51
|
McDonald JT, Kim J, Farmerie L, Johnson ML, Trovao NS, Arif S, Siew K, Tsoy S, Bram Y, Park J, Overbey E, Ryon K, Haltom J, Singh U, Enguita FJ, Zaksas V, Guarnieri JW, Topper M, Wallace DC, Meydan C, Baylin S, Meller R, Muratani M, Porterfield DM, Kaufman B, Mori MA, Walsh SB, Sigaudo-Roussel D, Mebarek S, Bottini M, Marquette CA, Wurtele ES, Schwartz RE, Galeano D, Mason CE, Grabham P, Beheshti A. Space radiation damage rescued by inhibition of key spaceflight associated miRNAs. Nat Commun 2024; 15:4825. [PMID: 38862542 PMCID: PMC11166944 DOI: 10.1038/s41467-024-48920-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/17/2024] [Indexed: 06/13/2024] Open
Abstract
Our previous research revealed a key microRNA signature that is associated with spaceflight that can be used as a biomarker and to develop countermeasure treatments to mitigate the damage caused by space radiation. Here, we expand on this work to determine the biological factors rescued by the countermeasure treatment. We performed RNA-sequencing and transcriptomic analysis on 3D microvessel cell cultures exposed to simulated deep space radiation (0.5 Gy of Galactic Cosmic Radiation) with and without the antagonists to three microRNAs: miR-16-5p, miR-125b-5p, and let-7a-5p (i.e., antagomirs). Significant reduction of inflammation and DNA double strand breaks (DSBs) activity and rescue of mitochondria functions are observed after antagomir treatment. Using data from astronaut participants in the NASA Twin Study, Inspiration4, and JAXA missions, we reveal the genes and pathways implicated in the action of these antagomirs are altered in humans. Our findings indicate a countermeasure strategy that can potentially be utilized by astronauts in spaceflight missions to mitigate space radiation damage.
Collapse
Affiliation(s)
- J Tyson McDonald
- Department of Radiation Medicine, Georgetown University School of Medicine, Washington, D.C, USA
| | - JangKeun Kim
- Department of Physiology, Biophysics and Systems Biology and the WorldQuant Initiative, Weill Cornell Medicine, New York, NY, USA
| | - Lily Farmerie
- Vascular Medicine Institute at the University of Pittsburgh Department of Medicine, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Meghan L Johnson
- Vascular Medicine Institute at the University of Pittsburgh Department of Medicine, Pittsburgh, PA, USA
| | - Nidia S Trovao
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Shehbeel Arif
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Keith Siew
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
| | - Sergey Tsoy
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Yaron Bram
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jiwoon Park
- Department of Physiology, Biophysics and Systems Biology and the WorldQuant Initiative, Weill Cornell Medicine, New York, NY, USA
| | - Eliah Overbey
- Department of Physiology, Biophysics and Systems Biology and the WorldQuant Initiative, Weill Cornell Medicine, New York, NY, USA
| | - Krista Ryon
- Department of Physiology, Biophysics and Systems Biology and the WorldQuant Initiative, Weill Cornell Medicine, New York, NY, USA
| | - Jeffrey Haltom
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Urminder Singh
- Bioinformatics and Computational Biology Program, Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 90011, USA
| | - Francisco J Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Victoria Zaksas
- Center for Translational Data Science, University of Chicago, Chicago, IL, 60637, USA
- Clever Research Lab, Springfield, IL, 62704, USA
| | - Joseph W Guarnieri
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Michael Topper
- Departments of Oncology and Medicine and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Douglas C Wallace
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Division of Human Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Cem Meydan
- Department of Physiology, Biophysics and Systems Biology and the WorldQuant Initiative, Weill Cornell Medicine, New York, NY, USA
| | - Stephen Baylin
- Departments of Oncology and Medicine and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Robert Meller
- Neuroscience Institute, Department of Neurobiology/ Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Masafumi Muratani
- Transborder Medical Research Center, University of Tsukuba, Ibaraki, 305-8575, Japan
- Department of Genome Biology, Institute of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - D Marshall Porterfield
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Brett Kaufman
- Vascular Medicine Institute at the University of Pittsburgh Department of Medicine, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas, Campinas, SP, Brazil
- Obesity and Comorbidities Research Center (OCRC), Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Stephen B Walsh
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
| | | | - Saida Mebarek
- ICBMS, UMR5246, CNRS, INSA, CPE-Lyon, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Massimo Bottini
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Christophe A Marquette
- 3d.FAB, CNRS, INSA, CPE-Lyon, UMR5246, ICBMS, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Eve Syrkin Wurtele
- Bioinformatics and Computational Biology Program, Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 90011, USA
- Genetics Program, Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 90011, USA
| | - Robert E Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Diego Galeano
- Facultad de Ingeniería, Universidad Nacional de Asunción, San Lorenzo, Paraguay
| | - Christopher E Mason
- Department of Physiology, Biophysics and Systems Biology and the WorldQuant Initiative, Weill Cornell Medicine, New York, NY, USA
| | - Peter Grabham
- Center for Radiological Research, College of Physicians and Surgeons, Columbia University, New York, NY, 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, US.
| |
Collapse
|
52
|
Houerbi N, Kim J, Overbey EG, Batra R, Schweickart A, Patras L, Lucotti S, Ryon KA, Najjar D, Meydan C, Damle N, Chin C, Narayanan SA, Guarnieri JW, Widjaja G, Beheshti A, Tobias G, Vatter F, Hirschberg JW, Kleinman A, Afshin EE, MacKay M, Chen Q, Miller D, Gajadhar AS, Williamson L, Tandel P, Yang Q, Chu J, Benz R, Siddiqui A, Hornburg D, Gross S, Shirah B, Krumsiek J, Mateus J, Mao X, Matei I, Mason CE. Secretome profiling reveals acute changes in oxidative stress, brain homeostasis, and coagulation following short-duration spaceflight. Nat Commun 2024; 15:4862. [PMID: 38862464 PMCID: PMC11166969 DOI: 10.1038/s41467-024-48841-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/15/2024] [Indexed: 06/13/2024] Open
Abstract
As spaceflight becomes more common with commercial crews, blood-based measures of crew health can guide both astronaut biomedicine and countermeasures. By profiling plasma proteins, metabolites, and extracellular vesicles/particles (EVPs) from the SpaceX Inspiration4 crew, we generated "spaceflight secretome profiles," which showed significant differences in coagulation, oxidative stress, and brain-enriched proteins. While >93% of differentially abundant proteins (DAPs) in vesicles and metabolites recovered within six months, the majority (73%) of plasma DAPs were still perturbed post-flight. Moreover, these proteomic alterations correlated better with peripheral blood mononuclear cells than whole blood, suggesting that immune cells contribute more DAPs than erythrocytes. Finally, to discern possible mechanisms leading to brain-enriched protein detection and blood-brain barrier (BBB) disruption, we examined protein changes in dissected brains of spaceflight mice, which showed increases in PECAM-1, a marker of BBB integrity. These data highlight how even short-duration spaceflight can disrupt human and murine physiology and identify spaceflight biomarkers that can guide countermeasure development.
Collapse
Affiliation(s)
- Nadia Houerbi
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Eliah G Overbey
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Richa Batra
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Annalise Schweickart
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional Biology and Medicine program, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Laura Patras
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Serena Lucotti
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Krista A Ryon
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Deena Najjar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Namita Damle
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Christopher Chin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - S Anand Narayanan
- Department of Nutrition & Integrative Physiology, Florida State University, Tallahassee, FL, USA
| | - Joseph W Guarnieri
- Center of Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Gabrielle Widjaja
- Center of Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Afshin Beheshti
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Gabriel Tobias
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Seer, Inc., Redwood City, CA, 94065, USA
| | - Fanny Vatter
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Seer, Inc., Redwood City, CA, 94065, USA
| | | | - Ashley Kleinman
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Evan E Afshin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Matthew MacKay
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Qiuying Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Dawson Miller
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | | | | | | | - Qiu Yang
- Seer, Inc., Redwood City, CA, 94065, USA
| | | | - Ryan Benz
- Seer, Inc., Redwood City, CA, 94065, USA
| | | | | | - Steven Gross
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Bader Shirah
- Department of Neuroscience, King Faisal Specialist Hospital & Research Centre, Jeddah, Saudi Arabia
| | - Jan Krumsiek
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional Biology and Medicine program, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jaime Mateus
- Space Exploration Technologies Corporation (SpaceX), Hawthorne, CA, USA
| | - Xiao Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA, 92350, USA
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA.
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA.
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
- Tri-Institutional Biology and Medicine program, Weill Cornell Medicine, New York, NY, 10021, USA.
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
- WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
53
|
Wu F, Du H, Overbey E, Kim J, Makhijani P, Martin N, Lerner CA, Nguyen K, Baechle J, Valentino TR, Fuentealba M, Bartleson JM, Halaweh H, Winer S, Meydan C, Garrett-Bakelman F, Sayed N, Melov S, Muratani M, Gerencser AA, Kasler HG, Beheshti A, Mason CE, Furman D, Winer DA. Single-cell analysis identifies conserved features of immune dysfunction in simulated microgravity and spaceflight. Nat Commun 2024; 15:4795. [PMID: 38862487 PMCID: PMC11166937 DOI: 10.1038/s41467-023-42013-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 09/27/2023] [Indexed: 06/13/2024] Open
Abstract
Microgravity is associated with immunological dysfunction, though the mechanisms are poorly understood. Here, using single-cell analysis of human peripheral blood mononuclear cells (PBMCs) exposed to short term (25 hours) simulated microgravity, we characterize altered genes and pathways at basal and stimulated states with a Toll-like Receptor-7/8 agonist. We validate single-cell analysis by RNA sequencing and super-resolution microscopy, and against data from the Inspiration-4 (I4) mission, JAXA (Cell-Free Epigenome) mission, Twins study, and spleens from mice on the International Space Station. Overall, microgravity alters specific pathways for optimal immunity, including the cytoskeleton, interferon signaling, pyroptosis, temperature-shock, innate inflammation (e.g., Coronavirus pathogenesis pathway and IL-6 signaling), nuclear receptors, and sirtuin signaling. Microgravity directs monocyte inflammatory parameters, and impairs T cell and NK cell functionality. Using machine learning, we identify numerous compounds linking microgravity to immune cell transcription, and demonstrate that the flavonol, quercetin, can reverse most abnormal pathways. These results define immune cell alterations in microgravity, and provide opportunities for countermeasures to maintain normal immunity in space.
Collapse
Grants
- R01 MH117406 NIMH NIH HHS
- T32 AG000266 NIA NIH HHS
- This work was supported in part through funds derived from the Buck Institute for Research on Aging (D.A.W., D.F.), and the Huiying Memorial Foundation (D.A.W.). T.V. and J.B. are funded by a T32 NIH fellowship grant (NIA T32 AG000266). C.E.M. thanks the Scientific Computing Unit (SCU) at WCM, the WorldQuant Foundation, NASA (NNX14AH50G, NNX17AB26G, 80NSSC22K0254, NNH18ZTT001N-FG2, 80NSSC22K0254, NNX16AO69A), the National Institutes of Health (R01MH117406), and LLS (MCL7001-18, LLS 9238-16).
Collapse
Affiliation(s)
- Fei Wu
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Huixun Du
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Eliah Overbey
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10021, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Priya Makhijani
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Nicolas Martin
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Chad A Lerner
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Khiem Nguyen
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Jordan Baechle
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | | | | | | | - Heather Halaweh
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Shawn Winer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Francine Garrett-Bakelman
- Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Simon Melov
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - 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
| | | | | | - Afshin Beheshti
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94043, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, 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, New York, NY, 10021, USA.
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, 94945, USA.
- Stanford 1000 Immunomes Project, Stanford University School of Medicine, Stanford, CA, USA.
- Institute for Research in Translational Medicine, Universidad Austral, CONICET, Pilar, Buenos Aires, Argentina.
| | - Daniel A Winer
- Buck Institute for Research on Aging, Novato, CA, 94945, USA.
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA.
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, M5G 1L7, Canada.
| |
Collapse
|
54
|
Kim J, Tierney BT, Overbey EG, Dantas E, Fuentealba M, Park J, Narayanan SA, Wu F, Najjar D, Chin CR, Meydan C, Loy C, Mathyk B, Klotz R, Ortiz V, Nguyen K, Ryon KA, Damle N, Houerbi N, Patras LI, Schanzer N, Hutchinson GA, Foox J, Bhattacharya C, Mackay M, Afshin EE, Hirschberg JW, Kleinman AS, Schmidt JC, Schmidt CM, Schmidt MA, Beheshti A, Matei I, Lyden D, Mullane S, Asadi A, Lenz JS, Mzava O, Yu M, Ganesan S, De Vlaminck I, Melnick AM, Barisic D, Winer DA, Zwart SR, Crucian BE, Smith SM, Mateus J, Furman D, Mason CE. Single-cell multi-ome and immune profiles of the Inspiration4 crew reveal conserved, cell-type, and sex-specific responses to spaceflight. Nat Commun 2024; 15:4954. [PMID: 38862516 PMCID: PMC11166952 DOI: 10.1038/s41467-024-49211-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
Spaceflight induces an immune response in astronauts. To better characterize this effect, we generated single-cell, multi-ome, cell-free RNA (cfRNA), biochemical, and hematology data for the SpaceX Inspiration4 (I4) mission crew. We found that 18 cytokines/chemokines related to inflammation, aging, and muscle homeostasis changed after spaceflight. In I4 single-cell multi-omics data, we identified a "spaceflight signature" of gene expression characterized by enrichment in oxidative phosphorylation, UV response, immune function, and TCF21 pathways. We confirmed the presence of this signature in independent datasets, including the NASA Twins Study, the I4 skin spatial transcriptomics, and 817 NASA GeneLab mouse transcriptomes. Finally, we observed that (1) T cells showed an up-regulation of FOXP3, (2) MHC class I genes exhibited long-term suppression, and (3) infection-related immune pathways were associated with microbiome shifts. In summary, this study reveals conserved and distinct immune disruptions occurring and details a roadmap for potential countermeasures to preserve astronaut health.
Collapse
Affiliation(s)
- JangKeun Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Braden T Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Eliah G Overbey
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Center for STEM, University of Austin, Austin, TX, USA
- BioAstra, Inc, New York, NY, USA
| | - Ezequiel Dantas
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Matias Fuentealba
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Jiwoon Park
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - S Anand Narayanan
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Fei Wu
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Deena Najjar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
| | - Christopher R Chin
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Tri-Institutional Biology and Medicine Program, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Conor Loy
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, NY, 14850, USA
| | - Begum Mathyk
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Remi Klotz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Veronica Ortiz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Khiem Nguyen
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Krista A Ryon
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
| | - Namita Damle
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
| | - Nadia Houerbi
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Laura I Patras
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Nathan Schanzer
- School of Medicine, New York Medical College, Valhalla, NY, 10595, USA
| | - Gwyneth A Hutchinson
- NASA Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, 94720, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jonathan Foox
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Chandrima Bhattacharya
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Tri-Institutional Biology and Medicine Program, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Matthew Mackay
- Tri-Institutional Biology and Medicine Program, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Evan E Afshin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jeremy Wain Hirschberg
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Ashley S Kleinman
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Julian C Schmidt
- Sovaris Aerospace, Boulder, CO, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, CO, USA
| | - Caleb M Schmidt
- Sovaris Aerospace, Boulder, CO, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, CO, USA
- Department of Systems Engineering, Colorado State University, Fort Collins, CO, USA
| | - Michael A Schmidt
- Sovaris Aerospace, Boulder, CO, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, CO, USA
| | - Afshin Beheshti
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Irina Matei
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - David Lyden
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Sean Mullane
- Space Exploration Technologies Corporation (SpaceX), Hawthorne, CA, USA
| | - Amran Asadi
- Space Exploration Technologies Corporation (SpaceX), Hawthorne, CA, USA
| | - Joan S Lenz
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, NY, 14850, USA
| | - Omary Mzava
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, NY, 14850, USA
| | - Min Yu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Saravanan Ganesan
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Iwijn De Vlaminck
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, NY, 14850, USA
| | - Ari M Melnick
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Darko Barisic
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Daniel A Winer
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Sara R Zwart
- University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
| | - Brian E Crucian
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Human Health and Performance Directorate, 2101 NASA Parkway, Houston, TX, 77058, USA
| | - Scott M Smith
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Human Health and Performance Directorate, 2101 NASA Parkway, Houston, TX, 77058, USA
| | - Jaime Mateus
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - David Furman
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, 94945, USA.
- Stanford 1000 Immunomes Project, Stanford School of Medicine, Stanford, CA, 94306, USA.
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral, CONICET, Pilar, Argentina.
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA.
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA.
- Tri-Institutional Biology and Medicine Program, Weill Cornell Medicine, New York, NY, 10021, USA.
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
- WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
55
|
Borg J, Loy C, Kim J, Buhagiar A, Chin C, Damle N, De Vlaminck I, Felice A, Liu T, Matei I, Meydan C, Muratani M, Mzava O, Overbey E, Ryon KA, Smith SM, Tierney BT, Trudel G, Zwart SR, Beheshti A, Mason CE, Borg J. Spatiotemporal expression and control of haemoglobin in space. Nat Commun 2024; 15:4927. [PMID: 38862545 PMCID: PMC11166948 DOI: 10.1038/s41467-024-49289-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 05/31/2024] [Indexed: 06/13/2024] Open
Abstract
It is now widely recognised that the environment in space activates a diverse set of genes involved in regulating fundamental cellular pathways. This includes the activation of genes associated with blood homoeostasis and erythropoiesis, with a particular emphasis on those involved in globin chain production. Haemoglobin biology provides an intriguing model for studying space omics, as it has been extensively explored at multiple -omic levels, spanning DNA, RNA, and protein analyses, in both experimental and clinical contexts. In this study, we examined the developmental expression of haemoglobin over time and space using a unique suite of multi-omic datasets available on NASA GeneLab, from the NASA Twins Study, the JAXA CFE study, and the Inspiration4 mission. Our findings reveal significant variations in globin gene expression corresponding to the distinct spatiotemporal characteristics of the collected samples. This study sheds light on the dynamic nature of globin gene regulation in response to the space environment and provides valuable insights into the broader implications of space omics research.
Collapse
Affiliation(s)
- Josef Borg
- Faculty of Health Sciences, University of Malta, Msida, MSD2080, Malta
| | - Conor Loy
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Alfred Buhagiar
- Faculty of Health Sciences, University of Malta, Msida, MSD2080, Malta
| | - Christopher Chin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Namita Damle
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Iwijn De Vlaminck
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Alex Felice
- Department of Surgery, Faculty of Medicine and Surgery, University of Malta, Msida, MSD2080, Malta
| | - Tammy Liu
- Ottawa Hospital Research Institute, Department of Medicine, Ottawa, Ontario, Canada
| | - Irina Matei
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Masafumi Muratani
- Department of Genome Biology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Omary Mzava
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Eliah Overbey
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Krista A Ryon
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Scott M Smith
- Biomedical Research and Environmental Sciences Division, Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX, USA
| | - Braden T Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Guy Trudel
- Ottawa Hospital Research Institute, Department of Medicine, Ottawa, Ontario, Canada
| | - Sara R Zwart
- Biomedical Research and Environmental Sciences Division, Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX, USA
- University of Texas Medical Branch, Galveston, TX, USA
| | - Afshin Beheshti
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10065, USA.
| | - Joseph Borg
- Faculty of Health Sciences, University of Malta, Msida, MSD2080, Malta.
| |
Collapse
|
56
|
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
|
57
|
Overbey EG, Ryon K, Kim J, Tierney BT, Klotz R, Ortiz V, Mullane S, Schmidt JC, MacKay M, Damle N, Najjar D, Matei I, Patras L, Garcia Medina JS, Kleinman AS, Wain Hirschberg J, Proszynski J, Narayanan SA, Schmidt CM, Afshin EE, Innes L, Saldarriaga MM, Schmidt MA, Granstein RD, Shirah B, Yu M, Lyden D, Mateus J, Mason CE. Collection of biospecimens from the inspiration4 mission establishes the standards for the space omics and medical atlas (SOMA). Nat Commun 2024; 15:4964. [PMID: 38862509 PMCID: PMC11166662 DOI: 10.1038/s41467-024-48806-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/15/2024] [Indexed: 06/13/2024] Open
Abstract
The SpaceX Inspiration4 mission provided a unique opportunity to study the impact of spaceflight on the human body. Biospecimen samples were collected from four crew members longitudinally before (Launch: L-92, L-44, L-3 days), during (Flight Day: FD1, FD2, FD3), and after (Return: R + 1, R + 45, R + 82, R + 194 days) spaceflight, spanning a total of 289 days across 2021-2022. The collection process included venous whole blood, capillary dried blood spot cards, saliva, urine, stool, body swabs, capsule swabs, SpaceX Dragon capsule HEPA filter, and skin biopsies. Venous whole blood was further processed to obtain aliquots of serum, plasma, extracellular vesicles and particles, and peripheral blood mononuclear cells. In total, 2,911 sample aliquots were shipped to our central lab at Weill Cornell Medicine for downstream assays and biobanking. This paper provides an overview of the extensive biospecimen collection and highlights their processing procedures and long-term biobanking techniques, facilitating future molecular tests and evaluations.As such, this study details a robust framework for obtaining and preserving high-quality human, microbial, and environmental samples for aerospace medicine in the Space Omics and Medical Atlas (SOMA) initiative, which can aid future human spaceflight and space biology experiments.
Collapse
Affiliation(s)
- Eliah G Overbey
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- BioAstra, Inc, New York, NY, USA
- Center for STEM, University of Austin, Austin, TX, 78701, USA
| | - Krista Ryon
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Braden T Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Remi Klotz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Veronica Ortiz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sean Mullane
- Space Exploration Technologies Corporation, Hawthorne, CA, USA
| | - Julian C Schmidt
- Sovaris Aerospace, Boulder, Colorado, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, Colorado, USA
| | - Matthew MacKay
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Namita Damle
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Deena Najjar
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Laura Patras
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - J Sebastian Garcia Medina
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Ashley S Kleinman
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jeremy Wain Hirschberg
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jacqueline Proszynski
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - S Anand Narayanan
- Florida State University, College of Education, Health, and Human Sciences, Department of Health, Nutrition, and Food Sciences, Tallahassee, FL, USA
| | - Caleb M Schmidt
- Sovaris Aerospace, Boulder, Colorado, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, Colorado, USA
- Department of Systems Engineering, Colorado State University, Fort Collins, Colorado, USA
| | - Evan E Afshin
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Lucinda Innes
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | | | - Michael A Schmidt
- Sovaris Aerospace, Boulder, Colorado, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, Colorado, USA
| | | | - Bader Shirah
- Department of Neuroscience, King Faisal Specialist Hospital & Research Centre, Jeddah, Saudi Arabia
| | - Min Yu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - David Lyden
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Jaime Mateus
- Space Exploration Technologies Corporation, Hawthorne, CA, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
- BioAstra, Inc, New York, NY, USA.
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
- WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
58
|
Seylani A, Galsinh AS, Tasoula A, I AR, Camera A, Calleja-Agius J, Borg J, Goel C, Kim J, Clark KB, Das S, Arif S, Boerrigter M, Coffey C, Szewczyk N, Mason CE, Manoli M, Karouia F, Schwertz H, Beheshti A, Tulodziecki D. Ethical considerations for the age of non-governmental space exploration. Nat Commun 2024; 15:4774. [PMID: 38862473 PMCID: PMC11166968 DOI: 10.1038/s41467-023-44357-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 12/05/2023] [Indexed: 06/13/2024] Open
Abstract
Mounting ambitions and capabilities for public and private, non-government sector crewed space exploration bring with them an increasingly diverse set of space travelers, raising new and nontrivial ethical, legal, and medical policy and practice concerns which are still relatively underexplored. In this piece, we lay out several pressing issues related to ethical considerations for selecting space travelers and conducting human subject research on them, especially in the context of non-governmental and commercial/private space operations.
Collapse
Affiliation(s)
- Allen Seylani
- School of Medicine, University of California, Riverside. 92521 Botanical Garden Dr, Riverside, CA, 92507, USA
| | - Aman Singh Galsinh
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, AB24 3FX, UK
| | - Alexia Tasoula
- Department of Life Science Engineering, FH Technikum, Vienna, Austria
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Anu R I
- Department of Cancer Biology and Therapeutics, MVR Cancer Centre and Research Institute, Calicut, India
- Department of Clinical Biochemistry, MVR Cancer Centre and Research Institute, Calicut, India
| | - Andrea Camera
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jean Calleja-Agius
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD2080, Msida, Malta
| | - Joseph Borg
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, MSD2080, Msida, Malta
| | - Chirag Goel
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - JangKeun Kim
- Department of Physiology & Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Kevin B Clark
- Cures Within Reach, Chicago, IL, 60602, USA
- Peace Innovation Institute, The Hague 2511, Netherlands & Stanford University, Palo Alto, CA, 94305, USA
- Biometrics and Nanotechnology Councils, Institute for Electrical and Electronics Engineers, New York, NY, 10016-5997, USA
| | - Saswati Das
- Department of Biochemistry, Atal Bihari Vajpayee Institute of Medical Sciences, New Delhi, India
| | - Shehbeel Arif
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Caroline Coffey
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Nathaniel Szewczyk
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Christopher E Mason
- Department of Physiology & Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Maria Manoli
- School of Law, University of Aberdeen, Aberdeen, AB24 3UB, UK
| | - Fathi Karouia
- Blue Marble Space Institute for 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
| | - Hansjörg Schwertz
- Molecular Medicine Program at the University of Utah, Salt Lake City, UT, 84112, USA.
- Division of Occupational Medicine at the University of Utah, Salt Lake City, UT, 84112, USA.
- Occupational Medicine at Billings Clinic Bozeman, Bozeman, MT, 59715, 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, US.
| | - Dana Tulodziecki
- Department of Philosophy, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
59
|
Murgia M, Rittweger J, Reggiani C, Bottinelli R, Mann M, Schiaffino S, Narici MV. Spaceflight on the ISS changed the skeletal muscle proteome of two astronauts. NPJ Microgravity 2024; 10:60. [PMID: 38839773 PMCID: PMC11153545 DOI: 10.1038/s41526-024-00406-3] [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: 02/05/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
Skeletal muscle undergoes atrophy and loss of force during long space missions, when astronauts are persistently exposed to altered gravity and increased ionizing radiation. We previously carried out mass spectrometry-based proteomics from skeletal muscle biopsies of two astronauts, taken before and after a mission on the International Space Station. The experiments were part of an effort to find similarities between spaceflight and bed rest, a ground-based model of unloading, focused on proteins located at the costameres. We here extend the data analysis of the astronaut dataset and show compartment-resolved changes in the mitochondrial proteome, remodeling of the extracellular matrix and of the antioxidant response. The astronauts differed in their level of onboard physical exercise, which correlated with their respective preservation of muscle mass and force at landing in previous analyses. We show that the mitochondrial proteome downregulation during spaceflight, particularly the inner membrane and matrix, was dramatic for both astronauts. The expression of autophagy regulators and reactive oxygen species scavengers, however, showed partially opposite expression trends in the two subjects, possibly correlating with their level of onboard exercise. As mitochondria are primarily affected in many different tissues during spaceflight, we hypothesize that reactive oxygen species (ROS) rather than mechanical unloading per se could be the primary cause of skeletal muscle mitochondrial damage in space. Onboard physical exercise might have a strong direct effect on the prevention of muscle atrophy through mechanotransduction and a subsidiary effect on mitochondrial quality control, possibly through upregulation of autophagy and anti-oxidant responses.
Collapse
Affiliation(s)
- Marta Murgia
- Department of Biomedical Sciences, University of Padova, 35131, Padua, Italy.
- Max-Planck-Institute of Biochemistry, 82152, Martinsried, Germany.
| | - Jörn Rittweger
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
- Department of Pediatrics and Adolescent Medicine, University Hospital Cologne, Cologne, Germany
| | - Carlo Reggiani
- Department of Biomedical Sciences, University of Padova, 35131, Padua, Italy
- Science and Research Center Koper, Institute for Kinesiology Research, 6000, Koper, Slovenia
| | - Roberto Bottinelli
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Matthias Mann
- Department of Biomedical Sciences, University of Padova, 35131, Padua, Italy
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Marco V Narici
- Department of Biomedical Sciences, University of Padova, 35131, Padua, Italy
- Science and Research Center Koper, Institute for Kinesiology Research, 6000, Koper, Slovenia
- CIR-MYO Myology Center, 35121, Padua, Italy
| |
Collapse
|
60
|
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
|
61
|
Itkin T, Unger K, Barak Y, Yovel A, Stekolshchik L, Ego L, Aydinov Y, Gerchman Y, Sapir A. Exploiting the Unique Biology of Caenorhabditis elegans to Launch Neurodegeneration Studies in Space. ASTROBIOLOGY 2024; 24:579-589. [PMID: 38917419 DOI: 10.1089/ast.2023.0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
The 21st century is likely to be the first century in which large-scale short- and long-term space missions become common. Accordingly, an ever-increasing body of research is focusing on understanding the effects of current and future space expeditions on human physiology in health and disease. Yet the complex experimental environment, the small number of participants, and the high cost of space missions are among the primary factors that hinder a better understanding of the impact of space missions on human physiology. The goal of our research was to develop a cost-effective, compact, and easy-to-manipulate system to address questions related to human health and disease in space. This initiative was part of the Ramon SpaceLab program, an annual research-based learning program designed to cultivate high school students' involvement in space exploration by facilitating experiments aboard the International Space Station (ISS). In the present study, we used the nematode Caenorhabditis elegans (C. elegans), a well-suited model organism, to investigate the effect of space missions on neurodegeneration-related processes. Our study specifically focused on the level of aggregation of Huntington's disease-causing polyglutamine stretch-containing (PolyQ) proteins in C. elegans muscles, the canonical system for studying neurodegeneration in this organism. We compared animals expressing PolyQ proteins grown onboard the ISS with their genetically identical siblings grown on Earth and observed a significant difference in the number of aggregates between the two populations. Currently, it is challenging to determine whether this effect stems from developmental or morphological differences between the cultures or is a result of life in space. Nevertheless, our results serve as a proof of concept and open a new avenue for utilizing C. elegans to address various open questions in space studies, including the effects of space conditions on the onset and development of neurodegenerative diseases.
Collapse
Affiliation(s)
- Tatyana Itkin
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Ksenia Unger
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Yair Barak
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Amit Yovel
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Liya Stekolshchik
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Linoy Ego
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Yana Aydinov
- Science, Technology, Engineering, and Mathematics Program, Shakim High School, Nahariya, Israel
| | - Yoram Gerchman
- Department of Biology and the Environment, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
- Institute of Evolution, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
- Oranim Academic College, Kiryat Tivon, Israel
| | - Amir Sapir
- Department of Biology and the Environment, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
62
|
Kaur J, Kaur J, Nigam A. Extremophiles in Space Exploration. Indian J Microbiol 2024; 64:418-428. [PMID: 39010991 PMCID: PMC11246395 DOI: 10.1007/s12088-024-01297-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/28/2024] [Indexed: 07/17/2024] Open
Abstract
In the era of deep space exploration, extremophile research represents a key area of research w.r.t space survival. This review thus delves into the intriguing realm of 'Space and Astro Microbiology', providing insights into microbial survival, resilience, and behavioral adaptations in space-like environments. This discussion encompasses the modified behavior of extremophilic microorganisms, influencing virulence, stress resistance, and gene expression. It then shifts to recent studies on the International Space Station and simulated microgravity, revealing microbial responses that impact drug susceptibility, antibiotic resistance, and its commercial implications. The review then transitions into Astro microbiology, exploring the possibilities of interplanetary transit, lithopanspermia, and terraforming. Debates on life's origin and recent Martian meteorite discoveries are noted. We also discuss Proactive Inoculation Protocols for selecting adaptable microorganisms as terraforming pioneers. The discussion concludes with a note on microbes' role as bioengineers in bioregenerative life support systems, in recycling organic waste for sustainable space travel; and in promoting optimal plant growth to prepare Martian and lunar basalt. This piece emphasizes the transformative impact of microbes on the future of space exploration.
Collapse
Affiliation(s)
- Jasvinder Kaur
- Gargi College, University of Delhi, Siri Fort Road, New Delhi, 110 049 India
| | - Jaspreet Kaur
- Maitreyi College, University of Delhi, New Delhi, 110 021 India
| | - Aeshna Nigam
- Shivaji College, University of Delhi, New Delhi, 110 027 India
| |
Collapse
|
63
|
Pirjanian NA, Kalpana K, Kruglikov I, Mesci P, Stoudemire J, Grisanti P, Noggle SA, Loring JF, Fossati V. Establishing Neural Organoid Cultures for Investigating the Effects of Microgravity in Low-Earth Orbit (LEO). Methods Mol Biol 2024. [PMID: 38801498 DOI: 10.1007/7651_2024_550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Recent findings from studies involving astronauts and animal models indicate that microgravity increases immune cell activity and potentially alters the white and gray matter of the central nervous system (CNS). To further investigate the impact of microgravity on CNS cells, we established cultures of three-dimensional neural organoids containing isogenic microglia, the brain's resident immune cells, and sent them onboard the International Space Station. When using induced pluripotent stem cell (iPSC) lines from individuals affected by neuroinflammatory and neurodegenerative diseases such as multiple sclerosis (MS) and Parkinson's disease (PD), these cultures can provide novel insights into pathogenic pathways that may be exacerbated by microgravity. We have devised a cryovial culture strategy that enables organoids to be maintained through space travel and onboard the International Space Station (ISS) without the need for medium or carbon dioxide exchange. Here, we provide a comprehensive description of all the steps involved: generating various types of neural organoids, establishing long-term cultures, arranging plans for shipment to the Kennedy Space Center (KSC), and ultimately preparing organoids for launch into low-Earth orbit (LEO) and return to Earth for post-flight analyses.
Collapse
Affiliation(s)
| | - Kriti Kalpana
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Ilya Kruglikov
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | | | | | | | - Scott A Noggle
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Jeanne F Loring
- National Stem Cell Foundation, Louisville, KY, USA
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY, USA.
| |
Collapse
|
64
|
Salazar M, Joly S, Anglada-Escudé G, Ribas L. Epigenetic and physiological alterations in zebrafish subjected to hypergravity. PLoS One 2024; 19:e0300310. [PMID: 38776274 PMCID: PMC11111069 DOI: 10.1371/journal.pone.0300310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/27/2024] [Indexed: 05/24/2024] Open
Abstract
Gravity is one of the most constant environmental factors across Earth's evolution and all organisms are adapted to it. Consequently, spatial exploration has captured the interest in studying the biological changes that physiological alterations are caused by gravity. In the last two decades, epigenetics has explained how environmental cues can alter gene functions in organisms. Although many studies addressed gravity, the underlying biological and molecular mechanisms that occur in altered gravity for those epigenetics-related mechanisms, are mostly inexistent. The present study addressed the effects of hypergravity on development, behavior, gene expression, and most importantly, on the epigenetic changes in a worldwide animal model, the zebrafish (Danio rerio). To perform hypergravity experiments, a custom-centrifuge simulating the large diameter centrifuge (100 rpm ~ 3 g) was designed and zebrafish embryos were exposed during 5 days post fertilization (dpf). Results showed a significant decrease in survival at 2 dpf but no significance in the hatching rate. Physiological and morphological alterations including fish position, movement frequency, and swimming behavior showed significant changes due to hypergravity. Epigenetic studies showed significant hypermethylation of the genome of the zebrafish larvae subjected to 5 days of hypergravity. Downregulation of the gene expression of three epigenetic-related genes (dnmt1, dnmt3, and tet1), although not significant, was further observed. Taken altogether, gravity alterations affected biological responses including epigenetics in fish, providing a valuable roadmap of the putative hazards of living beyond Earth.
Collapse
Affiliation(s)
- Marcela Salazar
- Department of Renewable Marine Resources, Institut de Ciències del Mar—Consejo Superior de Investigaciones Científicas (ICM-CSIC), Barcelona, Spain
| | - Silvia Joly
- Department of Renewable Marine Resources, Institut de Ciències del Mar—Consejo Superior de Investigaciones Científicas (ICM-CSIC), Barcelona, Spain
| | - Guillem Anglada-Escudé
- Department of Astrophysics, Institut de Ciències de l’Espai—Consejo Superior de Investigaciones Científicas (ICE-CSIC), UAB Campus at Cerdanyola del Vallès, Barcelona, Spain
- Institut d’Estudis Espacials de Catalunya–IEEC/CERCA, Gran Capità, 2–4, Edifici Nexus, Despatx 201, Barcelona, Spain
| | - Laia Ribas
- Department of Renewable Marine Resources, Institut de Ciències del Mar—Consejo Superior de Investigaciones Científicas (ICM-CSIC), Barcelona, Spain
| |
Collapse
|
65
|
Riego ML, Meher PK, Brzozowska B, Akuwudike P, Bucher M, Oestreicher U, Lundholm L, Wojcik A. Chromosomal damage, gene expression and alternative transcription in human lymphocytes exposed to mixed ionizing radiation as encountered in space. Sci Rep 2024; 14:11502. [PMID: 38769353 PMCID: PMC11106305 DOI: 10.1038/s41598-024-62313-7] [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: 08/05/2023] [Accepted: 05/15/2024] [Indexed: 05/22/2024] Open
Abstract
Astronauts travelling in space will be exposed to mixed beams of particle radiation and photons. Exposure limits that correspond to defined cancer risk are calculated by multiplying absorbed doses by a radiation-type specific quality factor that reflects the biological effectiveness of the particle without considering possible interaction with photons. We have shown previously that alpha radiation and X-rays may interact resulting in synergistic DNA damage responses in human peripheral blood lymphocytes but the level of intra-individual variability was high. In order to assess the variability and validate the synergism, blood from two male donors was drawn at 9 time points during 3 seasons of the year and exposed to 0-2 Gy of X-rays, alpha particles or 1:1 mixture of both (half the dose each). DNA damage response was quantified by chromosomal aberrations and by mRNA levels of 3 radiation-responsive genes FDXR, CDKN1A and MDM2 measured 24 h post exposure. The quality of response in terms of differential expression of alternative transcripts was assessed by using two primer pairs per gene. A consistently higher than expected effect of mixed beams was found in both donors for chromosomal aberrations and gene expression with some seasonal variability for the latter. No synergy was detected for alternative transcription.
Collapse
Affiliation(s)
- Milagrosa López Riego
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, 106 91, Stockholm, Sweden
| | - Prabodha Kumar Meher
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, 106 91, Stockholm, Sweden
| | - Beata Brzozowska
- Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Pamela Akuwudike
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, 106 91, Stockholm, Sweden
| | - Martin Bucher
- Federal Office for Radiation Protection, Oberschleissheim, Germany
| | | | - Lovisa Lundholm
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, 106 91, Stockholm, Sweden
| | - Andrzej Wojcik
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, 106 91, Stockholm, Sweden.
- Institute of Biology, Jan Kochanowski University, Kielce, Poland.
| |
Collapse
|
66
|
Sanders LM, Grigorev KA, Scott RT, Saravia-Butler AM, Polo SHL, Gilbert R, Overbey EG, Kim J, Mason CE, Costes SV. Inspiration4 data access through the NASA Open Science Data Repository. NPJ Microgravity 2024; 10:56. [PMID: 38744887 PMCID: PMC11094041 DOI: 10.1038/s41526-024-00393-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/03/2024] [Indexed: 05/16/2024] Open
Abstract
The increasing accessibility of commercial and private space travel necessitates a profound understanding of its impact on human health. The NASA Open Science Data Repository (OSDR) provides transparent and FAIR access to biological studies, notably the SpaceX Inspiration4 (I4) mission, which amassed extensive data from civilian astronauts. This dataset encompasses omics and clinical assays, facilitating comprehensive research on space-induced biological responses. These data allow for multi-modal, longitudinal assessments, bridging the gap between human and model organism studies. Crucially, community-driven data standards established by NASA's OSDR Analysis Working Groups empower artificial intelligence and machine learning to glean invaluable insights, guiding future mission planning and health risk mitigation. This article presents a concise guide to access and analyze I4 data in OSDR, including programmatic access through GLOpenAPI. This pioneering effort establishes a precedent for post-mission health monitoring programs within space agencies, propelling research in the burgeoning field of commercial space travel's impact on human physiology.
Collapse
Affiliation(s)
- Lauren M Sanders
- Space Biosciences Research Branch, NASA Ames Research Center, Moffett Field, CA, USA
- Blue Marble Space, Seattle, WA, USA
| | - Kirill A Grigorev
- Space Biosciences Research Branch, NASA Ames Research Center, Moffett Field, CA, USA
- Blue Marble Space, Seattle, WA, USA
| | - Ryan T Scott
- Space Biosciences Research Branch, NASA Ames Research Center, Moffett Field, CA, USA
- KBR, Houston, TX, USA
| | - Amanda M Saravia-Butler
- Space Biosciences Research Branch, NASA Ames Research Center, Moffett Field, CA, USA
- KBR, Houston, TX, USA
| | - San-Huei Lai Polo
- Space Biosciences Research Branch, NASA Ames Research Center, Moffett Field, CA, USA
- KBR, Houston, TX, USA
| | - Rachel Gilbert
- Space Biosciences Research Branch, NASA Ames Research Center, Moffett Field, CA, USA
- KBR, Houston, TX, USA
| | - Eliah G Overbey
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Center for STEM, University of Austin, Austin, TX, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA
| | - Sylvain V Costes
- Space Biosciences Research Branch, NASA Ames Research Center, Moffett Field, CA, USA.
| |
Collapse
|
67
|
Trudel G, Stratis D, Rocheleau L, Pelchat M, Laneuville O. Transcriptomic evidence of erythropoietic adaptation from the International Space Station and from an Earth-based space analog. NPJ Microgravity 2024; 10:55. [PMID: 38740795 DOI: 10.1038/s41526-024-00400-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/25/2024] [Indexed: 05/16/2024] Open
Abstract
Space anemia affects astronauts and the underlying molecular alterations remain unknown. We evaluated the response of erythropoiesis-modulating genes to spaceflight through the analysis of leukocyte transcriptomes from astronauts during long-duration spaceflight and from an Earth model of microgravity. Differential expression analysis identified 50 genes encoding ribosomal proteins with reduced expression at the transition to bed rest and increased during the bed rest phase; a similar trend was observed in astronauts. Additional genes associated with anemia (15 genes), erythrocyte maturation (3 genes), and hemoglobin (6 genes) were down-regulated during bed rest and increased during reambulation. Transcript levels of the erythropoiesis transcription factor GATA1 and nine of most enriched erythrocyte proteins increased at reambulation after bed rest and at return to Earth from space. Dynamic changes of the leukocyte transcriptome composition while in microgravity and during reambulation supported an erythropoietic modulation accompanying the hemolysis of space anemia and of immobility-induced anemia.
Collapse
Affiliation(s)
- Guy Trudel
- Bone and Joint Research Laboratory, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.
- Department of Medicine, Division of Physiatry, Faculty of Medicine, University of Ottawa, 505 Smyth Road, Ottawa, ON, K1H 8M2, Canada.
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| | - Daniel Stratis
- Department of Biology, Faculty of Science, University of Ottawa, 30 Marie Curie Private Drive, Ottawa, ON, K1N 6N5, Canada.
| | - Lynda Rocheleau
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Martin Pelchat
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| | - Odette Laneuville
- Bone and Joint Research Laboratory, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.
- Department of Biology, Faculty of Science, University of Ottawa, 30 Marie Curie Private Drive, Ottawa, ON, K1N 6N5, Canada.
| |
Collapse
|
68
|
Schmauch E, Piening B, Mohebnasab M, Xia B, Zhu C, Stern J, Zhang W, Dowdell AK, Kim JI, Andrijevic D, Khalil K, Jaffe IS, Loza BL, Gragert L, Camellato BR, Oliveira MF, O'Brien DP, Chen HM, Weldon E, Gao H, Gandla D, Chang A, Bhatt R, Gao S, Lin X, Reddy KP, Kagermazova L, Habara AH, Widawsky S, Liang FX, Sall J, Loupy A, Heguy A, Taylor SEB, Zhu Y, Michael B, Jiang L, Jian R, Chong AS, Fairchild RL, Linna-Kuosmanen S, Kaikkonen MU, Tatapudi V, Lorber M, Ayares D, Mangiola M, Narula N, Moazami N, Pass H, Herati RS, Griesemer A, Kellis M, Snyder MP, Montgomery RA, Boeke JD, Keating BJ. Integrative multi-omics profiling in human decedents receiving pig heart xenografts. Nat Med 2024; 30:1448-1460. [PMID: 38760586 DOI: 10.1038/s41591-024-02972-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 04/03/2024] [Indexed: 05/19/2024]
Abstract
In a previous study, heart xenografts from 10-gene-edited pigs transplanted into two human decedents did not show evidence of acute-onset cellular- or antibody-mediated rejection. Here, to better understand the detailed molecular landscape following xenotransplantation, we carried out bulk and single-cell transcriptomics, lipidomics, proteomics and metabolomics on blood samples obtained from the transplanted decedents every 6 h, as well as histological and transcriptomic tissue profiling. We observed substantial early immune responses in peripheral blood mononuclear cells and xenograft tissue obtained from decedent 1 (male), associated with downstream T cell and natural killer cell activity. Longitudinal analyses indicated the presence of ischemia reperfusion injury, exacerbated by inadequate immunosuppression of T cells, consistent with previous findings of perioperative cardiac xenograft dysfunction in pig-to-nonhuman primate studies. Moreover, at 42 h after transplantation, substantial alterations in cellular metabolism and liver-damage pathways occurred, correlating with profound organ-wide physiological dysfunction. By contrast, relatively minor changes in RNA, protein, lipid and metabolism profiles were observed in decedent 2 (female) as compared to decedent 1. Overall, these multi-omics analyses delineate distinct responses to cardiac xenotransplantation in the two human decedents and reveal new insights into early molecular and immune responses after xenotransplantation. These findings may aid in the development of targeted therapeutic approaches to limit ischemia reperfusion injury-related phenotypes and improve outcomes.
Collapse
Affiliation(s)
- Eloi Schmauch
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | - Brian Piening
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | - Maedeh Mohebnasab
- Division of Molecular Genetics Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Bo Xia
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
- Society of Fellows, Harvard University, Cambridge, MA, USA
| | - Chenchen Zhu
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Jeffrey Stern
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Weimin Zhang
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
| | - Alexa K Dowdell
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | - Jacqueline I Kim
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - David Andrijevic
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Karen Khalil
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - Ian S Jaffe
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Bao-Li Loza
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Loren Gragert
- Division of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | | | - Han M Chen
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Elaina Weldon
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Hui Gao
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Divya Gandla
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Chang
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Riyana Bhatt
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah Gao
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiangping Lin
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Kriyana P Reddy
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Alawi H Habara
- Department of Biochemistry, College of Medicine, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Sophie Widawsky
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Feng-Xia Liang
- DART Microscopy Laboratory, NYU Langone Health, New York, NY, USA
| | - Joseph Sall
- DART Microscopy Laboratory, NYU Langone Health, New York, NY, USA
| | - Alexandre Loupy
- Université Paris Cité, Paris Institute for Transplantation and Organ Regeneration, Paris, France
| | - Adriana Heguy
- Genome Technology Center, NYU Langone Health, New York, NY, USA
| | | | - Yinan Zhu
- Division of Molecular Genetics Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Basil Michael
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Lihua Jiang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ruiqi Jian
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Robert L Fairchild
- Department of Inflammation and Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Suvi Linna-Kuosmanen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Minna U Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Vasishta Tatapudi
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | | | | | - Massimo Mangiola
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - Navneet Narula
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Nader Moazami
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
| | - Harvey Pass
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
| | - Ramin S Herati
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Adam Griesemer
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | | | - Robert A Montgomery
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Jef D Boeke
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, Brooklyn, NY, USA
| | - Brendan J Keating
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA.
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA.
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
69
|
Kimura Y, Nakai Y, Ino Y, Akiyama T, Moriyama K, Ohira T, Saito T, Inaba Y, Kumagai K, Ryo A, Hirano H. Identification of gravity-responsive serum proteins in spaceflight mice using a quantitative proteomic approach with data-independent acquisition mass spectrometry. Proteomics 2024; 24:e2300214. [PMID: 38475964 DOI: 10.1002/pmic.202300214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024]
Abstract
Physical inactivity associated with gravity unloading, such as microgravity during spaceflight and hindlimb unloading (HU), can cause various physiological changes. In this study, we attempted to identify serum proteins whose levels fluctuated in response to gravity unloading. First, we quantitatively assessed changes in the serum proteome profiles of spaceflight mice using mass spectrometry with data-independent acquisition. The serum levels of several proteins involved in the responses to estrogen and glucocorticoid, blood vessel maturation, osteoblast differentiation, and ossification were changed by microgravity exposure. Furthermore, a collective evaluation of serum proteomic data from spaceflight and HU mice identified 30 serum proteins, including Mmp2, Igfbp2, Tnc, Cdh5, and Pmel, whose levels varied to a similar extent in both gravity unloading models. These changes in serum levels could be involved in the physiological changes induced by gravity unloading. A collective evaluation of serum, femur, and soleus muscle proteome data of spaceflight mice also showed 24 serum proteins, including Igfbp5, Igfbp3, and Postn, whose levels could be associated with biological changes induced by microgravity. This study examined serum proteome profiles in response to gravity unloading, and may help deepen our understanding of microgravity adaptation mechanisms during prolonged spaceflight missions.
Collapse
Affiliation(s)
- Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Yusuke Nakai
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Yoko Ino
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Tomoko Akiyama
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Kayano Moriyama
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Takashi Ohira
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | | | - Yutaka Inaba
- Department of Orthopaedic Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Ken Kumagai
- Department of Orthopaedic Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Akihide Ryo
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Hisashi Hirano
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| |
Collapse
|
70
|
Rudolf AM, Hood WR. Mitochondrial stress in the spaceflight environment. Mitochondrion 2024; 76:101855. [PMID: 38403094 DOI: 10.1016/j.mito.2024.101855] [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: 07/08/2023] [Revised: 02/12/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
Space is a challenging environment that deregulates individual homeostasis. The main external hazards associated with spaceflight include ionizing space radiation, microgravity, isolation and confinement, distance from Earth, and hostile environment. Characterizing the biological responses to spaceflight environment is essential to validate the health risks, and to develop effective protection strategies. Mitochondria energetics is a key mechanism underpinning many physiological, ecological and evolutionary processes. Moreover, mitochondrial stress can be considered one of the fundamental features of space travel. So, we attempt to synthesize key information regarding the extensive effects of spaceflight on mitochondria. In summary, mitochondria are affected by all of the five main hazards of spaceflight at multiple levels, including their morphology, respiratory function, protein, and genetics, in various tissues and organ systems. We emphasize that investigating mitochondrial biology in spaceflight conditions should become the central focus of research on the impacts of spaceflight on human health, as this approach will help resolve numerous challenges of space health and combat several health disorders associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Agata M Rudolf
- Department of Biological Sciences, Auburn University, Auburn, AL, USA; Space Technology Centre, AGH University of Science and Technology, Krakow, Poland.
| | - Wendy R Hood
- Department of Biological Sciences, Auburn University, Auburn, AL, USA
| |
Collapse
|
71
|
Zhang J, Wang X, Fu Z, Xing C, Wang Z, Yang H, Li J, Liu M, Dong L, Zhang X, Li Y, Wang J, Long J, Liu J, Wang S, Li J, Gao F. Long-term simulated microgravity fosters carotid aging-like changes via Piezo1. Cardiovasc Res 2024; 120:548-559. [PMID: 38271270 DOI: 10.1093/cvr/cvae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/05/2023] [Accepted: 11/30/2023] [Indexed: 01/27/2024] Open
Abstract
AIMS Elucidating the impacts of long-term spaceflight on cardiovascular health is urgently needed in face of the rapid development of human space exploration. Recent reports including the NASA Twins Study on vascular deconditioning and aging of astronauts in spaceflight are controversial. The aims of this study were to elucidate whether long-term microgravity promotes vascular aging and the underlying mechanisms. METHODS AND RESULTS Hindlimb unloading (HU) by tail suspension was used to simulate microgravity in rats and mice. The dynamic changes of carotid stiffness in rats during 8 weeks of HU were determined. Simulated microgravity led to carotid artery aging-like changes as evidenced by increased stiffness, thickness, fibrosis, and elevated senescence biomarkers in the HU rats. Specific deletion of the mechanotransducer Piezo1 in vascular smooth muscles significantly blunted these aging-like changes in mice. Mechanistically, mechanical stretch-induced activation of Piezo1 elevated microRNA-582-5p in vascular smooth muscle cells, with resultant enhanced synthetic cell phenotype and increased collagen deposition via PTEN/PI3K/Akt signalling. Importantly, inhibition of miRNA-582-5p alleviated carotid fibrosis and stiffness not only in HU rats but also in aged rats. CONCLUSIONS Long-term simulated microgravity induces carotid aging-like changes via the mechanotransducer Piezo1-initiated and miRNA-mediated mechanism.
Collapse
MESH Headings
- Animals
- Aging/metabolism
- Aging/pathology
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Carotid Arteries/physiopathology
- Cells, Cultured
- Disease Models, Animal
- Fibrosis
- Hindlimb Suspension
- Ion Channels/metabolism
- Ion Channels/genetics
- Mechanotransduction, Cellular/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- MicroRNAs/metabolism
- MicroRNAs/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Phosphatidylinositol 3-Kinases/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- PTEN Phosphohydrolase/metabolism
- PTEN Phosphohydrolase/genetics
- Rats, Sprague-Dawley
- Signal Transduction
- Time Factors
- Vascular Remodeling
- Vascular Stiffness
- Weightlessness Simulation
Collapse
Affiliation(s)
- Jiaxin Zhang
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, China
| | - Xinpei Wang
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Zihao Fu
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Changyang Xing
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhen Wang
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Hongyan Yang
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Jiahui Li
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Meijie Liu
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Ling Dong
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Xing Zhang
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Yongzhi Li
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Jiaping Wang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Jiangang Long
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jiankang Liu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Shengpeng Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Jia Li
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
- Key Laboratory of Hazard Assessment and Control in Special Operational Environment of Ministry of Education, School of Public Health, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| | - Feng Gao
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, 169 Changlexi Road, Xi'an 710032, China
| |
Collapse
|
72
|
Zhang Y, Zhao L, Sun Y. Using single-sample networks to identify the contrasting patterns of gene interactions and reveal the radiation dose-dependent effects in multiple tissues of spaceflight mice. NPJ Microgravity 2024; 10:45. [PMID: 38575629 PMCID: PMC10995210 DOI: 10.1038/s41526-024-00383-7] [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: 11/02/2023] [Accepted: 03/08/2024] [Indexed: 04/06/2024] Open
Abstract
Transcriptome profiles are sensitive to space stressors and serve as valuable indicators of the biological effects during spaceflight. Herein, we transformed the expression profiles into gene interaction patterns by single-sample networks (SSNs) and performed the integrated analysis on the 301 spaceflight and 290 ground control samples, which were obtained from the GeneLab platform. Specifically, an individual SSN was established for each sample. Based on the topological structures of 591 SSNs, the differentially interacted genes (DIGs) were identified between spaceflights and ground controls. The results showed that spaceflight disrupted the gene interaction patterns in mice and resulted in significant enrichment of biological processes such as protein/amino acid metabolism and nucleic acid (DNA/RNA) metabolism (P-value < 0.05). We observed that the mice exposed to radiation doses within the three intervals (4.66-7.14, 7.592-8.295, 8.49-22.099 mGy) exhibited similar gene interaction patterns. Low and medium doses resulted in changes to the circadian rhythm, while the damaging effects on genetic material became more pronounced in higher doses. The gene interaction patterns in response to space stressors varied among different tissues, with the spleen, lung, and skin being the most responsive to space radiation (P-value < 0.01). The changes observed in gene networks during spaceflight conditions might contribute to the development of various diseases, such as mental disorders, depression, and metabolic disorders, among others. Additionally, organisms activated specific gene networks in response to virus reactivation. We identified several hub genes that were associated with circadian rhythms, suggesting that spaceflight could lead to substantial circadian rhythm dysregulation.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, 116026, Dalian, Liaoning, China
| | - Lei Zhao
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, 116026, Dalian, Liaoning, China.
| | - Yeqing Sun
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, 116026, Dalian, Liaoning, China.
| |
Collapse
|
73
|
Sharma S, Risen S, Gilberto VS, Boland S, Chatterjee A, Moreno JA, Nagpal P. Targeted-Neuroinflammation Mitigation Using Inflammasome-Inhibiting Nanoligomers is Therapeutic in an Experimental Autoimmune Encephalomyelitis Mouse Model. ACS Chem Neurosci 2024; 15:1596-1608. [PMID: 38526238 DOI: 10.1021/acschemneuro.4c00024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024] Open
Abstract
Multiple sclerosis (MS) is a debilitating autoimmune disease that impacts millions of patients worldwide, disproportionately impacting women (4:1), and often presenting at highly productive stages of life. This disease affects the spinal cord and brain and is characterized by severe neuroinflammation, demyelination, and subsequent neuronal damage, resulting in symptoms like loss of mobility. While untargeted and pan-immunosuppressive therapies have proven to be disease-modifying and manage (or prolong the time between) symptoms in many patients, a significant fraction are unable to achieve remission. Recent work has suggested that targeted neuroinflammation mitigation through selective inflammasome inhibition can offer relief to patients while preserving key components of immune function. Here, we show a screening of potential therapeutic targets using inflammasome-inhibiting Nanoligomers (NF-κB1, TNFR1, TNF-α, IL-6) that meet or far-exceed commercially available small-molecule counterparts like ruxolitinib, MCC950, and deucravacitinib. Using the human brain organoid model, top Nanoligomer combinations (NF-κB1 + TNFR1: NI111, and NF-κB1 + NLRP3: NI112) were shown to significantly reduce neuroinflammation without any observable negative impact on organoid function. Further testing of these top Nanoligomer combinations in an aggressive experimental autoimmune encephalomyelitis (EAE) mouse model for MS using intraperitoneal (IP) injections showed that NF-κB1 and NLRP3 targeting Nanoligomer combination NI112 rescues mice without observable loss of mobility or disability, minimal inflammation in brain and spinal cord histology, and minimal to no immune cell infiltration of the spinal cord and no demyelination, similar to or at par with mice that received no EAE injections (negative control). Mice receiving NI111 (NF-κB1 + TNFR1) also showed reduced neuroinflammation compared to saline (sham)-treated EAE mice and at par/similar to other inflammasome-inhibiting small molecule treatments, although it was significantly higher than NI112 leading to subsequent worsening clinical outcomes. Furthermore, treatment with an oral formulation of NI112 at lower doses showed a significant reduction in EAE severity, albeit with higher variance owing to administration and formulation/fill-and-finish variability. Overall, these results point to the potential of further development and testing of these inflammasome-targeting Nanoliogmers as an effective neuroinflammation treatment for multiple neurodegenerative diseases and potentially benefit several patients suffering from such debilitating autoimmune diseases like MS.
Collapse
Affiliation(s)
- Sadhana Sharma
- Sachi Bio, Colorado Technology Center, 685 S Arthur Avenue, Louisville, Colorado 80027, United States
| | - Sydney Risen
- Environmental & Radiological Health Sciences, and Brain Research Center, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Vincenzo S Gilberto
- Sachi Bio, Colorado Technology Center, 685 S Arthur Avenue, Louisville, Colorado 80027, United States
| | - Sean Boland
- Environmental & Radiological Health Sciences, and Brain Research Center, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Anushree Chatterjee
- Sachi Bio, Colorado Technology Center, 685 S Arthur Avenue, Louisville, Colorado 80027, United States
| | - Julie A Moreno
- Environmental & Radiological Health Sciences, and Brain Research Center, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Prashant Nagpal
- Sachi Bio, Colorado Technology Center, 685 S Arthur Avenue, Louisville, Colorado 80027, United States
| |
Collapse
|
74
|
Mason CE, Sierra MA, Feng HJ, Bailey SM. Telomeres and aging: on and off the planet! Biogerontology 2024; 25:313-327. [PMID: 38581556 PMCID: PMC10998805 DOI: 10.1007/s10522-024-10098-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 04/08/2024]
Abstract
Improving human healthspan in our rapidly aging population has never been more imperative. Telomeres, protective "caps" at the ends of linear chromosomes, are essential for maintaining genome stability of eukaryotic genomes. Due to their physical location and the "end-replication problem" first envisioned by Dr. Alexey Olovnikov, telomeres shorten with cell division, the implications of which are remarkably profound. Telomeres are hallmarks and molecular drivers of aging, as well as fundamental integrating components of the cumulative effects of genetic, lifestyle, and environmental factors that erode telomere length over time. Ongoing telomere attrition and the resulting limit to replicative potential imposed by cellular senescence serves a powerful tumor suppressor function, and also underlies aging and a spectrum of age-related degenerative pathologies, including reduced fertility, dementias, cardiovascular disease and cancer. However, very little data exists regarding the extraordinary stressors and exposures associated with long-duration space exploration and eventual habitation of other planets, nor how such missions will influence telomeres, reproduction, health, disease risk, and aging. Here, we briefly review our current understanding, which has advanced significantly in recent years as a result of the NASA Twins Study, the most comprehensive evaluation of human health effects associated with spaceflight ever conducted. Thus, the Twins Study is at the forefront of personalized space medicine approaches for astronauts and sets the stage for subsequent missions. We also extrapolate from current understanding to future missions, highlighting potential biological and biochemical strategies that may enable human survival, and consider the prospect of longevity in the extreme environment of space.
Collapse
Affiliation(s)
- Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA
| | - Maria A Sierra
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional Computational Biology & Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Henry J Feng
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Susan M Bailey
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
75
|
Wubshet NH, Cai G, Chen SJ, Sullivan M, Reeves M, Mays D, Harrison M, Varnado P, Yang B, Arreguin-Martinez E, Qu Y, Lin SS, Duran P, Aguilar C, Giza S, Clements T, Liu AP. Cellular mechanotransduction of human osteoblasts in microgravity. NPJ Microgravity 2024; 10:35. [PMID: 38514677 PMCID: PMC10957960 DOI: 10.1038/s41526-024-00386-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/13/2024] [Indexed: 03/23/2024] Open
Abstract
Astronauts experience significant and rapid bone loss as a result of an extended stay in space, making the International Space Station (ISS) the perfect laboratory for studying osteoporosis due to the accelerated nature of bone loss on the ISS. This prompts the question, how does the lack of load due to zero-gravity propagate to bone-forming cells, human fetal osteoblasts (hFOBs), altering their maturation to mineralization? Here, we aim to study the mechanotransduction mechanisms by which bone loss occurs in microgravity. Two automated experiments, microfluidic chips capable of measuring single-cell mechanics via aspiration and cell spheroids incubated in pressure-controlled chambers, were each integrated into a CubeLab deployed to the ISS National Laboratory. For the first experiment, we report protrusion measurements of aspirated cells after exposure to microgravity at the ISS and compare these results to ground control conducted inside the CubeLab. We found slightly elongated protrusions for space samples compared to ground samples indicating softening of hFOB cells in microgravity. In the second experiment, we encapsulated osteoblast spheroids in collagen gel and incubated the samples in pressure-controlled chambers. We found that microgravity significantly reduced filamentous actin levels in the hFOB spheroids. When subjected to pressure, the spheroids exhibited increased pSMAD1/5/9 expression, regardless of the microgravity condition. Moreover, microgravity reduced YAP expression, while pressure increased YAP levels, thus restoring YAP expression for spheroids in microgravity. Our study provides insights into the influence of microgravity on the mechanical properties of bone cells and the impact of compressive pressure on cell signaling in space.
Collapse
Affiliation(s)
- Nadab H Wubshet
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Grace Cai
- Applied Physics Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Samuel J Chen
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | | | | | | | | | - Benjamin Yang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Yunjia Qu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shan-Shan Lin
- Applied Physics Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Pamela Duran
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Carlos Aguilar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | | | - Allen P Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Applied Physics Program, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biophysics, University of Michigan, Ann Arbor, MI, 48109, USA.
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
76
|
Archer SN, Möller-Levet C, Bonmatí-Carrión MÁ, Laing EE, Dijk DJ. Extensive dynamic changes in the human transcriptome and its circadian organization during prolonged bed rest. iScience 2024; 27:109331. [PMID: 38487016 PMCID: PMC10937834 DOI: 10.1016/j.isci.2024.109331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/11/2023] [Accepted: 02/20/2024] [Indexed: 03/17/2024] Open
Abstract
Physiological and molecular processes including the transcriptome change across the 24-h day, driven by molecular circadian clocks and behavioral and systemic factors. It is not known how the temporal organization of the human transcriptome responds to a long-lasting challenge. This may, however, provide insights into adaptation, disease, and recovery. We investigated the human 24-h time series transcriptome in 20 individuals during a 90-day constant bed rest protocol. We show that the protocol affected 91% of the transcriptome with 76% of the transcriptome still affected after 10 days of recovery. Dimensionality-reduction approaches revealed that many affected transcripts were associated with mRNA translation and immune function. The number, amplitude, and phase of rhythmic transcripts, including clock genes, varied significantly across the challenge. These findings of long-lasting changes in the temporal organization of the transcriptome have implications for understanding the mechanisms underlying health consequences of conditions such as microgravity and bed rest.
Collapse
Affiliation(s)
- Simon N. Archer
- Surrey Sleep Research Centre, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
| | - Carla Möller-Levet
- Bioinformatics Core Facility, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
| | - María-Ángeles Bonmatí-Carrión
- Surrey Sleep Research Centre, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
- Chronobiology Laboratory, Department of Physiology, University of Murcia, Murcia, Spain
- Ciber Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Emma E. Laing
- Department of Microbiology, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
| | - Derk-Jan Dijk
- Surrey Sleep Research Centre, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
- UK Dementia Research Institute Care Research & Technology Centre, Imperial College London & University of Surrey, Guildford, UK
| |
Collapse
|
77
|
Zhang Y, Du X, Zhao L, Sun Y. Construction of dose prediction model and identification of sensitive genes for space radiation based on single-sample networks under spaceflight conditions. Int J Radiat Biol 2024; 100:777-790. [PMID: 38471034 DOI: 10.1080/09553002.2024.2327393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/01/2024] [Indexed: 03/14/2024]
Abstract
PURPOSE To identify sensitive genes for space radiation, we integrated the transcriptomic samples of spaceflight mice from GeneLab and predicted the radiation doses absorbed by individuals in space. METHODS AND MATERIALS A single-sample network (SSN) for each individual sample was constructed. Then, using machine learning and genetic algorithms, we built the regression models to predict the absorbed dose equivalent based on the topological structure of SSNs. Moreover, we analyzed the SSNs from each tissue and compared the similarities and differences among them. RESULTS Our model exhibited excellent performance with the following metrics: R 2 = 0.980 , MSE = 6.74 e - 04 , and the Pearson correlation coefficient of 0.990 (p value <.0001) between predicted and actual values. We identified 20 key genes, the majority of which had been proven to be associated with radiation. However, we uniquely established them as space radiation sensitive genes for the first time. Through further analysis of the SSNs, we discovered that the different tissues exhibited distinct mechanisms in response to space stressors. CONCLUSIONS The topology structures of SSNs effectively predicted radiation doses under spaceflight conditions, and the SSNs revealed the gene regulatory patterns within the organisms under space stressors.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning, China
| | - Xiaohui Du
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning, China
| | - Lei Zhao
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning, China
| | - Yeqing Sun
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning, China
| |
Collapse
|
78
|
Wubshet NH, Cai G, Chen SJ, Sullivan M, Reeves M, Mays D, Harrison M, Varnado P, Yang B, Arreguin-Martinez E, Qu Y, Lin SS, Duran P, Aguilar C, Giza S, Clements T, Liu AP. Cellular mechanotransduction of human osteoblasts in microgravity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.03.583164. [PMID: 38464311 PMCID: PMC10925314 DOI: 10.1101/2024.03.03.583164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Astronauts experience significant and rapid bone loss as a result of an extended stay in space, making the International Space Station (ISS) the perfect laboratory for studying osteoporosis due to the accelerated nature of bone loss on the ISS. This prompts the question, how does the lack of load due to zero-gravity propagate to bone-forming cells, human fetal osteoblasts (hFOBs), altering their maturation to mineralization? Here, we aim to study the mechanotransduction mechanisms by which bone loss occurs in microgravity. Two automated experiments, 4 microfluidic chips capable of measuring single-cell mechanics of hFOBs via aspiration and cell spheroids incubated in pressure-controlled chambers, were each integrated into a CubeLab deployed to the ISS National Laboratory. For the first experiment, we report protrusion measurements of aspirated cells after exposure to microgravity at the ISS and compare these results to ground control conducted inside the CubeLab. Our analysis revealed slightly elongated protrusions for space samples compared to ground samples indicating softening of hFOB cells in microgravity. In the second experiment, we encapsulated osteoblast spheroids in collagen gel and incubated the samples in pressure-controlled chambers. We found that microgravity significantly reduced filamentous actin levels in the hFOB spheroids. When subjected to pressure, the spheroids exhibited increased pSMAD1/5/9 expression, regardless of the microgravity condition. Moreover, microgravity reduced YAP expression, while pressure increased YAP levels, thus restoring YAP expression for spheroids in microgravity. Our study provides insights into the influence of microgravity on the mechanical properties of bone cells and the impact of compressive pressure on cell behavior and signaling in space.
Collapse
Affiliation(s)
- Nadab H. Wubshet
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Grace Cai
- Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| | - Samuel J. Chen
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | | - Benjamin Yang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | - Yunjia Qu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Shan-Shan Lin
- Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| | - Pamela Duran
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Carlos Aguilar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Allen P. Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
- Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
79
|
Zhao L, Li Z, Huang B, Mi D, Xu D, Sun Y. Integrating evolutionarily conserved mechanism of response to radiation for exploring novel Caenorhabditis elegans radiation-responsive genes for estimation of radiation dose associated with spaceflight. CHEMOSPHERE 2024; 351:141148. [PMID: 38211791 DOI: 10.1016/j.chemosphere.2024.141148] [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: 09/18/2023] [Revised: 12/09/2023] [Accepted: 01/06/2024] [Indexed: 01/13/2024]
Abstract
During space exploration, space radiation is widely recognized as an inescapable perilous stressor, owing to its capacity to induce genomic DNA damage and escalate the likelihood of detrimental health outcomes. Rapid and reliable estimation of space radiation dose holds paramount significance in accurately assessing the health risks associated with spaceflight. However, the identification of space radiation-responsive genes, with their potential to serve as early indicators for diagnosing radiation dose associated with spaceflight, continues to pose a significant challenge. In this study, based on the evolutionarily conserved mechanism of radiation response, an in silico analysis method of homologous comparison was performed to identify the Caenorhabditis elegans orthologues of human radiation-responsive genes with possible roles in the major processes of response to radiation, and thereby to explore the potential C. elegans radiation-responsive genes for evaluating the levels of space radiation exposure. The results showed that there were 60 known C. elegans radiation-responsive genes and 211 C. elegans orthologues of human radiation-responsive genes implicated in the major processes of response to radiation. Through an investigation of all available transcriptomic datasets obtained from space-flown C. elegans, it was observed that the expression levels of the majority of these putative C. elegans radiation-responsive genes identified in this study were notably changed across various spaceflight conditions. Furthermore, this study indicated that within the identified genes, 19 known C. elegans radiation-responsive genes and 40 newly identified C. elegans orthologues of human radiation-responsive genes exhibited a remarkable positive correlation with the duration of spaceflight. Moreover, a noteworthy presence of substantial multi-collinearity among the majority of these identified genes was observed. This observation lends support to the possibility of treating each identified gene as an independent indicator of radiation dose in space. Ultimately, a subset of 15 potential radiation-responsive genes was identified, presenting the most promising indicators for estimation of radiation dose associated with spaceflight in C. elegans.
Collapse
Affiliation(s)
- Lei Zhao
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, 116026, Liaoning, China.
| | - Zejun Li
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, 116026, Liaoning, China
| | - Baohang Huang
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, 116026, Liaoning, China
| | - Dong Mi
- College of Science, Dalian Maritime University, Dalian, 116026, Liaoning, China
| | - Dan Xu
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, 116026, Liaoning, China
| | - Yeqing Sun
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, 116026, Liaoning, China.
| |
Collapse
|
80
|
Garcia-Medina JS, Sienkiewicz K, Narayanan SA, Overbey EG, Grigorev K, Ryon KA, Burke M, Proszynski J, Tierney B, Schmidt CM, Mencia-Trinchant N, Klotz R, Ortiz V, Foox J, Chin C, Najjar D, Matei I, Chan I, Cruchaga C, Kleinman A, Kim J, Lucaci A, Loy C, Mzava O, De Vlaminck I, Singaraju A, Taylor LE, Schmidt JC, Schmidt MA, Blease K, Moreno J, Boddicker A, Zhao J, Lajoie B, Altomare A, Kruglyak S, Levy S, Yu M, Hassane DC, Bailey SM, Bolton K, Mateus J, Mason CE. Genome and clonal hematopoiesis stability contrasts with immune, cfDNA, mitochondrial, and telomere length changes during short duration spaceflight. PRECISION CLINICAL MEDICINE 2024; 7:pbae007. [PMID: 38634106 PMCID: PMC11022651 DOI: 10.1093/pcmedi/pbae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/24/2024] [Indexed: 04/19/2024] Open
Abstract
Background The Inspiration4 (I4) mission, the first all-civilian orbital flight mission, investigated the physiological effects of short-duration spaceflight through a multi-omic approach. Despite advances, there remains much to learn about human adaptation to spaceflight's unique challenges, including microgravity, immune system perturbations, and radiation exposure. Methods To provide a detailed genetics analysis of the mission, we collected dried blood spots pre-, during, and post-flight for DNA extraction. Telomere length was measured by quantitative PCR, while whole genome and cfDNA sequencing provided insight into genomic stability and immune adaptations. A robust bioinformatic pipeline was used for data analysis, including variant calling to assess mutational burden. Result Telomere elongation occurred during spaceflight and shortened after return to Earth. Cell-free DNA analysis revealed increased immune cell signatures post-flight. No significant clonal hematopoiesis of indeterminate potential (CHIP) or whole-genome instability was observed. The long-term gene expression changes across immune cells suggested cellular adaptations to the space environment persisting months post-flight. Conclusion Our findings provide valuable insights into the physiological consequences of short-duration spaceflight, with telomere dynamics and immune cell gene expression adapting to spaceflight and persisting after return to Earth. CHIP sequencing data will serve as a reference point for studying the early development of CHIP in astronauts, an understudied phenomenon as previous studies have focused on career astronauts. This study will serve as a reference point for future commercial and non-commercial spaceflight, low Earth orbit (LEO) missions, and deep-space exploration.
Collapse
Affiliation(s)
- J Sebastian Garcia-Medina
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, 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
| | - Karolina Sienkiewicz
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, 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
| | - S Anand Narayanan
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL 32306, USA
| | - Eliah G Overbey
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, 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
- BioAstra Inc, New York, NY, USA
| | - Kirill Grigorev
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, 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
| | - Krista A Ryon
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Marissa Burke
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Jacqueline Proszynski
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Braden Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, 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
| | - Caleb M Schmidt
- Sovaris Aerospace, Boulder, CO 80302, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, CO 80302, USA
- Department of Systems Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Nuria Mencia-Trinchant
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Remi Klotz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Veronica Ortiz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jonathan Foox
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, 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
| | - Christopher Chin
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, 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
- BioAstra Inc, New York, NY, USA
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, NY 10021, USA
- WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Deena Najjar
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Irenaeus Chan
- Washington University St. Louis Oncology Division, St. Louis, MO 63100, USA
| | - Carlos Cruchaga
- Washington University St. Louis Oncology Division, St. Louis, MO 63100, USA
| | - Ashley Kleinman
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, 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
| | - Alexander Lucaci
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Conor Loy
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Omary Mzava
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Anvita Singaraju
- Department of Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Lynn E Taylor
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Julian C Schmidt
- Sovaris Aerospace, Boulder, CO 80302, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, CO 80302, USA
| | - Michael A Schmidt
- Sovaris Aerospace, Boulder, CO 80302, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, CO 80302, USA
| | | | - Juan Moreno
- Element Biosciences, San Diego, CA 10055, USA
| | | | - Junhua Zhao
- Element Biosciences, San Diego, CA 10055, USA
| | | | | | | | - Shawn Levy
- Element Biosciences, San Diego, CA 10055, USA
| | - Min Yu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Duane C Hassane
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Susan M Bailey
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO 80523, USA
| | - Kelly Bolton
- Washington University St. Louis Oncology Division, St. Louis, MO 63100, USA
| | - Jaime Mateus
- Space Exploration Technologies Corporation, Hawthorne, CA 90250, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, 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
- BioAstra Inc, New York, NY, USA
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, NY 10021, USA
- WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
81
|
Buescher FM, Schmitz MT, Frett T, Kramme J, de Boni L, Elmenhorst EM, Mulder E, Moestl S, Heusser K, Frings-Meuthen P, Jordan J, Rittweger J, Pesta D. Effects of 30 days bed rest and exercise countermeasures on PBMC bioenergetics. Acta Physiol (Oxf) 2024; 240:e14102. [PMID: 38294173 DOI: 10.1111/apha.14102] [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: 11/10/2023] [Revised: 12/27/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024]
Abstract
AIM Altered mitochondrial function across various tissues is a key determinant of spaceflight-induced physical deconditioning. In comparison to tissue biopsies, blood cell bioenergetics holds promise as a systemic and more readily accessible biomarker, which was evaluated during head-down tilt bed rest (HDTBR), an established ground-based analog for spaceflight-induced physiological changes in humans. More specifically, this study explored the effects of HDTBR and an exercise countermeasure on mitochondrial respiration in peripheral blood mononuclear cells (PBMCs). METHODS We subjected 24 healthy participants to a strict 30-day HDTBR protocol. The control group (n = 12) underwent HDTBR only, while the countermeasure group (n = 12) engaged in regular supine cycling exercise followed by veno-occlusive thigh cuffs post-exercise for 6 h. We assessed routine blood parameters 14 days before bed rest, the respiratory capacity of PBMCs via high-resolution respirometry, and citrate synthase activity 2 days before and at day 30 of bed rest. We confirmed PBMC composition by flow cytometry. RESULTS The change of the PBMC maximal oxidative phosphorylation capacity (OXPHOS) amounted to an 11% increase in the countermeasure group, while it decreased by 10% in the control group (p = 0.04). The limitation of OXPHOS increased in control only while other respiratory states were not affected by either intervention. Correlation analysis revealed positive associations between white blood cells, lymphocytes, and basophils with PBMC bioenergetics in both groups. CONCLUSION This study reveals that a regular exercise countermeasure has a positive impact on PBMC mitochondrial function, confirming the potential application of blood cell bioenergetics for human spaceflight.
Collapse
Affiliation(s)
- F-M Buescher
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
| | - M T Schmitz
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
- Institute of Medical Biometry, Informatics and Epidemiology, Medical Faculty, University of Bonn, Bonn, Germany
| | - T Frett
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
| | - J Kramme
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
| | - L de Boni
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
| | - E M Elmenhorst
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
| | - E Mulder
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
| | - S Moestl
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
| | - K Heusser
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
| | - P Frings-Meuthen
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
| | - J Jordan
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
- Medical Faculty, University of Cologne, Cologne, Germany
| | - J Rittweger
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
| | - D Pesta
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
- Medical Faculty, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
82
|
Sarma MS, Shelhamer M. The human biology of spaceflight. Am J Hum Biol 2024; 36:e24048. [PMID: 38337152 PMCID: PMC10940193 DOI: 10.1002/ajhb.24048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
To expand the human exploration footprint and reach Mars in the 2030s, we must explore how humans survive and thrive in demanding, unusual, and novel ecologies (i.e., extreme environments). In the extreme conditions encountered during human spaceflight, there is a need to understand human functioning and response in a more rigorous theoretically informed way. Current models of human performance in space-relevant environments and human space science are often operationally focused, with emphasis on acute physiological or behavioral outcomes. However, integrating current perspectives in human biology allows for a more holistic and complete understanding of how humans function over a range of time in an extreme environment. Here, we show how the use of evolution-informed frameworks (i.e., models of life history theory to organize the adaptive pressures of spaceflight and biocultural perspectives) coupled with the use of mixed-methodological toolkits can shape models that better encompass the scope of biobehavioral human adjustment to long-duration space travel and extra-terrestrial habitation. Further, we discuss how we can marry human biology perspectives with the rigorous programmatic structures developed for spaceflight to model other unknown and nascent extremes.
Collapse
Affiliation(s)
- Mallika S. Sarma
- Human Spaceflight Lab, Johns Hopkins School of Medicine, Baltimore, MD 21215
| | - Mark Shelhamer
- Human Spaceflight Lab, Johns Hopkins School of Medicine, Baltimore, MD 21215
| |
Collapse
|
83
|
Campisi M, Cannella L, Pavanello S. Cosmic chronometers: Is spaceflight a catalyst for biological ageing? Ageing Res Rev 2024; 95:102227. [PMID: 38346506 DOI: 10.1016/j.arr.2024.102227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/05/2024] [Accepted: 02/06/2024] [Indexed: 02/22/2024]
Abstract
Astronauts returning from space missions often exhibit health issues mirroring age-related conditions, suggesting spaceflight as a potential driver of biological ageing and age-related diseases. To unravel the underlying mechanisms of these conditions, this comprehensive review explores the impact of the space "exposome" on the twelve hallmarks of ageing. Through a meticulous analysis encompassing both space environments and terrestrial analogs, we aim to decipher how different conditions influence ageing hallmarks. Utilizing PubMed, we identified 189 studies and 60 meet screening criteria. Research on biological ageing in space has focused on genomic instability, chronic inflammation, and deregulated nutrient sensing. Spaceflight consistently induces genomic instability, linked to prolonged exposure to ionizing radiation, triggers pro-inflammatory and immune alterations, resembling conditions in isolated simulations. Nutrient sensing pathways reveal increased systemic insulin-like growth-factor-1. Microbiome studies indicate imbalances favoring opportunistic species during spaceflight. Telomere dynamics present intriguing patterns, with lengthening during missions and rapid shortening upon return. Despite a pro-ageing trend, some protective mechanisms emerge. Countermeasures, encompassing dietary adjustments, prebiotics, postbiotics, symbiotics, tailored exercises, meditation, and anti-inflammatory supplements, exhibit potential. Spaceflight's impact on ageing is intricate, with diverse findings challenging established beliefs. Multidisciplinary studies provide guidance for future research in this field.
Collapse
Affiliation(s)
- Manuela Campisi
- Occupational Medicine, Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Luana Cannella
- Occupational Medicine, Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Sofia Pavanello
- Occupational Medicine, Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy.
| |
Collapse
|
84
|
Schoenrock B, Muckelt PE, Hastermann M, Albracht K, MacGregor R, Martin D, Gunga HC, Salanova M, Stokes MJ, Warner MB, Blottner D. Muscle stiffness indicating mission crew health in space. Sci Rep 2024; 14:4196. [PMID: 38378866 PMCID: PMC10879143 DOI: 10.1038/s41598-024-54759-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/16/2024] [Indexed: 02/22/2024] Open
Abstract
Muscle function is compromised by gravitational unloading in space affecting overall musculoskeletal health. Astronauts perform daily exercise programmes to mitigate these effects but knowing which muscles to target would optimise effectiveness. Accurate inflight assessment to inform exercise programmes is critical due to lack of technologies suitable for spaceflight. Changes in mechanical properties indicate muscle health status and can be measured rapidly and non-invasively using novel technology. A hand-held MyotonPRO device enabled monitoring of muscle health for the first time in spaceflight (> 180 days). Greater/maintained stiffness indicated countermeasures were effective. Tissue stiffness was preserved in the majority of muscles (neck, shoulder, back, thigh) but Tibialis Anterior (foot lever muscle) stiffness decreased inflight vs. preflight (p < 0.0001; mean difference 149 N/m) in all 12 crewmembers. The calf muscles showed opposing effects, Gastrocnemius increasing in stiffness Soleus decreasing. Selective stiffness decrements indicate lack of preservation despite daily inflight countermeasures. This calls for more targeted exercises for lower leg muscles with vital roles as ankle joint stabilizers and in gait. Muscle stiffness is a digital biomarker for risk monitoring during future planetary explorations (Moon, Mars), for healthcare management in challenging environments or clinical disorders in people on Earth, to enable effective tailored exercise programmes.
Collapse
Affiliation(s)
- Britt Schoenrock
- NeuroMuscular System & Signaling Group, Berlin Center of Space Medicine and Extreme Environments, 10115 Berlin, Germany, Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany, 10115, Berlin, Germany
| | - Paul E Muckelt
- School of Health Sciences, University of Southampton, Southampton, UK
| | - Maria Hastermann
- Experimental and Clinical Research Center (ECRC) and NeuroCure Clinical Research Center (NCRC), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | - Hans-Christian Gunga
- Institute of Physiology, Berlin Center of Space Medicine and Extreme Environments, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany, Berlin, Germany
| | - Michele Salanova
- NeuroMuscular System & Signaling Group, Berlin Center of Space Medicine and Extreme Environments, 10115 Berlin, Germany, Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany, 10115, Berlin, Germany
| | - Maria J Stokes
- School of Health Sciences, University of Southampton, Southampton, UK
| | - Martin B Warner
- School of Health Sciences, University of Southampton, Southampton, UK
| | - Dieter Blottner
- NeuroMuscular System & Signaling Group, Berlin Center of Space Medicine and Extreme Environments, 10115 Berlin, Germany, Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany, 10115, Berlin, Germany.
| |
Collapse
|
85
|
Barcenilla BB, Kundel I, Hall E, Hilty N, Ulianich P, Cook J, Turley J, Yerram M, Min JH, Castillo-González C, Shippen DE. Telomere dynamics and oxidative stress in Arabidopsis grown in lunar regolith simulant. FRONTIERS IN PLANT SCIENCE 2024; 15:1351613. [PMID: 38434436 PMCID: PMC10908177 DOI: 10.3389/fpls.2024.1351613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024]
Abstract
NASA envisions a future where humans establish a thriving colony on the Moon by 2050. Plants will be essential for this endeavor, but little is known about their adaptation to extraterrestrial bodies. The capacity to grow plants in lunar regolith would represent a major step towards this goal by minimizing the reliance on resources transported from Earth. Recent studies reveal that Arabidopsis thaliana can germinate and grow on genuine lunar regolith as well as on lunar regolith simulant. However, plants arrest in vegetative development and activate a variety of stress response pathways, most notably the oxidative stress response. Telomeres are hotspots for oxidative damage in the genome and a marker of fitness in many organisms. Here we examine A. thaliana growth on a lunar regolith simulant and the impact of this resource on plant physiology and on telomere dynamics, telomerase enzyme activity and genome oxidation. We report that plants successfully set seed and generate a viable second plant generation if the lunar regolith simulant is pre-washed with an antioxidant cocktail. However, plants sustain a higher degree of genome oxidation and decreased biomass relative to conventional Earth soil cultivation. Moreover, telomerase activity substantially declines and telomeres shorten in plants grown in lunar regolith simulant, implying that genome integrity may not be sustainable over the long-term. Overcoming these challenges will be an important goal in ensuring success on the lunar frontier.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Dorothy E. Shippen
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| |
Collapse
|
86
|
Tomsia M, Cieśla J, Śmieszek J, Florek S, Macionga A, Michalczyk K, Stygar D. Long-term space missions' effects on the human organism: what we do know and what requires further research. Front Physiol 2024; 15:1284644. [PMID: 38415007 PMCID: PMC10896920 DOI: 10.3389/fphys.2024.1284644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 01/22/2024] [Indexed: 02/29/2024] Open
Abstract
Space has always fascinated people. Many years have passed since the first spaceflight, and in addition to the enormous technological progress, the level of understanding of human physiology in space is also increasing. The presented paper aims to summarize the recent research findings on the influence of the space environment (microgravity, pressure differences, cosmic radiation, etc.) on the human body systems during short-term and long-term space missions. The review also presents the biggest challenges and problems that must be solved in order to extend safely the time of human stay in space. In the era of increasing engineering capabilities, plans to colonize other planets, and the growing interest in commercial space flights, the most topical issues of modern medicine seems to be understanding the effects of long-term stay in space, and finding solutions to minimize the harmful effects of the space environment on the human body.
Collapse
Affiliation(s)
- Marcin Tomsia
- Department of Forensic Medicine and Forensic Toxicology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Julia Cieśla
- School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Joanna Śmieszek
- School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Szymon Florek
- School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Agata Macionga
- School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Katarzyna Michalczyk
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Dominika Stygar
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
- SLU University Animal Hospital, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
87
|
Mortazavi SMJ, Said-Salman I, Mortazavi AR, El Khatib S, Sihver L. How the adaptation of the human microbiome to harsh space environment can determine the chances of success for a space mission to Mars and beyond. Front Microbiol 2024; 14:1237564. [PMID: 38390219 PMCID: PMC10881706 DOI: 10.3389/fmicb.2023.1237564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/05/2023] [Indexed: 02/24/2024] Open
Abstract
The ability of human cells to adapt to space radiation is essential for the well-being of astronauts during long-distance space expeditions, such as voyages to Mars or other deep space destinations. However, the adaptation of the microbiomes should not be overlooked. Microorganisms inside an astronaut's body, or inside the space station or other spacecraft, will also be exposed to radiation, which may induce resistance to antibiotics, UV, heat, desiccation, and other life-threatening factors. Therefore, it is essential to consider the potential effects of radiation not only on humans but also on their microbiomes to develop effective risk reduction strategies for space missions. Studying the human microbiome in space missions can have several potential benefits, including but not limited to a better understanding of the major effects space travel has on human health, developing new technologies for monitoring health and developing new radiation therapies and treatments. While radioadaptive response in astronauts' cells can lead to resistance against high levels of space radiation, radioadaptive response in their microbiome can lead to resistance against UV, heat, desiccation, antibiotics, and radiation. As astronauts and their microbiomes compete to adapt to the space environment. The microorganisms may emerge as the winners, leading to life-threatening situations due to lethal infections. Therefore, understanding the magnitude of the adaptation of microorganisms before launching a space mission is crucial to be able to develop effective strategies to mitigate the risks associated with radiation exposure. Ensuring the safety and well-being of astronauts during long-duration space missions and minimizing the risks linked with radiation exposure can be achieved by adopting this approach.
Collapse
Affiliation(s)
- Seyed Mohammad Javad Mortazavi
- Ionizing and non-ionizing radiation protection research center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ilham Said-Salman
- Department of Biological and Chemical Sciences, School of Arts & Sciences, Lebanese International University, Saida, Lebanon
- Department of Biological and Chemical Sciences, International University of Beirut, Beirut, Lebanon
| | | | - Sami El Khatib
- Department of Biomedical Sciences, School of Arts and Sciences, Lebanese International University, Beirut, Lebanon
- Center for Applied Mathematics and Bioinformatics (CAMB) at Gulf University for Science and Technology, Kuwait City, Kuwait
| | - Lembit Sihver
- Department of Radiation Dosimetry, Nuclear Physics Institute (NPI) of the Czech Academy of Sciences (CAS), Prague, Czechia
- Department of Radiation Physics, Technische Universität Wien Atominstitut, Vienna, Austria
| |
Collapse
|
88
|
Lecoq PE, Dupuis C, Mousset X, Benoit-Gonnin X, Peyrin JM, Aider JL. Influence of microgravity on spontaneous calcium activity of primary hippocampal neurons grown in microfluidic chips. NPJ Microgravity 2024; 10:15. [PMID: 38321051 PMCID: PMC10847089 DOI: 10.1038/s41526-024-00355-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 01/10/2024] [Indexed: 02/08/2024] Open
Abstract
The influence of variations of gravity, either hypergravity or microgravity, on the brain of astronauts is a major concern for long journeys in space, to the Moon or to Mars, or simply long-duration missions on the ISS (International Space Station). Monitoring brain activity, before and after ISS missions already demonstrated important and long term effects on the brains of astronauts. In this study, we focus on the influence of gravity variations at the cellular level on primary hippocampal neurons. A dedicated setup has been designed and built to perform live calcium imaging during parabolic flights. During a CNES (Centre National d'Etudes Spatiales) parabolic flight campaign, we were able to observe and monitor the calcium activity of 2D networks of neurons inside microfluidic devices during gravity changes over different parabolas. Our preliminary results clearly indicate a modification of the calcium activity associated to variations of gravity.
Collapse
Affiliation(s)
- Pierre-Ewen Lecoq
- PMMH, ESPCI Paris - PSL, Paris, 75005, France.
- Neurosciences Paris Seine IBPS, UMR8246, Inserm U1130, Sorbonne University, 4 Place Jussieu, Paris, 75005, France.
| | - Chloé Dupuis
- PMMH, ESPCI Paris - PSL, Paris, 75005, France
- Neurosciences Paris Seine IBPS, UMR8246, Inserm U1130, Sorbonne University, 4 Place Jussieu, Paris, 75005, France
| | - Xavier Mousset
- PMMH, ESPCI Paris - PSL, Paris, 75005, France
- Neurosciences Paris Seine IBPS, UMR8246, Inserm U1130, Sorbonne University, 4 Place Jussieu, Paris, 75005, France
| | | | - Jean-Michel Peyrin
- Neurosciences Paris Seine IBPS, UMR8246, Inserm U1130, Sorbonne University, 4 Place Jussieu, Paris, 75005, France.
| | | |
Collapse
|
89
|
Duscher AA, Vroom MM, Foster JS. Impact of modeled microgravity stress on innate immunity in a beneficial animal-microbe symbiosis. Sci Rep 2024; 14:2912. [PMID: 38316910 PMCID: PMC10844198 DOI: 10.1038/s41598-024-53477-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/31/2024] [Indexed: 02/07/2024] Open
Abstract
The innate immune response is the first line of defense for all animals to not only detect invading microbes and toxins but also sense and interface with the environment. One such environment that can significantly affect innate immunity is spaceflight. In this study, we explored the impact of microgravity stress on key elements of the NFκB innate immune pathway. The symbiosis between the bobtail squid Euprymna scolopes and its beneficial symbiont Vibrio fischeri was used as a model system under a simulated microgravity environment. The expression of genes associated with the NFκB pathway was monitored over time as the symbiosis progressed. Results revealed that although the onset of the symbiosis was the major driver in the differential expression of NFκB signaling, the stress of simulated low-shear microgravity also caused a dysregulation of expression. Several genes were expressed at earlier time points suggesting that elements of the E. scolopes NFκB pathway are stress-inducible, whereas expression of other pathway components was delayed. The results provide new insights into the role of NFκB signaling in the squid-vibrio symbiosis, and how the stress of microgravity negatively impacts the host immune response. Together, these results provide a foundation to develop mitigation strategies to maintain host-microbe homeostasis during spaceflight.
Collapse
Affiliation(s)
- Alexandrea A Duscher
- Department of Microbiology and Cell Science, Space Life Science Lab, University of Florida, Merritt Island, FL, 32953, USA
- Chesapeake Bay Governor's School, Warsaw, VA, 22572, USA
| | - Madeline M Vroom
- Department of Microbiology and Cell Science, Space Life Science Lab, University of Florida, Merritt Island, FL, 32953, USA
- Vaxxinity, Space Life Sciences Lab, Merritt Island, FL, 32953, USA
| | - Jamie S Foster
- Department of Microbiology and Cell Science, Space Life Science Lab, University of Florida, Merritt Island, FL, 32953, USA.
| |
Collapse
|
90
|
Gan X, Zhao J, Li S, Kan G, Zhang Y, Wang B, Zhang P, Ma X, Tian H, Liao M, Ju D, Xu S, Chen X, Guo J. Simulated space environmental factors of weightlessness, noise and low atmospheric pressure differentially affect the diurnal rhythm and the gut microbiome. LIFE SCIENCES IN SPACE RESEARCH 2024; 40:115-125. [PMID: 38245336 DOI: 10.1016/j.lssr.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/21/2023] [Accepted: 09/19/2023] [Indexed: 01/22/2024]
Abstract
The circadian clock extensively regulates physiology and behavior. In space, astronauts encounter many environmental factors that are dramatically different from those on Earth; however, the effects of these factors on circadian rhythms and the mechanisms remain largely unknown. The present study aimed to investigate the changes in the mouse diurnal rhythm and gut microbiome under simulated space capsule conditions, including microgravity, noise and low atmospheric pressure (LAP). Noise and LAP were loaded in the capsule while the conditions in the animal room remained constant. The mice in the capsule showed disturbed locomotor rhythms and faster adaptation to a 6-h phase advance. RNA sequencing of hypothalamus samples containing the suprachiasmatic nucleus (SCN) revealed that microgravity simulated by hind limb unloading (HU) and exposure to noise and LAP led to decreases in the quantities of differentially expressed genes (DEGs), including circadian clock genes. Changes in the rhythmicity of genes implicated in pathways of cardiovascular deconditioning and more concentrated phases were found under HU or noise and LAP. Furthermore, 16S rRNA sequencing revealed dysbiosis in the gut microbiome, and noise and LAP may repress the temporal discrepancy in the microbiome community structure induced by microgravity. Changes in diurnal oscillations were observed in a number of gut bacteria with critical physiological consequences on metabolism and immunodefense. We also found that the superimposition of noise and LAP may repress normal changes in global gene expression and adaptation in the gut microbiome. Our data demonstrate that in addition to microgravity, exposure to noise and LAP affect the robustness of circadian rhythms and the community structure of the gut microbiome, and these factors may interfere with each other in their adaptation to respective conditions. These findings are important for furthering our understanding of the alterations in circadian rhythms in the complex environment of space.
Collapse
Affiliation(s)
- Xihui Gan
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, School of Life Sciences, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Jianwei Zhao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Silin Li
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, School of Life Sciences, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Guanghan Kan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yin Zhang
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, School of Life Sciences, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Bo Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Peng Zhang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Xiaohong Ma
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, School of Life Sciences, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Hongni Tian
- National Institute of Biological Sciences, Beijing, China
| | - Meimei Liao
- National Institute of Biological Sciences, Beijing, China
| | - Dapeng Ju
- National Institute of Biological Sciences, Beijing, China
| | - Shuihong Xu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Xiaoping Chen
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China; National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China.
| | - Jinhu Guo
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, School of Life Sciences, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
91
|
Neikirk K, Stephens DC, Beasley HK, Marshall AG, Gaddy JA, Damo SM, Hinton A. Is space the final frontier for mitochondrial study? Biotechniques 2024; 76:46-51. [PMID: 38084381 DOI: 10.2144/btn-2023-0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Abstract
Tweetable abstract This perspective considers several avenues for future research on mitochondrial dynamics, stress, and DNA in outer space.
Collapse
Affiliation(s)
- Kit Neikirk
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Dominique C Stephens
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
- Department of Life & Physical Sciences, Fisk University, Nashville, TN 37208, USA
| | - Heather K Beasley
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Andrea G Marshall
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Jennifer A Gaddy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Steven M Damo
- Department of Life & Physical Sciences, Fisk University, Nashville, TN 37208, USA
| | - Antentor Hinton
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
92
|
Dontre AJ. Weighing the impact of microgravity on vestibular and visual functions. LIFE SCIENCES IN SPACE RESEARCH 2024; 40:51-61. [PMID: 38245348 DOI: 10.1016/j.lssr.2023.12.003] [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: 10/25/2023] [Revised: 12/03/2023] [Accepted: 12/27/2023] [Indexed: 01/22/2024]
Abstract
Numerous technological challenges have been overcome to realize human space exploration. As mission durations gradually lengthen, the next obstacle is a set of physical limitations. Extended exposure to microgravity poses multiple threats to various bodily systems. Two of these systems are of particular concern for the success of future space missions. The vestibular system includes the otolith organs, which are stimulated in gravity but unloaded in microgravity. This impairs perception, posture, and coordination, all of which are relevant to mission success. Similarly, vision is impaired in many space travelers due to possible intracranial pressure changes or fluid shifts in the brain. As humankind prepares for extended missions to Mars and beyond, it is imperative to compensate for these perils in prolonged weightlessness. Possible countermeasures are considered such as exercise regimens, improved nutrition, and artificial gravity achieved with a centrifuge or spacecraft rotation.
Collapse
Affiliation(s)
- Alexander J Dontre
- School of Psychology, Fielding Graduate University, 2020 De La Vina Street, Santa Barbara, CA 93105, USA; Department of Communications, Behavioral, and Natural Sciences, Franklin University, 201 South Grant Avenue, Columbus, OH 43215, USA.
| |
Collapse
|
93
|
Noirungsee N, Changkhong S, Phinyo K, Suwannajak C, Tanakul N, Inwongwan S. Genome-scale metabolic modelling of extremophiles and its applications in astrobiological environments. ENVIRONMENTAL MICROBIOLOGY REPORTS 2024; 16:e13231. [PMID: 38192220 PMCID: PMC10866088 DOI: 10.1111/1758-2229.13231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 12/19/2023] [Indexed: 01/10/2024]
Abstract
Metabolic modelling approaches have become the powerful tools in modern biology. These mathematical models are widely used to predict metabolic phenotypes of the organisms or communities of interest, and to identify metabolic targets in metabolic engineering. Apart from a broad range of industrial applications, the possibility of using metabolic modelling in the contexts of astrobiology are poorly explored. In this mini-review, we consolidated the concepts and related applications of applying metabolic modelling in studying organisms in space-related environments, specifically the extremophilic microbes. We recapitulated the current state of the art in metabolic modelling approaches and their advantages in the astrobiological context. Our review encompassed the applications of metabolic modelling in the theoretical investigation of the origin of life within prebiotic environments, as well as the compilation of existing uses of genome-scale metabolic models of extremophiles. Furthermore, we emphasize the current challenges associated with applying this technique in extreme environments, and conclude this review by discussing the potential implementation of metabolic models to explore theoretically optimal metabolic networks under various space conditions. Through this mini-review, our aim is to highlight the potential of metabolic modelling in advancing the study of astrobiology.
Collapse
Affiliation(s)
- Nuttapol Noirungsee
- Department of Biology, Faculty of ScienceChiang Mai UniversityChiang MaiThailand
- Research Center of Microbial Diversity and Sustainable Utilizations, Faculty of ScienceChiang Mai UniversityChiang MaiThailand
| | - Sakunthip Changkhong
- Department of Biology, Faculty of ScienceChiang Mai UniversityChiang MaiThailand
- Department of Thoracic SurgeryUniversity Hospital ZurichZurichSwitzerland
| | - Kittiya Phinyo
- Department of Biology, Faculty of ScienceChiang Mai UniversityChiang MaiThailand
- Research group on Earth—Space Ecology (ESE), Faculty of ScienceChiang Mai UniversityChiang MaiThailand
- Office of Research AdministrationChiang Mai UniversityChiang MaiThailand
| | | | - Nahathai Tanakul
- National Astronomical Research Institute of ThailandChiang MaiThailand
| | - Sahutchai Inwongwan
- Department of Biology, Faculty of ScienceChiang Mai UniversityChiang MaiThailand
- Research Center of Microbial Diversity and Sustainable Utilizations, Faculty of ScienceChiang Mai UniversityChiang MaiThailand
| |
Collapse
|
94
|
Berliner AJ, Zezulka S, Hutchinson GA, Bertoldo S, Cockell CS, Arkin AP. Domains of life sciences in spacefaring: what, where, and how to get involved. NPJ Microgravity 2024; 10:12. [PMID: 38287000 PMCID: PMC10825151 DOI: 10.1038/s41526-024-00354-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 01/11/2024] [Indexed: 01/31/2024] Open
Affiliation(s)
- Aaron J Berliner
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, USA.
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA.
- Program in Aerospace Engineering, University of California Berkeley, Berkeley, CA, USA.
| | - Spencer Zezulka
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, USA
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA
- School of Information, University of California Berkeley, Berkeley, CA, USA
| | - Gwyneth A Hutchinson
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, USA
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | - Sophia Bertoldo
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, USA
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | - Charles S Cockell
- UK Centre for Astrobiology, School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| | - Adam P Arkin
- Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, USA.
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA.
| |
Collapse
|
95
|
Anthony KM, Collins JM, Love SAM, Stewart JD, Buchheit SF, Gondalia R, Schwartz GG, Huang DY, Meliker JR, Zhang Z, Barac A, Desai P, Hayden KM, Honigberg MC, Jaiswal S, Natarajan P, Bick AG, Kooperberg C, Manson JE, Reiner AP, Whitsel EA. Radon Exposure, Clonal Hematopoiesis, and Stroke Susceptibility in the Women's Health Initiative. Neurology 2024; 102:e208055. [PMID: 38170948 PMCID: PMC10870742 DOI: 10.1212/wnl.0000000000208055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 10/30/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Studies suggest that clonal hematopoiesis of indeterminate potential (CHIP) may increase risk of hematologic malignancy and cardiovascular disease, including stroke. However, few studies have investigated plausible environmental risk factors for CHIP such as radon, despite the climate-related increases in and documented infrequency of testing for this common indoor air pollutant.The purpose of this study was to estimate the risk of CHIP related to radon, an established environmental mutagen. METHODS We linked geocoded addresses of 10,799 Women's Health Initiative Trans-Omics for Precision Medicine (WHI TOPMed) participants to US Environmental Protection Agency-predicted, county-level, indoor average screening radon concentrations, categorized as follows: Zone 1 (>4 pCi/L), Zone 2 (2-4 pCi/L), and Zone 3 (<2 pCi/L). We defined CHIP as the presence of one or more leukemogenic driver mutations with variant allele frequency >0.02. We identified prevalent and incident ischemic and hemorrhagic strokes; subtyped ischemic stroke using Trial of ORG 10172 in Acute Stroke Treatment (TOAST) criteria; and then estimated radon-related risk of CHIP as an odds ratio (OR) and 95% CI using multivariable-adjusted, design-weighted logistic regression stratified by age, race/ethnicity, smoking status, and stroke type/subtype. RESULTS The percentages of participants with CHIP in Zones 1, 2, and 3 were 9.0%, 8.4%, and 7.7%, respectively (ptrend = 0.06). Among participants with ischemic stroke, Zones 2 and 1 were associated with higher estimated risks of CHIP relative to Zone 3: 1.39 (1.15-1.68) and 1.46 (1.15-1.87), but not among participants with hemorrhagic stroke: 0.98 (0.68-1.40) and 1.03 (0.70-1.52), or without stroke: 1.04 (0.74-1.46) and 0.95 (0.63-1.42), respectively (pinteraction = 0.03). Corresponding estimates were particularly high among TOAST-subtyped cardioembolism: 1.78 (1.30-2.47) and 1.88 (1.31-2.72), or other ischemic etiologies: 1.37 (1.06-1.78) and 1.50 (1.11-2.04), but not small vessel occlusion: 1.05 (0.74-1.49) and 1.00 (0.68-1.47), respectively (pinteraction = 0.10). Observed patterns of association among strata were insensitive to attrition weighting, ancestry adjustment, prevalent stroke exclusion, separate analysis of DNMT3A driver mutations, and substitution with 3 alternative estimates of radon exposure. DISCUSSION The robust elevation of radon-related risk of CHIP among postmenopausal women who develop incident cardioembolic stroke is consistent with a potential role of somatic genomic mutation in this societally burdensome form of cerebrovascular disease, although the mechanism has yet to be confirmed.
Collapse
Affiliation(s)
- Kurtis M Anthony
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Jason M Collins
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Shelly-Ann M Love
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - James D Stewart
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Sophie F Buchheit
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Rahul Gondalia
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Gary G Schwartz
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - David Y Huang
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Jaymie R Meliker
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Zhenzhen Zhang
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Ana Barac
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Pinkal Desai
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Kathleen M Hayden
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Michael C Honigberg
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Siddhartha Jaiswal
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Pradeep Natarajan
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Alexander G Bick
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Charles Kooperberg
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - JoAnn E Manson
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Alexander P Reiner
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Eric A Whitsel
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| |
Collapse
|
96
|
Perdyan A, Jąkalski M, Horbacz M, Beheshti A, Mieczkowski J. Chromosomal positioning and epigenetic architecture influence DNA methylation patterns triggered by galactic cosmic radiation. Sci Rep 2024; 14:1324. [PMID: 38225252 PMCID: PMC10789781 DOI: 10.1038/s41598-024-51756-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/09/2024] [Indexed: 01/17/2024] Open
Abstract
Despite surging interest in space travel in recent decades, the impacts of prolonged, elevated exposure to galactic cosmic radiation (GCR) on human health remain poorly understood. This form of ionizing radiation causes significant changes to biological systems including damage to DNA structure by altering epigenetic phenotype with emphasis on DNA methylation. Building on previous work by Kennedy et al. (Sci Rep 8(1): 6709. 10.1038/S41598-018-24755-8), we evaluated spatial DNA methylation patterns triggered by high-LET (56Fe, 28Si) and low-LET (X-ray) radiation and the influence of chromosome positioning and epigenetic architecture in distinct radial layers of cell nucleus. Next, we validated our results using gene expression data of mice irradiated with simulated GCR and JAXA astronauts. We showed that primarily 56Fe induces a persistent DNA methylation increase whereas 28Si and X-ray induce a decrease DNA methylation which is not persistent with time. Moreover, we highlighted the role of nuclear chromatin architecture in cell response to external radiation. In summary, our study provides novel insights towards epigenetic and transcriptomic response as well as chromatin multidimensional structure influence on galactic cosmic radiation damage.
Collapse
Affiliation(s)
- Adrian Perdyan
- International Research Agenda 3P - Medicine Laboratory, Medical University of Gdansk, Marii Sklodowskiej Curie 3a, 80-210, Gdansk, Poland
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Marcin Jąkalski
- International Research Agenda 3P - Medicine Laboratory, Medical University of Gdansk, Marii Sklodowskiej Curie 3a, 80-210, Gdansk, Poland
| | - Monika Horbacz
- International Research Agenda 3P - Medicine Laboratory, Medical University of Gdansk, Marii Sklodowskiej Curie 3a, 80-210, Gdansk, Poland
| | - Afshin Beheshti
- Space Biosciences Division, NASA Ames Research Center, Blue Marble Space Institute of Science, Moffett Field, CA, 94035, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jakub Mieczkowski
- International Research Agenda 3P - Medicine Laboratory, Medical University of Gdansk, Marii Sklodowskiej Curie 3a, 80-210, Gdansk, Poland.
| |
Collapse
|
97
|
Little RB, Snowton M, Uziel O. Cancer cells possess different isotopic enrichment: Isotopic induced functionalizations of normal DNA mutations leading to cancer. Heliyon 2024; 10:e23519. [PMID: 38170065 PMCID: PMC10758867 DOI: 10.1016/j.heliyon.2023.e23519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/19/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Although the dynamics of telomeres during the life expectancy of normal cells has been extensively studied, there are still some unresolved issues regarding this research field. For example, the conditions required for telomere shortening leading to malignant transformations are not fully understood. In this work, we mass analyzed DNA of normal and cancer cells for comparing telomere isotopic compositions of white blood cells and cancer cells. We have found that the 1327 Da and 1672 Da characteristic telomere mass to charges cause differential mass distributions of about 1 Da among normal cells relative to cancer cells. These isotopic differences are consistent with a prior theory according to which replacing primordial, common isotopes of 1H, 12C, 14N, 16O, 24Mg, 31P and/or 32S by nonprimordial, uncommon isotopes of 2D, 13C, 15N, 17O, 25Mg and/or 33S leads to altered enzymatic dynamics. This replacement may subsequently modulate DNA and telomere codons resulting in transformation of normal cells to cancer cells (in 15 N depletion in telomeres dependent manner). The prior theory and the current data are consistent also with a recently observed non-uniform methylation pattern of the DNA of cancer cells relative to a more uniform methylation in the DNA of normal cells. We observe further evidence of nonprimordial isotopic accelerations of acetylations, methylations, hydroxylations and aminations of nucleosides with alterations of phosphorylations of nucleotides; which may explain the induction of mutations at the DNA, RNA and proteins leading to cancer and more general alterations of DNA, which are associated with aging. This difference in mass spectra between normal and cancer DNA may stem from different functionalizations and isotopic enrichments affecting the motion derived from nuclear magnetic moments (NMMs). We suggest that this phenomenon may lead to malignant transformation.
Collapse
Affiliation(s)
| | - Maiya Snowton
- Department of Natural Science, Stillman College, Tuscaloosa, AL, USA
| | - Orit Uziel
- The Felsenstein Medical Research Center, Rabin Medical Center and Faculty of Medicine, Tel-Aviv University, Israel
| |
Collapse
|
98
|
Afshari N, Koturbash I, Boerma M, Newhauser W, Kratz M, Willey J, Williams J, Chancellor J. A Review of Numerical Models of Radiation Injury and Repair Considering Subcellular Targets and the Extracellular Microenvironment. Int J Mol Sci 2024; 25:1015. [PMID: 38256089 PMCID: PMC10816679 DOI: 10.3390/ijms25021015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Astronauts in space are subject to continuous exposure to ionizing radiation. There is concern about the acute and late-occurring adverse health effects that astronauts could incur following a protracted exposure to the space radiation environment. Therefore, it is vital to consider the current tools and models used to describe and study the organic consequences of ionizing radiation exposure. It is equally important to see where these models could be improved. Historically, radiobiological models focused on how radiation damages nuclear deoxyribonucleic acid (DNA) and the role DNA repair mechanisms play in resulting biological effects, building on the hypotheses of Crowther and Lea from the 1940s and 1960s, and they neglected other subcellular targets outside of nuclear DNA. The development of these models and the current state of knowledge about radiation effects impacting astronauts in orbit, as well as how the radiation environment and cellular microenvironment are incorporated into these radiobiological models, aid our understanding of the influence space travel may have on astronaut health. It is vital to consider the current tools and models used to describe the organic consequences of ionizing radiation exposure and identify where they can be further improved.
Collapse
Affiliation(s)
- Nousha Afshari
- Department of Physics and Astronomy, Louisiana State University, Baton Rouge, LA 70803, USA; (N.A.); (W.N.)
| | - Igor Koturbash
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Marjan Boerma
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Wayne Newhauser
- Department of Physics and Astronomy, Louisiana State University, Baton Rouge, LA 70803, USA; (N.A.); (W.N.)
| | - Maria Kratz
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA 70803, USA;
| | - Jeffrey Willey
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA;
| | - Jacqueline Williams
- School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Jeffery Chancellor
- Department of Physics and Astronomy, Louisiana State University, Baton Rouge, LA 70803, USA; (N.A.); (W.N.)
- Department of Preventive Medicine and Population Health, University of Texas Medical Branch, Galveston, TX 77555, USA
- Outer Space Institute, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
99
|
Caddy HT, Kelsey LJ, Parker LP, Green DJ, Doyle BJ. Modelling large scale artery haemodynamics from the heart to the eye in response to simulated microgravity. NPJ Microgravity 2024; 10:7. [PMID: 38218868 PMCID: PMC10787773 DOI: 10.1038/s41526-024-00348-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024] Open
Abstract
We investigated variations in haemodynamics in response to simulated microgravity across a semi-subject-specific three-dimensional (3D) continuous arterial network connecting the heart to the eye using computational fluid dynamics (CFD) simulations. Using this model we simulated pulsatile blood flow in an upright Earth gravity case and a simulated microgravity case. Under simulated microgravity, regional time-averaged wall shear stress (TAWSS) increased and oscillatory shear index (OSI) decreased in upper body arteries, whilst the opposite was observed in the lower body. Between cases, uniform changes in TAWSS and OSI were found in the retina across diameters. This work demonstrates that 3D CFD simulations can be performed across continuously connected networks of small and large arteries. Simulated results exhibited similarities to low dimensional spaceflight simulations and measured data-specifically that blood flow and shear stress decrease towards the lower limbs and increase towards the cerebrovasculature and eyes in response to simulated microgravity, relative to an upright position in Earth gravity.
Collapse
Affiliation(s)
- Harrison T Caddy
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, Australia and the UWA Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
- School of Human Sciences (Exercise and Sport Sciences), The University of Western Australia, Perth, WA, Australia
| | - Lachlan J Kelsey
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, Australia and the UWA Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
- School of Engineering, The University of Western Australia, Perth, WA, Australia
| | - Louis P Parker
- FLOW, Department of Engineering Mechanics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Daniel J Green
- School of Human Sciences (Exercise and Sport Sciences), The University of Western Australia, Perth, WA, Australia
| | - Barry J Doyle
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, Queen Elizabeth II Medical Centre, Nedlands, Australia and the UWA Centre for Medical Research, The University of Western Australia, Perth, WA, Australia.
- School of Engineering, The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
100
|
Park E, Evans MA, Walsh K. Regulators of clonal hematopoiesis and physiological consequences of this condition. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:3. [PMID: 39119355 PMCID: PMC11309374 DOI: 10.20517/jca.2023.39] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Clonal hematopoiesis (CH) is a prevalent condition that results from somatic mutations in hematopoietic stem cells. When these mutations occur in "driver" genes, they can potentially confer fitness advantages to the affected cells, leading to a clonal expansion. While most clonal expansions of mutant cells are generally considered to be asymptomatic since they do not impact overall blood cell numbers, CH carriers face long-term risks of all-cause mortality and age-associated diseases, including cardiovascular disease and hematological malignancies. While considerable research has focused on understanding the association between CH and these diseases, less attention has been given to exploring the regulatory factors that contribute to the expansion of the driver gene clone. This review focuses on the association between environmental stressors and inherited genetic risk factors in the context of CH development. A better understanding of how these stressors impact CH development will facilitate mechanistic studies and potentially lead to new therapeutic avenues to treat individuals with this condition.
Collapse
Affiliation(s)
- Eunbee Park
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Megan A. Evans
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Kenneth Walsh
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| |
Collapse
|