Abstract
The recent acceleration of commercial, private, and multi-national spaceflight has created an unprecedented level of activity in low Earth orbit (LEO), concomitant with the highest-ever number of crewed missions entering space and preparations for exploration-class (>1 year) missions. Such rapid advancement into space from many new companies, countries, and space-related entities has enabled a“Second Space Age.” This new era is also poised to leverage, for the first time, modern tools and methods of molecular biology and precision medicine, thus enabling precision aerospace medicine for the crews. The applications of these biomedical technologies and algorithms are diverse, encompassing multi-omic, single-cell, and spatial biology tools to investigate human and microbial responses to spaceflight. Additionally, they extend to the development of new imaging techniques, real-time cognitive assessments, physiological monitoring, and personalized risk profiles tailored for astronauts. Furthermore, these technologies enable advancements in pharmacogenomics (PGx), as well as the identification of novel spaceflight biomarkers and the development of corresponding countermeasures. In this review, we highlight some of the recent biomedical research from the National Aeronautics and Space Administration (NASA), Japan Aerospace Exploration Agency (JAXA), European Space Agency (ESA), and other space agencies, and also detail the commercial spaceflight sector’s (e.g. SpaceX, Blue Origin, Axiom, Sierra Space) entrance into aerospace medicine and space biology, the first aerospace medicine biobank, and the myriad upcoming missions that will utilize these tools to ensure a permanent human presence beyond LEO, venturing out to other planets and moons.
Introduction
The launch of the Russian satellite, Sputnik, in 1957 and the establishment of the National Aeronautics and Space Administration (NASA) in 1958 marked the beginning of the first Space Age. This era not only changed humanity, but also reshaped our relationship with our Moon, solar system, and search for new stars. The Union of Soviet Socialist Republics (USSR) and the U.S.A. fiercely competed in space launches (Fig. 1, inset) during the Cold War, evolving from short missions to the first space stations (e.g., Salyut 1 by USSR and Skylab by USA). Eventually, more countries created capacity for space exploration (Fig. 1), which introduced a wider range of genetic, medical, and ethnic backgrounds among the humans who have flown into space.
Figure 1. A historic overview of space launches.

(inset) The launches that defined the first space age, from 1957 to 2022, broken down by the country of origin. (Main) The exponential increase in launches marks the Second Space Age, driven more by commercial launches. The number of launches (y-axis) per year (x-axis) is plotted with the color annotated as the United States (blue), Union of Soviet Socialist Republics (USSR)/Russia (purple), China (red), and other countries (green).
Moreover, while Russian cosmonaut Valentina Tereshkova was the first female in space in 19631, the first American female was not sent into space until 1983 (astronaut Sally Ride). Sex-specific differences in spaceflight’s effects have gained attention as more females have entered space. Notably, females appear to be less affected by spaceflight-associated neuro-ocular syndrome (SANS), yet more affected in other modalities, such as vascular responses and possible cancer risk2. However, comprehensive studies on cell-specific and genetic changes in both sexes only began in 2021, revealing differences crucial for mission planning3–5.
Astronaut selection, traditionally performed by government agencies like NASA, JAXA, and ESA, has expanded from the U.S. military’s selection in 1959 to include scientists in 19626. Current astronaut criteria typically involve citizenship, advanced degrees, and physical, cognitive, and stress testing. The private sector’s involvement, starting with Orbital Sciences Corporation’s Pegasus mission in 1990, has reshaped space exploration [1]. The private sector’s contributions to spaceflight technology and crew health research expanded with the entry of companies like Blue Origin, Virgin Galactic, and SpaceX. In 2021, SpaceX’s Inspiration4 marked the first fully private, crewed orbital mission, emphasizing the growing trend of civilian astronauts7–9. Then, 2022 and 2023 set records for the most launches into space by both commercial and government agencies (n=188, n=196, respectively)10. The SpaceX Starship, the largest rocket ever built, reached orbit in 2023, highlighting the accelerated pace of spaceflight technologies and new economies for space exploration11.
These spaceflight developments are not just a difference of scale; they represent a substantive difference in the speed, type, and degree of access to space. For example, after more than 20 years of continuous human presence in space onboard the solitary International Space Station (ISS), there is now another orbiting space station (Tiangong) from the Chinese National Space Administration (CNSA) and five orbital platforms being planned by Axiom, Northrop Grumman, Sierra Space-Blue Origin, VAST, and Voyager-Nanoracks. Furthermore, additional research platforms are currently in development beyond LEO, including the NASA-led Lunar Gateway space station orbiting the Moon which will have Canadian Space Agency (CSA), ESA, Mohammed Bin Rashid Space Centre, and JAXA partners, and permanent Lunar habitats by NASA Artemis program, as well as Lunar habitats by the CNSA and ROSCOSMOS (led by Russian government). By the late 2030s, the Mars Base Camp orbital platform by an aerospace company (Lockheed Martin) is planned for an orbit around Mars that can provide continual access to the surface12 (Table 1).
Table 1. Upcoming LEO and interplanetary missions in the next decades.
Current mission plans include those led by non-government actors (Non-Gov), NASA (GOV (US)), and non-US governments (Gov). The mission destinations are listed on the top of each category (Asteroids, exoplanets, Gas Giants, Low Earth Orbit, Mars, Moon, Venus and Mercury). Asteroids related missions will be conducted mainly by both NASA and European Space Agency (ESA) with the specific information found here109–113. The exoplanets missions will be conducted by Breakthrough Initiatives114. Gas giants missions will be conducted by NASA, ESA115–117 and China National Space Administration (CNSA). Low Earth Orbit missions indicated in this figure will be done by Indian Crewed Spaceflight (ISRO)118 and Virgin Galactic119. Several agencies are planning Mars missions which include: Lockheed Martin (LM)120, United Arab Emirates (UAE) Space Agency121, ISRO122, NASA123,124, CNSA125, Japan Aerospace Exploration Agency (JAXA)126, ESA127, and SpaceX128. The Moon missions will be conducted by NASA Artemis program (with support from ESA)129–131, China (CNSA)/ROSCOSMOS132,133, and JAXA134. Both NASA135,136 and ESA137 are planning Venus missions. There is also a joint ESA/JAXA Mercury mission138.
| Destination | Mission name | Mission details | Agency | Agency type | Mission type | links |
|---|---|---|---|---|---|---|
| Asteroids / Kuiper Belt | DART | DART launch (2021); Asteroid Didymos impact (2022) | NASA | Gov (US) | Flyby | https://science.nasa.gov/mission/dart/ |
| DART/Hera | Hera launch to visit DART (2024); Hera arrives at Didymos site (2026) | ESA | Gov | Flyby | https://www.heramission.space | |
| Lucy | Launch (2021); Inner-Main Belt (2025); L4 Trojan Cloud (2027); L5 Trojan Cloud (2033) | NASA | Gov (US) | Flyby | https://science.nasa.gov/mission/lucy/ | |
| Hayabusa2 | Launch (2014); Asteroid Ryugu sample return (2020); Asteroid (98943)2001 CC21 (2026); Asteroid 1998 KY26 (2031). | JAXA | Gov | Flyby | https://science.nasa.gov/mission/hayabusa-2/ | |
| New Horizons | Launch (2006); Pluto (2015); Arrakoth (2019); Kuiper belt (2023 onward) | NASA | Gov (US) | Flyby | https://science.nasa.gov/mission/new-horizons/ | |
| OSIRIS-Rex | OSIRIS-Rex asteroid sample return: launch (2016); return (2023) | NASA | Gov (US) | Sample return mission | https://science.nasa.gov/mission/osiris-rex/ | |
| Psyche | launch of Psyche asteroid probe (2023); arrival (2026); completion (2028) | NASA | Gov (US) | Orbiter | https://www.jpl.nasa.gov/missions/psyche | |
| Exoplanets | Starshot | launch of Breakthrough Starshot (2036); arrival (2061); signal returns (2065) | Breakthrough Initiatives | Non-Gov | Flyby | https://breakthroughinitiatives.org/initiative/3 |
| Gas Giants | Dragonfly | launch of Dragonfly lander (2027); arrival on Titan (2034) | NASA | Gov (US) | Lander | https://dragonfly.jhuapl.edu |
| Europa Clipper | Launch (2024); Arrival (2028) | NASA | Gov (US) | Orbiter | https://europa.nasa.gov | |
| PERSEUS | Launch (2031); Arrival (2043) | NASA | Gov (US) | Orbiter | https://ntrs.nasa.gov/citations/47115782563137 | |
| JUICE | launch (2023); arrival (2030); orbit Ganymede (2034) | ESA | Gov | Orbiter | https://www.esa.int/Science_Exploration/Space_Science/Juice | |
| Tianwen-4 | Launch (2029); Jupiter orbit (2035); Uranus flyby probe (2045) | CNSA | Orbiter and Flyby | |||
| Low Earth Orbit | Gaganyaan | launch (2023) | ISRO | Gov | Crewed spacecraft | https://www.isro.gov.in/Gaganyaan.html |
| Space Tours | first tour (2023) | Virgin Galactic | Non-Gov | Crewed spacecraft | https://brochure.virgingalactic.com/spaceflight/ | |
| Mars | Mars BaseCamp | launch (2028); return (2031) | LM | Non-Gov | Deep space habitat | https://www.lockheedmartin.com/en-us/products/mars-base-camp.html |
| Hope | Launch (2020); Arrival (2021); Completion (2024) | UAE | Gov | Orbiter | https://www.emiratesmarsmission.ae/hope-probe/instruments/ | |
| Mangalyaan 2 / Mars Orbiter Mission (MOM) | Launch (2024) | ISRO | Gov | Orbiter | https://www.youtube.com/watch?v=H7NReDapIks and https://www.isro.gov.in/MarsOrbiterMissionSpacecraft.htmlnifies,the%20way%20for%20future%20explorations. | |
| Perseverance | Launch (2020); Arrival (2021); Collections (2021–2025). | NASA | Gov (US) | Rover | https://mars.nasa.gov/mars2020/ | |
| Mars Sample Return (MSR) | Launch (2026); Arrival (2028); Return (2032) | NASA | Gov (US) | Retrieval | https://mars.nasa.gov/msr/ | |
| Tianwen | Tianwen-1 Launch (2020); Arrival (2021); Tianwen-2 (2025); Tianwen-3 Sample Retrieval (2030) | CNSA | Gov | Rover | https://nssdc.gsfc.nasa.gov/nmc/spacecraft/display.action?id=2020-049A#:~:text=Tianwen%201%20(formerly%20Huoxing%201,reaches%20Mars%20in%20February%202021. | |
| MMX | launch (2024); orbit (2025); return (2029) | JAXA | Gov | Orbiter | https://www.mmx.jaxa.jp/en/ | |
| Rosalind Franklin | launch (2028); arrival (2030) | ESA | Gov | Rover | https://www.esa.int/Science_Exploration/Human_and_Robotic_Exploration/Exploration/ExoMars/ExoMars_rover | |
| Starship - uncrewed lander | launch (2027) | SpaceX | Non-Gov | Uncrewed lander | https://www.spacex.com/vehicles/starship/ | |
| Starship - crewed lander | launch first crew (2027) | SpaceX | Non-Gov | Crewed lander | https://www.spacex.com/vehicles/starship/ | |
| Moon | Argonaut | Argonaut 1 (2031); Argonaut 2 (2033); Argonaut 3 (2035) | ESA | Gov | Uncrewed spacecraft | https://www.esa.int/Science_Exploration/Human_and_Robotic_Exploration/Exploration/Argonaut |
| Artemis | Artemis 1 (2022); Artemis 2 (2025); Gateway (2026); Artemis 3 (2027); Artemis 4 (2030); Artemis 5 (2031); Artemis 6 (2032) | NASA | Gov | Uncrewed spacecraft | https://www.nasa.gov/humans-in-space/artemis/ | |
| Moonlight | Moonlight (2024) | ESA | Gov | Uncrewed Satellites | https://www.esa.int/ESA_Multimedia/Videos/2022/11/What_is_ESA_s_Moonlight_initiative | |
| Chandrayaan | Chandrayaan-3 (2023); Chandrayaan-4 (2028); Chandrayaan-5 (2030); Chandrayaan-6 (2032) | ISRO | Gov | South pole, drilling, and sample return missions | https://www.isro.gov.in/Chandrayaan3_Details.html | |
| Chang'e | Chang'e 5 (2020) | CNSA | Gov | Sample return mission | https://nssdc.gsfc.nasa.gov/nmc/spacecraft/display.action?id=2018-103A | |
| Chang'e | Chang'e 6 (2025) | CNSA | Gov | Lander | https://nssdc.gsfc.nasa.gov/planetary/lunar/cnsa_moon_future.html | |
| Chang'e | Chang'e 7 / Rashid II (2026) | CNSA/MBRSC | Gov | Lander | https://nssdc.gsfc.nasa.gov/planetary/lunar/cnsa_moon_future.html | |
| Chang'e | Chang'e 8 (2027) | CNSA | Gov | Lander | https://nssdc.gsfc.nasa.gov/planetary/lunar/cnsa_moon_future.html | |
| ILRS | Launch (2026) | CNSA/ROSCOSMOS | Gov | Lander | https://www.cnsa.gov.cn/english/n6465652/n6465653/c6812150/content.html | |
| IM | IM-1 (2024); IM-2 (2025); IM-3 (2026) | Commercial | Non-Gov | |||
| Russia Lunar | launch test and lunar soil return (2027) | ROSCOSMOS | Gov | Lander | ||
| Russia Lunar | launch crew (2029) | ROSCOSMOS | Gov | Lander | ||
| SLIM | SLIM (2022) | JAXA | Gov | Lander | https://global.jaxa.jp/projects/sas/slim/ | |
| VIPER | Launch (2024) | NASA | Gov (US) | Lander | https://science.nasa.gov/mission/viper | |
| Venus | DAVINCI | Launch (2029) | NASA | Gov (US) | Flyby | https://ssed.gsfc.nasa.gov/davinci/ |
| Envision | Launch (2031) | ESA | Gov | Orbiter | https://www.esa.int/Science_Exploration/Space_Science/EnVision_factsheet | |
| Vertias | Launch (2031) | NASA | Gov (US) | Orbiter | https://www.jpl.nasa.gov/missions/veritas | |
| Mercury | BepiColombo | Launch (2018); Landing (2025) | ESA/JAXA | Gov (US)/Gov | Orbiter | https://www.esa.int/Science_Exploration/Space_Science/BepiColombo |
Abbreviations: DART, Double Asteroid Redirection Test; PERSEUS, Plasma Environment, Radiation, Structure, And Evolution Of The Uranian System; JUICE, Jupiter Ice moons explorer at Jupiter, Ganymede, Callisto, and Europa; MMX, Martian Moons Exploration; Rosalind Franklin, part of the ExoMars programme; ILRS, International Lunar Research Station; VIPER, Volatiles Investigating Polar Exploration Rover.
These accelerating trends have arguably created a “Second Space Age” that features key differences from the first era. Specifically, (1) the commercial spaceflight sector is now leading many launches and missions instead of governmental agencies; (2) there is a log-level increase in the number of countries participating in space exploration (Fig 1); (3) the advanced cellular and molecular studies of the human body’s spaceflight response has surpassed the number of publications from the prior missions like NASA’s Twins Study13 (4) the biomedical, behavioral, and omics data from the astronauts can now be accessed through a Biobank and Biorepository8; (5) there is increased crew heterogeneity across age, sex, and race; and (6) a continued human presence will extend beyond LEO, including lunar bases and planetary missions (Table 1).This Second Space Age enables “precision astronaut medicine” and thus, the chance to create personalized countermeasures for astronauts. In addition, accessible astronaut biomedical data in Biobanks benefits research in both space and Earth-based contexts14,15, similar to the utility of the USA’s AllofUs Program and the UK Biobank.
In this Perspective, we highlight research from the “Space Omics and Medical Atlas (SOMA) across orbits” package, which features data collected from SpaceX’s Inspiration4 (I4) crew members, JAXA studies, NASA and ESA astronauts, and a comparison of these results with a large body of model organism data, cellular profiles, computational models, and countermeasures. The I4 mission, the first all-civilian spaceflight, provided unprecedented insights through multi-omics (RNA-seq, microbiomics, proteomics, etc.) and diverse medical assessments (neurobehavioral, cognitive, environmental). This mission generated nearly 3,000 samples and hundreds of terabytes of data, constituting the most extensive dataset for human space exploration to date, and the first mission with public access to paired astronaut data (SOMA portal) and samples (Biobank)8,15 In addition, The SOMA package spans blood measurements from the 1960s Mercury missions up to recent commercial missions in 2024, and features a wide range of molecular and cellular assays across humans, model organisms, and ground-based simulations (e.g., NASA Space Radiation Laboratory)16,17 performed by investigators across >100 institutions. These datasets show changes at the cellular, tissue, organismal and systematic levels (Table 2), and begin to map differences between populations (e.g. age, sex) and link specific countermeasures to each astronaut. We describe here the specific changes observed at each modality of biology, detail their significance, and link them to future missions and plans for the coming decades, with the aim to create a guide for potential countermeasures and tools essential for ensuring safe human space travel, particularly as mission durations, risks, and radiation levels escalate.
Table 2. The package of Space Omics and Space Omics and Medical Atlas (SOMA) across orbits.
The research and papers discussed in this manuscript are highlighted and categorized by different biological components which are: cellular, organ and tissue, and whole body. In addition, we categorize the countermeasures and computational research separately. Lastly, the annotation of astronaut data is included in the manuscripts.
| Main Assays | Key Cellular/Tissue Changes | Ref | Astronaut Data | |
|---|---|---|---|---|
| Cellular | ||||
| Mitochondria | RNA-seq | Plasma cell free (cf) RNA maps indicating mitochondrial dysfunction | 47 | JAXA |
| Whole-genome Sequencing | Mitochondrial DNA in plasma; genome stability | 32 | JAXA; I4; NASA twin | |
| Immune Cells | scRNA-seq | Immune dysfunction in space and simulated microgravity | 35 | JAXA; I4; NASA twin |
| Single cell multi-omics | Inflammation and chromatin changes in monocytes | 3, 151 | I4 | |
| Sex-specific immunomes | sexually dimorphic immune and endocrine kinetics | 33 | None | |
| Review | Macrophage alterations | 5 | None | |
| Behavioral assays and flow cytometry-based immune cell profiling | Monocyte driven changes | 139 | None | |
| RNA-seq | Haemoglobin dysregulation | 140 | JAXA; I4; NASA twin | |
| Chromosomes / Telomeres | Genomics and RNA-seq | Elevated telomeric RNA | 29, 32 | I4, NASA twin |
| Epigenetics & Transcriptomics | DNA methylation changes | 8, 22 | JAXA | |
| Epigenetic changes | Epitranscriptomics | RNA methylation increases and shifts | 141 | I4, NASA twin |
| Endocrine Effects | Multi-Omics | Insulin and Estrogen signaling | 4 | JAXA; I4 |
| Organs and tissues | ||||
| Heart | Omics & western blotting | Cardiac fibrosis and miRNA increases | 61 | JAXA; I4 |
| Clonal hematopoiesis of Indeterminate Potential (CHIP) | CHIP Hazard Ratios | 142 | None | |
| CHIP changes from spaceflight | 32 | I4; NASA twin | ||
| Skin | Spatial Multi-Omics | Inflammatory skin changes | 57 | I4; NASA twin |
| Transcriptomics | Skin health dysfunction | 56 | JAXA; I4; NASA twin | |
| Skeletal Muscle | Bioreactor | Development of muscle countermeasures | 54 | None |
| Transcriptomics | Sarcopenia | 52 | JAXA; I4 | |
| Brain | Spatial transcriptomics | Neurodegenerative disease | 59 | None |
| Multi-omics and exosome profiling | Oxidative and BBB stress | 60 | I4, NASA twin | |
| Behavioral Assays | Psychomotor vigilance | 58 | None | |
| Kidney | Multi-omics and spatial transcriptomics | Kidney dysfunction | 63 | JAXA; I4; NASA twin |
| Systemic, host–microbe, and whole-body impact | ||||
| Whole Body | Transcriptomics | Frailty gene expression in muscles | 51 | JAXA; I4 |
| Biospecimen protocols | Blood, urine, and skin | 9 | I4; NASA twin | |
| Biobank and data repository | Space omics & medical Atlas | 8 | I4, NASA twin; JAXA | |
| Physiological & molecular assays | Crew differences and I4 mobile imaging | 7 | I4, NASA twin | |
| Perspective | Whole Body | 91 | I4; NASA twin; JAXA | |
| Microbiome | Metagenomics and Metatranscriptomics | Microbial exchange | 73 | I4, NASA twin |
| Metagenomics | Microbial adaption to space | 72 | None | |
| Metagenomics | Microbial tracking on the ISS | 143 | None | |
| Countermeasures | ||||
| Drugs | RNA-seq and treatments with miRNA inhibitors | Immune & mitochondrial activation | 61, 62 | JAXA; I4; NASA twin |
| Genes | WGS, RNA, CRISPRa/i | Protective Alleles and Data Modeling | 97, 99 | I4; NASA twin |
| Computational and omics Tools | ||||
| Artificial Intelligence (AI) | Multi-omics & Machine Learning (ML) | Calcium uptake in muscles | 144 | None |
| Perspective | Precision Health | 145 | I4; NASA twin | |
| ML & transcriptomics | Liver dysfunction | 146 | None | |
| Perspective | AI in space research | 147 | None | |
| Omics Analysis | Transcriptomics | Muscle degradation | 148 | I4 |
| ML, CRISP, Transcriptomics | Liver dysfunction | 92 | None | |
| Perspective and ethics | ||||
| Ethics for Commercial Spaceflight | 98 | I4 | ||
| Open science integration for space biology research | 149 | I4, NASA twin, JAXA | ||
| Inspiration4 data availability on NASA’s open science platform | 150 | I4 | ||
| Women’s Health and Reproductive Systems | 152 | I4 | ||
Cellular Adaptations in response to Spaceflight
Spaceflight introduces hazards that result in diverse cellular and molecular changes18, primarily influenced by two factors: space radiation exposure and microgravity. Galactic cosmic radiation (GCR) is an unavoidable aspect of short- or long-term space missions, exposing astronauts to various atomic nuclei containing high linear energy transfer (LET) particles like 56Fe and 28Si, which pose significant health risks. The impact of radiation exposure includes distinct imprints on the human genome, transcriptome, and chromatin structure19,20. Understanding these effects is crucial for minimizing detrimental health outcomes21.
Perdyan et al.22 conducted a computational multi-omics analysis, investigating GCR’s effects on epigenetic23,24 and transcriptomic patterns using in vitro data from different bronchial epithelial cell lines exposed space radiation, in vivo data from mice exposed to whole body space radiation, and JAXA study astronauts’ data25 from NASA’s Open Science Data Repository (OSDR)/GeneLab platform26–28. Results showed that 56Fe induced DNA hypermethylation, while 28Si and X-ray exposure led to global DNA hypomethylation. Differentially methylated sites primarily accumulated in nuclear periphery, with minor DNA methylation changes in euchromatic regions. Persistent epigenome and transcriptomic changes that lasted up to 4 months post-landing were induced by 56Fe, but not by 28Si, in model organisms exposed to simulated GCR and JAXA study astronauts. The possible mechanisms behind the distinct 56Fe and 28Si responses can be examined in future studies.
Spaceflight-induced changes also extend to telomeres, the nucleoprotein complexes essential for maintaining genome stability. Previous work showed telomere elongation in NASA astronauts29–31, and recent studies shed light on the likely mechanisms behind this phenomenon8,32. Elevated levels of telomeric RNA (TERRA) in spaceflight samples suggest its role in facilitating telomeric recombination-mediated repair through the telomerase-independent ALT pathway29, and TERRA may also form dipeptide-repeat signaling proteins. These findings have broad implications for scenarios involving persistent telomeric DNA damage, such as space radiation exposure.
Chromosomal and telomeric damage induced by the space environment also has a direct impact on immune-related dysfunction. Burke et al.33 explored the effects of simulated GCR on murine models, revealing sexually dimorphic immune and endocrine responses. RNA sequencing also indicated sexually distinct sex-specific responses, with females showing more efficiently regulated inflammation profiles compared to males, which matches gene expression data from the I4 crew, and underscores the importance of personalized translational approaches for astronauts on exploration missions.
To further explore immune dysregulation in spaceflight, an extensive review by An et al. 5, highlighted the severe impact of the space environment on macrophages, central innate immune cells crucial for antigen removal and directing adaptive immune responses13,34. A single-cell multi-omics, and cytokine analysis of the I4 crews has identified 17 cytokines/chemokines related to inflammation and muscle homeostasis that increased after spaceflight and revealed changes in gene expression, chromatin accessibility, and TCR/BCR immune repertoire in response to spaceflight3,8,9. Differentially expressed genes (DEGs) were enriched for immune-metabolic pathways as well as chromatin modifications, and the immune cell types that were most impacted by spaceflight were CD14 and CD16 monocytes. Integrating with microbiome abundance data from the same crews has for the first time identified immune cell DEGs associated with microbiome shifts in taxonomy and viral activation3.
In addition to space radiation, microgravity can also impact the entire human immune system35. Single-cell RNA-seq analysis of human peripheral blood mononuclear cells (PBMCs) exposed to short-term simulated microgravity revealed core features of immune impairment. Comparative transcriptomics identified conserved features of immune dysfunction across simulated microgravity and spaceflight, including changes in pathways linked to cytoskeleton dynamics, pyroptosis, temperature-shock, proteostasis, nuclear receptors, interferon, IL-6, and sirtuin cascades.
Liquid biopsies, an alternative to traditional biopsies, extract cell-free (cf) nucleic acids from the blood or urine36,37, which emerge upon space-relevant stress38, aging39, metabolic disorders40, inflammation41, and DNA damage and clonal mutations42,43. These can detect changes earlier than protein biomarkers44, providing enhanced molecular heterogeneity resolution compared to standard tissue biopsies45. “Full-body molecular profiling” using cfDNA and cfRNA from liquid biopsies, coupled with clonal hematopoiesis mutation scans43,46, is a contemporary approach mapping spaceflight impact, ongoing in astronauts under the SOMA protocol (Table 3).
Table 3. Study design and biospecimen collection schemes for current omics-based flight studies.
A comparison of data generated as part of the NASA Human Research Program (HRP) Spaceflight Standard Measures and Omics Archive studies, Translational Research Institute for Space Health (TRISH) efforts, and the Cornell Space Omics and Medical Atlas (SOMA). Data generation protocols include Whole Genome Sequencing (WGS) in Clinical Laboratory Improvement Act (CLIA) labs, Pharmacogenomics (PGx), Whole Genome Bisulfite Sequencing (WGBS), Complete Blood Counts (CBC) with differential, Complete Metabolite Panel (CMP), biochemical assays with the Johnson Space Center (JSC) panel, extracellular vesicles and particles (EVPs), and B-cell receptor and T-cell receptor (BCR/TCR) repertoires. Some variations include Glycoproteomics (+Glyco) or poly-Adenylated (polyA) and ribosomal RNA-depleted (ribo-) RNA-sequencing (RNA-seq). Most samples are aliquoted and banked into long-term archives, including viably frozen cells in dimethyl sulfoxide (DMSO).
| Assays and Purpose | ||||
|---|---|---|---|---|
| Protocol | HRP / NASA | TRISH / Baylor | SOMA / Cornell | |
| Blood | Whole Blood | - | CLIA WGS and PGx | CLIA WGS and PGx |
| Blood cell count (CBC) | Blood cell count (CBC) | Blood cell count (CBC) | ||
| Metabolic Panel (CMP) | Metabolic Panel (CMP) | Metabolic panel (CMP) | ||
| Serum | Biochemistry (JSC panel) | Biochemistry (JSC panel) | Biochemistry (JSC panel) | |
| Plasma | Proteomics (+Glyc) | Proteomics | Proteomics (untargeted/targeted) | |
| Lipidomics | - | Lipidomics | ||
| Metabolomics | Metabolomics | Metabolomics | ||
| - | - | Exosome/EVPs Profiles and Proteins | ||
| PBMCs | - | - | Viably Frozen Cells (DMSO) | |
| - | - | Telomere Length | ||
| - | - | Clonal Hematopoiesis Panel | ||
| - | Single-Cell RNA-seq | Single-Cell RNA-seq | ||
| - | - | Single-Cell ATAC-seq | ||
| Functional Immune Assessment | Immune profiling | Single-Cell (BCR/TCR)-seq | ||
| cfDNA | - | - | Cell-free DNA sequencing | |
| cfRNA | - | - | Cell-free RNA sequencing | |
| PAXgene RNA | RNA-seq | RNA-seq | RNA-seq (polyA, ribo-) | |
| - | - | Direct RNA sequencing | ||
| Cheek Epithelia | Buccal Swab | WGS | - | Meta(Genome/Transcriptome) |
| - | - | Metabolomics | ||
| Urine | 24-hr-void | Proteomics | - | Proteomics |
| Lipidomics | - | Lipidomics | ||
| Metabolomics | - | Metabolomics | ||
| Biochemistry (JSC panel) | - | Biochemistry (JSC panel) | ||
| Morning void | - | Dipstick | Dipstick | |
| - | 16S | Metagenomics | ||
| - | - | Proteomics | ||
| - | Metabolomics | Metabolomics | ||
| - | - | Exosomes | ||
| - | - | Cell-free DNA/RNA sequencing | ||
| - | - | Biochemistry (JSC panel) | ||
| Saliva 1-day | Crude Saliva | Immune and qPCR viral panel | - | Immune and JSC qPCR viral panel |
| Oragene | WGBS | 16S | Meta(Genome/Transcriptome) | |
| Microbiome | Body Swabs | Metagenome | 16S | Meta(Genome/Transcriptome) |
| Saliva | Metagenome | 16S | Meta(Genome/Transcriptome) | |
| Fecal | Metagenome | 16S | Meta(Genome/Transcriptome) | |
| Vaginal | - | - | Meta(Genome/Transcriptome) | |
| Spacecraft | Swabs | - | - | Environmental data |
| - | - | Meta(Genome/Transcriptome) | ||
| Hair Follicles | Hair | - | - | Telomere Length |
| - | - | Nucleic Acid Banking | ||
| - | - | Meta(Genome/Transcriptome) | ||
| Semen | Sperm | - | - | Concentration, Size, Count, Motility, Morphology |
| Skin Biopsy | 3mm punch | - | - | Spatial transcriptome/proteome |
| - | - | Histology & morphology | ||
JAXA’s Cell-Free Epigenome (CFE) Study47 conducted an 11-time point liquid biopsy study with six astronauts who resided on ISS for >120 days. The study showed that cfRNA in plasma can capture longitudinal gene expression profiles of stressed or lysed internal tissues. The cfRNA analysis before, during, and after spaceflight also revealed mitochondrial dysregulation in space36, supporting previous studies13,48,49. The cfDNA analysis revealed a significant increase in relative mitochondrial DNA copy numbers during spaceflight, returning to baseline post-flight36, replicating NASA’s Twins Study findings13. The association of the extracellular mitochondria (exMT)-enriched fraction with the CD36 scavenger receptor and the release of exMT-containing extracellular vesicles into the plasma during spaceflight indicated systemic metabolic stress responses to the space environment. These results suggest exMT as a potential biomarker to assess tissue responses in spaceflight and to decipher tissues undergoing apoptosis, and reinforce theories that mitochondrial dysregulation is a central feature increasing spaceflight health risks.
Mitochondrial and immune function are interconnected, impacting insulin and estrogen signaling, and posing heightened health risks for the female reproductive system50. An integrated analysis of murine, JAXA cfRNA, and I4 scRNA-seq data revealed altered mRNA levels during and after spaceflight, affecting mitochondrial metabolic pathways, particularly lipid metabolism and oxidative stress4. These changes contribute to heightened health risks associated with reproductive hormone synthesis. Mitochondrial dysfunction in response to spaceflight was further supported by a comprehensive multi-omics analysis on I4 crew specimens3, 32. Distinct alterations in macrophages, neutrophils, and CD4+ T-cells, along with elevated interleukin-6 (IL-6) levels, were observed in scRNA-seq data, suggesting their potential impact on mitochondrial regulation, even in the relatively short I4 mission.
Organ and Tissue Responses in Spaceflight
The cellular changes that occur during spaceflight illustrate a consistent story of immune perturbation, DNA damage, and mitochondrial stress, evidenced across cellular, model organism, and astronaut models. Given the widespread cellular and molecular changes, studies have examined the combined impact of spaceflight at the organ and tissue levels. Here we will highlight the studies utilizing both existing data from model organism studies and astronaut data from the Twins, I4, and JAXA missions.
Muscle health is a crucial aspect of space research18, given the abnormal changes it undergoes during extended space missions, involving microgravity and radiation exposure. These changes can result in muscle mass decline and bone density loss, posing challenges for astronauts’ recovery upon returning to Earth and potentially accelerating biological decline or frailty47. These issues mirror the sarcopenia, characterized by muscle loss and frailty and often observed in older adults, and current countermeasures are relatively ineffective51. Castañeda et al.52 identified key genes associated with sarcopenia and found these genes to be dysregulated when comparing human cells sent to ISS and astronaut data from JAXA and I4 missions. Interestingly, skin expression profiling in I4 astronauts revealed deregulation of genes related to muscle loss, suggesting that skin data could serve as informative indicators of muscle-related gene deregulation53. The study further predicted potential countermeasure drugs targeting sarcopenia-associated genes 52.
In an additional study addressing muscle loss, Kamal et al.54 developed a new microgravity bioreactor using the StrexCell® system to release a daily bout of uniaxial cyclic stretch, that elicits changes in tensile loading on skeletal muscle myotubes. They provided evidence of a new uniaxial bioreactor for skeletal muscle loading and unloading that could be used for the study of mechanotransduction in skeletal muscle during future spaceflight. The StrexCell bioreactor system could also be used to test new countermeasure strategies against the adverse effects of microgravity and also could help in studies of aging55.
Skin-related issues, such as inflammation and discomfort during spaceflight, are well-known, but molecular insights and mitigation strategies are limited. Two manuscripts in this package enhance our understanding of skin changes during long- and short-duration spaceflight, featuring the first astronaut skin biopsies. Cope et al.56 conducted a comprehensive analysis using transcriptomic skin data from OSDR, correlated rodent and astronaut data from various missions, and identified responsive pathways in cell cycle regulation, lipogenesis, DNA damage, and mitochondrial dysregulation. In a second study, Park et al.57 analyzed 3mm human skin biopsies before and after spaceflight, revealing metabolic changes, DNA repair, cell cycle alterations, and immune system activation. Inflammatory responses and immune deregulation, driven by KRAS, were observed across skin tissue layers, consistent with cellular responses in previous studies.
Beyond muscle and skin, studies have delved into molecular changes affecting the central nervous system (CNS) and neuronal tissues, caused by exposure to GCR and microgravity. Desai et al.58 simulated acute and chronic GCR exposure on murine models, and observed differences in psychomotor vigilance. The study highlighted potential adverse effects on attentional processes and reaction time, emphasizing the importance of cognitive and neurological metrics for in-flight mission decision-making. The investigation also explored the link between GCR exposure effects on neurocognitive performance and neurotransmitter abnormalities affecting circuit connectivity. Chronic GCR exposure was found to increase levels of neurotransmitters within the prefrontal cortex, indicating potential interventions targeting dopamine pathways to restore homeostatic signaling in the irradiated brain.
Masarapu et al.59 and Houerbi et al.60 examined brain alterations in ISS and ground control murine models using Spatial Transcriptomics and single-cell multiomics (RNA-seq and ATAC-seq). These studies provided evidence of spaceflight-induced disruptions in neurogenesis, neuronal development, synaptogenesis, and neurodegeneration, sharing similarities with changes observed in aging and neurodegenerative diseases. Spatial transcriptomic data suggested a disrupted blood-brain barrier (BBB) in rodents during flight, underscoring the importance of continued monitoring for brain health in future crews.
Cardiovascular tissues and related organs are also severely impacted by the space environment and subject to elevated health risks. Paar et al.61 investigated the impact of space radiation on the heart, focusing on GCR-induced cardiac fibrosis. Activation of fibrosis-associated genes and pathways, including TGF-β1, was observed in blood samples from I4 Mission and JAXA CFE Study astronauts. Simulated GCR experiments in mice revealed time-dependent regulation of fibrotic processes, indicating the potential for developing novel countermeasures targeting various fibrotic markers related to spaceflight response. The study explored the influence of circulating microRNAs (miRNAs) linked to spaceflight-associated cardiovascular risks61,62, and tested antagomirs targeting miR-16–5p, miR-125b-5p, and let-7a-5p to mitigate cardiac fibrosis. The treatment restored TGF-β1 and COL1 signaling to control levels, highlighting the potential for developing novel countermeasures (below section).
The kidney, often understudied in spaceflight, was the focus of a comprehensive study by Siew et al.63. The I4 crew members exhibited changes in urinary chemistry during spaceflight, associated with primary alterations in ion transporter regulation. Diverse approaches revealed functional and structural renal remodeling in spaceflight, including morphometry, imaging, and multi-omics on rodent kidneys from the ISS, simulated ground analog experiments, and the I4 data,. Acute GCR exposure demonstrated markers of mitochondrial distress and early proteinuria, suggesting glomerular and proximal tubule dysfunction. These findings suggest the possibility of transient, maladaptive nephron remodeling that might lead to progressive kidney damage during long-duration deep space missions, underscoring the importance of appropriate mitigation strategies.
Recognizing the varied radiosensitivity of each tissue/organ is crucial for targeted research and countermeasures. Radiosensitive organs, including hematopoietic-related organs, reproductive systems, gastrointestinal system, epidermis, and eyes, exhibit the greatest sensitivity (and risk from)to space radiation64. As deep space missions become more feasible, understanding and mitigating the risks posed by constant exposure to low-dose space radiation becomes imperative. Mitochondrial exhaustion due to inflammation and immune suppression64 becomes a concern, particularly for organs less sensitive to radiation, like the brain and muscles, which also requires monitoring in spaceflight.
Systemic Effects of Spaceflight
With a better understanding of how the space environment impacts humans at both the cellular and organ/tissue levels, the overall biological response at the whole body, host-microbial, and systemic levels can be better understood and linked to prior work18. For example, understanding how spaceflight can advance aging and impact overall frailty can leverage the wide range of studies and indicate a systemic change. Camera et al.51 focused on establishing a frailty index for humans during spaceflight, which also links to well-defined hallmarks of aging39,51,65, including: mitochondrial dysfunction, telomere alterations, genomic instability, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, cellular senescence, stem cell exhaustion, and altered intercellular communication. Studies in this package link some aspects of spaceflight to the hallmarks of aging pathology, indicating signs of premature aging for some missions. The systemic impact of this can contribute to advanced muscle loss or sarcopenia, cardiovascular health risks (such as fibrosis), clonal hematopoiesis, immune dysfunction, CNS issues, and more. Camera et al.51 created the “frailty index” using data from NASA’s OSDR27, from different mouse missions flown to the ISS, missions with cell culture flown to the ISS and simulated human microgravity experiments (i.e. bedrest studies66), and astronaut data from the JAXA study and I4 mission. Camera et al.51 focuses mainly on the impacts of frailty on muscle tissue, which revealed a key set of genes associated with an early frailty phenotype. Specifically, they noticed key changes with interferon inflammatory response, metabolic disorders, hypoxia response, and increased cellular senescence.
The I4 mission provided a vast amount of both physiological and molecular data from the four civilian astronauts, spanning the six research projects, thousands of samples, and three mission phases (Table 3)7. Key measurements include multi-omics and virome analysis associated with spaceflight, organ ultrasound imaging, and comprehensive cardiovascular and neurocognitive assessments. Systemic alterations were evident post-flight, particularly in human PBMCs, showing thousands of DEGs at R+1. Notably, CD14+ and CD16+ monocytes displayed the most significant changes in gene expression, which were linked to regions of more open chromatin, including genes associated with DNA repair, immune activation, and nucleosome organization8. Physiological changes were recorded using handheld ultrasound devices for autonomous imaging of the urinary bladder, internal jugular vein, and eyes. Generally, short-duration spaceflight did not induce significant physiologic changes post-flight relative to pre-flight. However, crewmembers, even without space motion sickness, exhibited consistent vertical ocular misalignment post-flight, contrasting with pre-flight conditions. Cardiovascular function, activity levels, and energy expenditure were objectively measured using the Apple Watch Series 6, marking its inaugural use in spaceflight. Neurocognitive performance was assayed using a battery of ten cognitive tests developed for astronauts that has been deployed in both spaceflight and ground-based spaceflight analog studies.
Although the effects of short-duration spaceflight on cardiovascular function and neurocognitive performance were modest, there were marked interindividual differences in response to spaceflight, consistent with previous research67,68. Significant changes in heart rate, heart rate variability, energy expenditure, and activity levels occurred across mission phases. Furthermore, the spacecraft environment can impact crew physiology and neurobehavioral functions68 and three out of the four I4 crew exhibited positive associations between CO2 levels and heart rate variability in-flight. Moreover, cfRNA and cfDNA profiles revealed that cells with the greatest lysis arose from the hematopoietic system8,60, which mirrors the radiation risk of this system. Overall, the findings from the orbital mission demonstrate that the collection of high quality biomedical and behavioral data are feasible in a commercial crew with rapid training, and how systemic and whole-body level analysis from omics and biometrics data generates rich profiles on impact of spaceflight on the human body.
During spaceflight, alterations in host-microbial interactions have a systemic impact, particularly as microorganisms adapt to novel and extreme environments by incorporating new genetic material, particularly through bacteriophages69. Bacteriophages, upon inserting viral DNA into hosts, can become dormant (prophages), leading to modified host genotypes with gene disruption70, silencing, and chromosomal rearrangement, thereby influencing host gene expression8. Prophages facilitate the transfer of bacterial genes, including virulence and antibiotic resistance genes, toxins, effector proteins, and regulatory proteins, among cells71. Irby et al.72 investigated prophage presence and function in genomes of bacteria isolated from the ISS compared to terrestrial counterparts, exploring their contribution to microbial adaptation in the spaceflight-built environment. Analyzing ten bacterial species from five ISS sampling campaigns, they identified significant spaceflight-related differences in mobile genetic elements, particularly prophages. While transposes are common in terrestrial strains, they are notably absent in ISS strains. Instead, ISS strains exhibit an increased prevalence of Mu-like phages and unclassified phages. This variation implies that selective pressures unique to the space environment, such as limited nutrient availability and heightened genetic diversity, promote microbial survival under these conditions. Overall, the study indicated that prophage-encoded functions correlated with increased microbial persistence on the ISS, providing insights into potential mechanisms for microbial adaptation to this unique environment.
The I4 mission also created the largest astronaut microbiome study to date73, spanning 750 samples across 10 time points, with shotgun metagenomics and metatranscriptomics performed for each sample. Data from Tierney et al. showed a microbiome architecture of spaceflight that was characterized by time-dependent and taxonomically-divergent microbiome alterations across both time and space (including strain exchange with the SpaceX Dragon spacecraft). They also observed pan-phyletic viral activation and signs of persistent changes that, in the oral microbiome, yielded plaque-associated species with strong associations to immune cell gene expression. Further, they found enrichments of microbial genes associated with antibiotic production, toxin antitoxin systems, and stress response enriched universally across the body sites, and were correlated with some of the T-cell and B-cell expression dynamics in the crew.
Countermeasure Development for Spaceflight
There are limited medical countermeasure options specifically designed to decrease the negative effects of radiation exposure in astronauts due to spaceflight. Currently, there are three FDA-approved medical countermeasures, Neupogen, Neulasta, and Leukine, which are intended to improve survival following exposure to an acute myelosuppressive radiation dose74. These countermeasures improve the likelihood of survival by mitigating neutropenia and thrombocytopenia associated with acute radiation sickness. However, their effectiveness has primarily been studied in the context of photon irradiation, with limited evaluations for proton or other radiation qualities experienced during spaceflight, such as GCR. Additionally, while the FDA-approved radioprotectant Ethyol (Amifostine) is available to reduce xerostomia post-radiotherapy for head and neck cancers, its utility in mitigating space radiation effects is limited due to its parenteral administration, short half-life, and side effects18.
Addressing the challenges posed by space radiation and microgravity, Paar et al.61 explored the potential of miRNA inhibitors as a countermeasure. Inhibitors targeting specific miRNAs (miR-16–5p, miR-125b-5p, let-7a-5p) were tested to alleviate cardiac fibrosis in mice exposed to simulated space radiation and microgravity. A complementary study by McDonald et al.62 identified these miRNAs based on a previously established circulating miRNA signature associated with the space environment75. Using a 3D human model for microvessel physiology, inhibition of these miRNAs demonstrated significant preservation of the human microvessel structure, reducing DNA damage and stress after exposure to simulated Galactic Cosmic Rays (GCR). This approach, supported by observations in both 3D human microvasculature tissue model and astronaut data from missions like JAXA and Inspiration4, indicates the potential effectiveness of miRNA inhibitors in countering specific challenges encountered during spaceflight.
Expanding countermeasures to address skin-related issues observed in various datasets, including spatial transcriptomics from the I4 mission, JAXA CFE, and murine models56, offers insights into potential interventions. Altered expression of FLG and CASP14, genes known to modulate skin permeability, during and after flight indicate that these genes may be involved in water loss and responses to irritants, allergens, and microbial products during spaceflight. FLG loss-of-function mutations are associated with conditions like atopic dermatitis. This can be treated by dupilumab, which inhibits interleukins 4 and 13, and thereby upregulates FLG expression and restores epidermal barrier function. This drug could be explored for in-flight and post-flight treatment to restore skin barrier function76.
Interestingly, miRNA-based countermeasures offer innovative potential to mitigate space radiation damage; however, extensive pre-clinical and clinical trials are essential before human implementation. Meanwhile, repurposed drugs are being explored as countermeasures for spaceflight-related damage, particularly addressing symptoms from solar particle events (SPE)77. Anti-nausea medications like Ondansetron, granisetron, palonosetron, Imodium®, Neupogen®, corticosteroid cream, and dolasetron are considered for mitigating SPE symptoms (e.g. nausea, vomiting, diarrhea, radiation dermatitis, neutropenia). Flavonoid supplements (e.g., apigenin78) and vitamin D79, along with exercise80, are also investigated as countermeasures to reduce inflammation81 and mitigate spaceflight damage. Until specific miRNA-based treatments are developed, a combination of FDA-approved drugs, nutritional supplements, and microbial interventions82 may be explored for comprehensive mitigation of spaceflight-induced damage.
Astronaut precision medicine (APM) emerges as an actionable countermeasure involving tailoring treatment and prevention to individual characteristics, encompassing molecular, physiological, morphological, and behavioral aspects83,84. Pharmacogenomics (PGx), a cornerstone of APM, examines gene variants influencing drug metabolism85,86, optimizing drug safety and efficacy for individual astronauts. Developing PGx profiles of astronauts and crews could ensure personalized drug regimens, enhancing mission safety and effectiveness.
This principle can be applied to many of the drugs in a mission formulary. Importantly, these types of drug responses can be predicted and personalized. The application of PGx (drug-gene interaction) should also be accompanied by careful attention to drug-drug, drug-nutrient, drug-food, drug-microbe, and drug-herb interactions. These can be systematically assessed for individuals and crewsand can be implemented using large cohort databases and routine sequencing for the crews. Addressing these interactions removes another potential impediment to astronaut health, safety, and performance.
Applying APM/PGx to space missions involves molecular phenotyping to characterize functionally related molecular networks (FCN)83. By addressing dysregulations before space missions, APM aims to prevent their impact on health, safety, and performance in the space environment. Targeting specific gut microbe-produced substances, such as the elevated neurotoxin and nephrotoxin p-cresol observed in the NASA Twins Study87, enables dietary countermeasures, including fiber and resistant starch, to lower p-cresol production. APM may also address challenges like space-associated neuro-ocular syndrome (SANS) by characterizing genotypes and metabolites related to the one-carbon molecular network.
Beyond the pharmacological and physical countermeasures, genetic and epigenetic tools have emerged as innovative approaches to mitigate spaceflight-associated risks. CRISPR technologies, utilizing Cas9 and other Cas systems, allows precise modification of somatic cells to correct or replace disease-driving genes. Specifically, recent clinical trials have successfully treated conditions like beta-Thalassemia and sickle cell disease by deleting repressor genes for fetal hemoglobin88. Epigenetic modification systems, utilizing deactivated Cas9 (dCas9)89, fused with histone or DNA modifiers, such as DNMT3A or TET1, enable targeted modification of gene expression, providing a means for permanent or transient genetic alterations related to spaceflight. These advancements may play a crucial role in addressing long-term challenges for human settlement on other planets90.
Computational and omics Tools in Spaceflight Research
Advanced computational methods, omics platforms, and new algorithms play a crucial role in understanding factors related to spaceflight health. Casaletto et al.91 utilized machine learning techniques, specifically the Causal Research and Inference Search Platform (CRISP), to predict features causally linked to a binary response variable, employing prediction invariance as a guiding principle. By applying CRISP to gene expression data from NASA’s OSDR, they identified genes and molecular targets associated with lipid density phenotype in space-flown rodents. This approach unveiled novel insights not captured by traditional systems biology methods, particularly in addressing liver dysfunction. The SOMA Resource paper8 also features four data portals and tutorials on data usage, to help discover more biology and replicate across missions. The study highlights the importance of a causal inference framework based on environment invariance for robust feature identification, emphasizing its applicability to various tissues, phenotypes, and omics data. Continued advancements in computational and biological tools are crucial for comprehending spaceflight’s impact and developing effective countermeasures.
Limitations associated with space research
While the NASA’s Twins Study13 marked a significant stride in clinical genomics and multiomics analysis during spaceflight, limitations on crew size and follow-up were evident. The I4 and JAXA studies, with n = 4 for I4, n = 6 for JAXA, and n = 14 for an ISS astronaut study on bone marrow92, have expanded the subject pool but still face constraints, especially when considering sex-specific analyses. The inherent challenges of limited human subjects in space experiments persist due to constrained flight opportunities, regulatory restrictions, and cost considerations.
Notwithstanding these challenges, meticulous planning, procedures, and analysis, coupled with a skilled team, have demonstrated the generation of valuable insights from I4 and JAXA studies. Ground-based studies and control cohorts, including those like HI-SEAS and analog astronauts in EXPAND, alongside collaborations with initiatives such as the UK Biobank and commercial entities like Pheno.AI and the Human Phenome Project, continue to enhance our understanding despite the inherent limitations in human subject numbers for space research.
Outlook
While data from the various missions, computational tools, and model organisms provide valuable insights into the impacts of spaceflight, significant challenges persist. While some molecular signatures are consistent across both short and long-term missions (e.g. IL-6, IL-10 increases in plasma, telomere elongation, mitochondrial stress), others appear specific to extended exposure and chronic space radiation (e.g. CRP spikes). The increasing radiation burden observed in current missions like I4 and future missions (Fig. 2), highlights the necessity for precision medicine strategies tailored to individual astronauts, ensuring the right treatment at the right time for the specific mission.
Figure 2. Radiation levels of Inspiration4 mission, NASA’s Twins Study and other exposures.

The effective accumulated radiation dose is provided in millisieverts (mSv). The low linear energy transfer (LET) radiation (or terrestrial radiation) is denoted by the green bars. The radiation levels experienced during the Inspiration4 mission and Scott Kelly year-long mission (NASA’s Twins Study) are indicated by orange bars. The estimated radiation dose of a 3-year future mission to Mars is depicted by a red bar. All other high LET radiation doses are indicated by the blue bars.
Previous work has identified mitochondrial dysfunction as a key driver of systemic damages during spaceflight48, including inflammation, immune suppression, cardiovascular dysfunction, muscle atrophy, bone loss, and circadian rhythm disruption. While these systemic stresses appear universal, individuals experience varying degrees of dysregulation, necessitating astronaut-specific precision medicine to ensure safe space travel for all. Data from I4 and JAXA missions reveal both universal changes (increased inflammation and mitochondrial stress), independent of sex and ethnicity, and sex-specific variations (insulin and estrogen changes in females)4,8,33. By aggregating these findings, we can annotate systemic changes and construct a molecular fingerprint for key alterations indicated that individualized astronaut healthcare is crucial.
While conventional countermeasures focus primarily on pharmacological interventions, emerging approaches utilizing RNA biology, omics-based methods, and gene therapies offer promising avenues for active defense93–95. These advancements, coupled with genomic tools and personalized activation of specific alleles88, hold the potential to address individual health challenges encountered in space. However, careful consideration must be given to ethical concerns like informed consent96, crew ownership of data97–99, and adherence to full Institutional Review Board (IRB) protocols as research in this evolving landscape progresses, especially for long-duration missions (Fig. 3) and applies to ground studies as well.
Figure 3. Long-duration missions enabled by heavy lift rockets.

(a) The orbital trajectory and future missions enabled by the current Dragon capsule parameters. (b) Extra-lunar orbital trajectory that would approach the Lagrange point 1 (L1) closer to the sun and up to 1.54M km from the Earth (center blue diamond). The moon’s orbit is shown in dotted lines around the Earth. (c) The orbital trajectory for a three-planet mission in 2033 that would flyby Mars twice and also Venus (flyby) within about 18 months. The launch dates and approximate orbital timings (left) are shown around the planetary orbits (dotted line circles) and the flight path (yellow line). The sun is shown in the middle of the figure.
Indeed, ground-based analog studies continue to complement spaceflight experiments100–107, providing valuable insights into human responses to the space environment. As space research advances, integrating data from individuals of diverse ages, sexes, and lifestyles is essential to facilitate a comprehensive understanding of genetic and epigenetic associations with space adaptation. Efficient subject stratification will be crucial for the successful evaluation of future medical interventions.
The data and new discoveries described above are exciting, but beg the question: How will we know when we’ve reached the end of the Second Space Age? Perhaps, it could happen within a matter of decades. China and the US have both announced plans for a crewed mission to Mars (no earlier than 2035 and 2039, respectively), as well as for active work to return samples from Mars (Table 1). New trajectories enabled by heavy-lift rockets like the Starship can enable missions that span longer lunar arcs (Fig 3b), or threeplanets in one trip (Fig 3c)108 and future missions will be enabled by the current SpaceX Dragon parameters for crew and resources (Fig. 3a), enabling humans to travel farther than they have ever gone before. When successful, these events will signal the shift of humanity from a LEO-focused species to an interplanetary one, with instruments, missions, and crews moving around the planets of our first solar system. Indeed, by 2050, there should likely be: (1) orbital satellites around all planets in our solar system (Table 1); (2) a permanent presence of humans on the Moon; (3) the first crewed visit to another planet (e.g. Mars); (4) exchange of materials and samples between planets; and (5) plans to send probes to other stars. When that celestial stage is set, we will enter the next Space Age, when humans are permanent travelers and explorers in space.
Acknowledgements:
Special thanks to Dr. Jack Miller for assisting with the radiation doses in Figure 2. CEM thanks Igor Tulchinsky and the WorldQuant Foundation, NASA (NNX14AH50G, NNX17AB26G, 80NSSC22K0254, NNH18ZTT001N-FG2, 80NSSC19K0432, NNX13AE45G), the National Institutes of Health (R01MH117406, P01CA214274 R01CA249054), the LLS (MCL7001–18, LLS 9238–16), and the GI Research Foundation. J.K. thanks Boryung and their Global Space Healthcare Initiative and Humans In Space programs. A.B. was supported by NASA grant 16-ROSBFP_GL-0005: NNH16ZTT001N-FG Appendix G: Solicitation of Proposals for Flight and Ground Space Biology Research (Award Number: 80NSSC19K0883). SMB gratefully acknowledges funding from NASA (NNX14AB02G and 80NSSC19K0434). JJB thanks the National Institute of Aging for their ongoing support (5T32AG000266–23). RID and CLL thanks the National Aeronautics and Space Administration (NASA), NASA Johnson Space Center grant TXS0147017 (RID) and NASA NSCOR grant number NNX15AI22G (CLL) for funding this work. V.C. thanks ANID-Subdirección de Capital Humano/Doctorado Nacional/2022– 21220897 and FONDECYT 11190998; Proyecto Centro Basal ANID IMPACT:FB210024. A.P. was supported by the Walczak award funded by NAWA - Polish National Agency for Academic Exchange (Agreement No. BPN/WAL/2022/1/00024/U/00001). S.B.W. and K.S. acknowledges this work was partially funded by the UK Space Agency through a grant [ST/X000036/1] administered by the Science and Technology Facilities Council (STFC). S.B.W. is supported by Kidney Research UK [RP_017_20190306; ST_001_20221128; TF_007_20191202]. K.S. acknowledges this research was funded in part by the Wellcome Trust [Grant number 110282/Z/15/Z]. For the purpose of open access, the author has applied a CC BY public copyright license to any Author Accepted Manuscript version arising from this submission. T.L. acknowledges that portion of his research was carried out at the Jet Propulsion Laboratory, California Institute of Technology, under a contract with the National Aeronautics and Space Administration (80NM0018D0004).
Footnotes
Competing interests
CEM is a co-Founder of Cosmica Biosciences. SMB is a co-founder and Scientific Advisory Board member of KromaTiD, Inc.
Ethics and inclusion statement
This manuscript has included authors from all backgrounds from the scientific international community and the results are held at the highest ethical standards.
References
- 1.Whiting M Valentina Tereshkova and Sally Ride - Women Space Pioneers. NASA; http://www.nasa.gov/mediacast/valentina-tereshkova-and-sally-ride-women-space-pioneers (2018). [Google Scholar]
- 2.Mark S et al. The Impact of Sex and Gender on Adaptation to Space: Executive Summary. J Womens Health (Larchmt) 23, 941–947 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Kim J, et al. Single-cell multi-ome and immune profiles of the Inspiration4 crew reveal cell-type and sex-specific responses to spaceflight. Nat Commun (in press). The SpaceX Inspiration4 mission enabled state-of-the-art, single-cell multi-omics analysis of four astronauts, revealing that spaceflight-induced changes in gene expression, chromatin accessibility, and immune cell proportions, particularly in non-classical monocytes and NK cells, largely revert to normal within a few months post-flight, providing crucial data for future space missions and countermeasure development.
- 4.Mathyk B et al. Spaceflight alters insulin and estrogen signaling pathways. Comms Bio. (2024) doi: 10.1038/s42003-023-05213-2. [DOI] [Google Scholar]
- 5.An, et al. Influence of the Spaceflight Environment of Macrophage Lineages. npj Microgravity (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.60 Years Ago: NASA Selects A Second Group of Astronauts - NASA. https://www.nasa.gov/history/60-years-ago-nasa-selects-a-second-group-of-astronauts/ (2022). [Google Scholar]
- 7. Jones C, Overbey EG & et al. The SpaceX Inspiration4 mission reveals inflight molecular and physiological metrics from an all-civilian crew. Nature ((in press)). The SpaceX Inspiration4 mission, which included the first all-civilian crew and innovative in-flight experimental technologies, demonstrated that short-duration spaceflight induces temporary physiological and stress responses, supporting the likely safety of such missions and establishing a valuable biomedical database for future astronaut health countermeasures.
- 8. Overbey EG, et al. & et al. The Space Omics and Medical Atlas (SOMA): A comprehensive data resource and biobank for astronauts. Nature (in press). Spaceflight missions offer opportunities to study multi-omic and physiological changes in astronauts, as detailed in the Human Space Omics and Medical Atlas (SOMA), which presents comprehensive multi-omic samples and data from the SpaceX Inspiration4 crew, available through public and controlled-access biobanks and data repositories to accelerate biomedical discoveries for long-duration space missions
- 9. Overbey EG et al. Collection of Biospecimens from the Inspiration4 Mission Establishes the Standards for the Space Omics and Medical Atlas (SOMA). bioRxiv 2023.05.02.539108 (2023) doi: 10.1101/2023.05.02.539108. Collection of Biospecimens from the Inspiration4 Mission Establishes the Standards for the Space Omics and Medical Atlas (SOMA). This paper highlights the protocols used in the Inspirstion4 mission, which lay a foundation for precision astronaut medicine
- 10.Witze A 2022 was a record year for space launches. Nature 613, 426–426 (2023). [DOI] [PubMed] [Google Scholar]
- 11.The Starship Singularity | American Foreign Policy Council. https://www.afpc.org/publications/policy-papers/the-starship-singularity. [Google Scholar]
- 12.Cichan T et al. Mars Base Camp: An Architecture for Sending Humans to Mars. New Space 5, 203–218 (2017). [Google Scholar]
- 13.Garrett-Bakelman FE et al. The NASA Twins Study: A multidimensional analysis of a year-long human spaceflight. Science 364, eaau8650 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Urquieta E, Wu J, Hury J & Donoviel D Establishment of an open biomedical database for commercial spaceflight. Nat Med 28, 611–612 (2022). [DOI] [PubMed] [Google Scholar]
- 15.Open science in space. Nat Med 27, 1485–1485 (2021). [DOI] [PubMed] [Google Scholar]
- 16.Huff JL et al. Galactic cosmic ray simulation at the NASA space radiation laboratory - Progress, challenges and recommendations on mixed-field effects. Life Sci Space Res (Amst) 36, 90–104 (2023). [DOI] [PubMed] [Google Scholar]
- 17.Schimmerling W Genesis of the NASA Space Radiation Laboratory. Life Sci Space Res (Amst) 9, 2–11 (2016). [DOI] [PubMed] [Google Scholar]
- 18.Afshinnekoo E et al. Fundamental Biological Features of Spaceflight: Advancing the Field to Enable Deep-Space Exploration. Cell 183, 1162–1184 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Rydberg B Radiation-induced DNA damage and chromatin structure. Acta Oncol 40, 682–685 (2001). [DOI] [PubMed] [Google Scholar]
- 20.García-Nieto PE et al. Carcinogen susceptibility is regulated by genome architecture and predicts cancer mutagenesis. EMBO J 36, 2829–2843 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Okada R et al. Transcriptome analysis of gravitational effects on mouse skeletal muscles under microgravity and artificial 1 g onboard environment. Sci Rep 11, 9168 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.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 14, 1324 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kennedy EM et al. Galactic Cosmic Radiation Induces Persistent Epigenome Alterations Relevant to Human Lung Cancer. Sci Rep 8, 6709 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Rao SSP et al. A 3D Map of the Human Genome at Kilobase Resolution Reveals Principles of Chromatin Looping. Cell 159, 1665–1680 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Muratani M Cell-free RNA analysis of plasma samples collected from six astronauts in JAXA Cell-Free Epigenome (CFE) Study. (2022) doi: 10.26030/r2xr-h714. [DOI] [Google Scholar]
- 35. Wu F et al. & et al. Single Cell Analysis Identifies Conserved Features of Immune Dysfunction in Simulated Microgravity and Spaceflight. Nat Commun (2024) in press. Using single-cell analysis of human PBMCs exposed to short-term simulated microgravity, the team identified significant transcriptional alterations in immune cells, with monocytes showing the most pathway changes, increased retroviral and mycobacterial transcripts, and differences under stimulated conditions, providing insights into microgravity-induced immune dysfunction and potential countermeasures like quercetin
- 36.Heitzer E, Haque IS, Roberts CES & Speicher MR Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat Rev Genet 20, 71–88 (2019). [DOI] [PubMed] [Google Scholar]
- 37.Siravegna G, Marsoni S, Siena S & Bardelli A Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol 14, 531–548 (2017). [DOI] [PubMed] [Google Scholar]
- 38.Hummel EM et al. Cell-free DNA release under psychosocial and physical stress conditions. Transl Psychiatry 8, 236 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Teo YV et al. Cell-free DNA as a biomarker of aging. Aging Cell 18, e12890 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Drag MH & Kilpeläinen TO Cell-free DNA and RNA-measurement and applications in clinical diagnostics with focus on metabolic disorders. Physiol Genomics 53, 33–46 (2021). [DOI] [PubMed] [Google Scholar]
- 41.Zwirner K et al. Circulating cell-free DNA: A potential biomarker to differentiate inflammation and infection during radiochemotherapy. Radiother Oncol 129, 575–581 (2018). [DOI] [PubMed] [Google Scholar]
- 42.Pariset E et al. DNA Damage Baseline Predicts Resilience to Space Radiation and Radiotherapy. Cell Rep 33, 108434 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Brojakowska A et al. Retrospective analysis of somatic mutations and clonal hematopoiesis in astronauts. Commun Biol 5, 828 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Dawson S-J et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med 368, 1199–1209 (2013). [DOI] [PubMed] [Google Scholar]
- 45.Murtaza M et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 497, 108–112 (2013). [DOI] [PubMed] [Google Scholar]
- 46.Mencia-Trinchant N et al. Clonal Hematopoiesis Before, During, and After Human Spaceflight. Cell Rep 33, 108458 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Rutter L, et al. Release of CD36-associated cell-free mitochondrial DNA and RNA as a hallmark of space environment response. Nat Commun (2024) in press doi: 10.1038/s41467-023-41995-z.. A liquid biopsy study of six astronauts on >120-day ISS missions revealed mitochondria dysregulation in plasma cfRNA, identified a distinct extracellular mitochondria fraction associated with CD36, and showed broad tissue origins and systemic metabolic responses induced by microgravity, confirmed by mouse spaceflight data
- 48. da Silveira WA et al. Comprehensive Multi-omics Analysis Reveals Mitochondrial Stress as a Central Biological Hub for Spaceflight Impact. Cell 183, 1185–1201 e20 (2020). The first exosome profile of astronauts also showed enrichment of mtDNA and mtRNA from spaceflight
- 49.Bezdan D et al. Cell-free DNA (cfDNA) and Exosome Profiling from a Year-Long Human Spaceflight Reveals Circulating Biomarkers. iScience 23, 101844 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sangwung P, Petersen KF, Shulman GI & Knowles JW Mitochondrial Dysfunction, Insulin Resistance, and Potential Genetic Implications. Endocrinology 161, bqaa017 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Camera A et al. Identification and validation of multi-omics frailty biomarkers in human spaceflight. Comm Med (2024) in press doi: 10.1038/s41598-024-57948-5. Human space exploration causes molecular changes similar to aging, such as genomic instability and mitochondrial dysfunction, and using data from NASA’s GeneLab and astronaut missions, we identified gene expression changes related to frailty, suggesting that spaceflight may induce a frailty-like condition, warranting future studies on a frailty index for astronaut health
- 52.Caicedo A et al. Key Genes, Altered Pathways and Potential Treatments for Muscle Loss in Astronauts and Sarcopenic Patient. Research Square vol. PREPRINT (Version 1) Preprint at 10.21203/rs.3.rs-2819258/v1 (2023). [DOI] [Google Scholar]
- 53.Paul E et al. Disruption of Supv3L1 damages the skin and causes sarcopenia, loss of fat, and death. Mamm Genome 20, 92–108 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kamal K, Othman M, Kim J-H & Lawler J Bioreactor Development for Skeletal Muscle Hypertrophy and Atrophy by Manipulating Uniaxial Cyclic Strain: Proof of Concept. Res Sq (2023) doi: 10.21203/rs.3.rs-2437261/v1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Cannavo A et al. Are Skeletal Muscle Changes during Prolonged Space Flights Similar to Those Experienced by Frail and Sarcopenic Older Adults? Life (Basel) 12, 2139 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Cope H et al. More than a Feeling: Dermatological Changes Impacted by Spaceflight. Res Sq rs.3.rs-2367727 (2023) doi: 10.21203/rs.3.rs-2367727/v1. [DOI] [Google Scholar]
- 57. Park J, et al. Spatial multi-omics of human skin reveals KRAS and inflammatory responses to spaceflight. Nat Commun (2024). The first-ever skin biopsies from astronauts showed the disruption of KRAS pathways, movement of sub-dermal immune cell populations, distinct gene expression responses compares to whole blood profiles, and distinct inflammatory signatures
- 58.Desai RI et al. Complex 33-beam simulated galactic cosmic radiation exposure impacts cognitive function and prefrontal cortex neurotransmitter networks in male mice. Nat Commun 14, 7779 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Masarapu Y et al. Spatially resolved multiomics on the neuronal effects induced by spaceflight. Res Sq (2023) doi: 10.21203/rs.3.rs-2865086/v1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Houerbi N, Kim J & et al. Secretome profiling captures acute changes in oxidative stress, brain homeostasis and coagulation from spaceflight. Nat Commun (in press). As spaceflight becomes more common with commercial crews, blood-based measures of crew health, including plasma protein, metabolite, and EVP profiling from the SpaceX Inspiration4 crew, reveal significant and often lasting changes in coagulation, oxidative stress, and brain-enriched proteins, highlighting the potential for spaceflight biomarkers to guide astronaut biomedicine and countermeasures, as well as possible disruption in blood-brain barrier functions
- 61.Beheshti A et al. Countermeasures for cardiac fibrosis in space travel: It takes more than a towel for a hitchhiker’s guide to the galax. Research Square Preprint at 10.21203/rs.3.rs-2351744/v1 (2023). [DOI] [Google Scholar]
- 62. McDonald T et al. Space Radiation Damage Rescued by Inhibition of Key Spaceflight Associated miRNAs. Nature Commun (2024) in press. This manuscript demonstrated that antagomirs targeting key miRNAs associated with cardiovascular risk can reduce DNA double-strand break repair foci, inflammation, and DNA damage in 3D human microvasculature tissue models exposed to Galactic Cosmic Rays, potentially offering a countermeasure for space radiation effects, as supported by astronaut data from Inspiration4 and JAXA missions
- 63. Walsh S et al. Cosmic Kidney Disease: The Effects of Spaceflight and Galactic Cosmic Radiation on Renal Structure and Function. Res Sq (2023) doi: 10.21203/rs.3.rs-2982830/v1. Deep space environmental effects on kidney function, studied through mammalian samples from low Earth orbit and simulated galactic cosmic radiation, reveal renal tubular remodeling, mitochondrial dysfunction, impaired ion transport, and disrupted protein turnover, suggesting long-term spaceflight risks substantial kidney damage and associated health issues
- 64.Olde Engberink RHG et al. The kidney, volume homeostasis and osmoregulation in space: current perspective and knowledge gaps. npj Microgravity 9, 1–8 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.López-Otín C, Blasco MA, Partridge L, Serrano M & Kroemer G The Hallmarks of Aging. Cell 153, 1194–1217 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Cromwell RL et al. Overview of the NASA 70-day Bed Rest Study. Med Sci Sports Exerc 50, 1909–1919 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Meck JV, Reyes CJ, Perez SA, Goldberger AL & Ziegler MG Marked exacerbation of orthostatic intolerance after long- vs. short-duration spaceflight in veteran astronauts. Psychosom Med 63, 865–873 (2001). [DOI] [PubMed] [Google Scholar]
- 68.Tu D et al. Dynamic ensemble prediction of cognitive performance in spaceflight. Sci Rep 12, 11032 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Ramisetty BCM & Sudhakari PA Bacterial ‘Grounded’ Prophages: Hotspots for Genetic Renovation and Innovation. Front Genet 10, 65 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Koskella B & Brockhurst MA Bacteria–phage coevolution as a driver of ecological and evolutionary processes in microbial communities. FEMS Microbiol Rev 38, 916–931 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Vigil-Stenman T, Ininbergs K, Bergman B & Ekman M High abundance and expression of transposases in bacteria from the Baltic Sea. ISME J 11, 2611–2623 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Irby I & Broddrick J Microbial adaptation to spaceflight is correlated with bacteriophage-encoded functions. Res Sq (2023) doi: 10.21203/rs.3.rs-2602810/v1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Tierney B, Kim J & et al. The microbiome architecture of short-term spaceflight and its potential link to host immune activation. Nat Microbiol (2024). The largest microbiome profile to date featured >750 shotgun metagenome and metatranscriptome, revealing viral activation, rapid microbial transfer between crew members, and immune cell interactions associated with spaceflight
- 74.Singh VK, Romaine PLP & Seed TM Medical Countermeasures for Radiation Exposure and Related Injuries: Characterization of Medicines, FDA-Approval Status and Inclusion into the Strategic National Stockpile. Health Phys 108, 607–630 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Malkani S et al. Circulating miRNA Spaceflight Signature Reveals Targets for Countermeasure Development. Cell Rep 33, 108448 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.D’Ippolito D & Pisano M Dupilumab (Dupixent): An Interleukin-4 Receptor Antagonist for Atopic Dermatitis. P T 43, 532–535 (2018). [PMC free article] [PubMed] [Google Scholar]
- 77.Carnell LS Spaceflight medical countermeasures: a strategic approach for mitigating effects from solar particle events. International Journal of Radiation Biology 97, S125–S131 (2021). [DOI] [PubMed] [Google Scholar]
- 78.Peanlikhit T, Honikel L, Liu J, Zimmerman T & Rithidech K Countermeasure efficacy of apigenin for silicon-ion-induced early damage in blood and bone marrow of exposed C57BL/6J mice. Life Sciences in Space Research 35, 44–52 (2022). [DOI] [PubMed] [Google Scholar]
- 79.Smith SM, Zwart SR, Block G, Rice BL & Davis-Street JE The Nutritional Status of Astronauts Is Altered after Long-Term Space Flight Aboard the International Space Station1. The Journal of Nutrition 135, 437–443 (2005). [DOI] [PubMed] [Google Scholar]
- 80.Scott JM et al. Effects of exercise countermeasures on multisystem function in long duration spaceflight astronauts. npj Microgravity 9, 1–9 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Dynan WS, Chang PY, Sishc BJ & Elgart SR Breaking the limit: Biological countermeasures for space radiation exposure to enable long-duration spaceflight. Life Sciences in Space Research 35, 1–3 (2022). [DOI] [PubMed] [Google Scholar]
- 82. Meydan C et al. Improved gastrointestinal health for irritable bowel syndrome with metagenome-guided interventions. Precision Clinical Medicine 3, 136–146 (2020). This study showed how matching probiotics to the gut profiles of patients can improve patient metrics inflammation and discomfort. Such microbe-host matching can guide similar deployment for future spaceflight missions
- 83. Schmidt MA, Jones JA & Mason CE Optimizing human performance in extreme environments through precision medicine: From spaceflight to high-performance operations on Earth. Cambridge Prisms: Precision Medicine 1, e27 (2023). Precision medicine in spaceflight includes pharmacogenomics, pharmacokinetics, and matching the response of an astronaut to the blood chemistry, as well as other molecular personalization tools
- 84.Pavez Loriè E et al. The Future of Personalized Medicine in Space: From Observations to Countermeasures. Frontiers in Bioengineering and Biotechnology 9, (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Schmidt MA, Schmidt CM & Goodwin TJ Pharmacogenomics in Spaceflight. in Handbook of Space Pharmaceuticals (eds. Pathak Y, Araújo dos Santos M & Zea L) 1–39 (Springer International Publishing, Cham, 2018). doi: 10.1007/978-3-319-50909-9_26-1. [DOI] [Google Scholar]
- 86.Handbook of Space Pharmaceuticals. (Springer, 2022). [Google Scholar]
- 87.Schmidt MA, Meydan C, Schmidt CM, Afshinnekoo E & Mason CE Elevation of Gut-Derived p-Cresol During Spaceflight and its Effect on Drug Metabolism and Performance in Astronauts. 2020.11.10.374645 Preprint at 10.1101/2020.11.10.374645 (2020). [DOI] [Google Scholar]
- 88.Frangoul H et al. CRISPR-Cas9 Gene Editing for Sickle Cell Disease and β-Thalassemia. N Engl J Med 384, 252–260 (2021). [DOI] [PubMed] [Google Scholar]
- 89.Hilton IB et al. Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nat Biotechnol 33, 510–517 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Mason CE The Next 500 Years: Engineering Life to Reach New Worlds. (The MIT Press, 2021). doi: 10.7551/mitpress/12585.001.0001. This book details the insights from the NASA Twins Study, the genetic and epigenetic technologies that could enable life to survive on other planets beyond Earth, and the possible new planets and moons that could be the foci of future missions
- 91.Casaletto J et al. Analyzing the Relationship Between Gene Expression and Phenotype in Space-Flown Mice Using a Causal Inference Machine Learning Ensembl. Research Square Preprint at 10.21203/rs.3.rs-2332064/v1 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Liu T et al. Bone marrow adiposity modulation after long duration spaceflight in astronauts. Nat Commun 14, 4799 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Westover C et al. Multi-omics Analysis of Dsup Expressing Human Cells Reveals Open Chromatin Architectural Dynamics Underyling Radioprotection. 2020.11.10.373571 Preprint at 10.1101/2020.11.10.373571 (2022). [DOI] [Google Scholar]
- 94.Chavez C, Cruz-Becerra G, Fei J, Kassavetis GA & Kadonaga JT The tardigrade damage suppressor protein binds to nucleosomes and protects DNA from hydroxyl radicals. eLife 8, e47682 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Aguilar R et al. Multivalent binding of the tardigrade Dsup protein to chromatin promotes yeast survival and longevity upon exposure to oxidative damage. Res Sq rs.3.rs-3182883 (2023) doi: 10.21203/rs.3.rs-3182883/v1. [DOI] [Google Scholar]
- 96.Rahimzadeh V et al. Ethically cleared to launch? Science 381, 1408–1411 (2023). [DOI] [PubMed] [Google Scholar]
- 97. Rutter L, et al. & et al. Protective alleles and precision healthcare in crewed spaceflight. Nat Commun (in press). Protective genes confer the greatest adaptability for stressful environments, and this paper also describes dCas9 systems and other means to transiently activate genes that might be beneficial for spaceflight
- 98.Seylani A, Galsinh AS, Tasoula A & et al. Ethical Considerations for the Age of Non-Governmental Space Exploration. Nature Communications. Nat Commun (2024). doi: 10.1038/s41467-023-44357-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Rutter L, et al. & et al. Astronaut omics and the impact of space on the human body at scale. Nat Commun (2024). doi: 10.1038/s41467-024-47237-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Lieberman P, Morey A, Hochstadt J, Larson M & Mather S Mount Everest: a space analogue for speech monitoring of cognitive deficits and stress. Aviat Space Environ Med 76, B198–207 (2005). [PubMed] [Google Scholar]
- 101.Li L, Zhou Y, Zou S & Wang Y The Effects of High-Altitude Mountaineering on Cognitive Function in Mountaineers: A Meta-Analysis. Int J Environ Res Public Health 20, 5101 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Austermann K et al. Effects of antioxidant supplementation on bone mineral density, bone mineral content and bone structure in healthy men during 60 days of 6° head-down tilt bed rest: Results from a randomised controlled trial. Nutr Bull 48, 256–266 (2023). [DOI] [PubMed] [Google Scholar]
- 103.McGrath ER et al. Bone metabolism during strict head-down tilt bed rest and exposure to elevated levels of ambient CO2. NPJ Microgravity 8, 57 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.McGregor HR et al. Ophthalmic changes in a spaceflight analog are associated with brain functional reorganization. Human Brain Mapping 42, 4281–4297 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Strewe C et al. Sex differences in stress and immune responses during confinement in Antarctica. Biol Sex Differ 10, 20 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Steinach M et al. Sleep Quality Changes during Overwintering at the German Antarctic Stations Neumayer II and III: The Gender Factor. PLoS One 11, e0150099 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Nwanaji-Enwerem JC et al. A Longitudinal Epigenetic Aging and Leukocyte Analysis of Simulated Space Travel: The Mars-500 Mission. Cell Reports 33, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Price H, Shishko R, Mrozinski J & Woolley R Concept for 2033 Crewed Mars Orbital Mission with Venus Flyby. Journal of Spacecraft and Rockets 60, 49–58 (2023). [Google Scholar]
- 109.Hera Mission. Hera Mission https://www.heramission.space (2023).
- 110.Lucy - NASA Science. https://science.nasa.gov/mission/lucy/.
- 111.OSIRIS-REx - NASA Science. https://science.nasa.gov/mission/osiris-rex/.
- 112.Psyche Mission | A Mission to a Metal World. Psyche Mission https://psyche.asu.edu/. [Google Scholar]
- 113.NASA - NSSDCA - Spacecraft - Details. https://nssdc.gsfc.nasa.gov/nmc/spacecraft/display.action?id=2005-014A.
- 114.Breakthrough Initiatives. https://breakthroughinitiatives.org/initiative/3.
- 115.JHUAPL. Dragonfly. Dragonfly https://dragonfly.jhuapl.edu/.
- 116.NASA’s Europa Clipper. NASA’s Europa Clipper https://europa.nasa.gov/.
- 117.Juice. https://www.esa.int/Science_Exploration/Space_Science/Juice.
- 118.Gaganyaan. https://www.isro.gov.in/Gaganyaan.html.
- 119.Virgin Galactic| Spaceflight. https://brochure.virgingalactic.com/spaceflight/.
- 120. Mars Base Camp. Lockheed Martin https://www.lockheedmartin.com/en-us/products/mars-base-camp.html. This mission and station plan would enable continued orbital presence around Mars for human missions
- 121.Instruments | Emirates Mars Mission. https://www.emiratesmarsmission.ae/hope-probe/instruments/.
- 122.Frąckiewicz M Mangalyaan-2 Mission: ISRO’s Secret Plan for a Second Mars Mission. TS2 SPACE; https://ts2.pl/en/mangalyaan-2-mission-isros-secret-plan-for-a-second-mars-mission/ (2023). [Google Scholar]
- 123.mars.nasa.gov. Mars 2020 Perseverance Rover - NASA. https://mars.nasa.gov/mars2020/.
- 124.mars.nasa.gov. Mars Sample Return - NASA. https://mars.nasa.gov/msr/.
- 125.NASA - NSSDCA - Spacecraft - Details. https://nssdc.gsfc.nasa.gov/nmc/spacecraft/display.action?id=2020-049A#:~:text=Tianwen%201%20(formerly%20Huoxing%201,reaches%20Mars%20in%20February%202021.
- 126.MMX - Martian Moons eXploration. MMX - Martian Moons eXploration http://mmx.isas.jaxa.jp/.
- 127.ExoMars rover. https://www.esa.int/Science_Exploration/Human_and_Robotic_Exploration/Exploration/ExoMars/ExoMars_rover.
- 128.SpaceX. SpaceX http://www.spacex.com.
- 129.https://www.nasa.gov/wp-content/uploads/static/artemis/NASA: Artemis. https://www.nasa.gov/wp-content/uploads/static/artemis/NASA https://www.nasa.gov/specials/artemis/index.html.
- 130.Artemis I https://www.esa.int/Science_Exploration/Human_and_Robotic_Exploration/Orion/Artemis_I.
- 131.Artemis II. https://www.esa.int/Science_Exploration/Human_and_Robotic_Exploration/Orion/Artemis_II.
- 132.International Lunar Research Station (ILRS) Guide for Partnership. https://www.cnsa.gov.cn/english/n6465652/n6465653/c6812150/content.html.
- 133.Future Chinese Lunar Missions. https://nssdc.gsfc.nasa.gov/planetary/lunar/cnsa_moon_future.html.
- 134.JAXA | Smart Lander for Investigating Moon (SLIM). JAXA | Japan Aerospace Exploration Agency; https://global.jaxa.jp/projects/sas/slim/. [Google Scholar]
- 135.DAVINCI Homepage - Probe and Flyby Mission To Venus Atmosphere NASA Goddard. https://ssed.gsfc.nasa.gov/davinci/.
- 136.https://www.jpl.nasa.gov. VERITAS. NASA Jet Propulsion Laboratory (JPL) https://www.jpl.nasa.gov/missions/veritas. [Google Scholar]
- 137.EnVision factsheet. https://www.esa.int/Science_Exploration/Space_Science/EnVision_factsheet.
- 138.BepiColombo. https://www.esa.int/Science_Exploration/Space_Science/BepiColombo.
- 139.Rienecker KDA et al. Combined space stressors induce independent behavioral deficits predicted by early peripheral blood monocytes. Sci Rep 13, 1749 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Borg J et al. Spatiotemporal Expression and Control of Haemoglobin in Space. Res Sq (2023) doi: 10.21203/rs.3.rs-3083058/v1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Grigorev K, et al. Direct RNA sequencing of astronauts reveals spaceflight-associated epitranscriptome changes and stress-related transcriptional responses. Nat Commun (2024). This was the first application of native RNA sequence analysis to astronaut samples, and detailed the expression, isoform, and also RNA modification dynamics associated with spaceflight
- 142.Werneth CM, et al. Considering Clonal Hematopoiesis of Indeterminate Potential in Space Radiation Risk Analysis for Hematologic Cancers and Cardiovascular Disease. Commun Med (2024). doi: 10.1038/s43856-023-00408-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Simpson AC et al. Phylogenetic affiliations and genomic characterization of novel bacterial species and their abundance in the International Space Station. Res Sq rs.3.rs-3126314 (2023) doi: 10.21203/rs.3.rs-3126314/v1. [DOI] [Google Scholar]
- 144.Li K et al. Explainable machine learning identifies multi-omics signatures of muscle response to spaceflight in mice. npj Microgravity 9, 1–10 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Scott RT et al. Biomonitoring and precision health in deep space supported by artificial intelligence. Nat Mach Intell 5, 196–207 (2023). [Google Scholar]
- 146.Ilangovan H et al. Spaced Out Data No More: Genomic Harmonization Meets Machine Learning in Murine Livers. Res Sq (2023) doi: 10.21203/rs.3.rs-2827816/v1. [DOI] [Google Scholar]
- 147.Sanders LM et al. Biological research and self-driving labs in deep space supported by artificial intelligence. Nat Mach Intell 5, 208–219 (2023). [Google Scholar]
- 148.Adamopoulos, et al. NASA GeneLab derived microarrays studies of Mus Musculus and Homo sapiens organisms in altered gravitational conditions. npj Microgravity (in review). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Costes SV, Gentemann CL, Platts SH & Carnell LS Biological Horizons: Pioneering Open Science in the Cosmos. Nat Commun (in review). The impact of open science and the GeneLab data repositories have enabled dozens of papers to be quickly derived from the Inspiration4 mission, within only two years, providing a model for future spaceflight data and analysis collaboration
- 150.Sanders LM et al. Inspiration4 Data Access through the NASA Open Science Data Repository. npj Microgravity (in press). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Barisic D, Chin CR, Meydan C, Teater M, Tsialta I, Mlynarczyk C, Chadburn A, Wang X, Sarkozy M, Xia M, Carson SE, Raggiri S, Debek S, Pelzer B, Durmaz C, Deng Q, Lakra P, Rivas M, Steidl C, Scott DW, Weng AP, Mason CE, Green MR, Melnick A. ARID1A orchestrates SWI/SNF-mediated sequential binding of transcription factors with ARID1A loss driving pre-memory B cell fate and lymphomagenesis. Cancer Cell. 2024. Apr 8;42(4):583–604.e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Mathyk B, et al. Understanding how space travel affects the female reproductive system to the Moon and beyond. In press. npj Women’s Health (2024) doi: 10.1038/s44294-024-00009-z [DOI] [Google Scholar]
