- 1Laboratory of Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- 2Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
- 3Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States
- 4Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, United States
- 5Cancer Dormancy Institute, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, United States
- 6Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- 7Tumor Microenvironment Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, United States
- 8Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, United States
- 9Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, NY, United States
- 10The Marilyn and Stanely M. Katz Institute for Immunotherapy for Cancer and Inflammatory Disorders, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, United States
Spaceflight imposes a constellation of physiological challenges—cosmic radiation, microgravity, disrupted circadian rhythms, and psychosocial stress—that critically compromise astronaut health. Among the most vulnerable organs is the thymus, a cornerstone of immune system functionality, tasked with generating naive T cells essential for adaptive immunity. The thymus is particularly sensitive to spaceflight conditions, as its role in maintaining immune homeostasis is tightly regulated by a balance of systemic and local factors easily disrupted in space. Cosmic radiation, an omnipresent hazard beyond Earth’s magnetosphere, accelerates DNA damage and cellular senescence in thymic epithelial cells, impairing thymopoiesis and increasing the risk of immune dysregulation. Microgravity and circadian rhythm disruption exacerbate this by altering immune cell migration patterns and stromal support, critical for T-cell development. Psychosocial stressors, including prolonged isolation and mission-induced anxiety, further compound thymic atrophy by elevating systemic glucocorticoid levels. Ground-based analogs simulating cosmic radiation and microgravity have been instrumental in elucidating mechanisms of thymic involution and its downstream effects on immunity. These models reveal that long-duration missions result in diminished naive T-cell output, leaving astronauts vulnerable to infections and possibly at high risk for developing neoplasia. Advances in countermeasures, such as pharmacological interventions targeting thymic regeneration and bioengineering approaches to protect thymic architecture, are emerging as vital strategies to preserve immune resilience during prolonged space exploration. Focusing on the thymus as a central hub of immune vulnerability underscores its pivotal role in spaceflight-induced health risks. Understanding these dynamics will not only enhance the safety of human space missions but also provide critical insights into thymus biology under extreme conditions.
1 Overview of spaceflight stressors affecting the immune system
Astronauts experience hostile environmental changes and stressors during spaceflight, broadly classified into four distinct categories: cosmic radiation, microgravity, circadian derailment, and psychosocial stressors, the latter including social isolation, various constraints and fears, crew member conflicts, and extreme pressure for exceptional mission performance. Together, these factors have a significant impact on many physiological systems in the body, eventually posing an obstacle to long-term space missions (1–11).
Beyond the Earth’s protective magnetosphere, astronauts are exposed to elevated levels of galactic cosmic radiation (GCR) and solar energetic particles (SEP), both of which pose significant health risks. GCR, consisting of high-energy protons and atomic nuclei, and SEP, primarily composed of charged particles from the Sun, are highly penetrating, and can damage cellular structures, DNA, and tissues. Due to the challenges in replicating the precise radiation environment of space in Earth-based facilities, it remains difficult to fully assess the long-term health consequences of chronic exposure to GCR and SEP (12), estimated to be approximately 1mSV per day spent at the international space station (ISS) (13). Nevertheless, the primary risks associated with this exposure include an increased likelihood of cancer development, central nervous system (CNS) defects that contribute to cognitive and behavioral impairments, as well as neurological and cardiovascular disorders. Additionally, radiation exposure has been shown to lead to an acute or progressive decline in immune system functions, which can severely impact astronaut health and mission success (12).
In addition to the constant but low-dose GCR/SEP exposure, any gravitational forces less than 1x10-3 g, including those at ~1x10-6 g (i.e., microgravity), which are typically experienced during spaceflight, may impose additional stress, particularly to the musculoskeletal system (14–18). Indeed, during international space station (ISS) missions, astronauts experience a significant reduction in bone mineral density along with muscular atrophy, triggering the inclusion of physical training routines during spaceflight as an essential countermeasure (19–21). However, it has long been established that muscles and bones related to posture and weight are inherently linked to the gravitational load, and as such, can be severely affected by its perturbations (22–24). On the other side, the effect of microgravity on other organ systems, especially the immune system, are not appreciated, and key observations are only now beginning to emerge (25).
Spaceflight also presents unique challenges to circadian rhythms, primarily due to absence of a consistent 24-hour light-dark cycle. In spaceflight, the continuous artificial lighting and the lack of natural sunlight cues disturb the body’s internal clock, leading to fragmented sleep patterns and impaired performance (26–30). Such disruptions pose significant risks for long-term missions, as sleep disorders and desynchronized circadian rhythms can heighten the behavioral risks and psychiatric disorders (31). In addition, a number of studies over the years highlight a complex link between circadian rhythms and immune function (32–34), which may lead to severe symptoms, such as obesity, metabolic syndromes, cardiovascular disease, and cancer (31, 35–37), thus inferring that such disruptions may further impact health and resilience in space (31).
Psychosocial stressors in spaceflight, including isolation, confinement, and interpersonal challenges, can significantly impact astronaut health, including feelings of loneliness, anxiety, and depression, exacerbated by the absence of natural light, radiation effects, microgravity effects, and/or long-duration separation from family. Observational research from ISS missions, the Mars500 simulation, and other space-analogue environments, has documented significant psychological strain, mood swing, irritability, cognitive impacts, and interpersonal conflicts (38–43). Currently, there is ample evidence that psychosocial stressors can lead to the dysfunction of the immune system (44), although establishing direct causative link is challenging, due to the complex interplay of factors unique to spaceflight conditions.
In summary, the stressors encountered during spaceflight—cosmic radiation, microgravity, circadian disruption, and psychosocial challenges—can independently affect astronaut health, particularly the immune system. These factors likely interact with each other, amplifying their negative impact on immune functions. Understanding the mechanisms behind these interactions is therefore crucial for mitigating health risks on long-duration missions. A compromised immune system can hinder the ability to fight infections and recover from illness aboard a manned mission, making it essential to safeguard immune health for the success of space exploration.
2 Effects of spaceflight stressors on thymus homeostasis and involution
Several studies have shown that spaceflight stressors perturb immune system homeostasis and immunological responses to pathogens (45–58). The human alpha herpesviruses, such as herpes simplex virus (HSV-1) and varicella zoster virus (VZV), may enter a latent state in cranial nerve ganglia but can reactivate when stress impacts immune regulation (59, 60). During spaceflight, reactivation of viruses like Epstein-Barr (EBV), VZV, HSV-1, and cytomegalovirus (CMV) often occurs without symptoms, although live virus particles have been isolated, and viral shedding rates increase with mission length. As a consequence, extended missions (>180 days) could heighten the risk of developing symptomatic infections in astronauts, such as skin rash dermatitis, posing an incremental health concern and impairing their performance (61–67). Additional evidence of adverse immunological manifestations occurring during either short- or long-duration spaceflight missions comes from measured disturbances in immune-related cytokine levels in astronauts, such as tumor necrosis factor-α (TNFα) and interferon-γ (IFNγ) among others (58, 68, 69), which are also known to be essential for thymus homeostasis (70–72).
Studies on the impact of spaceflight on immune system development and lymphoid organ homeostasis are limited, and comprise of mixed observations from astronauts and rodents (73–75). With regards to the bone marrow, it has been demonstrated that spaceflight may disrupt both the mesenchymal (MSC) and hematopoietic stem cell (HSC) compartments, thus affecting the differentiation and maturation of descendant lineages, particularly B cells, myeloid cells, and erythrocytes (15, 20, 76, 77). An in-depth analysis of the impact of spaceflight on bone marrow homeostasis, however, is beyond the scope of this article, but the readers are encouraged to review relevant work by others (25, 78–81).
Concrete evidence that spaceflight affects thymic functions and causes involution has been recently demonstrated in a critical study that investigated the effects of long-term spaceflight in 16 astronauts during a median 184-day mission aboard the International Space Station (ISS) (82). Thymopoiesis was assessed in each astronaut at multiple timepoints by measuring T-cell receptor excision circles (TREC) (82), a molecular marker detectable in recent thymic emigrants (83–85). Samples were collected approximately 180 days before launch, within 2–4 hours of landing, and up to 180 days post-landing. A consistent and significant decline in thymopoiesis was observed immediately after landing, followed by a return to preflight levels within days to weeks, eventually stabilizing to the preflight range (82). Interestingly, the study identified an inverse correlation between cortisol levels and thymic output (82), suggesting that glucocorticoid-induced thymocyte apoptosis may in part contribute to reduced thymopoiesis during spaceflight. Thymic involution was also observed in experimental mice housed aboard the ISS for 35 days (86), and the Space Shuttle Atlantis for 13 days (87). Notably, significant thymic mass loss occurred only in the former, although DNA fragmentation assays indicated increased apoptosis in the thyme of mice exposed to spaceflight in the latter (86, 87). Therefore, mission duration is critical for a substantial impact on thymic integrity. Altogether, these findings suggest that extended space missions compromise immune and thymic function, and increase infection susceptibility. Additionally, they underscore not only the critical need for developing countermeasures to enhance immune resilience, but also the importance of developing faithful ground-based analogues to investigate in more detail immune dysfunctions from a more mechanistic perspective.
3 Ground-based models that recapitulate spaceflight-induced thymic involution
Given the high cost and limited opportunities to conduct spaceflight experiments with model organisms, ground-based models simulating spaceflight conditions have been developed as practical and accessible alternatives. These models aim to replicate key stressors encountered during spaceflight, as outlined above, offering insights into their physiological effects on thymus homeostasis. Studying the impact of spaceflight on immune system development is particularly challenging in humans due to ethical and logistical constraints. As a result, rodent models have become the primary choice for such investigations, providing valuable data, while serving as an approximation of human responses. In this section, we will thus describe and critically assess the most well-established ground-based models currently regarded as relatively equivalent to actual spaceflight conditions, highlighting their utility, limitations, and relevance to understanding spaceflight-induced stressors.
3.1 Cosmic radiation
Most studies investigating the impact of space radiation on the hematopoietic system have been conducted using monoenergetic electron and gamma-ray beams. Exposure of rats to gamma rays was performed on board of the satellite Cosmos-690 along with a control group receiving matched dosing on Earth (88). Hematopoietic assessments demonstrated a significantly enhanced effect in rats irradiated in spaceflight, when compared to rats irradiated on Earth, with severe suppression of bone marrow hematopoiesis and thymopoiesis (88). Along these lines, the exposure of rat thymocyte suspensions to Co-60 gamma-rays induced severe apoptosis and distinct morphological and functional changes in thymocytes, assessed via electron microscopy, DNA fragmentation assays, and biochemical assays (89). Further mechanistic insights revealed an activation of intracellular and intranuclear proteases, typical of the extrinsic apoptotic pathway, leading to the degradation of mitochondria and the release of pro-apoptotic factors (90). However, a similar study on Cosmos-690 that measured the combined effects of microgravity and ionizing radiation from a Cs-137 source did not reveal significant changes in thymus weight and spleen after irradiation, although bone marrow hematopoiesis was affected (91). In two separate studies, exposure to Fe-56 particles, or C-12 (6+) ions induced severe spleen and bone marrow defects, as well as thymic involution in adult female C57BL/6 or King-Ming strain mice, respectively, demonstrating varying degrees of susceptibility for lymphocyte populations (92, 93). Collectively, these studies demonstrate the pleiotropic effects of diverse monoenergetic ion sources on thymic structure and function, although the precise mechanistic insights behind the observed variability are not fully understood. It should be noted however, that most research to evaluate health risks from space radiation has been historically performed via acute exposure to such monoenergetic single-ion beams, as outlined in the studies above. Nevertheless, it has now been established that such exposures do not faithfully recapitulate the intricacies of the galactic ray environment in our solar system (12), and as such, should be interpreted with caution.
To address such concerns, ground-based GCR simulators have been developed to expose experimental animals and cell cultures to “mission-relevant” radiation doses. These simulators incorporate diverse vehicle and shielding configurations, high design fidelity, precise material characterization, mission duration considerations, and realistic solar conditions (94–101). The most advanced, developed by the NASA Space Radiation Laboratory (NSRL) at Brookhaven National Laboratory, delivers radiation doses comprising a mixture of protons (~65%-75%), helium ions (~10%-20%), and heavier ions (C, O, Si, Ti, Fe) (102). To more closely replicate the low-dose rates found in space, this system can additionally fractionate sequential field exposures over daily intervals for 2 to 6 weeks, allowing state-of-the-art cellular and animal model systems to be exposed to mission-relevant radiation (12). So far, sophisticated GCR simulation has been used to examine various organ system adaptations to space- and mission-relevant radiation doses, including the gastrointestinal, endocrine, cardiovascular, immune, ocular, and central nervous systems (103–111). To our knowledge however, there are currently no studies explicitly dedicated to evaluating thymus architecture and functions using GCR simulators.
Instead, most studies using GCR simulators have focused on the effects of mission-relevant GCR exposure on the bone marrow. For instance, mice exposed to mission-equivalent GCR doses showed increased osteoclast activity and trabecular bone loss, suggesting alterations in the endosteal niche (112, 113), which regulates hematopoiesis (114–116). Simulated SEP and GCR radiation also disrupted the ability of MSCs to support hematopoiesis and directly impaired human hematopoietic stem cell (HSC) functionality, inducing DNA damage and mutations. Sequential exposure to protons and iron ions, mimicking deep space radiation, was particularly harmful to HSC genome integrity. Notably, sequential exposure to protons and iron ions—mimicking the complexity of deep space radiation—proved significantly more harmful to HSC genome integrity and function than exposure to either particle type alone (117). These findings emphasize once again the importance of simulating the full spectrum of galactic cosmic radiation for accurate assessments. Collectively, these studies suggest that GCR may impact thymopoiesis indirectly by disrupting bone marrow hematopoiesis and the influx of early thymic progenitors. However, the possibility of direct effects of GCR/SEP on the thymus itself cannot be excluded, as indicated from astronaut observations and rodent experiments (25, 82, 118).
3.2 Microgravity
Development of ground-based models replicating microgravity is particularly challenging. Parabolic flights conducted on Earth on one side accurately replicate the gravitational conditions experienced in orbital spaceflight. Indeed, numerous experiments have explored the effects of microgravity on physiological systems using this approach (119). However, the duration of induced microgravity during parabolic flights is typically limited to several minutes, making it unsuitable for assessing long-term effects. Since the impact of microgravity on the thymus likely occurs over days, this model is likely inadequate for evaluating thymic responses.
For in vitro experiments, devices such as the clinostat and magnetic levitation are commonly used to simulate microgravity. These models are primarily restricted to cultured cell studies, thus posing an impediment to recapitulate the complex microenvironment of lymphoid organs. However, fetal thymic organ cultures, typically derived from E14 to E16 mouse embryos, can be maintained in vitro, and physiologically recapitulate stromal-thymocyte interactions (120), potentially enabling the study of simulated microgravity effects on thymocyte development. Indeed, a clinostat study demonstrated a reduction in CD4+CD8+ thymocytes after a 12-day fetal thymic organ culture (121). Nonetheless, these findings should be interpreted with caution, as the cellular composition of the fetal thymus differs significantly from the adult one, highlighting the need for further studies to understand the effects of microgravity on adult thymic function.
The hindlimb unloading (HU) model, also known as the tail suspension model, is frequently used to simulate weightlessness in rodents (122). This model removes weight-bearing from the hindlimbs, impacting the musculoskeletal system, and causing a redistribution of body fluids towards the head, analogous to the fluid shifts observed in humans under microgravity conditions. Besides musculoskeletal ramifications, short-term (2-day) HU in mice resulted in reduced thymic mass, with CD4+CD8+ thymocytes being particularly sensitive (123). The total number of mature single-positive (CD4+CD8- or CD4-CD8+) thymocytes was markedly reduced and accompanying TUNEL assays indicated an increase in apoptotic cells in the thymus (123). Combined with steroid receptor blocking experiments, these findings also suggested that corticosterone-dependent apoptosis is responsible for thymic cell reduction during short-term HU. Another study revealed that osteopontin is involved in HU-induced thymic apoptosis by regulating corticosterone levels during a 3-day HU (124). Subsequent studies have demonstrated that circulatory osteopontin can interfere with the hypothalamus-pituitary-adrenal (HPA) axis, thus regulating steroid hormone production and modulating stress responses (125), although the precise mechanisms behind this regulation have not been elucidated. In contrast, long-term HU does not selectively reduce CD4+CD8+ thymocytes, despite a decrease in overall thymic mass (126). Instead, long-term HU led to significant decline in medullary thymic epithelial cells (TECs), particularly those expressing high levels of CD80 and the autoimmune regulator (AIRE). Consistent with this, the expression of tissue-specific antigens was downregulated in the thymus of long-term HU-loaded mice (126). Together, these findings indicate that the effects of HU are distinctly time-dependent: short-term HU selectively induces corticosterone-driven apoptosis in CD4+CD8+ thymocytes, while long-term HU impacts all thymocytes in a non-selective manner, along with the AIRE+ mTEC (i.e., mTEChi) population, likely through apoptosis-independent mechanisms, despite the persistently elevated corticosterone levels in either condition.
Several studies have indicated that spaceflight induces thymic involution in mice with similar lesions to those observed in the HU model (74, 75, 86, 87, 118, 126), suggesting that hindlimb unloading may effectively replicate some aspects of spaceflight conditions. However, a key limitation of the HU model is that it not only simulates weightlessness, but also induces psychological stress in mice, which can act as a model for depression (127). This introduces complexity in interpreting results, because it becomes challenging to differentiate whether thymic atrophy is due to stress, musculoskeletal changes, fluid redistribution, or a combination of these factors. All these conditions are present under both microgravity and hindlimb unloading environments.
3.3 Circadian derailment
Circadian rhythms control many aspects of human physiology, affecting daily variations in body temperature, blood pressure, and hormone levels and coordinate function across different organ systems, including neurological, metabolic, endocrine, cardiovascular, and immune (128). Circadian rhythmicity in the body is entrained by photic cues and a tight network of central and peripheral clocks enabled by a neural pacemaker directly responsive to environmental and behavioral states such as the sleep-wake cycles, feeding, metabolic cues, and secretion of hormones (particularly glucocorticoids) (129–131).
Circadian derailment is considered a risk factor during space missions by NASA. During space flight, astronauts are exposed to changes in microgravity, which impose pathophysiological effects on circadian rhythmicity, leading to derailment as a consequence of disturbed sleep, wakefulness, and feeding patterns (30, 31). Astronauts working at the ISS experience 16 sunrises and sunsets within a 24-hour period, impairing the 24-hour diurnal cycle experienced on earth. Even more so, the profound workload during space missions, which requires astronauts to complete highly complex tasks during long periods of time, contributes to the disruption of sleep-wakefulness cycles that collectively affect the body’s physiological diurnal rhythms (132–135). Derailment of circadian rhythm affects human health as increased occurrence of cardiovascular disease (CVD) (136), metabolic disorders (137), and cancer (138–140) were reported to be associated with shift work or frequent time zone travel. Coupled with other hazards of spaceflight, derailment of circadian oscillations during space missions may result in considerable risk to astronaut health, including not only sleep deprivation and diminished alertness, loss of cognitive abilities, depression, and anxiety (141, 142), but also the development of metabolic syndrome, CVD and cancer.
The hematopoietic and immune systems are particularly sensitive to circadian derailment. Mobilization and trafficking of leukocytes and hematopoietic stem and progenitor cells (HSPCs) between lymphoid organs and other tissues in the body is tightly regulated by central and peripheral clocks (34, 143–145). Innate immune cells (including granulocytes, monocytes, and macrophages) and T and B cells exhibit strong circadian oscillations in peripheral blood, peaking during the behavioral rest phase (daytime in rodents, and at night in humans) (146–148). Oscillation of blood lymphocytes was demonstrated to depend on glucocorticoids, catecholamines, and hypoxia-inducible factor 1a (HIF-1a) (149–151) that mediate rhythmic expression of chemokine receptors (e.g., CXCR4, CXCR5, CCR7, CX3CR1) that oscillate in phase with tissue-specific chemokines (e.g., CXCL12 in bone marrow and lung and CCL21 in lymph nodes) and endothelial adhesion molecules, including P and E-Selectin, Intercellular adhesion molecule-1 (ICAM-1), ICAM-2, and vascular cell adhesion molecule-1 (VCAM-1) across lymphoid and other organs, (including liver, skin, gut and lung) (146). Besides leukocyte trafficking and recruitment into tissues, recent studies have demonstrated that innate and adaptive immune responses depend on circadian rhythmicity, including response to pathogens, B cell development, and T cell differentiation. Circadian control of immune response is not the scope of this review; a detailed summary of this topic can be found elsewhere (34, 152).
Whether and how derailment of circadian rhythmicity affects thymic function and T cell development is less known. Although it has been shown that loss of intrinsic circadian rhythms by deletion of the master clock regulator Brain and Muscle Arnt-like protein-1 (Bmal1) in thymocytes does not affect T cell development (153), CD4+ single positive (SP4) thymocyte emigration from the thymus was shown to be regulated by circadian rhythms, as well as rhythmic expression of emigration related-molecules sphingosine 1-phosphate receptor (S1PR1) and C-C chemokine receptor 2 (CCR2) (154). As spaceflight and altered microgravity were shown to induce thymic involution (75, 87), it is yet to be determined to what extent the derailment of circadian rhythmicity contributes to thymic dysfunction. Future studies utilizing ground-based models of acute and chronic jet lag will directly test this question and determine how circadian derailment affects thymic structure and functionality. However, as spaceflight is associated with microgravity disruption, which can also contribute to impaired circadian rhythmicity (155–158), a combination of jet lag with hindlimb unloading may be necessary to properly simulate spaceflight conditions that derail circadian rhythmicity. Furthermore, it will be important to show to what extent derailment of circadian rhythmicity during spaceflight contributes to the development of CVD and cancer, as defective immune response contributes to the pathogenicity of either condition.
3.4 Psychosocial stress
During prolonged space missions, astronauts are exposed to extreme environments for extended durations, potentially leading to adverse physical and mental health effects, such as depression and cognitive impairment. The concept of “long-term spaceflight composite stress” (LSCS) encapsulates the multifaceted sources of stress encountered in space (142). Among these, psychosocial stress stands out as a significant contributor, distinct from well-known hazards like cosmic radiation, microgravity, and circadian disruptions (142). Instead, it arises from factors such as social isolation, confinement in cramped and crowded spaces, cultural differences and conflicts among crew members, homesickness, performance anxiety, and persistent noise from the onboard equipment (e.g., fans, exercise machines, life-support systems) (142). However, studying the isolated effects of psychosocial stressors on normal physiology, and the immune system, in ground-based rodent models presents significant challenges. The multifactorial nature of these stressors is inherently difficult to replicate in controlled laboratory settings (159–164). Additionally, fundamental differences between humans and rodents further complicate such models: the human brain, with its unparalleled complexity and advanced cognitive and emotional capacities, processes psychosocial stimuli in ways that are not easily mirrored in rodent counterparts (165).
The effects of LSCS have been previously studied, although psycho-social factors have not been isolated from other spaceflight-associated stressors. For example, a 42-day simulation combining microgravity, isolation, noise, circadian rhythm disturbances, and low pressure demonstrated significant weight loss, anxiety, memory deficits, and depression in rats. These behavioral changes correlated with reduced postsynaptic density thickness and synaptic interface curvature, indicating impaired synaptic plasticity in the hippocampus of LSCS-exposed rats (166–168). While a connection between depression and immune system dysregulation is loosely supported (169), direct evidence linking LSCS to immunological and thymic functions is still lacking.
4 Conclusions, future perspectives, and translational (space-)blocks
Spaceflight-induced thymic involution is a complex phenomenon influenced by composite stressors, highlighting the necessity of developing faithful ground-based models to complement spaceflight research. The logistical and financial challenges of conducting rodent experiments in space make such models indispensable. However, most existing models focus on isolating single stressors, such as hindlimb unloading to simulate microgravity, or galactic cosmic ray (GCR) simulators to replicate radiation exposure. While these approaches provide valuable insights into the individual contributions of specific stressors to thymic dysfunction, they fail to replicate the multifactorial nature of the space environment, where these stressors act simultaneously. Multi-hit models, also known as long-term spaceflight composite stress (LSCS) models, which incorporate multiple stressors on the other side may offer a more comprehensive solution to this challenge, as they may reveal their synergistic or additive effects (73, 111, 142, 166, 170). Notably, certain ground-based models, such as the HU may be inherently multifactorial themselves, raising further concerns regarding their interpretation. As mentioned above HU introduces psychological stress to mice (127), thus making it a marginally LSCS model. While these models are promising, further studies are essential to determine their capacity to reliably replace spaceflight experiments, particularly in mimicking the intricate interplay of stressors experienced in space.
In ground-based models, thymic involution is primarily associated with loss of double-positive (CD4+CD8+) thymocytes, a sensitive subset that often serves as an early indicator of stress-induced thymic damage (171–177). Thymic involution in these models is typically observed following short-term exposures to stressors such as monoenergetic radiation beams or brief hindlimb unloading. Such changes are often driven by overstimulation of the hypothalamic-pituitary-adrenal (HPA) axis, resulting in elevated corticosterone or cortisol levels. However, space missions are expected to impose prolonged stressors, necessitating models that investigate the effects of extended exposures. Notably, ground-based experiments involving longer durations, such as extended hindlimb unloading or sophisticated galactic cosmic ray (GCR) simulations, reveal distinct thymic alterations. Beyond thymocyte loss, these exposures significantly affect the thymic stroma, particularly thymic epithelial cells (TECs) (25), which are critical for maintaining thymic architecture and supporting thymocyte development and selection (178–184). This shift underscores that long-term stressors may more profoundly impair the thymus by targeting its regenerative infrastructure rather than inducing acute thymocyte depletion. Accordingly, future countermeasures should prioritize the preservation and regeneration of the thymic stroma, especially TECs, to ensure the recovery and sustained functionality of the thymus during prolonged spaceflight.
Thymic involution during spaceflight poses both immediate and long-term risks to astronaut health (25). Interestingly, the recovery of thymic function shortly after returning to Earth was shown in astronauts (82), highlighting the organ’s regenerative capacity and intrinsic plasticity. However, during extended missions in deep space, the thymus may face sustained functional compromise. Prolonged thymic involution could lead to a diminished T-cell receptor repertoire, impaired immune surveillance, and weakened systemic immunity (173, 185–187). These effects may heighten susceptibility to infections, including reactivation of latent viruses, and potentially increase the long-term risk of cancer or other immunological diseases (25, 188, 189). While limited epidemiological data do not currently suggest a higher cancer incidence among astronauts compared to the general population (190–192), further monitoring and research are critical to comprehensively assess these risks. The thymus plays a particularly vital role in children, where it establishes a diverse and robust T-cell receptor repertoire (175). In adults, while peripheral expansion of existing T-cell clones predominates, the thymus remains essential for generating new T-cell receptor diversity, enhancing immune adaptability to novel pathogens, and even a subtle but prolonged thymic decline could potentially have significant consequences (193). For instance, a large study found that patients undergoing thymectomy as part of chest surgery had significantly reduced overall survival compared to those undergoing similar surgeries without thymectomy, underscoring critical role of the thymus in adult immunity (194). Therefore, to safeguard astronaut health in prolonged space exploration, it is imperative to prevent or mitigate spaceflight-induced thymic involution (52).
Despite the progress and advances using ground-based models to simulate spaceflight-induced thymic involution, critical questions still remain. How do specific thymic subsets, such as medullary thymic epithelial cells and early thymic progenitors, respond to prolonged low-dose, mixed-field galactic cosmic radiation (GCR)? What specific molecular pathways disrupted by GCR differentiate its effects from other forms of ionizing radiation, and which is the molecular basis for such differences? Moreover, the interplay between corticosterone-driven apoptosis and apoptosis-independent mechanisms affecting thymocytes and thymic architecture is still unclear. Moreover, the fidelity of these models raises questions: to what extent do fluid shifts and psychological stress in the HU model skew results away from microgravity’s true impact on the thymus? How can these variables be isolated? Future studies should integrate advanced molecular imaging and single-cell technologies, to gain an in-depth understanding of the mechanistic underpinnings behind spaceflight-induced thymic involution, and to support the development of rationalized countermeasures for astronaut health in long-term space missions.
Author contributions
WM: Writing – original draft, Writing – review & editing. MM: Writing – original draft, Writing – review & editing. TA: Writing – original draft, Writing – review & editing. GK: Conceptualization, Writing – original draft, Writing – review & editing.
Funding
The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This work is supported by the Montefiore-Einstein Comprehensive Cancer Center (GK, MM) and by CREST from the Japan Science and Technology Agency (JPMJCR2011 to TA).
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of this manuscript.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Demontis GC, Germani MM, Caiani EG, Barravecchia I, Passino C, Angeloni D. Human pathophysiological adaptations to the space environment. Front Physiol. (2017) 8:547. doi: 10.3389/fphys.2017.00547
2. 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:eaau8650. doi: 10.1126/science.aau8650
3. Lev MH. The long-term effects of spaceflight on human brain physiology. Radiology. (2020) 295:649–50. doi: 10.1148/radiol.2020201164
4. Shen M, Frishman WH. Effects of spaceflight on cardiovascular physiology and health. Cardiol Rev. (2019) 27:122–6. doi: 10.1097/CRD.0000000000000236
5. Ramachandran V, Wang R, Ramachandran SS, Ahmed AS, Phan K, Antonsen EL. Effects of spaceflight on cartilage: implications on spinal physiology. J Spine Surg. (2018) 4:433–45. doi: 10.21037/jss.2018.04.07
6. Trappe S. Effects of spaceflight, simulated spaceflight and countermeasures on single muscle fiber physiology. J Gravit Physiol. (2002) 9:P323–326.
7. Ronca AE, Alberts JR. Physiology of a microgravity environment selected contribution: effects of spaceflight during pregnancy on labor and birth at 1 G. J Appl Physiol. (1985) 89:849–54. doi: 10.1152/jappl.2000.89.2.849
8. Harris SA, Zhang M, Kidder LS, Evans GL, Spelsberg TC, Turner RT. Effects of orbital spaceflight on human osteoblastic cell physiology and gene expression. Bone. (2000) 26:325–31. doi: 10.1016/S8756-3282(00)00234-9
9. Dunn Rosenberg J, Jannasch A, Binsted K, Landry S. Biobehavioral and psychosocial stress changes during three 8-12 month spaceflight analog missions with Mars-like conditions of isolation and confinement. Front Physiol. (2022) 13:898841. doi: 10.3389/fphys.2022.898841
10. Schlaff CD, Helgeson MD, Wagner SC. Pathophysiologic spine adaptations and countermeasures for prolonged spaceflight. Clin Spine Surg. (2024) 37:43–8. doi: 10.1097/BSD.0000000000001488
11. Buoite Stella A, Ajcevic M, Furlanis G, Manganotti P. Neurophysiological adaptations to spaceflight and simulated microgravity. Clin Neurophysiol. (2021) 132:498–504. doi: 10.1016/j.clinph.2020.11.033
12. 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:e3000669. doi: 10.1371/journal.pbio.3000669
13. Cucinotta FA. Space radiation risks for astronauts on multiple International Space Station missions. PloS One. (2014) 9:e96099. doi: 10.1371/journal.pone.0096099
14. Juhl OJ, Buettmann EG, Friedman MA, DeNapoli RC, Hoppock GA, Donahue HJ. Update on the effects of microgravity on the musculoskeletal system. NPJ Microgravity. (2021) 7:28. doi: 10.1038/s41526-021-00158-4
15. Grimm D, Grosse J, Wehland M, Mann V, Reseland JE, Sundaresan A, et al. The impact of microgravity on bone in humans. Bone. (2016) 87:44–56. doi: 10.1016/j.bone.2015.12.057
16. Nagaraja MP, Risin D. The current state of bone loss research: data from spaceflight and microgravity simulators. J Cell Biochem. (2013) 114:1001–8. doi: 10.1002/jcb.24454
17. Smith SM, Heer M, Shackelford LC, Sibonga JD, Spatz J, Pietrzyk RA, et al. Bone metabolism and renal stone risk during International Space Station missions. Bone. (2015) 81:712–20. doi: 10.1016/j.bone.2015.10.002
18. Adamopoulos K, Koutsouris D, Zaravinos A, Lambrou GI. Gravitational influence on human living systems and the evolution of species on earth. Molecules. (2021) 26:2784. doi: 10.3390/molecules26092784
19. Dadwal UC, Maupin KA, Zamarioli A, Tucker A, Harris JS, Fischer JP, et al. The effects of spaceflight and fracture healing on distant skeletal sites. Sci Rep. (2019) 9:11419. doi: 10.1038/s41598-019-47695-3
20. Vico L, Hargens A. Skeletal changes during and after spaceflight. Nat Rev Rheumatol. (2018) 14:229–45. doi: 10.1038/nrrheum.2018.37
21. Yang J, Zhang G, Dong D, Shang P. Effects of iron overload and oxidative damage on the musculoskeletal system in the space environment: data from spaceflights and ground-based simulation models. Int J Mol Sci. (2018) 19:2608. doi: 10.3390/ijms19092608
22. Costa-Almeida R, Carvalho DTO, Ferreira MJS, Pesqueira T, Monici M, van Loon J, et al. Continuous exposure to simulated hypergravity-induced changes in proliferation, morphology, and gene expression of human tendon cells. Stem Cells Dev. (2018) 27:858–69. doi: 10.1089/scd.2017.0206
23. Kacena MA, Todd P, Gerstenfeld LC, Landis WJ. Experiments with osteoblasts cultured under hypergravity conditions. Microgravity Sci Technol. (2004) 15:28–34. doi: 10.1007/BF02870949
24. Ciofani G, Ricotti L, Rigosa J, Menciassi A, Mattoli V, Monici M. Hypergravity effects on myoblast proliferation and differentiation. J Biosci Bioeng. (2012) 113:258–61. doi: 10.1016/j.jbiosc.2011.09.025
25. 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. doi: 10.1038/s41526-020-0104-1
26. Monk TH, Buysse DJ, Billy BD, Kennedy KS, Willrich LM. Sleep and circadian rhythms in four orbiting astronauts. J Biol Rhythms. (1998) 13:188–201. doi: 10.1177/074873098129000039
27. Sulzman FM, Ferraro JS, Fuller CA, Moore-Ede MC, Klimovitsky V, Magedov V, et al. Thermoregulatory responses of rhesus monkeys during spaceflight. Physiol Behav. (1992) 51:585–91. doi: 10.1016/0031-9384(92)90184-4
28. Burgess HJ, Legasto CS, Fogg LF, Smith MR. Can small shifts in circadian phase affect performance? Appl Ergon. (2013) 44:109–11. doi: 10.1016/j.apergo.2012.05.007
29. Santy PA, Kapanka H, Davis JR, Stewart DF. Analysis of sleep on Shuttle missions. Aviat Space Environ Med. (1988) 59:1094–7.
30. Gundel A, Polyakov VV, Zulley J. The alteration of human sleep and circadian rhythms during spaceflight. J Sleep Res. (1997) 6:1–8. doi: 10.1046/j.1365-2869.1997.00028.x
31. Guo JH, Qu WM, Chen SG, Chen XP, Lv K, Huang ZL, et al. Keeping the right time in space: importance of circadian clock and sleep for physiology and performance of astronauts. Mil Med Res. (2014) 1:23. doi: 10.1186/2054-9369-1-23
32. Ince LM, Barnoud C, Lutes LK, Pick R, Wang C, Sinturel F, et al. Influence of circadian clocks on adaptive immunity and vaccination responses. Nat Commun. (2023) 14:476. doi: 10.1038/s41467-023-35979-2
33. Ding J, Chen P, Qi C. Circadian rhythm regulation in the immune system. Immunology. (2024) 171:525–33. doi: 10.1111/imm.13747
34. Wang C, Lutes LK, Barnoud C, Scheiermann C. The circadian immune system. Sci Immunol. (2022) 7:eabm2465. doi: 10.1126/sciimmunol.abm2465
35. Kolla BP, Auger RR. Jet lag and shift work sleep disorders: how to help reset the internal clock. Cleve Clin J Med. (2011) 78:675–84. doi: 10.3949/ccjm.78a.10083
36. Baron KG, Reid KJ. Circadian misalignment and health. Int Rev Psychiatry. (2014) 26:139–54. doi: 10.3109/09540261.2014.911149
37. Haus EL, Smolensky MH. Shift work and cancer risk: potential mechanistic roles of circadian disruption, light at night, and sleep deprivation. Sleep Med Rev. (2013) 17:273–84. doi: 10.1016/j.smrv.2012.08.003
38. Palinkas LA, Suedfeld P. Psychosocial issues in isolated and confined extreme environments. Neurosci Biobehav Rev. (2021) 126:413–29. doi: 10.1016/j.neubiorev.2021.03.032
39. Palinkas LA. Psychosocial issues in long-term space flight: overview. Gravit Space Biol Bull. (2001) 14:25–33.
40. Tafforin C. Confinement vs. isolation as analogue environments for Mars missions from a human ethology viewpoint. Aerosp Med Hum Perform. (2015) 86:131–5. doi: 10.3357/AMHP.4100.2015
41. Tafforin C. Time effects, cultural influences, and individual differences in crew behavior during the Mars-500 experiment. Aviat Space Environ Med. (2013) 84:1082–6. doi: 10.3357/asem.3692.2013
42. Oluwafemi FA, Abdelbaki R, Lai JC, Mora-Almanza JG, Afolayan EM. A review of astronaut mental health in manned missions: Potential interventions for cognitive and mental health challenges. Life Sci Space Res (Amst). (2021) 28:26–31. doi: 10.1016/j.lssr.2020.12.002
43. De la Torre GG, Groemer G, Diaz-Artiles A, Pattyn N, Van Cutsem J, Musilova M, et al. Space Analogs and Behavioral Health Performance Research review and recommendations checklist from ESA Topical Team. NPJ Microgravity. (2024) 10:98. doi: 10.1038/s41526-024-00437-w
44. Segerstrom SC, Miller GE. Psychological stress and the human immune system: a meta-analytic study of 30 years of inquiry. Psychol Bull. (2004) 130:601–30. doi: 10.1037/0033-2909.130.4.601
45. Tierney BT, Kim J, Overbey EG, Ryon KA, Foox J, Sierra MA, et al. Longitudinal multi-omics analysis of host microbiome architecture and immune responses during short-term spaceflight. Nat Microbiol. (2024) 9:1661–75. doi: 10.1038/s41564-024-01635-8
46. Kim J, Tierney BT, Overbey EG, Dantas E, Fuentealba M, Park J, et al. Single-cell multi-ome and immune profiles of the Inspiration4 crew reveal conserved, cell-type, and sex-specific responses to spaceflight. Nat Commun. (2024) 15:4954. doi: 10.1038/s41467-024-49211-2
47. Wu F, Du H, Overbey E, Kim J, Makhijani P, Martin N, et al. Single-cell analysis identifies conserved features of immune dysfunction in simulated microgravity and spaceflight. Nat Commun. (2024) 15:4795. doi: 10.1038/s41467-023-42013-y
48. Garcia-Medina JS, Sienkiewicz K, Narayanan SA, Overbey EG, Grigorev K, Ryon KA, et al. Genome and clonal hematopoiesis stability contrasts with immune, cfDNA, mitochondrial, and telomere length changes during short duration spaceflight. Precis Clin Med. (2024) 7:pbae007. doi: 10.1093/pcmedi/pbae007
49. Tierney BT, Kim J, Overbey EG, Ryon KA, Foox J, Sierra M, et al. Viral activation and ecological restructuring characterize a microbiome axis of spaceflight-associated immune activation. Res Sq. (2023), rs.3.rs–2493867. doi: 10.21203/rs.3.rs-2493867/v1
50. 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) 23:101747. doi: 10.1016/j.isci.2020.101747
51. Smith JK. IL-6 and the dysregulation of immune, bone, muscle, and metabolic homeostasis during spaceflight. NPJ Microgravity. (2018) 4:24. doi: 10.1038/s41526-018-0057-9
52. Crucian BE, Chouker 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. doi: 10.3389/fimmu.2018.01437
53. Crucian B, Simpson RJ, Mehta S, Stowe R, Chouker A, Hwang SA, et al. Terrestrial stress analogs for spaceflight associated immune system dysregulation. Brain Behav Immun. (2014) 39:23–32. doi: 10.1016/j.bbi.2014.01.011
54. Stowe RP, Sams CF, Pierson DL. Adrenocortical and immune responses following short- and long-duration spaceflight. Aviat Space Environ Med. (2011) 82:627–34. doi: 10.3357/asem.2980.2011
55. Crucian B, Sams C. Immune system dysregulation during spaceflight: clinical risk for exploration-class missions. J Leukoc Biol. (2009) 86:1017–8. doi: 10.1189/jlb.0709500
56. Gueguinou 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) 86:1027–38. doi: 10.1189/jlb.0309167
57. Baqai FP, Gridley DS, Slater JM, Luo-Owen X, Stodieck LS, Ferguson V, et al. Effects of spaceflight on innate immune function and antioxidant gene expression. J Appl Physiol (1985). (2009) 106:1935–42. doi: 10.1152/japplphysiol.91361.2008
58. Crucian BE, Stowe RP, Pierson DL, Sams CF. Immune system dysregulation following short- vs long-duration spaceflight. Aviat Space Environ Med. (2008) 79:835–43. doi: 10.3357/asem.2276.2008
59. Ostler JB, Sawant L, Harrison K, Jones C. Regulation of neurotropic herpesvirus productive infection and latency-reactivation cycle by glucocorticoid receptor and stress-induced transcription factors. Vitam Horm. (2021) 117:101–32. doi: 10.1016/bs.vh.2021.06.005
61. Mehta SK, Szpara ML, Rooney BV, Diak DM, Shipley MM, Renner DW, et al. Dermatitis during spaceflight associated with HSV-1 reactivation. Viruses. (2022) 14:789. doi: 10.3390/v14040789
62. Kunz HE, Makedonas G, Mehta SK, Tyring SK, Vangipuram R, Quiriarte H, et al. Zoster patients on earth and astronauts in space share similar immunologic profiles. Life Sci Space Res (Amst). (2020) 25:119–28. doi: 10.1016/j.lssr.2019.10.001
63. Rooney BV, Crucian BE, Pierson DL, Laudenslager ML, Mehta SK. Herpes virus reactivation in astronauts during spaceflight and its application on earth. Front Microbiol. (2019) 10:16. doi: 10.3389/fmicb.2019.00016
64. Mehta SK, Suresh R, Brandt K, Diak DM, Smith SM, Zwart SR, et al. Immune system dysregulation preceding a case of laboratory-confirmed zoster/dermatitis on board the International Space Station. J Allergy Clin Immunol Glob. (2024) 3:100244. doi: 10.1016/j.jacig.2024.100244
65. Voorhies AA, Mark Ott C, Mehta S, Pierson DL, Crucian BE, Feiveson A, et al. Study of the impact of long-duration space missions at the International Space Station on the astronaut microbiome. Sci Rep. (2019) 9:9911. doi: 10.1038/s41598-019-46303-8
66. Cohrs RJ, Mehta SK, Schmid DS, Gilden DH, Pierson DL. Asymptomatic reactivation and shed of infectious varicella zoster virus in astronauts. J Med Virol. (2008) 80:1116–22. doi: 10.1002/jmv.21173
67. 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) 72:174–9. doi: 10.1002/jmv.10555
68. 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) 34:778–86. doi: 10.1089/jir.2013.0129
69. 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. doi: 10.3389/fimmu.2021.725748
70. Martinez RJ, Hogquist KA. The role of interferon in the thymus. Curr Opin Immunol. (2023) 84:102389. doi: 10.1016/j.coi.2023.102389
71. Akiyama T, Shimo Y, Yanai H, Qin J, Ohshima D, Maruyama Y, et al. The tumor necrosis factor family receptors RANK and CD40 cooperatively establish the thymic medullary microenvironment and self-tolerance. Immunity. (2008) 29:423–37. doi: 10.1016/j.immuni.2008.06.015
72. Chatzidakis I, Mamalaki C. T cells as sources and targets of TNF: implications for immunity and autoimmunity. Curr Dir Autoimmun. (2010) 11:105–18. doi: 10.1159/000289200
73. Sonnenfeld G. Use of animal models for space flight physiology studies, with special focus on the immune system. Gravit Space Biol Bull. (2005) 18:31–5.
74. Pecaut MJ, Nelson GA, Peters LL, Kostenuik PJ, Bateman TA, Morony S, et al. Genetic models in applied physiology: selected contribution: effects of spaceflight on immunity in the C57BL/6 mouse. I. Immune population distributions. J Appl Physiol (1985). (2003) 94:2085–94. doi: 10.1152/japplphysiol.01052.2002
75. Gridley DS, Nelson GA, Peters LL, Kostenuik PJ, Bateman TA, Morony S, et al. Genetic models in applied physiology: selected contribution: effects of spaceflight on immunity in the C57BL/6 mouse. II. Activation, cytokines, erythrocytes, and platelets. J Appl Physiol (1985). (2003) 94:2095–103. doi: 10.1152/japplphysiol.01053.2002
76. Meyers VE, Zayzafoon M, Gonda SR, Gathings WE, McDonald JM. Modeled microgravity disrupts collagen I/integrin signaling during osteoblastic differentiation of human mesenchymal stem cells. J Cell Biochem. (2004) 93:697–707. doi: 10.1002/jcb.20229
77. Chen Z, Luo Q, Lin C, Kuang D, Song G. Simulated microgravity inhibits osteogenic differentiation of mesenchymal stem cells via depolymerizing F-actin to impede TAZ nuclear translocation. Sci Rep. (2016) 6:30322. doi: 10.1038/srep30322
78. Kernagis DN, Balcer-Kubiczek E, Bazyar S, Orschell CM, Jackson IL. Medical countermeasures for the hematopoietic-subsyndrome of acute radiation syndrome in space. Life Sci Space Res (Amst). (2022) 35:36–43. doi: 10.1016/j.lssr.2022.06.002
79. Sarkar R, Pampaloni F. In vitro models of bone marrow remodelling and immune dysfunction in space: present state and future directions. Biomedicines. (2022) 10:766. doi: 10.3390/biomedicines10040766
80. Ozcivici E. Effects of spaceflight on cells of bone marrow origin. Turk J Haematol. (2013) 30:1–7. doi: 10.4274/tjh.2012.0127
81. Rayman RB. Essential thrombocythemia: aeromedical considerations. Aviat Space Environ Med. (2009) 80:968–70. doi: 10.3357/asem.2476.2009
82. Benjamin CL, Stowe RP, St John L, Sams CF, Mehta SK, Crucian BE, et al. Decreases in thymopoiesis of astronauts returning from space flight. JCI Insight. (2016) 1:e88787. doi: 10.1172/jci.insight.88787
83. Aspinall R, Pido J, Andrew D. A simple method for the measurement of sjTREC levels in blood. Mech Ageing Dev. (2000) 121:59–67. doi: 10.1016/s0047-6374(00)00197-4
84. Ou X, Zhao H, Sun H, Yang Z, Xie B, Shi Y, et al. Detection and quantification of the age-related sjTREC decline in human peripheral blood. Int J Legal Med. (2011) 125:603–8. doi: 10.1007/s00414-010-0528-3
85. Xu Y, Xu L, Chen C, Zhang Y, Zeng C, Jin Z, et al. Age-related immune profile of the T cell receptor repertoire, thymic recent output function, and miRNAs. BioMed Res Int. (2020) 2020:5910823. doi: 10.1155/2020/5910823
86. Horie K, Kato T, Kudo T, Sasanuma H, Miyauchi M, Akiyama N, et al. Impact of spaceflight on the murine thymus and mitigation by exposure to artificial gravity during spaceflight. Sci Rep. (2019) 9:19866. doi: 10.1038/s41598-019-56432-9
87. 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:e75097. doi: 10.1371/journal.pone.0075097
88. Kalandarova MP, Verigo VV, Podlyzhnaya GN, Rodina GP, Serova LV, Chelnaya NA. Effect of irradiation in the space environment on the blood-forming system in rats. Life Sci Space Res. (1976) 14:179–83.
89. Klassen NV, Walker PR, Ross CK, Cygler J, Lach B. Two-stage cell shrinkage and the OER for radiation-induced apoptosis of rat thymocytes. Int J Radiat Biol. (1993) 64:571–81. doi: 10.1080/09553009314551791
90. Kutsyi MP, Kuznetsova EA, Gluiaeva NA, Gaziev AI. Effect of gamma-radiation and mitochondrial apoptogenic factors on nuclear protease activity. Radiats Biol Radioecol. (2002) 42:357–63.
91. Portugalov VV, Savina EA, Kaplansky AS, Yakovleva VI, Durnova GN, Pankova AS, et al. Discussion of the combined effect of weightlessness and ionizing radiation on the mammalian body: morphological data. Aviat Space Environ Med. (1977) 48:33–6.
92. Pecaut MJ, Dutta-Roy R, Smith AL, Jones TA, Nelson GA, Gridley DS. Acute effects of iron-particle radiation on immunity. Part I: Population distributions. Radiat Res. (2006) 165:68–77. doi: 10.1667/rr3493.1
93. Xie Y, Zhang H, Wang YL, Zhou QM, Qiu R, Yuan ZG, et al. Alterations of immune functions induced by 12C6+ ion irradiation in mice. Int J Radiat Biol. (2007) 83:577–81. doi: 10.1080/09553000701481774
94. Kramer R, Cassola VF, Khoury HJ, Vieira JW, Lima VJ, Brown KR. FASH and MASH: female and male adult human phantoms based on polygon mesh surfaces: II. Dosimetric calculations. Phys Med Biol. (2010) 55:163–89. doi: 10.1088/0031-9155/55/1/010
95. Kramer R, Khoury HJ, Vieira JW, Lima VJ. MAX06 and FAX06: update of two adult human phantoms for radiation protection dosimetry. Phys Med Biol. (2006) 51:3331–46. doi: 10.1088/0031-9155/51/14/003
96. Simonsen LC, Nealy JE, Townsend LW, Wilson JW. Space radiation dose estimates on the surface of Mars. J Spacecr Rockets. (1990) 27:353–4. doi: 10.2514/3.26149
97. Matthia D, Hassler DM, de Wet W, Ehresmann B, Firan A, Flores-McLaughlin J, et al. The radiation environment on the surface of Mars - Summary of model calculations and comparison to RAD data. Life Sci Space Res (Amst). (2017) 14:18–28. doi: 10.1016/j.lssr.2017.06.003
98. Slaba TC, Bahadori AA, Reddell BD, Singleterry RC, Clowdsley MS, Blattnig SR. Optimal shielding thickness for galactic cosmic ray environments. Life Sci Space Res (Amst). (2017) 12:1–15. doi: 10.1016/j.lssr.2016.12.003
99. Norbury JW, Slaba TC, Aghara S, Badavi FF, Blattnig SR, Clowdsley MS, et al. Advances in space radiation physics and transport at NASA. Life Sci Space Res (Amst). (2019) 22:98–124. doi: 10.1016/j.lssr.2019.07.003
100. Slaba TC, Blattnig SR, Norbury JW, Rusek A, La Tessa C. Reference field specification and preliminary beam selection strategy for accelerator-based GCR simulation. Life Sci Space Res (Amst). (2016) 8:52–67. doi: 10.1016/j.lssr.2016.01.001
101. Townsend LW, Adams JH, Blattnig SR, Clowdsley MS, Fry DJ, Jun I, et al. Solar particle event storm shelter requirements for missions beyond low Earth orbit. Life Sci Space Res (Amst). (2018) 17:32–9. doi: 10.1016/j.lssr.2018.02.002
102. Norbury JW, Schimmerling W, Slaba TC, Azzam EI, Badavi FF, Baiocco G, et al. Galactic cosmic ray simulation at the NASA Space Radiation Laboratory. Life Sci Space Res (Amst). (2016) 8:38–51. doi: 10.1016/j.lssr.2016.02.001
103. Suman S, Kumar S, Kallakury BVS, Moon BH, Angdisen J, Datta K, et al. Predominant contribution of the dose received from constituent heavy-ions in the induction of gastrointestinal tumorigenesis after simulated space radiation exposure. Radiat Environ Biophys. (2022) 61:631–7. doi: 10.1007/s00411-022-00997-z
104. Diaz J, Kuhlman BM, Edenhoffer NP, Evans AC, Martin KA, Guida P, et al. Immediate effects of acute Mars mission equivalent doses of SEP and GCR radiation on the murine gastrointestinal system-protective effects of curcumin-loaded nanolipoprotein particles (cNLPs). Front Astron Space Sci. (2023) 10:1117811. doi: 10.3389/fspas.2023.1117811
105. Yun S, Kiffer FC, Bancroft GL, Guzman CS, Soler I, Haas HA, et al. The longitudinal behavioral effects of acute exposure to galactic cosmic radiation in female C57BL/6J mice: Implications for deep space missions, female crews, and potential antioxidant countermeasures. J Neurochem. (2024) 169(1):e16225. doi: 10.1111/jnc.16225
106. Lenarczyk M, Kronenberg A, Mader M, Komorowski R, Hopewell JW, Baker JE. Exposure to multiple ion beams, broadly representative of galactic cosmic rays, causes perivascular cardiac fibrosis in mature male rats. PloS One. (2023) 18:e0283877. doi: 10.1371/journal.pone.0283877
107. 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. doi: 10.3389/fpubh.2023.1063250
108. Kleiman NJ, Edmondson EF, Weil MM, Fallgren CM, King A, Schmidt C, et al. Radiation cataract in Heterogeneous Stock mice after gamma-ray or HZE ion exposure. Life Sci Space Res (Amst). (2024) 40:97–105. doi: 10.1016/j.lssr.2023.09.004
109. Burke M, Wong K, Talyansky Y, Mhatre SD, Mitchell C, Juran CM, et al. Sexual dimorphism during integrative endocrine and immune responses to ionizing radiation in mice. Sci Rep. (2024) 14:7334. doi: 10.1038/s41598-023-33629-7
110. Almeida-Porada G, Rodman C, Kuhlman B, Brudvik E, Moon J, George S, et al. Exposure of the bone marrow microenvironment to simulated solar and galactic cosmic radiation induces biological bystander effects on human hematopoiesis. Stem Cells Dev. (2018) 27:1237–56. doi: 10.1089/scd.2018.0005
111. 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) 192:241–50. doi: 10.1667/RR15219.1
112. Kim HN, Richardson KK, Krager KJ, Ling W, Simmons P, Allen AR, et al. Simulated galactic cosmic rays modify mitochondrial metabolism in osteoclasts, increase osteoclastogenesis and cause trabecular bone loss in mice. Int J Mol Sci. (2021) 22:11711. doi: 10.3390/ijms222111711
113. Nelson GA. Space radiation and human exposures, A primer. Radiat Res. (2016) 185:349–58. doi: 10.1667/RR14311.1
114. Kunisaki Y, Bruns I, Scheiermann C, Ahmed J, Pinho S, Zhang D, et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature. (2013) 502:637–43. doi: 10.1038/nature12612
115. Lawson MA, McDonald MM, Kovacic N, Hua Khoo W, Terry RL, Down J, et al. Osteoclasts control reactivation of dormant myeloma cells by remodelling the endosteal niche. Nat Commun. (2015) 6:8983. doi: 10.1038/ncomms9983
116. Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity. (2006) 25:977–88. doi: 10.1016/j.immuni.2006.10.016
117. Rodman C, Almeida-Porada G, George SK, Moon J, Soker S, Pardee T, et al. In vitro and in vivo assessment of direct effects of simulated solar and galactic cosmic radiation on human hematopoietic stem/progenitor cells. Leukemia. (2017) 31:1398–407. doi: 10.1038/leu.2016.344
118. Han Y, Shi S, Liu S, Gu X. Effects of spaceflight on the spleen and thymus of mice: Gene pathway analysis and immune infiltration analysis. Math Biosci Eng. (2023) 20:8531–45. doi: 10.3934/mbe.2023374
119. Shelhamer M. Parabolic flight as a spaceflight analog. J Appl Physiol (1985). (2016) 120:1442–8. doi: 10.1152/japplphysiol.01046.2015
120. Jenkinson EJ, Anderson G. Fetal thymic organ cultures. Curr Opin Immunol. (1994) 6:293–7. doi: 10.1016/0952-7915(94)90104-x
121. Woods CC, Banks KE, Gruener R, DeLuca D. Loss of T cell precursors after spaceflight and exposure to vector-averaged gravity. FASEB J. (2003) 17:1526–8. doi: 10.1096/fj.02-0749fje
122. Globus RK, Morey-Holton E. Hindlimb unloading: rodent analog for microgravity. J Appl Physiol (1985). (2016) 120:1196–206. doi: 10.1152/japplphysiol.00997.2015
123. Wei LX, Zhou JN, Roberts AI, Shi YF. Lymphocyte reduction induced by hindlimb unloading: distinct mechanisms in the spleen and thymus. Cell Res. (2003) 13:465–71. doi: 10.1038/sj.cr.7290189
124. Wang KX, Shi Y, Denhardt DT. Osteopontin regulates hindlimb-unloading-induced lymphoid organ atrophy and weight loss by modulating corticosteroid production. Proc Natl Acad Sci U.S.A. (2007) 104:14777–82. doi: 10.1073/pnas.0703236104
125. Wang KX, Shi YF, Ron Y, Kazanecki CC, Denhardt DT. Plasma osteopontin modulates chronic restraint stress-induced thymus atrophy by regulating stress hormones: inhibition by an anti-osteopontin monoclonal antibody. J Immunol. (2009) 182:2485–91. doi: 10.4049/jimmunol.0803023
126. 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) 501:745–50. doi: 10.1016/j.bbrc.2018.05.060
127. Castagne V, Moser P, Roux S, Porsolt RD. Rodent models of depression: forced swim and tail suspension behavioral despair tests in rats and mice. Curr Protoc Neurosci. (2011) Chapter 8:Unit 8.10A. doi: 10.1002/0471142301.ns0810as55. Chapter 8, Unit 8 10A.
129. Buijs FN, León-Mercado L, Guzmán-Ruiz M, Guerrero-Vargas NN, Romo-Nava F, Buijs RM. The circadian system: A regulatory feedback network of periphery and brain. Physiology. (2016) 31:170–81. doi: 10.1152/physiol.00037.2015
130. Dumbell R, Matveeva O, Oster H. Circadian clocks, stress, and immunity. Front Endocrinol. (2016) 7. doi: 10.3389/fendo.2016.00037
131. Thaiss CA, Levy M, Korem T, Dohnalová L, Shapiro H, Jaitin DA, et al. Microbiota diurnal rhythmicity programs host transcriptome oscillations. Cell. (2016) 167:1495–1510.e1412. doi: 10.1016/j.cell.2016.11.003
132. Barger LK, Flynn-Evans EE, Kubey A, Walsh L, Ronda JM, Wang W, et al. Prevalence of sleep deficiency and use of hypnotic drugs in astronauts before, during, and after spaceflight: an observational study. Lancet Neurol. (2014) 13:904–12. doi: 10.1016/S1474-4422(14)70122-X
133. Dijk D-J, Neri DF, Wyatt JK, Ronda JM, Riel E, Ritz-De Cecco A, et al. Sleep, performance, circadian rhythms, and light-dark cycles during two space shuttle flights. Am J Physiology-Regulatory Integr Comp Physiol. (2001) 281:R1647–64. doi: 10.1152/ajpregu.2001.281.5.R1647
134. Flynn-Evans EE, Barger LK, Kubey AA, Sullivan JP, Czeisler CA. Circadian misalignment affects sleep and medication use before and during spaceflight. NPJ Microgravity. (2016) 2:15019. doi: 10.1038/npjmgrav.2015.19
135. Koller DP, Kasanin V, Flynn-Evans EE, Sullivan JP, Dijk D-J, Czeisler CA, et al. Altered sleep spindles and slow waves during space shuttle missions. NPJ Microgravity. (2021) 7:48. doi: 10.1038/s41526-021-00177-1
136. Smolensky MH, Portaluppi F, Manfredini R, Hermida RC, Tiseo R, Sackett-Lundeen LL, et al. Diurnal and twenty-four hour patterning of human diseases: Cardiac, vascular, and respiratory diseases, conditions, and syndromes. Sleep Med Rev. (2015) 21:3–11. doi: 10.1016/j.smrv.2014.07.001
137. Zimmet P, Alberti K, Stern N, Bilu C, El-Osta A, Einat H, et al. The Circadian Syndrome: is the Metabolic Syndrome and much more! J Intern Med. (2019) 286:181–91. doi: 10.1111/joim.12924
138. Kecklund G, Axelsson J. Health consequences of shift work and insufficient sleep. BMJ. (2016) 355:i5210. doi: 10.1136/bmj.i5210
139. Kwon P, Lundin J, Li W, Ray R, Littell C, Gao D, et al. Night shift work and lung cancer risk among female textile workers in Shanghai, China. J Occup Environ Hyg. (2015) 12:334–41. doi: 10.1080/15459624.2014.993472
140. Schernhammer ES, Feskanich D, Liang G, Han J. Rotating night-shift work and lung cancer risk among female nurses in the United States. Am J Epidemiol. (2013) 178:1434–41. doi: 10.1093/aje/kwt155
141. Wu B, Wang Y, Wu X, Liu D, Xu D, Wang F. On-orbit sleep problems of astronauts and countermeasures. Military Med Res. (2018) 5:17. doi: 10.1186/s40779-018-0165-6
142. Yin Y, Liu J, Fan Q, Zhao S, Wu X, Wang J, et al. Long-term spaceflight composite stress induces depression and cognitive impairment in astronauts-insights from neuroplasticity. Transl Psychiatry. (2023) 13:342. doi: 10.1038/s41398-023-02638-5
143. Méndez-Ferrer S, Lucas D, Battista M, Frenette PS. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. (2008) 452:442–7. doi: 10.1038/nature06685
144. Scheiermann C, Kunisaki Y, Lucas D, Chow A, Jang J-E, Zhang D, et al. Adrenergic nerves govern circadian leukocyte recruitment to tissues. Immunity. (2012) 37:290–301. doi: 10.1016/j.immuni.2012.05.021
145. Scheiermann C, Gibbs J, Ince L, Loudon A. Clocking in to immunity. Nat Rev Immunol. (2018) 18:423–37. doi: 10.1038/s41577-018-0008-4
146. He W, Holtkamp S, Hergenhan SM, Kraus K, de Juan A, Weber J, et al. Circadian expression of migratory factors establishes lineage-specific signatures that guide the homing of leukocyte subsets to tissues. Immunity. (2018) 49:1175–1190.e1177. doi: 10.1016/j.immuni.2018.10.007
147. Born J, Lange T, Hansen K, Mölle M, Fehm HL. Effects of sleep and circadian rhythm on human circulating immune cells. J Immunol. (1997) 158:4454–64. doi: 10.4049/jimmunol.158.9.4454
148. Lucas D, Battista M, Shi PA, Isola L, Frenette PS. Mobilized hematopoietic stem cell yield depends on species-specific circadian timing. Cell Stem Cell. (2008) 3:364–6. doi: 10.1016/j.stem.2008.09.004
149. Besedovsky L, Born J, Lange T. Endogenous glucocorticoid receptor signaling drives rhythmic changes in human T-cell subset numbers and the expression of the chemokine receptor CXCR4. FASEB J. (2014) 28:67–75. doi: 10.1096/fj.13-237958
150. Dimitrov S, Benedict C, Heutling D, Westermann J, Born J, Lange T. Cortisol and epinephrine control opposing circadian rhythms in T cell subsets. Blood. (2009) 113:5134–43. doi: 10.1182/blood-2008-11-190769
151. Zhao Y, Liu M, Chan XY, Tan SY, Subramaniam S, Fan Y, et al. Uncovering the mystery of opposite circadian rhythms between mouse and human leukocytes in humanized mice. Blood. (2017) 130:1995–2005. doi: 10.1182/blood-2017-04-778779
152. Haspel JA, Anafi R, Brown MK, Cermakian N, Depner C, Desplats P, et al. Perfect timing: circadian rhythms, sleep, and immunity — an NIH workshop summary. JCI Insight. (2020) 5:e131487. doi: 10.1172/jci.insight.131487
153. Hemmers S, Rudensky AY. The cell-intrinsic circadian clock is dispensable for lymphocyte differentiation and function. Cell Rep. (2015) 11:1339–49. doi: 10.1016/j.celrep.2015.04.058
154. Minaduola M, Aili A, Bao Y, Peng Z, Ge Q, Jin R. The circadian clock sets a spatial–temporal window for recent thymic emigrants. Immunol Cell Biol. (2022) 100:731–41. doi: 10.1111/imcb.12582
155. Chen L, Zhang B, Yang L, Bai Y-G, Song J-B, Ge Y-L, et al. BMAL1 disrupted intrinsic diurnal oscillation in rat cerebrovascular contractility of simulated microgravity rats by altering circadian regulation of miR-103/CaV1.2 signal pathway. Int J Mol Sci. (2019) 20:3947. doi: 10.3390/ijms20163947
156. Ranieri D, Cucina A, Bizzarri M, Alimandi M, Torrisi MR. Microgravity influences circadian clock oscillation in human keratinocytes. FEBS Open Bio. (2015) 5:717–23. doi: 10.1016/j.fob.2015.08.012
157. Ranieri D, Proietti S, Dinicola S, Masiello MG, Rosato B, Ricci G, et al. Simulated microgravity triggers epithelial mesenchymal transition in human keratinocytes. Sci Rep. (2017) 7:538. doi: 10.1038/s41598-017-00602-0
158. Yang S, Liu Y, Yang Y, Yang Z, Cheng S, Hou W, et al. Simulated microgravity influences circadian rhythm of NIH3T3 cells. Biol Rhythm Res. (2016) 47:897–907. doi: 10.1080/09291016.2016.1207391
159. Barroca NCB, Della Santa G, Suchecki D, Garcia-Cairasco N, Umeoka EHL. Challenges in the use of animal models and perspectives for a translational view of stress and psychopathologies. Neurosci Biobehav Rev. (2022) 140:104771. doi: 10.1016/j.neubiorev.2022.104771
160. Cryan JF, Sweeney FF. The age of anxiety: role of animal models of anxiolytic action in drug discovery. Br J Pharmacol. (2011) 164:1129–61. doi: 10.1111/j.1476-5381.2011.01362.x
161. Krishnan V, Nestler EJ. Animal models of depression: molecular perspectives. Curr Top Behav Neurosci. (2011) 7:121–47. doi: 10.1007/7854_2010_108
162. Harro J. Animal models of depression: pros and cons. Cell Tissue Res. (2019) 377:5–20. doi: 10.1007/s00441-018-2973-0
163. Petkovic A, Chaudhury D. Encore: Behavioural animal models of stress, depression and mood disorders. Front Behav Neurosci. (2022) 16:931964. doi: 10.3389/fnbeh.2022.931964
164. Tanaka M, Szabo A, Vecsei L. Preclinical modeling in depression and anxiety: Current challenges and future research directions. Adv Clin Exp Med. (2023) 32:505–9. doi: 10.17219/acem/165944
165. Gururajan A, Reif A, Cryan JF, Slattery DA. The future of rodent models in depression research. Nat Rev Neurosci. (2019) 20:686–701. doi: 10.1038/s41583-019-0221-6
166. Yin YS, Zhu YB, Liu JL, Fan QC, Wu XR, Zhao S, et al. Long-term spaceflight composite stress induces depressive behaviors in model rats through disrupting hippocampus synaptic plasticity. CNS Neurosci Ther. (2024) 30:e14438. doi: 10.1111/cns.14438
167. Wu X, Yin Y, Liu J, Zhu Y, Fan Q, Zhao S, et al. Baoyuan jieyu formula ameliorates depression-like behaviour in rats induced by simulated long-term spaceflight composite stress through regulating MAPK and BDNF pathways. Life Sci Space Res (Amst). (2021) 31:34–42. doi: 10.1016/j.lssr.2021.06.001
168. Yin Y, Wu X, Zhu Y, Liu J, Fan Q, Zhao S, et al. Protective effect of Baoyuan Jieyu formula on long-term spaceflight composite stress-induced depressive-like behavior and memory deficits through regulation of Ca(2+) channel currents. Life Sci Space Res (Amst). (2024) 40:135–42. doi: 10.1016/j.lssr.2023.07.002
169. Tubbs JD, Ding J, Baum L, Sham PC. Immune dysregulation in depression: Evidence from genome-wide association. Brain Behav Immun Health. (2020) 7:100108. doi: 10.1016/j.bbih.2020.100108
170. Wadhwa A, Moreno-Villanueva M, Crucian B, Wu H. Synergistic interplay between radiation and microgravity in spaceflight-related immunological health risks. Immun Ageing. (2024) 21:50. doi: 10.1186/s12979-024-00449-w
171. Ansari AR, Liu H. Acute thymic involution and mechanisms for recovery. Arch Immunol Ther Exp (Warsz). (2017) 65:401–20. doi: 10.1007/s00005-017-0462-x
172. Calder AE, Hince MN, Dudakov JA, Chidgey AP, Boyd RL. Thymic involution: where endocrinology meets immunology. Neuroimmunomodulation. (2011) 18:281–9. doi: 10.1159/000329496
173. Gulla S, Reddy MC, Reddy VC, Chitta S, Bhanoori M, Lomada D. Role of thymus in health and disease. Int Rev Immunol. (2023) 42:347–63. doi: 10.1080/08830185.2022.2064461
174. Kinsella S, Dudakov JA. When the damage is done: injury and repair in thymus function. Front Immunol. (2020) 11:1745. doi: 10.3389/fimmu.2020.01745
175. Lagou MK, Anastasiadou DP, Karagiannis GS. A proposed link between acute thymic involution and late adverse effects of chemotherapy. Front Immunol. (2022) 13:933547. doi: 10.3389/fimmu.2022.933547
176. Lagou MK, Karagiannis GS. Obesity-induced thymic involution and cancer risk. Semin Cancer Biol. (2023) 93:3–19. doi: 10.1016/j.semcancer.2023.04.008
177. Lynch HE, Goldberg GL, Chidgey A, Van den Brink MR, Boyd R, Sempowski GD. Thymic involution and immune reconstitution. Trends Immunol. (2009) 30:366–73. doi: 10.1016/j.it.2009.04.003
178. Abramson J, Anderson G. Thymic epithelial cells. Annu Rev Immunol. (2017) 35:85–118. doi: 10.1146/annurev-immunol-051116-052320
179. Alawam AS, Anderson G, Lucas B. Generation and regeneration of thymic epithelial cells. Front Immunol. (2020) 11:858. doi: 10.3389/fimmu.2020.00858
180. Anderson G, Takahama Y. Thymic epithelial cells: working class heroes for T cell development and repertoire selection. Trends Immunol. (2012) 33:256–63. doi: 10.1016/j.it.2012.03.005
181. Bhalla P, Su DM, van Oers NSC. Thymus functionality needs more than a few TECs. Front Immunol. (2022) 13:864777. doi: 10.3389/fimmu.2022.864777
182. Lepletier A, Chidgey AP, Savino W. Perspectives for improvement of the thymic microenvironment through manipulation of thymic epithelial cells: A mini-review. Gerontology. (2015) 61:504–14. doi: 10.1159/000375160
183. Manley NR, Richie ER, Blackburn CC, Condie BG, Sage J. Structure and function of the thymic microenvironment. Front Biosci (Landmark Ed). (2011) 16:2461–77. doi: 10.2741/3866
184. Nitta T, Murata S, Ueno T, Tanaka K, Takahama Y. Thymic microenvironments for T-cell repertoire formation. Adv Immunol. (2008) 99:59–94. doi: 10.1016/S0065-2776(08)00603-2
185. Palmer S, Albergante L, Blackburn CC, Newman TJ. Thymic involution and rising disease incidence with age. Proc Natl Acad Sci U.S.A. (2018) 115:1883–8. doi: 10.1073/pnas.1714478115
186. Rose NR. Thymus function, ageing and autoimmunity. Immunol Lett. (1994) 40:225–30. doi: 10.1016/0165-2478(94)00060-3
187. Park JE, Botting RA, Dominguez Conde C, Popescu DM, Lavaert M, Kunz DJ, et al. A cell atlas of human thymic development defines T cell repertoire formation. Science. (2020) 367:eaay3224. doi: 10.1126/science.aay3224
188. Chopp L, Redmond C, O'Shea JJ, Schwartz DM. From thymus to tissues and tumors: a review of T cell biology. J Allergy Clin Immunol. (2022) 151(1):81–97. doi: 10.1016/j.jaci.2022.10.011
189. Wang W, Thomas R, Sizova O, Su DM. Thymic function associated with cancer development, relapse, and antitumor immunity - A mini-review. Front Immunol. (2020) 11:773. doi: 10.3389/fimmu.2020.00773
190. Hamm PB, Billica RD, Johnson GS, Wear ML, Pool SL. Risk of cancer mortality among the Longitudinal Study of Astronaut Health (LSAH) participants. Aviat Space Environ Med. (1998) 69:142–4.
191. Hamm PB, Nicogossian AE, Pool SL, Wear ML, Billica RD. Design and current status of the longitudinal study of astronaut health. Aviat Space Environ Med. (2000) 71:564–70.
192. Peterson LE, Pepper LJ, Hamm PB, Gilbert SL. Longitudinal study of astronaut health: mortality in the years 1959-1991. Radiat Res. (1993) 133:257–64. doi: 10.2307/3578364
193. Palmer DB. The effect of age on thymic function. Front Immunol. (2013) 4:316. doi: 10.3389/fimmu.2013.00316
Keywords: thymus, spaceflight, involution, cosmic radiation, microgravity (μg), circadian rhythms, psychosocial stress
Citation: Muramatsu W, Maryanovich M, Akiyama T and Karagiannis GS (2025) Thymus ad astra, or spaceflight-induced thymic involution. Front. Immunol. 15:1534444. doi: 10.3389/fimmu.2024.1534444
Received: 25 November 2024; Accepted: 24 December 2024;
Published: 24 January 2025.
Edited by:
Miho Shinzawa, National Institutes of Health (NIH), United StatesReviewed by:
Krisztian Kvell, University of Pécs, HungaryCopyright © 2025 Muramatsu, Maryanovich, Akiyama and Karagiannis. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: George S. Karagiannis, Z2Vvcmdpb3Mua2FyYWdpYW5uaXNAZWluc3RlaW5tZWQuZWR1