1
|
Wu Z, Liu H, Yan L, Deng Y, Tian Z, Du Y, Zhao Y, Ma H, Deng Y, Li Y, Wang Z. Imaging of Gut Bacterial Macroscopic Changes in Simulated Microgravity-Exposed Rats via In Vivo Metabolic Labeling. Anal Chem 2024; 96:19758-19767. [PMID: 39591367 DOI: 10.1021/acs.analchem.4c05028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
The impact of the microgravity environment on gut bacteria has been widely recognized to induce notable gastrointestinal pathology during extended spaceflight. However, most current studies for gut microbiome homeostasis profiling are based on the 16S rRNA gene sequencing of fecal samples; this technology faces challenges in analyzing gut bacterial alterations in situ, dynamically, and with high spatiotemporal resolution. Herein, we present the utilization of bioorthogonal metabolic labeling for noninvasive imaging of gut bacterial macroscopic changes in simulated microgravity (SMG) rats. After being subsequently labeled with the metabolic reporters d-Ala-N3 and ICG-DBCO through click chemistry, it was shown that SMG can trigger obvious perturbation of gut bacteria, evidenced by the significant increase in the total bacterial content and spatial distribution variations. Such a difference was accompanied by the occurrence of intestinal inflammation and tissue damage. Compared with 16S rRNA genome analysis focusing on composition and diversity, the metabolic labeling strategy provides unprecedented insights into the macroscopic changes of the gut bacterial content and distribution under SMG. Our study will be helpful for investigating the biological implication of SMG-induced imbalance in gut bacteria, potentially promoting the deep investigation of the complex gastrointestinal pathology in space biomedicine.
Collapse
Affiliation(s)
- Zhujun Wu
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Huayan Liu
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Liben Yan
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Yifan Deng
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Zhongqin Tian
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yiyang Du
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yuankun Zhao
- Advanced Research Institute of Multidisciplinary Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Hong Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Yulin Deng
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Yujuan Li
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Zhimin Wang
- Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
2
|
Huang Q, Ma F, Jin Y, Gao D, Chang M, Sun P. The dynamic distribution of the rectal microbiota in Holstein dairy calves provides a framework for understanding early-life gut health. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 19:301-312. [PMID: 39640550 PMCID: PMC11617247 DOI: 10.1016/j.aninu.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 12/07/2024]
Abstract
The posterior intestinal microbiota plays a vital role in the growth and health of Holstein dairy calves. However, its establishment and dynamic changes during early development remain unclear. The aim of this study was to investigate microbial colonization and development in the rectum of calves within the first 70 d after birth. Here, 96 rectal content samples were collected from 8 Holstein dairy calves at 12 time points and analyzed using 16S rRNA gene sequencing. The microbial alpha diversity increased with age. The bacterial community displayed a distinct dynamic distribution. The phylum Proteobacteria was replaced by Firmicutes and Bacteroidetes after d 3. The colonization process of bacterial genera in the rectum of neonatal calves can be divided into 2 periods: the colonization period (stage 1: d 1 and stage 2: d 3) and the stable period (stage 3: d 7-14, stage 4: d 21-42, and stage 5: d 49-70). The fermentation pattern and metabolic function changed from propionate fermentation dominated by Shigella to lactic acid fermentation dominated by Lactobacillus, Blautia, and Oscillospira. The stable period was more comprehensive and complete than the colonization period. This study revealed the dynamic changes in the posterior intestinal microbiota of Holstein dairy calves during early development. The transition period (d 7-14) was identified as a key stage for early nutritional intervention, as the abundance of Lactobacillus increased and the abundance of harmful bacteria (such as Proteobacteria and Shigella) decreased. This study provides a framework for understanding early-life gut health and offers theoretical guidance for future research on host-microbe interactions and early nutritional interventions. It is suggested that nutritional interventions based on microbial characteristics at different stages be implemented to improve calf growth performance and immune function, which may contribute to the reduction of diarrhea and other gastrointestinal disorders during dairy production.
Collapse
Affiliation(s)
- Qi Huang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Fengtao Ma
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yuhang Jin
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Duo Gao
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Meinan Chang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Peng Sun
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
3
|
Marchal S, Choukér A, Bereiter-Hahn J, Kraus A, Grimm D, Krüger M. Challenges for the human immune system after leaving Earth. NPJ Microgravity 2024; 10:106. [PMID: 39557881 PMCID: PMC11574097 DOI: 10.1038/s41526-024-00446-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 11/02/2024] [Indexed: 11/20/2024] Open
Abstract
From the start of life on Earth, several immune defense mechanisms have evolved to guarantee cellular integrity, homeostasis, and host survival. All these sophisticated balances as shaped by and towards the environmental needs have occurred over hundreds of millions of years. Human spaceflight involves various health hazards, such as higher levels of radiation, altered gravity, isolation and confinement, living in tight quarters, and stress associated with being away from home. A growing body of evidence points towards immunological changes in astronauts, including heightened pro-inflammatory responses, reactivation of latent viruses, and cell-mediated alterations, reflecting a dysbalanced state in astronauts. Simultaneously, enhanced pathogenicity, virulence, and drug resistance properties of microorganisms tip the scale out of favor for prolonged stay in space. As we have learned from the past, we see potential for the human immune system, forged and maintained throughout evolutionary history, to adapt to the space exposome. It is unlikely that this will happen in the short time frames set for current space exploration missions. Instead, major risks to astronaut health need to be addressed first, before humans can safely evolve into the space environment.
Collapse
Affiliation(s)
- Shannon Marchal
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke University, Universitätsplatz 2, Magdeburg, Germany
| | - Alexander Choukér
- Laboratory of Translational Research "Stress and Immunity", Department of Anesthesiology, LMU University Hospital, LMU Munich, Marchioninistr. 15, Munich, Germany
| | - Jürgen Bereiter-Hahn
- Institute for Cell Biology and Neurosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Armin Kraus
- Clinic for Plastic, Aesthetic and Hand Surgery, University Hospital Magdeburg, Magdeburg, Germany
- Research Group "Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen" (MARS), Otto-von-Guericke University, Universitätsplatz 2, Magdeburg, Germany
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke University, Universitätsplatz 2, Magdeburg, Germany
- Research Group "Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen" (MARS), Otto-von-Guericke University, Universitätsplatz 2, Magdeburg, Germany
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke University, Universitätsplatz 2, Magdeburg, Germany.
- Research Group "Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen" (MARS), Otto-von-Guericke University, Universitätsplatz 2, Magdeburg, Germany.
| |
Collapse
|
4
|
Aksoyalp ZŞ, Temel A, Karpuz M. Pharmacological Innovations in Space: Challenges and Future Perspectives. Pharm Res 2024; 41:2095-2120. [PMID: 39532779 DOI: 10.1007/s11095-024-03788-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Since the first human experience in space, the interest in space research and medicine to explore universe is growing day by day. The extreme space conditions mainly radiation and microgravity effects on human physiology, antimicrobial susceptibility, and efficacy, safety, and stability of drugs. Therefore, the aim of this review is to address the impact of extreme space conditions, mainly microgravity and radiation, on human physiology and highlights the need for future approaches by evaluating the effectiveness of strategies to prevent or mitigate health problems. METHODS Published papers and NASA technical documents were searched in Pubmed and Google Scholar databases using the keywords ''antimicrobial susceptibility or drug resistance or drug stability or innovations or pharmacokinetic or pharmacodynamics'' and ''radiation or microgravity or space environments or space medicine or space pharmacy'' to prepare this review. RESULTS In this review, the challenges regarding physiological effects and drug-related problems are examined through the evaluation of extreme conditions in space. Medications used in spaceflight are summarized, and the role of pharmacists specializing in space medicine is briefly explained. Last but not least, to overcome the aforementioned issues, novel approaches have been addressed, such as personalised treatments, development of space-resistant formulations and various microbial applications. CONCLUSIONS Further research in the space medicine is required to facilitate the safe and healthy travel of humans to the Moon, Mars and other extraterrestrial destinations. One bear in mind that space research will contribute not only to the exploration of the universe, but also to the advancement of health and technological discoveries on Earth.
Collapse
Affiliation(s)
- Zinnet Şevval Aksoyalp
- Department of Pharmacology, Faculty of Pharmacy, Izmir Katip Celebi University, 35620, Izmir, Türkiye
| | - Aybala Temel
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Izmir Katip Celebi University, 35620, Izmir, Türkiye
| | - Merve Karpuz
- Department of Radiopharmacy, Faculty of Pharmacy, Izmir Katip Celebi University, 35620, Izmir, Türkiye.
| |
Collapse
|
5
|
Minoretti P, Fontana JM, Yilmaz Y. Pilots, Astronauts, and the Aerospace Microbiota: A Narrative Review of Occupational Impact. Cureus 2024; 16:e72268. [PMID: 39583430 PMCID: PMC11585399 DOI: 10.7759/cureus.72268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 10/23/2024] [Indexed: 11/26/2024] Open
Abstract
The human microbiota plays a crucial role in maintaining health and preventing disease; however, the effects of occupational exposure on the microbiota of aircrew and astronauts are not fully understood. This narrative review aims to synthesize the current knowledge on microbiota alterations in aerospace medicine, assess the potential of probiotics as a countermeasure, and identify key gaps that warrant further research. The references were identified through searching PubMed for English articles published between 2010 and 2024, using keywords related to microbiota, probiotics, aviation, spaceflight, pilots, and astronauts. Additionally, the bibliographies of relevant papers were reviewed. Studies in aerospace medicine were selected based on their focus on the occupational impact on microbiota and the use of probiotics in this context. For aircrew, initial studies indicate a decrease in beneficial gut bacteria, suggesting that probiotics could enhance gastrointestinal health, immunity, and overall well-being. However, unsupervised use of probiotics carries potential risks. Conversely, spaceflight induces significant changes in the gut, skin, oral, and nasal microbiota of astronauts, characterized by altered diversity and abundance of specific microbial taxa. These changes include a relative decrease in the abundance of beneficial gut bacteria, an increase in opportunistic pathogens, and evidence of microbial transfer between astronauts and spacecraft surfaces. While simulated space studies suggest the potential for probiotics to mitigate dysbiosis, direct testing done during actual spaceflight is lacking. The observed microbiota changes during spaceflight are associated with various health implications, including alterations in metabolic pathways and interactions between the microbial metabolic capabilities and the host's metabolism. In conclusion, this review highlights the profound impact of spaceflight on astronaut microbiota and the promising role of probiotics as an intervention in both space and aviation medicine. However, significant research gaps remain. These include elucidating the functional implications of microbial shifts, developing personalized countermeasures, and validating the efficacy of probiotics during spaceflight. Future studies should leverage advanced tools such as metagenomic analysis and longitudinal tracking of astronaut health to inform targeted interventions that support the well-being of aerospace personnel. Integrating data across different sites of the body and missions, facilitated by resources like the Space Omics and Medical Atlas (SOMA), can help identify consistent microbial changes induced by the unique occupational conditions of spaceflight and aviation. This integrated approach will be crucial for developing effective microbiota-based countermeasures to mitigate the occupational health risks associated with space and aviation.
Collapse
Affiliation(s)
| | | | - Yusuf Yilmaz
- Gastroenterology and Hepatology, Recep Tayyip Erdoğan University, Rize, TUR
| |
Collapse
|
6
|
Nickerson CA, McLean RJC, Barrila J, Yang J, Thornhill SG, Banken LL, Porterfield DM, Poste G, Pellis NR, Ott CM. Microbiology of human spaceflight: microbial responses to mechanical forces that impact health and habitat sustainability. Microbiol Mol Biol Rev 2024; 88:e0014423. [PMID: 39158275 PMCID: PMC11426028 DOI: 10.1128/mmbr.00144-23] [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] [Indexed: 08/20/2024] Open
Abstract
SUMMARYUnderstanding the dynamic adaptive plasticity of microorganisms has been advanced by studying their responses to extreme environments. Spaceflight research platforms provide a unique opportunity to study microbial characteristics in new extreme adaptational modes, including sustained exposure to reduced forces of gravity and associated low fluid shear force conditions. Under these conditions, unexpected microbial responses occur, including alterations in virulence, antibiotic and stress resistance, biofilm formation, metabolism, motility, and gene expression, which are not observed using conventional experimental approaches. Here, we review biological and physical mechanisms that regulate microbial responses to spaceflight and spaceflight analog environments from both the microbe and host-microbe perspective that are relevant to human health and habitat sustainability. We highlight instrumentation and technology used in spaceflight microbiology experiments, their limitations, and advances necessary to enable next-generation research. As spaceflight experiments are relatively rare, we discuss ground-based analogs that mimic aspects of microbial responses to reduced gravity in spaceflight, including those that reduce mechanical forces of fluid flow over cell surfaces which also simulate conditions encountered by microorganisms during their terrestrial lifecycles. As spaceflight mission durations increase with traditional astronauts and commercial space programs send civilian crews with underlying health conditions, microorganisms will continue to play increasingly critical roles in health and habitat sustainability, thus defining a new dimension of occupational health. The ability of microorganisms to adapt, survive, and evolve in the spaceflight environment is important for future human space endeavors and provides opportunities for innovative biological and technological advances to benefit life on Earth.
Collapse
Affiliation(s)
- Cheryl A. Nickerson
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, Arizona, USA
| | | | - Jennifer Barrila
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, Arizona, USA
| | - Jiseon Yang
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, Arizona, USA
| | | | - Laura L. Banken
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, Arizona, USA
| | - D. Marshall Porterfield
- Department of Agricultural & Biological Engineering, Purdue University, West Lafayette, Indiana, USA
| | - George Poste
- Complex Adaptive Systems Initiative, Arizona State University, Tempe, Arizona, USA
| | | | - C. Mark Ott
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, Texas, USA
| |
Collapse
|
7
|
Ibrahim Z, Khan NA, Siddiqui R, Qaisar R, Marzook H, Soares NC, Elmoselhi AB. Gut matters in microgravity: potential link of gut microbiota and its metabolites to cardiovascular and musculoskeletal well-being. Nutr Metab (Lond) 2024; 21:66. [PMID: 39123239 PMCID: PMC11316329 DOI: 10.1186/s12986-024-00836-6] [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: 02/08/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
The gut microbiota and its secreted metabolites play a significant role in cardiovascular and musculoskeletal health and diseases. The dysregulation of the intestinal microbiota poses a significant threat to cardiovascular and skeletal muscle well-being. Nonetheless, the precise molecular mechanisms underlying these changes remain unclear. Furthermore, microgravity presents several challenges to cardiovascular and musculoskeletal health compromising muscle strength, endothelial dysfunction, and metabolic changes. The purpose of this review is to critically examine the role of gut microbiota metabolites on cardiovascular and skeletal muscle functions and dysfunctions. It also explores the molecular mechanisms that drive microgravity-induced deconditioning in both cardiovascular and skeletal muscle. Key findings in this review highlight that several alterations in gut microbiota and secreted metabolites in microgravity mirror characteristics seen in cardiovascular and skeletal muscle diseases. Those alterations include increased levels of Firmicutes/Bacteroidetes (F/B) ratio, elevated lipopolysaccharide levels (LPS), increased in para-cresol (p-cresol) and secondary metabolites, along with reduction in bile acids and Akkermansia muciniphila bacteria. Highlighting the potential, modulating gut microbiota in microgravity conditions could play a significant role in mitigating cardiovascular and skeletal muscle diseases not only during space flight but also in prolonged bed rest scenarios here on Earth.
Collapse
Affiliation(s)
- Zeinab Ibrahim
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Naveed A Khan
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Ruqaiyyah Siddiqui
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS,, UK
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Rizwan Qaisar
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Hezlin Marzook
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Nelson C Soares
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid university of Medicine and Health Sciences, Dubai, 0000, United Arab Emirates
- Laboratory of Proteomics, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Av Padre Cruz, Lisbon, 1649-016, Portugal
| | - Adel B Elmoselhi
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE.
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
8
|
Rutter LA, Cope H, MacKay MJ, Herranz R, Das S, Ponomarev SA, Costes SV, Paul AM, Barker R, Taylor DM, Bezdan D, Szewczyk NJ, Muratani M, Mason CE, Giacomello S. Astronaut omics and the impact of space on the human body at scale. Nat Commun 2024; 15:4952. [PMID: 38862505 PMCID: PMC11166943 DOI: 10.1038/s41467-024-47237-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 03/22/2024] [Indexed: 06/13/2024] Open
Abstract
Future multi-year crewed planetary missions will motivate advances in aerospace nutrition and telehealth. On Earth, the Human Cell Atlas project aims to spatially map all cell types in the human body. Here, we propose that a parallel Human Cell Space Atlas could serve as an openly available, global resource for space life science research. As humanity becomes increasingly spacefaring, high-resolution omics on orbit could permit an advent of precision spaceflight healthcare. Alongside the scientific potential, we consider the complex ethical, cultural, and legal challenges intrinsic to the human space omics discipline, and how philosophical frameworks may benefit from international perspectives.
Collapse
Affiliation(s)
- Lindsay A Rutter
- Transborder Medical Research Center, University of Tsukuba, 305-8575, Tsukuba, Japan
- Department of Genome Biology, Institute of Medicine, University of Tsukuba, 305-8575, Tsukuba, Japan
- School of Chemistry, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Henry Cope
- School of Medicine, University of Nottingham, Derby, DE22 3DT, UK
| | - Matthew J MacKay
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
- The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Raúl Herranz
- Centro de Investigaciones Biológicas "Margarita Salas" (CSIC), Ramiro de Maeztu 9, Madrid, 28040, Spain
| | - Saswati Das
- Department of Biochemistry, Atal Bihari Vajpayee Institute of Medical Sciences & Dr. Ram Manohar Lohia Hospital, New Delhi, 110001, India
| | - Sergey A Ponomarev
- Department of Immunology and Microbiology, Institute for the Biomedical Problems, Russian Academy of Sciences, 123007, Moscow, Russia
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Amber M Paul
- Embry-Riddle Aeronautical University, Department of Human Factors and Behavioral Neurobiology, Daytona Beach, FL, 32114, USA
| | - Richard Barker
- Department of Botany, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Deanne M Taylor
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Daniela Bezdan
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, 72076, Germany
- NGS Competence Center Tübingen (NCCT), University of Tübingen, Tübingen, 72076, Germany
- yuri GmbH, Meckenbeuren, 88074, Germany
| | - Nathaniel J Szewczyk
- School of Medicine, University of Nottingham, Derby, DE22 3DT, UK
- Ohio Musculoskeletal and Neurological Institute (OMNI), Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Masafumi Muratani
- Transborder Medical Research Center, University of Tsukuba, 305-8575, Tsukuba, Japan
- Department of Genome Biology, Institute of Medicine, University of Tsukuba, 305-8575, Tsukuba, Japan
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA.
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA.
- The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10065, USA.
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10065, USA.
| | | |
Collapse
|
9
|
Wang M, Chen S, Zhong C, Liu L, Wang G, Huang X, Yang X, Yang H, Li L. The influence of simulated weightlessness on the composition and function of gut microbiota and bile acid metabolism products. LIFE SCIENCES IN SPACE RESEARCH 2024; 41:18-28. [PMID: 38670645 DOI: 10.1016/j.lssr.2024.01.002] [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: 08/29/2023] [Revised: 11/15/2023] [Accepted: 01/08/2024] [Indexed: 04/28/2024]
Abstract
The aim of this study was to investigate the effects of simulated weightlessness on gut microbiota, bile acid metabolism, and inflammatory cytokines compared to the control group. The study compared the changes in gut microbiota at the phylum and genus levels in the feces of control and weightlessness rats after 1 and 8 weeks using fecal 16S rRNA sequencing. In the weightlessness group, there was an increase in the proportion of anaerobic bacteria and biofilm-forming bacteria, and a decrease in the proportion of aerobic and Gram-negative bacteria. Further investigations explored the impact of weightlessness on bile acid metabolism products. The levels of glycine ursodeoxycholic acid, glycine chenodeoxycholic acid, glycine deoxycholic acid and glycine cholic acid levels were lower in rats undergoing weightlessness for 1 week compared to the control group.Moreover, the study examined the relationship between gut microbiota and bile acid metabolism products.It was observed that, unlike the control group, there were significant positive correlations between Planctomycetes, Proteobacteria, Synergistetes, and GUDCA levels in rats after 1 week of weightlessness. Finally, ELISA results indicated significant differences in the levels of MDA, GSH, NLRP3, and SIgA inflammatory cytokines between rats undergoing weightlessness for 1 week and the control group rats. Our research confirmed that the simulated weightlessness environment significantly affects the gut microbiota and bile acid metabolism in rats, potentially leading to changes in inflammatory cytokines and causing intestinal tissue inflammation. Further exploring the relationship between gut microbiota and bile acid metabolism under weightless conditions will be crucial for understanding the functional changes in the intestines caused by weightlessness.
Collapse
Affiliation(s)
- Min Wang
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, No. 9 Anxiang North Lane, Chaoyang District, Beijing 100101, China
| | - Sheng Chen
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, No. 9 Anxiang North Lane, Chaoyang District, Beijing 100101, China
| | - Changqing Zhong
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, No. 9 Anxiang North Lane, Chaoyang District, Beijing 100101, China
| | - Lei Liu
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, No. 9 Anxiang North Lane, Chaoyang District, Beijing 100101, China
| | - Guodong Wang
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, No. 9 Anxiang North Lane, Chaoyang District, Beijing 100101, China
| | - Xin Huang
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, No. 9 Anxiang North Lane, Chaoyang District, Beijing 100101, China
| | - Xiaoman Yang
- Department of Pathology, PLA Strategic Support Force Characteristic Medical Center, No. 9 Anxiang North Lane, Chaoyang District, Beijing 100101, China
| | - Heming Yang
- Department of General Surgery, PLA Strategic Support Force Characteristic Medical Center, No. 9 Anxiang North Lane, Chaoyang District, Beijing 100101, China.
| | - Lianyong Li
- Department of Gastroenterology, PLA Strategic Support Force Characteristic Medical Center, No. 9 Anxiang North Lane, Chaoyang District, Beijing 100101, China.
| |
Collapse
|
10
|
Liu Y, Xiao S, Wang D, Qin C, Wei H, Li D. A review on separation and application of plant-derived exosome-like nanoparticles. J Sep Sci 2024; 47:e2300669. [PMID: 38651549 DOI: 10.1002/jssc.202300669] [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: 09/12/2023] [Revised: 12/25/2023] [Accepted: 01/04/2024] [Indexed: 04/25/2024]
Abstract
Exosomes-like nanoparticles (ELNs) (exosomes or extracellular vesicles) are vesicle-like bodies secreted by cells. Plant ELNs (PENs) are membrane vesicles secreted by plant cells, with a lipid bilayer as the basic skeleton, enclosing various active substances such as proteins and nucleic acids, which have many physiological and pathological functions. Recent studies have found that the PENs are widespread within different plant species and their biological functions are increasingly recognized. The effective separation method is also necessary for its function and application. Ultracentrifugation, sucrose density gradient ultracentrifugation, ultrafiltration, polymer-based precipitation methods, etc., are commonly used methods for plant exosome-like nanoparticle extraction. In recent years, emerging methods such as size exclusion chromatography, immunoaffinity capture-based technique, and microfluidic technology have shown advancements compared to traditional methods. The standardized separation process for PENs continues to evolve. In this review, we summarized the recent progress in the biogenesis, components, separation methods, and some functions of PENs. When the research on the separation method of PENs and their unique biological structure is further studied. A brand-new idea for the efficient separation and utilization of PENs can be provided in the future, which has a very broad prospect.
Collapse
Affiliation(s)
- Ying Liu
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, China
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin, China
- Engineering Research Center of Forest Bio-Preparation, Ministry of Education, Northeast Forestry University, Harbin, China
| | - Siqiu Xiao
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, China
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin, China
- Engineering Research Center of Forest Bio-Preparation, Ministry of Education, Northeast Forestry University, Harbin, China
| | - Dianbing Wang
- Institute of Biophysics, Chinese Academy of Sciences, Research Center of Biomacromolecules, China Academy of Sciences, National Laboratory of Biomacromolecules, Beijing, China
| | - Chengyu Qin
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, China
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin, China
- Engineering Research Center of Forest Bio-Preparation, Ministry of Education, Northeast Forestry University, Harbin, China
| | - Hongling Wei
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, China
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin, China
- Engineering Research Center of Forest Bio-Preparation, Ministry of Education, Northeast Forestry University, Harbin, China
| | - Dewen Li
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, China
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin, China
- Engineering Research Center of Forest Bio-Preparation, Ministry of Education, Northeast Forestry University, Harbin, China
| |
Collapse
|
11
|
Rithidech KN, Peanlikhit T, Honikel L, Li J, Liu J, Karakach T, Zimmerman T, Welsh J. Consumption of Apigenin Prevents Radiation-induced Gut Dysbiosis in Male C57BL/6J Mice Exposed to Silicon Ions. Radiat Res 2024; 201:317-329. [PMID: 38373016 DOI: 10.1667/rade-23-00110.1] [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: 06/08/2023] [Accepted: 02/06/2024] [Indexed: 02/20/2024]
Abstract
The search for medical treatments to prevent radiation-induced damage to gastrointestinal tissue is crucial as such injuries can be fatal. This study aimed to investigate the effects of apigenin (AP) on the gut microbiome of irradiated mice, as it is a promising radiation countermeasure. Male C57BL/6J mice were divided into four groups, with six mice in each group. Two groups were given food with apigenin (20 mg/kg body weight or AP 20) before and after exposure to 0 or 50 cGy of silicon (28Si) ions, while another two groups of mice received regular diet without apigenin (0 mg/kg body weight or AP 0) before and after irradiation. The duodenum, the primary site for oral AP absorption, was collected from each mouse seven days after radiation exposure. Using 16S rRNA amplicon sequencing, we found significant differences in microbial diversity among groups. Firmicutes and Bacteroidetes were the major phyla for all groups, while actinobacterial and proteobacterial sequences represented only a small percentage. Mice not given dietary apigenin had a higher Firmicutes and Bacteroidetes (F/B) ratio and an imbalanced duodenal microbiota after exposure to radiation, while irradiated mice given apigenin had maintained homeostasis of the microbiota. Additionally, irradiated mice not given apigenin had decreased probiotic bacteria abundance and increased inflammation, while apigenin-supplemented mice had reduced inflammation and restored normal histological structure. In conclusion, our results demonstrate the potential of dietary apigenin as a countermeasure against radiation-induced gut injuries due to its anti-inflammatory activity, reduction of gut microbiota dysbiosis, and increase in probiotic bacteria (e.g., Lachnospiraceae, Muribaculaceae and Bifidobacteriaceae).
Collapse
Affiliation(s)
| | - Tanat Peanlikhit
- Pathology Department, Stony Brook University, Stony Brook, New York 11794-8691
| | - Louise Honikel
- Pathology Department, Stony Brook University, Stony Brook, New York 11794-8691
| | - Jinyu Li
- Pathology Department, Stony Brook University, Stony Brook, New York 11794-8691
| | - Jingxuan Liu
- Pathology Department, Stony Brook University, Stony Brook, New York 11794-8691
| | - Tobias Karakach
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada B3H 4R2
| | - Thomas Zimmerman
- Pathology Department, Stony Brook University, Stony Brook, New York 11794-8691
- Division of Laboratory Animal Resources, Stony Brook University, Stony Brook, New York 11794-8611
| | - James Welsh
- Department of Radiation Oncology, Loyola University Health System, Maywood, Illinois 60153
| |
Collapse
|
12
|
Campisi M, Cannella L, Pavanello S. Cosmic chronometers: Is spaceflight a catalyst for biological ageing? Ageing Res Rev 2024; 95:102227. [PMID: 38346506 DOI: 10.1016/j.arr.2024.102227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/05/2024] [Accepted: 02/06/2024] [Indexed: 02/22/2024]
Abstract
Astronauts returning from space missions often exhibit health issues mirroring age-related conditions, suggesting spaceflight as a potential driver of biological ageing and age-related diseases. To unravel the underlying mechanisms of these conditions, this comprehensive review explores the impact of the space "exposome" on the twelve hallmarks of ageing. Through a meticulous analysis encompassing both space environments and terrestrial analogs, we aim to decipher how different conditions influence ageing hallmarks. Utilizing PubMed, we identified 189 studies and 60 meet screening criteria. Research on biological ageing in space has focused on genomic instability, chronic inflammation, and deregulated nutrient sensing. Spaceflight consistently induces genomic instability, linked to prolonged exposure to ionizing radiation, triggers pro-inflammatory and immune alterations, resembling conditions in isolated simulations. Nutrient sensing pathways reveal increased systemic insulin-like growth-factor-1. Microbiome studies indicate imbalances favoring opportunistic species during spaceflight. Telomere dynamics present intriguing patterns, with lengthening during missions and rapid shortening upon return. Despite a pro-ageing trend, some protective mechanisms emerge. Countermeasures, encompassing dietary adjustments, prebiotics, postbiotics, symbiotics, tailored exercises, meditation, and anti-inflammatory supplements, exhibit potential. Spaceflight's impact on ageing is intricate, with diverse findings challenging established beliefs. Multidisciplinary studies provide guidance for future research in this field.
Collapse
Affiliation(s)
- Manuela Campisi
- Occupational Medicine, Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Luana Cannella
- Occupational Medicine, Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Sofia Pavanello
- Occupational Medicine, Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy.
| |
Collapse
|
13
|
Gan X, Zhao J, Li S, Kan G, Zhang Y, Wang B, Zhang P, Ma X, Tian H, Liao M, Ju D, Xu S, Chen X, Guo J. Simulated space environmental factors of weightlessness, noise and low atmospheric pressure differentially affect the diurnal rhythm and the gut microbiome. LIFE SCIENCES IN SPACE RESEARCH 2024; 40:115-125. [PMID: 38245336 DOI: 10.1016/j.lssr.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/21/2023] [Accepted: 09/19/2023] [Indexed: 01/22/2024]
Abstract
The circadian clock extensively regulates physiology and behavior. In space, astronauts encounter many environmental factors that are dramatically different from those on Earth; however, the effects of these factors on circadian rhythms and the mechanisms remain largely unknown. The present study aimed to investigate the changes in the mouse diurnal rhythm and gut microbiome under simulated space capsule conditions, including microgravity, noise and low atmospheric pressure (LAP). Noise and LAP were loaded in the capsule while the conditions in the animal room remained constant. The mice in the capsule showed disturbed locomotor rhythms and faster adaptation to a 6-h phase advance. RNA sequencing of hypothalamus samples containing the suprachiasmatic nucleus (SCN) revealed that microgravity simulated by hind limb unloading (HU) and exposure to noise and LAP led to decreases in the quantities of differentially expressed genes (DEGs), including circadian clock genes. Changes in the rhythmicity of genes implicated in pathways of cardiovascular deconditioning and more concentrated phases were found under HU or noise and LAP. Furthermore, 16S rRNA sequencing revealed dysbiosis in the gut microbiome, and noise and LAP may repress the temporal discrepancy in the microbiome community structure induced by microgravity. Changes in diurnal oscillations were observed in a number of gut bacteria with critical physiological consequences on metabolism and immunodefense. We also found that the superimposition of noise and LAP may repress normal changes in global gene expression and adaptation in the gut microbiome. Our data demonstrate that in addition to microgravity, exposure to noise and LAP affect the robustness of circadian rhythms and the community structure of the gut microbiome, and these factors may interfere with each other in their adaptation to respective conditions. These findings are important for furthering our understanding of the alterations in circadian rhythms in the complex environment of space.
Collapse
Affiliation(s)
- Xihui Gan
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, School of Life Sciences, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Jianwei Zhao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Silin Li
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, School of Life Sciences, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Guanghan Kan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yin Zhang
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, School of Life Sciences, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Bo Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Peng Zhang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Xiaohong Ma
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, School of Life Sciences, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Hongni Tian
- National Institute of Biological Sciences, Beijing, China
| | - Meimei Liao
- National Institute of Biological Sciences, Beijing, China
| | - Dapeng Ju
- National Institute of Biological Sciences, Beijing, China
| | - Shuihong Xu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Xiaoping Chen
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China; National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China.
| | - Jinhu Guo
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, School of Life Sciences, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
14
|
Liu Z, Luo G, Du R, Kan G, Han X, Zhong G, Xing W, Cui Y, Sun W, Li J, Li Y, Zhao D, Yuan X, Jin X, Han Y, Sun H, Ling S, Li Y. Simulated spaceflight-induced cardiac remodeling is modulated by gut microbial-derived trimethylamine N-oxide. iScience 2023; 26:108556. [PMID: 38125015 PMCID: PMC10730869 DOI: 10.1016/j.isci.2023.108556] [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: 08/28/2023] [Revised: 10/19/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Spaceflight is physically demanding and can negatively affect astronauts' health. It has been shown that the human gut microbiota and cardiac function are affected by spaceflight and simulated spaceflight. This study investigated the effects of the gut microbiota on simulated spaceflight-induced cardiac remodeling using 10° of head-down bed rest (HDBR) in rhesus macaques and 30° of hindlimb unloading (HU) in mice. The gut microbiota, fecal metabolites, and cardiac remodeling were markedly affected by HDBR in macaques and HU in mice, cardiac remodeling in control mice was affected by the gut microbiota of HU mice and that of HU mice was protected by the gut microbiota of control mice, and there was a correlation between cardiac remodeling and the gut microbial-derived metabolite trimethylamine N-oxide. These findings suggest that spaceflight can affect cardiac remodeling by modulating the gut microbiota and fecal metabolites.
Collapse
Affiliation(s)
- Zizhong Liu
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Gui Luo
- Department of Rheumatology and Immunology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ruikai Du
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Guanghan Kan
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Xuan Han
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Guohui Zhong
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Wenjuan Xing
- School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the Ministry of Education, Fourth Military Medical University, Xi’an, China
| | - Ying Cui
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Weijia Sun
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Jianwei Li
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Yuheng Li
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Dingsheng Zhao
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Xinxin Yuan
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Xiaoyan Jin
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Yanping Han
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Huiyuan Sun
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Shukuan Ling
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| | - Yingxian Li
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
15
|
Ramos-Nascimento A, Grenga L, Haange SB, Himmelmann A, Arndt FS, Ly YT, Miotello G, Pible O, Jehmlich N, Engelmann B, von Bergen M, Mulder E, Frings-Meuthen P, Hellweg CE, Jordan J, Rolle-Kampczyk U, Armengaud J, Moeller R. Human gut microbiome and metabolite dynamics under simulated microgravity. Gut Microbes 2023; 15:2259033. [PMID: 37749878 PMCID: PMC10524775 DOI: 10.1080/19490976.2023.2259033] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 09/11/2023] [Indexed: 09/27/2023] Open
Abstract
The Artificial Gravity Bed Rest - European Space Agency (AGBRESA) study was the first joint bed rest study by ESA, DLR, and NASA that examined the effect of simulated weightlessness on the human body and assessed the potential benefits of artificial gravity as a countermeasure in an analog of long-duration spaceflight. In this study, we investigated the impact of simulated microgravity on the gut microbiome of 12 participants during a 60-day head-down tilt bed rest at the :envihab facilities. Over 60 days of simulated microgravity resulted in a mild change in the gut microbiome, with distinct microbial patterns and pathway expression in the feces of the countermeasure group compared to the microgravity simulation-only group. Additionally, we found that the countermeasure protocols selectively increased the abundance of beneficial short-chain fatty acids in the gut, such as acetate, butyrate, and propionate. Some physiological signatures also included the modulation of taxa reported to be either beneficial or opportunistic, indicating a mild adaptation in the microbiome network balance. Our results suggest that monitoring the gut microbial catalog along with pathway clustering and metabolite profiling is an informative synergistic strategy to determine health disturbances and the outcome of countermeasure protocols for future space missions.
Collapse
Affiliation(s)
- Ana Ramos-Nascimento
- Institute of Aerospace Medicine, German Aerospace Center (DLR e.V.), Cologne, Germany
| | - Lucia Grenga
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Bagnols sur Cèze, France
| | - Sven-Bastiaan Haange
- Department of Metabolomics, UFZ-Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany
| | - Alexandra Himmelmann
- Institute of Aerospace Medicine, German Aerospace Center (DLR e.V.), Cologne, Germany
| | - Franca Sabine Arndt
- Institute of Aerospace Medicine, German Aerospace Center (DLR e.V.), Cologne, Germany
| | - Yen-Tran Ly
- Institute of Aerospace Medicine, German Aerospace Center (DLR e.V.), Cologne, Germany
| | - Guylaine Miotello
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Bagnols sur Cèze, France
| | - Olivier Pible
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Bagnols sur Cèze, France
| | - Nico Jehmlich
- Department of Metabolomics, UFZ-Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany
| | - Beatrice Engelmann
- Department of Metabolomics, UFZ-Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany
| | - Martin von Bergen
- Department of Metabolomics, UFZ-Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany
| | - Edwin Mulder
- Institute of Aerospace Medicine, German Aerospace Center (DLR e.V.), Cologne, Germany
| | - Petra Frings-Meuthen
- Institute of Aerospace Medicine, German Aerospace Center (DLR e.V.), Cologne, Germany
| | | | - Jens Jordan
- Institute of Aerospace Medicine, German Aerospace Center (DLR e.V.), Cologne, Germany
| | - Ulrike Rolle-Kampczyk
- Department of Metabolomics, UFZ-Helmholtz Centre for Environmental Research Leipzig, Leipzig, Germany
| | - Jean Armengaud
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Bagnols sur Cèze, France
| | - Ralf Moeller
- Institute of Aerospace Medicine, German Aerospace Center (DLR e.V.), Cologne, Germany
| |
Collapse
|
16
|
Fu Y, Zhang K, Shan F, Li J, Wang Y, Li X, Xu H, Qin Z, Zhang L. Metagenomic analysis of gut microbiome and resistome of Whooper and Black Swans: a one health perspective. BMC Genomics 2023; 24:635. [PMID: 37875797 PMCID: PMC10594901 DOI: 10.1186/s12864-023-09742-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 10/13/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND With the promotion of "One Health," the health of animals and their impact on the environment have become major concerns recently. Widely distributed in China, the whooper swans (Cygnus cygnus) and black swans (Cygnus atratus) are not only important to the ecological environment, but they may also potentially influence public health security. The metagenomic approach was adopted to uncover the impacts of the gut microbiota of swans on host and public health. RESULTS In this study, the intestinal microbiome and resistome of migratory whooper swans and captive-bred black swans were identified. The results revealed similar gut microbes and functional compositions in whooper and black swans. Interestingly, different bacteria and probiotics were enriched by overwintering whooper swans. We also found that Acinetobacter and Escherichia were significantly enriched in early wintering period swans and that clinically important pathogens were more abundant in black swans. Whooper swans and black swans are potential reservoirs of antibiotic resistance genes (ARGs) and novel ARGs, and the abundance of novel ARGs in whooper swans was significantly higher than that in black swans. Metagenomic assembly-based host tracking revealed that most ARG-carrying contigs originated from Proteobacteria (mainly Gammaproteobacteria). CONCLUSIONS The results revealed spatiotemporal changes in microbiome and resistome in swans, providing a reference for safeguarding public health security and preventing animal epidemics.
Collapse
Affiliation(s)
- Yin Fu
- College of Veterinary Medicine, Henan Agricultural University, No. 15 Longzihu University Area, Zhengzhou New District, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
- Ministry of Agriculture and Rural Areas Key Laboratory for Quality and Safety Control of Poultry Products, Zhengzhou, 450046, China
| | - Kaihui Zhang
- College of Veterinary Medicine, Henan Agricultural University, No. 15 Longzihu University Area, Zhengzhou New District, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
- Ministry of Agriculture and Rural Areas Key Laboratory for Quality and Safety Control of Poultry Products, Zhengzhou, 450046, China
| | - Fa Shan
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Junqiang Li
- College of Veterinary Medicine, Henan Agricultural University, No. 15 Longzihu University Area, Zhengzhou New District, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
- Ministry of Agriculture and Rural Areas Key Laboratory for Quality and Safety Control of Poultry Products, Zhengzhou, 450046, China
| | - Yilin Wang
- College of Veterinary Medicine, Henan Agricultural University, No. 15 Longzihu University Area, Zhengzhou New District, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
- Ministry of Agriculture and Rural Areas Key Laboratory for Quality and Safety Control of Poultry Products, Zhengzhou, 450046, China
| | - Xiaoying Li
- College of Veterinary Medicine, Henan Agricultural University, No. 15 Longzihu University Area, Zhengzhou New District, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
- Ministry of Agriculture and Rural Areas Key Laboratory for Quality and Safety Control of Poultry Products, Zhengzhou, 450046, China
| | - Huiyan Xu
- College of Veterinary Medicine, Henan Agricultural University, No. 15 Longzihu University Area, Zhengzhou New District, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
- Ministry of Agriculture and Rural Areas Key Laboratory for Quality and Safety Control of Poultry Products, Zhengzhou, 450046, China
| | - Ziyang Qin
- College of Veterinary Medicine, Henan Agricultural University, No. 15 Longzihu University Area, Zhengzhou New District, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
- Ministry of Agriculture and Rural Areas Key Laboratory for Quality and Safety Control of Poultry Products, Zhengzhou, 450046, China
| | - Longxian Zhang
- College of Veterinary Medicine, Henan Agricultural University, No. 15 Longzihu University Area, Zhengzhou New District, Zhengzhou, 450046, China.
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China.
- Ministry of Agriculture and Rural Areas Key Laboratory for Quality and Safety Control of Poultry Products, Zhengzhou, 450046, China.
| |
Collapse
|
17
|
Fu Y, Zhang K, Yang M, Li X, Chen Y, Li J, Xu H, Dhakal P, Zhang L. Metagenomic analysis reveals the relationship between intestinal protozoan parasites and the intestinal microecological balance in calves. Parasit Vectors 2023; 16:257. [PMID: 37525231 PMCID: PMC10388496 DOI: 10.1186/s13071-023-05877-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/07/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND A close connection between a protozoan parasite and the balance of the other gut microbes of the host has been demonstrated. The calves may be naturally co-infected with many parasites, and the co-effects of parasites on other intestinal microbes of calves remain unclear. This study aims to preliminarily reveal the relationship between intestinal parasites and other intestinal microbes in calves. METHODS Fecal samples were collected from four calves with bloody diarrhea, four calves with watery diarrhea, and seven normal calves, and the microbial flora of the samples were analyzed by whole-genome sequencing. Protozoal parasites were detected in the metagenome sequences and identified using polymerase chain reaction (PCR). RESULTS Cryptosporidium, Eimeria, Giardia, Blastocystis, and Entamoeba were detected by metagenomic analysis, and the identified species were Giardia duodenalis assemblage E, Cryptosporidium bovis, Cryptosporidium ryanae, Eimeria bovis, Eimeria subspherica, Entamoeba bovis, and Blastocystis ST2 and ST10. Metagenomic analysis showed that the intestinal microbes of calves with diarrhea were disordered, especially in calves with bloody diarrhea. Furthermore, different parasites show distinct relationships with the intestinal microecology. Cryptosporidium, Eimeria, and Giardia were negatively correlated with various intestinal bacteria but positively correlated with some fungi. However, Blastocystis and Entamoeba were positively associated with other gut microbes. Twenty-seven biomarkers not only were significantly enriched in bloody diarrhea, watery diarrhea, and normal calves but were also associated with Eimeria, Cryptosporidium, and Giardia. Only Eimeria showed a distinct relationship with seven genera of bacteria, which were significantly enriched in the healthy calves. All 18 genera of fungi were positively correlated with Cryptosporidium, Eimeria, and Giardia, which were also significantly enriched in calves with bloody diarrhea. Functional genes related to parasites and diseases were found mainly in fungi. CONCLUSIONS This study revealed the relationship between intestinal protozoan parasites and the other calf gut microbiome. Different intestinal protozoan parasites have diametrically opposite effects on other gut microecology, which not only affects bacteria in the gut, but also is significantly related to fungi and archaea.
Collapse
Affiliation(s)
- Yin Fu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
| | - Kaihui Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
| | - Mengyao Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
| | - Xiaoying Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
| | - Yuancai Chen
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
| | - Junqiang Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
| | - Huiyan Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
| | - Pitambar Dhakal
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China
| | - Longxian Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China.
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, China.
| |
Collapse
|
18
|
Klos B, Steinbach C, Ketel J, Lambert C, Penders J, Doré J, Enck P, Mack I. Effects of isolation and confinement on gastrointestinal microbiota-a systematic review. Front Nutr 2023; 10:1214016. [PMID: 37492598 PMCID: PMC10364611 DOI: 10.3389/fnut.2023.1214016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/21/2023] [Indexed: 07/27/2023] Open
Abstract
Purpose The gastrointestinal (GI) microbiota is a complex and dynamic ecosystem whose composition and function are influenced by many internal and external factors. Overall, the individual GI microbiota composition appears to be rather stable but can be influenced by extreme shifts in environmental exposures. To date, there is no systematic literature review that examines the effects of extreme environmental conditions, such as strict isolation and confinement, on the GI microbiota. Methods We conducted a systematic review to examine the effects of isolated and confined environments on the human GI microbiota. The literature search was conducted according to PRISMA criteria using PubMed, Web of Science and Cochrane Library. Relevant studies were identified based on exposure to isolated and confined environments, generally being also antigen-limited, for a minimum of 28 days and classified according to the microbiota analysis method (cultivation- or molecular based approaches) and the isolation habitat (space, space- or microgravity simulation such as MARS-500 or natural isolation such as Antarctica). Microbial shifts in abundance, alpha diversity and community structure in response to isolation were assessed. Results Regardless of the study habitat, inconsistent shifts in abundance of 40 different genera, mainly in the phylum Bacillota (formerly Firmicutes) were reported. Overall, the heterogeneity of studies was high. Reducing heterogeneity was neither possible by differentiating the microbiota analysis methods nor by subgrouping according to the isolation habitat. Alpha diversity evolved non-specifically, whereas the microbial community structure remained dissimilar despite partial convergence. The GI ecosystem returned to baseline levels following exposure, showing resilience irrespective of the experiment length. Conclusion An isolated and confined environment has a considerable impact on the GI microbiota composition in terms of diversity and relative abundances of dominant taxa. However, due to a limited number of studies with rather small sample sizes, it is important to approach an in-depth conclusion with caution, and results should be considered as a preliminary trend. The risk of dysbiosis and associated diseases should be considered when planning future projects in extreme environments. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42022357589.
Collapse
Affiliation(s)
- Bea Klos
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
| | - Christina Steinbach
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
| | - Jasmin Ketel
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
| | - Claude Lambert
- CIRI–Immunology Lab University Hospital, Saint-Étienne, France
- LCOMS/ENOSIS Université de Lorraine, Metz, France
| | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, Maastricht University Medical Center, CAPHRI Care and Public Health Research Institute, Maastricht, Netherlands
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, Maastricht University Medical Center, School of Nutrition and Translational Research in Metabolism, Maastricht, Netherlands
| | - Joël Doré
- UMR Micalis Institut, INRA, Paris-Saclay University, Jouy-En-Josas, France
| | - Paul Enck
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
| | - Isabelle Mack
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
19
|
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
|
20
|
Li Y, Liu Z, Luo G, Lan H, Chen P, Du R, Jing G, Liu L, Cui X, Li Y, Han Y, Xu J, Zhu H, Ling S, Li Y. Effects of 60 days of 6° head-down bed rest on the composition and function of the human gut microbiota. iScience 2023; 26:106615. [PMID: 37250329 PMCID: PMC10214410 DOI: 10.1016/j.isci.2023.106615] [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: 12/10/2022] [Revised: 01/28/2023] [Accepted: 04/04/2023] [Indexed: 05/31/2023] Open
Abstract
Spaceflight is rigorous and dangerous environment which can negatively affect astronauts' health and the entire mission. The 60 days of 6° head-down bed rest (HDBR) experiment provided us with an opportunity to trace the change of gut microbiota under simulated microgravity. The gut microbiota of volunteers was analyzed and characterized by 16S rRNA gene sequencing and metagenomic sequencing. Our results showed that the composition and function of the volunteers' gut microbiota were markedly was affected by 60 days of 6° HDBR. We further confirmed the species and diversity fluctuations. Resistance and virulence genes in the gut microbiota were also affected by 60 days of 6° HDBR, but the species attributions remained stable. The human gut microbiota affected by 60 days of 6° HDBR which was partially consistent with the effect of spaceflight, this implied that HDBR was a simulation of how spaceflight affects the human gut microbiota.
Collapse
Affiliation(s)
- Yixuan Li
- School of Life Sciences, Ludong University, Yantai, China
- Yantai Hi-tech Industrial Development Zone Center for Disease Control and Prevention, Yantai, China
| | - Zizhong Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Gui Luo
- Department of Rheumatology, Chinese PLA General Hospital, Beijing, China
| | - Haiyun Lan
- Key Lab of Space Nutrition and Food Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Pu Chen
- Key Lab of Space Nutrition and Food Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Ruikai Du
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Gongchao Jing
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Shandong Energy Institute, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, China
| | - Lu Liu
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Shandong Energy Institute, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, China
| | - Xiaohan Cui
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Shandong Energy Institute, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, China
| | - Yongzhi Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yanping Han
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Jian Xu
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Shandong Energy Institute, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, China
| | - Hongwei Zhu
- School of Life Sciences, Ludong University, Yantai, China
| | - Shukuan Ling
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang 325000, P.R. China
| | - Yingxian Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
21
|
Bharindwal S, Goswami N, Jha P, Pandey S, Jobby R. Prospective Use of Probiotics to Maintain Astronaut Health during Spaceflight. Life (Basel) 2023; 13:life13030727. [PMID: 36983881 PMCID: PMC10058446 DOI: 10.3390/life13030727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/30/2023] Open
Abstract
Maintaining an astronaut's health during space travel is crucial. Multiple studies have observed various changes in the gut microbiome and physiological health. Astronauts on board the International Space Station (ISS) had changes in the microbial communities in their gut, nose, and skin. Additionally, immune system cell alterations have been observed in astronauts with changes in neutrophils, monocytes, and T-cells. Probiotics help tackle these health issues caused during spaceflight by inhibiting pathogen adherence, enhancing epithelial barrier function by reducing permeability, and producing an anti-inflammatory effect. When exposed to microgravity, probiotics demonstrated a shorter lag phase, faster growth, improved acid tolerance, and bile resistance. A freeze-dried Lactobacillus casei strain Shirota capsule was tested for its stability on ISS for a month and has been shown to enhance innate immunity and balance intestinal microbiota. The usage of freeze-dried spores of B. subtilis proves to be advantageous to long-term spaceflight because it qualifies for all the aspects tested for commercial probiotics under simulated conditions. These results demonstrate a need to further study the effect of probiotics in simulated microgravity and spaceflight conditions and to apply them to overcome the effects caused by gut microbiome dysbiosis and issues that might occur during spaceflight.
Collapse
Affiliation(s)
- Sahaj Bharindwal
- Amity Centre of Excellence in Astrobiology, Amity University Mumbai, Mumbai 410206, Maharashtra, India
- Department of Biology, University of Naples Federico II, 80131 Naples, Italy
- Amity Institute of Biotechnology, Amity University Maharashtra, Mumbai 410206, Maharashtra, India
| | - Nidhi Goswami
- Amity Centre of Excellence in Astrobiology, Amity University Mumbai, Mumbai 410206, Maharashtra, India
- Amity Institute of Biotechnology, Amity University Maharashtra, Mumbai 410206, Maharashtra, India
| | - Pamela Jha
- Sunandan Divatia School of Science, NMIMS University Mumbai, Mumbai 400056, Maharashtra, India
| | - Siddharth Pandey
- Amity Centre of Excellence in Astrobiology, Amity University Mumbai, Mumbai 410206, Maharashtra, India
| | - Renitta Jobby
- Amity Centre of Excellence in Astrobiology, Amity University Mumbai, Mumbai 410206, Maharashtra, India
- Amity Institute of Biotechnology, Amity University Maharashtra, Mumbai 410206, Maharashtra, India
| |
Collapse
|
22
|
Theotokis P, Manthou ME, Deftereou TE, Miliaras D, Meditskou S. Addressing Spaceflight Biology through the Lens of a Histologist-Embryologist. Life (Basel) 2023; 13:life13020588. [PMID: 36836946 PMCID: PMC9965490 DOI: 10.3390/life13020588] [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: 11/28/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023] Open
Abstract
Embryogenesis and fetal development are highly delicate and error-prone processes in their core physiology, let alone if stress-associated factors and conditions are involved. Space radiation and altered gravity are factors that could radically affect fertility and pregnancy and compromise a physiological organogenesis. Unfortunately, there is a dearth of information examining the effects of cosmic exposures on reproductive and proliferating outcomes with regard to mammalian embryonic development. However, explicit attention has been given to investigations exploring discrete structures and neural networks such as the vestibular system, an entity that is viewed as the sixth sense and organically controls gravity beginning with the prenatal period. The role of the gut microbiome, a newly acknowledged field of research in the space community, is also being challenged to be added in forthcoming experimental protocols. This review discusses the data that have surfaced from simulations or actual space expeditions and addresses developmental adaptations at the histological level induced by an extraterrestrial milieu.
Collapse
Affiliation(s)
- Paschalis Theotokis
- Laboratory of Histology and Embryology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Maria Eleni Manthou
- Laboratory of Histology and Embryology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | | | - Dimosthenis Miliaras
- Laboratory of Histology and Embryology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Soultana Meditskou
- Laboratory of Histology and Embryology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Correspondence:
| |
Collapse
|
23
|
Yuan L, Zhang R, Li X, Gao C, Hu X, Hussain S, Zhang L, Wang M, Ma X, Pan Q, Lou X, Si S. Long-term simulated microgravity alters gut microbiota and metabolome in mice. Front Microbiol 2023; 14:1100747. [PMID: 37032862 PMCID: PMC10080065 DOI: 10.3389/fmicb.2023.1100747] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/09/2023] [Indexed: 04/11/2023] Open
Abstract
Spaceflight and microgravity has a significant impact on the immune, central nervous, bone, and muscle support and cardiovascular systems. However, limited studies are available on the adverse effects of long-term microgravity on the intestinal microbiota, metabolism, and its relationships. In this study, a ground-based simulated microgravity (SMG) mouse model was established to evaluate the impact of long-term microgravity on gut microbiota and metabolome. After 8 weeks of SMG, alterations of the intestinal microbiota and metabolites were detected using 16S rRNA sequencing and untargeted metabolomics. Compared to the control, no significant differences in α-diversity were observed at weeks 2, 4 and 8. Nevertheless, there were clear differences in community structures at different time points. The phylum Verrucomicrobia significantly declined from 2 to 8 weeks of SMG, yet the relative abundance of Actinobacteria and Deferribacteres expanded remarkably at weeks 8. SMG decreased the genus of Allobaculum and increased Bacteroides significantly throughout the period of 8 weeks. Besides, Genus Akkermansia, Gracilibacter, Prevotella, Odoribacter, Rothia, Sporosarcina, Gracilibacter, Clostridium, and Mucispirillum were identified as biomarkers for SMG group. Desulfovibrio_c21_c20, Akkermansia_muciniphila, and Ruminococcus_gnavus dropped at week 2, which tend to recover at week 4, except for Akkermansia_muciniphila. Bacteroides_uniformis and Faecalibacterium_prausnitzii declined significantly, while Ruminococcus_flavefaciens and Mucispirillum_schaedleri elevated at week 8. Furthermore, intestinal metabolome analysis showed that 129 were upregulated and 146 metabolites were downregulated in SMG. Long-term SMG most affected steroid hormone biosynthesis, tryptophan, cysteine, methionine, arginine, proline metabolism, and histidine metabolism. Correlated analysis suggested that the potential beneficial taxa Allobaculum, Akkermansia, and Faecalibacterium were negatively associated with tryptophan, histidine, arginine, and proline metabolism, but positively with steroid hormone biosynthesis. Yet Bacteroides, Lachnospiraceae_Clostridium, Rothia, Bilophila, and Coprococcus were positively correlated with arginine, proline, tryptophan, and histidine metabolism, while negatively associated with steroid hormone biosynthesis. These results suggest that Long-term SMG altered the community of intestinal microbiota, and then further disturbed intestinal metabolites and metabolic pathways, which have great potential to help understand and provide clues for revealing the mechanisms of long-term SMG involved diseases.
Collapse
Affiliation(s)
- Lu Yuan
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Rong Zhang
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Xinlou Li
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Caiyun Gao
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Xiangnan Hu
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Safdar Hussain
- Center for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Linlin Zhang
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Moye Wang
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Xiaoyu Ma
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Qiuxia Pan
- Department of Traditional Chinese Medicine, PLA Strategic Support Force Medical Center, Beijing, China
| | - Xiaotong Lou
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
- *Correspondence: Xiaotong Lou,
| | - Shaoyan Si
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
- Shaoyan Si,
| |
Collapse
|
24
|
Zong B, Wang Y, Wang J, Zhang P, Kan G, Li M, Feng J, Wang Y, Chen X, Jin R, Ge Q. Effects of long-term simulated microgravity on liver metabolism in rhesus macaques. FASEB J 2022; 36:e22536. [PMID: 36070186 DOI: 10.1096/fj.202200544rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/29/2022] [Accepted: 08/24/2022] [Indexed: 11/11/2022]
Abstract
The liver is an essential multifunctional organ and constantly communicates with nearly all the tissues in the body. Spaceflight or simulated microgravity has a significant impact on the livers of rodent models, including lipid accumulation and inflammatory cell infiltration. Whether similar liver lipotoxicity could occur in humans is not known, even though altered circulating cholesterol profile has been reported in astronauts. Using a 42-day head-down bed rest (HDBR) model in rhesus macaques, the present study investigated whether simulated microgravity alters the liver of non-human primates at the transcriptome and metabolome levels. Its association with stress and intestinal changes was also explored. Compared to the controls, the HDBR monkeys showed mild liver injury, elevated ANGPTL3 level in the plasma, and accumulation of fat vacuoles and inflammatory cells in the liver. Altered transcriptome signatures with up-regulation of genes in lipid metabolisms and down-regulation of genes in innate immune defense were also found in HDBR group-derived liver samples. The metabolic profiling of the liver revealed mildly disturbed fatty acid metabolism in the liver of HDBR monkeys. The intestinal dysbiosis, its associated endotoxemia and changes in the composition of bile acids, and elevated stress hormone in HDBR monkeys may contribute to the altered lipid metabolisms in the liver. These data indicate that liver metabolic functions and gut-liver axis should be closely monitored in prolonged spaceflight to facilitate strategy design to improve and maintain metabolic homeostasis.
Collapse
Affiliation(s)
- Beibei Zong
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yujia Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Jingyi Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Peng Zhang
- State Key Laboratory of Space Medicine Fundamentals and Application, Chinese Astronaut Research and Training Center, Beijing, China
| | - Guanghan Kan
- State Key Laboratory of Space Medicine Fundamentals and Application, Chinese Astronaut Research and Training Center, Beijing, China
| | - Mingyang Li
- Immunology Research Center, Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Juan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Yifan Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Xiaoping Chen
- State Key Laboratory of Space Medicine Fundamentals and Application, Chinese Astronaut Research and Training Center, Beijing, China.,National Key Laboratory of Human Factors Engineering, China Astronauts Research and Training Center, Beijing, China
| | - Rong Jin
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Qing Ge
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.,Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| |
Collapse
|
25
|
Cope H, Willis CR, MacKay MJ, Rutter LA, Toh LS, Williams PM, Herranz R, Borg J, Bezdan D, Giacomello S, Muratani M, Mason CE, Etheridge T, Szewczyk NJ. Routine omics collection is a golden opportunity for European human research in space and analog environments. PATTERNS 2022; 3:100550. [PMID: 36277820 PMCID: PMC9583032 DOI: 10.1016/j.patter.2022.100550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
26
|
Tesei D, Jewczynko A, Lynch AM, Urbaniak C. Understanding the Complexities and Changes of the Astronaut Microbiome for Successful Long-Duration Space Missions. Life (Basel) 2022; 12:life12040495. [PMID: 35454986 PMCID: PMC9031868 DOI: 10.3390/life12040495] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
During space missions, astronauts are faced with a variety of challenges that are unique to spaceflight and that have been known to cause physiological changes in humans over a period of time. Several of these changes occur at the microbiome level, a complex ensemble of microbial communities residing in various anatomic sites of the human body, with a pivotal role in regulating the health and behavior of the host. The microbiome is essential for day-to-day physiological activities, and alterations in microbiome composition and function have been linked to various human diseases. For these reasons, understanding the impact of spaceflight and space conditions on the microbiome of astronauts is important to assess significant health risks that can emerge during long-term missions and to develop countermeasures. Here, we review various conditions that are caused by long-term space exploration and discuss the role of the microbiome in promoting or ameliorating these conditions, as well as space-related factors that impact microbiome composition. The topics explored pertain to microgravity, radiation, immunity, bone health, cognitive function, gender differences and pharmacomicrobiomics. Connections are made between the trifecta of spaceflight, the host and the microbiome, and the significance of these interactions for successful long-term space missions.
Collapse
Affiliation(s)
- Donatella Tesei
- Department of Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria;
| | - Anna Jewczynko
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Anne M. Lynch
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Camilla Urbaniak
- ZIN Technologies Inc., Middleburg Heights, OH 44130, USA
- NASA Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA 91109, USA
- Correspondence:
| |
Collapse
|
27
|
Fan C, Zhang L, Jia S, Tang X, Fu H, Li W, Liu C, Zhang H, Cheng Q, Zhang Y. Seasonal variations in the composition and functional profiles of gut microbiota reflect dietary changes in plateau pikas. Integr Zool 2022; 17:379-395. [PMID: 35051309 PMCID: PMC9305894 DOI: 10.1111/1749-4877.12630] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Seasonal variations in gut microbiota of small mammals and how it is influenced by environmental variables is relatively poorly understood. We sampled 162 wild plateau pikas (Ochotona curzoniae) in four seasons over two and a half years and recorded the air temperature, precipitation, and nutrient content in edible vegetation at the sampling site. After conducting 16S rRNA and shotgun metagenomic sequencing, we found that the highest alpha diversity, the relative abundance of Firmicutes, and the simplest co-occurrence network occurred in winter, whereas that the highest relative abundance of Proteobacteria and the most complex network structure was observed in spring. The highest relative abundance of Verrucomicrobiota and Spirochaetota were seen in summer and autumn, respectively. Air temperature, precipitation, and the contents of crude protein, crude fiber, and polysaccharide in vegetation had significant effects on the seasonal changes in gut microbiota. Diet contributed more to microbial variation than climatic factors. Metagenomic analysis revealed that the amino acid metabolism pathway and axillary activity enzymes were most abundant in summer, while abundance of carbohydrate-binding modules and carbohydrate esterases were highest in spring. These microbial variations were related to the changes in dietary nutrition, indicating that gut microbiota of plateau pika contribute to the efficient use of food resources. This study provides new evidence of how external environmental factors affect the intestinal environment of small mammals. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Chao Fan
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, China.,College of Life Sciences, Qufu Normal University, Qufu, 273165, 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
| | - Shangang Jia
- College of Grassland Science and Technology, China Agricultural University, Beijing, 100193, China
| | - 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
| | - Haibo Fu
- 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
| | - Wenjing Li
- 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
| | - Chuanfa Liu
- 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
| | - He 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.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qi Cheng
- 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
| | - 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
|
28
|
Tong T, Niu X, Li Q, Ling Y, Li Z, Liu J, Zhang M, Bai Z, Xia R, Wu Z, Liu X. The Effect of Lactobacillus plantarum BW2013 on The Gut Microbiota in Mice Analyzed by 16S rRNA Amplicon Sequencing. Pol J Microbiol 2021; 70:235-243. [PMID: 34349813 PMCID: PMC8326986 DOI: 10.33073/pjm-2021-022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/16/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2022] Open
Abstract
Lactobacillus plantarum BW2013 was isolated from the fermented Chinese cabbage. This study aimed to test the effect of this strain on the gut microbiota in BALB/c mice by 16S rRNA amplicon sequencing. The mice were randomly allocated to the control group and three treatment groups of L. plantarum BW2013 (a low-dose group of 108 CFU/ml, a medium-dose group of 109 CFU/ml, and a high-dose group of 1010 CFU/ml). The weight of mice was recorded once a week, and the fecal samples were collected for 16S rRNA amplicon sequencing after 28 days of continuous treatment. Compared with the control group, the body weight gain in the treatment groups was not significant. The 16S rRNA amplicon sequencing analysis showed that both the Chao1 and ACE indexes increased slightly in the medium-dose group compared to the control group, but the difference was not significant. Based on PCoA results, there was no significant difference in β diversity between the treatment groups. Compared to the control group, the abundance of Bacteroidetes increased in the low-dose group. The abundance of Firmicutes increased in the medium-dose group. At the genus level, the abundance of Alloprevotella increased in the low-dose group compared to the control group. The increased abundance of Ruminococcaceae and decreased abundance of Candidatus_Saccharimonas was observed in the medium-dose group. Additionally, the abundance of Bacteroides increased, and Alistipes and Candidatus_Saccharimonas decreased in the high-dose group. These results indicated that L. plantarum BW2013 could ameliorate gut microbiota composition, but its effects vary with the dose.
Collapse
Affiliation(s)
- Tong Tong
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Xiaohui Niu
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Qian Li
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Yuxi Ling
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Zuming Li
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Jia Liu
- Internal Trade Food Science and Technology (Beijing) Co., Ltd, Beijing, China
| | - Michael Zhang
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Zhihui Bai
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Ran Xia
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Zhichao Wu
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Xiu Liu
- China National Research Institute of Food and Fermentation Industies Co., Ltd, Beijing, China
| |
Collapse
|
29
|
Sun P, Yang J, Wang B, Ma H, Zhang Y, Guo J, Chen X, Zhao J, Sun H, Yang J, Yang H, Cui Y. The effects of combined environmental factors on the intestinal flora of mice based on ground simulation experiments. Sci Rep 2021; 11:11373. [PMID: 34059794 PMCID: PMC8166921 DOI: 10.1038/s41598-021-91077-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 05/17/2021] [Indexed: 11/09/2022] Open
Abstract
The composition and function of intestinal microbial communities are important for human health. However, these intestinal floras are sensitive to changes in the environment. Adverse changes to intestinal flora can affect the health of astronauts, resulting in difficulties in implementing space missions. We randomly divided mice into three groups and placed each group in either a normal environment, simulated microgravity environment or a combined effects environment, which included simulated microgravity, low pressure and noise. Fecal samples of the mice were collected for follow-up analysis based on metagenomics technology. With the influence of different space environmental factors, the species composition at the phylum and genus levels were significantly affected by the combined effects environment, especially the abundance of the Firmicutes and Bacteroidetes. Furthermore, screening was conducted to identify biomarkers that could be regarded as environmental markers. And there have also been some noticeable changes in the function of intestinal floras. Moreover, the abundance of antibiotic resistance genes (ARGs) was also found to be changed under different environmental conditions, such as bacitracin and vancomycin. The combined effects environment could significantly affect the species composition, function, and the expression of ARGs of intestinal flora of mice which may provide a theoretical basis for space medical supervision and healthcare.
Collapse
Affiliation(s)
- Peiming Sun
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China
| | - Jiaqi Yang
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China
- Department of General Surgery, The 306th Hospital of Chinese PLA-Peking University Teaching Hospital, Chaoyang District, Beijing, 100101, China
| | - Bo Wang
- China Astronaut Research and Training Center, Haidian District, Beijing, 100094, China
| | - Huan Ma
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Haizhu District, Guangzhou, 510006, China
| | - Yin Zhang
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Haizhu District, Guangzhou, 510006, China
| | - Jinhu Guo
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Haizhu District, Guangzhou, 510006, China
| | - Xiaoping Chen
- China Astronaut Research and Training Center, Haidian District, Beijing, 100094, China
| | - Jianwei Zhao
- China Astronaut Research and Training Center, Haidian District, Beijing, 100094, China
| | - Hongwei Sun
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China
| | - Jianwu Yang
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China
| | - Heming Yang
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China.
| | - Yan Cui
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China.
| |
Collapse
|
30
|
Mikelsaar M, Mändar R. Commentary: Gut Microbiome and Space Travelers' Health: State of the Art and Possible Pro/Prebiotic Strategies for Long-Term Space Missions. Front Physiol 2021; 12:651977. [PMID: 33833693 PMCID: PMC8021706 DOI: 10.3389/fphys.2021.651977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/01/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Marika Mikelsaar
- Department of Microbiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Reet Mändar
- Department of Microbiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
31
|
Abstract
Scientists have invested considerable resources in the study of the microbiota of the human body. These microorganisms play pivotal roles in immunity and disease. Of which, probiotics are live beneficial microorganisms that keep your intestinal or lung microbiota healthy, and occupy a special role in combating the infections. Thus, it is critical to understand their contributions to these processes. Technology can facilitate advanced studies of the microbiota, including how it develops and its positive and negatives effects on the immune system. This paper investigates how several factors (e.g. birth delivery mode, metabolic activities, types of microorganisms, and immune system interactions) affect the microbiota, particularly in early life. The paper also discusses how gastrointestinal microbes in particular may be associated with certain disease processes, such as those related to schizophrenia, autism, and diabetes. Clinical studies show that certain probiotic strains, like Lactobacillus rhamnosus GG and Bifidobacterium animalis ssp. lactis help to prevent infection of pathogenic organisms (both bacterial and viral). This research may yield crucial contributions to disease prevention and public health. The dysbiosis may result in changes in the acquired immunity later on. The probiotic strains can prevent viral replication during SARS-CoV-2 or COVID-19 infection by reducing proinflammatory cytokines. There has been much interest into the intestinal flora as proposed by the diversity, volume, and proposed role in disease. Future research in the field of microbiome should be done in order to uncover their association to gut virome by noting both their influence on each other and relevant health and disease.
Collapse
|
32
|
Shi Z, Qin M, Huang L, Xu T, Chen Y, Hu Q, Peng S, Peng Z, Qu LN, Chen SG, Tuo QH, Liao DF, Wang XP, Wu RR, Yuan TF, Li YH, Liu XM. Human torpor: translating insights from nature into manned deep space expedition. Biol Rev Camb Philos Soc 2020; 96:642-672. [PMID: 33314677 DOI: 10.1111/brv.12671] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/09/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022]
Abstract
During a long-duration manned spaceflight mission, such as flying to Mars and beyond, all crew members will spend a long period in an independent spacecraft with closed-loop bioregenerative life-support systems. Saving resources and reducing medical risks, particularly in mental heath, are key technology gaps hampering human expedition into deep space. In the 1960s, several scientists proposed that an induced state of suppressed metabolism in humans, which mimics 'hibernation', could be an ideal solution to cope with many issues during spaceflight. In recent years, with the introduction of specific methods, it is becoming more feasible to induce an artificial hibernation-like state (synthetic torpor) in non-hibernating species. Natural torpor is a fascinating, yet enigmatic, physiological process in which metabolic rate (MR), body core temperature (Tb ) and behavioural activity are reduced to save energy during harsh seasonal conditions. It employs a complex central neural network to orchestrate a homeostatic state of hypometabolism, hypothermia and hypoactivity in response to environmental challenges. The anatomical and functional connections within the central nervous system (CNS) lie at the heart of controlling synthetic torpor. Although progress has been made, the precise mechanisms underlying the active regulation of the torpor-arousal transition, and their profound influence on neural function and behaviour, which are critical concerns for safe and reversible human torpor, remain poorly understood. In this review, we place particular emphasis on elaborating the central nervous mechanism orchestrating the torpor-arousal transition in both non-flying hibernating mammals and non-hibernating species, and aim to provide translational insights into long-duration manned spaceflight. In addition, identifying difficulties and challenges ahead will underscore important concerns in engineering synthetic torpor in humans. We believe that synthetic torpor may not be the only option for manned long-duration spaceflight, but it is the most achievable solution in the foreseeable future. Translating the available knowledge from natural torpor research will not only benefit manned spaceflight, but also many clinical settings attempting to manipulate energy metabolism and neurobehavioural functions.
Collapse
Affiliation(s)
- Zhe Shi
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.,Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China
| | - Meng Qin
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Lu Huang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632, China
| | - Tao Xu
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qin Hu
- College of Life Sciences and Bio-Engineering, Beijing University of Technology, Beijing, 100024, China
| | - Sha Peng
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Zhuang Peng
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Li-Na Qu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Shan-Guang Chen
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Qin-Hui Tuo
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Duan-Fang Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Xiao-Ping Wang
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Ren-Rong Wu
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China
| | - Ying-Hui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Xin-Min Liu
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.,Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| |
Collapse
|
33
|
Turroni S, Magnani M, Kc P, Lesnik P, Vidal H, Heer M. Gut Microbiome and Space Travelers' Health: State of the Art and Possible Pro/Prebiotic Strategies for Long-Term Space Missions. Front Physiol 2020; 11:553929. [PMID: 33013480 PMCID: PMC7505921 DOI: 10.3389/fphys.2020.553929] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/14/2020] [Indexed: 12/20/2022] Open
Abstract
The upcoming exploration missions will imply a much longer duration than any of the missions flown so far. In these missions, physiological adaptation to the new environment leads to changes in different body systems, such as the cardiovascular and musculoskeletal systems, metabolic and neurobehavioral health and immune function. To keep space travelers healthy on their trip to Moon, Mars and beyond and their return to Earth, a variety of countermeasures need to be provided to maintain body functionality. From research on the International Space Station (ISS) we know today, that for instance prescribing an adequate training regime for each individual with the devices available in the respective spacecraft is still a challenge. Nutrient supply is not yet optimal and must be optimized in exploration missions. Food intake is intrinsically linked to changes in the gut microbiome composition. Most of the microbes that inhabit our body supply ecosystem benefit to the host-microbe system, including production of important resources, bioconversion of nutrients, and protection against pathogenic microbes. The gut microbiome has also the ability to signal the host, regulating the processes of energy storage and appetite perception, and influencing immune and neurobehavioral function. The composition and functionality of the microbiome most likely changes during spaceflight. Supporting a healthy microbiome by respective measures in space travelers might maintain their health during the mission but also support rehabilitation when being back on Earth. In this review we are summarizing the changes in the gut microbiome observed in spaceflight and analog models, focusing particularly on the effects on metabolism, the musculoskeletal and immune systems and neurobehavioral disorders. Since space travelers are healthy volunteers, we focus on the potential of countermeasures based on pre- and probiotics supplements.
Collapse
Affiliation(s)
- Silvia Turroni
- Unit of Microbial Ecology of Health, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Marciane Magnani
- Laboratory of Microbial Processes in Foods, Department of Food Engineering, Federal University of Paraíba, João Pessoa, Brazil
| | - Pukar Kc
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Philippe Lesnik
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Paris, France.,Institute of Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, Paris, France
| | - Hubert Vidal
- CarMeN Laboratory, INSERM, INRA, Université Claude Bernard Lyon 1, Pierre-Benite, France
| | - Martina Heer
- International University of Applied Sciences, Bad Reichenhall, Germany.,Institute of Nutritional and Food Sciences, University of Bonn, Bonn, Germany
| |
Collapse
|
34
|
Wischer D, Schneider D, Poehlein A, Herrmann F, Oruc H, Meinhardt J, Wagner O, Ahmed R, Kharin S, Novikova N, Haag R, Daniel R, Grohmann E. Novel Antimicrobial Cellulose Fleece Inhibits Growth of Human-Derived Biofilm-Forming Staphylococci During the SIRIUS19 Simulated Space Mission. Front Microbiol 2020; 11:1626. [PMID: 32849336 PMCID: PMC7405646 DOI: 10.3389/fmicb.2020.01626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022] Open
Abstract
Two novel antimicrobial surface coatings were assessed for their lasting antibacterial effect under simulated space conditions during the SIRIUS-19 study. Because long-term space travel can affect the human immune system, astronauts are particularly susceptible to infectious disease. Moreover, the space flight environment can alter the composition of microbial communities within the spacecraft and increase bacterial virulence and resistance to antibiotics. In addition to protecting the crew from infection by human pathogens, prevention and elimination of bacterial contamination is important to avoid corrosion and damage of the technical equipment. The antimicrobial coating AGXX® consists of micro-galvanic cells composed of silver and ruthenium which damage bacterial cells through the release of reactive oxygen species. Over the last years, several studies on the antimicrobial effect of AGXX® have demonstrated an effective inhibition of growth and even complete elimination of many pathogenic bacteria – including multiresistant microorganisms – as well as their biofilms. The second antimicrobial coating, GOX, consists of chemically modified graphene oxide. Through a positive surface charge and its flexible scaffold, GOX can multivalently bind and immobilize bacteria via electrostatic attraction. Here, AGXX® and GOX were applied to non-metallic carriers not previously tested. The antimicrobial coated materials, as well as uncoated control samples, were exposed in the SIRIUS artificial space module and analyzed at different time points during the 4-months isolation study. Survival and growth of airborne heterotrophic, aerobic bacteria on the surfaces were assessed by cultivation-based methods, employing growth conditions suitable for potential human pathogens. Human-associated, biofilm-forming Staphylococcus spp. (S. hominis, S. haemolyticus, and S. epidermidis) strongly dominated at all time points, most were resistant against erythromycin, kanamycin, and ampicillin. AGXX® coatings completely inhibited growth of these opportunistic pathogens on all tested surface materials. Particularly, AGXX®-cellulose fleece achieved a clear reduction in bacterial load able to recover post contact. GOX-cellulose fleece effectively immobilized bacteria. Sequence analysis of 16S rRNA gene amplicons revealed that the isolated Staphylococcus spp. did not dominate the overall bacterial community, accounting for only 0.1–0.4% of all sequences. Instead, molecular data revealed Lactobacillus, Comamonas, Pseudomonas, Sporosarcina, and Bacillus as the dominant genera across all samples and time points.
Collapse
Affiliation(s)
- Daniela Wischer
- Faculty of Life Sciences and Technology, Department of Microbiology, Beuth University of Applied Sciences, Berlin, Germany
| | - Dominik Schneider
- Department of Genomic and Applied Microbiology, Göttingen Genomics Laboratory, Institute of Microbiology and Genetics, Georg-August-University Göttingen, Göttingen, Germany
| | - Anja Poehlein
- Department of Genomic and Applied Microbiology, Göttingen Genomics Laboratory, Institute of Microbiology and Genetics, Georg-August-University Göttingen, Göttingen, Germany
| | - Friederike Herrmann
- Faculty of Life Sciences and Technology, Department of Microbiology, Beuth University of Applied Sciences, Berlin, Germany
| | - Harun Oruc
- Faculty of Life Sciences and Technology, Department of Microbiology, Beuth University of Applied Sciences, Berlin, Germany
| | - Junias Meinhardt
- Faculty of Life Sciences and Technology, Department of Microbiology, Beuth University of Applied Sciences, Berlin, Germany
| | - Olaf Wagner
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Rameez Ahmed
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Sergey Kharin
- Institute of Biomedical Problems (IBMP), Moscow, Russia
| | | | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Rolf Daniel
- Department of Genomic and Applied Microbiology, Göttingen Genomics Laboratory, Institute of Microbiology and Genetics, Georg-August-University Göttingen, Göttingen, Germany
| | - Elisabeth Grohmann
- Faculty of Life Sciences and Technology, Department of Microbiology, Beuth University of Applied Sciences, Berlin, Germany
| |
Collapse
|