1
|
Gonzalez E, Lee MD, Tierney BT, Lipieta N, Flores P, Mishra M, Beckett L, Finkelstein A, Mo A, Walton P, Karouia F, Barker R, Jansen RJ, Green SJ, Weging S, Kelliher J, Singh NK, Bezdan D, Galazska J, Brereton NJB. Spaceflight alters host-gut microbiota interactions. NPJ Biofilms Microbiomes 2024; 10:71. [PMID: 39209868 PMCID: PMC11362537 DOI: 10.1038/s41522-024-00545-1] [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: 02/07/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
The ISS rodent habitat has provided crucial insights into the impact of spaceflight on mammals, inducing symptoms characteristic of liver disease, insulin resistance, osteopenia, and myopathy. Although these physiological responses can involve the microbiome on Earth, host-microbiota interactions during spaceflight are still being elucidated. We explore murine gut microbiota and host gene expression in the colon and liver after 29 and 56 days of spaceflight using multiomics. Metagenomics revealed significant changes in 44 microbiome species, including relative reductions in bile acid and butyrate metabolising bacteria like Extibacter muris and Dysosmobacter welbionis. Functional prediction indicate over-representation of fatty acid and bile acid metabolism, extracellular matrix interactions, and antibiotic resistance genes. Host gene expression described corresponding changes to bile acid and energy metabolism, and immune suppression. These changes imply that interactions at the host-gut microbiome interface contribute to spaceflight pathology and that these interactions might critically influence human health and long-duration spaceflight feasibility.
Collapse
Affiliation(s)
- E Gonzalez
- Microbiome Unit, Canadian Centre for Computational Genomics, Department of Human Genetics, McGill University, Montréal, Canada
- Centre for Microbiome Research, McGill University, Montréal, Canada
| | - M D Lee
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
| | - B T Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - N Lipieta
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - P Flores
- BioServe Space Technologies, University of Colorado Boulder, Boulder, CO, USA
| | - M Mishra
- Grossman School of Medicine, New York University, New York, USA
| | - L Beckett
- University of Nottingham, Nottingham, NG7 2RD, UK
| | - A Finkelstein
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - A Mo
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - P Walton
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - F Karouia
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Centre for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - R Barker
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Yuri GmbH, Wiesentalstr. 40, 88074, Meckenbeuren, Germany
- University of Wisconsin-Madison, Madison, WI, USA
| | - R J Jansen
- Department of Public Health, North Dakota State University, Fargo, ND, USA
- Genomics, Phenomics, and Bioinformatics Program, North Dakota State University, Fargo, ND, USA
| | - S J Green
- Genomics and Microbiome Core Facility, Rush University Medical Centre, 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - S Weging
- Institute of Computer Science, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - J Kelliher
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - N K Singh
- Department of Industrial Relations, Division of Occupational Safety and Health, Oakland, USA
| | - D Bezdan
- University of Wisconsin-Madison, Madison, WI, USA
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- NGS Competence Centre Tübingen (NCCT), University of Tübingen, Tübingen, Germany
| | - J Galazska
- Space Biosciences Research Branch, NASA Ames Research Centre, Moffett Field, CA, USA
| | - N J B Brereton
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
2
|
Xie Y, Liu F. The role of the gut microbiota in tumor, immunity, and immunotherapy. Front Immunol 2024; 15:1410928. [PMID: 38903520 PMCID: PMC11188355 DOI: 10.3389/fimmu.2024.1410928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
In recent years, with the deepening understanding of the gut microbiota, it has been recognized to play a significant role in the development and progression of diseases. Particularly in gastrointestinal tumors, the gut microbiota influences tumor growth by dysbiosis, release of bacterial toxins, and modulation of host signaling pathways and immune status. Immune checkpoint inhibitors (ICIs) have greatly improved cancer treatment efficacy by enhancing immune cell responses. Current clinical and preclinical studies have demonstrated that the gut microbiota and its metabolites can enhance the effectiveness of immunotherapy. Furthermore, certain gut microbiota can serve as biomarkers for predicting immunotherapy responses. Interventions targeting the gut microbiota for the treatment of gastrointestinal diseases, especially colorectal cancer (CRC), include fecal microbiota transplantation, probiotics, prebiotics, engineered bacteria, and dietary interventions. These approaches not only improve the efficacy of ICIs but also hold promise for enhancing immunotherapy outcomes. In this review, we primarily discuss the role of the gut microbiota and its metabolites in tumors, host immunity, and immunotherapy.
Collapse
Affiliation(s)
| | - Fang Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
3
|
Yang Y, Ma Q, Jin S, Huang B, Wang Z, Chen G. Identification of mapk genes, and their expression profiles in response to low salinity stress, in cobia (Rachycentron canadum). Comp Biochem Physiol B Biochem Mol Biol 2024; 271:110950. [PMID: 38307403 DOI: 10.1016/j.cbpb.2024.110950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/04/2024]
Abstract
Mitogen-activated protein kinases (MAPKs) are a class of protein kinases that regulate various physiological processes, and play a crucial role in maintaining the osmotic equilibrium of fish. The objective of this study was to identify and characterize the mapk family genes in cobia (Rachycentron canadum) and examine their expression profiles under different low salinity stress regimes (acute: from 30‰ to 10‰ in 1 h, sub-chronic: from 30‰ to 10‰ over 4 d). A total of 12 cobia mapk genes (Rcmapks) were identified and cloned, including six erk subfamily genes (Rcmapk1/3/4/6/7/15), three jnk subfamily genes (Rcmapk8/9/10) and three p38 mapk subfamily genes (Rcmapk 11/13/14). Domain analysis indicated that the RcMAPKs possessed the typical domains including S_TKc and PKc_like domain. Phylogenetic analysis revealed that the Rcmapks were most closely related to those of the turbot (Scophthalmus maximus). The tissue distribution of mapk genes in adult cobia and the expression patterns of Rcmapks under different low salinity stress regimes were investigated using quantitative real-time PCR (qRT-PCR). The results revealed that Rcmapk3/9/10/11/13/14 exhibited a relatively broad expression distribution across 14 different tissues. For all these genes the highest expression level was in the brain, except for Rcmapk14 (highly expressed in the stomach, gill, and skin). The genes Rcmapk1/6/15 showed significantly higher expression in the testis. Under acute low salinity stress, expression of Rcmapk1/3/6/7/9/11/13/14 was significantly altered in the gill, intestine, and trunk kidney, however, the aforementioned genes exhibited very different expression patterns among the three tissues. In the gill, most of the genes from the erk (Rcmapk3/6/7) and p38 mapk subfamily (Rcmapk11/13/14) were significantly up-regulated at almost all the time points (P < 0.05); Similarly, the expression of Rcmapk3/9/11/13/14 genes were significantly increased in the trunk kidney; while in the intestine, most of the altered genes (Rcmapk6/7/9/11/13/14) were significantly down-regulated at 1 h. Following the sub-chronic low salinity stress, expression of Rcmapk1/3/6/7/9/11/13/14 genes were significantly altered in all three tissues. These findings provide important reference data for elucidating the roles of cobia mapk family genes in response to low salinity stress.
Collapse
Affiliation(s)
- Yunsheng Yang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Qian Ma
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China.
| | - Shulei Jin
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Baosong Huang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhongliang Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Gang Chen
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| |
Collapse
|
4
|
Junaid M, Lu H, Li Y, Liu Y, Din AU, Qi Z, Xiong Y, Yan J. Novel Synergistic Probiotic Intervention: Transcriptomic and Metabolomic Analysis Reveals Ameliorative Effects on Immunity, Gut Barrier, and Metabolism of Mice during Salmonella typhimurium Infection. Genes (Basel) 2024; 15:435. [PMID: 38674370 PMCID: PMC11050207 DOI: 10.3390/genes15040435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Salmonella typhimurium (S. typhimurium), a prevalent cause of foodborne infection, induces significant changes in the host transcriptome and metabolome. The lack of therapeutics with minimal or no side effects prompts the scientific community to explore alternative therapies. This study investigates the therapeutic potential of a probiotic mixture comprising Lactobacillus acidophilus (L. acidophilus 1.3251) and Lactobacillus plantarum (L. plantarum 9513) against S. typhimurium, utilizing transcriptome and metabolomic analyses, a novel approach that has not been previously documented. Twenty-four SPF-BALB/c mice were divided into four groups: control negative group (CNG); positive control group (CPG); probiotic-supplemented non-challenged group (LAPG); and probiotic-supplemented Salmonella-challenged group (LAPST). An RNA-sequencing analysis of small intestinal (ileum) tissue revealed 2907 upregulated and 394 downregulated DEGs in the LAPST vs. CPG group. A functional analysis of DEGs highlighted their significantly altered gene ontology (GO) terms related to metabolism, gut integrity, cellular development, and immunity (p ≤ 0.05). The KEGG analysis showed that differentially expressed genes (DEGs) in the LAPST group were primarily involved in pathways related to gut integrity, immunity, and metabolism, such as MAPK, PI3K-Akt, AMPK, the tryptophan metabolism, the glycine, serine, and threonine metabolism, ECM-receptor interaction, and others. Additionally, the fecal metabolic analysis identified 1215 upregulated and 305 downregulated metabolites in the LAPST vs. CPG group, implying their involvement in KEGG pathways including bile secretion, propanoate metabolism, arginine and proline metabolism, amino acid biosynthesis, and protein digestion and absorption, which are vital for maintaining barrier integrity, immunity, and metabolism. In conclusion, these findings suggest that the administration of a probiotic mixture improves immunity, maintains gut homeostasis and barrier integrity, and enhances metabolism in Salmonella infection.
Collapse
Affiliation(s)
- Muhammad Junaid
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Hongyu Lu
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Yixiang Li
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Yu Liu
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Ahmad Ud Din
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA
| | - Zhongquan Qi
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Yi Xiong
- Guangxi Center for Animals Disease Control and Prevention, Nanning 530004, China
| | - Jianhua Yan
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| |
Collapse
|
5
|
Mortazavi SMJ, Said-Salman I, Mortazavi AR, El Khatib S, Sihver L. How the adaptation of the human microbiome to harsh space environment can determine the chances of success for a space mission to Mars and beyond. Front Microbiol 2024; 14:1237564. [PMID: 38390219 PMCID: PMC10881706 DOI: 10.3389/fmicb.2023.1237564] [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: 08/22/2023] [Accepted: 12/05/2023] [Indexed: 02/24/2024] Open
Abstract
The ability of human cells to adapt to space radiation is essential for the well-being of astronauts during long-distance space expeditions, such as voyages to Mars or other deep space destinations. However, the adaptation of the microbiomes should not be overlooked. Microorganisms inside an astronaut's body, or inside the space station or other spacecraft, will also be exposed to radiation, which may induce resistance to antibiotics, UV, heat, desiccation, and other life-threatening factors. Therefore, it is essential to consider the potential effects of radiation not only on humans but also on their microbiomes to develop effective risk reduction strategies for space missions. Studying the human microbiome in space missions can have several potential benefits, including but not limited to a better understanding of the major effects space travel has on human health, developing new technologies for monitoring health and developing new radiation therapies and treatments. While radioadaptive response in astronauts' cells can lead to resistance against high levels of space radiation, radioadaptive response in their microbiome can lead to resistance against UV, heat, desiccation, antibiotics, and radiation. As astronauts and their microbiomes compete to adapt to the space environment. The microorganisms may emerge as the winners, leading to life-threatening situations due to lethal infections. Therefore, understanding the magnitude of the adaptation of microorganisms before launching a space mission is crucial to be able to develop effective strategies to mitigate the risks associated with radiation exposure. Ensuring the safety and well-being of astronauts during long-duration space missions and minimizing the risks linked with radiation exposure can be achieved by adopting this approach.
Collapse
Affiliation(s)
- Seyed Mohammad Javad Mortazavi
- Ionizing and non-ionizing radiation protection research center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ilham Said-Salman
- Department of Biological and Chemical Sciences, School of Arts & Sciences, Lebanese International University, Saida, Lebanon
- Department of Biological and Chemical Sciences, International University of Beirut, Beirut, Lebanon
| | | | - Sami El Khatib
- Department of Biomedical Sciences, School of Arts and Sciences, Lebanese International University, Beirut, Lebanon
- Center for Applied Mathematics and Bioinformatics (CAMB) at Gulf University for Science and Technology, Kuwait City, Kuwait
| | - Lembit Sihver
- Department of Radiation Dosimetry, Nuclear Physics Institute (NPI) of the Czech Academy of Sciences (CAS), Prague, Czechia
- Department of Radiation Physics, Technische Universität Wien Atominstitut, Vienna, Austria
| |
Collapse
|
6
|
Majumder N, Ghosh S. 3D biofabrication and space: A 'far-fetched dream' or a 'forthcoming reality'? Biotechnol Adv 2023; 69:108273. [PMID: 37863444 DOI: 10.1016/j.biotechadv.2023.108273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/10/2023] [Accepted: 10/15/2023] [Indexed: 10/22/2023]
Abstract
The long duration space missions across the Low Earth Orbit (LEO) often expose the voyagers to an abrupt zero gravity influence. The severe extraterrestrial cosmic radiation directly causes a plethora of moderate to chronic healthcare crises. The only feasible solution to manage critical injuries on board is surgical interventions or immediate return to Earth. This led the group of space medicine practitioners to adopt principles from tissue engineering and develop human tissue equivalents as an immediate regenerative therapy on board. The current review explicitly demonstrates the constructive application of different tissue-engineered equivalents matured under the available ground-based microgravity simulation facilities. Further, it elucidates how augmenting the superiority of biomaterial-based 3D bioprinting technology can enhance their clinical applicability. Additionally, the regulatory role of weightlessness condition on the underlying cellular signaling pathways governing tissue morphogenesis has been critically discussed. This information will provide future directions on how 3D biofabrication can be used as a plausible tool for healing on-flight chronic health emergencies. Thus, in our review, we aimed to precisely debate whether 3D biofabrication is deployed to cater to on-flight healthcare anomalies or space-like conditions are being utilized for generating 3D bioprinted human tissue constructs for efficient drug screening and regenerative therapy.
Collapse
Affiliation(s)
- Nilotpal Majumder
- Regenerative Engineering Laboratory, Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Sourabh Ghosh
- Regenerative Engineering Laboratory, Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India.
| |
Collapse
|
7
|
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
|
8
|
Corydon TJ, Schulz H, Richter P, Strauch SM, Böhmer M, Ricciardi DA, Wehland M, Krüger M, Erzinger GS, Lebert M, Infanger M, Wise PM, Grimm D. Current Knowledge about the Impact of Microgravity on Gene Regulation. Cells 2023; 12:cells12071043. [PMID: 37048115 PMCID: PMC10093652 DOI: 10.3390/cells12071043] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/24/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
Microgravity (µg) has a massive impact on the health of space explorers. Microgravity changes the proliferation, differentiation, and growth of cells. As crewed spaceflights into deep space are being planned along with the commercialization of space travelling, researchers have focused on gene regulation in cells and organisms exposed to real (r-) and simulated (s-) µg. In particular, cancer and metastasis research benefits from the findings obtained under µg conditions. Gene regulation is a key factor in a cell or an organism’s ability to sustain life and respond to environmental changes. It is a universal process to control the amount, location, and timing in which genes are expressed. In this review, we provide an overview of µg-induced changes in the numerous mechanisms involved in gene regulation, including regulatory proteins, microRNAs, and the chemical modification of DNA. In particular, we discuss the current knowledge about the impact of microgravity on gene regulation in different types of bacteria, protists, fungi, animals, humans, and cells with a focus on the brain, eye, endothelium, immune system, cartilage, muscle, bone, and various cancers as well as recent findings in plants. Importantly, the obtained data clearly imply that µg experiments can support translational medicine on Earth.
Collapse
Affiliation(s)
- Thomas J. Corydon
- Department of Biomedicine, Aarhus University, Hoegh Guldbergs Gade 10, 8000 Aarhus, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Blvd. 99, 8200 Aarhus, Denmark
- Correspondence: ; Tel.: +45-28-992-179
| | - Herbert Schulz
- Department of Microgravity and Translational Regenerative Medicine, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- Clinic for Plastic, Aesthetic and Hand Surgery, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Peter Richter
- Gravitational Biology Group, Department of Biology, Friedrich-Alexander University, 91058 Erlangen, Germany
| | - Sebastian M. Strauch
- Postgraduate Program in Health and Environment, University of Joinville Region, Joinville 89219-710, SC, Brazil
| | - Maik Böhmer
- Institute for Molecular Biosciences, Johann Wolfgang Goethe Universität, 60438 Frankfurt am Main, Germany
| | - Dario A. Ricciardi
- Institute for Molecular Biosciences, Johann Wolfgang Goethe Universität, 60438 Frankfurt am Main, Germany
| | - Markus Wehland
- Department of Microgravity and Translational Regenerative Medicine, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- Clinic for Plastic, Aesthetic and Hand Surgery, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Gilmar S. Erzinger
- Postgraduate Program in Health and Environment, University of Joinville Region, Joinville 89219-710, SC, Brazil
| | - Michael Lebert
- Gravitational Biology Group, Department of Biology, Friedrich-Alexander University, 91058 Erlangen, Germany
| | - Manfred Infanger
- Department of Microgravity and Translational Regenerative Medicine, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- Clinic for Plastic, Aesthetic and Hand Surgery, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Petra M. Wise
- Department of Microgravity and Translational Regenerative Medicine, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- The Saban Research Institute, Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Blvd, Los Angeles, CA 90027, USA
| | - Daniela Grimm
- Department of Biomedicine, Aarhus University, Hoegh Guldbergs Gade 10, 8000 Aarhus, Denmark
- Department of Microgravity and Translational Regenerative Medicine, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- Clinic for Plastic, Aesthetic and Hand Surgery, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| |
Collapse
|
9
|
Niu YB, Zhang YH, Sun Y, Song XZ, Li ZH, Xie M, Mei QB, Li YH, Chen Q. Asperosaponin VI Protects Against Bone Loss Due to Hindlimb Unloading in Skeletally Growing Mice Through Regulating Microbial Dysbiosis Altering the 5-HT Pathway. Calcif Tissue Int 2023; 112:389-402. [PMID: 36595050 DOI: 10.1007/s00223-022-01057-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/21/2022] [Indexed: 01/04/2023]
Abstract
Osteoporosis is a complex multifactorial disease that can lead to an increased risk of fracture. However, selective and effective osteoporosis drugs are still lacking. We showed that Asperosaponin VI (AVI) has the implications to be further developed as an alternative supplement for the prevention and treatment of bone loss. AVI has been found to have beneficial effects on metabolic diseases such as bone loss, obesity, and atherosclerosis. Our study was designed to determine the effect and mechanism of action of AVI against bone loss through regulating microbial dysbiosis. A hindlimb unloading mouse model was established to determine the effect of AVI on bone microarchitecture, gut microbiota, and serum metabolites. Eighteen female C57BL/6 J mice were divided into three groups: control, hindlimb unloading with vehicle (HLU), and hindlimb unloading treated with AVI (HLU-AVI, 200 mg/kg/day). AVI was administrated orally for 4 weeks. The results demonstrated that AVI improved the bone microstructure by reversing the decrease in bone volume fraction and trabecular number, and the increase in trabecular separation and structure model index of cancellous bone in hindlimb suspension mice. The results of 16sRNA gene sequencing suggested that the therapeutic effect of AVI on bone loss may be achieved through it regulating the gut microbiota, especially certain specific microorganisms. Combined with the analysis of ELISA, immunohistochemistry, and serum metabolome results, it could be speculated that AVI played an important role in adjusting the balance of bone metabolism by influencing specific flora such as Clostridium and its metabolites to regulate the 5-hydroxytryptophan pathway. The study explored the novel mechanism of AVI against osteoporosis, and has implications for the further development of AVI as an alternative supplement for the prevention and treatment of bone loss.
Collapse
Affiliation(s)
- Y-B Niu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Y-H Zhang
- State Key Laboratory of Solidification Processing, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Y Sun
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, People's Republic of China
| | - X-Z Song
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Z-H Li
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - M Xie
- Department of Pharmacy, The First Naval Force Hospital of Southern Theatre Command, Zhanjiang, 524005, Guangdong, People's Republic of China
| | - Q-B Mei
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Y-H Li
- Department of Pathology, The First Naval Force Hospital of Southern Theatre Command, Zhanjiang, 524005, Guangdong, People's Republic of China.
| | - Q Chen
- State Key Laboratory of Solidification Processing, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China.
| |
Collapse
|
10
|
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: 7.0] [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
|
11
|
Bao X, Wang W, Chen X, Feng Y, Xu X, Sun G, Li B, Liu X, Li Z, Yang J. Exploration of immune response mechanisms in cadmium and copper co-exposed juvenile golden cuttlefish ( Sepia esculenta) based on transcriptome profiling. Front Immunol 2022; 13:963931. [PMID: 36211441 PMCID: PMC9538352 DOI: 10.3389/fimmu.2022.963931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/01/2022] [Indexed: 11/23/2022] Open
Abstract
Sepia esculenta is a popular economic cephalopod with high yield, delicious meat, and rich nutrition. With the rapid development of heavy industry and medical industry, a large amount of waste has been released into the ocean recklessly in recent years, inducing a significant increase in the content of heavy metals, especially cadmium (Cd) and copper (Cu), in the ocean. This phenomenon significantly affects the growth and development of S. esculenta, causing a serious blow to its artificial breeding. In this study, transcriptome analysis is used to initially explore immune response mechanisms of Cd and Cu co-exposed juvenile S. esculenta. The results show that 1,088 differentially expressed genes (DEGs) are identified. And DEGs functional enrichment analysis results suggests that co-exposure may promote inflammatory and innate immune responses in juvenile S. esculenta. Fifteen key genes that might regulate the immunity of S. esculenta are identified using protein-protein interaction (PPI) network and KEGG enrichment analyses, of which the three genes with the highest number of interactions or involve in more KEGG pathways are identified as hub genes that might significantly affect the immune response processes. Comprehensive analysis of PPI network and KEGG signaling pathway is used for the first time to explore co-exposed S. esculenta juvenile immune response processes. Our results preliminarily reveal immune response mechanisms of cephalopods exposed to heavy metals and provide a valuable resource for further understanding of mollusk immunity.
Collapse
Affiliation(s)
- Xiaokai Bao
- School of Agriculture, Ludong University, Yantai, China
| | - Weijun Wang
- School of Agriculture, Ludong University, Yantai, China
| | - Xipan Chen
- School of Agriculture, Ludong University, Yantai, China
| | - Yanwei Feng
- School of Agriculture, Ludong University, Yantai, China
| | - Xiaohui Xu
- School of Agriculture, Ludong University, Yantai, China
| | - Guohua Sun
- School of Agriculture, Ludong University, Yantai, China
| | - Bin Li
- School of Agriculture, Ludong University, Yantai, China
| | - Xiumei Liu
- College of Life Sciences, Yantai University, Yantai, China
| | - Zan Li
- School of Agriculture, Ludong University, Yantai, China
| | - Jianmin Yang
- School of Agriculture, Ludong University, Yantai, China
| |
Collapse
|
12
|
An R. MRTF may be the missing link in a multiscale mechanobiology approach toward macrophage dysfunction in space. Front Cell Dev Biol 2022; 10:997365. [PMID: 36172272 PMCID: PMC9510870 DOI: 10.3389/fcell.2022.997365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/10/2022] [Indexed: 11/23/2022] Open
Abstract
Macrophages exhibit impaired phagocytosis, adhesion, migration, and cytokine production in space, hindering their ability to elicit immune responses. Considering that the combined effect of spaceflight microgravity and radiation is multiscale and multifactorial in nature, it is expected that contradictory findings are common in the field. This theory paper reanalyzes research on the macrophage spaceflight response across multiple timescales from seconds to weeks, and spatial scales from the molecular, intracellular, extracellular, to the physiological. Key findings include time-dependence of both pro-inflammatory activation and integrin expression. Here, we introduce the time-dependent, intracellular localization of MRTF-A as a hypothetical confounder of macrophage activation. We discuss the mechanosensitive MRTF-A/SRF pathway dependence on the actin cytoskeleton/nucleoskeleton, microtubules, membrane mechanoreceptors, hypoxia, oxidative stress, and intracellular/extracellular crosstalk. By adopting a multiscale perspective, this paper provides the first mechanistic answer for a three-decade-old question regarding impaired cytokine secretion in microgravity—and strengthens the connection between the recent advances in mechanobiology, microgravity, and the spaceflight immune response. Finally, we hypothesize MRTF involvement and complications in treating spaceflight-induced cardiovascular, skeletal, and immune disease.
Collapse
Affiliation(s)
- Rocky An
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, United States
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
- *Correspondence: Rocky An,
| |
Collapse
|
13
|
Al KF, Chmiel JA, Stuivenberg GA, Reid G, Burton JP. Long-Duration Space Travel Support Must Consider Wider Influences to Conserve Microbiota Composition and Function. Life (Basel) 2022; 12:1163. [PMID: 36013342 PMCID: PMC9409767 DOI: 10.3390/life12081163] [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: 06/24/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 12/03/2022] Open
Abstract
The microbiota is important for immune modulation, nutrient acquisition, vitamin production, and other aspects for long-term human health. Isolated model organisms can lose microbial diversity over time and humans are likely the same. Decreasing microbial diversity and the subsequent loss of function may accelerate disease progression on Earth, and to an even greater degree in space. For this reason, maintaining a healthy microbiome during spaceflight has recently garnered consideration. Diet, lifestyle, and consumption of beneficial microbes can shape the microbiota, but the replenishment we attain from environmental exposure to microbes is important too. Probiotics, prebiotics, fermented foods, fecal microbiota transplantation (FMT), and other methods of microbiota modulation currently available may be of benefit for shorter trips, but may not be viable options to overcome the unique challenges faced in long-term space travel. Novel fermented food products with particular impact on gut health, immune modulation, and other space-targeted health outcomes are worthy of exploration. Further consideration of potential microbial replenishment to humans, including from environmental sources to maintain a healthy microbiome, may also be required.
Collapse
Affiliation(s)
- Kait F. Al
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 3K7, Canada; (K.F.A.); (J.A.C.); (G.A.S.); (G.R.)
| | - John A. Chmiel
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 3K7, Canada; (K.F.A.); (J.A.C.); (G.A.S.); (G.R.)
| | - Gerrit A. Stuivenberg
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 3K7, Canada; (K.F.A.); (J.A.C.); (G.A.S.); (G.R.)
| | - Gregor Reid
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 3K7, Canada; (K.F.A.); (J.A.C.); (G.A.S.); (G.R.)
- Department of Surgery, University of Western Ontario, London, ON N6A 4V2, Canada
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
| | - Jeremy P. Burton
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 3K7, Canada; (K.F.A.); (J.A.C.); (G.A.S.); (G.R.)
- Department of Surgery, University of Western Ontario, London, ON N6A 4V2, Canada
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
| |
Collapse
|
14
|
Exposure to Random Positioning Machine Alters the Mineralization Process and PTX3 Expression in the SAOS-2 Cell Line. Life (Basel) 2022; 12:life12050610. [PMID: 35629278 PMCID: PMC9143356 DOI: 10.3390/life12050610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/11/2022] [Accepted: 04/17/2022] [Indexed: 01/02/2023] Open
Abstract
Bone loss is among the most frequent changes seen in astronauts during space missions. Although weightlessness is known to cause high bone resorption and a rapid decrease in bone minerals and calcium, the underlying mechanisms are not yet fully understood. In our work, we investigated the influence of random positioning machine (RPM) exposure on the mineralization process in the SAOS-2 cell line, in osteogenic and non-osteogenic conditions, by examining changes in their mineralizing capacity and in the expression of PTX3, a positive regulator of bone mineralization. We analyzed cell viability by MTS assay and the mineralization process after staining with Toluidine Blue and Alizarin Red, while PTX3 expression was investigated by immunocytochemistry and western blotting analysis. Our results showed that RPM exposure increased cells’ viability and improved their mineralizing competence when not treated with osteogenic cocktail. In contrast, in osteogenic conditions, cells exposed to RPM showed a reduction in the presence of calcification-like structures, mineral deposits and PTX3 expression, suggesting that the effects of RPM exposure on mineralizing matrix deposition depend on the presence of osteogenic factors in the culture medium. Further studies will be needed to clarify the role of potential mineralization markers in the cellular response to the simulated biological effects of microgravity, paving the way for a new approach to treating osteoporosis in astronauts exposed to spaceflight.
Collapse
|
15
|
Lu SY, Guo S, Chai SB, Yang JQ, Yue Y, Li H, Yan HF, Zhang T, Sun PM, Sun HW, Zhou JL, Yang JW, Li ZP, Cui Y. Proteomic analysis of the effects of simulated microgravity in human gastric mucosal cells. LIFE SCIENCES IN SPACE RESEARCH 2022; 32:26-37. [PMID: 35065758 DOI: 10.1016/j.lssr.2021.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 06/14/2023]
Abstract
Microgravity is an ecological factor that affects the environment of the body. In this study, quantitative isobaric labeling (tandem mass tag) method was used to study the changes in human gastric mucosal cells under simulated microgravity for the first time. Comparative proteomic analysis identified 394 (202 upregulated and 192 downregulated) and 542 (286 upregulated and 256 downregulated) proteins differentially regulated by simulated microgravity after 3 and 7 days, respectively. Then the identified proteins were subjected to Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses for further exploration. The results of the analysis showed that the ribosomes of gastric mucosal cells were significantly impacted after exposure to simulated microgravity for 3 days, and the cells appeared to be in a state of stress and inflammation. Exposure to simulated microgravity for 7 days significantly affected the mitochondria of the cells, oxidative stress became more evident, while inflammation and weakened connections were observed in the cells. The results of this study highlighted the temporal response trend of gastric mucosal cells to the stressor of microgravity at the two time points of 3 and 7 days. These findings will provide insights into the development of methods to protect the gastric mucosa during space flight.
Collapse
Affiliation(s)
- Sheng-Yu Lu
- Department of General Surgery, The 306th Hospital of PLA-Peking University Teaching Hospital, Beijing 100101, China; Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Song Guo
- Department of General Surgery, The 306th Hospital of PLA-Peking University Teaching Hospital, Beijing 100101, China; Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Shao-Bin Chai
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Jia-Qi Yang
- Department of General Surgery, The 306th Hospital of PLA-Peking University Teaching Hospital, Beijing 100101, China; Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Yuan Yue
- Department of General Surgery, The 306th Hospital of PLA-Peking University Teaching Hospital, Beijing 100101, China; Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Hao Li
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Hong-Feng Yan
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Tao Zhang
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Pei-Ming Sun
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Hong-Wei Sun
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Jin-Lian Zhou
- Department of Pathology, Strategic Support Force Medical Center, Beijing 100101, China
| | - Jian-Wu Yang
- Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China
| | - Zheng-Peng Li
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yan Cui
- Department of General Surgery, The 306th Hospital of PLA-Peking University Teaching Hospital, Beijing 100101, China; Department of General Surgery, Strategic Support Force Medical Center, Beijing 100101, China.
| |
Collapse
|
16
|
Yuan J, Li X, Fang N, Li P, Zhang Z, Lin M, Hou Q. Perilla Leaf Extract (PLE) Attenuates COPD Airway Inflammation via the TLR4/Syk/PKC/NF-κB Pathway In Vivo and In Vitro. Front Pharmacol 2022; 12:763624. [PMID: 35058774 PMCID: PMC8764369 DOI: 10.3389/fphar.2021.763624] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/10/2021] [Indexed: 11/20/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex and heterogeneous disease characterized by persistent airflow limitation but still lacking effective treatments. Perilla frutescens (L.) Britt., an important traditional medicinal plant with excellent antioxidant and anti-inflammatory properties, is widely used for the treatment of respiratory disease in China. However, its protective activity and mechanism against COPD airway inflammation have not been fully studied. Here, the anti-inflammatory effects of the PLE were investigated, and its underlying mechanisms were then elucidated. The presented results suggested a notable effect of the PLE on airway inflammation of COPD, by significantly ameliorating inflammatory cell infiltration in lung tissue, lessening leukocytes (lymphocytes, neutrophils, and macrophages) and inflammatory mediators (interleukin 4 (IL-4), IL-6, IL-17A, interferon γ (IFN-γ), and tumor necrosis factor α (TNF-α)) in the bronchoalveolar lavage fluid (BALF) of cigarette smoke (CS)/lipopolysaccharide (LPS)-induced COPD mice in vivo and inhibiting the production of inflammatory factors (nitric oxide (NO), IL-6, and TNF-α) and intracellular reactive oxygen species (ROS) in LPS-stimulated RAW264.7 cells in vitro. For further extent, PLE treatment significantly suppressed the expression and phosphorylation of TLR4, Syk, PKC, and NF-κB p65 in vivo and their mRNA in vitro. Subsequently, by co-treating with their inhibitors in vitro, its potential mechanism via TLR4/Syk/PKC/NF-κB p65 signals was disclosed. In summary, the obtained results indicated a noteworthy effective activity of the PLE on COPD inflammation, and partly, the TLR4/Syk/PKC/NF-κB p65 axis might be the potential mechanism.
Collapse
Affiliation(s)
- Jiqiao Yuan
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuyu Li
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Fang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ping Li
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziqian Zhang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingbao Lin
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Hou
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
17
|
Effect of Microgravity Environment on Gut Microbiome and Angiogenesis. Life (Basel) 2021; 11:life11101008. [PMID: 34685381 PMCID: PMC8541308 DOI: 10.3390/life11101008] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/24/2022] Open
Abstract
Microgravity environments are known to cause a plethora of stressors to astronauts. Recently, it has become apparent that gut microbiome composition of astronauts is altered following space travel, and this is of significance given the important role of the gut microbiome in human health. Other changes observed in astronauts comprise reduced muscle strength and bone fragility, visual impairment, endothelial dysfunction, metabolic changes, behavior changes due to fatigue or stress and effects on mental well-being. However, the effects of microgravity on angiogenesis, as well as the connection with the gut microbiome are incompletely understood. Here, the potential association of angiogenesis with visual impairment, skeletal muscle and gut microbiome is proposed and explored. Furthermore, metabolites that are effectors of angiogenesis are deliberated upon along with their connection with gut bacterial metabolites. Targeting and modulating the gut microbiome may potentially have a profound influence on astronaut health, given its impact on overall human health, which is thus warranted given the likelihood of increased human activity in the solar system, and the determination to travel to Mars in future missions.
Collapse
|
18
|
ElGindi M, Sapudom J, Ibrahim IH, Al-Sayegh M, Chen W, Garcia-Sabaté A, Teo JCM. May the Force Be with You (Or Not): The Immune System under Microgravity. Cells 2021; 10:1941. [PMID: 34440709 PMCID: PMC8391211 DOI: 10.3390/cells10081941] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
All terrestrial organisms have evolved and adapted to thrive under Earth's gravitational force. Due to the increase of crewed space flights in recent years, it is vital to understand how the lack of gravitational forces affects organisms. It is known that astronauts who have been exposed to microgravity suffer from an array of pathological conditions including an impaired immune system, which is one of the most negatively affected by microgravity. However, at the cellular level a gap in knowledge exists, limiting our ability to understand immune impairment in space. This review highlights the most significant work done over the past 10 years detailing the effects of microgravity on cellular aspects of the immune system.
Collapse
Affiliation(s)
- Mei ElGindi
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates; (M.E.); (J.S.); (I.H.I.)
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates; (M.E.); (J.S.); (I.H.I.)
| | - Ibrahim Hamed Ibrahim
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates; (M.E.); (J.S.); (I.H.I.)
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates;
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA;
- Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA
| | - Anna Garcia-Sabaté
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates; (M.E.); (J.S.); (I.H.I.)
| | - Jeremy C. M. Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates; (M.E.); (J.S.); (I.H.I.)
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA;
- Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA
| |
Collapse
|
19
|
Simões MF, Antunes A. Microbial Pathogenicity in Space. Pathogens 2021; 10:450. [PMID: 33918768 PMCID: PMC8069885 DOI: 10.3390/pathogens10040450] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/04/2021] [Accepted: 04/08/2021] [Indexed: 12/25/2022] Open
Abstract
After a less dynamic period, space exploration is now booming. There has been a sharp increase in the number of current missions and also of those being planned for the near future. Microorganisms will be an inevitable component of these missions, mostly because they hitchhike, either attached to space technology, like spaceships or spacesuits, to organic matter and even to us (human microbiome), or to other life forms we carry on our missions. Basically, we never travel alone. Therefore, we need to have a clear understanding of how dangerous our "travel buddies" can be; given that, during space missions, our access to medical assistance and medical drugs will be very limited. Do we explore space together with pathogenic microorganisms? Do our hitchhikers adapt to the space conditions, as well as we do? Do they become pathogenic during that adaptation process? The current review intends to better clarify these questions in order to facilitate future activities in space. More technological advances are needed to guarantee the success of all missions and assure the reduction of any possible health and environmental risks for the astronauts and for the locations being explored.
Collapse
Affiliation(s)
- Marta Filipa Simões
- State Key Laboratory of Lunar and Planetary Sciences (SKLPlanets), Macau University of Science and Technology (MUST), Avenida Wai Long, Taipa, Macau, China;
- China National Space Administration (CNSA), Macau Center for Space Exploration and Science, Macau, China
| | - André Antunes
- State Key Laboratory of Lunar and Planetary Sciences (SKLPlanets), Macau University of Science and Technology (MUST), Avenida Wai Long, Taipa, Macau, China;
- China National Space Administration (CNSA), Macau Center for Space Exploration and Science, Macau, China
| |
Collapse
|
20
|
Gizard F, Fernandez A, De Vadder F. Interactions between gut microbiota and skeletal muscle. Nutr Metab Insights 2021; 13:1178638820980490. [PMID: 33402830 PMCID: PMC7745561 DOI: 10.1177/1178638820980490] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
The gut microbiota is now recognized as a major contributor to the host’s nutrition, metabolism, immunity, and neurological functions. Imbalanced microbiota (ie, dysbiosis) is linked to undernutrition-induced stunting, inflammatory and metabolic diseases, and cancers. Skeletal muscle also takes part in the interorgan crosstalk regulating substrate metabolism, immunity, and health. Here, we review the reciprocal influence of gut microbiota and skeletal muscle in relation to juvenile growth, performance, aging, and chronic diseases. Several routes involving the vascular system and organs such as the liver and adipose tissue connect the gut microbiota and skeletal muscle, with effects on fitness and health. Therapeutic perspectives arise from the health benefits observed with changes in gut microbiota and muscle activity, further encouraging multimodal therapeutic strategies.
Collapse
Affiliation(s)
- Florence Gizard
- Mammalian Cell Biology Group, Institute of Human Genetics UMR9002, CNRS-University of Montpellier, Montpellier, France
| | - Anne Fernandez
- Mammalian Cell Biology Group, Institute of Human Genetics UMR9002, CNRS-University of Montpellier, Montpellier, France
| | - Filipe De Vadder
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, École Normale Supérieure de Lyon, Centre National de la Recherche Scientifique, Université Claude Bernard Lyon 1, UMR5242, Lyon, France
| |
Collapse
|