Skip to main content

Synergistic interplay between radiation and microgravity in spaceflight-related immunological health risks

Abstract

Spaceflight poses a myriad of environmental stressors to astronauts´ physiology including microgravity and radiation. The individual impacts of microgravity and radiation on the immune system have been extensively investigated, though a comprehensive review on their combined effects on immune system outcomes is missing. Therefore, this review aims at understanding the synergistic, additive, and antagonistic interactions between microgravity and radiation and their impact on immune function as observed during spaceflight-analog studies such as rodent hindlimb unloading and cell culture rotating wall vessel models. These mimic some, but not all, of the physiological changes observed in astronauts during spaceflight and provide valuable information that should be considered when planning future missions. We provide guidelines for the design of further spaceflight-analog studies, incorporating influential factors such as age and sex for rodent models and standardizing the longitudinal evaluation of specific immunological alterations for both rodent and cellular models of spaceflight exposure.

Introduction

The spaceflight environment induces detrimental effects to the human body, resulting in a wide range of physiological and psychological changes. These range from DNA damage and cell cycle dysregulations, to neuro-ocular conditions, gastrointestinal microbiota alterations, circadian rhythm changes, inflammation, metabolomic changes, and more [1,2,3,4,5,6]. Two of the most significant stress factors in space are cosmic radiation and microgravity [7]. With the National Aeronautics & Space Administration (NASA) Artemis missions to the Moon and Mars, relative immunological risks from microgravity and radiation exposure will vary throughout the course of the mission. A nuanced understanding of the independent and interactive immunological effects of microgravity and radiation will be instrumental in predicting health risks accurately; this will point the way to prioritizing countermeasure development accordingly for astronaut health and safety. Therefore, the ultimate focus of this review is to detail the interactive immunological effects of combined exposure to microgravity and radiation observed through ground-based spaceflight-analog studies. Relevant observations obtained from astronauts on spaceflight missions to the International Space Station (ISS) will be compared and discussed (Table 1). Lastly, we provide guidelines for further systematic and rigorous exploration of immune dysregulations from spaceflight exposure.

Table 1 Comparing spaceflight-analog studies to cytokine perturbations in astronauts

Microgravity and its terrestrial analogs

Well-characterized risks of microgravity include bone loss, muscle atrophy, and cardiovascular and neuro-ocular fluid shifts that have been reviewed extensively elsewhere [6, 14]. Countermeasures have been effectively implemented against some of these effects, such as resistance exercises to prevent muscle weakening [15], though it may be difficult to continue these in newer deep-space exploration spacecraft [16]. There are obvious ethical, health, and logistical limitations to performing repeated and controlled studies exploring the effects of radiation and microgravity in humans, limiting the development of novel mission-critical countermeasures. Therefore, research on microgravity effects is often performed on the ground using analogs that have also been extensively described elsewhere [17, 18]. Briefly, the hindlimb unloading (HU) rodent model involves lifting the rodents’ hindlimbs off the cage floor by tethering its tail to a support bar above [19]. Forelimb weight-bearing is generally unperturbed. This results in a decreasing load borne by the hindlimbs contributing to bone and muscle atrophy [20]. HU also contributes to a cephalic fluid shift analogous to that observed in astronauts [19], enabling the study of cardiovascular, neurological, ophthalmic perturbations experienced in space [1].

More recently, the use of partial weight bearing (PWB) rodent models has been developed to mimic reduced quadrupedal weight loading [21,22,23]. By using a harness to lift all rodent limbs off the cage floor, the effects of gravity can be explored as a continuum between microgravity, partial unloading similar to that on Mars, and full loading as on Earth [24]. Many studies that make use of PWB suspension focus on musculoskeletal parameters, with a direct association between degree of gravity loading and musculoskeletal atrophy. However, there does not seem to be any threshold effect whereby skeletal and muscular disuse losses are minimized [25, 26]. The PWB does not create a cephalic fluid shift [27] as in the HU model and its use may be largely minimized to musculoskeletal studies as a result. Mortreux and colleagues did demonstrate that the PWB model did not induce a state of chronic inflammation but to date, no studies have examined the direct relationship between PWB and possible immune disruptions.

Cell culture analogs, such as a rotating wall vessel (RWV) or random positioning machine (RPM), represent a benchtop method to mimic a hypogravic environment for spaceflight-analog studies. A RWV is a 2D microgravity simulator that employs continuous rotation around the horizontal axis to establish a “free fall” through the cell suspension medium, thus creating a low shear but mixed fluid environment [28]. Similarly, a RPM is a 3D microgravity simulator that rotates along both the x- and y- axis at different and random speeds for each axis [29]. In doing so, the gravity vector experienced by the cells is temporally randomized to an average of zero [30, 31]. It is important to note that the rotation rate of the vessel must be faster than the biological process in study but not so fast as to introduce centrifugal forces that cause cellular accumulations in certain areas [32].

Some recent studies have also been successful in simulating microgravity without the use of rotation by instead using a magnetic field to counterbalance the gravity force. This also allows for the investigation of faster cellular processes that would otherwise be unobservable with RWV or RPM [33]. Positive magnetophoresis, where cells bind to a labelled magnetic bead, levitates the cell by acting at the cell membrane surface and thus does not truly simulate microgravity as internal structures do not experience the same forces [33]. Additionally, it necessitates the formation of dense cellular aggregates [34], and deviations from the locations of magnetic equilibrium may result in certain cells experiencing net forces of up to 2g [35], obfuscating the responses to microgravity being studied. Negative magnetophoresis on the other hand applies a magnetic force to the cell culture and medium, wholly counterbalancing the gravitational force to achieve truer weightlessness [33, 36]. Diamagnetic objects such as cells can experience stable magnetic levitation in the presence of strong magnetic fields, though these can induce various biological effects including weakened immune function [37,38,39].

Finally, neutral buoyancy, such as that achieved in parabolic flights with sub-orbital sounding rockets, also represents another technique employed for spaceflight-analog studies [30, 40]. The true weightlessness experienced reflects the closest approximation to spaceflight conditions outside of experiments conducted onboard the ISS. While this allows for investigation of certain immune functions such as metabolics [40] or signal transduction [41], parabolic flights achieve relatively short hypogravic-periods (seconds to minutes), limiting their practical uses for long-term functional immune assays. Nonetheless, they may represent an ideal study design to study the brief periods of launch and descent during which astronauts are exposed to hypergravitational states. Recent short-term hypergravity studies have indicated alterations in intrinsic apoptotic signaling pathways [42], cytoskeleton organization in T cells [43], post-transcriptional modifications in T cells [44], increased gene expression of Aire and RANK for medullary thymic epithelial cells [45], and even prevention of microgravity-induced impairments in T cell activation [46]. Mice exposed to long-term hypergravity conditions demonstrated disrupted intestinal microbiota [47], modified T cell receptor diversity [48, 49], and more [50]. As we move away from the dichotomous perspective of gravity as a binary (0g or 1g), and instead consider it on a continuum, long-term exposure to hypergravity may represent the next step in understanding the immunological effects of gravity in a dose-associated manner.

Space radiation and its terrestrial analogs

Radiation encountered by astronauts during spaceflight largely consists of galactic cosmic radiation (GCR) that originates outside the solar system [51]. Exposure to GCR consists of both proton and heavy-ion high-linear energy transfer (LET) radiation which can be more damaging to biological systems than X- or gamma rays of the same level of doses [52, 53]. Although protons and alpha particles make up the majority of the relative flux of GCR dosages, significant dosages also come from carbon, oxygen, silicon, iron, zirconium, barium, platinum, and lead ions [54]. Protective abilities of current spacecrafts to such exposures may be limited, as the GCR nuclei have been shown to easily pass through thicker shields without significant losses in intra-vehicular radiation dose [55,56,57]. Solar particle events (SPE) such as solar flares present another possible radiation hazard. They can transmit massive amounts of proton irradiation in a matter of hours or days, potentially causing acute radiation sicknesses including nausea, gastrointestinal pain and discomfort and faintness [58, 59]. In low Earth orbit (LEO) such as the orbit of the ISS, astronauts are also exposed to protons trapped in the Earth’s geomagnetic field [58]. Chronic exposure to radiation can increase the risk of immunosuppression [60], heart disease [61], carcinogenesis [62], and more [59].

Spaceflight-analog studies vary widely in their use of radiation exposures. Often, acute doses of proton or gamma radiation are utilized such as for SPE simulation, with a few studies employing heavy-ion high-LET exposures from the NASA Space Radiation laboratory (NSRL) at Brookhaven National Laboratory (BNL) for GCR simulation. The NSRL SPE model uses proton irradiation beams ranging in energy from 50 MeV/n (90.3% of the total dose) up to 150 MeV/n (0.15% of the total dose). This SPE model is similar to the fluence, or photon incidence per cross-sectional area, of the August 1972 SPE event and the energy spectrum of the March 1989 event [63]. The GCR simulation at NSRL is a one-hour exposure that utilizes different energy exposures of proton and alpha particle irradiation as well as heavy-ion deliveries of carbon, oxygen, silicon, titanium, and iron [64]. Finally, NSRL also has a simplified GCR simulation exposure of approximately twenty minutes consisting of proton, alpha particle, oxygen, silicon, and iron exposures [65]. It is not immediately clear whether the simplified GCR exposure adequately achieves the same physiological assaults on the immune system as the full GCR simulation as no direct comparative studies have been performed.

Astronaut health and safety remains the top priority for spaceflight missions, and recent advancements in computational modeling may offer promising avenues to simulate the effects of space radiation on biological systems. The same externally applied dose of radiation will result in different effective local doses and relative biological effects experienced by deeper organs. For example, thymic mass was elevated over a month after whole-body heavy-ion irradiation whereas spleen, liver, and lung masses were unchanged [66]. By incorporating data from space missions, ground-based experiments, and particle accelerator research, these models can aid in predicting differential effects of radiation doses and assessing potential health risks for astronauts [67].

Immune disruptions in astronauts

Spaceflight studies have observed a variety of immunological alterations ranging from T cell function, lymphocyte proliferation, and cytokine production summarized recently [68,69,70,71]. For instance, primary lymphoid organs such as bone marrow and the thymus indicate reduced T-cell maturation and proliferation during spaceflight [72]. Astronauts also have increased susceptibility to infection [73], which may be due to increased pathogen virulence [71, 74], viral reactivation [75], or viral shedding [76]. Astronauts experience decreased natural killer (NK) cell numbers and functional activity [77, 78]; changes in white blood cell populations such as neutrophils, monocytes, helper T cells, and B cells [71, 79, 80]; and dysregulated cytokine levels during or after spaceflight [77, 78, 81]. This compromised immunity, as seen in spaceflight travelers and analogs thereof [82, 83], may put them at risk for widespread pathogenic invasions in space [78, 84].

For short-duration missions, increased levels of the following cytokines have been observed in astronauts onboard the ISS: tumor necrosis factor-α (TNFα), interferon (IFN)-α, IFN-γ, interleukin (IL)-1b, IL-4, IL-10, and IL-12 [11]. It is also critical to note the differing cytokine disruptions in short-duration missions compared to long-duration missions. In long-duration missions, increased levels of TNFα, IL-1ra, IL-8, thrombopoietin, vascular endothelial growth factor (VEGF), C-C motif chemokine ligand (CCL) 2, CCL4, and C-X-C motif chemokine ligand (CXCL) 5 were observed. There were additionally no changes in plasma levels of IL-1a/b, IL-2, IL-4, IL-5, IL-17, CCL3, CCL5, IFNγ, granulocyte colony-stimulating factor (G-CSF), and basic fibroblast growth factor (FGF) [8]. Some reports have differed on whether levels of IL-10, IL-12, and granulocyte-macrophage (GM)-CSF have no change or decrease during long-duration spaceflight [8, 9].

As a result, concordance between observations in astronauts and spaceflight-analog studies has had differing successes (Table 1). Spaceflight-analog studies must be carefully designed in order to provide an accurate comparison for astronaut health and performance–i.e., short-term spaceflight-analog studies may lack the chronic immune perturbations necessary to identify certain dysfunctions. A key example here is an increase in IL-12 levels detected by human peripheral blood mononuclear cells (PBMCs) in concordance with observations from astronauts on short-duration spaceflight missions [11] but not long-duration missions [9].

It becomes obvious that both microgravity (µG) and space radiation (IR) in the space environment imply diverse impaired immune function [85,86,87]. However, the vast majority of spaceflight-analogs examine each risk individually; studies that investigate the combined physiological effects of µG and IR are critical for accurately assessing the risks associated with space exploration and for effective countermeasure development [1, 88]. It is difficult to directly determine the relative contribution of µG, IR and other stress factors to the observed changes on immune health in spaceflight [89]. Investigations with human subjects are limited by small sample sizes, relatively small number of flights available, and unstandardized experimental conditions [90]. Using spaceflight-analogs such as HU mouse models and RWV cell culture studies can therefore prove an effective way to understand the modulatory interactions between µG and IR for certain immune endpoints (Fig. 1). Indeed, evident biological effects of combined µG and IR have been previously reviewed in skeletal, ocular, central nervous system, cardiovascular, and stem cell responses using these models [1, 88, 91,92,93,94,95]. Herein we focus specifically on how µG-analogs and space IR-analogs independently and interactively modulate the immune system, rather than a simple summary of physiological changes observed.

Fig. 1
figure 1

Spaceflight and spaceflight-analog studies observe a diverse range of immunological perturbations from radiation and microgravity. Spaceflight-analog research utilizes space-like radiation and microgravity analogs such as rotating wall vessels and hindlimb suspension to understand the immunological perturbations that arise from spaceflight travel. These range from altered cytokine levels to changes in immune cell proliferation, activity, function, and more. Ultimately, spaceflight travelers experience vast immunological dysregulations which the spaceflight-analog research attempts to elucidate and mitigate

In vivo rodent models

Cytokines evoke vast inflammatory and anti-inflammatory signaling in the immune system, and regulation of their plasma levels is critical for proper immune function. Combined exposure rodent models indicate dysregulated cytokine levels similar to changes observed in astronauts’ plasmas collected while in space [9, 11, 12] (Fig. 2). For example, acute exposure to combined hindlimb unloading (HU) for simulated microgravity (sim-µG) and solar particle event-style radiation (IR) increased cytokine concentrations of IFN-α, IL-6, and TNFα, a synergistic change that was not as significantly observed in sim-µG-alone or IR-alone environments [13] (Fig. 3).

Fig. 2
figure 2

Serum levels of cytokines in astronauts during long- and short-duration spaceflight missions. Serum levels of (A) IFNα and (B) IL-7 in astronauts during long-duration spaceflight missions, from pre-launch (L), in-flight (FD and FLT), to return on Earth (R) [9]. Serum levels of (C) IL-12 and (D) TNFα in astronauts during short-duration spaceflight missions [11]. Astronauts experience significant aberrations in cytokine levels during spaceflight missions, regardless of duration. Data shown are mean ± standard deviation (SD). Significance was evaluated via a Student’s t test by comparing all other data points to L-180 baseline data. Figures re-created and modified with permission from [11] and [9] under a CC-BY unrestricted open access license

Fig. 3
figure 3

Synergistic effects of combined exposure spaceflight-analog rodent studies on cytokine levels and expression. Serum levels of (A) IFNα and (B) TNFα in rodents exposed to control or spaceflight-analog conditions of irradiation (IR), simulated microgravity (Sim-µG), or combined Sim-µG + IR exposure. Cytokine levels are significantly synergistically increased in combined spaceflight-analog studies. Data shown are the mean ± SD (n = 5/group in duplicate). Significance was evaluated with ANOVA analyses with post-hoc group comparisons using Bonferroni correction. Data shown are from a single experiment representative of three experiments. Figures re-created and modified with permission from [13] under a CC-BY unrestricted open access license

Studies on immunological endpoints for rodents flown on the ISS allow for more direct comparisons between spaceflight and ground-based spaceflight-analog observations. Female C57BL/6J mice flown on the STS-135 Space Shuttle Atlantis mission for 13 days were characterized for changes in splenic and thymic immune cell function and gene expression, adrenal catecholamine levels, and hepatic transcriptomics [96, 97]. Splenic mass and most splenic leukocyte subtypes were significantly depleted after spaceflight exposure, but demonstrated increased oxidative burst activity, phagocytosis, and gene expression patterns of endocytosis and peroxisome formation. Conversely, gene expression analysis indicated no increase in gene expression related to reactive oxygen species (ROS) metabolism. This discrepancy between functional activity and gene expression patterns is noteworthy, as other studies with rodents flown on the ISS also reported genetic downregulation of Nrf2 and Ptgs2, related to oxidative stress, and Tnf, related to the inflammatory response [98]. Post-spaceflight splenocytes also had decreased surface marker expression after ConA stimulation and decreased antigen presentation marker expression after Toll-like receptor (TLR) agonist stimulation [99]. In the thymus, no corresponding decrease in mass was observed, though there were higher levels of DNA fragmentation and changes in gene expression related to T cell activity and cancer immune surveillance. Both the ISS-flown rodents and some spaceflight-analog studies performed in rodents similarly reported decreased leukocyte populations [100, 101] and increased corticosterone levels [102]. However, other spaceflight-analog studies in rodents have reported differing observations such as no changes in splenic immune cells and decreases in thymic T cell populations after combined sim-µG + IR exposure [103]. This discrepancy may be due to the radiation shielding and other protective countermeasures on the ISS that cause the spaceflight studies to experience primarily microgravity exposure alone, as opposed to truly combined exposure. Their use as a direct comparison must therefore be approached cautiously. Depending on the particular spaceflight-analog study design, reproducible concordance with immunological perturbations measured in true spaceflight observations may or may not be possible.

Immune cell populations

Myeloid cells were affected by interactions between microgravity-analog and radiation exposure in spaceflight-analog studies. These interactions notably differed depending on the timepoint and the type of radiation exposure utilized: one or four days after exposure, and proton IR or gamma IR. For example, one day after sim-µG + proton-IR exposure (three days sim-µG, one 2.0 Gy 50 cGy/min proton-IR dose) there was an additive interaction between sim-µG and the proton-IR that slightly increased levels of serum CD14 markers for myeloid proliferation in female ICR mice [13]. Four days after the same sim-µG + proton-IR exposure, monocyte blood counts were decreased by nearly 70% even though individual microgravity-analog or radiation exposures had only minimally decreased the monocyte counts [100]. However, four days after sim-µG + gamma-IR (three days sim-µG, one 2.0 Gy 44 cGy/min gamma-IR dose), a synergistic interaction between sim-µG and gamma-IR significantly increased serum CD14 levels [13]. Neutrophil counts had no change after proton-IR alone and an increase after sim-µG-alone conditions, but after sim-µG + proton-IR, neutrophil count displayed a significant synergistic decrease in population fraction [100]. Granulocyte populations displayed an independent proton-IR-driven decrease 4 days post-exposure, but an interactive antagonistic decrease 16 days post-exposure [101]. Although the observed immune disruption is the same, the nuance of such synergistic interactions would ultimately be lost in single-hazard focused studies that do not incorporate both sim-µG and IR exposures into the study design.

In combined exposures, lymphoid cells display largely IR-driven decreases in population counts though some reports differ on the specific effects of sim-µG-alone exposures [100, 101]. Population counts of CD3+/CD8 + cytotoxic T cells were unchanged four days after exposure to sim-µG or proton-IR alone, but significantly decreased by approximately 50% in combined exposure [100] (Fig. 4). This is another key example of a synergistic interaction between radiation and microgravity perturbing immune ability that would not otherwise be obvious from studies investigating sim-µG or proton-IR alone. A similar synergistic pattern was observed for the decrease of the T lymphocyte proliferation index twenty-one days after exposure in female ICR mice [100] (Fig. 4). In male C57BL/6J mice, acute gamma-IR alone did not influence natural killer T cell (NK T) and natural killer 1 cell (NK 1) populations, whereas chronic sim-µG + gamma-IR exposure over thirty days caused synergistic decreases in thymic regulatory T cells (Treg), NK T cells, and NK 1 cells [103] (Fig. 4). Interestingly, these changes were not observed in splenic immune cell populations [103], so observed synergistic interactions are additionally specific to cell type, location, and IR dose. Immune effects are also clearly influenced by chronicity of exposure to spaceflight-analog conditions, radiation source, and post-exposure readaptation time.

Fig. 4
figure 4

Synergistic effects of combined exposure spaceflight-analog rodent studies on immune cell proliferation. Rodents were exposed to control or spaceflight-analog conditions of irradiation (IR), simulated microgravity (Sim-µG), or combined Sim-µG + IR exposure. (A) Population of CD3+/CD8 + cytotoxic T cells is greatly depleted in combined exposure compared to control. (B) T lymphocyte proliferation rate in rodents exposed to Sim-µG + IR is significantly lower than control, IR-alone, and Sim-µG-alone exposures. Data shown are the mean ± SD (n = 4–6/group) [100]. Populations proportions of (C) CD3+/CD335 + thymic regulatory T (Treg) cells, (D) CD27+/CD335- natural killer (NK) T cells, and (E) CD3+/CD335 + NK 1 cells are also altered in combined spaceflight-analog exposure compared to IR-alone exposure. Data shown are mean ± confidence interval (CI) (n = 8/group); significance was evaluated with ANOVA and post-hoc analyses [103]. Figures re-created and modified with permission from [100] under a CC-BY unrestricted open access license and [103] under an Elsevier user license within STM permissions guidelines

Sex differences

Such perturbations can significantly compromise the immune system’s functional ability to clear infections [73, 82, 83, 104]. Circulating levels of lipopolysaccharides (LPS) were synergistically increased four days after combined exposure in female ICR mice, a result of reduced gastrointestinal ability to contain gram-negative bacterial products [13]. Mice exposed to sim-µG + proton-IR had significantly increased bacterial blood counts after challenge with Pseudomonas aeruginosa or Klebsiella pneumoniae that was not reflected in the sim-µG-alone or proton-IR-alone conditions [102]. There was a further synergistic impairment of granulocyte proliferation from combined exposure [102], reflecting a state of immunocompromise and susceptibility to pathogenic invasions that astronauts are likely subject to as well [78, 82,83,84, 104].

This immune dysfunction is heightened in female mice, contributing to significantly poorer health outcomes after in vivo bacterial challenge. In sim-µG-alone, gamma-IR alone, and proton-IR alone conditions, CH3/HeN and Balb/c female mice had increased morbidity compared to respective male mice three to five days after systemic P. aeruginosa challenge. After similar challenge in combined sim-µG + proton-IR conditions, the sex difference was modulated: CH3/HeN female mice had 100% morbidity compared to 60% in males, whereas Balb/c female and male mice both had 100% morbidity. Interestingly, in sim-µG + gamma-IR conditions the observed sex-difference was decreased as Ch3/HeN female mice had 90% morbidity compared to 80% in males [102]. This points to an influence of mouse strain on the synergistic immunological response to bacterial challenge observed in sim-µG + IR conditions. It is yet unclear which strain may more closely resemble human immunological disruptions in space, how to more accurately “humanize” mouse immune models, and how sex-associated effects differ based on the immune biomarker in question [105,106,107]. The specific cause of such sex-associated morbidity differences is also unclear and necessitates further investigations to determine the spaceflight-related health risks for female astronauts, a rapidly growing cohort with potentially unique susceptibility risks [108,109,110,111].

Re-adaptations

A large question also remains of whether these observed immunological dysfunctions are present only during the spaceflight-analog exposure itself, or if they persist chronically even after return to terrestrial (normal weight-bearing and radiation-free) conditions. Cosmonauts experience persistently increased levels of von Willebrand factor, C-reactive protein, D-dimers, monocytes, and granulocytes even after a week of return to Earth [112].

Combined spaceflight-analog studies observe similar deficits: Mice underwent HU for 14 days with whole-body proton-IR (50 cGy, 150 MeV) on day 7, and then four or thirty days of re-adaptation. After four days of re-adaptation, increases in blood NK cell counts were primarily driven by IR, but by thirty days of re-adaptation there was a synergistic influence of the sim-µG + IR interaction decreasing NK cell counts in the blood [91]. These changes in interaction are notable for two reasons: (1) it illustrates that re-adaptation to Earth’s conditions is not a linear process of recovery to baseline, and (2) it suggests countermeasure development may need to focus on different exposure risks at different points during recovery. Similarly, endothelial nitric oxide (NO) synthase (eNOS) immunoreactivity was unchanged in single-exposure cohorts and slightly increased in only the combined sim-µG + IR exposure cohort after four days of re-adaptation; this eNOS reactivity was further synergistically increased after thirty days of recovery [91] (Fig. 5). In another study, rodents were exposed to sim-µG for 18 days and/or 0.5 Gy simplified GCR IR on day 7 to assay changes in immune cell populations such as monocytes, NK cells, and B cells in peripheral blood. Early effects (day 24) were driven by IR exposure, but late effects (day 133–147) were driven by sim-µG exposure [113]. Functionally, impairments in T cell activation both 4 days and 21 days after combined sim-µG (3 days of HU) and 2 Gy proton-IR were also driven independently by sim-µG exposure: no change in CD69 + markers was observed after IR, whereas significant decreases resulted after sim-µG and sim-µG + IR [100]. These results reinforce the dynamic temporal interplay between sim-µG and IR, both during and post-spaceflight; we must accordingly adjust our expectations and rehabilitation plans to optimize astronaut recovery after return from space travel.

Fig. 5
figure 5

Temporality of synergistic sim-µG + IR effects observed on endothelial nitric oxide (NO) synthase (eNOS) immunoreactivity. Mice were hindlimb unloaded for 14 days with whole-body proton-IR (50 cGy, 150 MeV) on day 7. eNOS immunoreactivity in rodents was assessed following (A) 4 days or (B) 30 days of recovery from control or spaceflight-analog conditions of irradiation (IR), simulated microgravity (Sim-µG), or combined Sim-µG + IR.  (C) eNOS immunoreactivity was slightly increased in only the combined sim-µG + IR exposure cohort after four days of re-adaptation. This was further significantly and synergistically increased thirty days after exposure. These results reinforce the dynamic temporal interplay between sim-µG and IR, both during and post-spaceflight [91]. Data shown are mean density of eNOS-positive endothelial cells ± standard error of the mean (SEM) (n = 8/group). Figures re-created and modified from [91] with express permission from the publisher

Single-hazard spaceflight-analog studies further underscore this modulation: 40 days after total-body 56Fe IR exposure, peripheral leukocyte counts were unchanged, CD8 + T cell counts were decreased, and spontaneous blastogenesis increased. Splenic lymphocyte counts on the other hand were significantly decreased, and both spontaneous and mitogen-induced blastogenesis was unchanged [66]. This highlights both persistent immunological dysregulation and differential recovery by immune organs after exposure to spaceflight-associated hazards. The mechanistic bases of these observations are poorly understood, necessitating further investigations into re-adaptation metrics with single-hazard and combined exposure spaceflight-analog studies. Particularly, experimental design of spaceflight-analog studies in rodents should incorporate both sexes, various strains, and various time points during and after spaceflight-analog exposure. This will illuminate the nuanced interplay between microgravity and radiation, point to mechanistic explanations, and provide a basis for countermeasure development against chronic immunological risks.

In vitro cell culture studies

The combined effects of microgravity and radiation have been extensively investigated in other physiological systems using rotating wall vessels [32, 114, 115]. Notably, some cell culture spaceflight-analog studies differ from findings in astronauts. Human peripheral blood mononuclear cells (PBMCs) were exposed to 2 min of 0.8-Gy or 2.0-Gy gamma-IR followed by 24 h of incubation in sim-µG with a RWV or 1g conditions; cytokine release of IL-1B, IL-7, and IL-12 was synergistically increased and detectable only after combined sim-µG + gamma-IR exposure [10] (Fig. 6). This reiterates both the perturbations in cytokine levels noted in astronauts [11] and the synergistic interactions thereof noted in rodent spaceflight-analog studies. However, the PBMC’s also demonstrated a synergistic increase in GM-CSF after combined exposure whereas long-duration astronauts either had no change [8] or a decrease [9] in GM-CSF levels reported. Congruence between spaceflight studies in vivo in rodents and in vitro in cell culture is similarly mixed. Compared to rodents flown on the ISS, spaceflight-analog cell culture studies have demonstrated similar increases in double-stranded DNA breaks, and ROS activity through extracellular signal-regulated kinase (ERK), mitogen-activated protein kinase phosphatase-1 (MKP-1), and caspase-3 activation [116, 117]. However, spaceflight-analog cell culture studies have also observed reduced cytokine secretion [118], mast cell degranulation [118], and both reduced [119] and increased [116, 117] apoptosis. The use of cell culture models as spaceflight-analogs must be approached cautiously, as they are likely unable to replicate systems-level physiological changes observed in humans and in rodents.

Fig. 6
figure 6

Synergistic effects of combined exposure spaceflight-analog cell studies and isoproterenol on cytokine expression. Cytokine concentration in cell culture medium measured using fluorescent secondary antibodies. Cells were exposed to gamma irradiation with 2 Gy or not, and subsequently incubated in 1 g or µg. After 24 h, cytokine concentration of (A) IL-12, (B) IL-7, (C) IL-1b, and (D) GM-CSF was measured. Radiation induced cytokine production in µg but not in 1 g. Isoproterenol treatment prior to radiation prevented abnormal expression of all cytokines. Data shown are mean ± SEM from 10 independent experiments; p-value threshold after correcting for false discovery rate is 0.0049 (significant differences indicated with *). The synergistic effect of isoproterenol and radiation in µg was significant for all four cytokines [10]. Figures re-created and modified with permission from [10] under a CC-BY unrestricted open access license

Gene expression and regulation

Such cell culture studies may nonetheless provide rich opportunity for elucidation of gene regulation and signaling pathways involved in the immune response during spaceflight-analog exposures. Lymphoblastoid TK6 cells were incubated for 24 h after control or 2-Gy gamma-IR exposure in static or sim-µG conditions using an RWV machine [91]. Combined sim-µG + gamma-IR exposure additively altered expression patterns of mRNA, miRNA, and lncRNA in lymphoblastoid TK6 cells, but synergistically altered expression levels of the RNAs and their target genes. Pathway analyses suggest dysregulation of genes involved in immune and inflammatory responses including LPS/TLR, TNFα, and nuclear factor-kappa B cell (NF-κB) signaling pathways [120]. This is not unexpected since it has been additionally reported that sim-µG-alone induces genes involves in the NF-kB pathways and cellular proliferation [121], while IR-alone induces genes involved in DNA damage and repair mechanisms [7, 122]. Although conducted in vitro, this may provide a mechanistic explanation for the effects observed in vivo such as increased LPS levels and altered cytokine levels in mice exposed to sim-µG + IR [13]. However, it is important to keep in mind practical limitations of these conclusions. In attempting to analyze the miRNA data from astronauts aboard the ISS, we have found that quantification analysis of the miRNA expression levels fluctuates significantly even between days, control subjects, and trials, making it difficult to draw significant conclusions from miRNA data (HW unpublished data).

DNA repair and apoptosis

DNA repair processes are required for intact immune function [123, 124] such as creating genetic diversity in developing T and B cells for adaptive immunity [125]. DNA damage such as double strand breaks, chromosomal aberrations, micronucleus formation, and genetic mutations are synergistically affected by combined microgravity and space radiation exposure, as reviewed recently [7]. For example, the repair process of double strand breaks in human peripheral blood lymphocytes was analyzed after IR exposure with a heavy-ion Cs source at 5.155 cGy/s, sim-µG with 24 h of RWV, or control conditions. There were significant decreases in the rate of DNA repair and increased apoptotic levels after sim-µG + IR exposure, highlighting genotoxic effects of combined sim-µG and IR exposure beyond single-hazard levels [116]. A recent study investigating the effects of combined simulated spaceflight exposure on astrocytes similarly indicated sim-µG (24 h of 2D clinostat incubation) had no significant influence on DNA double-strand break repair, whereas different doses of radiation from X-rays, 12C ions, or 56Fe ions induced dose-, LET-, and time-dependent deficits in DNA repair [126]. Gene expression analyses further indicated that the sim-µG exposure after 2-Gy X-ray IR resulted in downregulation of genes involved in DNA damage repair, mitosis, and cell proliferation [126]. This is similar to the altered gene expression relating to cell cycle progression and and senescence observed in rodents flown on the ISS for 15 days [98]. Genomic instability from diminished DNA damage repair may contribute further to the compromised immune status and increased infection susceptibility previously discussed in vivo [78, 82,83,84, 102, 104].

Programmed cell death is a critical response to DNA damage to preserve physiological cellular functioning. For example, lymphoblastoid TK6 cells that were incubated for 24 h after control or 4-Gy gamma-IR exposure in static or sim-µG conditions using an RWV machine were then assessed for genomic damage and apoptosis levels. Combined sim-µG + IR led to a significant increase in genomic mutation frequency but a significant reduction in apoptotic cell counts compared to IR-alone [119]; this points to an aberrantly increased frequency of damaged cells surviving the physiological apoptosis process. Human fibroblasts simultaneously exposed to sim-µG (24 h of RWV) and 1 Gy of Carbon-ion or X-ray radiation had decreased expression of cell cycle-suppression genes ABL1 and CDKN1A and increased expression of cell cycle-promoting genes. The authors therefore suggest that cells may successfully pass through cell cycle checkpoints despite DNA damage due to combined effects from sim-µG and carbon-ion IR [127]. Conversely, human B lymphoblasts exposed to 30 min of sim-µG with RWV and heavy-ion carbon IR exposure (300 MeV/u, 1 Gy/min) had decreased cell survival, increased apoptosis, and increased ROS-sensitive apoptosis signaling [117]. These functional changes in immune surveillance may be mediated through heavy ion radiation-induced intracellular ROS generation as the observed defects were reversed with antioxidant administration [117]. Therefore, although many spaceflight-analog studies indicate decreased apoptosis and therefore immune function, this cannot be definitively or mechanistically concluded. More combined exposure studies are needed in a variety of cell types and in co-culture with neighboring cell types to determine the complex signaling networks that govern the DNA damage and apoptotic responses in spaceflight.

Conclusions and other considerations

This review presents a unique and diverse set of immunological interactions that occur between microgravity and radiation exposure. Combined spaceflight-analog models such as hindlimb unloading and rotating wall vessels demonstrated largely similar immunological perturbations compared to astronauts onboard the ISS. Results from cell culture spaceflight-analog studies must be interpreted cautiously as they do not fully replicate all dysregulations that astronauts have experienced during spaceflight missions. Broadly though, the spaceflight-analog models point to additive effects in myeloid counts and lymphoblastoid RNA expression patterns; synergistic effects in cytokine levels, T and NK cell proliferation, and DNA repair ability; and some antagonistic effects in lymphoid and granulocyte counts. It becomes clear that the interactive nature between microgravity and radiation in spaceflight-analog studies differs with time, sex and age for rodent-analog studies, radiation dosage and type, and cell lines for immune markers. Few studies however effectively demonstrate convincing mechanistic explanations for this. Combined-exposure spaceflight-analog studies are likely to provide more accurate baseline understandings of observed immune dysfunction, allowing for improved risk prediction and mitigation for spaceflight missions. It must also be remembered that such complex immune markers are perturbed by much more than just microgravity and radiation.

Ageing

As we open the door to human space exploration beyond LEO, it will be critical to understand the risks of older astronauts and those undergoing long-term spaceflight in particular. Characterization of the immune response in an elderly astronaut (age 77) revealed increased viral reactivation, adrenocorticotropic hormone levels, cortisol levels, monocyte counts, and NK cell counts compared to younger crewmembers [128]. Dysregulations in several immune parameters such as IL-4, IL-10, IL-12, and IFNγ have shown to differ between astronauts in short-term and long-term spaceflight missions [8, 9, 11]. Closer examination of longitudinal immune changes is critical to preparing for NASA’s upcoming Artemis missions to the Moon and Mars.

Advanced age is also closely associated with pathophysiological immune aberrations such as persistent immune system activation, increased inflammatory states, immunosenescence, increased severity of infections, and more [129,130,131,132,133,134,135]. This can lead to increased morbidity rates with increasing age [136] and can have drastic consequences in spaceflight missions far removed from comprehensive healthcare access. Many spaceflight-associated changes observed in astronauts including genomic instability, dysfunctional protein homeostasis, mitochondrial dysfunction, stem cell depletion, impaired intercellular communication, and irregular gut microbiomes are similar to those observed in physiological ageing, as recently reviewed [137]. Notable age-associated immune perturbations that mimic those observed in astronauts include chronic inflammation [137], cytokine perturbations [132, 138], viral reactivation [139], cortisol increases [140], and NK cell function decreases [141] (Table 2). Indeed, Capri and colleagues recently described spaceflight missions as “accelerating aging” by contributing to such inflammatory activity [87]. However, if sufficient re-adaptation measures can be employed, this is not necessarily the case for astronauts whose immune function may normalize to baseline upon return to Earth.

Table 2 Comparing the immunological responses to ageing and spaceflight travel

Many of the spaceflight-analog studies described herein utilized younger rodents (5–12 weeks old) and assayed only short-term effects of spaceflight-exposure. Nonetheless, a mere three weeks of hindlimb unloading in young mice induced decreases in IgM repertoire and B cell lymphopoiesis similar to older mice who were not exposed to spaceflight-analog conditions [143, 144]. Rodent models of Alzheimer’s disease (AD) have also begun to elucidate the neural effects of age-associated comorbidities in spaceflight-analog studies. GCR exposures have been shown to differentially alter neuropathology and behavior in sex- and mutation-associated manners [145, 146]. Proton irradiation increases deposition of amyloid-beta plaques but did not change cytokine levels of IL-1, IL-6, CCL2, CXCL10, and TNFα [147]. A head-down bedrest (HDBR) study in older adults was recently conducted by the Canadian Space Agency to compare the effects of ageing and spaceflight-associated fluid shifts comprehensively and longitudinally, though results are still forthcoming [148]. Further comparative and longitudinal studies may prove informative in unravelling the relative contributions of microgravity and radiation to age-related immune effects.

Stress response

Various other endogenous factors, such as stress hormones or inflammatory signaling, can also further compromise immune function. For example, the Nrf2 transcription factor regulates expression of cytoprotective and antioxidative stress response genes. In Nrf-2 knockout mice that flew onboard the ISS for 31 days, spaceflight-induced immunosuppression and tissue inflammatory markers were markedly increased compared to wild-type mice [149]. This is a non-trivial limitation as the HU model has been shown to induce at least transient stress responses in rodents such as increased serum corticosterone or atrophy of lymphoid organs [19, 150,151,152]. Sim-µG, IR, and particularly combined sim-µG + IR increased serum total corticosterone levels in C3H/HeN female mice [102], indicating an additive activation of the adrenergic stress response. Conversely, female C57BL/6J flown for 13 days on the STS-135 mission demonstrated increased adrenal and hepatic corticosterone levels, but no changes in catecholamine levels [96]. This discrepancy and its impact on functional changes in immune activity should be investigated further, particularly as increased catecholamine, cortisol, and intracellular cAMP levels have been linked to modulations in several immunological processes such as lymphocyte activation or apoptotic cell death [153,154,155,156,157,158]. Stress hormones may also influence the immune system’s ability to produce antibodies [153, 159] and modulate immune cell function [160], for example by activating the NF-kB pathway which was already observed to be disrupted in sim-µG + IR cell culture models [120]. The effects of combined sim-µG and IR on these stress-induced pathways are not fully understood, though it may play a significant role in the cellular response to DNA damage repair [7]. Additional environmental exposures in the space environment can further complicate the stress-related immune effects observed, such as sleep deprivation [161] or long-term social isolation [162, 163].

Countermeasure development

Over the years of the International Space Station’s operations, major physiological improvements in immunity, stress, and viral reactivation have been made from updated cargo and resupply frequencies, exercise protocols, nutritional quality and supplementation, and schedule management, thoroughly reviewed elsewhere [164, 165]. By continuing to characterize this plethora of both acute and chronic immunological dysregulations indicated from both µG and IR and their synergistic interactions, it will become more feasible to develop in-flight countermeasures and post-flight readaptation measures. Astronauts have a heightened risk of exposure and immune damage from infectious diseases during space missions [71, 72, 102, 166,167,168]. Observations made by the spaceflight-analog studies discussed underscore the cellular, genetic, and functional immune dysregulations that contribute to these risks.

Beyond these administrative-based changes, pharmacological countermeasures may present a possible intervention to these immune risks. Isoproterenol, a sympathomimetic drug, decreased IR-induced apoptosis in PBMCs exposed to sim-µG. Similar to other findings discussed herein, neither sim-µG nor IR-alone significantly increased cytokine production, but in sim-µG + IR conditions, a highly significant increase was observed in IL-7, GM-CSF, IL-12, and IL-1b concentrations. Immediate prior administration of isoproterenol prevented cytokine increase and kept it within-control levels. The synergistic effect of isoproterenol + sim-µG + IR was significant for all four cytokines [10] (Fig. 6). These results suggest that prophylactic isoproterenol is synergistically able to prevent some sim-µG and IR-mediated immune changes. Similarly, antioxidant administration of N-acetyl cysteine and quercetin were able to reverse sim- µG + IR-induced inhibition of apoptosis in human B lymphoblasts, as discussed earlier [117]. These findings open the door to novel insight on regulatory pathways and possible pharmacological countermeasures to mitigate spaceflight-mediated damages.

However, it is important to keep in mind that weakened immune systems and microgravity or radiation-induced drug instabilities may inhibit the pharmacokinetics and efficacy of antimicrobial agents utilized; studies directly investigating these pharmacokinetics and pharmacodynamics in space are extremely limited though [169,170,171]. Established alterations in gut microbiome that occur as a result of spaceflight travel can influence bioavailability of drug absorption [172]. HDBR studies have also implicated slightly lower (but not statistically significant) ciprofloxacin absorption [173] and delayed absorption and clearance of pivmecillinam and benzylpenicillin [174, 175], though the clinical significance is unclear. Research on drug stability has focused on radioprotective packaging, optimal storage conditions, careful consideration of pharmacological formulation in liquid or solid forms, and multi-pronged pharmaceutical regimes such as non-antibiotics combined with tetracyclines to take advantage of their synergistic activities [169]. There is limited data on pharmaceuticals that can safely target both the microgravity and radiation-induced immune dysregulations since, as we have discussed, they can have independent mechanisms or pathways. Further investigations may prove useful in elucidating possible targets for pharmacological intervention, especially as inter-individual variability will notably influence the efficacy of developed countermeasures and the advent of personalized medicine in space [176].

Guidelines for standardized study design

Most studies examining stressors to astronaut physiology and performance examine individual risks despite growing evidence that many spaceflight hazards affect astronaut health in combination with one another. Beyond microgravity and radiation, this can include ageing, stress responses, social isolation, poor sleep quality or quantity, and disrupted circadian rhythms [177,178,179]. Synergistic interactions between microgravity and radiation in particular often differed depending on the time point, IR dose, IR type, sex, and strain used in the study. With such diversity of experimental design for spaceflight-analog studies, we obtain valuable metrics for many rodent models, time points, cell and tissue types, and more but meta-analytical comparisons become prohibitively difficult. There are clear diverse modulatory interactions within the immune systems that cannot be accurately assayed through studies focusing on single risks alone.

Some experimental designs also did not accurately reflect the simultaneous combined µG + IR exposure experienced in space: vast variability was present in whether spaceflight-analog systems were exposed to IR before, during, and/or after sim-µG. Immune dysregulations are dynamic; acute and/or sequential exposures may have contributed to the greater concordance of spaceflight-analog studies with observations from short-duration spaceflight missions. In the setting of preparing for NASA’s Artemis missions and long-duration human space exploration, longitudinal studies with chronic exposures will prove greatly informative.

Finally, irradiation doses in many of these studies ranged from 0.5 Gy to 2.0 Gy. Comparatively, astronauts on the ISS receive approximately, 1 mSv/day though this will likely double or triple with travel past LEO [56, 180]. The impacts of acute whole-body SPE exposure are difficult to calculate due to heterogeneities in radiation delivery to internal organs and extrapolating rodent model data across species [181]. Current estimates predict doses of <0.5 Gy-Eq to internal organs and dose delivery rates peaking at approximately 0.12 Gy-Eq/hr to hematopoietic organs such as bone marrow [182, 183]. The radiobiological effectiveness of the IR dose will differ based on IR source, whether gamma rays, X-rays, or charged particles such as protons or heavy ions [184,185,186,187,188]. Although proton-IR may effectively mimic SPE’s, gamma-IR is only one component of GCR and does not provide a truly comprehensive analog for GCR [189]. It is also difficult to predict how simultaneous SPE and GCR exposures may affect the pathophysiological changes to human health and performance [56, 190].

It is critical that future study designs more rigorously mimic the spaceflight environment. The spaceflight-analog study, whether rodents or cells, should incorporate both sim-µG conditions and chronic IR simultaneously, rather than sequentially as performed in many of the reported studies. PWB models may prove to be an attractive way to assess the effects of partial gravity on the immune system, as relevant for future missions on Mars. Radiation exposures should incorporate lower dose ranges below 0.5 Gy and from heavy-ion sources to simulate prolonged GCR exposure [189, 191]. Finally, a greater emphasis should be placed on longitudinal studies evaluating specific immunological alterations in diverse combined environments such as age, sex differences, circadian disturbances, nutrition, and more. Investigating countermeasures and re-adaptation metrics will be particularly informative in safeguarding astronaut health and performance.

As we move into a new age of deep space human exploration, exposure to combined environmental stressors will present critical health and safety risks for astronauts. There will remain a pressing need to further investigate the complex immunological health risks arising from microgravity, radiation, and more.

Data availability

Primary data sharing is not applicable to this manuscript as no datasets were generated during the current review. All datasets analyzed to recreate figures were retrieved from the National Institutes of Health PubMed index and have been cited in this manuscript for further reference.

Abbreviations

BNL:

Brookhaven National Laboratory

CCL:

C-C motif chemokine ligand

CXCL:

C-X-C motif chemokine ligand

CI:

Confidence interval

ERK:

Extracellular signal-regulated kinase

FGF:

Fibroblast growth factor

G-CSF:

Granulocyte colony stimulating factor

GM-CSF:

Granulocyte-macrophage colony stimulating factor

HDBR:

Head-down bedrest

IFN:

Interferon

IL:

Interleukin

IR:

Radiation

ISS:

International Space Station

GCR:

Galactic cosmic radiation

HU:

Hindlimb unloading

LEO:

Low Earth orbit

LET:

Linear energy transfer

LPS:

Lipopolysaccharide

MKP-1:

Mitogen-activated protein kinase phosphatase-1

NASA:

National Aeronautics and Space Administration

NF-kB:

Nuclear factor kappa light chain enhancer of activated B cells

NK:

Natural killer cells

NO:

Nitric oxide

NSRL:

NASA Space Radiation Laboratory

PBMC:

Peripheral blood mononuclear cell

PWB:

Partial weight bearing

ROS:

Reaction oxygen species

RWV:

Rotating wall vessel

SD:

Standard deviation

SEM:

Standard error of the mean

SPE:

Solar particle events

TLR:

Toll-like receptor

TNFα:

Tumor necrosis factor-α

µG:

Microgravity

VEGF:

Vascular endothelial growth factor

References

  1. Willey JS, Britten RA, Blaber E, Tahimic CGT, Chancellor J, Mortreux M, et al. The individual and combined effects of spaceflight radiation and microgravity on biologic systems and functional outcomes. J Environ Sci Health C Toxicol Carcinog. 2021;39(2):129–79.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Bezdan D, Grigorev K, Meydan C, Pelissier Vatter FA, Cioffi M, Rao V, et al. Cell-free DNA (cfDNA) and exosome profiling from a year-long human spaceflight reveals circulating biomarkers. iScience. 2020;23(12):101844.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Garrett-Bakelman FE, Darshi M, Green SJ, Gur RC, Lin L, Macias BR et al. The NASA Twins Study: a multidimensional analysis of a year-long human spaceflight. Science. 2019;364(6436).

  4. Gertz ML, Chin CR, Tomoiaga D, MacKay M, Chang C, Butler D, et al. Multi-omic, Single-Cell, and biochemical profiles of astronauts Guide Pharmacological Strategies for returning to gravity. Cell Rep. 2020;33(10):108429.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. da Silveira WA, Fazelinia H, Rosenthal SB, Laiakis EC, Kim MS, Meydan C, et al. Comprehensive multi-omics Analysis reveals mitochondrial stress as a Central Biological hub for spaceflight impact. Cell. 2020;183(5):1185–e20120.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Gupta U, Baig S, Majid A, Bell SM. The neurology of space flight; how does space flight effect the human nervous system? Life Sci Space Res (Amst). 2023;36:105–15.

    Article  PubMed  Google Scholar 

  7. Moreno-Villanueva M, Wong M, Lu T, Zhang Y, Wu H. Interplay of space radiation and microgravity in DNA damage and DNA damage response. NPJ Microgravity. 2017;3:14.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Crucian BE, Zwart SR, Mehta S, Uchakin P, Quiriarte HD, Pierson D, et al. Plasma cytokine concentrations indicate that in vivo hormonal regulation of immunity is altered during long-duration spaceflight. J Interferon Cytokine Res. 2014;34(10):778–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Krieger SS, Zwart SR, Mehta S, Wu H, Simpson RJ, Smith SM, et al. Alterations in saliva and plasma cytokine concentrations during long-duration spaceflight. Front Immunol. 2021;12:725748.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Moreno-Villanueva M, Feiveson AH, Krieger S, Kay Brinda A, von Scheven G, Bürkle A et al. Synergistic effects of Weightlessness, Isoproterenol, and Radiation on DNA damage response and cytokine production in Immune cells. Int J Mol Sci. 2018;19(11).

  11. Crucian B, Stowe R, Mehta S, Uchakin P, Quiriarte H, Pierson D, et al. Immune system dysregulation occurs during short duration spaceflight on board the space shuttle. J Clin Immunol. 2013;33(2):456–65.

    Article  CAS  PubMed  Google Scholar 

  12. Paul AM, Cheng-Campbell M, Blaber EA, Anand S, Bhattacharya S, Zwart SR, et al. Beyond low-earth orbit: characterizing Immune and microRNA differentials following simulated deep spaceflight conditions in mice. iScience. 2020;23(12):101747.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhou Y, Ni H, Li M, Sanzari JK, Diffenderfer ES, Lin L, et al. Effect of solar particle event radiation and hindlimb suspension on gastrointestinal tract bacterial translocation and immune activation. PLoS ONE. 2012;7(9):e44329.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Williams D, Kuipers A, Mukai C, Thirsk R. Acclimation during space flight: effects on human physiology. CMAJ. 2009;180(13):1317–23.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Mulavara AP, Peters BT, Miller CA, Kofman IS, Reschke MF, Taylor LC, et al. Physiological and functional alterations after spaceflight and Bed Rest. Med Sci Sports Exerc. 2018;50(9):1961–80.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Buckey JC, Thamer S, Lan M. Bone loss and kidney stone risk in weightlessness. Curr Opin Nephrol Hypertens. 2023;32(2):172–6.

    Article  PubMed  Google Scholar 

  17. Morey-Holton E, Globus RK, Kaplansky A, Durnova G. The hindlimb unloading rat model: literature overview, technique update and comparison with space flight data. Adv Space Biol Med. 2005;10:7–40.

    Article  PubMed  Google Scholar 

  18. Sonnenfeld G. Animal models for the study of the effects of spaceflight on the immune system. Adv Space Res. 2003;32(8):1473–6.

    Article  CAS  PubMed  Google Scholar 

  19. Morey-Holton ER, Globus RK. Hindlimb unloading rodent model: technical aspects. J Appl Physiol (1985). 2002;92(4):1367–77.

    Article  PubMed  Google Scholar 

  20. Farley A, Gnyubkin V, Vanden-Bossche A, Laroche N, Neefs M, Baatout S, et al. Unloading-Induced cortical bone loss is exacerbated by low-dose irradiation during a simulated Deep Space Exploration Mission. Calcif Tissue Int. 2020;107(2):170–9.

    Article  CAS  PubMed  Google Scholar 

  21. Mortreux M, Nagy JA, Ko FC, Bouxsein ML, Rutkove SB. A novel partial gravity ground-based analog for rats via quadrupedal unloading. J Appl Physiol (1985). 2018;125(1):175–82.

    Article  CAS  PubMed  Google Scholar 

  22. Mortreux M, Rosa-Caldwell ME. Approaching gravity as a Continuum using the rat partial weight-bearing model. Life (Basel). 2020;10(10).

  23. Wagner EB, Granzella NP, Saito H, Newman DJ, Young LR, Bouxsein ML. Partial weight suspension: a novel murine model for investigating adaptation to reduced musculoskeletal loading. J Appl Physiol (1985). 2010;109(2):350–7.

    Article  PubMed  Google Scholar 

  24. Rosa-Caldwell ME, Mortreux M, Wadhwa A, Kaiser UB, Sung DM, Bouxsein ML, et al. Influence of gonadectomy on muscle health in micro- and partial-gravity environments in rats. J Appl Physiol (1985). 2023;134(6):1438–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ko FC, Mortreux M, Riveros D, Nagy JA, Rutkove SB, Bouxsein ML. Dose-dependent skeletal deficits due to varied reductions in mechanical loading in rats. NPJ Microgravity. 2020;6:15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Swift JM, Lima F, Macias BR, Allen MR, Greene ES, Shirazi-Fard Y, et al. Partial weight bearing does not prevent musculoskeletal losses associated with disuse. Med Sci Sports Exerc. 2013;45(11):2052–60.

    Article  PubMed  Google Scholar 

  27. Mortreux M, Riveros D, Semple C, Bouxsein ML, Rutkove SB. The partial weight-bearing rat model using a pelvic harness does not impact stress or hindlimb blood flow. Acta Astronaut. 2020;168:249–55.

    Article  CAS  Google Scholar 

  28. Klaus DM. Clinostats and bioreactors. Gravit Space Biol Bull. 2001;14(2):55–64.

    CAS  PubMed  Google Scholar 

  29. Borst AG, van Loon JJWA. Technology and Developments for the Random Positioning Machine. RPM Microgravity Sci Technol. 2008;21(4):287.

    Article  Google Scholar 

  30. Grimm D, Egli M, Krüger M, Riwaldt S, Corydon TJ, Kopp S, et al. Tissue Engineering under Microgravity conditions-Use of Stem cells and Specialized cells. Stem Cells Dev. 2018;27(12):787–804.

    Article  PubMed  Google Scholar 

  31. de Korte M, Keating A, Wang C. Culturing lymphocytes in simulated microgravity using a Rotary Cell Culture System. J Vis Exp. 2022(186).

  32. Wuest SL, Richard S, Kopp S, Grimm D, Egli M. Simulated microgravity: critical review on the use of random positioning machines for mammalian cell culture. Biomed Res Int. 2015;2015:971474.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Anil-Inevi M, Yaman S, Yildiz AA, Mese G, Yalcin-Ozuysal O, Tekin HC, et al. Biofabrication of in situ self assembled 3D cell cultures in a weightlessness environment generated using magnetic levitation. Sci Rep. 2018;8(1):7239.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Haisler WL, Timm DM, Gage JA, Tseng H, Killian TC, Souza GR. Three-dimensional cell culturing by magnetic levitation. Nat Protoc. 2013;8(10):1940–9.

    Article  CAS  PubMed  Google Scholar 

  35. Hammer BE, Kidder LS, Williams PC, Xu WW. Magnetic levitation of MC3T3 osteoblast cells as a ground-based Simulation of Microgravity. Microgravity Sci Technol. 2009;21(4):311–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Berry MVGA. Of flying frogs and levitrons. Eur J Phys. 1997;18(4).

  37. Marycz K, Kornicka K, Röcken M. Static magnetic field (SMF) as a Regulator of Stem Cell Fate - New perspectives in Regenerative Medicine arising from an underestimated Tool. Stem Cell Rev Rep. 2018;14(6):785–92.

    Article  CAS  PubMed  Google Scholar 

  38. Rosen AD. Mechanism of action of moderate-intensity static magnetic fields on biological systems. Cell Biochem Biophys. 2003;39(2):163–73.

    Article  CAS  PubMed  Google Scholar 

  39. Novikov VV, Yablokova EV, Fesenko EE. A decrease of the respiratory burst in neutrophils after exposure to weak combined magnetic fields of a certain duration. Biophysics. 2020;65(1):82–7.

    Article  CAS  Google Scholar 

  40. Thiel CS, Vahlensieck C, Bradley T, Tauber S, Lehmann M, Ullrich O. Metabolic Dynamics in short- and long-term microgravity in human primary macrophages. Int J Mol Sci. 2021;22(13).

  41. Verhaar AP, Hoekstra E, Tjon AS, Utomo WK, Deuring JJ, Bakker ER, et al. Dichotomal effect of space flight-associated microgravity on stress-activated protein kinases in innate immunity. Sci Rep. 2014;4:5468.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lang A, Binneboessel S, Nienhaus F, Bruno RR, Wolff G, Piayda K, et al. Acute and short-term fluctuations in gravity are associated with changes in circulatory plasma protein levels. NPJ Microgravity. 2024;10(1):25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wernlé K, Thiel CS, Ullrich O. Increased H3K9me3 and F-Actin reorganization in the Rapid Adaptive response to Hypergravity in Human T lymphocytes. Int J Mol Sci. 2023;24(24).

  44. Vahlensieck C, Thiel CS, Pöschl D, Bradley T, Krammer S, Lauber B, et al. Post-transcriptional Dynamics is involved in Rapid Adaptation to Hypergravity in Jurkat T cells. Front Cell Dev Biol. 2022;10:933984.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Tateishi R, Akiyama N, Miyauchi M, Yoshinaga R, Sasanuma H, Kudo T, et al. Hypergravity provokes a Temporary reduction in CD4 + CD8 + thymocyte number and a persistent decrease in Medullary thymic epithelial cell frequency in mice. PLoS ONE. 2015;10(10):e0141650.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Moser D, Biere K, Liemersdorf C, Tuschen M, Hemmersbach R, Choukér A. Differential effects of hypergravity on immune dysfunctions induced by simulated microgravity. Faseb j. 2023;37(5):e22910.

    Article  CAS  PubMed  Google Scholar 

  47. Alauzet C, Cunat L, Wack M, Lozniewski A, Busby H, Agrinier N, et al. Hypergravity disrupts murine intestinal microbiota. Sci Rep. 2019;9(1):9410.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Ghislin S, Ouzren-Zarhloul N, Kaminski S, Frippiat JP. Hypergravity exposure during gestation modifies the TCRβ repertoire of newborn mice. Sci Rep. 2015;5:9318.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Calcagno G, Ouzren N, Kaminski S, Ghislin S, Frippiat JP. Chronic hypergravity induces a modification of histone H3 lysine 27 trimethylation at TCRβ locus in murine thymocytes. Int J Mol Sci. 2022;23(13).

  50. Bonetto V, Magnelli V, Sabbatini M, Caprì F, van Loon J, Tavella S, et al. The importance of gravity vector on adult mammalian organisms: effects of hypergravity on mouse testis. PLoS ONE. 2023;18(9):e0282625.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Simonsen LC, Slaba TC, Guida P, Rusek A. NASA’s first ground-based Galactic cosmic Ray Simulator: enabling a new era in space radiobiology research. PLoS Biol. 2020;18(5):e3000669.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Durante M, Cucinotta FA. Heavy ion carcinogenesis and human space exploration. Nat Rev Cancer. 2008;8(6):465–72.

    Article  CAS  PubMed  Google Scholar 

  53. Finkelstein SR, Patel R, Deland K, Mercer J, Starr B, Zhu D et al. (56) Fe ion exposure increases the incidence of lung and brain tumors at a similar rate in male and female mice. bioRxiv. 2023.

  54. Mewaldt RA. Elemental composition and energy spectra of galactic cosmic rays. United States; 1988. Contract No.: N–89-28454.

  55. Cucinotta FA, Kim M-HY, Ren L. Evaluating shielding effectiveness for reducing space radiation cancer risks. Radiat Meas. 2006;41(9):1173–85.

    Article  CAS  Google Scholar 

  56. Chancellor JC, Blue RS, Cengel KA, Auñón-Chancellor SM, Rubins KH, Katzgraber HG, et al. Limitations in predicting the space radiation health risk for exploration astronauts. NPJ Microgravity. 2018;4:8.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Cucinotta FA, Durante M. Cancer risk from exposure to galactic cosmic rays: implications for space exploration by human beings. Lancet Oncol. 2006;7(5):431–5.

    Article  CAS  PubMed  Google Scholar 

  58. Townsend LW, Fry RJM. Radiation protection guidance for activities in low-earth orbit. Adv Space Res. 2002;30(4):957–63.

    Article  CAS  PubMed  Google Scholar 

  59. National Research Council. Managing Space Radiation Risk in the New era of Space Exploration. Washington, DC: National Academies; 2008. p. 132.

    Google Scholar 

  60. Salminen A, Kaarniranta K, Kauppinen A, Photoaging. UV radiation-induced inflammation and immunosuppression accelerate the aging process in the skin. Inflamm Res. 2022;71(7–8):817–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Little MP, Azizova TV, Richardson DB, Tapio S, Bernier MO, Kreuzer M, et al. Ionising radiation and cardiovascular disease: systematic review and meta-analysis. BMJ. 2023;380:e072924.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Neale RE, Lucas RM, Byrne SN, Hollestein L, Rhodes LE, Yazar S, et al. The effects of exposure to solar radiation on human health. Photochem Photobiol Sci. 2023;22(5):1011–47.

    Article  CAS  PubMed  Google Scholar 

  63. NASA Space Radiation Laboratory. Solar Particle Event Simulation: Brookhaven National Laboratory. 2023 [ https://www.bnl.gov/nsrl/userguide/spe-simulation.php.

  64. NASA Space Radiation Laboratory. Galactic Cosmic Ray Simulation (GCRsim): Brookhaven National Laboratory. 2023 [ https://www.bnl.gov/nsrl/userguide/gcrsim.php.

  65. NASA Space Radiation Laboratory. Simplified Galactic Cosmic Ray Simulation (SimGCRsim): Brookhaven National Laboratory. 2023 [ https://www.bnl.gov/nsrl/userguide/simgcrsim.php.

  66. Gridley DS, Pecaut MJ. Changes in the distribution and function of leukocytes after whole-body iron ion irradiation. J Radiat Res. 2016;57(5):477–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Committee on Ethics P, Guidelines for Health Standards for, Long D, Exploration S, Board on Health Sciences P. Institute of M. Health Standards for Long Duration and Exploration Spaceflight: Ethics principles, responsibilities, and decision Framework. In: Kahn J, Liverman CT, McCoy MA, editors. National Academies Press (US). Copyright 2014 by the National Academy of Sciences. Washington (DC): All rights reserved.; 2014.

    Google Scholar 

  68. Guéguinou N, Huin-Schohn C, Bascove M, Bueb JL, Tschirhart E, Legrand-Frossi C, et al. Could spaceflight-associated immune system weakening preclude the expansion of human presence beyond Earth’s orbit? J Leukoc Biol. 2009;86(5):1027–38.

    Article  PubMed  Google Scholar 

  69. Crucian B, Stowe RP, Mehta S, Quiriarte H, Pierson D, Sams C. Alterations in adaptive immunity persist during long-duration spaceflight. NPJ Microgravity. 2015;1:15013.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Mehta SK, Laudenslager ML, Stowe RP, Crucian BE, Feiveson AH, Sams CF, et al. Latent virus reactivation in astronauts on the international space station. NPJ Microgravity. 2017;3:11.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Mann V, Sundaresan A, Mehta SK, Crucian B, Doursout MF, Devakottai S. Effects of microgravity and other space stressors in immunosuppression and viral reactivation with potential nervous system involvement. Neurol India. 2019;67(Supplement):S198–203.

    PubMed  Google Scholar 

  72. Akiyama T, Horie K, Hinoi E, Hiraiwa M, Kato A, Maekawa Y, et al. How does spaceflight affect the acquired immune system? NPJ Microgravity. 2020;6:14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Taylor PW. Impact of space flight on bacterial virulence and antibiotic susceptibility. Infect Drug Resist. 2015;8:249–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Nickerson CA, Ott CM, Mister SJ, Morrow BJ, Burns-Keliher L, Pierson DL. Microgravity as a novel environmental signal affecting Salmonella enterica serovar typhimurium virulence. Infect Immun. 2000;68(6):3147–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Mehta SK, Cohrs RJ, Forghani B, Zerbe G, Gilden DH, Pierson DL. Stress-induced subclinical reactivation of varicella zoster virus in astronauts. J Med Virol. 2004;72(1):174–9.

    Article  PubMed  Google Scholar 

  76. Pierson DL, Stowe RP, Phillips TM, Lugg DJ, Mehta SK. Epstein-Barr virus shedding by astronauts during space flight. Brain Behav Immun. 2005;19(3):235–42.

    Article  CAS  PubMed  Google Scholar 

  77. Konstantinova IV. Immune resistance of man in space flights. Acta Astronaut. 1991;23:123–7.

    Article  CAS  PubMed  Google Scholar 

  78. Levine DS, Greenleaf JE. Immunosuppression during spaceflight deconditioning. Aviat Space Environ Med. 1998;69(2):172–7.

    CAS  PubMed  Google Scholar 

  79. Stowe RP, Sams CF, Mehta SK, Kaur I, Jones ML, Feeback DL, et al. Leukocyte subsets and neutrophil function after short-term spaceflight. J Leukoc Biol. 1999;65(2):179–86.

    Article  CAS  PubMed  Google Scholar 

  80. Kaur I, Simons ER, Castro VA, Ott CM, Pierson DL. Changes in monocyte functions of astronauts. Brain Behav Immun. 2005;19(6):547–54.

    Article  CAS  PubMed  Google Scholar 

  81. Crucian BE, Cubbage ML, Sams CF. Altered cytokine production by specific human peripheral blood cell subsets immediately following space flight. J Interferon Cytokine Res. 2000;20(6):547–56.

    Article  CAS  PubMed  Google Scholar 

  82. Mehta SK, Pierson DL, Cooley H, Dubow R, Lugg D. Epstein-Barr virus reactivation associated with diminished cell-mediated immunity in antarctic expeditioners. J Med Virol. 2000;61(2):235–40.

    Article  CAS  PubMed  Google Scholar 

  83. Shearer WT, Lee BN, Cron SG, Rosenblatt HM, Smith EO, Lugg DJ, et al. Suppression of human anti-inflammatory plasma cytokines IL-10 and IL-1RA with elevation of proinflammatory cytokine IFN-gamma during the isolation of the Antarctic winter. J Allergy Clin Immunol. 2002;109(5):854–7.

    Article  CAS  PubMed  Google Scholar 

  84. Shearer WT, Zhang S, Reuben JM, Lee BN, Butel JS. Effects of radiation and latent virus on immune responses in a space flight model. J Allergy Clin Immunol. 2005;115(6):1297–303.

    Article  CAS  PubMed  Google Scholar 

  85. Furukawa S, Nagamatsu A, Nenoi M, Fujimori A, Kakinuma S, Katsube T, et al. Space Radiation Biology for living in space. Biomed Res Int. 2020;2020:4703286.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Shen M, Frishman WH. Effects of Spaceflight on Cardiovascular Physiology and Health. Cardiol Rev. 2019;27(3):122–6.

    Article  PubMed  Google Scholar 

  87. Capri M, Conte M, Ciurca E, Pirazzini C, Garagnani P, Santoro A, et al. Long-term human spaceflight and inflammaging: does it promote aging? Ageing Res Rev. 2023;87:101909.

    Article  CAS  PubMed  Google Scholar 

  88. Overbey EG, Paul AM, da Silveira WA, Tahimic CGT, Reinsch SS, Szewczyk N et al. Mice exposed to combined chronic low-dose irradiation and modeled microgravity develop long-term neurological sequelae. Int J Mol Sci. 2019;20(17).

  89. Sonnenfeld G. The immune system in space and microgravity. Med Sci Sports Exerc. 2002;34(12):2021–7.

    Article  CAS  PubMed  Google Scholar 

  90. Taylor GR. Overview of spaceflight immunology studies. J Leukoc Biol. 1993;54(3):179–88.

    Article  CAS  PubMed  Google Scholar 

  91. Mao XW, Boerma M, Rodriguez D, Campbell-Beachler M, Jones T, Stanbouly S, et al. Combined effects of low-dose Proton Radiation and simulated microgravity on the mouse retina and the hematopoietic system. Radiat Res. 2019;192(3):241–50.

    Article  CAS  PubMed  Google Scholar 

  92. Mao XW, Nishiyama NC, Pecaut MJ, Campbell-Beachler M, Gifford P, Haynes KE, et al. Simulated microgravity and Low-Dose/Low-Dose-rate Radiation induces oxidative damage in the mouse brain. Radiat Res. 2016;185(6):647–57.

    Article  CAS  PubMed  Google Scholar 

  93. Yumoto K, Globus RK, Mojarrab R, Arakaki J, Wang A, Searby ND, et al. Short-term effects of whole-body exposure to (56)fe ions in combination with musculoskeletal disuse on bone cells. Radiat Res. 2010;173(4):494–504.

    Article  CAS  PubMed  Google Scholar 

  94. Shanmugarajan S, Zhang Y, Moreno-Villanueva M, Clanton R, Rohde LH, Ramesh GT et al. Combined effects of simulated microgravity and Radiation exposure on Osteoclast Cell Fusion. Int J Mol Sci. 2017;18(11).

  95. Delp MD, Charvat JM, Limoli CL, Globus RK, Ghosh P. Apollo Lunar astronauts Show Higher Cardiovascular Disease Mortality: possible Deep Space Radiation effects on the vascular endothelium. Sci Rep. 2016;6:29901.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Pecaut MJ, Mao XW, Bellinger DL, Jonscher KR, Stodieck LS, Ferguson VL, et al. Is spaceflight-induced immune dysfunction linked to systemic changes in metabolism? PLoS ONE. 2017;12(5):e0174174.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Gridley DS, Mao XW, Stodieck LS, Ferguson VL, Bateman TA, Moldovan M, et al. Changes in mouse thymus and spleen after return from the STS-135 mission in space. PLoS ONE. 2013;8(9):e75097.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Kumar A, Tahimic CGT, Almeida EAC, Globus RK. Spaceflight modulates the expression of key oxidative stress and cell cycle related genes in heart. Int J Mol Sci. 2021;22(16).

  99. Hwang SA, Crucian B, Sams C, Actor JK, Post-Spaceflight. (STS-135) mouse splenocytes demonstrate altered activation properties and Surface Molecule expression. PLoS ONE. 2015;10(5):e0124380.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Sanzari JK, Romero-Weaver AL, James G, Krigsfeld G, Lin L, Diffenderfer ES, et al. Leukocyte activity is altered in a ground based murine model of microgravity and proton radiation exposure. PLoS ONE. 2013;8(8):e71757.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Romero-Weaver AL, Lin L, Carabe-Fernandez A, Kennedy AR. Effects of Solar Particle Event-Like Proton Radiation and/or simulated microgravity on circulating mouse blood cells. Gravit Space Res. 2014;2(1):42–53.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Li M, Holmes V, Zhou Y, Ni H, Sanzari JK, Kennedy AR, et al. Hindlimb suspension and SPE-like radiation impairs clearance of bacterial infections. PLoS ONE. 2014;9(1):e85665.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Sadhukhan R, Majumdar D, Garg S, Landes RD, McHargue V, Pawar SA, et al. Simultaneous exposure to chronic irradiation and simulated microgravity differentially alters immune cell phenotype in mouse thymus and spleen. Life Sci Space Res (Amst). 2021;28:66–73.

    Article  PubMed  Google Scholar 

  104. Taylor GR, Konstantinova I, Sonnenfeld G, Jennings R. Changes in the immune system during and after spaceflight. Adv Space Biol Med. 1997;6:1–32.

    Article  CAS  PubMed  Google Scholar 

  105. Tao L, Reese TA. Making mouse models that Reflect Human Immune responses. Trends Immunol. 2017;38(3):181–93.

    Article  CAS  PubMed  Google Scholar 

  106. Masopust D, Sivula CP, Jameson SC. Of mice, dirty mice, and men: using mice to understand human immunology. J Immunol. 2017;199(2):383–8.

    Article  CAS  PubMed  Google Scholar 

  107. Zhang B, Duan Z, Zhao Y. Mouse models with human immunity and their application in biomedical research. J Cell Mol Med. 2009;13(6):1043–58.

    Article  CAS  PubMed  Google Scholar 

  108. Kumar K, Moon BH, Datta K, Fornace AJ Jr., Suman S. Simulated galactic cosmic radiation (GCR)-induced expression of Spp1 coincide with mammary ductal cell proliferation and preneoplastic changes in apc(Min/+) mouse. Life Sci Space Res (Amst). 2023;36:116–22.

    Article  PubMed  Google Scholar 

  109. DeNapoli RC, Buettmann EG, Friedman MA, Lichtman AH, Donahue HJ. Global cannabinoid receptor 1 deficiency affects disuse-induced bone loss in a site-specific and sex-dependent manner. J Biomech. 2023;146:111414.

    Article  PubMed  Google Scholar 

  110. Drago-Ferrante R, Di Fiore R, Karouia F, Subbannayya Y, Das S, Aydogan Mathyk B et al. Extraterrestrial gynecology: could Spaceflight increase the risk of developing Cancer in female astronauts? An updated review. Int J Mol Sci. 2022;23(13).

  111. Reyes DP, Masterova KS, Walton M, Kerstman EL, Antonsen EL. Assessment of Sex-Dependent Medical outcomes during Spaceflight. J Womens Health (Larchmt). 2022;31(8):1145–55.

    Article  PubMed  Google Scholar 

  112. Kuzichkin DS, Nichiporuk IA, Zhuravleva OA, Markin AA, Rykova MP, Zhuravleva TV, et al. Endothelial dysfunction markers and immune response indices in cosmonauts’ blood after long-duration space flights. NPJ Microgravity. 2022;8(1):46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Rienecker KDA, Grue K, Paladini MS, Frias ES, Frattini V, Borlongan MC, et al. Combined space stressors induce independent behavioral deficits predicted by early peripheral blood monocytes. Sci Rep. 2023;13(1):1749.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Herranz R, Anken R, Boonstra J, Braun M, Christianen PC, de Geest M, et al. Ground-based facilities for simulation of microgravity: organism-specific recommendations for their use, and recommended terminology. Astrobiology. 2013;13(1):1–17.

    Article  PubMed  PubMed Central  Google Scholar 

  115. van Loon JJWA. Some history and use of the random positioning machine, RPM, in gravity related research. Adv Space Res. 2007;39(7):1161–5.

    Article  Google Scholar 

  116. Mognato M, Girardi C, Fabris S, Celotti L. DNA repair in modeled microgravity: double strand break rejoining activity in human lymphocytes irradiated with gamma-rays. Mutat Res. 2009;663(1–2):32–9.

    Article  CAS  PubMed  Google Scholar 

  117. Dang B, Yang Y, Zhang E, Li W, Mi X, Meng Y, et al. Simulated microgravity increases heavy ion radiation-induced apoptosis in human B lymphoblasts. Life Sci. 2014;97(2):123–8.

    Article  CAS  PubMed  Google Scholar 

  118. Kim M, Jang G, Kim KS, Shin J. Detrimental effects of simulated microgravity on mast cell homeostasis and function. Front Immunol. 2022;13:1055531.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Canova S, Fiorasi F, Mognato M, Grifalconi M, Reddi E, Russo A, et al. Modeled microgravity affects cell response to ionizing radiation and increases genomic damage. Radiat Res. 2005;163(2):191–9.

    Article  CAS  PubMed  Google Scholar 

  120. Fu H, Su F, Zhu J, Zheng X, Ge C. Effect of simulated microgravity and ionizing radiation on expression profiles of miRNA, lncRNA, and mRNA in human lymphoblastoid cells. Life Sci Space Res (Amst). 2020;24:1–8.

    Article  PubMed  Google Scholar 

  121. Jiang M, Wang H, Liu Z, Lin L, Wang L, Xie M, et al. Endoplasmic reticulum stress-dependent activation of iNOS/NO-NF-κB signaling and NLRP3 inflammasome contributes to endothelial inflammation and apoptosis associated with microgravity. Faseb j. 2020;34(8):10835–49.

    Article  CAS  PubMed  Google Scholar 

  122. Zhang Y, Lu T, Wong M, Wang X, Stodieck L, Karouia F, et al. Transient gene and microRNA expression profile changes of confluent human fibroblast cells in spaceflight. Faseb j. 2016;30(6):2211–24.

    Article  CAS  PubMed  Google Scholar 

  123. Nastasi C, Mannarino L, D’Incalci M. DNA damage response and Immune Defense. Int J Mol Sci. 2020;21(20).

  124. Bednarski JJ, Sleckman BP. At the intersection of DNA damage and immune responses. Nat Rev Immunol. 2019;19(4):231–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Fontes FL, Pinheiro DM, Oliveira AH, Oliveira RK, Lajus TB, Agnez-Lima LF. Role of DNA repair in host immune response and inflammation. Mutat Res Rev Mutat Res. 2015;763:246–57.

    Article  CAS  PubMed  Google Scholar 

  126. Roggan MD, Kronenberg J, Wollert E, Hoffmann S, Nisar H, Konda B, et al. Unraveling astrocyte behavior in the space brain: Radiation response of primary astrocytes. Front Public Health. 2023;11:1063250.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Ikeda H, Muratani M, Hidema J, Hada M, Fujiwara K, Souda H et al. Expression Profile of Cell cycle-related genes in human fibroblasts exposed simultaneously to Radiation and simulated microgravity. Int J Mol Sci. 2019;20(19).

  128. Stowe RP, Mehta SK, Ferrando AA, Feeback DL, Pierson DL. Immune responses and latent herpesvirus reactivation in spaceflight. Aviat Space Environ Med. 2001;72(10):884–91.

    CAS  PubMed  Google Scholar 

  129. Yao X, Li H, Leng SX. Inflammation and immune system alterations in frailty. Clin Geriatr Med. 2011;27(1):79–87.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Wertheimer AM, Bennett MS, Park B, Uhrlaub JL, Martinez C, Pulko V, et al. Aging and cytomegalovirus infection differentially and jointly affect distinct circulating T cell subsets in humans. J Immunol. 2014;192(5):2143–55.

    Article  CAS  PubMed  Google Scholar 

  131. Horiuchi S, Wilmoth JR. Age patterns of the life table aging rate for major causes of death in Japan, 1951–1990. J Gerontol Biol Sci Med Sci. 1997;52(1):B67–77.

    Article  CAS  Google Scholar 

  132. Ferrucci L, Fabbri E. Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty. Nat Rev Cardiol. 2018;15(9):505–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Palacios-Pedrero M, Osterhaus A, Becker T, Elbahesh H, Rimmelzwaan GF, Saletti G. Aging and options to halt declining immunity to Virus infections. Front Immunol. 2021;12:681449.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Pinti M, Appay V, Campisi J, Frasca D, Fülöp T, Sauce D, et al. Aging of the immune system: focus on inflammation and vaccination. Eur J Immunol. 2016;46(10):2286–301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Oh SJ, Lee JK, Shin OS. Aging and the Immune System: the impact of immunosenescence on viral infection, immunity and vaccine immunogenicity. Immune Netw. 2019;19(6):e37.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Slaets H, Fonteyn L, Eijnde BO, Hellings N. Train your T cells: how skeletal muscles and T cells keep each other fit during aging. Brain Behav Immun. 2023;110:237–44.

    Article  CAS  PubMed  Google Scholar 

  137. Campisi M, Cannella L, Pavanello S. Cosmic chronometers: is spaceflight a catalyst for biological ageing? Ageing Res Rev. 2024;95:102227.

    Article  CAS  PubMed  Google Scholar 

  138. Sayed N, Huang Y, Nguyen K, Krejciova-Rajaniemi Z, Grawe AP, Gao T, et al. An inflammatory aging clock (iAge) based on deep learning tracks multimorbidity, immunosenescence, frailty and cardiovascular aging. Nat Aging. 2021;1:598–615.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Thomasini RL, Pereira DS, Pereira FSM, Mateo EC, Mota TN, Guimarães GG, et al. Aged-associated cytomegalovirus and Epstein-Barr virus reactivation and cytomegalovirus relationship with the frailty syndrome in older women. PLoS ONE. 2017;12(7):e0180841.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Moffat SD, An Y, Resnick SM, Diamond MP, Ferrucci L. Longitudinal change in Cortisol Levels across the Adult Life Span. J Gerontol Biol Sci Med Sci. 2020;75(2):394–400.

    Article  CAS  Google Scholar 

  141. Brauning A, Rae M, Zhu G, Fulton E, Admasu TD, Stolzing A et al. Aging of the Immune System: Focus on Natural Killer Cells Phenotype and functions. Cells. 2022;11(6).

  142. Reynolds RJ, Day SM. Mortality of US astronauts: comparisons with professional athletes. Occup Environ Med. 2019;76(2):114–7.

    Article  PubMed  Google Scholar 

  143. Fonte C, Jacob P, Vanet A, Ghislin S, Frippiat JP. Hindlimb unloading, a physiological model of microgravity, modifies the murine bone marrow IgM repertoire in a similar manner as aging but less strongly. Immun Ageing. 2023;20(1):64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Lescale C, Schenten V, Djeghloul D, Bennabi M, Gaignier F, Vandamme K, et al. Hind limb unloading, a model of spaceflight conditions, leads to decreased B lymphopoiesis similar to aging. Faseb j. 2015;29(2):455–63.

    Article  CAS  PubMed  Google Scholar 

  145. Liu B, Hinshaw RG, Le KX, Park MA, Wang S, Belanger AP, et al. Space-like (56)Fe irradiation manifests mild, early sex-specific behavioral and neuropathological changes in wildtype and Alzheimer’s-like transgenic mice. Sci Rep. 2019;9(1):12118.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Hinshaw RG, Schroeder MK, Ciola J, Varma C, Colletti B, Liu B et al. High-Energy, Whole-Body Proton Irradiation Differentially Alters Long-Term Brain Pathology and Behavior Dependent on Sex and Alzheimer’s Disease Mutations. Int J Mol Sci. 2023;24(4).

  147. Rudobeck E, Bellone JA, Szücs A, Bonnick K, Mehrotra-Carter S, Badaut J, et al. Low-dose proton radiation effects in a transgenic mouse model of Alzheimer’s disease - implications for space travel. PLoS ONE. 2017;12(11):e0186168.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Hajj-Boutros G, Sonjak V, Faust A, Balram S, Lagacé JC, St-Martin P et al. Myths and methodologies: understanding the health impact of head down bedrest for the benefit of older adults and astronauts. Study protocol of the Canadian Bedrest Study. Exp Physiol. 2024.

  149. Shimizu R, Hirano I, Hasegawa A, Suzuki M, Otsuki A, Taguchi K, et al. Nrf2 alleviates spaceflight-induced immunosuppression and thrombotic microangiopathy in mice. Commun Biol. 2023;6(1):875.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Ortiz RM, Wang TJ, Wade CE. Influence of centrifugation and hindlimb suspension on testosterone and corticosterone excretion in rats. Aviat Space Environ Med. 1999;70(5):499–504.

    CAS  PubMed  Google Scholar 

  151. Horie K, Kudo T, Yoshinaga R, Akiyama N, Sasanuma H, Kobayashi TJ, et al. Long-term hindlimb unloading causes a preferential reduction of medullary thymic epithelial cells expressing autoimmune regulator (Aire). Biochem Biophys Res Commun. 2018;501(3):745–50.

    Article  CAS  PubMed  Google Scholar 

  152. Wei LX, Zhou JN, Roberts AI, Shi YF. Lymphocyte reduction induced by hindlimb unloading: distinct mechanisms in the spleen and thymus. Cell Res. 2003;13(6):465–71.

    Article  CAS  PubMed  Google Scholar 

  153. Padgett DA, Glaser R. How stress influences the immune response. Trends Immunol. 2003;24(8):444–8.

    Article  CAS  PubMed  Google Scholar 

  154. Bierhaus A, Wolf J, Andrassy M, Rohleder N, Humpert PM, Petrov D, et al. A mechanism converting psychosocial stress into mononuclear cell activation. Proc Natl Acad Sci U S A. 2003;100(4):1920–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Torgersen KM, Vang T, Abrahamsen H, Yaqub S, Taskén K. Molecular mechanisms for protein kinase A-mediated modulation of immune function. Cell Signal. 2002;14(1):1–9.

    Article  CAS  PubMed  Google Scholar 

  156. Gu C, Ma YC, Benjamin J, Littman D, Chao MV, Huang XY. Apoptotic signaling through the beta -adrenergic receptor. A new Gs effector pathway. J Biol Chem. 2000;275(27):20726–33.

    Article  CAS  PubMed  Google Scholar 

  157. Myklebust JH, Josefsen D, Blomhoff HK, Levy FO, Naderi S, Reed JC, et al. Activation of the cAMP signaling pathway increases apoptosis in human B-precursor cells and is associated with downregulation of Mcl-1 expression. J Cell Physiol. 1999;180(1):71–80.

    Article  CAS  PubMed  Google Scholar 

  158. Parvathenani LK, Buescher ES, Chacon-Cruz E, Beebe SJ. Type I cAMP-dependent protein kinase delays apoptosis in human neutrophils at a site upstream of caspase-3. J Biol Chem. 1998;273(12):6736–43.

    Article  CAS  PubMed  Google Scholar 

  159. Dragoş D, Tănăsescu MD. The effect of stress on the defense systems. J Med Life. 2010;3(1):10–8.

    PubMed  PubMed Central  Google Scholar 

  160. Marino F, Cosentino M. Adrenergic modulation of immune cells: an update. Amino Acids. 2013;45(1):55–71.

    Article  CAS  PubMed  Google Scholar 

  161. Shearer WT, Reuben JM, Mullington JM, Price NJ, Lee BN, Smith EO, et al. Soluble TNF-alpha receptor 1 and IL-6 plasma levels in humans subjected to the sleep deprivation model of spaceflight. J Allergy Clin Immunol. 2001;107(1):165–70.

    Article  CAS  PubMed  Google Scholar 

  162. Zilioli S, Jiang Y. Endocrine and immunomodulatory effects of social isolation and loneliness across adulthood. Psychoneuroendocrinology. 2021;128:105194.

    Article  CAS  PubMed  Google Scholar 

  163. Schmitt DA, Schaffar L. Isolation and confinement as a model for spaceflight immune changes. J Leukoc Biol. 1993;54(3):209–13.

    Article  CAS  PubMed  Google Scholar 

  164. Crucian BE, Makedonas G, Sams CF, Pierson DL, Simpson R, Stowe RP, et al. Countermeasures-based improvements in stress, Immune System Dysregulation and Latent Herpesvirus Reactivation onboard the International Space Station - Relevance for Deep Space missions and Terrestrial Medicine. Neurosci Biobehav Rev. 2020;115:68–76.

    Article  CAS  PubMed  Google Scholar 

  165. Crucian BE, Choukèr A, Simpson RJ, Mehta S, Marshall G, Smith SM, et al. Immune System Dysregulation during Spaceflight: potential countermeasures for Deep Space Exploration missions. Front Immunol. 2018;9:1437.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Cervantes JL, Hong BY. Dysbiosis and Immune Dysregulation in outer space. Int Rev Immunol. 2016;35(1):67–82.

    CAS  PubMed  Google Scholar 

  167. Vaishampayan A, Grohmann E. Multi-resistant biofilm-forming pathogens on the International Space Station. J Biosci. 2019;44(5).

  168. Mehta SK, Laudenslager ML, Stowe RP, Crucian BE, Sams CF, Pierson DL. Multiple latent viruses reactivate in astronauts during space shuttle missions. Brain Behav Immun. 2014;41:210–7.

    Article  CAS  PubMed  Google Scholar 

  169. Simon Á, Smarandache A, Iancu V, Pascu ML. Stability of Antimicrobial Drug Molecules in different gravitational and Radiation conditions in View of applications during outer space missions. Molecules. 2021;26(8).

  170. Du B, Daniels VR, Vaksman Z, Boyd JL, Crady C, Putcha L. Evaluation of physical and chemical changes in pharmaceuticals flown on space missions. Aaps j. 2011;13(2):299–308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Blue RS, Chancellor JC, Antonsen EL, Bayuse TM, Daniels VR, Wotring VE. Limitations in predicting radiation-induced pharmaceutical instability during long-duration spaceflight. NPJ Microgravity. 2019;5:15.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Turroni S, Magnani M, Kc P, Lesnik P, Vidal H, Heer M. Gut microbiome and space travelers’ health: state of the art and possible Pro/Prebiotic strategies for long-term space missions. Front Physiol. 2020;11:553929.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Schuck Edgar LGM, Derendorf Hartmut. Effect of simulated microgravity on the Disposition and tissue penetration of ciprofloxacin in healthy volunteers. J Clin Pharmacol. 2013;45(7):822–31.

    Article  Google Scholar 

  174. Rumble RH, Roberts MS, Scott AR. The effects of posture on the pharmacokinetics of intramuscular benzylpenicillin. Eur J Clin Pharmacol. 1988;33(6):629–35.

    Article  CAS  PubMed  Google Scholar 

  175. Andrews J, Kendall MJ, Mitchard M. Factors influencing the absorption and disposition of mecillinam and pivmecillinam in man. Br J Clin Pharmacol. 1976;3(4):627–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Pavez Loriè E, Baatout S, Choukér A, Buchheim JI, Baselet B, Dello Russo C, et al. The future of Personalized Medicine in Space: from observations to countermeasures. Front Bioeng Biotechnol. 2021;9:739747.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Basner M, Dinges DF, Mollicone DJ, Savelev I, Ecker AJ, Di Antonio A, et al. Psychological and behavioral changes during confinement in a 520-day simulated interplanetary mission to mars. PLoS ONE. 2014;9(3):e93298.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Petit G, Cebolla AM, Fattinger S, Petieau M, Summerer L, Cheron G, et al. Local sleep-like events during wakefulness and their relationship to decreased alertness in astronauts on ISS. NPJ Microgravity. 2019;5:10.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Brainard GC, Barger LK, Soler RR, Hanifin JP. The development of lighting countermeasures for sleep disruption and circadian misalignment during spaceflight. Curr Opin Pulm Med. 2016;22(6):535–44.

    Article  PubMed  Google Scholar 

  180. Zhang S, Wimmer-Schweingruber RF, Yu J, Wang C, Fu Q, Zou Y et al. First measurements of the radiation dose on the lunar surface. Sci Adv. 2020;6(39).

  181. Stepanenko V, Kaprin A, Ivanov S, Shegay P, Zhumadilov K, Petukhov A, et al. Internal doses in experimental mice and rats following exposure to neutron-activated (56)MnO(2) powder: results of an international, multicenter study. Radiat Environ Biophys. 2020;59(4):683–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Kim M-HY, Wilson JW, Cucinotta FA, Simonsen LC, Atwell W, Badavi FF et al. Contribution of High Charge and Energy (HZE) Ions During Solar-Particle Event of September 29, 1989. 1999, 1999.

  183. Hu S, Cucinotta FA. Characterization of the radiation-damaged precursor cells in bone marrow based on modeling of the peripheral blood granulocytes response. Health Phys. 2011;101(1):67–78.

    Article  CAS  PubMed  Google Scholar 

  184. Clapp NK, Darden EB Jr., Jernigan MC. Relative effects of whole-body sublethal doses of 60-MeV protons and 300-kVp X-rays on disease incidences in RF mice. Radiat Res. 1974;57(1):158–86.

    Article  CAS  PubMed  Google Scholar 

  185. Task Group on Radiation Quality Effects in Radiological Protection Committee 1 on Radiation Effects International Commission on Radiological Protection. Relative biological effectiveness (RBE), quality factor (Q), and radiation weighting factor (w(R)). A report of the International Commission on Radiological Protection. Ann ICRP. 2003;33(4):1–117.

    Google Scholar 

  186. George K, Willingham V, Wu H, Gridley D, Nelson G, Cucinotta FA. Chromosome aberrations in human lymphocytes induced by 250 MeV protons: effects of dose, dose rate and shielding. Adv Space Res. 2002;30(4):891–9.

    Article  CAS  PubMed  Google Scholar 

  187. Edwards AA, Lloyd DC, Prosser JS, Finnon P, Moquet JE. Chromosome aberrations induced in human lymphocytes by 8.7 MeV protons and 23.5 MeV helium-3 ions. Int J Radiat Biol Relat Stud Phys Chem Med. 1986;50(1):137–45.

    Article  CAS  PubMed  Google Scholar 

  188. George KA, Hada M, Cucinotta FA. Biological effectiveness of Accelerated protons for chromosome exchanges. Front Oncol. 2015;5:226.

    Article  PubMed  PubMed Central  Google Scholar 

  189. Huff JL, Poignant F, Rahmanian S, Khan N, Blakely EA, Britten RA, 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). 2023;36:90–104.

    Article  PubMed  Google Scholar 

  190. Chancellor JC, Scott GB, Sutton JP. Space Radiation: the Number one risk to Astronaut Health beyond Low Earth Orbit. Life (Basel). 2014;4(3):491–510.

    PubMed  Google Scholar 

  191. Safe Human Expeditions Beyond Low Earth Orbit (LEO). National Aeronautics and Space Administration (NASA). 2022. https://ntrs.nasa.gov/api/citations/20220002905/downloads/NESC-RP-20-01589_NASA-TM-20220002905final.pdf.

Download references

Acknowledgements

This work was supported by the National Aeronautics and Space Administration Human Research Program (HW), University Space Research Association internship program (AW), and Nnational Institutes of Health award number T32GM144273 from the National Institute of General Medical Sciences (AW). Figures were generated using BioRender.com, WebPlotDigitizer tool, and GraphPad Prism.

Funding

Funding provided by the National Aeronautics and Space Administration, University Space Research Association, and the National Institutes of Health. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Aeronautics and Space Administration, National Institute of General Medical Sciences, or National Institutes of Health. The funding bodies had no role in directing the design of the study, analysis, interpretation of data, writing the manuscript, or decision to publish.

Author information

Authors and Affiliations

Authors

Contributions

Conception and design of this review (AW, HW). Acquisition, analysis, or interpretation of data for this work (AW, MMV). Drafting of the work and all figures (AW) or revising it critically for intellectual content (AW, HW, MMV, BC). All authors approved of the final version of the manuscript and agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Corresponding author

Correspondence to Anna Wadhwa.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wadhwa, A., Moreno-Villanueva, M., Crucian, B. et al. Synergistic interplay between radiation and microgravity in spaceflight-related immunological health risks. Immun Ageing 21, 50 (2024). https://doi.org/10.1186/s12979-024-00449-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12979-024-00449-w

Keywords