1
|
Derobertmasure A, Toh LS, Wotring VE, Williams PM, Morbidelli L, Stingl JC, Vinken M, Ramadan R, Chhun S, Boutouyrie P. Pharmacological countermeasures for long-duration space missions: addressing cardiovascular challenges and advancing space-adapted healthcare. Eur J Pharm Sci 2025; 209:107063. [PMID: 40064402 DOI: 10.1016/j.ejps.2025.107063] [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: 11/29/2024] [Revised: 02/10/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Future long-duration crewed space missions beyond Low Earth Orbit (LEO) will bring new healthcare challenges for astronauts for which pharmacological countermeasures (pharmacological countermeasures) are crucial. This paper highlights current pharmacological countermeasures challenges described in the ESA SciSpacE Roadmap, with a focus on the cardiovascular system as a model to demonstrate the potential implication of the challenges and recommendations. New pharmacological approaches and procedures need to be adapted to spaceflight (spaceflight) conditions, including ethical and reglementary considerations. Potential strategies include combining pharmacological biomarkers such as pharmacogenomics with therapeutic drug monitoring, advancing microsampling techniques, and implementing a pharmacovigilance system to gain deep insights into pharmacokinetics/pharmacodynamics (PK/PD) spaceflight alteration on drug exposure. Emerging therapeutic approaches (such as long-term regimens) or manufacturing drugs in the space environment, can address specific issues related to drug storage and stability. The integration of biobanks and innovative technologies like organoids and organ-on-a-chip, artificial intelligence (AI), including machine learning will further enhance PK modelling leading to personalized treatments. These innovative pharmaceutical tools will also enable reciprocal game-changing healthcare developments to be made on Earth as well as in space and are essential to ensure space explorers receive safe effective pharmaceutical care.
Collapse
Affiliation(s)
- Audrey Derobertmasure
- Faculty of Medicine, Paris Cité University, INSERM PARCC, Service de Pharmacologie Clinique, Hôpital Européen Georges Pompidou Hospital (AP-HP), Paris, France
| | - Li Shean Toh
- School of Pharmacy, Faculty of Science, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Virginia E Wotring
- International Space University, 1 rue Jean-Dominique Cassini, Parc d'Innovation, 6700 Illkirch-Graffenstaden, France
| | - Philip M Williams
- School of Pharmacy, Faculty of Science, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Julia C Stingl
- Institute of Clinical Pharmacology, University Hospital of RWTH Aachen, Wendlingweg 2, 52064, Aachen, Germany
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Raghda Ramadan
- Interdisciplinary Biosciences Group, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Stephanie Chhun
- Faculty of Medicine, Paris Cité University, Paris, France; Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253; AP-HP, Laboratory of Immunology, Necker-Enfants Malades Hospital, Paris, France
| | - Pierre Boutouyrie
- Faculty of Medicine, Paris Cité University, INSERM PARCC, Service de Pharmacologie Clinique, Hôpital Européen Georges Pompidou Hospital (AP-HP), Paris, France.
| |
Collapse
|
2
|
Xiong Y, Guo J, Yu W, Zeng D, Song C, Zhou L, Anatolyevna NL, Baranenko D, Xiao D, Zhou Y, Lu W. Molecular Mechanism of Microgravity-Induced Intestinal Flora Dysbiosis on the Abnormalities of Liver and Brain Metabolism. Int J Mol Sci 2025; 26:3094. [PMID: 40243802 PMCID: PMC11988970 DOI: 10.3390/ijms26073094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/06/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Space flight has many adverse effects on the physiological functions of astronauts. Certain similarities have been observed in some physiological processes of rodents and astronauts in space, although there are also differences. These similarities make rodents helpful models for initial investigations into space-induced physiological changes. This study uses a 3D-Clinostat to simulate microgravity and explores the role of microgravity in space flight-induced liver and brain abnormalities by comparing changes in the gut microbiota, serum metabolites, and the function and physiological biochemistry of liver and brain tissues between the simulated microgravity (SMG) group mice and the wild type (WT) group mice. The study, based on hematoxylin-eosin (HE) staining, 16S sequencing technology, and non-targeted metabolomics analysis, shows that the gut tissue morphology of the SMG group mice is abnormal, and the structure of the gut microbiota and the serum metabolite profile are imbalanced. Furthermore, using PICRUST 2 technology, we have predicted the functions of the gut microbiota and serum metabolites, and the results indicate that the liver metabolism and functions (including lipid metabolism, amino acid metabolism, and sugar metabolism, etc.) of the SMG group mice are disrupted, and the brain tissue metabolism and functions (including neurotransmitters and hormone secretion, etc.) are abnormal, suggesting a close relationship between microgravity and liver metabolic dysfunction and brain dysfunction. Additionally, the high similarity in the structure of the gut microbiota and serum metabolite profile between the fecal microbiota transplant (FMT) group mice and the SMG group mice, and the physiological and biochemical differences in liver and brain tissues compared to the WT group mice, suggest that microgravity induces imbalances in the gut microbiota, which in turn triggers abnormalities in liver and brain metabolism and function. Finally, through MetaMapp analysis and Pearson correlation analysis, we found that valeric acid, a metabolite of gut microbiota, is more likely to be the key metabolite that relates to microgravity-induced gut microbiota abnormalities, disorders of amino acid and lipid metabolism, and further induced metabolic or functional disorders in the liver and brain. This study has significant practical application value for deepening the understanding of the adaptability of living organisms in the space environment.
Collapse
Affiliation(s)
- Yi Xiong
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; (Y.X.)
- National and Local Joint Engineering Laboratory for Synthesis, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Jianguo Guo
- National Human Diseases Animal Model Resource Center, National Center of Technology Innovation for Animal Model, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing 100021, China
| | - Wenchen Yu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; (Y.X.)
- National and Local Joint Engineering Laboratory for Synthesis, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Deyong Zeng
- National and Local Joint Engineering Laboratory for Synthesis, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Chenchen Song
- National Human Diseases Animal Model Resource Center, National Center of Technology Innovation for Animal Model, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing 100021, China
| | - Li Zhou
- National Human Diseases Animal Model Resource Center, National Center of Technology Innovation for Animal Model, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing 100021, China
| | - Nadtochii Liudmila Anatolyevna
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
- School of Life Sciences, International Research Centre Biotechnologies of the Third Millennium, ITMO University, St. Petersburg 197101, Russia
| | - Denis Baranenko
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
- School of Life Sciences, International Research Centre Biotechnologies of the Third Millennium, ITMO University, St. Petersburg 197101, Russia
| | - Dan Xiao
- National and Local Joint Engineering Laboratory for Synthesis, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Yingyu Zhou
- National and Local Joint Engineering Laboratory for Synthesis, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Weihong Lu
- National and Local Joint Engineering Laboratory for Synthesis, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| |
Collapse
|
3
|
Finch RH, Vitry G, Siew K, Walsh SB, Beheshti A, Hardiman G, da Silveira WA. Spaceflight causes strain-dependent gene expression changes in the kidneys of mice. NPJ Microgravity 2025; 11:11. [PMID: 40133368 PMCID: PMC11937539 DOI: 10.1038/s41526-025-00465-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Incidence of kidney stones in astronauts is a major risk factor associated with long-term missions, caused by increased blood calcium levels due to bone demineralisation triggered by microgravity and space radiation. Transcriptomic changes have been observed in tissues during spaceflight, including the kidney. We analysed kidney transcriptome patterns in two different strains of mice flown on the International Space Station, C57BL/6J and BALB/c. Here we show a link between spaceflight and transcriptome patterns associated with dysregulation of lipid and extracellular matrix metabolism and altered transforming growth factor-beta signalling. A stronger response was seen in C57BL/6J mice than BALB/c. Genetic differences in hyaluronan metabolism between strains may confer protection against extracellular matrix remodelling through the downregulation of epithelial-mesenchymal transition. We intend for our findings to contribute to the development of new countermeasures against kidney disease in astronauts and people here on Earth.
Collapse
Affiliation(s)
- Rebecca H Finch
- University of Staffordshire, Department of Sports and Science, School of Health, Education, Policing and Sciences, Science Centre, Leek Road, Stoke-on-Trent, ST4 2DF, UK
| | - Geraldine Vitry
- University of Staffordshire, Department of Sports and Science, School of Health, Education, Policing and Sciences, Science Centre, Leek Road, Stoke-on-Trent, ST4 2DF, UK
- International Space University, 1 Rue Jean-Dominique Cassini, 67400, Illkirch-Graffenstaden, France
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Department of Oncology, 3970 Reservoir Rd, NW, New Research Building EP11, Washington, DC, 20057, USA
| | - Keith Siew
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
| | - Stephen B Walsh
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
| | - Afshin Beheshti
- Center for Space Biomedicine, McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Gary Hardiman
- Faculty of Medicine, Health and Life Sciences, Institute for Global Food Security (IGFS), School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, BT9 5DL, UK
- Department of Medicine, Medical University of South Carolina, MSC 403, 171 Ashley Ave Suite 419, Charleston, SC, 29425, USA
| | - Willian A da Silveira
- University of Staffordshire, Department of Sports and Science, School of Health, Education, Policing and Sciences, Science Centre, Leek Road, Stoke-on-Trent, ST4 2DF, UK.
- International Space University, 1 Rue Jean-Dominique Cassini, 67400, Illkirch-Graffenstaden, France.
- School of Science, Engineering and Environment. University of Salford, Manchester, M5 4WT, UK.
| |
Collapse
|
4
|
Cheng L, Li Y, Yan J. Space biological and human survival: Investigations into plants, animals, microorganisms and their components and bioregenerative life support systems. LIFE SCIENCES IN SPACE RESEARCH 2025; 44:143-153. [PMID: 39864907 DOI: 10.1016/j.lssr.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 01/28/2025]
Abstract
Space life science has been a frontier discipline in the life sciences, aiming to study the life phenomena of earth organisms and their activity patterns under the special environment of space. This review summarizes studies in various key topics in space life science, namely, how microbiome changes in humans and plants, the development of space agriculture and the use of animal, plant and cell models to study the effect of space environments on physiology. We highlight the new possibilities of using high-quality protein crystals uniquely available when grown under space conditions to aid drug development on earth, and the state-of-the-art Bioregenerative Life Support Systems (BLSS) to achieve long term human survival in space.
Collapse
Affiliation(s)
- Lin Cheng
- Holosensor Medical Technology Ltd, Room 12, No. 1798, Zhonghuayuan West Road, Yushan Town, Suzhou 215000, PR China
| | - Yitong Li
- Holosensor Medical Technology Ltd, Room 12, No. 1798, Zhonghuayuan West Road, Yushan Town, Suzhou 215000, PR China
| | - Jing Yan
- Holosensor Medical Technology Ltd, Room 12, No. 1798, Zhonghuayuan West Road, Yushan Town, Suzhou 215000, PR China; Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
5
|
Casaletto JA, Scott RT, Myrick M, Mackintosh G, Chok H, Saravia-Butler A, Hoarfrost A, Galazka JM, Sanders LM, Costes SV. Analyzing the relationship between gene expression and phenotype in space-flown mice using a causal inference machine learning ensemble. Sci Rep 2025; 15:2363. [PMID: 39824847 PMCID: PMC11748630 DOI: 10.1038/s41598-024-81394-y] [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: 04/12/2024] [Accepted: 11/26/2024] [Indexed: 01/20/2025] Open
Abstract
Spaceflight has several detrimental effects on human and rodent health. For example, liver dysfunction is a common phenotype observed in space-flown rodents, and this dysfunction is partially reflected in transcriptomic changes. Studies linking transcriptomics with liver dysfunction rely on tools which exploit correlation, but these tools make no attempt to disambiguate true correlations from spurious ones. In this work, we use a machine learning ensemble of causal inference methods called the Causal Research and Inference Search Platform (CRISP) which was developed to predict causal features of a binary response variable from high-dimensional input. We used CRISP to identify genes robustly correlated with a lipid density phenotype using transcriptomic and histological data from the NASA Open Science Data Repository (OSDR). Our approach identified genes and molecular targets not predicted by previous traditional differential gene expression analyses. These genes are likely to play a pivotal role in the liver dysfunction observed in space-flown rodents, and this work opens the door to identifying novel countermeasures for space travel.
Collapse
Affiliation(s)
- James A Casaletto
- Blue Marble Space Institute of Science, NASA Ames, Mountain View, USA.
| | | | - Makenna Myrick
- Department of Chemistry, University of Florida, Gainesville, USA
| | | | - Hamed Chok
- Blue Marble Space Institute of Science, NASA Ames, Mountain View, USA
| | | | | | | | | | | |
Collapse
|
6
|
Zhong S, Zheng L, Wu Y, Sun S, Luo Q, Song G, Lü D, Long M. Rotating culture regulates the formation of HepaRG-derived liver organoids via YAP translocation. BMC Biol 2024; 22:262. [PMID: 39548509 PMCID: PMC11568593 DOI: 10.1186/s12915-024-02062-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Liver organoid serves as an alternative model for liver pathophysiology in carbohydrate or lipid metabolism and xenobiotic metabolism transformation. Biomechanical cues including spaceflight mission can affect liver organoid construction and their related functions, but their underlying mechanisms are not fully understood yet. Here, a rotating cell culture device, namely Rotating Flat Chamber (RFC), was specifically designed for adhering cells or cell aggregated to elucidate the effects of altered gravity vector on HepaRG-derived liver organoids construction. RESULTS The organoids so formed under RFC presented the fast growth rate and large projection area. Meanwhile, the expressions of two pluripotency markers of SOX9 and CD44 were enhanced. This finding was positively correlated with the increased YAP expression and nuclear translocation as well as the elevated α4β6-integrin expression. Inhibition of YAP expression and nuclear translocation decreased the expression of SOX9 and CD44 under RFC, thereby attenuating the pluripotency of HepaRG-derived liver organoids. CONCLUSIONS In conclusion, we proposed a novel liver organoid construction method using rotating culture, by which the pluripotency of liver organoids so constructed is mediated by α4β6-integrin and YAP translocation. This work furthered the understanding in how the gravity vector orientation affects the construction of liver organoids and the related mechanotransductive pathways.
Collapse
Affiliation(s)
- Shaoyu Zhong
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Lu Zheng
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi Wu
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shujin Sun
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Dongyuan Lü
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Mian Long
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
7
|
Manna OM, Burgio S, Picone D, Carista A, Pitruzzella A, Fucarino A, Bucchieri F. Microgravity and Human Body: Unraveling the Potential Role of Heat-Shock Proteins in Spaceflight and Future Space Missions. BIOLOGY 2024; 13:921. [PMID: 39596876 PMCID: PMC11591694 DOI: 10.3390/biology13110921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
In recent years, the increasing number of long-duration space missions has prompted the scientific community to undertake a more comprehensive examination of the impact of microgravity on the human body during spaceflight. This review aims to assess the current knowledge regarding the consequences of exposure to an extreme environment, like microgravity, on the human body, focusing on the role of heat-shock proteins (HSPs). Previous studies have demonstrated that long-term exposure to microgravity during spaceflight can cause various changes in the human body, such as muscle atrophy, changes in muscle fiber composition, cardiovascular function, bone density, and even immune system functions. It has been postulated that heat-shock proteins (HSPs) may play a role in mitigating the harmful effects of microgravity-induced stress. According to past studies, heat-shock proteins (HSPs) are upregulated under simulated microgravity conditions. This upregulation assists in the maintenance of the proper folding and function of other proteins during stressful conditions, thereby safeguarding the physiological systems of organisms from the detrimental effects of microgravity. HSPs could also be used as biomarkers to assess the level of cellular stress in tissues and cells exposed to microgravity. Therefore, modulation of HSPs by drugs and genetic or environmental techniques could prove to be a potential therapeutic strategy to reduce the negative physiological consequences of long-duration spaceflight in astronauts.
Collapse
Affiliation(s)
- Olga Maria Manna
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90146 Palermo, Italy
| | - Stefano Burgio
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90146 Palermo, Italy
- Department of Medicine and Surgery, Kore University of Enna, 94100 Enna, Italy
| | - Domiziana Picone
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy
| | - Adelaide Carista
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy
| | - Alessandro Pitruzzella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy
| | - Alberto Fucarino
- Department of Theoretical and Applied Sciences, eCampus University, 22060 Novedrate, Italy
| | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
8
|
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
|
9
|
Finch RH, Vitry G, Siew K, Walsh SB, Behesti A, Hardiman G, da Silveira WA. Spaceflight causes strain dependent gene expression changes associated with lipid and extracellular matrix dysregulation in the mouse kidney in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.13.584781. [PMID: 38559158 PMCID: PMC10979940 DOI: 10.1101/2024.03.13.584781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
To explore new worlds we must ensure humans can survive and thrive in the space environment. Incidence of kidney stones in astronauts is a major risk factor associated with long term missions, caused by increased blood calcium levels due to bone demineralisation triggered by microgravity and space radiation. Transcriptomic changes have been observed in other tissues during spaceflight, including the kidney. We analysed kidney transcriptome patterns in two different strains of mice flown on the International Space Station, C57BL/6J and BALB/c. Here we show a link between spaceflight and transcriptome patterns associated with dysregulation of lipid and extracellular matrix metabolism and altered transforming growth factor-beta signalling. A stronger response was seen in C57BL/6J mice than BALB/c. Genetic differences in hyaluronan metabolism between strains may confer protection against extracellular matrix remodelling through downregulation of epithelial-mesenchymal transition. We intend for our findings to contribute to development of new countermeasures against kidney disease in astronauts and people here on Earth.
Collapse
|
10
|
Mathyk BA, Tabetah M, Karim R, Zaksas V, Kim J, Anu RI, Muratani M, Tasoula A, Singh RS, Chen YK, Overbey E, Park J, Cope H, Fazelinia H, Povero D, Borg J, Klotz RV, Yu M, Young SL, Mason CE, Szewczyk N, St Clair RM, Karouia F, Beheshti A. Spaceflight induces changes in gene expression profiles linked to insulin and estrogen. Commun Biol 2024; 7:692. [PMID: 38862620 PMCID: PMC11166981 DOI: 10.1038/s42003-023-05213-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/03/2023] [Indexed: 06/13/2024] Open
Abstract
Organismal adaptations to spaceflight have been characterized at the molecular level in model organisms, including Drosophila and C. elegans. Here, we extend molecular work to energy metabolism and sex hormone signaling in mice and humans. We found spaceflight induced changes in insulin and estrogen signaling in rodents and humans. Murine changes were most prominent in the liver, where we observed inhibition of insulin and estrogen receptor signaling with concomitant hepatic insulin resistance and steatosis. Based on the metabolic demand, metabolic pathways mediated by insulin and estrogen vary among muscles, specifically between the soleus and extensor digitorum longus. In humans, spaceflight induced changes in insulin and estrogen related genes and pathways. Pathway analysis demonstrated spaceflight induced changes in insulin resistance, estrogen signaling, stress response, and viral infection. These data strongly suggest the need for further research on the metabolic and reproductive endocrinologic effects of space travel, if we are to become a successful interplanetary species.
Collapse
Affiliation(s)
- Begum Aydogan Mathyk
- Department of Obstetrics and Gynecology, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| | - Marshall Tabetah
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Rashid Karim
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, OH, 45220, USA
- Novartis Institutes for Biomedical Research, 181 Massachusetts Ave, Cambridge, MA, 02139, USA
| | - Victoria Zaksas
- Center for Translational Data Science, University of Chicago, Chicago, IL, 60637, USA
- Clever Research Lab, Springfield, IL, 62704, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics and World Quant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA
| | - R I Anu
- Department of Cancer Biology & Therapeutics, Precision Oncology and Multi-omics clinic, Genetic counseling clinic. Department of Clinical Biochemistry, MVR Cancer Centre and Research Institute, Calicut, India
| | - Masafumi Muratani
- Transborder Medical Research Center, University of Tsukuba, Ibaraki, 305-8575, Japan
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Alexia Tasoula
- Department of Life Science Engineering, FH Technikum, Vienna, Austria
| | | | - Yen-Kai Chen
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Eliah Overbey
- Department of Physiology and Biophysics and World Quant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jiwoon Park
- Department of Physiology and Biophysics and World Quant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Henry Cope
- School of Medicine, University of Nottingham, Derby, DE22 3DT, UK
| | - Hossein Fazelinia
- Department of Biomedical and Health Informatics and Proteomics Core Facility, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Davide Povero
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Joseph Borg
- Department of Applied Biomedical Science, Faculty of Health Sciences, Msida, MSD2090, Malta
| | - Remi V Klotz
- Department of Stem Cell Biology & Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Min Yu
- Department of Stem Cell Biology & Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Steven L Young
- Division of Reproductive Endocrinology and Infertility, Duke School of Medicine, Durham, NC, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics and World Quant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Nathaniel Szewczyk
- School of Medicine, University of Nottingham, Derby, DE22 3DT, UK
- Ohio Musculoskeletal and Neurological Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Riley M St Clair
- Department of Life Sciences, Quest University, Squamish, BC, Canada
| | - Fathi Karouia
- Blue Marble Space Institute of Science, Exobiology Branch, NASA Ames Research Center, Moffett Field, CA, USA
- Space Research Within Reach, San Francisco, CA, USA; Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Afshin Beheshti
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.
| |
Collapse
|
11
|
Wu W, Ren J, Han M, Huang B. Influence of gut microbiome on metabolic diseases: a new perspective based on microgravity. J Diabetes Metab Disord 2024; 23:353-364. [PMID: 38932858 PMCID: PMC11196560 DOI: 10.1007/s40200-024-01394-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/28/2024] [Indexed: 06/28/2024]
Abstract
Purpose Microgravity, characterized by gravity levels of 10-3-10-6g, has been found to significantly impair various physiological systems in astronauts, including cardiovascular function, bone density, and metabolism. With the recent surge in human spaceflight, understanding the impact of microgravity on biological health has become paramount. Methods A comprehensive literature search was performed using the PubMed database to identify relevant publications pertaining to the interplay between gut microbiome, microgravity, space environment, and metabolic diseases. Results This comprehensive review primarily focuses on the progress made in investigating the gut microbiome and its association with metabolic diseases under microgravity conditions. Microgravity induces notable alterations in the composition, diversity, and functionality of the gut microbiome. These changes hold direct implications for metabolic disorders such as cardiovascular disease (CVD), bone metabolism disorders, energy metabolism dysregulation, liver dysfunction, and complications during pregnancy. Conclusion This novel perspective is crucial for preparing for deep space exploration and interstellar migration, where understanding the complex interplay between the gut microbiome and metabolic health becomes indispensable.
Collapse
Affiliation(s)
- Wanxin Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui China
| | - Junjie Ren
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, Anhui China
| | - Maozhen Han
- School of Life Sciences, Anhui Medical University, Hefei, 230032 Anhui China
| | - Binbin Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui China
| |
Collapse
|
12
|
Cialdai F, Brown AM, Baumann CW, Angeloni D, Baatout S, Benchoua A, Bereiter-Hahn J, Bottai D, Buchheim JI, Calvaruso M, Carnero-Diaz E, Castiglioni S, Cavalieri D, Ceccarelli G, Choukér A, Ciofani G, Coppola G, Cusella G, Degl'Innocenti A, Desaphy JF, Frippiat JP, Gelinsky M, Genchi G, Grano M, Grimm D, Guignandon A, Hahn C, Hatton J, Herranz R, Hellweg CE, Iorio CS, Karapantsios T, van Loon J, Lulli M, Maier J, Malda J, Mamaca E, Morbidelli L, van Ombergen A, Osterman A, Ovsianikov A, Pampaloni F, Pavezlorie E, Pereda-Campos V, Przybyla C, Puhl C, Rettberg P, Risaliti C, Rizzo AM, Robson-Brown K, Rossi L, Russo G, Salvetti A, Santucci D, Sperl M, Strollo F, Tabury K, Tavella S, Thielemann C, Willaert R, Szewczyk NJ, Monici M. How do gravity alterations affect animal and human systems at a cellular/tissue level? NPJ Microgravity 2023; 9:84. [PMID: 37865644 PMCID: PMC10590411 DOI: 10.1038/s41526-023-00330-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 10/11/2023] [Indexed: 10/23/2023] Open
Abstract
The present white paper concerns the indications and recommendations of the SciSpacE Science Community to make progress in filling the gaps of knowledge that prevent us from answering the question: "How Do Gravity Alterations Affect Animal and Human Systems at a Cellular/Tissue Level?" This is one of the five major scientific issues of the ESA roadmap "Biology in Space and Analogue Environments". Despite the many studies conducted so far on spaceflight adaptation mechanisms and related pathophysiological alterations observed in astronauts, we are not yet able to elaborate a synthetic integrated model of the many changes occurring at different system and functional levels. Consequently, it is difficult to develop credible models for predicting long-term consequences of human adaptation to the space environment, as well as to implement medical support plans for long-term missions and a strategy for preventing the possible health risks due to prolonged exposure to spaceflight beyond the low Earth orbit (LEO). The research activities suggested by the scientific community have the aim to overcome these problems by striving to connect biological and physiological aspects in a more holistic view of space adaptation effects.
Collapse
Affiliation(s)
- Francesca Cialdai
- ASAcampus Joint Laboratory, ASA Res. Div., DSBSC-University of Florence, Florence, Italy
| | - Austin M Brown
- Honors Tutorial College, Ohio University, Athens, OH, USA
| | - Cory W Baumann
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Debora Angeloni
- Inst. of Biorobotics, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Sarah Baatout
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN) Boeretang 200, 2400, Mol, Belgium
| | | | - Juergen Bereiter-Hahn
- Inst. for Cell and Neurobiol, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Daniele Bottai
- Dept. Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Judith-Irina Buchheim
- Laboratory of "Translational Research, Stress & Immunity", Department of Anesthesiology, LMU University Hospital Munich, Munich, Germany
| | - Marco Calvaruso
- Inst. Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Cefalù, Italy
| | - Eugénie Carnero-Diaz
- Inst. Systematic, Evolution, Biodiversity, Sorbonne University, NMNH, CNRS, EPHE, UA, Paris, France
| | - Sara Castiglioni
- Dept. of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | | | - Gabriele Ceccarelli
- Dept of Public Health, Experimental Medicine and Forensic, University of Pavia, Pavia, Italy
| | - Alexander Choukér
- Laboratory of "Translational Research, Stress & Immunity", Department of Anesthesiology, LMU University Hospital Munich, Munich, Germany
| | - Gianni Ciofani
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, 56025, Pontedera (PI), Italy
| | - Giuseppe Coppola
- Institute of Applied Science and Intelligent Sistems - CNR, Naples, Italy
| | - Gabriella Cusella
- Dept of Public Health, Experimental Medicine and Forensic, University of Pavia, Pavia, Italy
| | - Andrea Degl'Innocenti
- Dept Medical Biotechnologies, University of Siena, Siena, Italy
- Smart Bio-Interfaces, IIT, Pontedera (PI), Italy
| | - Jean-Francois Desaphy
- Dept. Precision and Regenerative Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Jean-Pol Frippiat
- Stress, Immunity, Pathogens Laboratory, SIMPA, Université de Lorraine, Nancy, France
| | - Michael Gelinsky
- Centre for Translational Bone, Joint & Soft Tissue Research, TU Dresden, Dresden, Germany
| | - Giada Genchi
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, 56025, Pontedera (PI), Italy
| | - Maria Grano
- Dept. Precision and Regenerative Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Daniela Grimm
- Dept. Microgravity and Translational Regenerative Medicine, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Dept of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Alain Guignandon
- SAINBIOSE, INSERM U1059, Université Jean Monnet, F-42000, Saint-Etienne, France
| | | | | | - Raúl Herranz
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Christine E Hellweg
- Radiation Biology Dept., Inst. of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | | | | | - Jack van Loon
- Amsterdam University Medical Center, ACTA/VU, Amsterdam, The Netherlands
| | - Matteo Lulli
- Dept. Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Jeanette Maier
- Dept. of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Jos Malda
- Dept. Orthopaedics, Univ. Med. Center Utrecht & Dept. Clinical Sciences, Utrecht Univ, Utrecht, The Netherlands
| | - Emina Mamaca
- European and International Affairs Dept, Ifremer centre Bretagne, Plouzané, France
| | | | | | - Andreas Osterman
- Max von Pettenkofer Institute, Virology, LMU Munich & DZIF, Partner Site Munich, Munich, Germany
| | - Aleksandr Ovsianikov
- 3D Printing and Biofabrication, Inst. Materials Science and Technology, TU Wien, Vienna, Austria
| | - Francesco Pampaloni
- Buchmann Inst. for Molecular Life Sciences, Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Elizabeth Pavezlorie
- Ludwig Boltzmann Inst. for Traumatology, Res. Center in Cooperation with AUVA, Vienna, Austria
| | - Veronica Pereda-Campos
- GSBMS/URU EVOLSAN - Medecine Evolutive, Université Paul Sabatier Toulouse III, Toulouse, France
| | - Cyrille Przybyla
- MARBEC, Univ Montpellier, CNRS, Ifremer, IRD, Palavas les Flots, France
| | - Christopher Puhl
- Space Applications NV/SA for European Space Agency, Houston, USA
| | - Petra Rettberg
- DLR, Inst of Aerospace Medicine, Research Group Astrobiology, Köln, Germany
| | - Chiara Risaliti
- ASAcampus Joint Laboratory, ASA Res. Div., DSBSC-University of Florence, Florence, Italy
| | - Angela Maria Rizzo
- Dept. of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Kate Robson-Brown
- Dept of Engineering Mathematics, and Dept of Anthropology and Archaeology, University of Bristol, Bristol, UK
| | - Leonardo Rossi
- Dept. Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giorgio Russo
- Inst. Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Cefalù, Italy
| | | | - Daniela Santucci
- Center for Behavioural Sciences and Mental Health, Ist. Superiore Sanità, Rome, Italy
| | | | - Felice Strollo
- Endocrinology and Metabolism Unit, IRCCS San Raffaele Pisana, Rome, Italy
| | - Kevin Tabury
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN) Boeretang 200, 2400, Mol, Belgium
| | - Sara Tavella
- IRCCS Ospedale Policlinico San Martino and University of Genoa, DIMES, Genoa, Italy
| | | | - Ronnie Willaert
- Research Group NAMI and NANO, Vrije Universiteit Brussels, Brussels, Belgium
| | | | - Monica Monici
- ASAcampus Joint Laboratory, ASA Res. Div., DSBSC-University of Florence, Florence, Italy.
| |
Collapse
|
13
|
Xiong Y, Ma C, Li Q, Zhang W, Zhao H, Ren P, Zhang K, Lei X. Melatonin ameliorates simulated-microgravity-induced mitochondrial dysfunction and lipid metabolism dysregulation in hepatocytes. FASEB J 2023; 37:e23132. [PMID: 37552471 DOI: 10.1096/fj.202301137r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/12/2023] [Accepted: 07/26/2023] [Indexed: 08/09/2023]
Abstract
The liver is an essential multifunctional organ, which constantly communicates with nearly all tissues. It has raised the concern that microgravity exposure can lead to liver dysfunction and metabolic syndromes. However, molecular mechanisms and intervention measures of the adverse effects of microgravity on hepatocytes are limited. In this study, we utilized the random positioning machine culture system to investigate the adverse effects on hepatocytes under simulated microgravity (SMG). Our results showed that SMG impaired hepatocyte viability, causing cell cycle arrest and apoptosis. Compared to normal gravity, it also triggered lipid accumulation, elevated triglyceride (TG) and ROS levels, and impaired mitochondria function in hepatocytes. Furthermore, RNA sequencing results showed that SMG upregulated genes implicated in lipid metabolisms, including PPARγ, PLIN2, CD36, FABPs, etc. Importantly, all these defects can be suppressed by melatonin, a potent antioxidant secreted by the pineal gland, suggesting its potential use of therapeutic intervention.
Collapse
Affiliation(s)
- Yue Xiong
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chiyuan Ma
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qin Li
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wenya Zhang
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huashan Zhao
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Peigen Ren
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ke Zhang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, China
| | - Xiaohua Lei
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
14
|
Kremsky I, Ali S, Stanbouly S, Holley J, Justinen S, Pecaut M, Crapo J, Mao X. Spaceflight-Induced Gene Expression Profiles in the Mouse Brain Are Attenuated by Treatment with the Antioxidant BuOE. Int J Mol Sci 2023; 24:13569. [PMID: 37686374 PMCID: PMC10487739 DOI: 10.3390/ijms241713569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
The demands of deep space pose a health risk to the central nervous system that has long been a concern when sending humans to space. While little is known about how spaceflight affects transcription spatially in the brain, a greater understanding of this process has the potential to aid strategies that mitigate the effects of spaceflight on the brain. Therefore, we performed GeoMx Digital Spatial Profiling of mouse brains subjected to either spaceflight or grounded controls. Four brain regions were selected: Cortex, Frontal Cortex, Corunu Ammonis I, and Dentate Gyrus. Antioxidants have emerged as a potential means of attenuating the effects of spaceflight, so we treated a subset of the mice with a superoxide dismutase mimic, MnTnBuOE-2-PyP 5+ (BuOE). Our analysis revealed hundreds of differentially expressed genes due to spaceflight in each of the four brain regions. Both common and region-specific transcriptomic responses were observed. Metabolic pathways and pathways sensitive to oxidative stress were enriched in the four brain regions due to spaceflight. These findings enhance our understanding of brain regional variation in susceptibility to spaceflight conditions. BuOE reduced the transcriptomic effects of spaceflight at a large number of genes, suggesting that this compound may attenuate oxidative stress-induced brain damage caused by the spaceflight environment.
Collapse
Affiliation(s)
- Isaac Kremsky
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Samir Ali
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
| | - Seta Stanbouly
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
| | - Jacob Holley
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
| | - Stephen Justinen
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
| | - Michael Pecaut
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
| | - James Crapo
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, University of Colorado Denver, Denver, CO 80206, USA;
| | - Xiaowen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (I.K.); (S.A.); (S.S.); (J.H.); (S.J.); (M.P.)
| |
Collapse
|
15
|
Sanders LM, Scott RT, Yang JH, Qutub AA, Garcia Martin H, Berrios DC, Hastings JJA, Rask J, Mackintosh G, Hoarfrost AL, Chalk S, Kalantari J, Khezeli K, Antonsen EL, Babdor J, Barker R, Baranzini SE, Beheshti A, Delgado-Aparicio GM, Glicksberg BS, Greene CS, Haendel M, Hamid AA, Heller P, Jamieson D, Jarvis KJ, Komarova SV, Komorowski M, Kothiyal P, Mahabal A, Manor U, Mason CE, Matar M, Mias GI, Miller J, Myers JG, Nelson C, Oribello J, Park SM, Parsons-Wingerter P, Prabhu RK, Reynolds RJ, Saravia-Butler A, Saria S, Sawyer A, Singh NK, Snyder M, Soboczenski F, Soman K, Theriot CA, Van Valen D, Venkateswaran K, Warren L, Worthey L, Zitnik M, Costes SV. Biological research and self-driving labs in deep space supported by artificial intelligence. NAT MACH INTELL 2023. [DOI: 10.1038/s42256-023-00618-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
16
|
Cope H, Elsborg J, Demharter S, Mcdonald JT, Wernecke C, Parthasarathy H, Unadkat H, Chatrathi M, Claudio J, Reinsch S, Zwart S, Smith S, Heer M, Muratani M, Meydan C, Overbey E, Kim J, Park J, Schisler J, Mason C, Szewczyk N, Willis C, Salam A, Beheshti A. More than a Feeling: Dermatological Changes Impacted by Spaceflight. RESEARCH SQUARE 2023:rs.3.rs-2367727. [PMID: 36798347 PMCID: PMC9934743 DOI: 10.21203/rs.3.rs-2367727/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Spaceflight poses a unique set of challenges to humans and the hostile Spaceflight environment can induce a wide range of increased health risks, including dermatological issues. The biology driving the frequency of skin issues in astronauts is currently not well understood. To address this issue, we used a systems biology approach utilizing NASA's Open Science Data Repository (OSDR) on spaceflown murine transcriptomic datasets focused on the skin, biomedical profiles from fifty NASA astronauts, and confirmation via transcriptomic data from JAXA astronauts, the NASA Twins Study, and the first civilian commercial mission, Inspiration4. Key biological changes related to skin health, DNA damage & repair, and mitochondrial dysregulation were determined to be involved with skin health risks during Spaceflight. Additionally, a machine learning model was utilized to determine key genes driving Spaceflight response in the skin. These results can be used for determining potential countermeasures to mitigate Spaceflight damage to the skin.
Collapse
|
17
|
Li W, Shu X, Zhang X, Zhang Z, Sun S, Li N, Long M. Potential Roles of YAP/TAZ Mechanotransduction in Spaceflight-Induced Liver Dysfunction. Int J Mol Sci 2023; 24:ijms24032197. [PMID: 36768527 PMCID: PMC9917057 DOI: 10.3390/ijms24032197] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Microgravity exposure during spaceflight causes the disordered regulation of liver function, presenting a specialized mechano-biological coupling process. While YAP/TAZ serves as a typical mechanosensitive pathway involved in hepatocyte metabolism, it remains unclear whether and how it is correlated with microgravity-induced liver dysfunction. Here, we discussed liver function alterations induced by spaceflight or simulated effects of microgravity on Earth. The roles of YAP/TAZ serving as a potential bridge in connecting liver metabolism with microgravity were specifically summarized. Existing evidence indicated that YAP/TAZ target gene expressions were affected by mechanotransductive pathways and phase separation, reasonably speculating that microgravity might regulate YAP/TAZ activation by disrupting these pathways via cytoskeletal remodeling or nuclear deformation, or disturbing condensates formation via diffusion limit, and then breaking liver homeostasis.
Collapse
Affiliation(s)
- Wang Li
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
- School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyu Shu
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
- School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyu Zhang
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
- School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ziliang Zhang
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Shujin Sun
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
- School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ning Li
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
- School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence: (N.L.); (M.L.)
| | - Mian Long
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
- School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence: (N.L.); (M.L.)
| |
Collapse
|
18
|
Cahill T, da Silveira WA, Renaud L, Wang H, Williamson T, Chung D, Chan S, Overton I, Hardiman G. Investigating the effects of chronic low-dose radiation exposure in the liver of a hypothermic zebrafish model. Sci Rep 2023; 13:918. [PMID: 36650199 PMCID: PMC9845366 DOI: 10.1038/s41598-022-26976-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/22/2022] [Indexed: 01/18/2023] Open
Abstract
Mankind's quest for a manned mission to Mars is placing increased emphasis on the development of innovative radio-protective countermeasures for long-term space travel. Hibernation confers radio-protective effects in hibernating animals, and this has led to the investigation of synthetic torpor to mitigate the deleterious effects of chronic low-dose-rate radiation exposure. Here we describe an induced torpor model we developed using the zebrafish. We explored the effects of radiation exposure on this model with a focus on the liver. Transcriptomic and behavioural analyses were performed. Radiation exposure resulted in transcriptomic perturbations in lipid metabolism and absorption, wound healing, immune response, and fibrogenic pathways. Induced torpor reduced metabolism and increased pro-survival, anti-apoptotic, and DNA repair pathways. Coupled with radiation exposure, induced torpor led to a stress response but also revealed maintenance of DNA repair mechanisms, pro-survival and anti-apoptotic signals. To further characterise our model of induced torpor, the zebrafish model was compared with hepatic transcriptomic data from hibernating grizzly bears (Ursus arctos horribilis) and active controls revealing conserved responses in gene expression associated with anti-apoptotic processes, DNA damage repair, cell survival, proliferation, and antioxidant response. Similarly, the radiation group was compared with space-flown mice revealing shared changes in lipid metabolism.
Collapse
Affiliation(s)
- Thomas Cahill
- School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, Belfast, BT9 5DL, UK
| | - Willian Abraham da Silveira
- School of Health, Science and Wellbeing, Department of Biological Sciences, Science Centre, Staffordshire University, Leek Road, Stoke-On-Trent, ST4 2DF, UK
- International Space University, 1 Rue Jean-Dominique Cassini, 67400, Illkirch-Graffenstaden, France
| | - Ludivine Renaud
- Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Hao Wang
- School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, Belfast, BT9 5DL, UK
| | - Tucker Williamson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Dongjun Chung
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Sherine Chan
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
- JLABS at the Children's National Research and Innovation Campus, Washington, DC, 20012, USA
| | - Ian Overton
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE, UK
| | - Gary Hardiman
- School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, Belfast, BT9 5DL, UK.
- Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
19
|
Manaig YJY, Criado-Mesas L, Esteve-Codina A, Mármol-Sánchez E, Castelló A, Sánchez A, Folch JM. Identifying miRNA-mRNA regulatory networks on extreme n-6/n-3 polyunsaturated fatty acid ratio expression profiles in porcine skeletal muscle. PLoS One 2023; 18:e0283231. [PMID: 37141193 PMCID: PMC10159129 DOI: 10.1371/journal.pone.0283231] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/06/2023] [Indexed: 05/05/2023] Open
Abstract
Omega-3 (n-3) and omega-6 (n-6) polyunsaturated fatty acids (PUFAs) are essential fatty acids with antagonistic inflammatory functions that play vital roles in metabolic health and immune response. Current commercial swine diets tend to over-supplement with n-6 PUFAs, which may increase the likelihood of developing inflammatory diseases and affect the overall well-being of the animals. However, it is still poorly understood how n-6/n-3 PUFA ratios affect the porcine transcriptome expression and how messenger RNAs (mRNAs) and microRNAs (miRNAs) might regulate biological processes related to PUFA metabolism. On account of this, we selected a total of 20 Iberian × Duroc crossbred pigs with extreme values for n-6/n-3 FA ratio (10 high vs 10 low), and longissimus dorsi muscle samples were used to identify differentially expressed mRNAs and miRNAs. The observed differentially expressed mRNAs were associated to biological pathways related to muscle growth and immunomodulation, while the differentially expressed microRNAs (ssc-miR-30a-3p, ssc-miR-30e-3p, ssc-miR-15b and ssc-miR-7142-3p) were correlated to adipogenesis and immunity. Relevant miRNA-to-mRNA regulatory networks were also predicted (i.e., mir15b to ARRDC3; mir-7142-3p to METTL21C), and linked to lipolysis, obesity, myogenesis, and protein degradation. The n-6/n-3 PUFA ratio differences in pig skeletal muscle revealed genes, miRNAs and enriched pathways involved in lipid metabolism, cell proliferation and inflammation.
Collapse
Affiliation(s)
- Yron Joseph Yabut Manaig
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB Consortium, Bellaterra, Barcelona, Spain
- Department of Veterinary Medicine and Animal Sciences, Università degli Studi di Milano, Lodi, Italy
| | - Lourdes Criado-Mesas
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB Consortium, Bellaterra, Barcelona, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Emilio Mármol-Sánchez
- Department of Molecular Biosciences, Science for Life Laboratory, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- Centre for Palaeogenetics, Stockholm, Sweden
| | - Anna Castelló
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB Consortium, Bellaterra, Barcelona, Spain
| | - Armand Sánchez
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB Consortium, Bellaterra, Barcelona, Spain
| | - Josep M Folch
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB Consortium, Bellaterra, Barcelona, Spain
| |
Collapse
|
20
|
Krittanawong C, Singh NK, Scheuring RA, Urquieta E, Bershad EM, Macaulay TR, Kaplin S, Dunn C, Kry SF, Russomano T, Shepanek M, Stowe RP, Kirkpatrick AW, Broderick TJ, Sibonga JD, Lee AG, Crucian BE. Human Health during Space Travel: State-of-the-Art Review. Cells 2022; 12:cells12010040. [PMID: 36611835 PMCID: PMC9818606 DOI: 10.3390/cells12010040] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
The field of human space travel is in the midst of a dramatic revolution. Upcoming missions are looking to push the boundaries of space travel, with plans to travel for longer distances and durations than ever before. Both the National Aeronautics and Space Administration (NASA) and several commercial space companies (e.g., Blue Origin, SpaceX, Virgin Galactic) have already started the process of preparing for long-distance, long-duration space exploration and currently plan to explore inner solar planets (e.g., Mars) by the 2030s. With the emergence of space tourism, space travel has materialized as a potential new, exciting frontier of business, hospitality, medicine, and technology in the coming years. However, current evidence regarding human health in space is very limited, particularly pertaining to short-term and long-term space travel. This review synthesizes developments across the continuum of space health including prior studies and unpublished data from NASA related to each individual organ system, and medical screening prior to space travel. We categorized the extraterrestrial environment into exogenous (e.g., space radiation and microgravity) and endogenous processes (e.g., alteration of humans' natural circadian rhythm and mental health due to confinement, isolation, immobilization, and lack of social interaction) and their various effects on human health. The aim of this review is to explore the potential health challenges associated with space travel and how they may be overcome in order to enable new paradigms for space health, as well as the use of emerging Artificial Intelligence based (AI) technology to propel future space health research.
Collapse
Affiliation(s)
- Chayakrit Krittanawong
- Department of Medicine and Center for Space Medicine, Section of Cardiology, Baylor College of Medicine, Houston, TX 77030, USA
- Translational Research Institute for Space Health, Houston, TX 77030, USA
- Department of Cardiovascular Diseases, New York University School of Medicine, New York, NY 10016, USA
- Correspondence: or (C.K.); (B.E.C.); Tel.: +1-713-798-4951 (C.K.); +1-281-483-0123 (B.E.C.)
| | - Nitin Kumar Singh
- Biotechnology and Planetary Protection Group, Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA 91109, USA
| | | | - Emmanuel Urquieta
- Translational Research Institute for Space Health, Houston, TX 77030, USA
- Department of Emergency Medicine and Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Eric M. Bershad
- Department of Neurology, Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Scott Kaplin
- Department of Cardiovascular Diseases, New York University School of Medicine, New York, NY 10016, USA
| | - Carly Dunn
- Department of Dermatology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephen F. Kry
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Marc Shepanek
- Office of the Chief Health and Medical Officer, NASA, Washington, DC 20546, USA
| | | | - Andrew W. Kirkpatrick
- Department of Surgery and Critical Care Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | | | - Jean D. Sibonga
- Division of Biomedical Research and Environmental Sciences, NASA Lyndon B. Johnson Space Center, Houston, TX 77058, USA
| | - Andrew G. Lee
- Department of Ophthalmology, University of Texas Medical Branch School of Medicine, Galveston, TX 77555, USA
- Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Ophthalmology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Ophthalmology, Texas A and M College of Medicine, College Station, TX 77807, USA
- Department of Ophthalmology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
- Departments of Ophthalmology, Neurology, and Neurosurgery, Weill Cornell Medicine, New York, NY 10021, USA
| | - Brian E. Crucian
- National Aeronautics and Space Administration (NASA) Johnson Space Center, Human Health and Performance Directorate, Houston, TX 77058, USA
- Correspondence: or (C.K.); (B.E.C.); Tel.: +1-713-798-4951 (C.K.); +1-281-483-0123 (B.E.C.)
| |
Collapse
|
21
|
Vinken M. Hepatology in space: Effects of spaceflight and simulated microgravity on the liver. Liver Int 2022; 42:2599-2606. [PMID: 36183343 DOI: 10.1111/liv.15444] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/22/2022] [Accepted: 09/29/2022] [Indexed: 12/13/2022]
Abstract
Microgravity as experienced during spaceflight affects a number of physiological processes in various organs. However, effects on the liver have yet been poorly documented. Nevertheless, the liver is a metabolically highly active organ involved in carbohydrate metabolism, lipid metabolism and xenobiotic biotransformation. The present paper provides an overview of the effects of microgravity on the liver observed in experimental animals during actual spaceflight and upon simulation of microgravity on Earth. These include (i) induction of liver injury and inflammation associated with apoptosis and oxidative stress, (ii) changes in liver carbohydrate metabolism resulting in the onset of a diabetogenic phenotype, (iii) modifications in hepatic lipid metabolism leading to early non-alcoholic fatty liver disease and (iv) alterations of the hepatic xenobiotic biotransformation machinery. Although most of these observations remain to be fully validated in humans, appropriate measures to counteract liver pathogenesis should be considered, especially in view of long-term space missions.
Collapse
Affiliation(s)
- Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
22
|
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
|
23
|
Dello Russo C, Bandiera T, Monici M, Surdo L, Yip VLM, Wotring V, Morbidelli L. Physiological adaptations affecting drug pharmacokinetics in space: what do we really know? A critical review of the literature. Br J Pharmacol 2022; 179:2538-2557. [PMID: 35170019 PMCID: PMC9314132 DOI: 10.1111/bph.15822] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 01/13/2022] [Accepted: 02/10/2022] [Indexed: 12/03/2022] Open
Abstract
As human spaceflight progresses with extended mission durations, the demand for effective and safe drugs will necessarily increase. To date, the accepted medications used during missions (for space motion sickness, sleep disturbances, allergies, pain, and sinus congestion) are administered under the assumption that they act as safely and efficaciously as on Earth. However, physiological changes have been documented in human subjects in spaceflight involving fluid shifts, muscle and bone loss, immune system dysregulation, and adjustments in the gastrointestinal tract and metabolism. These alterations may change the pharmacokinetics (PK) and pharmacodynamics of commonly used medications. Frustratingly, the information gained from bed rest studies and from in-flight observations is incomplete and also demonstrates a high variability in drug PK. Therefore, the objectives of this review are to report (i) the impact of the space environmental stressors on human physiology in relation to PK; (ii) the state-of-the-art on experimental data in space and/or in ground-based models; (iii) the validation of ground-based models for PK studies; and (iv) the identification of research gaps.
Collapse
Affiliation(s)
- Cinzia Dello Russo
- Department of Healthcare Surveillance and Bioethics, Section of PharmacologyUniversità Cattolica del Sacro CuoreRomeItaly
- Fondazione Policlinico Universitario A. Gemelli IRCCSRomeItaly
- MRC Centre for Drug Safety Science and Wolfson Centre for Personalised Medicine, Institute of Systems, Molecular and Integrative Biology (ISMIB)University of LiverpoolLiverpoolUK
| | - Tiziano Bandiera
- D3‐PharmaChemistry LineIstituto Italiano di Tecnologia (IIT)GenoaItaly
| | - Monica Monici
- ASAcampus Joint Laboratory, ASA Res. Div. & Dept. of Experimental and Clinical Biomedical Sciences “Mario Serio”University of FlorenceFlorenceItaly
| | - Leonardo Surdo
- Space Applications Services NV/SA for the European Space AgencyNoordwijkThe Netherlands
| | - Vincent Lai Ming Yip
- MRC Centre for Drug Safety Science and Wolfson Centre for Personalised Medicine, Institute of Systems, Molecular and Integrative Biology (ISMIB)University of LiverpoolLiverpoolUK
| | | | | |
Collapse
|
24
|
Uruno A, Saigusa D, Suzuki T, Yumoto A, Nakamura T, Matsukawa N, Yamazaki T, Saito R, Taguchi K, Suzuki M, Suzuki N, Otsuki A, Katsuoka F, Hishinuma E, Okada R, Koshiba S, Tomioka Y, Shimizu R, Shirakawa M, Kensler TW, Shiba D, Yamamoto M. Nrf2 plays a critical role in the metabolic response during and after spaceflight. Commun Biol 2021; 4:1381. [PMID: 34887485 PMCID: PMC8660801 DOI: 10.1038/s42003-021-02904-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 11/16/2021] [Indexed: 11/09/2022] Open
Abstract
Space travel induces stresses that contribute to health problems, as well as inducing the expression of Nrf2 (NF-E2-related factor-2) target genes that mediate adaptive responses to oxidative and other stress responses. The volume of epididymal white adipose tissue (eWAT) in mice increases during spaceflight, a change that is attenuated by Nrf2 knockout. We conducted metabolome analyses of plasma from wild-type and Nrf2 knockout mice collected at pre-flight, in-flight and post-flight time points, as well as tissues collected post-flight to clarify the metabolic responses during and after spaceflight and the contribution of Nrf2 to these responses. Plasma glycerophospholipid and sphingolipid levels were elevated during spaceflight, whereas triacylglycerol levels were lower after spaceflight. In wild-type mouse eWAT, triacylglycerol levels were increased, but phosphatidylcholine levels were decreased, and these changes were attenuated in Nrf2 knockout mice. Transcriptome analyses revealed marked changes in the expression of lipid-related genes in the liver and eWAT after spaceflight and the effects of Nrf2 knockout on these changes. Based on these results, we concluded that space stress provokes significant responses in lipid metabolism during and after spaceflight; Nrf2 plays critical roles in these responses.
Collapse
Affiliation(s)
- Akira Uruno
- grid.69566.3a0000 0001 2248 6943Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan ,grid.69566.3a0000 0001 2248 6943Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Daisuke Saigusa
- grid.69566.3a0000 0001 2248 6943Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan ,grid.69566.3a0000 0001 2248 6943Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takafumi Suzuki
- grid.69566.3a0000 0001 2248 6943Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akane Yumoto
- JEM Utilization Center, Human Spaceflight Technology Directorate, JAXA, Tsukuba, Japan
| | - Tomohiro Nakamura
- grid.69566.3a0000 0001 2248 6943Department of Health Record Informatics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Naomi Matsukawa
- grid.69566.3a0000 0001 2248 6943Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Takahiro Yamazaki
- grid.69566.3a0000 0001 2248 6943Laboratory of Oncology, Pharmacy Practice and Sciences, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ristumi Saito
- grid.69566.3a0000 0001 2248 6943Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan ,grid.69566.3a0000 0001 2248 6943Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keiko Taguchi
- grid.69566.3a0000 0001 2248 6943Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan ,grid.69566.3a0000 0001 2248 6943Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan ,grid.69566.3a0000 0001 2248 6943Advanced Research Center for Innovations in Next-GEneration Medicine (INGEM), Tohoku University, Sendai, Japan
| | - Mikiko Suzuki
- grid.69566.3a0000 0001 2248 6943Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan ,grid.69566.3a0000 0001 2248 6943Center for Radioisotope Sciences, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Norio Suzuki
- grid.69566.3a0000 0001 2248 6943Division of Oxygen Biology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akihito Otsuki
- grid.69566.3a0000 0001 2248 6943Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan ,grid.69566.3a0000 0001 2248 6943Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Fumiki Katsuoka
- grid.69566.3a0000 0001 2248 6943Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan ,grid.69566.3a0000 0001 2248 6943Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan ,grid.69566.3a0000 0001 2248 6943Advanced Research Center for Innovations in Next-GEneration Medicine (INGEM), Tohoku University, Sendai, Japan
| | - Eiji Hishinuma
- grid.69566.3a0000 0001 2248 6943Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan ,grid.69566.3a0000 0001 2248 6943Advanced Research Center for Innovations in Next-GEneration Medicine (INGEM), Tohoku University, Sendai, Japan
| | - Risa Okada
- JEM Utilization Center, Human Spaceflight Technology Directorate, JAXA, Tsukuba, Japan
| | - Seizo Koshiba
- grid.69566.3a0000 0001 2248 6943Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan ,grid.69566.3a0000 0001 2248 6943Advanced Research Center for Innovations in Next-GEneration Medicine (INGEM), Tohoku University, Sendai, Japan
| | - Yoshihisa Tomioka
- grid.69566.3a0000 0001 2248 6943Laboratory of Oncology, Pharmacy Practice and Sciences, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ritsuko Shimizu
- grid.69566.3a0000 0001 2248 6943Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan ,grid.69566.3a0000 0001 2248 6943Department of Molecular Hematology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masaki Shirakawa
- JEM Utilization Center, Human Spaceflight Technology Directorate, JAXA, Tsukuba, Japan
| | - Thomas W. Kensler
- grid.270240.30000 0001 2180 1622Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | - Dai Shiba
- JEM Utilization Center, Human Spaceflight Technology Directorate, JAXA, Tsukuba, Japan.
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan. .,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
25
|
Barnette BL, Yu Y, Ullrich RL, Emmett MR. Mitochondrial Effects in the Liver of C57BL/6 Mice by Low Dose, High Energy, High Charge Irradiation. Int J Mol Sci 2021; 22:ijms222111806. [PMID: 34769236 PMCID: PMC8584048 DOI: 10.3390/ijms222111806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/19/2021] [Accepted: 10/25/2021] [Indexed: 12/19/2022] Open
Abstract
Galactic cosmic rays are primarily composed of protons (85%), helium (14%), and high charge/high energy ions (HZEs) such as 56Fe, 28Si, and 16O. HZE exposure is a major risk factor for astronauts during deep-space travel due to the possibility of HZE-induced cancer. A systems biology integrated omics approach encompassing transcriptomics, proteomics, lipidomics, and functional biochemical assays was used to identify microenvironmental changes induced by HZE exposure. C57BL/6 mice were placed into six treatment groups and received the following irradiation treatments: 600 MeV/n 56Fe (0.2 Gy), 1 GeV/n 16O (0.2 Gy), 350 MeV/n 28Si (0.2 Gy), 137Cs (1.0 Gy) gamma rays, 137Cs (3.0 Gy) gamma rays, and sham irradiation. Left liver lobes were collected at 30, 60, 120, 270, and 360 days post-irradiation. Analysis of transcriptomic and proteomic data utilizing ingenuity pathway analysis identified multiple pathways involved in mitochondrial function that were altered after HZE irradiation. Lipids also exhibited changes that were linked to mitochondrial function. Molecular assays for mitochondrial Complex I activity showed significant decreases in activity after HZE exposure. HZE-induced mitochondrial dysfunction suggests an increased risk for deep space travel. Microenvironmental and pathway analysis as performed in this research identified possible targets for countermeasures to mitigate risk.
Collapse
Affiliation(s)
- Brooke L. Barnette
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA;
| | - Yongjia Yu
- Department of Radiation Oncology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA;
| | - Robert L. Ullrich
- The Radiation Effects Research Foundation (RERF), Hiroshima 732-0815, Japan;
| | - Mark R. Emmett
- Department of Radiation Oncology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA;
- Department of Pathology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA
- Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, TX 77555, USA
- Correspondence: ; Tel.: +1-(409)-747-1943
| |
Collapse
|
26
|
Gómez X, Sanon S, Zambrano K, Asquel S, Bassantes M, Morales JE, Otáñez G, Pomaquero C, Villarroel S, Zurita A, Calvache C, Celi K, Contreras T, Corrales D, Naciph MB, Peña J, Caicedo A. Key points for the development of antioxidant cocktails to prevent cellular stress and damage caused by reactive oxygen species (ROS) during manned space missions. NPJ Microgravity 2021; 7:35. [PMID: 34556658 PMCID: PMC8460669 DOI: 10.1038/s41526-021-00162-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 08/03/2021] [Indexed: 02/08/2023] Open
Abstract
Exposure to microgravity and ionizing radiation during spaceflight missions causes excessive reactive oxygen species (ROS) production that contributes to cellular stress and damage in astronauts. Average spaceflight mission time is expected to lengthen as humanity aims to visit other planets. However, longer missions or spaceflights will undoubtedly lead to an increment in microgravity, ionizing radiation and ROS production. Strategies to minimize ROS damage are necessary to maintain the health of astronauts, future space colonists, and tourists during and after spaceflight missions. An antioxidant cocktail formulated to prevent or mitigate ROS damage during space exploration could help maintain the health of space explorers. We propose key points to consider when developing an antioxidant cocktail. We discuss how ROS damages our body and organs, the genetic predisposition of astronauts to its damage, characteristics and evidence of the effectiveness of antioxidants to combat excess ROS, differences in drug metabolism when on Earth and in space that could modify antioxidant effects, and the characteristics and efficacy of common antioxidants. Based on this information we propose a workflow for assessing astronaut resistance to ROS damage, infight monitoring of ROS production, and an antioxidant cocktail. Developing an antioxidant cocktail represents a big challenge to translate current medical practices from an Earth setting to space. The key points presented in this review could promote the development of different antioxidant formulations to maintain space explorers' health in the future.
Collapse
Affiliation(s)
- Xavier Gómez
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
- Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador
- Mito-Act Research Consortium, Quito, Ecuador
| | - Serena Sanon
- Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador
- Cornell University, Ithaca, NY, USA
- Mito-Act Research Consortium, Quito, Ecuador
| | - Kevin Zambrano
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
- Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador
- Mito-Act Research Consortium, Quito, Ecuador
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Samira Asquel
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Mariuxi Bassantes
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Julián E Morales
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Gabriela Otáñez
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Core Pomaquero
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Sarah Villarroel
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Alejandro Zurita
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Carlos Calvache
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Kathlyn Celi
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Terry Contreras
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Dylan Corrales
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - María Belén Naciph
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - José Peña
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador
| | - Andrés Caicedo
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador.
- Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador.
- Mito-Act Research Consortium, Quito, Ecuador.
- Sistemas Médicos SIME, Universidad San Francisco de Quito USFQ, Quito, Ecuador.
| |
Collapse
|
27
|
Cahill T, Cope H, Bass JJ, Overbey EG, Gilbert R, da Silveira WA, Paul AM, Mishra T, Herranz R, Reinsch SS, Costes SV, Hardiman G, Szewczyk NJ, Tahimic CGT. Mammalian and Invertebrate Models as Complementary Tools for Gaining Mechanistic Insight on Muscle Responses to Spaceflight. Int J Mol Sci 2021; 22:ijms22179470. [PMID: 34502375 PMCID: PMC8430797 DOI: 10.3390/ijms22179470] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
Bioinformatics approaches have proven useful in understanding biological responses to spaceflight. Spaceflight experiments remain resource intensive and rare. One outstanding issue is how to maximize scientific output from a limited number of omics datasets from traditional animal models including nematodes, fruitfly, and rodents. The utility of omics data from invertebrate models in anticipating mammalian responses to spaceflight has not been fully explored. Hence, we performed comparative analyses of transcriptomes of soleus and extensor digitorum longus (EDL) in mice that underwent 37 days of spaceflight. Results indicate shared stress responses and altered circadian rhythm. EDL showed more robust growth signals and Pde2a downregulation, possibly underlying its resistance to atrophy versus soleus. Spaceflight and hindlimb unloading mice shared differential regulation of proliferation, circadian, and neuronal signaling. Shared gene regulation in muscles of humans on bedrest and space flown rodents suggest targets for mitigating muscle atrophy in space and on Earth. Spaceflight responses of C. elegans were more similar to EDL. Discrete life stages of D. melanogaster have distinct utility in anticipating EDL and soleus responses. In summary, spaceflight leads to shared and discrete molecular responses between muscle types and invertebrate models may augment mechanistic knowledge gained from rodent spaceflight and ground-based studies.
Collapse
Affiliation(s)
- Thomas Cahill
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.); (G.H.)
| | - Henry Cope
- Nottingham Biomedical Research Centre (BRC), School of Computer Science, University of Nottingham, Nottingham NG7 2QL, UK;
| | - Joseph J. Bass
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), University of Nottingham, Nottingham NG7 2QL, UK; (J.J.B.); (N.J.S.)
| | - Eliah G. Overbey
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA;
| | - Rachel Gilbert
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA; (R.G.); (A.M.P.); (S.S.R.); (S.V.C.)
- Universities Space Research Association, Columbia, MD 21046, USA
| | - Willian Abraham da Silveira
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.); (G.H.)
- Department of Biological Sciences, School of Life Sciences and Education, Staffordshire University, Stoke-on-Trent ST4 2DF, UK
| | - Amber M. Paul
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA; (R.G.); (A.M.P.); (S.S.R.); (S.V.C.)
- Department of Human Factors and Behavioral Neurobiology, Embry-Riddle Aeronautical University, Daytona Beach, FL 32114, USA
- Blue Marble Space Institute of Science, Seattle, WA 98104, USA
| | - Tejaswini Mishra
- Department of Genetics, Stanford University School of Medicine, Palo Alto, CA 94305, USA;
| | - Raúl Herranz
- Centro de Investigaciones Biológicas Margarita Salas–CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain;
| | - Sigrid S. Reinsch
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA; (R.G.); (A.M.P.); (S.S.R.); (S.V.C.)
| | - Sylvain V. Costes
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA; (R.G.); (A.M.P.); (S.S.R.); (S.V.C.)
| | - Gary Hardiman
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.); (G.H.)
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Nathaniel J. Szewczyk
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), University of Nottingham, Nottingham NG7 2QL, UK; (J.J.B.); (N.J.S.)
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Candice G. T. Tahimic
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA; (R.G.); (A.M.P.); (S.S.R.); (S.V.C.)
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
- Correspondence:
| |
Collapse
|
28
|
Laiakis EC, Shuryak I, Deziel A, Wang YW, Barnette BL, Yu Y, Ullrich RL, Fornace AJ, Emmett MR. Effects of Low Dose Space Radiation Exposures on the Splenic Metabolome. Int J Mol Sci 2021; 22:3070. [PMID: 33802822 PMCID: PMC8002539 DOI: 10.3390/ijms22063070] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Future space missions will include a return to the Moon and long duration deep space roundtrip missions to Mars. Leaving the protection that Low Earth Orbit provides will unavoidably expose astronauts to higher cumulative doses of space radiation, in addition to other stressors, e.g., microgravity. Immune regulation is known to be impacted by both radiation and spaceflight and it remains to be seen whether prolonged effects that will be encountered in deep space can have an adverse impact on health. In this study, we investigated the effects in the overall metabolism of three different low dose radiation exposures (γ-rays, 16O, and 56Fe) in spleens from male C57BL/6 mice at 1, 2, and 4 months after exposure. Forty metabolites were identified with significant enrichment in purine metabolism, tricarboxylic acid cycle, fatty acids, acylcarnitines, and amino acids. Early perturbations were more prominent in the γ irradiated samples, while later responses shifted towards more prominent responses in groups with high energy particle irradiations. Regression analysis showed a positive correlation of the abundance of identified fatty acids with time and a negative association with γ-rays, while the degradation pathway of purines was positively associated with time. Taken together, there is a strong suggestion of mitochondrial implication and the possibility of long-term effects on DNA repair and nucleotide pools following radiation exposure.
Collapse
Affiliation(s)
- Evagelia C. Laiakis
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | - Igor Shuryak
- Center for Radiological Research, Columbia University, New York, NY 10032, USA;
| | - Annabella Deziel
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
| | - Yi-Wen Wang
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
| | - Brooke L. Barnette
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.L.B.); (Y.Y.); (M.R.E.)
| | - Yongjia Yu
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.L.B.); (Y.Y.); (M.R.E.)
| | | | - Albert J. Fornace
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | - Mark R. Emmett
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.L.B.); (Y.Y.); (M.R.E.)
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
29
|
Afshinnekoo E, Scott RT, MacKay MJ, Pariset E, Cekanaviciute E, Barker R, Gilroy S, Hassane D, Smith SM, Zwart SR, Nelman-Gonzalez M, Crucian BE, Ponomarev SA, Orlov OI, Shiba D, Muratani M, Yamamoto M, Richards SE, Vaishampayan PA, Meydan C, Foox J, Myrrhe J, Istasse E, Singh N, Venkateswaran K, Keune JA, Ray HE, Basner M, Miller J, Vitaterna MH, Taylor DM, Wallace D, Rubins K, Bailey SM, Grabham P, Costes SV, Mason CE, Beheshti A. Fundamental Biological Features of Spaceflight: Advancing the Field to Enable Deep-Space Exploration. Cell 2020; 183:1162-1184. [PMID: 33242416 PMCID: PMC8441988 DOI: 10.1016/j.cell.2020.10.050] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022]
Abstract
Research on astronaut health and model organisms have revealed six features of spaceflight biology that guide our current understanding of fundamental molecular changes that occur during space travel. The features include oxidative stress, DNA damage, mitochondrial dysregulation, epigenetic changes (including gene regulation), telomere length alterations, and microbiome shifts. Here we review the known hazards of human spaceflight, how spaceflight affects living systems through these six fundamental features, and the associated health risks of space exploration. We also discuss the essential issues related to the health and safety of astronauts involved in future missions, especially planned long-duration and Martian missions.
Collapse
Affiliation(s)
- Ebrahim Afshinnekoo
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ryan T Scott
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Matthew J MacKay
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Eloise Pariset
- Universities Space Research Association (USRA), Mountain View, CA 94043, USA; Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Egle Cekanaviciute
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Richard Barker
- Department of Botany, University of Wisconsin, Madison, WI 53706, USA
| | - Simon Gilroy
- Department of Botany, University of Wisconsin, Madison, WI 53706, USA
| | | | - Scott M Smith
- Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Sara R Zwart
- Department of Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mayra Nelman-Gonzalez
- KBR, Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Brian E Crucian
- Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Sergey A Ponomarev
- Institute for the Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia
| | - Oleg I Orlov
- Institute for the Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia
| | - Dai Shiba
- JEM Utilization Center, Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency (JAXA), Ibaraki 305-8505, Japan
| | - Masafumi Muratani
- Transborder Medical Research Center, and Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan; Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Stephanie E Richards
- Bionetics, NASA Kennedy Space Center, Kennedy Space Center, Merritt Island, FL 32899, USA
| | - Parag A Vaishampayan
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jonathan Foox
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jacqueline Myrrhe
- European Space Agency, Research and Payloads Group, Data Exploitation and Utilisation Strategy Office, 2200 AG Noordwijk, the Netherlands
| | - Eric Istasse
- European Space Agency, Research and Payloads Group, Data Exploitation and Utilisation Strategy Office, 2200 AG Noordwijk, the Netherlands
| | - Nitin Singh
- Biotechnology and Planetary Protection Group, Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA 91109, USA
| | - Kasthuri Venkateswaran
- Biotechnology and Planetary Protection Group, Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA 91109, USA
| | - Jessica A Keune
- Space Medicine Operations Division, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Hami E Ray
- ASRC Federal Space and Defense, Inc., Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Mathias Basner
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jack Miller
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Martha Hotz Vitaterna
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL 60208, USA; Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Deanne M Taylor
- Department of Biomedical Informatics, The Children's Hospital of Philadelphia, PA 19104, USA; Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; The Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; The Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathleen Rubins
- Astronaut Office, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Susan M Bailey
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| | - Peter Grabham
- Center for Radiological Research, Department of Oncology, College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA.
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA.
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA; The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, NY 10021, USA.
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
30
|
Lai Polo SH, Saravia-Butler AM, Boyko V, Dinh MT, Chen YC, Fogle H, Reinsch SS, Ray S, Chakravarty K, Marcu O, Chen RB, Costes SV, Galazka JM. RNAseq Analysis of Rodent Spaceflight Experiments Is Confounded by Sample Collection Techniques. iScience 2020; 23:101733. [PMID: 33376967 PMCID: PMC7756143 DOI: 10.1016/j.isci.2020.101733] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/04/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
To understand the physiological changes that occur in response to spaceflight, mice are transported to the International Space Station (ISS) and housed for variable periods of time before euthanasia on-orbit or return to Earth. Sample collection under such difficult conditions introduces confounding factors that need to be identified and addressed. We found large changes in the transcriptome of mouse tissues dissected and preserved on-orbit compared with tissues from mice euthanized on-orbit, preserved, and dissected after return to Earth. Changes due to preservation method eclipsed those between flight and ground samples, making it difficult to identify spaceflight-specific changes. Follow-on experiments to interrogate the roles of euthanasia methods, tissue and carcass preservation protocols, and library preparation methods suggested that differences due to preservation protocols are exacerbated when coupled with polyA selection. This has important implications for the interpretation of existing datasets and the design of future experiments. Experimentation is necessary to understand how organisms respond to space Specialized protocols are used for preserving biological samples on the ISS RNAseq datasets are impacted by preservation protocols used on the ISS Impacts can be alleviated with improved carcass preservation protocols
Collapse
Affiliation(s)
- San-Huei Lai Polo
- KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA.,NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Amanda M Saravia-Butler
- NASA Ames Research Center, Moffett Field, CA 94035, USA.,Logyx, LLC, Mountain View, CA 94043, USA
| | - Valery Boyko
- NASA Ames Research Center, Moffett Field, CA 94035, USA.,The Bionetics Corporation, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Marie T Dinh
- NASA Ames Research Center, Moffett Field, CA 94035, USA.,Logyx, LLC, Mountain View, CA 94043, USA
| | - Yi-Chun Chen
- KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA.,NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Homer Fogle
- NASA Ames Research Center, Moffett Field, CA 94035, USA.,The Bionetics Corporation, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | | | - Shayoni Ray
- NGM Biopharmaceuticals, South San Francisco, CA 94080, USA
| | | | - Oana Marcu
- Carl Sagan Center, SETI Institute, Mountain View, CA 94043, USA
| | - Rick B Chen
- KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA.,NASA Ames Research Center, Moffett Field, CA 94035, USA
| | | | | |
Collapse
|
31
|
Yang JQ, Jiang N, Li ZP, Guo S, Chen ZY, Li BB, Chai SB, Lu SY, Yan HF, Sun PM, Zhang T, Sun HW, Yang JW, Zhou JL, Yang HM, Cui Y. The effects of microgravity on the digestive system and the new insights it brings to the life sciences. LIFE SCIENCES IN SPACE RESEARCH 2020; 27:74-82. [PMID: 34756233 DOI: 10.1016/j.lssr.2020.07.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/04/2020] [Accepted: 07/28/2020] [Indexed: 06/13/2023]
Abstract
BACKGROUND Weightlessness is a component of the complex space environment. It exerts adverse effects on the human body, and may pose unknown challenges to the implementation of space missions. The regular function of the digestive system is an important checkpoint for astronauts to conduct missions. Simulated microgravity can recreate the changes experienced by the human body in a weightless environment in space to a certain extent, providing technical support for the exploration of its mechanism and a practical method for other scientific research. METHODS AND MATERIALS In the present study, we reviewed and discussed the latest research on the effects of weightlessness or simulated microgravity on the digestive system, as well as the current challenges and future expectations for progress in medical science and further space exploration. RESULTS A series of studies have investigated the effects of weightlessness on the human digestive system. On one hand, weightlessness and the changing space environment may exert certain adverse effects on the human body. Studies based on cells or animals have demonstrated the complex effects on the human digestive system in response to weightlessness. On the other hand, a microgravity environment also facilitates the ideation of novel concepts for research in the domain of life science. CONCLUSION The effects of weightlessness on the digestive system are considerably complicated. The emergence of methods that help simulate a weightless environment provides a more convenient alternative for assessing the impact and the mechanism underlying the effect of weightlessness on the human body. In addition, the simulated microgravity environment facilitates the ideation of novel concepts for application in regenerative medicine and other fields of life science.
Collapse
Affiliation(s)
- Jia-Qi Yang
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China; Department of General Surgery, the 306th Hospital of Chinese PLA-Peking University Teaching Hospital, Beijing 100101, China
| | - Nan Jiang
- The Center for Hepatopancreatobiliary Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Zheng-Peng Li
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China
| | - Song Guo
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China; Department of General Surgery, the 306th Hospital of Chinese PLA-Peking University Teaching Hospital, Beijing 100101, China
| | - Zheng-Yang Chen
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China; Department of General Surgery, the 306th Hospital of Chinese PLA-Peking University Teaching Hospital, Beijing 100101, China
| | - Bin-Bin Li
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China
| | - Shao-Bin Chai
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China
| | - Sheng-Yu Lu
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China; Department of General Surgery, the 306th Hospital of Chinese PLA-Peking University Teaching Hospital, Beijing 100101, China
| | - Hong-Feng Yan
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China
| | - Pei-Ming Sun
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China
| | - Tao Zhang
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China
| | - Hong-Wei Sun
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China
| | - Jian-Wu Yang
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China
| | - Jin-Lian Zhou
- Department of Pathology, the Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China.
| | - He-Ming Yang
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China
| | - Yan Cui
- Department of General Surgery, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China.
| |
Collapse
|
32
|
Liu H, Guo J, Li Y, Zhang Y, Wang J, Gao J, Deng Y, Li Y. Investigation on Intestinal Proteins and Drug Metabolizing Enzymes in Simulated Microgravity Rats by a Proteomics Method. Molecules 2020; 25:E4391. [PMID: 32987831 PMCID: PMC7582489 DOI: 10.3390/molecules25194391] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/28/2022] Open
Abstract
The present study aimed to investigate the change of intestinal mucosa proteins, especially the alteration of intestinal drug metabolizing enzymes (IDMEs) following 14-day simulated microgravity. Morey-Holton tail-suspension analog was used to simulate microgravity. Intestinal mucosa proteins of rats were determined by label-free quantitative proteomic strategy. A total of 335 differentially expressed proteins (DEPs) were identified, 190 DEPs were upregulated, and 145 DEPs were downregulated. According to bioinformatic analysis, most of DEPs exhibited hydrolase, oxidoreductase, transferase, ligase, or lyase catalytic activity. DEPs were mainly enriched in metabolic pathways, including metabolism of amino acid, glucose, and carbon. Moreover, 11 of DEPs were involved in exogenous drug and xenobiotics metabolism. Owing to the importance of IDMEs for the efficacy and safety of oral drugs, the expression of cytochrome P450 1A2 (CYP1A2), CYP2D1, CYP3A2, CYP2E1, alcohol dehydrogenase 1 (ADH1), and glutathione S-transferase mu 5 (GSTM5) in rat intestine mucosa was determined by Western-blot. The activity of ADH, aldehyde dehydrogenase (ALDH) and GST was evaluated. Compared with control rats, the expression of CYP1A2, CYP2D1, CYP3A2, and ADH1 in the simulated microgravity (SMG) group of rats were dramatically decreased by 33.16%, 21.93%, 48.49%, and 22.83%, respectively. GSTM5 was significantly upregulated by 53.14% and CYP2E1 expression did not show a dramatical change in SMG group rats. Moreover, 14-day SMG reduced ADH activity, while ALDH and GST activities was not altered remarkably. It could be concluded that SMG dramatically affected the expression and activity of some IDMEs, which might alter the efficacy or safety of their substrate drugs under microgravity. The present study provided some preliminary information on IDMEs under microgravity. It revealed the potential effect of SMG on intestinal metabolism, which may be helpful to understand the intestinal health of astronauts and medication use.
Collapse
Affiliation(s)
- Huayan Liu
- School of Life Science, Beijing Institute of Technology, No.5 Zhongguancun South Street, Haidian District, Beijing 100081, China; (H.L.); (J.G.); (Y.D.)
| | - Jingjing Guo
- School of Life Science, Beijing Institute of Technology, No.5 Zhongguancun South Street, Haidian District, Beijing 100081, China; (H.L.); (J.G.); (Y.D.)
| | - Yujuan Li
- School of Life Science, Beijing Institute of Technology, No.5 Zhongguancun South Street, Haidian District, Beijing 100081, China; (H.L.); (J.G.); (Y.D.)
| | - Yushi Zhang
- Institute of Chinese Materia Medica, No.16 Dongzhimen Neinan Street, Dongcheng District, Beijing 100081, China;
| | - Jiaping Wang
- Astronaut Research and Training Center of China, No.109 Youyi Road, Haidian District, Beijing 100094, China; (J.W.); (J.G.)
| | - Jianyi Gao
- Astronaut Research and Training Center of China, No.109 Youyi Road, Haidian District, Beijing 100094, China; (J.W.); (J.G.)
| | - Yulin Deng
- School of Life Science, Beijing Institute of Technology, No.5 Zhongguancun South Street, Haidian District, Beijing 100081, China; (H.L.); (J.G.); (Y.D.)
| | - Yongzhi Li
- Astronaut Research and Training Center of China, No.109 Youyi Road, Haidian District, Beijing 100094, China; (J.W.); (J.G.)
| |
Collapse
|
33
|
Affiliation(s)
- T. Cahill
- Faculty of Medicine, Health and Life Sciences School of Biological Sciences Institute for Global Food Security (IGFS) Belfast UK
| | - G. Hardiman
- Faculty of Medicine, Health and Life Sciences School of Biological Sciences Institute for Global Food Security (IGFS) Belfast UK
- Department of Medicine Medical University of South Carolina Charleston SC USA
| |
Collapse
|
34
|
Takahashi S, Luo Y, Ranjit S, Xie C, Libby AE, Orlicky DJ, Dvornikov A, Wang XX, Myakala K, Jones BA, Bhasin K, Wang D, McManaman JL, Krausz KW, Gratton E, Ir D, Robertson CE, Frank DN, Gonzalez FJ, Levi M. Bile acid sequestration reverses liver injury and prevents progression of nonalcoholic steatohepatitis in Western diet-fed mice. J Biol Chem 2020; 295:4733-4747. [PMID: 32075905 DOI: 10.1074/jbc.ra119.011913] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease is a rapidly rising problem in the 21st century and is a leading cause of chronic liver disease that can lead to end-stage liver diseases, including cirrhosis and hepatocellular cancer. Despite this rising epidemic, no pharmacological treatment has yet been established to treat this disease. The rapidly increasing prevalence of nonalcoholic fatty liver disease and its aggressive form, nonalcoholic steatohepatitis (NASH), requires novel therapeutic approaches to prevent disease progression. Alterations in microbiome dynamics and dysbiosis play an important role in liver disease and may represent targetable pathways to treat liver disorders. Improving microbiome properties or restoring normal bile acid metabolism may prevent or slow the progression of liver diseases such as NASH. Importantly, aberrant systemic circulation of bile acids can greatly disrupt metabolic homeostasis. Bile acid sequestrants are orally administered polymers that bind bile acids in the intestine, forming nonabsorbable complexes. Bile acid sequestrants interrupt intestinal reabsorption of bile acids, decreasing their circulating levels. We determined that treatment with the bile acid sequestrant sevelamer reversed the liver injury and prevented the progression of NASH, including steatosis, inflammation, and fibrosis in a Western diet-induced NASH mouse model. Metabolomics and microbiome analysis revealed that this beneficial effect is associated with changes in the microbiota population and bile acid composition, including reversing microbiota complexity in cecum by increasing Lactobacillus and decreased Desulfovibrio The net effect of these changes was improvement in liver function and markers of liver injury and the positive effects of reversal of insulin resistance.
Collapse
Affiliation(s)
- Shogo Takahashi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, D.C., 20057.,National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Yuhuan Luo
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Suman Ranjit
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, D.C., 20057.,Department of Biomedical Engineering, Laboratory for Fluorescence Dynamics, University of California at Irvine, Irvine, California 92697
| | - Cen Xie
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Andrew E Libby
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, D.C., 20057
| | - David J Orlicky
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Alexander Dvornikov
- Department of Biomedical Engineering, Laboratory for Fluorescence Dynamics, University of California at Irvine, Irvine, California 92697
| | - Xiaoxin X Wang
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, D.C., 20057
| | - Komuraiah Myakala
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, D.C., 20057
| | - Bryce A Jones
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, D.C., 20057.,Department of Pharmacology and Physiology, Georgetown University, Washington, D.C., 20057
| | - Kanchan Bhasin
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, D.C., 20057
| | - Dong Wang
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - James L McManaman
- Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045.,Graduate Program in Integrated Physiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Kristopher W Krausz
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Enrico Gratton
- Department of Biomedical Engineering, Laboratory for Fluorescence Dynamics, University of California at Irvine, Irvine, California 92697
| | - Diana Ir
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Charles E Robertson
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Daniel N Frank
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Frank J Gonzalez
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, D.C., 20057
| |
Collapse
|
35
|
Validation of a New Rodent Experimental System to Investigate Consequences of Long Duration Space Habitation. Sci Rep 2020; 10:2336. [PMID: 32047211 PMCID: PMC7012842 DOI: 10.1038/s41598-020-58898-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 01/22/2020] [Indexed: 12/18/2022] Open
Abstract
Animal models are useful for exploring the health consequences of prolonged spaceflight. Capabilities were developed to perform experiments in low earth orbit with on-board sample recovery, thereby avoiding complications caused by return to Earth. For NASA’s Rodent Research-1 mission, female mice (ten 32 wk C57BL/6NTac; ten 16 wk C57BL/6J) were launched on an unmanned vehicle, then resided on the International Space Station for 21/22d or 37d in microgravity. Mice were euthanized on-orbit, livers and spleens dissected, and remaining tissues frozen in situ for later analyses. Mice appeared healthy by daily video health checks and body, adrenal, and spleen weights of 37d-flight (FLT) mice did not differ from ground controls housed in flight hardware (GC), while thymus weights were 35% greater in FLT than GC. Mice exposed to 37d of spaceflight displayed elevated liver mass (33%) and select enzyme activities compared to GC, whereas 21/22d-FLT mice did not. FLT mice appeared more physically active than respective GC while soleus muscle showed expected atrophy. RNA and enzyme activity levels in tissues recovered on-orbit were of acceptable quality. Thus, this system establishes a new capability for conducting long-duration experiments in space, enables sample recovery on-orbit, and avoids triggering standard indices of chronic stress.
Collapse
|
36
|
McDonald JT, Stainforth R, Miller J, Cahill T, da Silveira WA, Rathi KS, Hardiman G, Taylor D, Costes SV, Chauhan V, Meller R, Beheshti A. NASA GeneLab Platform Utilized for Biological Response to Space Radiation in Animal Models. Cancers (Basel) 2020; 12:E381. [PMID: 32045996 PMCID: PMC7072278 DOI: 10.3390/cancers12020381] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Ionizing radiation from galactic cosmic rays (GCR) is one of the major risk factors that will impact the health of astronauts on extended missions outside the protective effects of the Earth's magnetic field. The NASA GeneLab project has detailed information on radiation exposure using animal models with curated dosimetry information for spaceflight experiments. Methods: We analyzed multiple GeneLab omics datasets associated with both ground-based and spaceflight radiation studies that included in vivo and in vitro approaches. A range of ions from protons to iron particles with doses from 0.1 to 1.0 Gy for ground studies, as well as samples flown in low Earth orbit (LEO) with total doses of 1.0 mGy to 30 mGy, were utilized. Results: From this analysis, we were able to identify distinct biological signatures associating specific ions with specific biological responses due to radiation exposure in space. For example, we discovered changes in mitochondrial function, ribosomal assembly, and immune pathways as a function of dose. Conclusions: We provided a summary of how the GeneLab's rich database of omics experiments with animal models can be used to generate novel hypotheses to better understand human health risks from GCR exposures.
Collapse
Affiliation(s)
| | - Robert Stainforth
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, ON K1A-1C1, Canada; (R.S.); (V.C.)
| | - Jack Miller
- KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA;
| | - Thomas Cahill
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.)
| | - Willian A. da Silveira
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.)
| | - Komal S. Rathi
- Department of Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Gary Hardiman
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.)
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Deanne Taylor
- Department of Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- The Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- The Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sylvain V. Costes
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA;
| | - Vinita Chauhan
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, ON K1A-1C1, Canada; (R.S.); (V.C.)
| | - Robert Meller
- Department of Neurobiology and Pharmacology, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Afshin Beheshti
- KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA;
| |
Collapse
|
37
|
Fu H, Su F, Zhu J, Zheng X, Ge C. Effect of simulated microgravity and ionizing radiation on expression profiles of miRNA, lncRNA, and mRNA in human lymphoblastoid cells. LIFE SCIENCES IN SPACE RESEARCH 2020; 24:1-8. [PMID: 31987473 DOI: 10.1016/j.lssr.2019.10.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/26/2019] [Accepted: 10/19/2019] [Indexed: 06/10/2023]
Abstract
In space, multiple unique environmental factors, particularly microgravity and space radiation, pose a constant threat to astronaut health. MicroRNAs (miRNAs) and long noncoding RNAs (lncRNAs) are functional RNAs that play critical roles in regulating multiple cellular processes. To gain insight into the role of non-coding RNAs in response to radiation and microgravity, we analyzed RNA expression profiles in human lymphoblastoid TK6 cells incubated for 24 h under static or rotating conditions to stimulate microgravity in space, after 2-Gy γ-ray irradiation. The expression of 14 lncRNAs and 17 mRNAs (differentially-expressed genes, DEGs) was found to be significantly downregulated under simulated microgravity conditions. In contrast, irradiation upregulated 55 lncRNAs and 56 DEGs, whereas only one lncRNA, but no DEGs, was downregulated. Furthermore, two miRNAs, 70 lncRNAs, and 87 DEGs showed significantly altered expression in response to simulated microgravity after irradiation, and these changes were independently induced by irradiation and simulated microgravity. GO enrichment and KEGG pathway analyses indicated that the associated target genes showed similar patterns to the noncoding RNAs and were suggested to be involved in the immune/inflammatory response including LPS/TLR, TNF, and NF-κB signaling pathways. However, synergistic effects on RNA expression and cellular responses were also observed with a combination of simulated microgravity and irradiation based on microarray and RT-PCR analysis. Together, our results indicate that simulated microgravity and irradiation additively alter expression patterns but synergistically modulate the expression levels of RNAs and their target genes in human lymphoblastoid cells.
Collapse
Affiliation(s)
- Hanjiang Fu
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, #27 Taiping Rd. Haidian Dist., Beijing 100850, China
| | - Fei Su
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, #27 Taiping Rd. Haidian Dist., Beijing 100850, China
| | - Jie Zhu
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, #27 Taiping Rd. Haidian Dist., Beijing 100850, China
| | - Xiaofei Zheng
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, #27 Taiping Rd. Haidian Dist., Beijing 100850, China.
| | - Changhui Ge
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, #27 Taiping Rd. Haidian Dist., Beijing 100850, China.
| |
Collapse
|
38
|
Multi-omics analysis of multiple missions to space reveal a theme of lipid dysregulation in mouse liver. Sci Rep 2019; 9:19195. [PMID: 31844325 PMCID: PMC6915713 DOI: 10.1038/s41598-019-55869-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/30/2019] [Indexed: 12/13/2022] Open
Abstract
Spaceflight has several detrimental effects on the physiology of astronauts, many of which are recapitulated in rodent models. Mouse studies performed on the Space Shuttle showed disruption of lipid metabolism in liver. However, given that these animals were not sacrificed on-orbit and instead returned live to earth, it is unclear if these disruptions were solely induced by space stressors (e.g. microgravity, space radiation) or in part explained by the stress of return to Earth. In this work we analyzed three liver datasets from two different strains of mice (C57BL/6 (Jackson) & BALB/c (Taconic)) flown aboard the International Space Station (ISS). Notably, these animals were sacrificed on-orbit and exposed to varying spaceflight durations (i.e. 21, 37, and 42 days vs 13 days for the Shuttle mice). Oil Red O (ORO) staining showed abnormal lipid accumulation in all space-flown mice compared to ground controls regardless of strain or exposure duration. Similarly, transcriptomic analysis by RNA-sequencing revealed several pathways that were affected in both strains related to increased lipid metabolism, fatty acid metabolism, lipid and fatty acid processing, lipid catabolic processing, and lipid localization. In addition, key upstream regulators were predicted to be commonly regulated across all conditions including Glucagon (GCG) and Insulin (INS). Moreover, quantitative proteomic analysis showed that a number of lipid related proteins were changed in the livers during spaceflight. Taken together, these data indicate that activation of lipotoxic pathways are the result of space stressors alone and this activation occurs in various genetic backgrounds during spaceflight exposures of weeks to months. If similar responses occur in humans, a prolonged change of these pathways may result in the development of liver disease and should be investigated further.
Collapse
|
39
|
Li BB, Chen ZY, Guo S, Sun HW, Cui Y. Progress in research of digestive system trauma and stress injury under microgravity environment. Shijie Huaren Xiaohua Zazhi 2019; 27:1088-1094. [DOI: 10.11569/wcjd.v27.i17.1088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The last two decades have witnessed the rapid develop-ment of China's manned spaceflight industry. Studies have showed that the weightlessness environment has a series of adverse effects on the human body. Due to the complexity of the structure and function of the digestive system, the impact of weightlessness on the digestive system has certain particularity. How to ensure the steady state of the digestive system during astronaut's space mission and in the training under simulated weightlessness needs to be studied urgently. This review focuses on the progress in the research of digestive system trauma, stress injury, and repair under microgravity environment.
Collapse
Affiliation(s)
- Bin-Bin Li
- Department of General Surgery, The PLA 306 Teaching Hospital of Anhui Medical University, Chaoyang District, Beijing 100101, China
| | - Zheng-Yang Chen
- Department of General Surgery, The 306 Hospital of PLA-Peking University Teaching Hospital, Chaoyang District, Beijing 100101, China
| | - Song Guo
- Department of General Surgery, The 306 Hospital of PLA-Peking University Teaching Hospital, Chaoyang District, Beijing 100101, China
| | - Hong-Wei Sun
- Department of General Surgery, The 306 Hospital of PLA, Chaoyang District, Beijing 100101, China
| | - Yan Cui
- Department of General Surgery, The 306 Hospital of PLA, Chaoyang District, Beijing 100101, China
| |
Collapse
|
40
|
Medications in Space: In Search of a Pharmacologist's Guide to the Galaxy. Pharm Res 2019; 36:148. [PMID: 31414302 DOI: 10.1007/s11095-019-2679-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/30/2019] [Indexed: 12/23/2022]
Abstract
Medications have been used during space missions for more than half a century, yet our understanding of the effects of spaceflight on drug pharmacokinetics and pharmacodynamics is poor. The space environment induces time-dependent alterations in human physiology that include fluid shifts, cardiovascular deconditioning, bone and muscle density loss, and impaired immunity. This review presents the current knowledge on the physiological effects of spaceflight that can translate into altered drug disposition and activity and eventually to inadequate treatment. It describes findings from studies in astronauts along with mechanistic studies in animal models and in vitro systems. Future missions into deeper space and the emergence of commercial spaceflight will require a more detailed understanding of space pharmacology to optimize treatment in astronauts and space travelers.
Collapse
|
41
|
Wang Y, Zhao W, Shi J, Wang J, Hao J, Pang X, Huang X, Chen X, Li Y, Jin R, Ge Q. Intestinal microbiota contributes to altered glucose metabolism in simulated microgravity mouse model. FASEB J 2019; 33:10140-10151. [DOI: 10.1096/fj.201900238rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Yifan Wang
- Department of ImmunologySchool of Basic Medical SciencesNHC Key Laboratory of Medical ImmunologyPeking University Beijing China
| | - Weijia Zhao
- Department of ImmunologySchool of Basic Medical SciencesNHC Key Laboratory of Medical ImmunologyPeking University Beijing China
| | - Junxiu Shi
- Department of Developmental Cell BiologyKey Laboratory of Cell BiologyMinistry of Public HealthKey Laboratory of Medical Cell BiologyMinistry of EducationChina Medical University Shenyang China
| | - Jiachi Wang
- Department of Developmental Cell BiologyKey Laboratory of Cell BiologyMinistry of Public HealthKey Laboratory of Medical Cell BiologyMinistry of EducationChina Medical University Shenyang China
| | - Jie Hao
- Department of ImmunologySchool of Basic Medical SciencesNHC Key Laboratory of Medical ImmunologyPeking University Beijing China
| | - Xuewen Pang
- Department of ImmunologySchool of Basic Medical SciencesNHC Key Laboratory of Medical ImmunologyPeking University Beijing China
| | - Xiaojun Huang
- Beijing Key Laboratory of Hematopoietic Stem Cell TransplantationPeking University People's HospitalInstitute of Hematology Beijing China
| | - Xiaoping Chen
- State Key Laboratory of Space Medicine Fundamentals and ApplicationChinese Astronaut Research and Training Center Beijing China
| | - Yongzhi Li
- State Key Laboratory of Space Medicine Fundamentals and ApplicationChinese Astronaut Research and Training Center Beijing China
| | - Rong Jin
- Department of ImmunologySchool of Basic Medical SciencesNHC Key Laboratory of Medical ImmunologyPeking University Beijing China
| | - Qing Ge
- Department of ImmunologySchool of Basic Medical SciencesNHC Key Laboratory of Medical ImmunologyPeking University Beijing China
- Department of Integration of Chinese and Western MedicineSchool of Basic Medical SciencesPeking University Beijing China
| |
Collapse
|
42
|
Mao XW, Sandberg LB, Gridley DS, Herrmann EC, Zhang G, Raghavan R, Zubarev RA, Zhang B, Stodieck LS, Ferguson VL, Bateman TA, Pecaut MJ. Proteomic Analysis of Mouse Brain Subjected to Spaceflight. Int J Mol Sci 2018; 20:ijms20010007. [PMID: 30577490 PMCID: PMC6337482 DOI: 10.3390/ijms20010007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/11/2018] [Accepted: 12/17/2018] [Indexed: 01/01/2023] Open
Abstract
There is evidence that spaceflight poses acute and late risks to the central nervous system. To explore possible mechanisms, the proteomic changes following spaceflight in mouse brain were characterized. Space Shuttle Atlantis (STS-135) was launched from the Kennedy Space Center (KSC) on a 13-day mission. Within 3–5 h after landing, brain tissue was collected to evaluate protein expression profiles using quantitative proteomic analysis. Our results showed that there were 26 proteins that were significantly altered after spaceflight in the gray and/or white matter. While there was no overlap between the white and gray matter in terms of individual proteins, there was overlap in terms of function, synaptic plasticity, vesical activity, protein/organelle transport, and metabolism. Our data demonstrate that exposure to the spaceflight environment induces significant changes in protein expression related to neuronal structure and metabolic function. This might lead to a significant impact on brain structural and functional integrity that could affect the outcome of space missions.
Collapse
Affiliation(s)
- Xiao Wen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| | - Lawrence B Sandberg
- Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Daila S Gridley
- Department of Basic Sciences, Division of Biomedical Engineering Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| | - E Clifford Herrmann
- Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Guangyu Zhang
- Department of Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Ravi Raghavan
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Roman A Zubarev
- Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, SE 17177 Stockholm, Sweden.
- Department of Pharmacological and Technological Chemistry, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia.
| | - Bo Zhang
- Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, SE 17177 Stockholm, Sweden.
- Department of Pharmacological and Technological Chemistry, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia.
| | - Louis S Stodieck
- BioServe Space Technologies, University of Colorado at Boulder, Boulder, CO 80303, USA.
| | - Virginia L Ferguson
- BioServe Space Technologies, University of Colorado at Boulder, Boulder, CO 80303, USA.
| | - Ted A Bateman
- Department of Bioengineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Michael J Pecaut
- Department of Basic Sciences, Division of Biomedical Engineering Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| |
Collapse
|
43
|
Radiation-Induced Reactions in The Liver - Modulation of Radiation Effects by Lifestyle-Related Factors. Int J Mol Sci 2018; 19:ijms19123855. [PMID: 30513990 PMCID: PMC6321068 DOI: 10.3390/ijms19123855] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022] Open
Abstract
Radiation has a wide variety of effects on the liver. Fibrosis is a concern in medical fields as one of the acute effects of high-dose irradiation, such as with cancer radiotherapies. Cancer is also an important concern following exposure to radiation. The liver has an active metabolism and reacts to radiations. In addition, effects are modulated by many environmental factors, such as high-calorie foods or alcohol beverages. Adaptations to other environmental conditions could also influence the effects of radiation. Reactions to radiation may not be optimally regulated under conditions modulated by the environment, possibly leading to dysregulation, disease or cancer. Here, we introduce some reactions to ionizing radiation in the liver, as demonstrated primarily in animal experiments. In addition, modulation of radiation-induced effects in the liver due to factors such as obesity, alcohol drinking, or supplements derived from foods are reviewed. Perspectives on medical applications by modulations of radiation effects are also discussed.
Collapse
|
44
|
Strollo F, Gentile S, Strollo G, Mambro A, Vernikos J. Recent Progress in Space Physiology and Aging. Front Physiol 2018; 9:1551. [PMID: 30483144 PMCID: PMC6240610 DOI: 10.3389/fphys.2018.01551] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/16/2018] [Indexed: 12/17/2022] Open
Abstract
Astronauts coming back from long-term space missions present with different health problems potentially affecting mission performance, involving all functional systems and organs and closely resembling those found in the elderly. This review points out the most recent advances in the literature in areas of expertise in which specific research groups were particularly creative, and as they relate to aging and to possible benefits on Earth for disabled people. The update of new findings and approaches in space research refers especially to neuro-immuno-endocrine-metabolic interactions, optic nerve edema, motion sickness and muscle-tendon-bone interplay and aims at providing the curious - and even possibly naïve young researchers – with a source of inspiration and of creative ideas for translational research.
Collapse
Affiliation(s)
| | - Sandro Gentile
- Campania University "Luigi Vanvitelli" and Nefrocenter Research Network, Naples, Italy
| | | | - Andrea Mambro
- Anesthesiology and Resuscitation Unit, "Misercordia" Hospital, Grosseto, Italy
| | | |
Collapse
|
45
|
Genchi GG, Degl'Innocenti A, Salgarella AR, Pezzini I, Marino A, Menciassi A, Piccirillo S, Balsamo M, Ciofani G. Modulation of gene expression in rat muscle cells following treatment with nanoceria in different gravity regimes. Nanomedicine (Lond) 2018; 13:2821-2833. [PMID: 30334476 DOI: 10.2217/nnm-2018-0316] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM Oxidative stress (OS) is strictly associated with senescence/pathogenesis of biological systems. As putative countermeasure to environmental OS, cerium oxide nanoparticles (nanoceria [NC]) were administered to muscle cells on ground and aboard the International Space Station. MATERIALS & METHODS Transcriptional analyses were conducted through microarray technology and hierarchical clustering. Venn diagram and gene ontology analyses were also performed on selected gene lists. RESULTS Adaptive responses to both NC administration and to permanence in real microgravity conditions occurred. Enrichment in the biological processes related to aging, body fat development and mesodermal tissue proliferation for NC-treated samples were found. CONCLUSION Nanotechnology antioxidants promise applications to pathological conditions governed by OS on Earth and in life-hostile environments (low Earth orbit and deep space).
Collapse
Affiliation(s)
- Giada Graziana Genchi
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy
| | - Andrea Degl'Innocenti
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy
| | - Alice Rita Salgarella
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy
| | - Ilaria Pezzini
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy
| | - Attilio Marino
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy
| | - Arianna Menciassi
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy
| | - Sara Piccirillo
- Agenzia Spaziale Italiana, Via del Politecnico snc, Roma 00133, Italy
| | - Michele Balsamo
- Kayser Italia S.r.l., Via di Popogna 501, Livorno 57128, Italy
| | - Gianni Ciofani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy.,Politecnico di Torino, Department of Aerospace & Mechanical Engineering, Corso Duca degli Abruzzi 24, Torino 10129, Italy
| |
Collapse
|
46
|
Ray S, Gebre S, Fogle H, Berrios DC, Tran PB, Galazka JM, Costes SV. GeneLab: Omics database for spaceflight experiments. Bioinformatics 2018; 35:1753-1759. [DOI: 10.1093/bioinformatics/bty884] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/09/2018] [Accepted: 10/16/2018] [Indexed: 12/16/2022] Open
Affiliation(s)
- Shayoni Ray
- Space Biosciences Division, USRA/NASA Ames Research Center, Moffett Field, CA, USA
| | - Samrawit Gebre
- Space Biosciences Division, KBRwyle/NASA Ames Research Center, Moffett Field, CA, USA
| | - Homer Fogle
- Space Biosciences Division, KBRwyle/NASA Ames Research Center, Moffett Field, CA, USA
| | - Daniel C Berrios
- Space Biosciences Division, USRA/NASA Ames Research Center, Moffett Field, CA, USA
| | - Peter B Tran
- Intelligent Systems Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Jonathan M Galazka
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| |
Collapse
|
47
|
A microRNA signature and TGF-β1 response were identified as the key master regulators for spaceflight response. PLoS One 2018; 13:e0199621. [PMID: 30044882 PMCID: PMC6059388 DOI: 10.1371/journal.pone.0199621] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/03/2018] [Indexed: 12/30/2022] Open
Abstract
Translating fundamental biological discoveries from NASA Space Biology program into health risk from space flights has been an ongoing challenge. We propose to use NASA GeneLab database to gain new knowledge on potential systemic responses to space. Unbiased systems biology analysis of transcriptomic data from seven different rodent datasets reveals for the first time the existence of potential “master regulators” coordinating a systemic response to microgravity and/or space radiation with TGF-β1 being the most common regulator. We hypothesized the space environment leads to the release of biomolecules circulating inside the blood stream. Through datamining we identified 13 candidate microRNAs (miRNA) which are common in all studies and directly interact with TGF-β1 that can be potential circulating factors impacting space biology. This study exemplifies the utility of the GeneLab data repository to aid in the process of performing novel hypothesis–based research.
Collapse
|
48
|
Global transcriptomic analysis suggests carbon dioxide as an environmental stressor in spaceflight: A systems biology GeneLab case study. Sci Rep 2018. [PMID: 29520055 PMCID: PMC5843582 DOI: 10.1038/s41598-018-22613-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Spaceflight introduces a combination of environmental stressors, including microgravity, ionizing radiation, changes in diet and altered atmospheric gas composition. In order to understand the impact of each environmental component on astronauts it is important to investigate potential influences in isolation. Rodent spaceflight experiments involve both standard vivarium cages and animal enclosure modules (AEMs), which are cages used to house rodents in spaceflight. Ground control AEMs are engineered to match the spaceflight environment. There are limited studies examining the biological response invariably due to the configuration of AEM and vivarium housing. To investigate the innate global transcriptomic patterns of rodents housed in spaceflight-matched AEM compared to standard vivarium cages we utilized publicly available data from the NASA GeneLab repository. Using a systems biology approach, we observed that AEM housing was associated with significant transcriptomic differences, including reduced metabolism, altered immune responses, and activation of possible tumorigenic pathways. Although we did not perform any functional studies, our findings revealed a mild hypoxic phenotype in AEM, possibly due to atmospheric carbon dioxide that was increased to match conditions in spaceflight. Our investigation illustrates the process of generating new hypotheses and informing future experimental research by repurposing multiple space-flown datasets.
Collapse
|
49
|
Friedman JE, Dobrinskikh E, Alfonso‐Garcia A, Fast A, Janssen RC, Soderborg TK, Anderson AL, Reisz JA, D'Alessandro A, Frank DN, Robertson CE, de la Houssaye BA, Johnson LK, Orlicky DJ, Wang XX, Levi M, Potma EO, El Kasmi KC, Jonscher KR. Pyrroloquinoline quinone prevents developmental programming of microbial dysbiosis and macrophage polarization to attenuate liver fibrosis in offspring of obese mice. Hepatol Commun 2018; 2:313-328. [PMID: 29507905 PMCID: PMC5831029 DOI: 10.1002/hep4.1139] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 11/02/2017] [Accepted: 12/05/2017] [Indexed: 12/17/2022] Open
Abstract
Increasingly, evidence suggests that exposure to maternal obesity creates an inflammatory environment in utero, exerting long-lasting postnatal signatures on the juvenile innate immune system and microbiome that may predispose offspring to development of fatty liver disease. We found that exposure to a maternal Western-style diet (WD) accelerated fibrogenesis in the liver of offspring and was associated with early recruitment of proinflammatory macrophages at 8-12 weeks and microbial dysbiosis as early as 3 weeks of age. We further demonstrated that bone marrow-derived macrophages (BMDMs) were polarized toward an inflammatory state at 8 weeks of age and that a potent antioxidant, pyrroloquinoline quinone (PQQ), reversed BMDM metabolic reprogramming from glycolytic toward oxidative metabolism by restoring trichloroacetic acid cycle function at isocitrate dehydrogenase. This resulted in reduced inflammation and inhibited collagen fibril formation in the liver at 20 weeks of age, even when PQQ was withdrawn at 3 weeks of age. Beginning at 3 weeks of age, WD-fed mice developed a decreased abundance of Parabacteroides and Lactobacillus, together with increased Ruminococcus and decreased tight junction gene expression by 20 weeks, whereas microbiota of mice exposed to PQQ retained compositional stability with age, which was associated with improved liver health. Conclusion: Exposure to a maternal WD induces early gut dysbiosis and disrupts intestinal tight junctions, resulting in BMDM polarization and induction of proinflammatory and profibrotic programs in the offspring that persist into adulthood. Disrupted macrophage and microbiota function can be attenuated by short-term maternal treatment with PQQ prior to weaning, suggesting that reshaping the early gut microbiota in combination with reprogramming macrophages during early weaning may alleviate the sustained proinflammatory environment, preventing the rapid progression of nonalcoholic fatty liver disease to nonalcoholic steatohepatitis in offspring of obese mothers. (Hepatology Communications 2018;2:313-328).
Collapse
Affiliation(s)
| | - Evgenia Dobrinskikh
- Division of Renal Diseases and Hypertension, Department of MedicineUniversity of Colorado Anschutz Medical CampusAuroraCO
| | - Alba Alfonso‐Garcia
- Department of Biomedical Engineering and Beckman Laser InstituteUniversity of CaliforniaIrvine, IrvineCA
| | - Alexander Fast
- Department of Biomedical Engineering and Beckman Laser InstituteUniversity of CaliforniaIrvine, IrvineCA
| | | | | | - Aimee L. Anderson
- Children's Hospital Colorado, Digestive Disease Institute and Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics
| | | | | | | | | | | | | | | | - Xiaoxin X. Wang
- Division of Renal Diseases and Hypertension, Department of MedicineUniversity of Colorado Anschutz Medical CampusAuroraCO
| | - Moshe Levi
- Division of Renal Diseases and Hypertension, Department of MedicineUniversity of Colorado Anschutz Medical CampusAuroraCO
| | - Eric O. Potma
- Department of Biomedical Engineering and Beckman Laser InstituteUniversity of CaliforniaIrvine, IrvineCA
| | - Karim C. El Kasmi
- Children's Hospital Colorado, Digestive Disease Institute and Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics
| | - Karen R. Jonscher
- Department of AnesthesiologyUniversity of Colorado Anschutz Medical CampusAuroraCO
| |
Collapse
|
50
|
Gene-metabolite profile integration to understand the cause of spaceflight induced immunodeficiency. NPJ Microgravity 2018; 4:4. [PMID: 29387784 PMCID: PMC5788863 DOI: 10.1038/s41526-017-0038-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 11/06/2017] [Accepted: 12/28/2017] [Indexed: 12/15/2022] Open
Abstract
Spaceflight presents a spectrum of stresses very different from those associated with terrestrial conditions. Our previous study (BMC Genom. 15: 659, 2014) integrated the expressions of mRNAs, microRNAs, and proteins and results indicated that microgravity induces an immunosuppressive state that can facilitate opportunistic pathogenic attack. However, the existing data are not sufficient for elucidating the molecular drivers of the given immunosuppressed state. To meet this knowledge gap, we focused on the metabolite profile of spaceflown human cells. Independent studies have attributed cellular energy deficiency as a major cause of compromised immunity of the host, and metabolites that are closely associated with energy production could be a robust signature of atypical energy fluctuation. Our protocol involved inoculation of human endothelial cells in cell culture modules in spaceflight and on the ground concurrently. Ten days later, the cells in space and on the ground were exposed to lipopolysaccharide (LPS), a ubiquitous membrane endotoxin of Gram-negative bacteria. Nucleic acids, proteins, and metabolites were collected 4 and 8 h post-LPS exposure. Untargeted profiling of metabolites was followed by targeted identification of amino acids and knowledge integration with gene expression profiles. Consistent with the past reports associating microgravity with increased energy expenditure, we identified several markers linked to energy deficiency, including various amino acids such as tryptophan, creatinine, dopamine, and glycine, and cofactors such as lactate and pyruvate. The present study revealed a molecular architecture linking energy metabolism and immunodeficiency in microgravity. The energy-deficient condition potentially cascaded into dysregulation of protein metabolism and impairment of host immunity. This project is limited by a small sample size. Although a strict statistical screening was carefully implemented, the present results further emphasize the need for additional studies with larger sample sizes. Validating this hypothesis using an in vivo model is essential to extend the knowledge towards identifying markers of diagnostic and therapeutic value. Human cells challenged with a bacterial toxin show more signs of energy deficiency when flown in space than when cultured on the ground. Rasha Hammamieh from the US Army Center for Environmental Health Research in Frederick, Maryland, and colleagues exposed human endothelial cells in spaceflight to lipopolysaccharide, an immune response-triggering part of the bacterial membrane. They then collected nucleic acids, proteins and metabolites 4 and 8 h later, and saw a molecular architecture consistent with increased energy expenditure compared to matched control cells grown on Earth. Combined with the researchers’ previous finding that microgravity can induce an immunosuppressive state, the results suggest that energy imbalances potentially lead to problems with protein metabolism that ultimately impair the immune system. The authors propose that reversing this energy depletion could help enhance the immune health of astronauts.
Collapse
|