut microbiota is associated with the pathogenesis of Polycystic Ovary Syndrome (PCOS), and that improper diet can aggravate these changes. This study thus aimed to investigate the effects of a high-fat/high-fructose (HF/HFr) diet on the gut microbial community and their metabolites in prepubertal female mice with letrozole (LET)-induced PCOS. We also tested the correlations between the relative abundance of microbial taxa and selected PCOS parameters. RESEARCH METHODS & PROCEDURES Thirty-two C57BL/6 mice were randomly divided into four groups (n = 8) and implanted with LET or a placebo, with simultaneous administration of a HF/HFr diet or standard diet (StD) for 5 wk. The blood and intestinal contents were collected after the sacrifice. RESULTS Placebo + HF/HFr and LET + HF/HFr had significantly higher microbial alpha diversity than either group fed StD. The LET-implanted mice fed StD had a significantly higher abundance of Prevotellaceae_UCG-001 than the placebo mice fed StD. Both groups fed the HF/HFr diet had significantly lower fecal levels of short-chain fatty acids than the placebo mice fed StD, while the LET + HF/HFr animals had significantly higher concentrations of lipopolysaccharides in blood serum than either the placebo or LET mice fed StD. Opposite correlations were observed between Turicibacter and Lactobacillus and the lipid profile, CONCLUSION: HF/HFr diet had a much stronger effect on the composition of the intestinal microbiota of prepubertal mice than LET itself.
Collapse
Affiliation(s)
| | - Anna Maria Malinowska
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Ewa Pruszyńska-Oszmałek
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, Poznań, Poland
| | - Paweł Antoni Kołodziejski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, Poznań, Poland
| | | | - Joanna Bajerska
- Department of Human Nutrition and Dietetics, Poznań University of Life Sciences, Poznań, Poland.
| |
Collapse
|
11
|
Jiang ST, Sun YH, Li Y, Wang MQ, Wang XY, Dong YF. Gut microbiota is necessary for pair-housing to protect against post-stroke depression in mice. Exp Neurol 2024; 378:114834. [PMID: 38789022 DOI: 10.1016/j.expneurol.2024.114834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/06/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
The goal of this study is to investigate the role of microbiota-gut-brain axis involved in the protective effect of pair-housing on post-stroke depression (PSD). PSD model was induced by occluding the middle cerebral artery (MCAO) plus restraint stress for four weeks. At three days after MCAO, the mice were restrained 2 h per day. For pair-housing (PH), each mouse was pair housed with a healthy isosexual cohabitor for four weeks. While in the other PH group, their drinking water was replaced with antibiotic water. On day 35 to day 40, anxiety- and depression-like behaviors (sucrose consumption, open field test, forced swim test, and tail-suspension test) were conducted. Results showed pair-housed mice had better performance on anxiety- and depression-like behaviors than the PSD mice, and the richness and diversity of intestinal flora were also improved. However, drinking antibiotic water reversed the effects of pair-housing. Furthermore, pair-housing had an obvious improvement in gut barrier disorder and inflammation caused by PSD. Particularly, they showed significant decreases in CD8 lymphocytes and mRNA levels of pro-inflammatory cytokines (TNF-a, IL-1β and IL-6), while IL-10 mRNA was upregulated. In addition, pair-housing significantly reduced activated microglia and increased Nissl's body in the hippocampus of PSD mice. However, all these improvements were worse in the pair-housed mice administrated with antibiotic water. We conclude that pair-housing significantly improves PSD in association with enhanced functions of microbiota-gut-brain axis, and homeostasis of gut microbiota is indispensable for the protective effect of pair-housing on PSD.
Collapse
Affiliation(s)
- Su-Ting Jiang
- Department of Medical Care, School of Nursing, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yao-Huan Sun
- Department of Medical Care, School of Nursing, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ya Li
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Meng-Qing Wang
- Department of Pathology and Pathophysiology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xu-Yang Wang
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Yin-Feng Dong
- Department of Pathology and Pathophysiology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
12
|
de Paiva IHR, da Silva RS, Mendonça IP, de Souza JRB, Peixoto CA. Semaglutide Attenuates Anxious and Depressive-Like Behaviors and Reverses the Cognitive Impairment in a Type 2 Diabetes Mellitus Mouse Model Via the Microbiota-Gut-Brain Axis. J Neuroimmune Pharmacol 2024; 19:36. [PMID: 39042202 DOI: 10.1007/s11481-024-10142-w] [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: 08/09/2023] [Accepted: 07/14/2024] [Indexed: 07/24/2024]
Abstract
Newly conducted research suggests that metabolic disorders, like diabetes and obesity, play a significant role as risk factors for psychiatric disorders. This connection presents a potential avenue for creating novel antidepressant medications by repurposing drugs originally developed to address antidiabetic conditions. Earlier investigations have shown that GLP-1 (Glucagon-like Peptide-1) analogs exhibit neuroprotective qualities in various models of neurological diseases, encompassing conditions such as Alzheimer's disease, Parkinson's disease, and stroke. Moreover, GLP-1 analogs have demonstrated the capability to enhance neurogenesis, a process recognized for its significance in memory formation and the cognitive and emotional aspects of information processing. Nonetheless, whether semaglutide holds efficacy as both an antidepressant and anxiolytic agent remains uncertain. To address this, our study focused on a mouse model of depression linked to type 2 diabetes induced by a High Fat Diet (HFD). In this model, we administered semaglutide (0.05 mg/Kg intraperitoneally) on a weekly basis to evaluate its potential as a therapeutic option for depression and anxiety. Diabetic mice had higher blood glucose, lipidic profile, and insulin resistance. Moreover, mice fed HFD showed higher serum interleukin (IL)-1β and lipopolysaccharide (LPS) associated with impaired humor and cognition. The analysis of behavioral responses revealed that the administration of semaglutide effectively mitigated depressive- and anxiety-like behaviors, concurrently demonstrating an enhancement in cognitive function. Additionally, semaglutide treatment protected synaptic plasticity and reversed the hippocampal neuroinflammation induced by HFD fed, improving activation of the insulin pathway, demonstrating the protective effects of semaglutide. We also found that semaglutide treatment decreased astrogliosis and microgliosis in the dentate gyrus region of the hippocampus. In addition, semaglutide prevented the DM2-induced impairments of pro-opiomelanocortin (POMC), and G-protein-coupled receptor 43 (GPR43) and simultaneously increased the NeuN + and Glucagon-like Peptide-1 receptor (GLP-1R+) neurons in the hippocampus. Our data also showed that semaglutide increased the serotonin (5-HT) and serotonin transporter (5-HTT) and glutamatergic receptors in the hippocampus. At last, semaglutide changed the gut microbiota profile (increasing Bacterioidetes, Bacteroides acidifaciens, and Blautia coccoides) and decreased leaky gut, improving the gut-brain axis. Taken together, semaglutide has the potential to act as a therapeutic tool for depression and anxiety.
Collapse
MESH Headings
- Animals
- Glucagon-Like Peptides/pharmacology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/psychology
- Diabetes Mellitus, Type 2/metabolism
- Mice
- Cognitive Dysfunction/drug therapy
- Cognitive Dysfunction/prevention & control
- Cognitive Dysfunction/etiology
- Cognitive Dysfunction/metabolism
- Depression/drug therapy
- Depression/psychology
- Depression/metabolism
- Male
- Anxiety/drug therapy
- Anxiety/psychology
- Anxiety/etiology
- Gastrointestinal Microbiome/drug effects
- Mice, Inbred C57BL
- Brain-Gut Axis/drug effects
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/psychology
- Diabetes Mellitus, Experimental/metabolism
- Disease Models, Animal
- Antidepressive Agents/pharmacology
- Antidepressive Agents/therapeutic use
Collapse
Affiliation(s)
- Igor Henrique Rodrigues de Paiva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Av. Moraes Rego s/n, Recife CEP, PE, 50670-420, Brazil.
- Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, PE, Brazil.
| | - Rodrigo Soares da Silva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Av. Moraes Rego s/n, Recife CEP, PE, 50670-420, Brazil
- Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | - Ingrid Prata Mendonça
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Av. Moraes Rego s/n, Recife CEP, PE, 50670-420, Brazil
- Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | | | - Christina Alves Peixoto
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Av. Moraes Rego s/n, Recife CEP, PE, 50670-420, Brazil.
- Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Recife, Brazil.
| |
Collapse
|
13
|
Wilde J, Slack E, Foster KR. Host control of the microbiome: Mechanisms, evolution, and disease. Science 2024; 385:eadi3338. [PMID: 39024451 DOI: 10.1126/science.adi3338] [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: 04/19/2023] [Accepted: 05/29/2024] [Indexed: 07/20/2024]
Abstract
Many species, including humans, host communities of symbiotic microbes. There is a vast literature on the ways these microbiomes affect hosts, but here we argue for an increased focus on how hosts affect their microbiomes. Hosts exert control over their symbionts through diverse mechanisms, including immunity, barrier function, physiological homeostasis, and transit. These mechanisms enable hosts to shape the ecology and evolution of microbiomes and generate natural selection for microbial traits that benefit the host. Our microbiomes result from a perpetual tension between host control and symbiont evolution, and we can leverage the host's evolved abilities to regulate the microbiota to prevent and treat disease. The study of host control will be central to our ability to both understand and manipulate microbiotas for better health.
Collapse
Affiliation(s)
- Jacob Wilde
- Department of Biology, University of Oxford, Oxford, UK
| | - Emma Slack
- Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Basel Institute for Child Health, Basel, Switzerland
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Kevin R Foster
- Department of Biology, University of Oxford, Oxford, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| |
Collapse
|
14
|
Chesworth R, Yim HCH, Watt G, El-Omar E, Karl T. Cannabidiol (CBD) facilitates cocaine extinction and ameliorates cocaine-induced changes to the gut microbiome in male C57BL/6JArc mice. Prog Neuropsychopharmacol Biol Psychiatry 2024; 133:111014. [PMID: 38649130 DOI: 10.1016/j.pnpbp.2024.111014] [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: 02/11/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
Cocaine use disorder (CUD) is a global health problem with no approved medications. One potential treatment target is the gut microbiome, but it is unknown if cocaine induces long-lasting effects on gut microbes. A novel therapeutic candidate for CUD, cannabidiol (CBD), can improve gut function in rodent models. It is possible that protective effects of CBD against cocaine use are mediated by improving gut health. We examined this question in this experiment. Cocaine conditioned place preference (CPP) was conducted in adult male C57BL/6JArc mice. Mice were treated with vehicle or 20 mg/kg CBD prior to all cocaine CPP sessions (N = 11-13/group). Mice were tested drug free 1, 14 and 28 days after cessation of cocaine and CBD treatment. Fecal samples were collected prior to drug treatment and after each test session. Gut microbiome analyses were conducted using 16 s rRNA sequencing and correlated with behavioural parameters. We found a persistent preference for a cocaine-environment in mice, and long-lasting changes to gut microbe alpha diversity. Cocaine caused persistent changes to beta diversity which lasted for 4 weeks. CBD treatment reduced cocaine-environment preference during abstinence from cocaine and returned gut beta diversity measures to control levels. CBD treatment increased the relative abundance of Firmicutes phyla and Oscillospira genus, but decreased Bacteroidetes phyla and Bacteroides acidifaciens species. Preference score in cocaine-treated mice was positively correlated with abundance of Actinobacteria, whereas in mice treated with CBD and cocaine, the preference score was negatively correlated with Tenericutes abundance. Here we show that CBD facilitates cocaine extinction memory and reverses persistent cocaine-induced changes to gut microbe diversity. Furthermore, CBD increases the abundance of gut microbes which have anti-inflammatory properties. This suggests that CBD may act via the gut to reduce the memory of cocaine reward. Our data suggest that improving gut health and using CBD could limit cocaine abuse.
Collapse
Affiliation(s)
- Rose Chesworth
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.
| | - Howard Chi-Ho Yim
- St George and Sutherland Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW, Sydney, Australia; UNSW Microbiome Research Centre, St George and Sutherland Clinical School, UNSW, Sydney, Australia; Department of Gastroenterology and Hepatology, St George Hospital, Sydney, Australia
| | - Georgia Watt
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Emad El-Omar
- St George and Sutherland Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW, Sydney, Australia; UNSW Microbiome Research Centre, St George and Sutherland Clinical School, UNSW, Sydney, Australia; Department of Gastroenterology and Hepatology, St George Hospital, Sydney, Australia
| | - Tim Karl
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|
15
|
Bornbusch SL, Power ML, Schulkin J, Drea CM, Maslanka MT, Muletz-Wolz CR. Integrating microbiome science and evolutionary medicine into animal health and conservation. Biol Rev Camb Philos Soc 2024; 99:458-477. [PMID: 37956701 DOI: 10.1111/brv.13030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023]
Abstract
Microbiome science has provided groundbreaking insights into human and animal health. Similarly, evolutionary medicine - the incorporation of eco-evolutionary concepts into primarily human medical theory and practice - is increasingly recognised for its novel perspectives on modern diseases. Studies of host-microbe relationships have been expanded beyond humans to include a wide range of animal taxa, adding new facets to our understanding of animal ecology, evolution, behaviour, and health. In this review, we propose that a broader application of evolutionary medicine, combined with microbiome science, can provide valuable and innovative perspectives on animal care and conservation. First, we draw on classic ecological principles, such as alternative stable states, to propose an eco-evolutionary framework for understanding variation in animal microbiomes and their role in animal health and wellbeing. With a focus on mammalian gut microbiomes, we apply this framework to populations of animals under human care, with particular relevance to the many animal species that suffer diseases linked to gut microbial dysfunction (e.g. gut distress and infection, autoimmune disorders, obesity). We discuss diet and microbial landscapes (i.e. the microbes in the animal's external environment), as two factors that are (i) proposed to represent evolutionary mismatches for captive animals, (ii) linked to gut microbiome structure and function, and (iii) potentially best understood from an evolutionary medicine perspective. Keeping within our evolutionary framework, we highlight the potential benefits - and pitfalls - of modern microbial therapies, such as pre- and probiotics, faecal microbiota transplants, and microbial rewilding. We discuss the limited, yet growing, empirical evidence for the use of microbial therapies to modulate animal gut microbiomes beneficially. Interspersed throughout, we propose 12 actionable steps, grounded in evolutionary medicine, that can be applied to practical animal care and management. We encourage that these actionable steps be paired with integration of eco-evolutionary perspectives into our definitions of appropriate animal care standards. The evolutionary perspectives proposed herein may be best appreciated when applied to the broad diversity of species under human care, rather than when solely focused on humans. We urge animal care professionals, veterinarians, nutritionists, scientists, and others to collaborate on these efforts, allowing for simultaneous care of animal patients and the generation of valuable empirical data.
Collapse
Affiliation(s)
- Sally L Bornbusch
- Center for Conservation Genomics, Smithsonian's National Zoo and Conservation Biology Institute, 3001 Connecticut Ave. NW, Washington, DC, 20008, USA
- Department of Nutrition Science, Smithsonian's National Zoo and Conservation Biology Institute, 3001 Connecticut Ave. NW, Washington, DC, 20008, USA
| | - Michael L Power
- Center for Species Survival, Smithsonian's National Zoo and Conservation Biology Institute, Washington, 3001 Connecticut Ave. NW, Washington, DC, 20008, USA
| | - Jay Schulkin
- Department of Obstetrics & Gynecology, University of Washington School of Medicine, 1959 NE Pacific St., Box 356460, Seattle, WA, 98195, USA
| | - Christine M Drea
- Department of Evolutionary Anthropology, Duke University, 104 Biological Sciences, Campus Box 90383, Durham, NC, 27708, USA
| | - Michael T Maslanka
- Department of Nutrition Science, Smithsonian's National Zoo and Conservation Biology Institute, 3001 Connecticut Ave. NW, Washington, DC, 20008, USA
| | - Carly R Muletz-Wolz
- Center for Conservation Genomics, Smithsonian's National Zoo and Conservation Biology Institute, 3001 Connecticut Ave. NW, Washington, DC, 20008, USA
| |
Collapse
|
16
|
Li Z, He H, Chen M, Ni M, Guo C, Wan Z, Zhou J, Wang Z, Wang Y, Cai H, Li M, Sun H, Xu H. Novel mechanism of Clostridium butyricum alleviated coprophagy prevention-induced intestinal inflammation in rabbit. Int Immunopharmacol 2024; 130:111773. [PMID: 38430808 DOI: 10.1016/j.intimp.2024.111773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
As bacteria synthesize nutrients primarily in the cecum, coprophagy is indispensable for supplying rabbits with essential nutrients. Recent research has demonstrated its pivotal role in maintaining intestinal microbiota homeostasis and immune regulation in rabbits, although the specific mechanism remains unknown. Here, we used coprophagy prevention (CP) to investigate the effects of coprophagy on the cecum homeostasis and microbiota in New Zealand white rabbits. Furthermore, whether supplementation of Clostridium butyricum (C. butyricum) may alleviate the cecum inflammation and apoptosis caused by CP was also explored. Four groups were randomly assigned: control (Con), sham-coprophagy prevention (SCP), coprophagy prevention (CP), and CP and C. butyricum addition (CPCB). Compared to Con and SCP, CP augmented cecum inflammation and apoptosis, as well as bacterial adhesion to the cecal epithelial mucosa, while decreasing the expression of tight junction proteins (ZO-1, occluding, and claudin-1). The relative abundance of short-chain fatty acids (SCFAs)-producing bacteria was significantly decreased in the CP group. Inversely, there was an increase in the Firmicutes/Bacteroidetes ratio and the relative abundance of Christensenellaceae_R-7_group. Additionally, CP increased the levels of Flagellin, IFN-γ, TNF-a, and IL-1β in cecum contents and promoted the expression of TLR5/MyD88/NF-κB pathway in cecum tissues. However, the CPCB group showed significant improvements in all parameters compared to the CP group. Dietary C. butyricum supplementation significantly increased the production of SCFAs, particularly butyric acid, triggering anti-inflammatory, tissue repairing, and barrier-protective responses. Notably, CPCB effectively mitigated CP-induced apoptosis and inflammation. In summary, CP disrupts the cecum epithelial barrier and induces inflammation in New Zealand white rabbits, but these effects can be alleviated by C. butyricum supplementation. This process appears to be largely associated with the TLR5/MyD88/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhichao Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Hui He
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Mengjuan Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Mengke Ni
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Chaohui Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Zhiyi Wan
- College of Biological Sciences, China Agricultural University, No.2 Yuan Ming Yuan West Road, Beijing 100193, PR China
| | - Jianshe Zhou
- Institute of Fisheries Science, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa 850032, PR China
| | - Zhitong Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Yaling Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Hanfang Cai
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Ming Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China.
| | - HuiZeng Sun
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, PR China.
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, PR China.
| |
Collapse
|
17
|
Zhang L, Tang X, Fan C, Ren S, Cheng Q, Zhou H, Liu K, Jia S, Zhang Y. Dysbiosis of Gut Microbiome Aggravated Male Infertility in Captivity of Plateau Pika. Biomolecules 2024; 14:403. [PMID: 38672421 PMCID: PMC11047922 DOI: 10.3390/biom14040403] [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: 01/30/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Captivity is an important and efficient technique for rescuing endangered species. However, it induces infertility, and the underlying mechanism remains obscure. This study used the plateau pika (Ochotona curzoniae) as a model to integrate physiological, metagenomic, metabolomic, and transcriptome analyses and explore whether dysbiosis of the gut microbiota induced by artificial food exacerbates infertility in captive wild animals. Results revealed that captivity significantly decreased testosterone levels and the testicle weight/body weight ratio. RNA sequencing revealed abnormal gene expression profiles in the testicles of captive animals. The microbial α-diversity and Firmicutes/Bacteroidetes ratio were drastically decreased in the captivity group. Bacteroidetes and Muribaculaceae abundance notably increased in captive pikas. Metagenomic analysis revealed that the alteration of flora increased the capacity for carbohydrate degradation in captivity. The levels of microbe metabolites' short-chain fatty acids (SCFAs) were significantly high in the captive group. Increasing SCFAs influenced the immune response of captivity plateau pikas; pro-inflammatory cytokines were upregulated in captivity. The inflammation ultimately contributed to male infertility. In addition, a positive correlation was observed between Gastranaerophilales family abundance and testosterone concentration. Our results provide evidence for the interactions between artificial food, the gut microbiota, and male infertility in pikas and benefit the application of gut microbiota interference in threatened and endangered species.
Collapse
Affiliation(s)
- Liangzhi Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; (L.Z.); (X.T.); (C.F.); (S.R.); (Q.C.)
| | - Xianjiang Tang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; (L.Z.); (X.T.); (C.F.); (S.R.); (Q.C.)
| | - Chao Fan
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; (L.Z.); (X.T.); (C.F.); (S.R.); (Q.C.)
| | - Shi’en Ren
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; (L.Z.); (X.T.); (C.F.); (S.R.); (Q.C.)
| | - Qi Cheng
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; (L.Z.); (X.T.); (C.F.); (S.R.); (Q.C.)
| | - Huakun Zhou
- Key Laboratory of Restoration Ecology of Cold Area in Qinghai Province, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China;
| | - Kai Liu
- Qinghai Provincial Grassland Station, Xining 810008, China;
| | - Shangang Jia
- College of Grassland Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yanming Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; (L.Z.); (X.T.); (C.F.); (S.R.); (Q.C.)
| |
Collapse
|
18
|
Lu W, Jiang C, Chen Y, Lu Z, Xu X, Zhu L, Xi H, Ye G, Yan C, Chen J, Zhang J, Zuo L, Huang Q. Altered metabolome and microbiome associated with compromised intestinal barrier induced hepatic lipid metabolic disorder in mice after subacute and subchronic ozone exposure. ENVIRONMENT INTERNATIONAL 2024; 185:108559. [PMID: 38461778 DOI: 10.1016/j.envint.2024.108559] [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: 12/02/2023] [Revised: 02/05/2024] [Accepted: 03/04/2024] [Indexed: 03/12/2024]
Abstract
Exposure to ozone has been associated with metabolic disorders in humans, but the underlying mechanism remains unclear. In this study, the role of the gut-liver axis and the potential mechanism behind the metabolic disorder were investigated by histological examination, microbiome and metabolome approaches in mice during the subacute (4-week) and subchronic (12-week) exposure to 0.5 ppm and 2.5 ppm ozone. Ozone exposure resulted in slowed weight gain and reduced hepatic lipid contents in a dose-dependent manner. After exposure to ozone, the number of intestinal goblet cells decreased, while the number of tuft cells increased. Tight junction protein zonula occludens-1 (ZO-1) was significantly downregulated, and the apoptosis of epithelial cells increased with compensatory proliferation, indicating a compromised chemical and physical layer of the intestinal barrier. The hepatic and cecal metabolic profiles were altered, primarily related to lipid metabolism and oxidative stress. The abundance of Muribaculaceae increased dose-dependently in both colon and cecum, and was associated with the decrease of metabolites such as bile acids, betaine, and L-carnitine, which subsequently disrupted the intestinal barrier and lipid metabolism. Overall, this study found that subacute and subchronic exposure to ozone induced metabolic disorder via disturbing the gut-liver axis, especially the intestinal barrier. These findings provide new mechanistic understanding of the health risks associated with environmental ozone exposure and other oxidative stressors.
Collapse
Affiliation(s)
- Wenjia Lu
- Xiamen Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chonggui Jiang
- Innovation and Entrepreneurship Laboratory for college students, Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Yajie Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Zhonghua Lu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xueli Xu
- Xiamen Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liting Zhu
- Xiamen Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haotong Xi
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Guozhu Ye
- Xiamen Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Changzhou Yan
- Xiamen Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Jinsheng Chen
- Center for Excellence in Regional Atmospheric Environment, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Jie Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Li Zuo
- Innovation and Entrepreneurship Laboratory for college students, Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei 230032, China.
| | - Qiansheng Huang
- Xiamen Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China; National Basic Science Data Center, Beijing 100190, China.
| |
Collapse
|
19
|
Xiao Y, Feng J, Jia J, Li J, Zhou Y, Song Z, Guan F, Li X, Liu L. Vitamin K1 ameliorates lipopolysaccharide-triggered skeletal muscle damage revealed by faecal bacteria transplantation. J Cachexia Sarcopenia Muscle 2024; 15:81-97. [PMID: 38018317 PMCID: PMC10834346 DOI: 10.1002/jcsm.13379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 08/03/2023] [Accepted: 09/25/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Sepsis-associated muscle weakness is common in patients of intensive care units (ICUs), and it is closely associated with poor outcomes. The mechanism of sepsis-induced muscle weakness is unclear. Recent studies have found that gut microbiota and metabolites are involved in the regulation of skeletal muscle mass and metabolism. This study aimed to investigate the effects of gut microbiota and metabolites on sepsis-associated muscle weakness. METHODS In a lipopolysaccharide (LPS)-induced inflammation mouse model, mice with different sensitivities to LPS-induced inflammation were considered as donor mice for the faecal microbiota transplantation (FMT) assay, and recipient mice were divided into sensitive (Sen) and resistant (Res) groups. Skeletal muscle mass and function, as well as colonic barrier integrity were tested and gut microbiota and metabolite composition were analysed in both groups of mice. The effect of intestinal differential metabolite vitamin K1 on LPS-triggered muscle damage was investigated, and the underlying mechanism was explored. RESULTS Recipients exhibited varying LPS-triggered muscle damage and intestinal barrier disruption. Tibialis anterior (TA) muscle of Sen exhibited upregulated expression levels of MuRF-1 (0.825 ± 0.063 vs. 0.304 ± 0.293, P = 0.0141) and MAFbx (1.055 ± 0.079 vs. 0.456 ± 0.3, P = 0.0092). Colonic tight junction proteins ZO-1 (0.550 ± 0.087 vs. 0.842 ± 0.094, P = 0.0492) and occludin (0.284 ± 0.057 vs. 0.664 ± 0.191, P = 0.0487) were significantly downregulated in the Sen group. Metabolomic analysis showed significantly higher vitamin K1 in the faeces (P = 0.0195) and serum of the Res group (P = 0.0079) than those of the Sen group. After vitamin K1 intervention, muscle atrophy-related protein expression downregulated (P < 0.05). Meanwhile SIRT1 protein expression were upregulated (0.320 ± 0.035 vs. 0.685 ± 0.081, P = 0.0281) and pNF-κB protein expression were downregulated (0.815 ± 0.295 vs. 0.258 ± 0.130, P = 0.0308). PI3K (0.365 ± 0.142 vs. 0.763 ± 0.013, P = 0.0475), pAKT (0.493 ± 0.159 vs. 1.183 ± 0.344, P = 0.0254) and pmTOR (0.509 ± 0.088 vs. 1.110 ± 0.190, P = 0.0368) protein expression levels were upregulated in TA muscle. Meanwhile, vitamin K1 attenuated serum inflammatory factor levels. CONCLUSIONS Vitamin K1 might ameliorate LPS-triggered skeletal muscle damage by antagonizing NF-κB-mediated inflammation through upregulation of SIRT1 and regulating the balance between protein synthesis and catabolism.
Collapse
Affiliation(s)
- Yuru Xiao
- Department of AnesthesiologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Anesthesiology and Critical Care Medicine Key Laboratory of LuzhouSouthwest Medical UniversityLuzhouChina
| | - Jianguo Feng
- Department of AnesthesiologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Anesthesiology and Critical Care Medicine Key Laboratory of LuzhouSouthwest Medical UniversityLuzhouChina
| | - Jing Jia
- Department of AnesthesiologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Anesthesiology and Critical Care Medicine Key Laboratory of LuzhouSouthwest Medical UniversityLuzhouChina
| | - Jie Li
- Department of AnesthesiologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Yingshun Zhou
- Laboratory of Pathogen and MicrobiologySouthwest Medical UniversityLuzhouChina
| | - Zhangyong Song
- Department of Pathogenic BiologySouthwest Medical UniversityLuzhouChina
| | - Fasheng Guan
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Xuexin Li
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Li Liu
- Department of AnesthesiologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
20
|
Shankar A, Deal CK, McCahon S, Callegari K, Seitz T, Yan L, Drown DM, Williams CT. SAD rats: Effects of short photoperiod and carbohydrate consumption on sleep, liver steatosis, and the gut microbiome in diurnal grass rats. Chronobiol Int 2024; 41:93-104. [PMID: 38047486 PMCID: PMC10843721 DOI: 10.1080/07420528.2023.2288223] [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: 08/16/2023] [Revised: 10/24/2023] [Accepted: 11/22/2023] [Indexed: 12/05/2023]
Abstract
Seasonal affective disorder (SAD) is a recurrent depression triggered by exposure to short photoperiods, with a subset of patients reporting hypersomnia, increased appetite, and carbohydrate craving. Dysfunction of the microbiota - gut - brain axis is frequently associated with depressive disorders, but its role in SAD is unknown. Nile grass rats (Arvicanthis niloticus) are potentially useful for exploring the pathophysiology of SAD, as they are diurnal and have been found to exhibit anhedonia and affective-like behavior in response to short photoperiods. Further, given grass rats have been found to spontaneously develop metabolic syndrome, they may be particularly susceptible to environmental triggers of metabolic dysbiosis. We conducted a 2 × 2 factorial design experiment to test the effects of short photoperiod (4 h:20 h Light:Dark (LD) vs. neutral 12:12 LD), access to a high concentration (8%) sucrose solution, and the interaction between the two, on activity, sleep, liver steatosis, and the gut microbiome of grass rats. We found that animals on short photoperiods maintained robust diel rhythms and similar subjective day lengths as controls in neutral photoperiods but showed disrupted activity and sleep patterns (i.e. a return to sleep after an initial bout of activity that occurs ~ 13 h before lights off). We found no evidence that photoperiod influenced sucrose consumption. By the end of the experiment, some grass rats were overweight and exhibited signs of non-alcoholic fatty liver disease (NAFLD) with micro- and macro-steatosis. However, neither photoperiod nor access to sucrose solution significantly affected the degree of liver steatosis. The gut microbiome of grass rats varied substantially among individuals, but most variation was attributable to parental effects and the microbiome was unaffected by photoperiod or access to sucrose. Our study indicates short photoperiod leads to disrupted activity and sleep in grass rats but does not impact sucrose consumption or exacerbate metabolic dysbiosis and NAFLD.
Collapse
Affiliation(s)
- Anusha Shankar
- Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks AK 99775, USA
- Current: Lab of Ornithology, Cornell University, Ithaca, NY 14850, USA
| | - Cole K. Deal
- Department of Biology, Colorado State University, Fort Collins, CO 80526, USA
| | - Shelby McCahon
- Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks AK 99775, USA
| | - Kyle Callegari
- Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks AK 99775, USA
| | - Taylor Seitz
- Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks AK 99775, USA
| | - Lily Yan
- Department of Psychology, Michigan State University, East Lansing, MI 48824, USA
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| | - Devin M. Drown
- Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks AK 99775, USA
- Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks AK 99775, USA
| | - Cory T. Williams
- Department of Biology, Colorado State University, Fort Collins, CO 80526, USA
| |
Collapse
|
21
|
Li Z, Chen M, Zhang R, Wang Z, He H, Wan Z, Li H, Cai H, Chen Z, Li M, Xu H. Clostridium butyricum Ameliorates the Effect of Coprophagy Prevention on Hepatic Lipid Synthesis in Rabbits via the Gut-Liver Axis. Int J Mol Sci 2023; 24:17554. [PMID: 38139382 PMCID: PMC10744194 DOI: 10.3390/ijms242417554] [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: 11/05/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Coprophagy prevention (CP) affects the growth performance, hepatic lipid synthesis, and gut microbiota in rabbits. Supplementation with Clostridium butyricum (C. butyricum, Strain number: CCTCC M 2019962) has been found to improve growth performance in rabbits. However, it remains unknown whether C. butyricum can ameliorate the effects of CP on hepatic lipid synthesis and the underlying mechanisms are yet to be elucidated. Therefore, this study aimed to investigate the impact of CP on hepatic lipid synthesis and the underlying mechanism based on the gut-liver axis. The findings revealed that supplementation with C. butyricum could reverse CP-related growth performance, lipid accumulation, bile acid synthesis, and inflammation. Furthermore, C. butyricum exerted protective effects on the gut by preserving intestinal barrier integrity and modulating gut microbiota composition; these factors may represent potential mechanisms through which C. butyricum improves CP-related outcomes. Specifically, C. butyricum reshaped the microbiota by increasing butyric acid levels, thereby maintaining secondary bile acid (deoxycholic acid, chenodeoxycholic acid) balance and attenuating the inhibitory effects of the FXR/SHP pathway on lipid synthesis (SREBP1c/ApoA1). Moreover, the activation of butyrate/GPR43pathway by C. butyricum reduced damage to the intestinal barrier (ZO-1/Occludin/Claudin1) and restored the gut immune microenvironment in CP rabbits. In summary, supplementation with C. butyricum can alleviate the adverse effects of CP on growth performance and hepatic lipid synthesis by modulating the gut-liver axis.
Collapse
Affiliation(s)
- Zhichao Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Z.L.); (M.C.); (R.Z.); (Z.W.); (H.H.); (H.L.); (H.C.)
| | - Mengjuan Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Z.L.); (M.C.); (R.Z.); (Z.W.); (H.H.); (H.L.); (H.C.)
| | - Ran Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Z.L.); (M.C.); (R.Z.); (Z.W.); (H.H.); (H.L.); (H.C.)
| | - Zhitong Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Z.L.); (M.C.); (R.Z.); (Z.W.); (H.H.); (H.L.); (H.C.)
| | - Hui He
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Z.L.); (M.C.); (R.Z.); (Z.W.); (H.H.); (H.L.); (H.C.)
| | - Zhiyi Wan
- College of Biological Sciences, China Agricultural University, No. 2 Yuan Ming Yuan West Road, Beijing 100193, China;
| | - Hengjian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Z.L.); (M.C.); (R.Z.); (Z.W.); (H.H.); (H.L.); (H.C.)
| | - Hanfang Cai
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Z.L.); (M.C.); (R.Z.); (Z.W.); (H.H.); (H.L.); (H.C.)
| | - Zhi Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China;
| | - Ming Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Z.L.); (M.C.); (R.Z.); (Z.W.); (H.H.); (H.L.); (H.C.)
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Z.L.); (M.C.); (R.Z.); (Z.W.); (H.H.); (H.L.); (H.C.)
| |
Collapse
|
22
|
Tang X, Zhang L, Ren S, Zhao Y, Zhang Y. Temporal and geographic distribution of gut microbial enterotypes associated with host thermogenesis characteristics in plateau pikas. Microbiol Spectr 2023; 11:e0002023. [PMID: 37815332 PMCID: PMC10715161 DOI: 10.1128/spectrum.00020-23] [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: 01/03/2023] [Accepted: 08/28/2023] [Indexed: 10/11/2023] Open
Abstract
IMPORTANCE The gut microbiotas of small mammals play an important role in host energy homeostasis. However, it is still unknown whether small mammals with different enterotypes show differences in thermogenesis characteristics. Our study confirmed that plateau pikas with different bacterial enterotypes harbored distinct thermogenesis capabilities and employed various strategies against cold environments. Additionally, we also found that pikas with different fungal enterotypes may display differences in coprophagy.
Collapse
Affiliation(s)
- Xianjiang Tang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
- University of Chinese Academy of Sciences, College of Life Sciences, Beijing, China
| | - Liangzhi Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | - Shi'en Ren
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
- University of Chinese Academy of Sciences, College of Life Sciences, Beijing, China
| | - Yaqi Zhao
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
- University of Chinese Academy of Sciences, College of Life Sciences, Beijing, China
| | - Yanming Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| |
Collapse
|
23
|
Raymond S, St Clair CC. Urban Magpies Frequently Feed on Coyote Scats and May Spread an Emerging Zoonotic Tapeworm. ECOHEALTH 2023; 20:441-452. [PMID: 38109036 DOI: 10.1007/s10393-023-01664-5] [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: 12/16/2022] [Accepted: 10/30/2023] [Indexed: 12/19/2023]
Abstract
Allocoprophagy, in which animals feed on the feces of other individuals or species, has been little studied in vertebrates, despite its relevance to parasite transmission. These relationships may be especially important in cities, where animal density, disease incidence, and spatial overlap of humans and wildlife increase. Our goal was to document the incidence and predictors of coprophagy by black-billed magpies (Pica hudsonia) at coyote (Canis latrans) scats in Edmonton, Canada. We detected scats by following coyote trails and recorded whether coprophagy had occurred. We used multiple logistic regression to determine the top contextual and environmental predictors of coprophagy. Of 668 coyote scats, 37.3% had apparently been fed on. Coprophagy was more likely in winter and when scats were not fresh and did not contain vegetation or garbage. Environmental predictors of coprophagy included proximity to other coyote scats and playgrounds, distance from water and maintained trails, abundant natural land cover, and proximity to encampments of people experiencing homelessness. Our results reveal that magpies frequently access coyote scat and often do so near human-use areas. In Edmonton, where > 50% of coyotes are infected with a zoonotic tapeworm, coprophagy likely causes magpies to transport parasites with implications for zoonotic disease risk.
Collapse
Affiliation(s)
- Sage Raymond
- Department of Biological Sciences, University of Alberta, 77 University Campus, Edmonton, AB, T6G 2R3, Canada.
| | - Colleen Cassady St Clair
- Department of Biological Sciences, University of Alberta, 77 University Campus, Edmonton, AB, T6G 2R3, Canada
| |
Collapse
|
24
|
Valdivia C, Newton JA, von Beeren C, O'Donnell S, Kronauer DJC, Russell JA, Łukasik P. Microbial symbionts are shared between ants and their associated beetles. Environ Microbiol 2023; 25:3466-3483. [PMID: 37968789 DOI: 10.1111/1462-2920.16544] [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: 05/11/2023] [Accepted: 10/31/2023] [Indexed: 11/17/2023]
Abstract
The transmission of microbial symbionts across animal species could strongly affect their biology and evolution, but our understanding of transmission patterns and dynamics is limited. Army ants (Formicidae: Dorylinae) and their hundreds of closely associated insect guest species (myrmecophiles) can provide unique insights into interspecific microbial symbiont sharing. Here, we compared the microbiota of workers and larvae of the army ant Eciton burchellii with those of 13 myrmecophile beetle species using 16S rRNA amplicon sequencing. We found that the previously characterized specialized bacterial symbionts of army ant workers were largely absent from ant larvae and myrmecophiles, whose microbial communities were usually dominated by Rickettsia, Wolbachia, Rickettsiella and/or Weissella. Strikingly, different species of myrmecophiles and ant larvae often shared identical 16S rRNA genotypes of these common bacteria. Protein-coding gene sequences confirmed the close relationship of Weissella strains colonizing army ant larvae, some workers and several myrmecophile species. Unexpectedly, these strains were also similar to strains infecting dissimilar animals inhabiting very different habitats: trout and whales. Together, our data show that closely interacting species can share much of their microbiota, and some versatile microbial species can inhabit and possibly transmit across a diverse range of hosts and environments.
Collapse
Affiliation(s)
- Catalina Valdivia
- Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, Kraków, Poland
| | - Justin A Newton
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Christoph von Beeren
- Department of Biology, Technical University of Darmstadt, Darmstadt, Germany
- Laboratory of Social Evolution and Behavior, The Rockefeller University, New York, New York, USA
| | - Sean O'Donnell
- Department of Biodiversity, Earth & Environmental Science, Drexel University, Philadelphia, Pennsylvania, USA
| | - Daniel J C Kronauer
- Laboratory of Social Evolution and Behavior, The Rockefeller University, New York, New York, USA
- Howard Hughes Medical Institute, New York, New York, USA
| | - Jacob A Russell
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Piotr Łukasik
- Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, Kraków, Poland
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
25
|
Gao WT, Liu JX, Wang DH, Sun HJ, Zhang XY. Melatonin reduced colon inflammation but had no effect on energy metabolism in ageing Mongolian gerbils (Meriones unguiculatus). Comp Biochem Physiol C Toxicol Pharmacol 2023; 273:109731. [PMID: 37611884 DOI: 10.1016/j.cbpc.2023.109731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/21/2023] [Accepted: 08/20/2023] [Indexed: 08/25/2023]
Abstract
In photoperiod-sensitive wild animals, the secretion of melatonin (MT) is modulated by external photoperiod, and MT affects inflammation and the ageing process. The beneficial effects of MT in delaying the progress of ageing have been reported in laboratory mice and rats. However, little is known about MT in wild mammals. In the current study, we investigated energy metabolism, microbial community structure and colon homeostasis in ageing Mongolian gerbils (Meriones unguiculatus) through exogenous supplementation of MT to test the hypothesis that MT has beneficial effects on gut homeostasis in ageing gerbils. Exogenous MT supplementation had no effect on energy metabolism in Mongolian gerbils but reduced the levels of circulating tumor necrosis factor-α (TNF-α), immune globulin G (IgG) and corticosterone (CORT). The increase in the level of inflammation in ageing animals was related to changes in the structure and diversity of the gut microbiota. At the genus level, the relative abundance of Prevotella, Treponema, Corynebacterium, and Sphingomonas was increased in ageing animals and decreased significantly by the treatment of MT. Christensenella and Lactobacillus were attenuated in ageing animals, and tended to be enhanced by MT treatment. Functions related to glycosphingolipid biosynthesis-ganglio series and lipopolysaccharide biosynthesis (metabolisms of cofactors, vitamins and glycan) were increased in ageing animals and decreased significantly by the treatment of MT. Our data suggest that a supplement of MT could improve colon homeostasis through changing the composition of gut microbiota and reducing inflammation in ageing gerbils.
Collapse
Affiliation(s)
- Wen-Ting Gao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan 250358, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin-Xiu Liu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; School of Life Sciences, Shenyang Normal University, Shenyang 110034, China
| | - De-Hua Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; School of Life Sciences, Shandong University, Qingdao 266237, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hai-Ji Sun
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan 250358, China.
| | - Xue-Ying Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
26
|
Tang X, Zhang L, Ren S, Zhao Y, Liu K, Zhang Y. Stochastic Processes Derive Gut Fungi Community Assembly of Plateau Pikas ( Ochotona curzoniae) along Altitudinal Gradients across Warm and Cold Seasons. J Fungi (Basel) 2023; 9:1032. [PMID: 37888290 PMCID: PMC10607853 DOI: 10.3390/jof9101032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/05/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023] Open
Abstract
Although fungi occupy only a small proportion of the microbial community in the intestinal tract of mammals, they play important roles in host fat accumulation, nutrition metabolism, metabolic health, and immune development. Here, we investigated the dynamics and assembly of gut fungal communities in plateau pikas inhabiting six altitudinal gradients across warm and cold seasons. We found that the relative abundances of Podospora and Sporormiella significantly decreased with altitudinal gradients in the warm season, whereas the relative abundance of Sarocladium significantly increased. Alpha diversity significantly decreased with increasing altitudinal gradient in the warm and cold seasons. Distance-decay analysis showed that fungal community similarities were significantly and negatively correlated with elevation. The co-occurrence network complexity significantly decreased along the altitudinal gradients as the total number of nodes, number of edges, and degree of nodes significantly decreased. Both the null and neutral model analyses showed that stochastic or neutral processes dominated the gut fungal community assembly in both seasons and that ecological drift was the main ecological process explaining the variation in the gut fungal community across different plateau pikas. Homogeneous selection played a weak role in structuring gut fungal community assembly during the warm season. Collectively, these results expand our understanding of the distribution patterns of gut fungal communities and elucidate the mechanisms that maintain fungal diversity in the gut ecosystems of small mammals.
Collapse
Affiliation(s)
- Xianjiang Tang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining 810008, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liangzhi Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining 810008, China
| | - Shien Ren
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining 810008, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaqi Zhao
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining 810008, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kai Liu
- Qinghai Provincial Grassland Station, Xining 810008, China
| | - Yanming Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining 810008, China
| |
Collapse
|
27
|
Klure DM, Dearing MD. Seasonal restructuring facilitates compositional convergence of gut microbiota in free-ranging rodents. FEMS Microbiol Ecol 2023; 99:fiad127. [PMID: 37838471 PMCID: PMC10622585 DOI: 10.1093/femsec/fiad127] [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: 01/11/2023] [Revised: 08/22/2023] [Accepted: 10/11/2023] [Indexed: 10/16/2023] Open
Abstract
Gut microbes provide essential services to their host and shifts in their composition can impact host fitness. However, despite advances in our understanding of how microbes are assembled in the gut, we understand little about the stability of these communities within individuals, nor what factors influence its composition over the life of an animal. For this reason, we conducted a longitudinal survey of the gut microbial communities of individual free-ranging woodrats (Neotoma spp.) across a hybrid zone in the Mojave Desert, USA, using amplicon sequencing approaches to characterize gut microbial profiles and diet. We found that gut microbial communities were individualized and experienced compositional restructuring as a result of seasonal transitions and changes in diet. Turnover of gut microbiota was highest amongst bacterial subspecies and was much lower at the rank of Family, suggesting there may be selection for conservation of core microbial functions in the woodrat gut. Lastly, we identified an abundant core gut bacterial community that may aid woodrats in metabolizing a diet of plants and their specialized metabolites. These results demonstrate that the gut microbial communities of woodrats are highly dynamic and experience seasonal restructuring which may facilitate adaptive plasticity in response to changes in diet.
Collapse
Affiliation(s)
- Dylan M Klure
- School of Biological Sciences, University of Utah, 257 S 1400 E rm 201, Salt Lake City, UT, 84112, United States
| | - M Denise Dearing
- School of Biological Sciences, University of Utah, 257 S 1400 E rm 201, Salt Lake City, UT, 84112, United States
| |
Collapse
|
28
|
Videvall E, Bensch HM, Engelbrecht A, Cloete S, Cornwallis CK. Coprophagy rapidly matures juvenile gut microbiota in a precocial bird. Evol Lett 2023; 7:240-251. [PMID: 37475750 PMCID: PMC10355177 DOI: 10.1093/evlett/qrad021] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 07/22/2023] Open
Abstract
Coprophagy is a behavior where animals consume feces, and has been observed across a wide range of species, including birds and mammals. The phenomenon is particularly prevalent in juveniles, but the reasons for this remain unclear. One hypothesis is that coprophagy enables offspring to acquire beneficial gut microbes that aid development. However, despite the potential importance of this behavior, studies investigating the effects in juveniles are rare. Here we experimentally test this idea by examining how ingestion of adult feces by ostrich chicks affects their gut microbiota development, growth, feeding behavior, pathogen abundance, and mortality. We conducted extensive longitudinal experiments for 8 weeks, repeated over 2 years. It involved 240 chicks, of which 128 were provided daily access to fresh fecal material from adults and 112 were simultaneously given a control treatment. Repeated measures, behavioral observations, and DNA metabarcoding of the microbial gut community, both prior to and over the course of the experiment, allowed us to evaluate multiple aspects of the behavior. The results show that coprophagy causes (a) marked shifts to the juvenile gut microbiota, including a major increase in diversity and rapid maturation of the microbial composition, (b) higher growth rates (fecal-supplemented chicks became 9.4% heavier at 8 weeks old), (c) changes to overall feeding behavior but no differences in feed intake, (d) lower abundance of a common gut pathogen (Clostridium colinum), and (e) lower mortality associated with gut disease. Together, our results suggest that the behavior of coprophagy in juveniles is highly beneficial and may have evolved to accelerate the development of gut microbiota.
Collapse
Affiliation(s)
- Elin Videvall
- Corresponding author: Animal Ecology, Department of Ecology and Genetics, Uppsala University, Uppsala, Sweden.
| | - Hanna M Bensch
- Department of Biology, Lund University, Lund, Sweden
- Department of Biology and Environmental Science, Linneaus University, Kalmar, Sweden
| | - Anel Engelbrecht
- Directorate Animal Sciences, Western Cape Department of Agriculture, Oudtshoorn, South Africa
| | - Schalk Cloete
- Directorate Animal Sciences, Western Cape Department of Agriculture, Oudtshoorn, South Africa
- Department of Animal Sciences, Stellenbosch University, Stellenbosch, South Africa
| | | |
Collapse
|
29
|
Mukilan M. Impact of Pseudomonas aeruginosa, Bacillus subtilis, Staphylococcus aureus, and Escherichia coli Oral Infusions on Cognitive Memory Decline in Mild Cognitive Impairment. JOURNAL OF EXPERIMENTAL BIOLOGY AND AGRICULTURAL SCIENCES 2023; 11:581-592. [DOI: 10.18006/2023.11(3).581.592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Synaptic plasticity is a result of changes in the neuronal circuits which may result in the formation of protein-dependent (long-term memory (LTM) formation) and protein-independent (short-term memory (STM) formation) memories. This STM formation is based on existing proteins, but LTM formation depends on RNA and protein synthesis within the neuronal cells. This RNA and protein synthesis may depend on stimulus exposure like odour, taste, and other environmental stimuli. The present study is aimed to show the impact of oral bacterial infusions on cognitive memory formation through pre and post-infusive behavioural analysis. The results of the study revealed that oral infusions of Pseudomonas aeruginosa, Bacillus subtilis, Staphylococcus aureus and Escherichia coli result in impaired cognitive learning and memory formation. This impaired cognitive memory formation is shown with the help of two-step (pre and post-infusive) behavioural analysis. Pre-infusive behavioural study shows no decline in cognitive learning and memory formation before oral microbial infusions in a serene habituated environment. After oral microbial infusions, a post-infusive behavioural analysis may reveal a memory decline in the treated group. Comparative two-step behavioural analysis indicates that P. aeruginosa infusions strongly impact cognitive memory decline compared to the other three groups. This cognitive memory decline may happen due to the production of primary/secondary metabolites within the animal gut and their transportation to the CNS through the blood-brain barrier. The outcome of the present study states that poor oral hygiene plays a significant role in cognitive memory decline concerning mild cognitive impairment (MCI).
Collapse
|
30
|
Klatt KC, Bass K, Speakman JR, Hall KD. Chowing down: diet considerations in rodent models of metabolic disease. LIFE METABOLISM 2023; 2:load013. [PMID: 37485302 PMCID: PMC10361708 DOI: 10.1093/lifemeta/load013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/28/2023] [Accepted: 04/13/2023] [Indexed: 07/25/2023]
Abstract
Diet plays a substantial role in the etiology, progression, and treatment of chronic disease and is best considered as a multifaceted set of modifiable input variables with pleiotropic effects on a variety of biological pathways spanning multiple organ systems. This brief review discusses key issues related to the design and conduct of diet interventions in rodent models of metabolic disease and their implications for interpreting experiments. We also make specific recommendations to improve rodent diet studies to help better understand the role of diet on metabolic physiology and thereby improve our understanding of metabolic disease.
Collapse
Affiliation(s)
- Kevin C Klatt
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA 94720, United States
| | - Kevin Bass
- Garrison Institute of Aging, Texas Tech University Health Science Center, Lubbock, TX 79430, United States
| | - John R Speakman
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Kevin D Hall
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| |
Collapse
|
31
|
Bedree JK, Kerns K, Chen T, Lima BP, Liu G, Ha P, Shi J, Pan HC, Kim JK, Tran L, Minot SS, Hendrickson EL, Lamont EI, Schulte F, Hardt M, Stephens D, Patel M, Kokaras A, Stodieck L, Shirazi-Fard Y, Wu B, Kwak JH, Ting K, Soo C, McLean JS, He X, Shi W. Specific host metabolite and gut microbiome alterations are associated with bone loss during spaceflight. Cell Rep 2023; 42:112299. [PMID: 37080202 PMCID: PMC10344367 DOI: 10.1016/j.celrep.2023.112299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 10/30/2022] [Accepted: 03/07/2023] [Indexed: 04/22/2023] Open
Abstract
Understanding the axis of the human microbiome and physiological homeostasis is an essential task in managing deep-space-travel-associated health risks. The NASA-led Rodent Research 5 mission enabled an ancillary investigation of the gut microbiome, varying exposure to microgravity (flight) relative to ground controls in the context of previously shown bone mineral density (BMD) loss that was observed in these flight groups. We demonstrate elevated abundance of Lactobacillus murinus and Dorea sp. during microgravity exposure relative to ground control through whole-genome sequencing and 16S rRNA analyses. Specific functionally assigned gene clusters of L. murinus and Dorea sp. capable of producing metabolites, lactic acid, leucine/isoleucine, and glutathione are enriched. These metabolites are elevated in the microgravity-exposed host serum as shown by liquid chromatography-tandem mass spectrometry (LC-MS/MS) metabolomic analysis. Along with BMD loss, ELISA reveals increases in osteocalcin and reductions in tartrate-resistant acid phosphatase 5b signifying additional loss of bone homeostasis in flight.
Collapse
Affiliation(s)
- Joseph K Bedree
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA.
| | - Kristopher Kerns
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Tsute Chen
- Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Bruno P Lima
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Guo Liu
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Pin Ha
- Section of Orthodontics, Division of Growth & Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Plastic and Reconstructive Surgery, School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jiayu Shi
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hsin Chuan Pan
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jong Kil Kim
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Luan Tran
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samuel S Minot
- Microbiome Research Initiative, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Erik L Hendrickson
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Eleanor I Lamont
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Fabian Schulte
- Forsyth Center for Salivary Diagnostics, Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA; Harvard School of Dental Medicine, Department of Developmental Biology, Boston, MA 02115, USA
| | - Markus Hardt
- Forsyth Center for Salivary Diagnostics, Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA; Harvard School of Dental Medicine, Department of Developmental Biology, Boston, MA 02115, USA
| | - Danielle Stephens
- Multiplex Core, Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA
| | - Michele Patel
- Multiplex Core, Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA
| | - Alexis Kokaras
- Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA
| | - Louis Stodieck
- BioServe Space Technologies, Department of Aerospace Engineering Sciences, University of Colorado, Boulder, CO 80303, USA
| | - Yasaman Shirazi-Fard
- Bone and Signaling Laboratory, Space Biosciences Division, NASA Ames Research Center, Mail Stop 288-2, Moffett Field, CA 94035, USA
| | - Benjamin Wu
- Department of Bioengineering, School of Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jin Hee Kwak
- Section of Orthodontics, Division of Growth & Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kang Ting
- Section of Orthodontics, Division of Growth & Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthopedic Surgery, School of Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jeffrey S McLean
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Xuesong He
- Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Wenyuan Shi
- Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA.
| |
Collapse
|
32
|
Wang Z, He H, Chen M, Ni M, Yuan D, Cai H, Chen Z, Li M, Xu H. Impact of coprophagy prevention on the growth performance, serum biochemistry, and intestinal microbiome of rabbits. BMC Microbiol 2023; 23:125. [PMID: 37165350 PMCID: PMC10170819 DOI: 10.1186/s12866-023-02869-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 04/23/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Coprophagy plays a vital role in maintaining growth and development in many small herbivores. Here, we constructed a coprophagy model by dividing rabbits into three groups, namely, control group (CON), sham-coprophagy prevention group (SCP), and coprophagy prevention group (CP), to explore the effects of coprophagy prevention on growth performance and cecal microecology in rabbits. RESULTS Results showed that CP treatment decreased the feed utilization and growth performance of rabbits. Serum total cholesterol and total triglyceride in the CP group were remarkably lower than those in the other two groups. Furthermore, CP treatment destroyed cecum villi and reduced the content of short-chain fatty acids (SCFAs) in cecum contents. Gut microbiota profiling showed significant differences in the phylum and genus composition of cecal microorganisms among the three groups. At the genus level, the abundance of Oscillospira and Ruminococcus decreased significantly in the CP group. Enrichment analysis of metabolic pathways showed a significantly up-regulated differential metabolic pathway (PWY-7315, dTDP-N-acetylthomosamine biosynthesis) in the CP group compared with that in the CON group. Correlation analysis showed that the serum biochemical parameters were positively correlated with the abundance of Oscillospira, Sutterella, and Butyricimonas but negatively correlated with the abundance of Oxalobacte and Desulfovibrio. Meanwhile, the abundance of Butyricimonas and Parabacteroidesde was positively correlated with the concentration of butyric acid in the cecum. CONCLUSIONS In summary, coprophagy prevention had negative effects on serum biochemistry and gut microbiota, ultimately decreasing the growth performance of rabbits. The findings provide evidence for further revealing the biological significance of coprophagy in small herbivorous mammals.
Collapse
Affiliation(s)
- Zhitong Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Hui He
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Mengjuan Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Mengke Ni
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Dongdong Yuan
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Hanfang Cai
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Zhi Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225000, China
| | - Ming Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China.
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China.
| |
Collapse
|
33
|
Sha H, He X, Yan K, Li J, Li X, Xie Y, Yang Y, Deng Y, Li G, Yang J. Blocking coprophagy increases the levels of inflammation and depression in healthy mice as well as mice receiving fecal microbiota transplantation from disease model mice donors. APMIS 2023. [PMID: 37145345 DOI: 10.1111/apm.13326] [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: 04/17/2022] [Accepted: 04/18/2023] [Indexed: 05/06/2023]
Abstract
Rodents have been extensively used as animal models in microbiome studies. However, all rodents have a habitual nature called coprophagy, a phenomenon that they self-reinoculate feces into their gastrointestinal tract. Recent studies have shown that blocking coprophagy can alter rodents' diversity of gut microbiota, metabolism, neurochemistry, and cognitive behavior. However, whether rodents' coprophagy behavior affects the levels of inflammation and depression is unclear. In order to address this problem, we first blocked coprophagy in healthy mice. It displayed an increase in the levels of depression, verified by depressive-like behaviors and mood-related indicators, and inflammation, verified by the increased levels of the pro-inflammatory cytokine, in coprophagy-blocked mice. Furthermore, we transplanted fecal microbiota from chronic restraint stress (CRS) depression model mice and lipopolysaccharide (LPS) inflammation model mice to healthy recipient mice, respectively. It showed that the disease-like phenotypes in the coprophagy-blocked group were worse than those in the coprophagy-unblocked group, including severer depressive symptoms and higher levels of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α and IFN-γ) in serum, prefrontal cortex (PFC), and hippocampus (HIP). These findings showed that blocking coprophagy in mice not only increased the levels of inflammation and depression in healthy mice but also aggravated inflammation and depression induced by fecal microbiota from disease donors. The discovery may provide a vital reference for future research involving FMT in rodents.
Collapse
Affiliation(s)
- Haoran Sha
- Grade 2020, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Xiaoyi He
- Department of Anatomy, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Kai Yan
- Department of Anatomy, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jiakang Li
- Grade 2017, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Xu Li
- Grade 2018, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yinyin Xie
- Grade 2018, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yousheng Yang
- Grade 2018, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yajuan Deng
- Grade 2018, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Guoying Li
- Guangdong Medical Association, Guangzhou, Guangdong, China
| | - Junhua Yang
- Department of Anatomy, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Guangdong Key Laboratory of Bioactive Drug Research, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
34
|
Tannic Acid Induces Intestinal Dysfunction and Intestinal Microbial Dysregulation in Brandt's Voles ( Lasiopodomys brandtii). Animals (Basel) 2023; 13:ani13040586. [PMID: 36830373 PMCID: PMC9951651 DOI: 10.3390/ani13040586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 02/10/2023] Open
Abstract
Brandt's vole (Lasiopodomys brandtii) is a small herbivorous mammal that feeds on plants rich in secondary metabolites (PSMs), including tannins. However, plant defense mechanisms against herbivory by Brandt's voles are not clearly established. This study aimed to investigate the effects of dietary tannic acid (TA) on the growth performance, intestinal morphology, digestive enzyme activities, cecal fermentation, intestinal barrier function, and gut microbiota in Brandt's voles. The results showed that TA significantly hindered body weight gain, reduced daily food intake, changed the intestinal morphology, reduced digestive enzyme activity, and increased the serum zonulin levels (p < 0.05). The number of intestinal goblet and mast cells and the levels of serum cytokines and immunoglobulins (IgA, IgG, TNF-α, IL-6, and duodenal SlgA) were all reduced by TA (p < 0.05). Moreover, TA altered β-diversity in the colonic microbial community (p < 0.05). In conclusion, the results indicate that TA could damage the intestinal function of Brandt's voles by altering their intestinal morphology, decreasing digestive ability and intestinal barrier function, and altering microbiota composition. Our study investigated the effects of natural PSMs on the intestinal function of wildlife and improved our general understanding of plant-herbivore interactions and the ecological role of PSMs.
Collapse
|
35
|
Cheney AM, Costello SM, Pinkham NV, Waldum A, Broadaway SC, Cotrina-Vidal M, Mergy M, Tripet B, Kominsky DJ, Grifka-Walk HM, Kaufmann H, Norcliffe-Kaufmann L, Peach JT, Bothner B, Lefcort F, Copié V, Walk ST. Gut microbiome dysbiosis drives metabolic dysfunction in Familial dysautonomia. Nat Commun 2023; 14:218. [PMID: 36639365 PMCID: PMC9839693 DOI: 10.1038/s41467-023-35787-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 12/18/2022] [Indexed: 01/15/2023] Open
Abstract
Familial dysautonomia (FD) is a rare genetic neurologic disorder caused by impaired neuronal development and progressive degeneration of both the peripheral and central nervous systems. FD is monogenic, with >99.4% of patients sharing an identical point mutation in the elongator acetyltransferase complex subunit 1 (ELP1) gene, providing a relatively simple genetic background in which to identify modifiable factors that influence pathology. Gastrointestinal symptoms and metabolic deficits are common among FD patients, which supports the hypothesis that the gut microbiome and metabolome are altered and dysfunctional compared to healthy individuals. Here we show significant differences in gut microbiome composition (16 S rRNA gene sequencing of stool samples) and NMR-based stool and serum metabolomes between a cohort of FD patients (~14% of patients worldwide) and their cohabitating, healthy relatives. We show that key observations in human subjects are recapitulated in a neuron-specific Elp1-deficient mouse model, and that cohousing mutant and littermate control mice ameliorates gut microbiome dysbiosis, improves deficits in gut transit, and reduces disease severity. Our results provide evidence that neurologic deficits in FD alter the structure and function of the gut microbiome, which shifts overall host metabolism to perpetuate further neurodegeneration.
Collapse
Affiliation(s)
- Alexandra M Cheney
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, USA
| | - Stephanann M Costello
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, USA
| | - Nicholas V Pinkham
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Annie Waldum
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, USA
| | - Susan C Broadaway
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Maria Cotrina-Vidal
- Department of Neurology, New York University School of Medicine, New York, NY, USA
| | - Marc Mergy
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Brian Tripet
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, USA
| | - Douglas J Kominsky
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Heather M Grifka-Walk
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Horacio Kaufmann
- Department of Neurology, New York University School of Medicine, New York, NY, USA
| | | | - Jesse T Peach
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, USA
| | - Brian Bothner
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, USA
| | - Frances Lefcort
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA.
| | - Valérie Copié
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, USA.
| | - Seth T Walk
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA.
| |
Collapse
|
36
|
Recruitment of Muscle Genes as an Effect of Brown Adipose Tissue Ablation in Cold-Acclimated Brandt's Voles ( Lasiopodomys brandtii). Int J Mol Sci 2022; 24:ijms24010342. [PMID: 36613791 PMCID: PMC9820317 DOI: 10.3390/ijms24010342] [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: 11/10/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Skeletal muscle-based nonshivering thermogenesis (NST) plays an important role in the regulation and maintenance of body temperature in birds and large mammals, which do not contain brown adipose tissue (BAT). However, the relative contribution of muscle-based NST to thermoregulation is not clearly elucidated in wild small mammals, which have evolved an obligate thermogenic organ of BAT. In this study, we investigated whether muscle would become an important site of NST when BAT function is conditionally minimized in Brandt's voles (Lasiopodomys brandtii). We surgically removed interscapular BAT (iBAT, which constitutes 52%~56% of total BAT) and exposed the voles to prolonged cold (4 °C) for 28 days. The iBAT-ablated voles were able to maintain the same levels of NST and body temperature (~37.9 °C) during the entire period of cold acclimation as sham voles. The expression of uncoupling protein 1 (UCP1) and its transcriptional regulators at both protein and mRNA levels in the iBAT of cold-acclimated voles was higher than that in the warm group. However, no difference was observed in the protein or mRNA levels of these thermogenesis-related markers except for PGC-1α in other sites of BAT (including infrascapular region, neck, and axilla) between warm and cold groups either in sham or iBAT-ablated voles. The iBAT-ablated voles showed higher UCP1 expression in white adipose tissue (WAT) than sham voles during cold acclimation. The expression of sarcolipin (SLN) and sarcoplasmic endoplasmic reticulum Ca2+-dependent adenosine triphosphatase (SERCA) in skeletal muscles was higher in cold than in warm, but no alteration in phospholamban (PLB) and phosphorylated-PLB (P-PLB) was observed. Additionally, there was increased in iBAT-ablated voles compared to that in the sham group in cold. Moreover, these iBAT-ablated voles underwent extensive remodeling of mitochondria and genes of key components related with mitochondrial metabolism. These data collectively indicate that recruitment of skeletal muscle-based thermogenesis may compensate for BAT impairment and suggest a functional interaction between the two forms of thermogenic processes of iBAT and skeletal muscle in wild small mammals for coping cold stress.
Collapse
|
37
|
Li Z, Li R, Li J, Wang Z, He H, Yan D, Yu L, Li H, Li M, Xu H. Coprophagy Prevention Affects the Reproductive Performance in New Zealand White Rabbits Is Mediated through Nox4-ROS-NF κB Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8999899. [PMID: 39282150 PMCID: PMC11401658 DOI: 10.1155/2022/8999899] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 09/18/2024]
Abstract
Coprophagy is of great significance to the growth, development, and reproductive performance of rabbits. This study is aimed at exploring the effect of coprophagy on the reproductive performance of New Zealand white rabbits by coprophagy prevention (CP). The results showed that CP treatment significantly decreased the growth and development performance of female rabbits and the live birth rate of embryos. The results of blood biochemical indexes showed that CP treatment significantly increased the contents of serum ALB, ALP, and MDA, while serum SOD activity was significantly decreased. Transcriptome analysis showed that GO terms were mainly enriched in transport function and reproductive function after CP treatment. In addition, KEGG results showed that inflammation related signal pathways were activated and the expression level of genes related to tight junction proteins was downregulated by CP treatment. Concurrently, western blot further confirmed the results of KEGG. In short, fecal feeding is an important survival strategy for some small rodents, coprophagy prevention will affect the inflammatory level of the body, change the oxidative stress level of the body, and then activate NOX4-ROS-NF-κB pathway, increase the expression level of adhesion protein ICAM-1 and VCAM-1, lead to the damage of uterine epithelial barrier, and then affect the reproductive performance of rabbits.
Collapse
Affiliation(s)
- Zhichao Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - RuiTing Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Jing Li
- Animal Health Supervision Institute of Biyang, Henan 463700, China
| | - Zhitong Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Hui He
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Duo Yan
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Lei Yu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Hengjian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Ming Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| |
Collapse
|
38
|
Moraitou M, Forsythe A, Fellows Yates JA, Brealey JC, Warinner C, Guschanski K. Ecology, Not Host Phylogeny, Shapes the Oral Microbiome in Closely Related Species. Mol Biol Evol 2022; 39:msac263. [PMID: 36472532 PMCID: PMC9778846 DOI: 10.1093/molbev/msac263] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Host-associated microbiomes are essential for a multitude of biological processes. Placed at the contact zone between external and internal environments, the little-studied oral microbiome has important roles in host physiology and health. Here, we investigate the roles of host evolutionary relationships and ecology in shaping the oral microbiome in three closely related gorilla subspecies (mountain, Grauer's, and western lowland gorillas) using shotgun metagenomics of 46 museum-preserved dental calculus samples. We find that the oral microbiomes of mountain gorillas are functionally and taxonomically distinct from the other two subspecies, despite close evolutionary relationships and geographic proximity with Grauer's gorillas. Grauer's gorillas show intermediate bacterial taxonomic and functional, and dietary profiles. Altitudinal differences in gorilla subspecies ranges appear to explain these patterns, suggesting a close connection between dental calculus microbiomes and the environment, likely mediated through diet. This is further supported by the presence of gorilla subspecies-specific phyllosphere/rhizosphere taxa in the oral microbiome. Mountain gorillas show a high abundance of nitrate-reducing oral taxa, which may promote adaptation to a high-altitude lifestyle by modulating blood pressure. Our results suggest that ecology, rather than evolutionary relationships and geographic distribution, shape the oral microbiome in these closely related species.
Collapse
Affiliation(s)
- Markella Moraitou
- Animal Ecology, Department of Ecology and Genetics, Uppsala University, 75236 Uppsala, Sweden
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, United Kingdom
| | - Adrian Forsythe
- Animal Ecology, Department of Ecology and Genetics, Uppsala University, 75236 Uppsala, Sweden
| | - James A Fellows Yates
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
- Department of Paleobiotechnology, Leibniz Institute for Natural Product Research and Infection Biology Hans Knöll Institute, 07745 Jena, Germany
| | - Jaelle C Brealey
- Department of Natural History, NTNU University Museum, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Christina Warinner
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
- Department of Paleobiotechnology, Leibniz Institute for Natural Product Research and Infection Biology Hans Knöll Institute, 07745 Jena, Germany
- Faculty of Biological Sciences, Friedrich Schiller University, 07743 Jena, Germany
- Department of Anthropology, Harvard University, Cambridge, MA 02138, USA
| | - Katerina Guschanski
- Animal Ecology, Department of Ecology and Genetics, Uppsala University, 75236 Uppsala, Sweden
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, United Kingdom
- Science for Life Laboratory, 75237 Uppsala, Sweden
| |
Collapse
|
39
|
Li Z, He H, Ni M, Wang Z, Guo C, Niu Y, Xing S, Song M, Wang Y, Jiang Y, Yu L, Li M, Xu H. Microbiome-Metabolome Analysis of the Immune Microenvironment of the Cecal Contents, Soft Feces, and Hard Feces of Hyplus Rabbits. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5725442. [PMID: 36466090 PMCID: PMC9713467 DOI: 10.1155/2022/5725442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/16/2022] [Indexed: 01/14/2024]
Abstract
The intestinal microbiota and its metabolites play vital roles in host growth, development, and immune regulation. This study analyzed the microbial community distribution and the cytokine and short-chain fatty acid (SCFA) content of cecal contents (Con group), soft feces (SF group), and hard feces (HF group) of 60-day-old Hyplus rabbits and verified the effect of soft feces on the cecal immune microenvironment by coprophagy prevention (CP). The results showed that there were significant differences in the levels of phylum and genus composition, cytokines, and SCFAs among the Con group, SF group, and HF group. The correlation analysis of cytokines and SCFAs with differential microbial communities showed that Muribaculaceae, Ruminococcaceae_UCG-014, Ruminococcaceae_NK4A214_group, and Christensenellaceae_R-7_Group are closely related to cytokines and SCFAs. After CP treatment, the contents of propionic acid, butyric acid, IL-4, and IL-10 in cecum decreased significantly, whereas TNF-α and IL-1β increased significantly. Moreover, the inhibition of coprophagy led to the downregulation of the expression levels of tight junction proteins (Claudin-1, Occludin, and ZO-1) related to intestinal inflammation and intestinal barrier function, and the ring-like structure of ZO-1 was disrupted. In conclusion, coprophagy can not only help rabbits obtain more probiotics and SCFAs but also play an essential role in improving the immune microenvironment of cecum.
Collapse
Affiliation(s)
- Zhichao Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Hui He
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Mengke Ni
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Zhouyan Wang
- Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Chaohui Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yufang Niu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Shanshan Xing
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Mingkun Song
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yaling Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yixuan Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Lei Yu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Ming Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| |
Collapse
|
40
|
Bartochowski P, Gayrard N, Bornes S, Druart C, Argilés A, Cordaillat-Simmons M, Duranton F. Gut–Kidney Axis Investigations in Animal Models of Chronic Kidney Disease. Toxins (Basel) 2022; 14:toxins14090626. [PMID: 36136564 PMCID: PMC9502418 DOI: 10.3390/toxins14090626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/01/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic kidney disease (CKD) is an incurable disease in which renal function gradually declines, resulting in no noticeable symptoms during the early stages and a life-threatening disorder in the latest stage. The changes that accompany renal failure are likely to influence the gut microbiota, or the ecosystem of micro-organisms resident in the intestine. Altered gut microbiota can display metabolic changes and become harmful to the host. To study the gut–kidney axis in vivo, animal models should ideally reproduce the disorders affecting both the host and the gut microbiota. Murine models of CKD, but not dog, manifest slowed gut transit, similarly to patient. Animal models of CKD also reproduce altered intestinal barrier function, as well as the resulting leaky gut syndrome and bacterial translocation. CKD animal models replicate metabolic but not compositional changes in the gut microbiota. Researchers investigating the gut–kidney axis should pay attention to the selection of the animal model (disease induction method, species) and the setting of the experimental design (control group, sterilization method, individually ventilated cages) that have been shown to influence gut microbiota.
Collapse
Affiliation(s)
- Piotr Bartochowski
- RD Néphrologie SAS, 34090 Montpellier, France
- BC2M, Faculty of Pharmacy, University of Montpellier, 34090 Montpellier, France
| | - Nathalie Gayrard
- RD Néphrologie SAS, 34090 Montpellier, France
- BC2M, Faculty of Pharmacy, University of Montpellier, 34090 Montpellier, France
- Correspondence:
| | - Stéphanie Bornes
- Université Clermont Auvergne, Inrae, Vetagro Sup, UMRF0545, 15000 Aurillac, France
| | - Céline Druart
- Pharmabiotic Research Institute (PRI), 11100 Narbonne, France
| | - Angel Argilés
- RD Néphrologie SAS, 34090 Montpellier, France
- BC2M, Faculty of Pharmacy, University of Montpellier, 34090 Montpellier, France
| | | | - Flore Duranton
- RD Néphrologie SAS, 34090 Montpellier, France
- BC2M, Faculty of Pharmacy, University of Montpellier, 34090 Montpellier, France
| |
Collapse
|
41
|
Bello-Medina PC, Corona-Cervantes K, Zavala Torres NG, González A, Pérez-Morales M, González-Franco DA, Gómez A, García-Mena J, Díaz-Cintra S, Pacheco-López G. Chronic-Antibiotics Induced Gut Microbiota Dysbiosis Rescues Memory Impairment and Reduces β-Amyloid Aggregation in a Preclinical Alzheimer's Disease Model. Int J Mol Sci 2022; 23:8209. [PMID: 35897785 PMCID: PMC9331718 DOI: 10.3390/ijms23158209] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/08/2022] [Accepted: 07/12/2022] [Indexed: 02/03/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial pathology characterized by β-amyloid (Aβ) deposits, Tau hyperphosphorylation, neuroinflammatory response, and cognitive deficit. Changes in the bacterial gut microbiota (BGM) have been reported as a possible etiological factor of AD. We assessed in offspring (F1) 3xTg, the effect of BGM dysbiosisdysbiosis in mothers (F0) at gestation and F1 from lactation up to the age of 5 months on Aβ and Tau levels in the hippocampus, as well as on spatial memory at the early symptomatic stage of AD. We found that BGM dysbiosisdysbiosis with antibiotics (Abx) treatment in F0 was vertically transferred to their F1 3xTg mice, as observed on postnatal day (PD) 30 and 150. On PD150, we observed a delay in spatial memory impairment and Aβ deposits, but not in Tau and pTau protein in the hippocampus at the early symptomatic stage of AD. These effects are correlated with relative abundance of bacteria and alpha diversity, and are specific to bacterial consortia. Our results suggest that this specific BGM could reduce neuroinflammatory responses related to cerebral amyloidosis and cognitive deficit and activate metabolic pathways associated with the biosynthesis of triggering or protective molecules for AD.
Collapse
Affiliation(s)
- Paola C. Bello-Medina
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico;
- Biological and Health Sciences Division, Campus Lerma, Metropolitan Autonomus University (UAM), Lerma 52005, Mexico; (A.G.); (M.P.-M.); (D.A.G.-F.); (A.G.); (G.P.-L.)
| | - Karina Corona-Cervantes
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Zacatenco, Mexico City 07360, Mexico; (K.C.-C.); (N.G.Z.T.)
| | - Norma Gabriela Zavala Torres
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Zacatenco, Mexico City 07360, Mexico; (K.C.-C.); (N.G.Z.T.)
| | - Antonio González
- Biological and Health Sciences Division, Campus Lerma, Metropolitan Autonomus University (UAM), Lerma 52005, Mexico; (A.G.); (M.P.-M.); (D.A.G.-F.); (A.G.); (G.P.-L.)
| | - Marcel Pérez-Morales
- Biological and Health Sciences Division, Campus Lerma, Metropolitan Autonomus University (UAM), Lerma 52005, Mexico; (A.G.); (M.P.-M.); (D.A.G.-F.); (A.G.); (G.P.-L.)
| | - Diego A. González-Franco
- Biological and Health Sciences Division, Campus Lerma, Metropolitan Autonomus University (UAM), Lerma 52005, Mexico; (A.G.); (M.P.-M.); (D.A.G.-F.); (A.G.); (G.P.-L.)
| | - Astrid Gómez
- Biological and Health Sciences Division, Campus Lerma, Metropolitan Autonomus University (UAM), Lerma 52005, Mexico; (A.G.); (M.P.-M.); (D.A.G.-F.); (A.G.); (G.P.-L.)
| | - Jaime García-Mena
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Zacatenco, Mexico City 07360, Mexico; (K.C.-C.); (N.G.Z.T.)
| | - Sofía Díaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico;
| | - Gustavo Pacheco-López
- Biological and Health Sciences Division, Campus Lerma, Metropolitan Autonomus University (UAM), Lerma 52005, Mexico; (A.G.); (M.P.-M.); (D.A.G.-F.); (A.G.); (G.P.-L.)
| |
Collapse
|
42
|
Song G, Wang Y, Wang Y, Jiang Y, Sun S, Cai H, Sun G, Li M, Bionaz M, Xu H. Coprophagy Prevention Decreases the Reproductive Performance and Granulosa Cell Apoptosis via Regulation of CTSB Gene in Rabbits. Front Physiol 2022; 13:926795. [PMID: 35923240 PMCID: PMC9341522 DOI: 10.3389/fphys.2022.926795] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/23/2022] [Indexed: 11/22/2022] Open
Abstract
Coprophagy is an instinctive behavior in rabbit with important effects on growth and reproductive performance. The underlying mechanism of this effect in rabbit is unknown. Here, we used Elizabeth circle as a coprophagy preventing model in female rabbits and assess feed intake, growth, and reproductive performance. We found that preventing coprophagy did not affect feed intake but decreased body weight and weight of several organs and tissues and resulted in complete reproductive failure during the late pregnancy period, accompanied by reduced levels of plasma progesterone. RNA-seq analysis of rabbit ovarian tissues revealed that preventing coprophagy affected significantly 241 genes (DEGs), with the large majority being downregulated. Bioinformatic analyses revealed that those DEGs are mostly involved in apoptosis, immune response, and metabolic pathways. Among DEGs, the lysosomal cysteine protease cathepsin B (CTSB) was significantly downregulated in the coprophagy prevention group. Further studies using siRNA and adenovirus overexpression systems revealed that CTSB promotes the proliferation of rabbit granulosa cells (GCS) and prevents apoptosis. Measurement of transcripts coding for proteins related to apoptosis revealed a minor transcriptomic effect of CTSB, indicating that its effect is likely post-transcriptional. Overexpression of CTSB increased secretion of progesterone and estradiol, partly via upregulation of CYP19A1 while inhibition of CTSB decreased progesterone secretion partly via downregulation of the StAR gene. In conclusion, our study demonstrated the detrimental effect on reproduction by preventing coprophagy with a main role for this response played by CTSB on the granulosa cells of the ovary.
Collapse
Affiliation(s)
- Guohua Song
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Yadong Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Yaling Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Yixuan Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Shuaijie Sun
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Hanfang Cai
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Guirong Sun
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Ming Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- *Correspondence: Huifen Xu, ; Massimo Bionaz, ; Ming Li,
| | - Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
- *Correspondence: Huifen Xu, ; Massimo Bionaz, ; Ming Li,
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- *Correspondence: Huifen Xu, ; Massimo Bionaz, ; Ming Li,
| |
Collapse
|
43
|
Marclay M, Dwyer E, Suchodolski JS, Lidbury JA, Steiner JM, Gaschen FP. Recovery of Fecal Microbiome and Bile Acids in Healthy Dogs after Tylosin Administration with and without Fecal Microbiota Transplantation. Vet Sci 2022; 9:vetsci9070324. [PMID: 35878341 PMCID: PMC9318503 DOI: 10.3390/vetsci9070324] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/06/2022] [Accepted: 06/15/2022] [Indexed: 12/02/2022] Open
Abstract
Antibiotics cause gut dysbiosis and bile acid dysmetabolism in dogs. The effect of fecal microbiota transplantation (FMT) on microbiome and metabolome recovery is unknown. This prospective, randomized, placebo-controlled study included sixteen healthy purpose-bred dogs. All dogs received tylosin 20 mg/kg PO once daily (days 1–7) and were randomly assigned to either receive one FMT via enema (day 8), daily oral FMT capsules (days 8–21), or daily placebo capsules (days 8–21). Fecal samples were frozen at regular intervals until day 42. Quantitative PCR for 8 bacterial taxa was performed to calculate the fecal dysbiosis index (FDI) and fecal concentrations of unconjugated bile acids (UBA) were measured using gas chromatography-mass spectrometry. Tylosin altered the abundance of most evaluated bacteria and induced a significant decrease in secondary bile acid concentrations at day 7 in all dogs. However, most parameters returned to their baseline by day 14 in all dogs. In conclusion, tylosin markedly impacted fecal microbiota and bile acid concentrations, although return to baseline values was quick after the antibiotic was discontinued. Overall, FMT did not accelerate recovery of measured parameters. Further studies are warranted to confirm the value of FMT in accelerating microbiota recovery in antibiotic-associated dysbiosis in dogs.
Collapse
Affiliation(s)
- Margaux Marclay
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (M.M.); (E.D.)
- Medi-Vet SA Vétérinaire, 1007 Lausanne, Switzerland
| | - Elizabeth Dwyer
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (M.M.); (E.D.)
- Austin Veterinary Emergency and Specialty, Austin, TX 78730, USA
| | - Jan S. Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77840, USA; (J.S.S.); (J.A.L.); (J.M.S.)
| | - Jonathan A. Lidbury
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77840, USA; (J.S.S.); (J.A.L.); (J.M.S.)
| | - Joerg M. Steiner
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77840, USA; (J.S.S.); (J.A.L.); (J.M.S.)
| | - Frederic P. Gaschen
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (M.M.); (E.D.)
- Correspondence:
| |
Collapse
|
44
|
Bi T, Zhang L, Zhan L, Feng R, Zhao T, Ren W, Hang T, Zhou W, Lu X. Integrated Analyses of Microbiomics and Metabolomics Explore the Effect of Gut Microbiota Transplantation on Diabetes-Associated Cognitive Decline in Zucker Diabetic Fatty Rats. Front Aging Neurosci 2022; 14:913002. [PMID: 35721013 PMCID: PMC9204715 DOI: 10.3389/fnagi.2022.913002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetes-associated cognitive decline (DACD), one of the complications of type 2 diabetes (T2DM), correlates significantly with the disorder in glycolipid metabolism, insulin/leptin resistance, and accumulation of β-amyloid (Aβ). Although gut microbiota transplantation (GMT), a novel non-invasive physiotherapy strategy, has been a promising intervention to alleviate the symptoms of T2DM, its protective effect on progressive cognitive decline remains elusive. Here, we transplanted the gut microbiota of healthy or cognitive decline donor rats into ZDF or LZ rats, and integrated microbiomics and metabolomics to evaluate the directional effect of the gut microbiota on the recipient rats. The basal metabolism phenotype changed in ZDF rats instead of in LZ rats. One possible mechanism is that the microbiota and metabolites alter the structure of the intestinal tract, stimulate the brain insulin and leptin signaling pathways, and regulate the deposition of Aβ in the brain. It is worth noting that 10 species of genera, such as Parabacteroides, Blautia, and Lactobacillus, can regulate 20 kinds of metabolites, such as propanoic acid, acetic acid, and citramalic acid, and having a significant improvement on the cognitive behavior of ZDF rats. In addition, the correlation analysis indicated the gut microbiota and metabolites are highly associated with host phenotypes affected by GMT. In summary, our study indicates that altering the microbiota-gut-brain axis by reshaping the composition of gut microbiota is a viable strategy that has great potential for improving cognitive function and combatting DACD.
Collapse
Affiliation(s)
- Tingting Bi
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lijing Zhang
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Libin Zhan
- Center for Innovative Engineering Technology in Traditional Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, China
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
- *Correspondence: Libin Zhan,
| | - Ruiqi Feng
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tian Zhao
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiming Ren
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tianyi Hang
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wen Zhou
- School of Traditional Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoguang Lu
- Department of Emergency Medicine, Zhongshan Hospital, Dalian University, Dalian, China
- Xiaoguang Lu,
| |
Collapse
|
45
|
Zhang XY, Wang DH. Gut Microbial Community and Host Thermoregulation in Small Mammals. Front Physiol 2022; 13:888324. [PMID: 35480035 PMCID: PMC9035535 DOI: 10.3389/fphys.2022.888324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
The endotherms, particularly the small mammals living in the polar region and temperate zone, are faced with extreme challenges for maintaining stable core body temperatures in harsh cold winter. The non-hibernating small mammals increase metabolic rate including obligatory thermogenesis (basal/resting metabolic rate, BMR/RMR) and regulatory thermogenesis (mainly nonshivering thermogenesis, NST, in brown adipose tissue and skeletal muscle) to maintain thermal homeostasis in cold conditions. A substantial amount of evidence indicates that the symbiotic gut microbiota are sensitive to air temperature, and play an important function in cold-induced thermoregulation, via bacterial metabolites and byproducts such as short-chain fatty acids and secondary bile acids. Cold signal is sensed by specific thermosensitive transient receptor potential channels (thermo-TRPs), and then norepinephrine (NE) is released from sympathetic nervous system (SNS) and thyroid hormones also increase to induce NST. Meanwhile, these neurotransmitters and hormones can regulate the diversity and compositions of the gut microbiota. Therefore, cold-induced NST is controlled by both Thermo-TRPs—SNS—gut microbiota axis and thyroid—gut microbiota axis. Besides physiological thermoregulation, small mammals also rely on behavioral regulation, such as huddling and coprophagy, to maintain energy and thermal homeostasis, and the gut microbial community is involved in these processes. The present review summarized the recent progress in the gut microbiota and host physiological and behavioral thermoregulation in small mammals for better understanding the evolution and adaption of holobionts (host and symbiotic microorganism). The coevolution of host-microorganism symbionts promotes individual survival, population maintenance, and species coexistence in the ecosystems with complicated, variable environments.
Collapse
Affiliation(s)
- Xue-Ying Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - De-Hua Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, Shandong University, Qingdao, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: De-Hua Wang,
| |
Collapse
|
46
|
The microbiota-gut-kidney axis mediates host osmoregulation in a small desert mammal. NPJ Biofilms Microbiomes 2022; 8:16. [PMID: 35379849 PMCID: PMC8980004 DOI: 10.1038/s41522-022-00280-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 02/21/2022] [Indexed: 12/17/2022] Open
Abstract
Regulating sodium and water balances is crucial for survival of small, desert mammals. Studies demonstrate that the gut microbiota and their metabolites are involved in host energy homeostasis, but little is known on the interactions among salt loading, gut microbiota, and osmoregulation. The aim of this study was to fill this gap. Mongolian gerbils (Meriones unguiculatus) were offered drinking water (Con) and either water containing moderate (4%, MS) or high NaCl (8%, HS) ad libitum. Intake of HS reduced α diversity of the microbial community and, at the genus level, reduced the relative abundances of Rikenella and Christensenella but increased Atopobium. To confirm the function of gut microbiota in host osmoregulation, we transplanted caecal microbiota in HS gerbils. To cope with salt loading, the gerbils concentrated urine, resulting in negative energy balance and systemic inflammation. The HS gerbils increased hypothalamic arginine vasopressin and intestinal and renal aquaporin 2 to support water retention, and reduced intestinal and renal epithelial sodium channel α to promote sodium excretion. However, HS gerbils with caecal microbiota transplant (CMT) from Con donors maintained energy balance and osmoregulation, and had a much reduced systemic inflammation. Further, CMT from Con donors to HS recipients reshaped the gut microbiota, particularly by reducing Parabacteroides distasonis and Prevotella copri, and increasing Lactobacillus reuteri abundances, with a resulting increase in bacterial metabolites such as butyrate. These findings highlight a vital role of the microbiota-gut-kidney axis in mediating salt-related osmoregulation, allowing small mammals to adapt to high salt loads in a desert habitat.
Collapse
|
47
|
Chua KO, Fatima I, Lau YY, Hong KW, Yin WF, Mardaryev A, Chan KG, Chang CY. Bacterial microbiome of faecal samples of naked mole-rat collected from the toilet chamber. BMC Res Notes 2022; 15:107. [PMID: 35303951 PMCID: PMC8932300 DOI: 10.1186/s13104-022-06000-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/09/2022] [Indexed: 01/04/2023] Open
Abstract
Objective The naked mole rats (NMRs, Heterocephalus glaber) are subterranean rodents that belong to the family Bathyergidae. They gained the attention of the scientific community for their exceptionally long lifespan of up to 30 years and have become an animal model of biomedical research on neurodegenerative diseases, aging and cancer. NMRs dig and survive in a maze of underground tunnels and chambers and demarcate toilet chambers for defecation and urination. Due to their coprophagic behaviours, we believed that the toilet chamber might play a role in maintaining optimal health of the NMRs. A 16S rRNA gene amplicon sequencing was performed to characterize the bacterial microbiome of faecal samples collected from the toilet chamber of a laboratory NMR colony. Results Four faecal samples were collected at different time points from the same toilet chamber of a laboratory NMR colony for analysis. The 16S rRNA gene amplicon sequencing revealed that bacterial phyla Firmicutes and Bacteroidetes were the dominant taxa in the bacterial microbiome of NMRs. The relative abundance of the bacterial taxa shifted substantially between time points, indicating a dynamic microbiome in the toilet chamber. The data provided an insight to the faecal microbiome of NMRs in the toilet chamber. Supplementary Information The online version contains supplementary material available at 10.1186/s13104-022-06000-8.
Collapse
Affiliation(s)
- Kah-Ooi Chua
- Centre for Research in Biotechnology for Agriculture, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Iqra Fatima
- Centre for Skin Sciences, School of Chemistry and Biosciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Yin Yin Lau
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University Kuala Lumpur, 56000, Kuala Lumpur, Malaysia
| | - Kar Wai Hong
- International Genome Centre, Jiangsu University, Zhenjiang, China.,Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Wai-Fong Yin
- Institute of Marine Sciences, Shantou University, Shantou, 515063, China
| | - Andrei Mardaryev
- Centre for Skin Sciences, School of Chemistry and Biosciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Kok-Gan Chan
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University Kuala Lumpur, 56000, Kuala Lumpur, Malaysia. .,International Genome Centre, Jiangsu University, Zhenjiang, China. .,Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Chien-Yi Chang
- School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4BW, UK.
| |
Collapse
|
48
|
Lin Y, Yan G, Feng F, Wang M, Long F. Characterization of intestinal microbiota and serum metabolites in patients with mild hepatic encephalopathy. Open Life Sci 2022; 17:139-154. [PMID: 35350446 PMCID: PMC8919820 DOI: 10.1515/biol-2021-0140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 10/15/2021] [Accepted: 11/18/2021] [Indexed: 11/15/2022] Open
Abstract
Mild micro-hepatic encephalopathy (MHE) is a severe complication of cirrhosis. At present, there are differences in the consistency of detection strategies and treatment directions for MHE. The characteristic changes in intestinal microbiota and serum metabolites in MHE patients and the possible relevant interaction mechanisms would inevitably affect the developmental direction of MHE. Therefore, the changes in the characteristics of intestinal microbiota and serum metabolites of MHE patients were determined, and the possible interactions between them were analyzed. Stool and serum tests were performed on both the MHE patients and healthy individuals. The 16S rRNA gene high-throughput sequencing and bioinformatics analyses were used to analyze the differences in intestinal microbiota in MHE patients. The serum metabolites were detected using liquid LC-MS/MS (liquid chromatography-mass spectrometry) technology, and the differences in the metabolic networks of blood metabolites in MHE patients were analyzed. A comprehensive bioinformatics analysis approach was adopted to identify the composition and characteristics of microbiota and serum metabolites and the possible correlation between them. The main characteristics of the structural imbalance in the intestinal microbiota of MHE patients included a decrease in the number of beneficial bacteria at the levels of phylum, class, order, family, and genus and an increase in the pathogenic bacteria, resulting in substantial changes in the relative abundances of bacteria in the intestinal microbiota. The main predicted functions that showed significant differences included chromosome, amino acid-related enzymes, methane metabolism, and arginine and proline metabolism. The detection of serum metabolites resulted in 10 different metabolites, including taurocholic acid, citrulline, d-phenyl-lactic acid, l-tyrosine, benzoate, phenylalanine, linoleic acid, eicosapedienic acid, alpha-dimorphecolic acid, and dehydroepiandrosterone. The subsequent metabolite pathways analysis showed differences in the metabolism of linoleic acid, phenyl-propane, caffeine, arginine, proline, glycine, serine, threonine, tyrosine, and pyrimidine compared to the control group. In summary, it seems that the changes in the microbiome that we have identified have resulted in corresponding changes to the serum metabolome. In turn, this may represent changes in the absorption of metabolites from the gut or reflect the changed metabolic capacity of the MHE liver or both. There were characteristic changes in the intestinal microbiota and serum metabolites in the MHE patients. There might be a related interaction mechanism between the two, which would provide evidence and direction for the detection and treatment strategies of MHE.
Collapse
Affiliation(s)
- Yong Lin
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China
| | - Gengjie Yan
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China
| | - Feng Feng
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China
| | - Minggang Wang
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China
| | - Fuli Long
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China
| |
Collapse
|
49
|
Zajac DJ, Green SJ, Johnson LA, Estus S. APOE genetics influence murine gut microbiome. Sci Rep 2022; 12:1906. [PMID: 35115575 PMCID: PMC8814305 DOI: 10.1038/s41598-022-05763-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/17/2022] [Indexed: 01/03/2023] Open
Abstract
Apolipoprotein E (APOE) alleles impact pathogenesis and risk for multiple human diseases, making them primary targets for disease treatment and prevention. Previously, we and others reported an association between APOE alleles and the gut microbiome. Here, we evaluated effects of APOE heterozygosity and tested whether these overall results extended to mice maintained under ideal conditions for microbiome analyses. To model human APOE alleles, this study used APOE targeted replacement (TR) mice on a C57Bl/6 background. To minimize genetic drift, homozygous APOE3 mice were crossed to homozygous APOE2 or homozygous APOE4 mice prior to the study, and the resulting heterozygous progeny crossed further to generate the study mice. To maximize environmental homogeneity, mice with mixed genotypes were housed together and used bedding from the cages was mixed and added back as a portion of new bedding. Fecal samples were obtained from mice at 3-, 5- and 7-months of age, and microbiota analyzed by 16S ribosomal RNA gene amplicon sequencing. Linear discriminant analysis of effect size (LefSe) identified taxa associated with APOE status, depicted as cladograms to show phylogenetic relatedness. The influence of APOE status was tested on alpha-diversity (Shannon H index) and beta-diversity (principal coordinate analyses and PERMANOVA). Individual taxa associated with APOE status were identified by classical univariate analysis. Whether findings in the APOE mice were replicated in humans was evaluated by using published microbiome genome wide association data. Cladograms revealed robust differences with APOE in male mice and limited differences in female mice. The richness and evenness (alpha-diversity) and microbial community composition (beta-diversity) of the fecal microbiome was robustly associated with APOE status in male but not female mice. Classical univariate analysis revealed individual taxa that were significantly increased or decreased with APOE, illustrating a stepwise APOE2-APOE3-APOE4 pattern of association with heterozygous animals trending as intermediate in the stepwise pattern. The relative abundance of bacteria from the class Clostridia, order Clostridiales, family Ruminococacceae and related genera increased with APOE2 status. The relative abundance of Erysipelotrichia increased with APOE4 status, a finding that extended to humans. In this study, wherein mice were maintained in an ideal fashion for microbiome studies, gut microbiome profiles were strongly and significantly associated with APOE status in male APOE-TR mice. Erysipelotrichia are increased with APOE4 in both mice and humans. APOE allelic effects appeared generally intermediate in heterozygous animals. Further evaluation of these findings in humans, as well as studies evaluating the impact of the APOE-associated microbiota on disease-relevant phenotypes, will be necessary to determine if alterations in the gut microbiome represent a novel mechanism whereby APOE alleles impact disease.
Collapse
Affiliation(s)
- Diana J Zajac
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Stefan J Green
- Genome Research Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, USA
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL, USA
| | - Lance A Johnson
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Steven Estus
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.
- Department of Physiology, University of Kentucky, 789 S. Limestone, Rm. 537, Lexington, KY, 40536, USA.
| |
Collapse
|
50
|
Metagenomic Comparisons between Soft and Hard Feces of Plateau Pikas ( Ochotona curzoniae). Animals (Basel) 2022; 12:ani12020149. [PMID: 35049773 PMCID: PMC8772556 DOI: 10.3390/ani12020149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Plateau pika produces hard and soft feces with different morphology, component and microbial structure. Hard feces had more abundant Firmicutes, while soft feces had more abundant Akkermansia. The differences of microbial communities between hard and soft feces were mainly driven by core microbomes. Soft feces had a comprehensive advances in predict functional pathways compared to hard feces, these strengthened functional pathways were closely associated with metabolism of energy, vitamins, and amino acid. Our study preliminarily explored the differences in microbial structure and function between hard and soft feces, provided a foundation for future systematic explorations of the cecotrophy. Abstract The division of hard and soft feces is an effective digestion strategy in the order Lagomorpha. Although previous studies have reported that hard and soft feces differ in morphology and component, the discrepancy in the microbiome remains unclear. This study explored the microbiomes of hard and soft feces in plateau pikas by sequencing the V3 and V4 regions of 16S rDNA. We found that hard feces harbored higher Firmicutes, while soft feces harbored higher Akkermansia. Increased rare bacterial taxa were observed in hard feces compared with soft feces. Moreover, hard and soft feces displayed a greater difference in terms of core operational taxonomy units (OTUs) compared to the total OTUs. The soft feces showed enhancements in all predicted Kyoto Encyclopedia of Genes and Genomes (KEGG) functions, indicating an advancing microbial metabolism compared to hard feces. The significantly upregulated pathways in soft feces were mainly enriched in metabolism of energy and carbohydrate, glycan biosynthesis, cofactors and vitamins, and amino acids—all of which are associated with increased contents of microbial proteins, vitamins, and short-chain fatty acids. Our study reports, for the first time, the differential microbiomes between hard and soft feces of pikas and provides direction for the future studies on cecotrophy.
Collapse
|