Abstract
Both microgravity and radiation exposure in the spaceflight environment have been identified as hazards to astronaut health and performance. Substantial study has been focused on understanding the biology and risks associated with prolonged exposure to microgravity, and the hazards presented by radiation from galactic cosmic rays (GCR) and solar particle events (SPEs) outside of low earth orbit (LEO). To date, the majority of the ground-based analogues (e.g., rodent or cell culture studies) that investigate the biology of and risks associated with spaceflight hazards will focus on an individual hazard in isolation. However, astronauts will face these challenges simultaneously Combined hazard studies are necessary for understanding the risks astronauts face as they travel outside of LEO, and are also critical for countermeasure development. The focus of this review is to describe biologic and functional outcomes from ground-based analogue models for microgravity and radiation, specifically highlighting the combined effects of radiation and reduced weight-bearing from rodent ground-based tail suspension via hind limb unloading (HLU) and partial weight-bearing (PWB) models, although in vitro and spaceflight results are discussed as appropriate. The review focuses on the skeletal, ocular, central nervous system (CNS), cardiovascular, and stem cells responses.
Keywords: Hind limb unloading, tail suspension, radiation, spaceflight, cognition, cardiovascular, SANS, bone, stem cell
1. INTRODUCTION
Both microgravity and radiation exposure in the spaceflight environment have been identified as challenges to astronaut health and performance1–6. While substantial study has been focused on understanding the biology and risks associated with prolonged exposure to microgravity and the hazards presented by radiation within and outside of low earth orbit (LEO), other hazards exist that can impair individual and team performance and well-being. These include the experience of isolation from social groups and networks in confined and extreme environments7–10 combined with lack of sleep quality and quantity with disrupted circadian cycles3,8,11–13. NASA’s research portfolio which examines spaceflight hazards to astronaut health and performance heavily rely on ground-based analogues using rodent models, with most studies focusing on a single hazard. There are ethical and obvious health limitations in performing controlled experiments that examine radiation and/or microgravity effects in astronauts. Hence, the use of ground-based analogues to study spaceflight hazards have been useful in gaining insight into both mission-critical and post-mission effects of spaceflight environmental challenges on astronauts’ tissues, organs, systems, and functional performance. Astronaut health must be maintained in order to ensure success and quality of life as NASA directs resources to planned missions to the moon, Mars, and extended use of the International Space Station (ISS). The ultimate focus of this review is to describe physiologic outcomes from ground-based analogue models for microgravity and radiation, specifically highlighting the combined effects of radiation and reduced weight-bearing from rodent ground-based tail suspension via hind limb unloading (HLU) and partial weight-bearing (PWB) models, although in vitro and spaceflight results are discussed as appropriate and in comparison with HLU results. In addition, the limitations of the HLU model as a microgravity analogue, within and across systems will be discussed.
Loss of gravitational loading on the body and across tissues results in multisystem injury6. Among musculoskeletal tissues that exhibit a dynamic response to loading14–16, exposure to microgravity results in: muscle atrophy17,18, loss of bone mineral density primarily at load-bearing skeletal elements (e.g., pelvis, femur, vertebrae)19–21, and increased risk of herniation of the intervertebral disks due to increased fluid volume22,23. These negative effects also could be compounded by spinal muscle atrophy18 and affected spinal ligaments, ultimately leading to pain23–25. Cardiovascular responses to microgravity include redistribution of blood cranially, increasing arterial pressure at the head and lowering pressure at the distal leg elements26; though the response may be transient. This altered pressure gradient may contribute to spaceflight-associated neuro-ocular syndrome3 in which optic disk edema, optic globe flattening, thickening or folding of the choroid layer, ischemic regions of the retina, and hyperopic visual disturbance may occur27. With these and other systems degraded with exposure to microgravity, as reviewed herein, ground-based analogues are essential for identifying and developing countermeasures. To this end, the rodent HLU model is a primary surrogate for “microgravity”.
The HLU model was originally created as a ground-based analogue for reduced weight-bearing on skeletal tissues in order to study musculoskeletal deficits caused by spaceflight28. Briefly, the hind limbs are lifted off of the cage floor as the tail is tethered to a support bar within the cage1,28,29. Many studies use traction tape to tether and lift the tail and hind limbs off the ground; however other methods for tail suspension exist, for instance pinning of the tail to a tether30. The standard is to raise the tail so that the torso is at a 30° angle relative to the substrate to permit relatively normal forelimb loading, while decreasing load applied across the hind limbs28. Bone and muscle atrophy unsurprisingly occur with the reduction in weight-bearing31–35. This approach of tail suspension has been considered standard in part because it results in other physiologic responses analogous to the body’s response in microgravity, such as a cephalic fluid shift28,36–38. Because of the ensuing fluid shift, reductions in weight-bearing and resulting cardiovascular and metabolic adaptations within and across systems2,39, this approach has been widely adopted as a microgravity analogue compared to other disuse models of osteoporosis, such as neurectomy/nerve crush to induce degrees of paralysis40–42, or limb immobilization models43,44.
While the HLU model remains the gold-standard for ground-based microgravity research, more recently developed models exist that permit investigating gravity as a continuum, using PWB via quadrupedal unloading in both mice45 and rats46–49. The PWB model was principally designed to measure the dose-response relationship between degree of mechanical loading/weight-bearing and the musculoskeletal response and, thus far, studies have demonstrated a clear dose-dependent response but failed to detect the existence of a safe gravity threshold to minimize musculoskeletal deconditioning50–52. The rat PWB model uses of a pelvic harness in lieu of a traditional tail suspension, with limited obstruction of blood flow or creating a cephalic fluid shift as observed from existing studies53, and represents an environment to assess new countermeasures, both therapeutic54 and diagnostic55
Space Radiation Environment and Dose Estimates:
Space explorers outside of low-Earth Orbit (LEO) will be exposed to separate sources of ionizing radiation, energetic protons associated with a Solar Particle Event (SPE) and Galactic Cosmic Rays (GCR)56,57. NASA is currently planning for a sustainable presence on the lunar surface and exploration class missions to Mars, far away from the protection of Earth’s magnetic field. The nature of the radiation exposures that astronauts encounter will likely change to include higher GCR and possible SPE exposures57.
GCR nuclei originate outside our solar system and are high-LET (linear energy transfer), ions with enough energy to easily pass through typical spacecraft with minimal energy loss58. The considerable ionization power of GCR ions makes them the primary antagonist for possible late effects to multiple physiologic systems. During spaceflight in interplanetary space, every cell nucleus will be traversed by a hydrogen ion or delta ray every few days, and by the heavier GCR ion every few months59,60. Despite their relatively low frequency, the heavy ions contribute a significant amount to the GCR dose that astronauts will incur outside of LEO. Thicker shielding could provide protection but is limited by the capabilities of spacecraft launch systems. In fact, studies have shown that even if the amount of, e.g., aluminum, shielding is increased, there will not be a significant reduction of the intra-vehicular radiation dose57,60,61. Astronauts currently on the International Space Station (ISS) are exposed to on average about 1 mSv/day, and even have emergency plans to shield themselves with water bags should an SPE occur. As we travel further outside of LEO, however, it is expected that this daily dose will increase by a factor of 2–3 with limited additional protective measures for SPEs due to spacecraft design and consumables57,62,63.
While background GCR radiation is a concern, SPEs represent an immediate acute exposure of radiation dosage especially if they occur during extravehicular activity. SPEs can produce energetic particles at levels that are orders of magnitude higher than ambient exposures64,65. SPEs consist of high-energy protons that emanate from the Sun from regions of magnetic instability57,62. The ability to predict the occurrence or magnitude of future SPEs, and the likely doses received by exposed crew, are limited66,67. The acute radiobiological effects of whole-body SPE exposures are not well understood. They are complicated by the inhomogeneous distribution of radiation doses to sensitive organs and difficulty with extrapolating animal model data to humans56,57. Additionally, it is unknown how the human health response to SPEs will be affected by concurrent GCR exposure56,57. Based on calculated exposures in the context above, even large SPEs are predicted to deliver doses of <0.5Gy-Eq to internal organs and skin doses of <2.5Gy, with rates of delivery peaking at a dose rate of approximately 0.12Gy-Eq/hr to blood-forming organs66,68. For an SPE similar in magnitude to the 1972 event, an astronaut crew would incur an intra-vehicular skin dose roughly equal to NASA’s 1-year spaceflight radiation permissible exposure limit (3.0Gy-Eq/skin and 500mGy-Eq to blood forming organs64,69,70. Additionally, the dose to blood-forming organs would approach the 1-year permissible exposure limit for these organs (0.5Gy-Eq)69.
Combined Radiation and Reduced Weight-Bearing Overview:
As astronauts will face multiple hazards simultaneously during long duration missions outside of LEO, it is necessary to test the individual and combined effects of these hazards in order to best determine health risk and develop countermeasures. To date, several ground-based rodent studies have combined HLU with radiation exposure to assess combined biologic effects71–73. As detailed herein, while the reduced weight-bearing model is generally HLU (with some PWB), radiation sources vary greatly (primarily photon exposures, with some proton and a few high-Z high-energy ions (HZE) exposures at NASA Space Radiation laboratory (NSRL)). Of note, the majority of studies have performed a period of HLU followed by radiation exposure or initiate the study with radiation exposure and then enroll rodents into HLU35,71–78. A few studies examine outcomes after simultaneously delivering radiation with HLU29,31,72,73,79–84. For these, rodents are irradiated in the beamline at the NSRL, or at other facilities with low-dose rate gamma rays or photons.
2. SKELETAL RESPONSE
Bone Loss in Microgravity and Reduced Weight-Bearing:
Spaceflight environmental conditions, particularly microgravity and low-dose radiation, represent a risk to astronaut bone health. In the absence of mitigation strategies, spaceflight can lead to decrements in bone mass and strength85. Crew members returning from a 4–6 month mission on the ISS had reduced areal bone mineral density (aBMD) of the hip and spine. The rate of bone loss was found to be site-specific, with a 1.4–1.5% and 0.9% decrease in aBMD of the hip and spine respectively per month in the ISS. In the hip, the rate of loss was higher in cancellous bone (2.2–2.7%/month) compared to cortical bone (0.4–0.5%/month)86. In contrast, trabecular vBMD and mass decreased by 14.4–16.5 % throughout the mission duration87. Bone strength in the proximal femur also was reduced as determined by finite element modeling for stance and fall loading21. These decrements in skeletal structure and strength may increase the risk of fractures, which in turn can be catastrophic to future long duration spaceflight missions where there will be operational constraints on medical care and equipment.
Animal models have been used to gain insight on spaceflight-induced bone loss and its underlying mechanisms. As noted, because of the dynamic response of bone to loading, one of the advantages of using animal models, particularly from ground-based HLU, is that brisk bone loss can be achieved (within weeks to months) upon exposure to simulated microgravity28,36,39,88 and partial weight-bearing45,48,50,89. The magnitude of bone loss achieved by HLU varies across studies which is unsurprising given differences in methodologies, duration and age at onset of HU, as well as sex and strain of animals used. Seven days of HLU in 17 week old male C57BL/6J mice led to ~15% decrease in % cancellous bone volume (BV/TV) in the proximal tibia78. In female C57BL/6J mice, one month of HLU led to a 74% reduction in % bone volume in the proximal tibia35. A 21 day HLU period in 12 week old male C57BL/6 mice led to decreases in BMD as measured at different sites along the femur90. The magnitude of HLU-induced decreases in BMD ranged from 8–11% as measured from the proximal to distal end of the femur90. The appendicular bones of humans and rodents have notable differences in anatomy and stance loading, in addition to dissimilarities in the timing and duration of postnatal skeletal growth. Hence, anticipating the magnitude of bone loss in humans from a rodent model for skeletal disuse is not straightforward. Nevertheless, the results from the HLU model are generally consistent with findings in humans that microgravity can accelerate bone loss.
Space Radiation-induced Bone Damage:
Bone loss also has been examined in animal models for space radiation exposure. Historically, bone was considered radiation resistant, and thus the dose threshold for damage was assumed to be high91,92. However, studies performed in the early portion of the 2000’s identified late bone loss in mice after 2 Gy exposures to multiple qualities of radiation (e.g., protons, iron ions, carbon ions, and photons), with the consideration that 2 Gy represented an appropriate comparative dose between clinical fractions and spaceflight exposures93. Later studies typically made use of a lower reference dose of ionizing radiation (e.g. 1–2 Gy 137Cs)73,81,94, a single representative species of GCR35,84,95,96 or sequential exposure to two ion species72,97. These spaceflight relevant studies identified that while osteoblast activity is lower after exposure (which historically was assumed to be the mediator of late bone damage after high dose exposure), osteoclast activity was increased as an early response98,99, leading to acute bone loss78,98. The dose threshold for bone loss after simulated spaceflight radiation was determined to be low, with mixed fragment doses <0.5 Sv (protons and helium ions) causing late bone (and muscle) damage100, and individual doses of 0.5 Gy 56Fe and protons causing acute bone loss in mice101. These investigations have thus confirmed that bone is a radiation sensitive tissue, with the outcome being loss of architecture and density. However, one study has reported that radiation can have positive effects on bone microarchitecture74. Skeletally mature (16 week old) female mice exposed to 0.5 Gy 56Fe exhibited increased cancellous % bone volume (BV/TV), trabecular thickness (Tb.Th.) and trabecular number (Tb.N.) in the distal femur at 21 days post-irradiation. These results highlight that while radiation is generally considered disadvantageous, many questions remain regarding how low dose radiation could affect bone, including questions regarding bone material and mechanical properties that occur with alterations in architectural properties. These questions should be addressed in the context of complex, mixed beams of energetic particles to improve the model spaceflight-relevant radiation exposures. Recently, the NSRL has developed a GCR simulation involving a more complex combination of multiple ion species that approximates the anticipated exposure of crew based on current spacecraft design102.
Impact of Combined Exposure of Space Radiation and Simulated Microgravity on Bone Structure and Function:
Published studies on the combined effects of microgravity and radiation on skeletal integrity differ in experiment designs, making comparison and validation of results across studies quite challenging. Despite the limited number of reports and heterogeneity in experiment designs, two major conclusions can be drawn from collective findings. Firstly, findings from animal models predict that bone loss due to microgravity and ionizing radiation exposure can be additive under certain conditions. Secondly, skeletal parameters may have differential sensitivities to combined spaceflight factors. In some studies that have used representative high linear energy transfer (LET) species in combination with HLU, additive effects were seen in a subset of skeletal structural parameters but not in biomechanical properties. For example, 16-week old, male C57BL/6J mice that underwent 14 days of HLU displayed decreased cancellous and cortical BV/TV in tibia versus normally loaded controls while similarly aged cohorts exposed to 50 cGy proton or 50 cGy proton + 10 cGy 16O did not show such decrements. Yet when HLU was combined with 50 cGy proton + 10 cGy 16O exposure, further decrements in BV/TV were observed. Biomechanical properties such as maximal force, measured by three-point bending, was negatively impacted by HLU but not by radiation. However, no further deficits were observed when HLU and radiation were combined72. These findings are generally consistent with another report which made use of similarly aged female mice35 exposed to 100 cGy of protons in combination with four weeks of HLU. Combined microgravity and radiation also can lead to skeletal impairments at the cellular level. Skeletal homeostasis is achieved through the balance in the activity of bone-forming osteoblasts and bone-resorbing osteoclasts. Exposure to 0.5 Gy 56Fe was found to exacerbate HLU-induced deficits in alkaline phosphatase activity of marrow-derived ex vivo osteoblast cultures, suggesting that microgravity in combination with radiation exposure can impair osteoblastogenesis96. Additionally, two weeks of HLU combined with 1 Gy 56Fe delivered on Day 3 led to enhanced bone loss versus exposure to each single hazard. This study also identified potential mechanisms for bone loss in the hind limb from each challenge, related to endothelial dependent vasodilation of the feed arteries of the gastrocnemius83. Specifically, it was identified that altered vasodilation associated with bone loss was mediated by altered nitric oxide (NO) synthase signaling, although occurring in divergent manners between hazards: with HLU, alterations occurred to reduced production and concentration of NO, and radiation-induced alterations were due to increased NO quenching.
Long duration missions will involve extended periods of exposure to low doses of space radiation. Most of the available data on the effects of radiation on the skeleton were generated from acute exposures. Published results from fractionated or sustained exposure models remain a rarity. One group has examined the effects of 0.5 Gy 28Si delivered as an acute dose or three fractionated doses of 0.17 Gy in combination with PWB at one sixth body weight95. The PWB-induced decrements in both endocortical and periosteal % mineralizing surfaces of cortical bone sites appeared to be worsened by 28Si although standard deviations were wide, therefore necessitating additional confirmatory studies95. In general, neither acute nor fractionated exposure exacerbated the effects of partial weight-bearing on femoral BV/TV and femoral neck biomechanical properties such as load to failure and stiffness. Others who have simultaneously applied continuous exposure to low dose rate photons (8.5cGy 137Cs) exhibited no radiation response, only bone loss due to HLU over the course of a 20 day study81.
The long-term consequences of combined microgravity and radiation on skeletal health is unclear given that most investigations have focused on immediate time points (e.g. assessment after unloading). However, there is evidence that combined exposure to HLU and HZE (0.5 Gy) can negatively impact skeletal recovery after a period of unloading. In one such study84, animals that received an acute dose of 0.5 Gy 56Fe within a 14-day period of HLU displayed persistent deficits in vertebral trabecular morphology (structural model index, SMI) after a 28-day period of re-ambulation. However, cancellous BV/TV of vertebra of these animals were comparable to untreated controls after the same time period.
Humans living in space experience circadian misalignment103. The interaction of the circadian clock with microgravity and space radiation has not been well examined with alterations in bone as an outcome. One investigation exposed rats to an acute, high dose of radiation (4 Gy 137Cs), with ultradian cycles (45 min light/45 min dark), HLU or all factors combined. Ultradian cycles were sufficient to induce decrements in bone biomechanical properties but did not lead to bone loss. In addition, ultraradian rhythms did not worsen the negative effects of HLU and radiation on bone structure and strength.
Countermeasures in place to prevent osteopenia during ISS missions include load-bearing exercises. Strides also have been made in identifying other promising candidate countermeasures for mitigating spaceflight-induced bone loss. Antioxidants are often considered as candidate countermeasures to protect against radiation and/or HLU-induced bone loss: as previously noted altered NO concentration (and expanded upon in Section 5; Cardiovascular Response) is associated with radiation and HLU83, and biomarkers for oxidative stress in the marrow in another study has been identified to increase after radiation but not HLU78, highlighting some inconsistencies in the literature. In a rodent model, pre-feeding with an antioxidant-rich dietary supplement (dried plum) prevented bone loss and decrements in bone strength resulting from HLU, ionizing radiation and in combination94. These results suggest some shared mechanisms underlying microgravity and radiation-induced bone loss. However, while antioxidants such as dihydrolipoic acid (DHLA) or an antioxidant cocktail have been shown to be less efficacious at reducing bone loss in mice early after 2 Gy gamma ray exposure,97 alpha-lipoic acid78 has been shown to reduce early bone loss after 2 Gy gamma rays. Collectively, these findings suggest that antioxidants have varying efficacies in preventing the negative effects of spaceflight stressors on bone. Future studies are needed to better understand the underlying mechanisms for the protective effects of promising antioxidant-based countermeasures for spaceflight.
Additional and Future Considerations:
Although much has been achieved in understanding the effects of spaceflight on bone health, follow-up studies are needed to validate the abovementioned findings using improved radiation exposure paradigms such as the recently developed multi-ion GCR simulation102. The use of animals that better match the age of mission crew when performing spaceflight simulation studies will facilitate translation of rodent data to humans in space. In addition, standardization of methodologies and caging designs for conducting HLU and partial weight-bearing studies also are important to allow for comparison and interpretation of findings across studies and research groups. More mechanistic investigations also are needed to understand the molecular underpinnings of the skeletal response to combined spaceflight factors. Bioinformatics approaches that allow for the assessment of global changes at the genome, transcriptome and proteome levels are particularly useful in identifying novel pathways that underlie the skeletal response to spaceflight. To date, omics data from skeletal tissue is rare. Of the two traditionally mechanosensitive tissues, muscle vastly outnumbers bone or bone cell transcriptomic datasets found on the NASA GeneLab database. Increasing the availability of bone omics datasets will facilitate improved understanding of skeletal signaling in response to spaceflight.
Initiatives to study bone health in space have predominantly focused on how spaceflight negatively impacts skeletal structure and biomechanical properties with the overarching goal of understanding how they contribute to fracture risk. While it is important to address remaining knowledge gaps on fracture risk in follow-up studies, investigations by the space research community also need to broaden to reflect our growing understanding of skeletal function. Beyond providing structural support, the skeleton is an endocrine organ that can crosstalk with other tissues to maintain health and homeostasis. A number of studies demonstrate the ability of bone to function as an endocrine organ (reviewed in104–106 to regulate a variety of physiological processes including glucose metabolism107–109, appetite suppression110, cognition and behavior111,112. The organism’s ability to coordinate the function of multiple tissues via bone-derived factors is essential to keep up with the demands of daily living. For example, bone-derived lipocalin-2 (LCN2) regulates glucose homeostasis via endocrine action on major metabolic organs, and also can cross the blood-brain barrier to control appetite via its binding to the melanocortin receptor (MC4R) in the hypothalamus110. In addition, LCN2 can exert pro-inflammatory actions on a variety of cell types including vascular cells113. Osteocalcin (OCN), another bone-derived hormone can modulate cognition and anxiety-like behavior111,112. Mice heterozygous for an OCN null allele showed deficits in cognition while administration of OCN improved memory and decreased anxiety-like behaviors112. In addition, OCN has been shown to mediate aspects of the acute stress response. In the presence of stressors, it is thought that OCN participates in signaling to inhibit the parasympathetic branch of the autonomic nervous system to allow the sympathetic pathway to predominate, in turn promoting flight or fight responses114. The biological processes regulated by these bone-derived hormones are critical for human health and performance in space missions. Hence, it is important to begin to address whether combined spaceflight factors can perturb signaling mediated by bone-derived hormones. More studies also are needed to understand the role of bone crosstalk with other organs in mediating the physiological changes attributed to spaceflight.
3. OCULAR RESPONSE
There has been an increase in the incidence of the ocular problem reported in astronauts during and after space shuttle missions or orbits aboard ISS3. As previously noted, this syndrome, known as spaceflight associated neuro-ocular syndrome (SANS), is characterized by pathophysiology symptoms including optic disc edema, globe flattening, choroidal and retinal folds, hyperopic refractive error shifts, and nerve fiber layer infarcts (i.e., cotton wool spots)115–117. In the last decade, over 30% of astronauts flying long-duration ISS missions have presented with one or more of these ocular disturbances118. Most recently, the NASA twin study found variations in choroidal and total retinal thickness, which was suggestive of retinal edema and choroidal folds in the twin exposed to spaceflight119. There is concern that degradation of visual function as a result of space flight may compromise both mission goals and long-term quality of life after space travel.
Ocular damage and retinal degeneration can be promoted by many factors including aging, ischemia, fluctuation in oxygen tension, oxidative stress, and increased intraocular pressure120. Visual disturbances associated with space travel may be due to exposure from altered gravitation changes and ionizing radiation116,120–122. Although some ocular changes experienced by astronauts have been measured and evaluated117,118, validation was difficult due to the limited subject cohort size and test constraints on ISS. Furthermore, the underlying mechanisms of these ocular disturbance and factors contributing to the development of damage are currently unclear. Comprehensive ground-based rodent study models to simulate space condition including low-dose ionizing radiation and microgravity is warranted to determine the impact of the space environment on ocular structure and function.
Microgravity-induced Ocular Damage:
Microgravity induces a cephalic shift in body fluids, an increase in cephalad shifting of body fluids, and alterations in tissue perfusion121,123,124. Increasingly, evidence suggests that both actual microgravity encountered by astronauts in space, as well as modeled microgravity on Earth, have been shown to induce many deleterious physiological effects including changes in ocular structure and function125,126. After long-duration spaceflight, morphological changes in the optic nerve and surrounding tissues have been reported127. One study showed that even in transient microgravity conditions, as produced by parabolic flight, changes in retinal vasculature occur117. Microgravity may also induce an increase in intraocular pressure (IOP)128. A more recent study reports that in astronauts, there is an acute increase in IOP upon entering weightlessness, but that it normalizes to ground-based levels after a few days of flight118. Despite reported observations, cellular mechanisms of microgravity in inducing the unique physiological and pathological ocular responses have not yet been well-understood. Moreover, head-down tilt (HDT) during bed rest as a ground-based, human analog for microgravity has not confirmed some of SANS findings in astronauts129.
Space Radiation-induced Ocular Damage:
The adverse effects of radiation on the retina130–133 and retinal vasculature132,134 have been reported by multiple investigators who have documented structural, histopathological, and functional alterations in the affected retina after irradiation exposure. Despite reported studies, most of the animal and clinical investigations from which the current knowledge of radiation-induced ocular injury were obtained, are from relatively high doses of photon radiation. More recently, some ground-based studies have been conducted in rodent models to investigate low-dose space radiation-induced changes of ocular structure and function. Studies show dose-dependent increases in apoptosis in the retina following space radiation exposure. Data revealed that exposure to proton radiation-induced oxidative stress and apoptosis in the retina at a dose as low as 0.5Gy124. Analysis of the microvasculature in the rat retina showed a time- and dose-dependent, progressive loss of endothelial cells and microvessel length over for two years after proton irradiation135. Low doses of 16O ions also elicited apoptosis in the mouse retinal endothelial cells with the most robust changes observed after 0.1 Gy exposure compared to controls136.
Impact of Combined Exposure of Space Radiation and Simulated Microgravity on Ocular Structure and Function:
One of the main concerns for long-term deep manned space missions are health risks associated with combined exposure to microgravity environment and low-dose/low-dose-rate (LDR) radiation above levels normally found on earth due to GCRs. It is an important contribution for risk assessment to determine whether the low dose radiation response is modulated by simulated microgravity. The study design using a ground-based animal model to assess the biological effects of the spaceflight condition, combining space-like radiation exposure and microgravity is a more accurate model to simulate environmental stressors inherent to the spaceflight environment, providing a more actual risk assessment for astronauts. To our knowledge, ground studies to examine the impact of the simulated space flight conditions and underlying mechanism(s) of potential interaction on retinal structure and functional damage are very limited.
In one study, mice were HLU for 7 days, then whole-body irradiated with protons at 0.5 Gy, followed by HLU for an additional 7 days. The data showed SPE-like exposures of proton irradiation alone or combined with simulated microgravity has a significant impact on retinal endothelial cell survival71.
Additional and Future Considerations:
As noted earlier, factors and their interactions that contribute to detrimental ophthalmic changes to the spaceflight environment are not well-investigated. More studies are needed to expose animals to microgravity simulation and low-dose space radiation simultaneously over at least 4 weeks which simulate the duration of the ISS mission. This will allow the data to be extrapolated more accurately to estimate potential risks to astronauts in the space flight environment. To simulate key aspects of space radiation exposures, further rodent study designs should consider exposing animals to both the charged particle composition of the radiation field and its low dose rate for ocular response measurements. The observation periods for the study especially for degenerative tissues need to be extended for a longer time after radiation/unloading exposure to characterize the readaptation and chronic deficits. One of the complications associated with determining the response of stress insults is the latency between exposure and the expression of injury (e.g., cell loss or dysfunction). In order to obtain accurate data for the development and progression of the injury response, it is necessary to quantify changes over a long period.
The most profound physiological response and adaptation to the microgravity environment is the redistribution of fluid121. The mechanisms by which fluid shift in the spacecraft environment that could affect ocular function were less studied. Dedicated studies are needed to identify models to address this important question regarding the impact of the fluid shift on ocular structure, physiology, and visual function. Further studies are also needed to specifically assess ocular perfusion pressure and ocular hemodynamics in appropriate animal models. Electrophysiological assessment using electroretinogram (ERG) or other functional endpoints will be helpful to determine retina functional changes related to observed structural alteration.
Underlying cellular mechanisms of spaceflight environment, in facilitating ocular damage remain unclear. Some lines of evidence suggest that one of the mechanisms involved in response to spaceflight, including changes in the gravity vector, is likely due to oxidative stress137,138. Studies have shown that exposure to microgravity during spaceflights is associated with increased oxidative stress markers reflecting damage in lipid which results in lipid peroxidation in both humans and rodents139,140.
To expand our knowledge about the effects of spaceflight condition on the eyes and possible mechanisms associated with these changes, integrated omics profiling technologies such as genomics, proteomics and metabolomics are beneficial to determine sets of differentially expressed genes (DEGs), differentially expressed proteins, metabolomic/lipidomic signatures and the pathways that lead to pathological and possible degenerative changes. Recently, RNA sequencing from a spaceflight study detected 600 DEGs in murine spaceflight retinas, which were enriched for genes related to visual perception, the phototransduction pathway, and numerous retina and photoreceptor phenotype categories141.
Delineating differential gene and protein expression and their relationship to overall pathophysiological and functional changes in the ocular tissue will provide a basis for the discovery and development of biomarkers and pathways for neurovascular changes in response to spaceflight condition. Herein, using the “omics”-based molecular phenotyping approach for characterizing biosignatures associated with low-dose space radiation, simulated microgravity, and other space environmental stressors will help a deeper understanding of the underlying mechanisms responsible for the ocular structural and pathophysiological changes.
4. CENTRAL NERVOUS SYSTEM (CNS) RESPONSE
Importance of CNS Functionality to Mission Success:
Any deterioration in the ability of the astronauts to perceive or respond to changes in their situation could have disastrous consequences, as could changes in the mental health of the astronauts. The ability of astronauts to successfully complete a deep space mission, such as the ones planned for Mars, will thus be highly dependent upon a fully functional CNS. Not surprisingly, NASA has devoted considerable efforts to establishing the impact of social isolation, stress, sleep disturbances/loss and microgravity on various aspects of astronaut performance (cognition, sensorimotor response, social interaction, sleep) both during space flight and in rodent ground-based analogs. In many cases there are well established procedures and interventions to detect and mitigate these stressor related issues. However, there remains a high degree of uncertainty about the impact that exposure to space radiation will have on the cognitive and psychological capabilities of astronauts.
Microgravity Effects on the CNS:
Microgravity is also a major stressor on the CNS, inducing changes in the structure of the brain (rotation of the cerebral aqueduct, changes in ventricular volume, and narrowing of cerebrospinal fluid (CSF) spaces at the vertex142, and a cephalic fluid shift. It also produces significant effects on the brain, particularly in cerebellar, sensorimotor, and vestibular brain regions (Reviewed in143). Brain activity may also change in response to the need for increased processing required for postural stabilization, and integration of conflicting vestibular information in the microgravity environment144. Despite all these changes, the evidence that prolonged microgravity leads to a permanent loss of cognitive function is sparse. Astronauts report a “Space fog” for 1–2 days into a mission, but this typically resolves.
However, at the cellular level, simulated145 microgravity result in persistent changes in the mitochondrial function and lipid metabolism of human oligodendrocytes. Oligodendrocytes are essential for providing metabolic support to neurons, rapidly transferring (through cytoplasmic “myelinic” channels and monocarboxylate transporters) short-carbon-chain energy metabolites like pyruvate and lactate to neurons146. Such microgravity induced metabolic perturbations are likely to be deleterious to neuronal function in their own right, but will likely exacerbate changes in neuronal functionality with combined exposure to other spaceflight environmental hazards, such as radiation.
Space Radiation alters Neurophysiology and Neurocognitive Performance:
There is an ever-growing body of evidence from ground-based rodent studies that radiation exposure impairs performance in many cognitive processes, ranging from relatively fundamental processes to complex analogs/homologs of human cognitive tasks. Even in the one study that demonstrates an apparent radiation-induced improvement in pattern separations skills147, mice demonstrated reduced associative memory formation ability, and the apparent improvement may be attributable to an increase in sparsely encoded hippocampal-dependent memory. Thus, the overall consensus from ground-based rodent studies is that radiation exposure impacts performance in multiple cognitive tasks, utilizing multiple cognitive process governed by multiple brain regions. Mechanistic studies have revealed multiple changes in neurophysiological processes and dendritic structure within most brain regions investigated148–159. Moreover, there may be a loss of connectivity between brain regions149.
Space Radiation Alone Impacts Cognitive Processes Deemed to Be of Operational Significance:
While it could be argued that the loss of the ability to perform a task like novel object recognition may not have any operational significance, the radiation-induced loss of performance in rodent versions of tests widely used to assess attention and cognitive flexibility in humans cannot be so easily discounted. Astronauts are routinely screened during space flight for performance in a 10-test battery of cognitive tasks that NASA deemed necessary for mission success. Seven of the tasks in the “fit-for-duty” performance battery160,161 assess some aspect of executive function. Executive function, in lay terms, can be summarized as the “Triple A”: the ability to Assess, Adapt and Achieve. More technically, executive functions are a set of higher order cognitive abilities that animals utilize to keep information ‘in mind’, attend to appropriate cues (e.g., nonverbal and verbal working memory stimuli), update information as contingencies change and invoke alternative, more appropriate responses to new situations.
The rodent version of the psychomotor vigilance test (rPVT) is virtually identical to the PVT test that is part of NASA’s “fit-for-duty” performance battery on the ISS160,161. PVT performance is sensitive to fatigue, drug use, and age162,163, and exposure to mission-relevant (25 cGy) doses of protons results in deficits in accuracy, impulsivity and lapses in attention, all of which are indicative of deficits in sustained attention164. Such lapses in attention account for 80% of flight accidents in the Navy and Marine Corps165.
A key process that allows humans to rapidly and efficiently adapt to different situations is task- or set-shifting. An attentional set is formed when complex stimuli must be discriminated and classified as relevant or irrelevant to a particular task/situation. Set-shifting can be simplistically thought of as the ability to relearn what the most important discriminating stimulus (for a particular endpoint) is in a changing environment. Attentional set shifting (ATSET) thus enables subjects to rapidly adapt and respond to changes in the environment, and to perceive what is important for survival or completion of a task, skills that are required to deal with a sudden emergency. The Wisconsin Card Sorting Test (WCST) has been widely used to assess task switching in humans166, with task switching deficits being increased in patients with Parkinson’s disease167,168 and autism169. The intra-dimensional (IDS)/extra-dimensional (EDS) set shifting task is a modification of WSCT that assesses set-shifting abilities in rodents170. Performance in the ATSET assay is impaired after exposure to ≤ 15 cGy of 1 GeV/n 56Fe155,171, 1 GeV/n 48Ti150,151, 600 MeV/n 28Si148 and protracted low dose rate neutrons172.
Astronauts on deep space missions will have to act more autonomously than on previous missions, especially when rapid responses to unexpected problems are required. Creative problem solving skills will thus be of great importance to astronauts on a mission to Mars. Recent studies have shown that low (≤18 cGy) doses of 600 MeV/n 28Si and 252Cf-generated neutrons impact creative problem solving in rats148,172. Rather worryingly at the individual level, poor creative problem solving performance in the irradiated rats was not necessarily associated with poor ATSET performance, and vice versa148,172. Previously we have shown that while space radiation exposure impairs both spatial memory and ATSET performance, however, when the relative performance of individual rats in each task was compared there was no correlation between space radiation-induced loss of performance in each task173. These data suggest that risk assessments for radiation-induced neurocognitive impairment derived from a single cognitive domain may greatly underestimate the severity of the problem.
Establishing Space Radiation-resilience and Incidence of Severe Cognitive Impairment:
A notable feature of the rat-based studies was the marked inter-individual differences in cognitive flexibility performance of the irradiated rats, with some rats having performance metrics comparable to shams, while others completely fail to reach criterion in the cognitive tasks148,151,155,164,171. Analysis of the individual animal performance data using kernel density estimation to generate a performance probability profile, has allowed for an estimation of how frequently severe cognitive impairments are induced171,172,174. In these studies, the level of performance that was considered to represent severe ATSET impairment was set at the 5th percentile of the sham cohort performance profile (conceptually analogous to a Z-score of −2). Translating ground-based rodent studies into tangible risk estimates for astronauts remains an enormous challenge, but should similar neurocognitive impairments occur in astronauts exposed to low space radiation doses, a Numbers-Needed-to-Harm analysis (of the rodent data) predicts that ~30% of the astronauts could develop severe cognitive flexibility decrements171,172.
Combining Spaceflight Hazards on Space Radiation-induced Cognitive Impairment:
To date, this is a largely uninvestigated, but much needed, field of study for the effect of simulating multiple aspects of the space environment on behavioral impairment. Relatively little has been documented regarding cognitive functioning after extended exposure to combined spaceflight condition, and findings have been inconsistent. One study showed that mice exposed to HLU displayed behaviors suggesting abnormal exploration and/or high risk-taking behavior in the elevated zero maze. However, low-dose radiation exposure did not exacerbate HLU-induced behavioral changes where mouse was exposed to combination of low-dose gamma radiation and simulated microgravity by HLU175. In an in vitro study, synergistic changes of reduction in neuronal network integrity and cell survival induced by simulated space radiation and microgravity were observed176. Another study documented that learning and memory abilities were significantly reduced in rats under a simulated spaceflight hazards including microgravity, isolation confinement, noises, and altered circadian rhythms177. The potential interactions between space radiation, microgravity and other space environmental stressors should be addressed with comprehensive models including neuroimaging, electrophysiology, biological, and clinical data178.
Additional and Future Considerations:
Both sleep deprivation and sleep fragmentation (SF) have been linked to reduced neurocognitive functioning in humans and animals, and has a major impact on performance in multiple cognitive domains179–182. Two recent studies have established how inadequate/disturbed sleep alters the severity of space radiation-induced impairment of executive function. A single session of fragmented sleep uncovered latent ATSET performance deficits in rats exposed to both protracted neutron172 and Si183 irradiation that had no obvious defects in performance under rested wakefulness conditions. SF selectively impaired performance in the more complex set shifting stages of the ATSET test in the both neutron and Si irradiated rats. Set shifting performance has rarely been impacted by exposure in studies conducted with rats tested under rested wakefulness conditions. Thus, radiation-induced cognitive impairment may not be fully evident in normally rested rats, substantially underestimating the level of impairment that may occur when astronauts are on mission. Cognitive testing may thus have to be conducted under both rested wakefulness and SF conditions to get a more accurate assessment of radiation-induced neurocognitive impairment. Further ground-based studies on the impact of radiation on cognition in rodents that are under concomitant exposure to space flight stressors such as stress and social isolation are being initiated. Based upon the exacerbating effects of sleep reduction on the incidence, severity and type of cognitive impairments induced, it would seem likely that concomitant exposure to these other space flight stressors would result in similar “negative interactions”.
5. CARDIOVASCULAR RESPONSE
Cardiovascular Response in Microgravity and Reduced Weight-Bearing:
The vascular endothelium is an important regulator of vascular tone in arterial, venous, and lymphatic vessels, and is vital for the protection of arteries from the development of atherosclerotic plaque. The production of NO by the vascular endothelium through the NO synthase (eNOS) signaling pathway is a critical component for these vascular functions. Limited information is available regarding the effects of spaceflight on the vascular endothelium in humans. Lee et al.(184) reported that long-duration spaceflight in low Earth orbit elevated biomarkers of systemic oxidative stress and inflammation during flight, and these circulating biomarkers returned to preflight levels soon after landing. Despite the increase in systemic oxidative stress and inflammation, the astronauts showed no corresponding change in brachial artery endothelium-dependent vasodilation. Although these results suggest that spaceflight-induced elevations in oxidative stress and inflammation do not adversely impact vascular endothelial function, it is important to note that the source of the circulating biomarkers is unknown and may originate from specific or unique portions of the circulation, such as the mesenteric vascular bed. Therefore, results obtained from large conduit arteries from non-weight bearing limbs may not be indicative of changes associated with specific vascular beds, including the coronary and cerebral circulations.
In animal studies, spaceflight-induced apoptosis of vascular endothelial cells has been shown to occur in the retina and brain, and this loss of endothelial cells correspond with markers of disrupted barrier function of the blood-retinal185 and blood-brain barriers120,185,186. Impairment of arterial endothelium-dependent vasodilator function has also been reported to occur in cerebral and mesenteric arteries following spaceflight187,188. HLU has been shown to diminish cerebral artery endothelium-dependent vasodilation through the eNOS signaling pathway189 and possibly through an upregulation of vascular cell adhesion molecule-1190.
Although animal studies demonstrate that spaceflight can have acute adverse effects on the vasculature, studies of mortality among US astronauts exposed to low-earth orbit, relative to the general population, non-astronaut NASA employees, and nonflight astronauts, indicate the risk of death due to cardiovascular disease is not elevated191,192. Thus, spaceflight appears to have few long-term adverse consequences on the cardiovascular system. These mortality data are, however, somewhat limited in that most astronauts included flew in space for relatively short periods of time and remained predominantly in low Earth orbit. The risk for developing cardiovascular disease may be elevated as mission duration increases or as exploration goes beyond the Earth’s magnetosphere.
Space Radiation-induced Cardiovascular Response:
Emerging epidemiological research demonstrates that low-dose environmental, occupational, and medical radiation exposure increases the risk of mortality due to ischemic coronary artery and cerebrovascular disease193,194. However, these risk estimates of Earth-based irradiation are largely derived from low linear energy transfer radiation exposures, which have some fundamentally different properties from charged HZE particles present in the deep space environment. HZE ions, for example, produce greater adverse effects on cellular physiology through increased genetic alterations and perturbations to redox metabolism, leading to persistent elevations in oxidative stress195. Oxidative stress can impair vascular and cardiac function by direct oxidative damage or by activating cell signaling pathways that can lead to abnormal contractile, inflammatory, proliferative, or remodeling properties.
In animal studies, vascular endothelial cells have been shown to be especially sensitive to the effects of radiation. For example, Soucy et al.196,197 demonstrated that simulated space irradiation with 56Fe ions impaired endothelium-dependent vasodilation of the aorta and increased aortic stiffness; both these effects were secondary to the formation of reactive oxygen species. Other studies have likewise shown that simulated space radiation induces damage to vascular endothelial cells198,199 which could lead to diminished barrier function and accelerated development of atherosclerotic plaque. Indeed, research by Yu et al.200 has shown 56Fe irradiated portions of the aorta in apolipoprotein E-deficient mice have accelerated atherogenesis in the targeted regions.
In addition, space radiation has an adverse impact on cardiac tissue. Low dose whole body irradiation with protons or 56Fe ions were reported to produce myocardial DNA methylation201. 56Fe irradiation has also been shown to diminish left ventricular function and increase infarction size and mortality rate in mice with a surgically induced myocardial infarction202. Collectively, these studies indicate that the damaging effects of space radiation can occur within the myocardium as well as the vasculature.
Combined Effects of Simulated Space Radiation and Weightlessness on Vascular Function:
The possible synergistic impact of space radiation combined with microgravity-induced weightlessness on degenerative cardiovascular disease in astronauts is poorly understood. To assess this possible synergy, Ghosh et al.83 examined the single and combined effects of simulated space radiation with 56Fe ions and simulated weightlessness using HLU on endothelium-dependent vasodilation of mouse gastrocnemius muscle resistance arteries soon after the cessation of the treatments. Both 56Fe ion irradiation and HLU alone each impaired endothelium-dependent vasodilation, but this impairment was potentiated when the two treatments were combined. The endothelial dysfunction occurred primarily through the eNOS signaling pathway with 56Fe ion irradiation and HLU, but with 56Fe ion exposure the deficit was the apparent consequence of diminished anti-oxidant capacity and greater pro-oxidant protein expression in the artery, while with HLU the deficit in endothelium-dependent vasodilation was the apparent consequence of reduced eNOS protein content. These data indicate that a short-term consequence of space radiation exposure and weightlessness is the synergistic impairment of the vascular endothelium.
In a follow-up study to determine the long-term effects of simulated space radiation and weightlessness on vascular health, Delp et al.192 repeated the abovementioned studies of Ghosh et al.83, but gave the mice a 6–7 month recovery period, the human equivalent of 18–20 years, before the vascular studies were conducted. The results demonstrated that vascular impairment of endothelial function was not sustained in the HLU mice, which is consistent with the collective US astronaut cardiovascular mortality findings191,192. However, impairment of endothelium-dependent vasodilation persisted in the irradiated mice. This persistent radiation-induced endothelial dysfunction occurred through the eNOS signaling pathway and was associated with greater expression of the pro-oxidant protein xanthine oxidase. These data indicate that there are no long-term vascular consequences to weightless, but that the radiation effects of deep space travel on the vascular endothelium may endure through persistent elevations in oxidative stress192.
Additional and Future Considerations:
Understanding of the singular and combined effects of space-associated weightlessness and irradiation is only now beginning to emerge, and many areas of research are open to further investigation. For example, vascular alterations induced by radiation may not be uniform throughout the circulation, given that the local biochemical milieu surrounding blood vessels and local mechanical forces (e.g., blood flow and shear stress) may interact with the local radiation effects to produce variable responses in different regions of the body, such as in the coronary and cerebral circulations. Much of what is known about the effects of spaceflight on the cardiovascular system has come from the study of arterial vessels, but more research is needed to understand the effects of weightlessness and space radiation on venous and lymphatic vessels. This is particularly true given the likely involvement of venous and lymphatic vessels in the development of spaceflight associated neuro-ocular syndrome115–118 and venous thrombosis203,204 reported to occur in astronauts during long-duration spaceflight. Finally, little is known about possible sex-specific differences, or the effectiveness of various countermeasures (e.g., exercise, antioxidants, and nutraceuticals) on cardiovascular alterations associated with weightlessness and deep space irradiation. All these areas of research are vital as human exploration of deep space is being pursued.
6. STEM CELL RESPONSE
Tissue regeneration is both a highly mechanosensitive and radiosensitive process, relying heavily on mechanical cues imposed by Earth’s gravity205 and protection by the magnetosphere from harmful ionizing radiation. Data obtained from spaceflight experiments have documented the extensive effects of spaceflight stressors on progenitor cell populations and stem cell differentiation capabilities. These cellular changes often propagate into physiological defects which may pose significant health risks to astronauts both during and after sustained spaceflight missions. This section focuses on stem cell responses across physiologic systems, primarily through use of both ground-based HLU models, but also simulated microgravity devices (e.g., rotary cell culture systems, rotating wall vessels, and 3D clinostats).
The role of gravity in maintaining tissue homeostasis has become apparent with expanded study examining the effects of spaceflight on mammalian physiology. Dysfunction in stem cell populations contribute to many Earth-based disease conditions and can be enhanced by aging, oxidative stress, and genetic predisposition206,207. Adult stem cell populations are found in multiple physiological systems throughout the body and are surrounded by a highly organized and regulated microenvironment consisting of supporting cells and factors, resulting in the formation of a stem cell niche208. Following injury, damage, or normal cell attrition, stem cells within the niche receive signals resulting in transition to an active state and initiation of the differentiation process209. Therefore, in order for regeneration of damaged tissues to occur, resident stem cell pools must be activated and induced to differentiate into lineage specific cells210. Such activation signals may be biochemical or mechanical in nature, and therefore may be affected by exposure to microgravity. Spaceflight exposure may result in premature aging of specific physiological systems, and loss of stem cell functions may contribute to the initial observed tissue degeneration but more importantly, may be linked to regenerative deficits during long-duration spaceflight exposure beyond LEO.
Microgravity-induced Stem Cell Alterations:
Several studies specifically investigating the effects of microgravity on embryonic stem cell function have identified a deficit in differentiation capabilities in both true and simulated microgravity conditions211,212. Specifically, in spaceflight-induced microgravity mouse embryonic stem cells (ESCs) appear to maintain proliferative functions while failing to express genes required for germ layer differentiation212,213. In simulated microgravity experiments with ESCs, results are ambiguous, with some studies reporting increased differentiation, while other studies report decreased adhesion and cell death. Parabolic flight is another method that has been used to simulate microgravity, with an added hypergravity component. Mouse ESCs exposed to parabolic flight showed significant alterations to gene expression, including cell proliferation, apoptosis and transforming growth factor-beta (TGF- β) signaling214. These cells also demonstrated increased differentiation into cardiomyocyte colonies following parabolic flight, as has been shown in several studies on the Space Shuttle212,214.
Maintenance of stemness during microgravity exposure has also been demonstrated in several other stem cell populations including cardiovascular progenitor cells, mesenchymal stem cells (MSCs), hematopoietic (HSCs), and adipose derived stem cells. Specifically, studies using neonatal and adult cardiovascular progenitor cells exposed to microgravity on ISS and simulated microgravity in a clinostat resulted in altered cytoskeletal organization and migration in both cell populations215,216. Several of these responses were found to be regulated by miRNAs, thereby indicating that miRNAs may be a key mediator of the cellular response to spaceflight exposure215–217. MicroRNAs (miRNAs) are highly conserved non-coding RNA molecules that are involved in post-transcriptional regulation of gene expression. They function via base-pairing with complementary strands of mRNA, in turn silencing them by cleavage, destabilization, or hindering translation. Furthermore, the authors found reduced yes-associated protein 1 (YAP1) and Tafazzin (TAZ) signaling that can function to regulate transcription and is affected by mechanical load217. Neonatal but not adult cardiovascular progenitor cells exposed to spaceflight exhibited increased expression of markers for early cardiovascular development and enhanced proliferative potential, possibly mediated through miRNA signaling215,216.
YAP and TAZ signaling also play an important role in MSCs, resulting in the expression of Runx2 and initiation of osteogenesis. The effect of microgravity on MSCs has been widely studied. Studies investigating YAP and TAZ signaling under simulated microgravity have shown inhibition of osteogenic differentiation by MSCs due to lack of TAZ nuclear translocation218,219.
Some of the most well-documented changes to MSCs are morphological and include alterations to the cytoskeletal architecture, adhesion properties and migration patterns. In simulated microgravity studies, hMSCs were found to be flatter with disruption of the actin cytoskeleton, redistribution for vinculin and increased integrin expression220,221. Some reports have indicated increased proliferation of hMSCs cultured under simulated microgravity while others have reported decreased proliferation with reduced expression of cell cycle related genes during simulated microgravity and true spaceflight exposure222,223. Furthermore, hMSC cultures subjected to unloading via clinorotation have been shown to exhibit preservation of their dedifferentiated state as well as successful trans-differentiation222,224. These studies highlight the differences between culture systems and the need for standardized methods to assess the role of microgravity on cell and stem cell functions.
Several investigators have confirmed the inhibition of stem cell differentiation in vivo, as expressed in the osteochondrogenic precursor stem cell lineage in mouse models225. Mice exposed to spaceflight on the BionM1 capsule demonstrated reduced MSC commitment and corresponding increase in differentiation of the MSC lineage into osteoblasts and adipocytes following reloading226. Further experimentation of microgravity-induced altered gene expression suggests miR-132–3p as a potential inhibitor of osteoblastic differentiation225. Induction of cellular senescence is another potential contributor to the permanent proliferative state incurred by many cell populations under microgravity simulations. Minimal research has been conducted to elucidate the specific molecular modulators involved in spaceflight-induced cellular senescence; however transcriptomic analysis of space-flown mice suggests a p53-independent induction of the p21 senescence pathway227. Nevertheless, further experimentation is needed to fully elucidate these molecular pathways. Another interesting phenomenon is the change in lineage commitment of MSCs from the osteogenic lineage to adipogenic lineages similar to that which occurs during aging228. Several studies conducted using directed differentiation of MSCs under simulated microgravity have shown a preferential commitment to the adipogenic lineage through a variety of mechanisms including suppression of microfilament formation and RhoA activity229.
The hematopoietic system is of critical importance for immune function and red blood cell production, while also contributing to the maintenance of the skeletal system. Several studies have investigated alterations to the immune system and indicated that spaceflight results in shifts in immune cell phenotypes, reduction in circulating T cells, and decreased T cell activation, decrease in B cell numbers, and decreased numbers and cytotoxicity of NK cells230–233. As HSCs are the primary contributors of erythropoiesis, myelopoiesis, and lymphopoiesis234, it is critical to understand the effects of microgravity on basic HSC function and correlate these changes to altered immune functions observed during spaceflight.
Studies investigating hematopoietic lineage cells in astronauts found decreases in NK cells and reticulocytes following short duration spaceflight234. Post-flight assessment of astronauts indicate deficiencies in thymopoiesis235, a reduction in erythroid and myeloid progenitor cells236, and altered whole-blood transcriptomic and metabolomic profiles237. Interestingly HLU studies found significant changes to bone marrow HSC populations including increases in long-term HSCs (LT-HSCs), short-term HSCs (ST-HSCs), multipotent progenitor (MPP) cells and neutrophils but observed decreases in B-lineage cells, NK cells and erythrocytes. No recovery in HSCs and neutrophils following a 28-d reloading period was observed and repopulation assays showed significant impairment of the reconstitution ability of HSCs exposed to HLU234. Interestingly, significant alterations in myeloid colony forming units was also observed in mice following exposure to spaceflight on the BionM1 capsule238. Mice flown on the BionM1 capsule demonstrated a decrease in hematopoietic cells and a suppression of primitive multipotent progenitors that were not found to be reversible during a 7-day recovery period226. Human HSCs exposed to simulated microgravity have shown impairment of DNA damage repair mechanisms and accumulation of double stranded DNA breaks239. The differentiation capacity of these cells into dendritic cells was also impaired239. These studies highlight the reduced hematopoietic functions of HSCs following spaceflight and may demonstrate that altered immune responses in response to spaceflight exposure may originate, in part from deficits to HSC function.
The effects of microgravity on neural precursor cells has also been gaining interest in recent years. Several studies have been conducted using simulated microgravity, including differentiation studies investigating the role of microgravity on human ESC-derived neural organoids240. Organoids exposed to simulated microgravity exhibited deficits in expression of forebrain markers indicating that microgravity directs differentiation towards caudal neural progenitors240. Interesting, as demonstrated in other stem cell populations, Wnt signaling may be a key signaling mechanism involved in this response240.
Overall, studies investigating the impact of microgravity on stem cell function suggest that microgravity critically affects basic cell functions including alterations to cell morphology, cytoskeletal organization, gene expression profiles, differentiation and apoptosis related pathways218,223,232,241,242, contributing to the growing concern that the health of both stem cells and terminally differentiated lineage populations is threatened during spaceflight exposure. Findings that highlight the inability of stem cells to differentiate towards certain lineages emphasize the detrimental effect of microgravity on tissue regenerative processes and the need to develop therapeutic countermeasures before long-duration spaceflight missions are attempted.
Radiation-induced Stem Cell Dysfunction:
The effect of cosmic radiation on stem cell function is not well understood and studies have focused primarily on MSC/HSC populations. Multiple studies demonstrate a notable impact on primitive and differentiated hematopoietic cell lines after GCR/solar energetic particles (SEP) radiation exposure243,244. The progenitors of erythroid, T-lymphocytes, and B-lymphocyte lineages were seen to demonstrate radiosensitivity244, highlighting the potential of radiation exposure to destabilize immune function and efficiency. Whole-body blood cell counts after proton radiation confirm these findings indicating decreased amounts of white blood cells, lymphocytes, and neutrophils up to 100 days post-irradiation244. Additionally, HSCs may be affected through their corresponding microenvironment, influenced by adjacent irradiated cell populations through the bystander effect. Coculture experiments of unirradiated HSC and irradiated MSC mixtures show that HSC populations experience indirect effects of GCR/SEP radiation of MSCs along with the previously established direct effects resulting from HSC irradiation243. These results clearly display the concept that microenvironmental changes in the bone marrow induced by radiation exposure significantly impact bone marrow progenitor cells both directly and indirectly.
Several studies investigated the neurogenic risks of simulated space radiation environments, emphasizing a significant decrease in neural stem and progenitor cells245,246. Neurogenesis is required for cognitive strength, mood regulation, and neurological stability, thereby stressing the importance of characterizing any cosmic radiation-induced effects on neural stem cells (NSCs)247. While both NSCs and proliferating neural progenitor cells were negatively affected by cosmic radiation, quiescent NSCs were found to be particularly vulnerable compared to their rapidly dividing progeny245. This suggests that mechanisms unrelated to cellular replication may be impacted by GCR, such as oxidative stress, and implies long-term negative effects to stem cell populations and their differentiated progeny. HZE of GCR are shown to be directly associated with mitochondrial dysfunction and ROS production, causing damage and death of affected cells247. NSCs in the hippocampal region may be especially susceptible to cosmic radiation-induced oxidative damage, attenuating neurogenesis and impairing cognitive function. Furthermore, murine cerebral exposure to heavy ion radiation with high linear energy transfer (high-LET) results in a premature aging phenotype through accelerated p21-mediated neuronal cell senescence246. This phenomenon is also evident among other somatic stem cells, including endothelial progenitor cells, osteochondral progenitor cells, and lung progenitor cells248. These findings report the neurogenic risks of spaceflight radiation exposure and identify the need for further research into adult stem cell populations and the propagated tissue-based effects.
GCR-induced alterations in ESC functions have not been studied in great detail. A genomic analysis of murine ESCs during spaceflight reveals suppression of global gene expression and increased spontaneous chromosomal aberrations249. These genomic defects may contribute to increased ESC dysfunction or reprogramming. Furthermore, since ESCs are vital for repairing diseased tissue, loss of ESC stemness or differentiation abilities can adversely impact tissue regeneration and wound healing.
Combined Effects of Radiation and Microgravity on Stem Cells:
Stem cell regulation has been defined for individual spaceflight stressors (i.e. microgravity and radiation); however one of the fundamental questions to be answered in regenerative space biology is whether these stressors have synergistic effects on progenitor populations. Several studies describing tissue-level compounded effects by microgravity and radiation in the brain, retina, and skeleton have been reported250,251. It is possible that these tissue-based effects stem from aberrations in progenitor cell populations. Research conducted using gamma and heavy ion radiation demonstrated deficits in osteoblastogenesis due to increased oxidative stress in progenitor populations78,96,252. Additionally, as previously noted in Section 2, combined exposure of mice to HLU and radiation demonstrated depletion of progenitor populations that were rescued by administration of an antioxidant rich diet, the dried plum diet94. Recent research details that microgravity-induced osteoclast progenitor cell counts are modulated by space-standardized radiation251.
Other studies investigate the compounded effects of microgravity and proton irradiation on immune function and leukocyte activity. Lymphocytes are considered one of the most sensitive mammalian blood cell types to ionizing radiation exposure; thus it is important to characterize any microgravity-associated augmentation to this sensitivity. One study utilized the HLU model and solar particle event-like proton radiation to conclude that spaceflight factors do have a synergistic effect on the proliferation rate and activation rate of isolated splenic T lymphocytes. They found that combining HLU with proton irradiation significantly suppresses splenic T lymphocyte activation 21 days post exposure while diminishing the proliferation rate77. However, results are controversial, as another study reports no interplay between radiation and rotating wall vessel-simulated microgravity in human lymphocytes253.
Several research groups suggest that miRNAs may contribute to the regulation of compounded spaceflight effects254. Certain miRNAs may be differentially expressed depending on the combination of spaceflight factors inflicted. One such group demonstrates this by analyzing miRNA transcription profiles in peripheral blood lymphocytes exposed to simulated spaceflight. They conclude that simulated microgravity modulated the effect of radiation exposure by downregulating radiosensitive miRNAs, including important promoters of osteoblastic stem cell differentiation (miRNA-144), iPSC generation (miRNA-200a), and embryonic stem cell differentiation (miRNA-7)255. Lastly, research involving the effects of radiation and microgravity on human lymphoblastoid TK6 cells reveal no differentially expressed miRNAs under isolated factors. However, an interactive effect between the spaceflight conditions is exhibited, resulting in differentially expressed miRNA-15b and miRNA-221, both of which promotes stemness and tumorgenicity of cancer cells256. Taken together, research involving the interplay of radiation and spaceflight in stem cell functionality indicates a synergistic relationship, compounding the effects seen by each isolated factor.
Additional and Future Considerations:
Due to the extensive applications and potential benefits offered by stem cells therapies, research into these populations has increased over the past 10 years; however, we have only recently begun characterizing the effect of spaceflight stressors on stem cell populations and tissue regeneration. A majority of these studies have investigated alterations to HSCs or MSCs, leaving studies into tissue-specific and ESCs during spaceflight lacking257. Additionally, availability of true-spaceflight research focused on stem cell populations is limited due to limited resources and technology, therefore as noted researches must rely on ground-based models. As noted, these models fail to encompass all of the stressors and physiological effects associated with true-spaceflight, including full-body mechanical unloading, changes in fluid dynamics and systemic alterations258. As a result, research conducted to establish the compounded effects of microgravity, radiation, and other spaceflight-associated environmental changes on stem cell function are needed.
Recent developments in research programs led by the ISS National Laboratory in collaboration with National Institutes of Health (NIH) and National Science Foundation (NSF), have expanded the capacity for stem cell research to be conducted in space. Specifically, these programs have increased interest in Earth-based applications and have specifically called for the use of micro-physiological systems and organ-based cultures in space. These programs, and foundational studies characterizing the maintenance of stem cell properties in populations exposed to microgravity conditions, have presented a unique opportunity for the use of stem cell populations cultured under microgravity conditions for tissue engineering and regenerative medicine applications. This has been explored in multiple paradigms, including the use of cranial-derived MSCs as a potential cell therapy for the CNS259, and is reviewed extensively elsewhere258,260. Although stem cells have been shown in multiple studies to retain their stem cell characteristics and exhibit delayed differentiation, there are also many other changes that have been observed related to microgravity exposure261,262. These alterations may result in unintended consequences from using stem cell populations cultured under microgravity conditions in cell-based therapies.
As we start to explore the Solar System beyond LEO, from the Moon to Mars, expansion of studies in LEO investigating the effects of microgravity on stem cell function to the combined effects of microgravity and space radiation will be critical for understanding tissue regeneration capabilities. Maintenance of stemness in microgravity also poses a significant risk for the maintenance of tissue homeostasis, and for tissue repair and regeneration during long-duration spaceflight exposure. Exposure of radio-sensitive stem cell populations to deep-space radiation poses a significant concern for permanent and irreversible damage to stem cell populations while also posing a risk for stem cell depletion. Accumulation of DNA damage in stem cell populations that are activated upon stimulation, is also a cause for concern. However, in-depth research into the effects of combined spaceflight stressors on the numerous stem cell populations found in mammalian organisms is lacking. Furthermore, studies using whole animals and human cell populations (such as iPSCs) are also lacking; resulting in an incomplete picture of the effects of combined spaceflight stressors on stem cell populations. Although several studies have touched on the molecular mechanisms driving the observed changes, an in-depth understanding of the local and systemic mechanisms are still to be elucidated. Therefore, as new opportunities for spaceflight experiments are explored, it is critical to pursue the use of spaceflight for Earth-based benefits (including understanding basic stem cell function in addition to exploration of clinical applications) in addition to research enabling human exploration.
7. CONCLUSIONS AND ADDITIONAL CONSIDERATIONS REGARDING THE HLU MODEL
In LEO, the influence of microgravity on physiological systems is the most prominent spaceflight factor. Due to limited access to space, many studies have been conducted utilizing models of microgravity, including HLU in rodents, and simulation devices based on randomizing the gravity vector, or subjecting cell masses to consistent free fall conditions. Specific analysis of simulation devices is beyond the scope of this review; however, it is important to note that the utility of these devices to simulate microgravity is still debated223, partially due to the question of whether individual mammalian cells have the ability to sense gravity, or whether they sense their mechanical environment (which is altered by gravity) instead. Although these methods of simulating microgravity during spaceflight present unique drawbacks and limitations, they all generate results indicating that microgravity-facilitated mechanical unloading alters stem cell behavior and activity. This highlights the need to continue, and expand upon, studies focused on basic stem cell function under true microgravity conditions, in order to elucidate the potential physiological effects that these changes may have during long-duration spaceflight212,263–265.
Limitations exist regarding the use of HLU. Hind limb unloading via tail suspension is a good instrument to study bone demineralization associated with microgravity in rats and mice. In rats, HLU leads to many of the cardiovascular changes that occur in humans during spaceflight, including the cephalic fluid shift266,267. HLU invokes many of the changes in blood vessel structure and region-specific changes in blood flow that are seen in rats during space flight189,268 and also results in changes in the ultrastructure of the choroid plexus and CSF production that closely resemble those seen in rats that have been in space flight269,270. Factors such as altered neuroendocrine functions, behavioral deficits, and increased stress levels271,272 should be considered when designing studies and interpreting results. Stress is clearly a feature that astronauts will face during spaceflight), though astronauts typically have a high tolerance for stress. Porphyria, which is a sign of distress in laboratory rodents273 is a common, though transient, trait of rodents during HLU procedure28. HLU models can increase stress response as indicated by elevated serum corticosterone that is associated with atrophy of lymphoid organs36,274–276, although these responses are not always consistent. A primary technical report detailing aspects regarding HLU28 recommends housing single rodents at between 24.5C-25.5°C to mitigate any toxicity profiles (e.g., bone loss, impaired ability to thermoregulate31,277). Moreover, based on experience of all of the authors, attrition of rats from HLU studies (e.g., sudden loss of rodents during the study, excessive porphyria, or persistent weight loss) occurs more frequently if housing is below this temperature range.
With a few exceptions34,80, studies involving HLU generally involve isolating rodents via single housing because of jig constraints35,278, which could impact immune or stress responses36. Moreover, single housing in social species like rodents can profoundly alter physiology. For example, isolation can lead to anxiety279, impairments in memory280, upregulation of neuroendocrine responses to stress281–283,284 and altered immunity285,286. As flight studies typically involve group housed mice vs single housed conditions, one group has performed a side by side comparison of HLU under single and socially-housed conditions36. Results from this study reveal that deficits in skeletal structure typically attributed to HLU generally occur at a similar magnitude in single and socially-housed animals. This suggests that bone loss as observed in the HLU model results primarily from gravity-dependent mechanisms. In contrast, some immune system responses to HLU are differentially impacted by the social environment. Moreover, isolation impacts neural systems that mediate mission critical cognitive domains. For example, isolation has been observed to reduce makers of neuroplasticity in the rodent hippocampus and prefrontal cortex287–289. In addition, isolation has been observed to increase markers of neuroinflammation and cell loss in these brain structures287,290,291, increases redox stress, proinflammatory tumor necrosis factor α (TNFα) levels (in the hippocampus)290, and reduces Parvalbumin (PV)+ interneurons in CA2 and CA3292. Thus, factors such as room temperature, social housing, and stress responses should be considered when performing and interpreting individual and combined hazard investigations, such as HLU + space radiation.
While there are many facsimiles of flight stressors that can be used in rodents to mimic the neurological and psychological impacts seen in humans, modeling microgravity effects on the CNS using HLU is problematic. As noted, microgravity is documented to impact many aspects of mammalian physiology, including the skeletal, microbiome, gut mucosa, sensorimotor and ocular systems (e.g.,28,76,117,293–295). All of these are likely to have some to major impacts on the brain, either directly or indirectly, and interact with the sequelae from other flight stressors. Obviously, with accepted limitations, there are no ground-based systems that can be used on rodents to fully simulate microgravity. As noted, simulated microgravity145 results in persistent changes in the mitochondrial function and lipid metabolism within oligodendrocytes. While clinostats seem to do a reasonable job of simulating microgravity effects in oligodendrocytes145 no such devices are readily available for rodents.
Thus while HLU may induce some of the physical and phenotypic changes associated with space flight, it remains unclear if HLU induces similar signaling and molecular changes (e.g. increase in fatty acid metabolism that will impact neuronal function and thus cognition) as with microgravity. Moreover, whatever responses are induced as a result of the HLU, these occur in an animal that can be assumed to be under stress – although this in itself is a characteristic response in astronauts during spaceflight. The impact of such stress may invoke compensatory mechanisms that may, or may not, occur in humans while in space. Thus as noted, limitations should be considered with use of the HLU model. While PWB represents an alternative model and offers the possibility to investigate a wider range of mechanical loads, more studies are needed to investigate the response of multiple organ systems to reduced weight-bearing. For example, radiations have only been administered in PWB female mice, and more studies are needed to assess the benefits of this model to radiation research in larger outbred rodents. Further studies should also more completely profile the stress response resulting from PWB.
ACKNOWLEDGEMENTS
This project was supported by: Willey – NASA Space Biology grant # NNX15AB50G, NIH/NIAMS 1R21AR072806-01, and the Translational Research Institute for Space Health through NASA NNX16AO69A for award EO005. Britten – NASA grant #s NNX16AC40G, NNX15AI22G, and NNX14AE73G. Mortreux – NASA grant #s NNX16AL36G and 80NSSC19K1598. Delp - NASA grant #s NNX16AC28G, NNX15AE86G, 80NSSC19K1322, and 80NSSC19K1599. Mao - NASA Space Biology grant # NNX15AB41G, NASA 80NSSC18K0310, 80NSSC20K0986 and LLU Department of Basic Sciences.
REFERENCES
- 1.Walb MC, Black PJ, Payne VS, Munley MT, Willey JS. A reproducible radiation delivery method for unanesthetized rodents during periods of hind limb unloading. Life Sci Space Res (Amst). 2015;6:10–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Tahimic CGT, Globus RK. Redox Signaling and Its Impact on Skeletal and Vascular Responses to Spaceflight. Int J Mol Sci. 2017;18(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Stepanek J, Blue RS, Parazynski S. Space Medicine in the Era of Civilian Spaceflight. N Engl J Med. 2019;380(11):1053–1060. [DOI] [PubMed] [Google Scholar]
- 4.Kiffer F, Boerma M, Allen A. Behavioral effects of space radiation: A comprehensive review of animal studies. Life Sci Space Res (Amst). 2019;21:1–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Blue RS, Chancellor JC, Suresh R, et al. Challenges in Clinical Management of Radiation-Induced Illnesses During Exploration Spaceflight. Aerosp Med Hum Perform. 2019;90(11):966–977. [DOI] [PubMed] [Google Scholar]
- 6.da Silveira WA, Fazelinia H, Rosenthal SB, et al. Comprehensive Multi-omics Analysis Reveals Mitochondrial Stress as a Central Biological Hub for Spaceflight Impact. Cell. 2020;183(5):1185–1201 e1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Weber J, Javelle F, Klein T, et al. Neurophysiological, neuropsychological, and cognitive effects of 30 days of isolation. Exp Brain Res. 2019;237(6):1563–1573. [DOI] [PubMed] [Google Scholar]
- 8.Petit G, Cebolla AM, Fattinger S, et al. Local sleep-like events during wakefulness and their relationship to decreased alertness in astronauts on ISS. NPJ Microgravity. 2019;5:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Pendergraft JG, Carter DR, Tseng S, Landon LB, Slack KJ, Shuffler ML. Learning From the Past to Advance the Future: The Adaptation and Resilience of NASA’s Spaceflight Multiteam Systems Across Four Eras of Spaceflight. Front Psychol. 2019;10:1633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Basner M, Dinges DF, Mollicone DJ, et al. Psychological and behavioral changes during confinement in a 520-day simulated interplanetary mission to mars. PloS one. 2014;9(3):e93298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mairesse O, MacDonald-Nethercott E, Neu D, et al. Preparing for Mars: human sleep and performance during a 13 month stay in Antarctica. Sleep. 2019;42(1). [DOI] [PubMed] [Google Scholar]
- 12.Gonfalone A Sleep on manned space flights: Zero gravity reduces sleep duration. Pathophysiology. 2016;23(4):259–263. [DOI] [PubMed] [Google Scholar]
- 13.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):535–544. [DOI] [PubMed] [Google Scholar]
- 14.Lang T, Van Loon J, Bloomfield S, et al. Towards human exploration of space: the THESEUS review series on muscle and bone research priorities. NPJ Microgravity. 2017;3:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Lloyd SA, Morony SE, Ferguson VL, et al. Osteoprotegerin is an effective countermeasure for spaceflight-induced bone loss in mice. Bone. 2015;81:562–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Shirazi-Fard Y, Metzger CE, Kwaczala AT, Judex S, Bloomfield SA, Hogan HA. Moderate intensity resistive exercise improves metaphyseal cancellous bone recovery following an initial disuse period, but does not mitigate decrements during a subsequent disuse period in adult rats. Bone. 2014;66:296–305. [DOI] [PubMed] [Google Scholar]
- 17.Smith RC, Cramer MS, Mitchell PJ, et al. Inhibition of myostatin prevents microgravity-induced loss of skeletal muscle mass and strength. PloS one. 2020;15(4):e0230818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Burkhart K, Allaire B, Bouxsein ML. Negative Effects of Long-duration Spaceflight on Paraspinal Muscle Morphology. Spine (Phila Pa 1976). 2019;44(12):879–886. [DOI] [PubMed] [Google Scholar]
- 19.DeLong A, Friedman MA, Tucker SM, et al. Protective Effects of Controlled Mechanical Loading of Bone in C57BL6/J Mice Subject to Disuse. JBMR Plus. 2020;4(3):e10322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Vico L, Hargens A. Skeletal changes during and after spaceflight. Nat Rev Rheumatol. 2018;14(4):229–245. [DOI] [PubMed] [Google Scholar]
- 21.Keyak JH, Koyama AK, LeBlanc A, Lu Y, Lang TF. Reduction in proximal femoral strength due to long-duration spaceflight. Bone. 2009;44(3):449–453. [DOI] [PubMed] [Google Scholar]
- 22.Belavy DL, Adams M, Brisby H, et al. Disc herniations in astronauts: What causes them, and what does it tell us about herniation on earth? Eur Spine J. 2016;25(1):144–154. [DOI] [PubMed] [Google Scholar]
- 23.Johnston SL, Campbell MR, Scheuring R, Feiveson AH. Risk of herniated nucleus pulposus among U.S. astronauts. Aviat Space Environ Med. 2010;81(6):566–574. [DOI] [PubMed] [Google Scholar]
- 24.Bailey JF, Miller SL, Khieu K, et al. From the international space station to the clinic: how prolonged unloading may disrupt lumbar spine stability. Spine J. 2018;18(1):7–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Chang DG, Healey RM, Snyder AJ, et al. Lumbar Spine Paraspinal Muscle and Intervertebral Disc Height Changes in Astronauts After Long-Duration Spaceflight on the International Space Station. Spine (Phila Pa 1976). 2016;41(24):1917–1924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Shen M, Frishman WH. Effects of Spaceflight on Cardiovascular Physiology and Health. Cardiol Rev. 2019;27(3):122–126. [DOI] [PubMed] [Google Scholar]
- 27.Lee AG, Mader TH, Gibson CR, et al. Spaceflight associated neuro-ocular syndrome (SANS) and the neuro-ophthalmologic effects of microgravity: a review and an update. NPJ Microgravity. 2020;6:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Morey-Holton ER, Globus RK. Hindlimb unloading rodent model: technical aspects. J Appl Physiol (1985). 2002;92(4):1367–1377. [DOI] [PubMed] [Google Scholar]
- 29.Willey JS, Kwok AT, Moore JE, et al. Spaceflight-Relevant Challenges of Radiation and/or Reduced Weight Bearing Cause Arthritic Responses in Knee Articular Cartilage. Radiat Res. 2016;186(4):333–344. [DOI] [PubMed] [Google Scholar]
- 30.Knox M, Fluckey JD, Bennett P, Peterson CA, Dupont-Versteegden EE. Hindlimb unloading in adult rats using an alternative tail harness design. Aviat Space Environ Med. 2004;75(8):692–696. [PubMed] [Google Scholar]
- 31.Farley A, Gnyubkin V, Vanden-Bossche A, 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):170–179. [DOI] [PubMed] [Google Scholar]
- 32.Colaianni G, Mongelli T, Cuscito C, et al. Irisin prevents and restores bone loss and muscle atrophy in hind-limb suspended mice. Sci Rep. 2017;7(1):2811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wang J, Wang X, Feng W. Reloading Promotes Recovery of Disuse Muscle Loss by Inhibiting TGFbeta Pathway Activation in Rats After Hind Limb Suspension. Am J Phys Med Rehabil. 2017;96(6):430–437. [DOI] [PubMed] [Google Scholar]
- 34.Lloyd SA, Lang CH, Zhang Y, et al. Interdependence of muscle atrophy and bone loss induced by mechanical unloading. J Bone Miner Res. 2014;29(5):1118–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lloyd SA, Bandstra ER, Willey JS, et al. Effect of proton irradiation followed by hindlimb unloading on bone in mature mice: a model of long-duration spaceflight. Bone. 2012;51(4):756–764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Tahimic CGT, Paul AM, Schreurs AS, et al. Influence of Social Isolation During Prolonged Simulated Weightlessness by Hindlimb Unloading. Front Physiol. 2019;10:1147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chowdhury P, Long A, Harris G, Soulsby ME, Dobretsov M. Animal model of simulated microgravity: a comparative study of hindlimb unloading via tail versus pelvic suspension. Physiol Rep. 2013;1(1):e00012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Summers SM, Hayashi Y, Nguyen SV, Nguyen TM, Purdy RE. Hindlimb unweighting induces changes in the p38MAPK contractile pathway of the rat abdominal aorta. J Appl Physiol (1985). 2009;107(1):121–127. [DOI] [PubMed] [Google Scholar]
- 39.Globus RK, Morey-Holton E. Hindlimb unloading: rodent analog for microgravity. J Appl Physiol (1985). 2016;120(10):1196–1206. [DOI] [PubMed] [Google Scholar]
- 40.Piet J, Hu D, Baron R, Shefelbine SJ. Bone adaptation compensates resorption when sciatic neurectomy is followed by low magnitude induced loading. Bone. 2019;120:487–494. [DOI] [PubMed] [Google Scholar]
- 41.Ma X, Lv J, Sun X, et al. Naringin ameliorates bone loss induced by sciatic neurectomy and increases Semaphorin 3A expression in denervated bone. Sci Rep. 2016;6:24562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bateman TA, Dunstan CR, Lacey DL, Ferguson VL, Ayers RA, Simske SJ. Osteoprotegerin ameliorates sciatic nerve crush induced bone loss. J Orthop Res. 2001;19(4):518–523. [DOI] [PubMed] [Google Scholar]
- 43.Vegger JB, Nielsen ES, Bruel A, Thomsen JS. Additive effect of PTH (1–34) and zoledronate in the prevention of disuse osteopenia in rats. Bone. 2014;66:287–295. [DOI] [PubMed] [Google Scholar]
- 44.Khajuria DK, Disha C, Razdan R, Mahapatra DR, Vasireddi R. Prophylactic Effects of Propranolol versus the Standard Therapy on a New Model of Disuse Osteoporosis in Rats. Sci Pharm. 2014;82(2):357–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.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):350–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Semple C, Riveros D, Nagy JA, Rutkove SB, Mortreux M. Partial Weight-Bearing in Female Rats: Proof of Concept in a Martian-Gravity Analog. Front Physiol. 2020;11:302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Mortreux M, Riveros D, Bouxsein ML, Rutkove SB. Mimicking a Space Mission to Mars Using Hindlimb Unloading and Partial Weight Bearing in Rats. J Vis Exp. 2019(146). [DOI] [PubMed] [Google Scholar]
- 48.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):175–182. [DOI] [PubMed] [Google Scholar]
- 49.Mortreux M, Rosa-Caldwell ME. Approaching Gravity as a Continuum Using the Rat Partial Weight-Bearing Model. Life (Basel). 2020;10(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Mortreux M, Ko FC, Riveros D, Bouxsein ML, Rutkove SB. Longitudinal time course of muscle impairments during partial weight-bearing in rats. NPJ Microgravity. 2019;5:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Swift JM, Lima F, Macias BR, et al. Partial weight bearing does not prevent musculoskeletal losses associated with disuse. Medicine and science in sports and exercise. 2013;45(11):2052–2060. [DOI] [PubMed] [Google Scholar]
- 53.Mortreux M, Riverosa D, Semple C, Bouxsein ML, Rudkove SB. The partial weight-bearing rat model using a pelvic harness does not impact stress or hindlimb blood flow. Acta Astronautica. 2000;168(March 2020):249–255. [Google Scholar]
- 54.Mortreux M, Riveros D, Bouxsein ML, Rutkove SB. A Moderate Daily Dose of Resveratrol Mitigates Muscle Deconditioning in a Martian Gravity Analog. Front Physiol. 2019;10:899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Semple C, Riveros D, Sung DM, Nagy JA, Rutkove SB, Mortreux M. Using Electrical Impedance Myography as a Biomarker of Muscle Deconditioning in Rats Exposed to Micro- and Partial-Gravity Analogs. Front Physiol. 2020;11:557796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Chancellor J, Scott G, Sutton J. Space radiation: the number one risk to astronaut health beyond low earth orbit. Life. 2014;4(3):491–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Chancellor JC, Blue RS, Cengel KA, et al. Limitations in predicting the space radiation health risk for exploration astronauts. npj Microgravity. 2018;4(1):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Cucinotta FA, Kim M-HY, Ren L. Evaluating shielding effectiveness for reducing space radiation cancer risks. Radiation Measurements. 2006;41(9–10):1173–1185. [Google Scholar]
- 59.Cucinotta FA, Schimmerling W, Wilson JW, et al. Space radiation cancer risk projections for exploration missions: uncertainty reduction and mitigation. NASA JSC-29295. 2001:4–75. [Google Scholar]
- 60.Cucinotta FA, Durante M. Cancer risk from exposure to galactic cosmic rays: implications for space exploration by human beings. The lancet oncology. 2006;7(5):431–435. [DOI] [PubMed] [Google Scholar]
- 61.Chancellor JC, Guetersloh SB, Blue RS, Cengel KA, Ford JR, Katzgraber HG. Targeted Nuclear Spallation from Moderator Block Design for a Ground-Based Space Radiation Analog. arXiv preprint arXiv:170602727. 2017. [Google Scholar]
- 62.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(1):1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zhang S, Wimmer-Schweingruber RF, Yu J, et al. First measurements of the radiation dose on the lunar surface. Science Advances. 2020;6(39):eaaz1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.NCRP. Guidance on radiation received in space activities. NCRP Report No. 98; 1988.
- 65.Wilson JW, Cucinotta F, Shinn J, et al. Shielding from solar particle event exposures in deep space. Radiation measurements. 1999;30(3):361–382. [DOI] [PubMed] [Google Scholar]
- 66.Kim M, Wilson J, Cucinotta F, et al. Contribution of high charge and energy (HZE) Ions during solar-particle event of September 29. 1989.
- 67.Kim M-HY, De Angelis G, Cucinotta FA. Probabilistic assessment of radiation risk for astronauts in space missions. Acta Astronautica. 2011;68(7–8):747–759. [Google Scholar]
- 68.Hu S, Cucinotta FA. Characterization of the radiation-damaged precursor cells in bone marrow based on modeling of the peripheral blood granulocytes response. Health physics. 2011;101(1):67–78. [DOI] [PubMed] [Google Scholar]
- 69.Cucinotta FA, Durante M. Risk of radiation carcinogenesis. Human health and performance risks of space exploration missions NASA SP-2009–3405 Houston: National Aeronautics and Space Administration. 2009:119–170. [Google Scholar]
- 70.NCRP. Information needed to make radiation protection recommendations for space missions beyond low-earth orbit. 2006.
- 71.Mao XW, Boerma M, Rodriguez D, 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):241–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Krause AR, Speacht TL, Zhang Y, Lang CH, Donahue HJ. Simulated space radiation sensitizes bone but not muscle to the catabolic effects of mechanical unloading. PloS one. 2017;12(8):e0182403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Prisby RD, Alwood JS, Behnke BJ, et al. Effects of hindlimb unloading and ionizing radiation on skeletal muscle resistance artery vasodilation and its relation to cancellous bone in mice. J Appl Physiol (1985). 2016;120(2):97–106. [DOI] [PubMed] [Google Scholar]
- 74.Bokhari RS, Metzger CE, Black JM, et al. Positive impact of low-dose, high-energy radiation on bone in partial- and/or full-weightbearing mice. NPJ Microgravity. 2019;5:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Xu D, Zhao X, Li Y, et al. The combined effects of X-ray radiation and hindlimb suspension on bone loss. J Radiat Res. 2014;55(4):720–725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Li M, Holmes V, Zhou Y, et al. Hindlimb suspension and SPE-like radiation impairs clearance of bacterial infections. PloS one. 2014;9(1):e85665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Sanzari JK, Romero-Weaver AL, James G, et al. Leukocyte activity is altered in a ground based murine model of microgravity and proton radiation exposure. PloS one. 2013;8(8):e71757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kondo H, Yumoto K, Alwood JS, et al. Oxidative stress and gamma radiation-induced cancellous bone loss with musculoskeletal disuse. J Appl Physiol (1985). 2010;108(1):152–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Overbey EG, Paul AM, da Silveira WA, 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kwok AT, Moore JE, Rosas S, et al. Knee and Hip Joint Cartilage Damage from Combined Spaceflight Hazards of Low-Dose Radiation Less than 1 Gy and Prolonged Hindlimb Unloading. Radiat Res. 2019;191(6):497–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Yu K, Doherty AH, Genik PC, et al. Mimicking the effects of spaceflight on bone: Combined effects of disuse and chronic low-dose rate radiation exposure on bone mass in mice. Life Sci Space Res (Amst). 2017;15:62–68. [DOI] [PubMed] [Google Scholar]
- 82.Mao XW, Nishiyama NC, Campbell-Beachler M, et al. Role of NADPH Oxidase as a Mediator of Oxidative Damage in Low-Dose Irradiated and Hindlimb-Unloaded Mice. Radiat Res. 2017;188(4):392–399. [DOI] [PubMed] [Google Scholar]
- 83.Ghosh P, Behnke BJ, Stabley JN, et al. Effects of High-LET Radiation Exposure and Hindlimb Unloading on Skeletal Muscle Resistance Artery Vasomotor Properties and Cancellous Bone Microarchitecture in Mice. Radiat Res. 2016;185(3):257–266. [DOI] [PubMed] [Google Scholar]
- 84.Alwood JS, Yumoto K, Mojarrab R, et al. Heavy ion irradiation and unloading effects on mouse lumbar vertebral microarchitecture, mechanical properties and tissue stresses. Bone. 2010;47(2):248–255. [DOI] [PubMed] [Google Scholar]
- 85.Sibonga J, Matsumoto T, Jones J, et al. Resistive exercise in astronauts on prolonged spaceflights provides partial protection against spaceflight-induced bone loss. Bone. 2019;128:112037. [DOI] [PubMed] [Google Scholar]
- 86.Lang T, LeBlanc A, Evans H, Lu Y, Genant H, Yu A. Cortical and trabecular bone mineral loss from the spine and hip in long-duration spaceflight. J Bone Miner Res. 2004;19(6):1006–1012. [DOI] [PubMed] [Google Scholar]
- 87.Lang TF, Leblanc AD, Evans HJ, Lu Y. Adaptation of the proximal femur to skeletal reloading after long-duration spaceflight. J Bone Miner Res. 2006;21(8):1224–1230. [DOI] [PubMed] [Google Scholar]
- 88.Ferreira JA, Crissey JM, Brown M. An alternant method to the traditional NASA hindlimb unloading model in mice. J Vis Exp. 2011(49). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ellman R, Spatz J, Cloutier A, Palme R, Christiansen BA, Bouxsein ML. Partial reductions in mechanical loading yield proportional changes in bone density, bone architecture, and muscle mass. J Bone Miner Res. 2013;28(4):875–885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Nakajima K, Matsunaga S, Morioka T, et al. Effects of unloading by tail suspension on biological apatite crystallite alignment in mouse femur. Dent Mater J. 2020;39(4):670–677. [DOI] [PubMed] [Google Scholar]
- 91.Willey JS, Lloyd SA, Nelson GA, Bateman TA. Space radiation and bone loss. Gravitational and Space Biology. 2011;25(1):15–21. [PMC free article] [PubMed] [Google Scholar]
- 92.Willey JS, Lloyd SA, Nelson GA, Bateman TA. Ionizing Radiation and Bone Loss: Space Exploration and Clinical Therapy Applications. Clin Rev Bone Miner Metab. 2011;9(1):54–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Hamilton SA, Pecaut MJ, Gridley DS, et al. A murine model for bone loss from therapeutic and space-relevant sources of radiation. J Appl Physiol. 2006;101(3):789–793. [DOI] [PubMed] [Google Scholar]
- 94.Steczina S, Tahimic CGT, Pendleton M, et al. Dietary countermeasure mitigates simulated spaceflight-induced osteopenia in mice. Sci Rep. 2020;10(1):6484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Macias BR, Lima F, Swift JM, et al. Simulating the Lunar Environment: Partial Weightbearing and High-LET Radiation-Induce Bone Loss and Increase Sclerostin-Positive Osteocytes. Radiat Res. 2016;186(3):254–263. [DOI] [PubMed] [Google Scholar]
- 96.Yumoto K, Globus RK, Mojarrab R, 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. [DOI] [PubMed] [Google Scholar]
- 97.Schreurs AS, Shirazi-Fard Y, Shahnazari M, et al. Dried plum diet protects from bone loss caused by ionizing radiation. Sci Rep. 2016;6:21343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Kondo H, Searby ND, Mojarrab R, et al. Total-body irradiation of postpubertal mice with (137)Cs acutely compromises the microarchitecture of cancellous bone and increases osteoclasts. Radiat Res. 2009;171(3):283–289. [DOI] [PubMed] [Google Scholar]
- 99.Willey JS, Lloyd SA, Robbins ME, et al. Early increase in osteoclast number in mice after whole-body irradiation with 2 Gy X rays. Radiat Res. 2008;170(3):388–392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Bandstra ER, Thompson RW, Nelson GA, et al. Musculoskeletal Changes in Mice from 20–50 cGy of Simulated Galactic Cosmic Rays. Radiat Res. 2009;172(7):In press. [DOI] [PubMed] [Google Scholar]
- 101.Alwood JS, Tran LH, Schreurs AS, et al. Dose- and Ion-Dependent Effects in the Oxidative Stress Response to Space-Like Radiation Exposure in the Skeletal System. Int J Mol Sci. 2017;18(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.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] [PMC free article] [PubMed] [Google Scholar]
- 104.Oldknow KJ, MacRae VE, Farquharson C. Endocrine role of bone: recent and emerging perspectives beyond osteocalcin. J Endocrinol. 2015;225(1):R1–19. [DOI] [PubMed] [Google Scholar]
- 105.DiGirolamo DJ, Clemens TL, Kousteni S. The skeleton as an endocrine organ. Nat Rev Rheumatol. 2012;8(11):674–683. [DOI] [PubMed] [Google Scholar]
- 106.Guntur AR, Rosen CJ. Bone as an endocrine organ. Endocr Pract. 2012;18(5):758–762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Wei J, Ferron M, Clarke CJ, et al. Bone-specific insulin resistance disrupts whole-body glucose homeostasis via decreased osteocalcin activation. J Clin Invest. 2014;124(4):1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Kode A, Mosialou I, Silva BC, et al. FoxO1 protein cooperates with ATF4 protein in osteoblasts to control glucose homeostasis. J Biol Chem. 2012;287(12):8757–8768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Rached MT, Kode A, Silva BC, et al. FoxO1 expression in osteoblasts regulates glucose homeostasis through regulation of osteocalcin in mice. J Clin Invest. 2010;120(1):357–368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Mosialou I, Shikhel S, Liu JM, et al. MC4R-dependent suppression of appetite by bone-derived lipocalin 2. Nature. 2017;543(7645):385–390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Khrimian L, Obri A, Karsenty G. Modulation of cognition and anxiety-like behavior by bone remodeling. Mol Metab. 2017;6(12):1610–1615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Khrimian L, Obri A, Ramos-Brossier M, et al. Gpr158 mediates osteocalcin’s regulation of cognition. J Exp Med. 2017;214(10):2859–2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Eilenberg W, Stojkovic S, Piechota-Polanczyk A, et al. Neutrophil Gelatinase-Associated Lipocalin (NGAL) is Associated with Symptomatic Carotid Atherosclerosis and Drives Pro-inflammatory State In Vitro. Eur J Vasc Endovasc Surg. 2016;51(5):623–631. [DOI] [PubMed] [Google Scholar]
- 114.Berger JM, Singh P, Khrimian L, et al. Mediation of the Acute Stress Response by the Skeleton. Cell Metab. 2019;30(5):890–902 e898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Lee AG, Mader TH, Gibson CR, Tarver W. Space Flight-Associated Neuro-ocular Syndrome. JAMA Ophthalmol. 2017;135(9):992–994. [DOI] [PubMed] [Google Scholar]
- 116.Mader TH, Gibson CR, Miller NR, Subramanian PS, Patel NB, Lee AG. An overview of spaceflight-associated neuro-ocular syndrome (SANS). Neurol India. 2019;67(Supplement):S206–S211. [DOI] [PubMed] [Google Scholar]
- 117.Mader TH, Gibson CR, Pass AF, et al. Optic disc edema, globe flattening, choroidal folds, and hyperopic shifts observed in astronauts after long-duration space flight. Ophthalmology. 2011;118(10):2058–2069. [DOI] [PubMed] [Google Scholar]
- 118.Huang AS, Stenger MB, Macias BR. Gravitational Influence on Intraocular Pressure: Implications for Spaceflight and Disease. J Glaucoma. 2019;28(8):756–764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Garrett-Bakelman FE, Darshi M, Green SJ, et al. The NASA Twins Study: A multidimensional analysis of a year-long human spaceflight. Science. 2019;364(6436). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Mao XW, Nishiyama NC, Byrum SD, et al. Characterization of mouse ocular response to a 35-day spaceflight mission: Evidence of blood-retinal barrier disruption and ocular adaptations. Sci Rep. 2019;9(1):8215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Nelson ES, Mulugeta L, Myers JG. Microgravity-induced fluid shift and ophthalmic changes. Life (Basel). 2014;4(4):621–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Philpott DE, Corbett R, Turnbill C, et al. Cosmic ray effects on the eyes of rats flown on Cosmos No. 782, experimental K-007. Aviat Space Environ Med. 1978;49(1 Pt 1):19–28. [PubMed] [Google Scholar]
- 123.Taylor CR, Hanna M, Behnke BJ, et al. Spaceflight-induced alterations in cerebral artery vasoconstrictor, mechanical, and structural properties: implications for elevated cerebral perfusion and intracranial pressure. Faseb J. 2013;27(6):2282–2292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Zhang LF, Hargens AR. Spaceflight-Induced Intracranial Hypertension and Visual Impairment: Pathophysiology and Countermeasures. Physiol Rev. 2018;98(1):59–87. [DOI] [PubMed] [Google Scholar]
- 125.Taibbi G, Cromwell RL, Kapoor KG, Godley BF, Vizzeri G. The effect of microgravity on ocular structures and visual function: a review. Surv Ophthalmol. 2013;58(2):155–163. [DOI] [PubMed] [Google Scholar]
- 126.Zhao D, He Z, Vingrys AJ, Bui BV, Nguyen CT. The effect of intraocular and intracranial pressure on retinal structure and function in rats. Physiol Rep. 2015;3(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Patel N, Pass A, Mason S, Gibson CR, Otto C. Optical Coherence Tomography Analysis of the Optic Nerve Head and Surrounding Structures in Long-Duration International Space Station Astronauts. JAMA Ophthalmol. 2018;136(2):193–200. [DOI] [PubMed] [Google Scholar]
- 128.Morgan WH, Balaratnasingam C, Lind CR, et al. Cerebrospinal fluid pressure and the eye. Br J Ophthalmol. 2016;100(1):71–77. [DOI] [PubMed] [Google Scholar]
- 129.Taibbi G, Cromwell RL, Zanello SB, et al. Ocular Outcomes Comparison Between 14- and 70-Day Head-Down-Tilt Bed Rest. Invest Ophthalmol Vis Sci. 2016;57(2):495–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Frizziero L, Parrozzani R, Midena G, et al. Hyperreflective Intraretinal Spots in Radiation Macular Edema on Spectral Domain Optical Coherence Tomography. Retina. 2016;36(9):1664–1669. [DOI] [PubMed] [Google Scholar]
- 131.Mayer M, Kaiser N, Layer PG, Frohns F. Cell Cycle Regulation and Apoptotic Responses of the Embryonic Chick Retina by Ionizing Radiation. PloS one. 2016;11(5):e0155093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Toutounchian JJ, Steinle JJ, Makena PS, et al. Modulation of radiation injury response in retinal endothelial cells by quinic acid derivative KZ-41 involves p38 MAPK. PloS one. 2014;9(6):e100210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Vinogradova Iu V, Tronov VA, Liakhova KN, Poplinskaia VA, Ostrovskii MA. [Damage and functional recovery of the mouse retina after exposure to ionizing radiation and methylnitrosourea]. Radiats Biol Radioecol. 2014;54(4):385–392. [PubMed] [Google Scholar]
- 134.Fedirko PA, Babenko TF, Dorichevska RY, Garkava NA. Retinal Vascular Pathology Risk Development in the Irradiated at Different Ages as a Result of Chernobyl Npp Accident. Probl Radiac Med Radiobiol. 2015;20:467–573. [PubMed] [Google Scholar]
- 135.Mao XW, Archambeau JO, Kubinova L, Boyle S, Petersen G, Grove R. Quantification of rat retinal growth and vascular population changes after single and split doses of proton irradiation: translational study using stereology methods. Radiat Res. 2003;160(1):5–13. [DOI] [PubMed] [Google Scholar]
- 136.Mao XW, Boerma M, Rodriguez D, et al. Acute Effect of Low-Dose Space Radiation on Mouse Retina and Retinal Endothelial Cells. Radiat Res. 2018;190(1):45–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Chen HL, Qu LN, Li QD, Bi L, Huang ZM, Li YH. [Simulated microgravity-induced oxidative stress in different areas of rat brain.]. Sheng Li Xue Bao. 2009;61(2):108–114. [PubMed] [Google Scholar]
- 138.Zhang R, Ran HH, Ma J, Bai YG, Lin LJ. NAD(P)H oxidase inhibiting with apocynin improved vascular reactivity in tail-suspended hindlimb unweighting rat. J Physiol Biochem. 2012;68(1):99–105. [DOI] [PubMed] [Google Scholar]
- 139.Yang TB, Zhong P, Qu LN, Yuan YH. [Space flight and peroxidative damage]. Space Med Med Eng (Beijing). 2003;16(6):455–458. [PubMed] [Google Scholar]
- 140.Zhan H, Chen LM, Xin YM, Tang GX, Wen J. Effects of tea polyphenols on cerebral lipid peroxidation, liver and renal functions in rats after repeated +Gz stress. Space Med Med Eng (Beijing). 1999;12(1):1–5. [PubMed] [Google Scholar]
- 141.Overbey EG, da Silveira WA, Stanbouly S, et al. Spaceflight influences gene expression, photoreceptor integrity, and oxidative stress-related damage in the murine retina. Sci Rep. 2019;9(1):13304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Roberts DR, Albrecht MH, Collins HR, et al. Effects of Spaceflight on Astronaut Brain Structure as Indicated on MRI. N Engl J Med. 2017;377(18):1746–1753. [DOI] [PubMed] [Google Scholar]
- 143.Van Ombergen A, Demertzi A, Tomilovskaya E, et al. The effect of spaceflight and microgravity on the human brain. J Neurol. 2017;264(Suppl 1):18–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Cebolla AM, Petieau M, Dan B, Balazs L, McIntyre J, Cheron G. “Cerebellar contribution to visuo-attentional alpha rhythm: insights from weightlessness”. Sci Rep. 2016;6:37824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Espinosa-Jeffrey A, Nguyen K, Kumar S, et al. Simulated microgravity enhances oligodendrocyte mitochondrial function and lipid metabolism. J Neurosci Res. 2016;94(12):1434–1450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Philips T, Rothstein JD. Oligodendroglia: metabolic supporters of neurons. J Clin Invest. 2017;127(9):3271–3280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Whoolery CW, Yun S, Reynolds RP, et al. Multi-domain cognitive assessment of male mice shows space radiation is not harmful to high-level cognition and actually improves pattern separation. Sci Rep. 2020;10(1):2737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Britten RA, Duncan VD, Fesshaye A, Rudobeck E, Nelson GA, Vlkolinsky R. Altered Cognitive Flexibility and Synaptic Plasticity in the Rat Prefrontal Cortex after Exposure to Low (</=15 cGy) Doses of (28)Si Radiation. Radiat Res. 2020;193(3):223–235. [DOI] [PubMed] [Google Scholar]
- 149.Parihar VK, Maroso M, Syage A, et al. Persistent nature of alterations in cognition and neuronal circuit excitability after exposure to simulated cosmic radiation in mice. Exp Neurol. 2018;305:44–55. [DOI] [PubMed] [Google Scholar]
- 150.Parihar VK, Allen BD, Caressi C, et al. Cosmic radiation exposure and persistent cognitive dysfunction. Sci Rep. 2016;6:34774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Hadley MM, Davis LK, Jewell JS, Miller VD, Britten RA. Exposure to Mission-Relevant Doses of 1 GeV/n (48)Ti Particles Impairs Attentional Set-Shifting Performance in Retired Breeder Rats. Radiat Res. 2016;185(1):13–19. [DOI] [PubMed] [Google Scholar]
- 152.Bellone JA, Rudobeck E, Hartman RE, Szucs A, Vlkolinsky R. A Single Low Dose of Proton Radiation Induces Long-Term Behavioral and Electrophysiological Changes in Mice. Radiat Res. 2015;184(2):193–202. [DOI] [PubMed] [Google Scholar]
- 153.Sokolova IV, Schneider CJ, Bezaire M, Soltesz I, Vlkolinsky R, Nelson GA. Proton radiation alters intrinsic and synaptic properties of CA1 pyramidal neurons of the mouse hippocampus. Radiat Res. 2015;183(2):208–218. [DOI] [PubMed] [Google Scholar]
- 154.Marty VN, Vlkolinsky R, Minassian N, Cohen T, Nelson GA, Spigelman I. Radiation-induced alterations in synaptic neurotransmission of dentate granule cells depend on the dose and species of charged particles. Radiat Res. 2014;182(6):653–665. [DOI] [PubMed] [Google Scholar]
- 155.Britten RA, Davis LK, Jewell JS, et al. Exposure to mission relevant doses of 1 GeV/Nucleon (56)Fe particles leads to impairment of attentional set-shifting performance in socially mature rats. Radiat Res. 2014;182(3):292–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Rudobeck E, Nelson GA, Sokolova IV, Vlkolinsky R. (28)silicon radiation impairs neuronal output in CA1 neurons of mouse ventral hippocampus without altering dendritic excitability. Radiat Res. 2014;181(4):407–415. [DOI] [PubMed] [Google Scholar]
- 157.Machida M, Lonart G, Britten RA. Low (60 cGy) doses of (56)Fe HZE-particle radiation lead to a persistent reduction in the glutamatergic readily releasable pool in rat hippocampal synaptosomes. Radiat Res. 2010;174(5):618–623. [DOI] [PubMed] [Google Scholar]
- 158.Villasana L, Rosenberg J, Raber J. Sex-dependent effects of 56Fe irradiation on contextual fear conditioning in C57BL/6J mice. Hippocampus. 2010;20(1):19–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Vlkolinsky R, Krucker T, Nelson GA, Obenaus A. (56)Fe-particle radiation reduces neuronal output and attenuates lipopolysaccharide-induced inhibition of long-term potentiation in the mouse hippocampus. Radiat Res. 2008;169(5):523–530. [DOI] [PubMed] [Google Scholar]
- 160.Moore TM, Basner M, Nasrini J, et al. Validation of the Cognition Test Battery for Spaceflight in a Sample of Highly Educated Adults. Aerosp Med Hum Perform. 2017;88(10):937–946. [DOI] [PubMed] [Google Scholar]
- 161.Basner M, Savitt A, Moore TM, et al. Development and Validation of the Cognition Test Battery for Spaceflight. Aerosp Med Hum Perform. 2015;86(11):942–952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Lim J, Dinges DF. Sleep deprivation and vigilant attention. Ann N Y Acad Sci. 2008;1129:305–322. [DOI] [PubMed] [Google Scholar]
- 163.Blatter K, Graw P, Munch M, Knoblauch V, Wirz-Justice A, Cajochen C. Gender and age differences in psychomotor vigilance performance under differential sleep pressure conditions. Behav Brain Res. 2006;168(2):312–317. [DOI] [PubMed] [Google Scholar]
- 164.Davis CM, DeCicco-Skinner KL, Roma PG, Hienz RD. Individual differences in attentional deficits and dopaminergic protein levels following exposure to proton radiation. Radiat Res. 2014;181(3):258–271. [DOI] [PubMed] [Google Scholar]
- 165.Wiegmann DA, Shappell SA. Human error and crew resource management failures in Naval aviation mishaps: a review of U.S. Naval Safety Center data, 1990–96. Aviat Space Environ Med. 1999;70(12):1147–1151. [PubMed] [Google Scholar]
- 166.Eling P, Derckx K, Maes R. On the historical and conceptual background of the Wisconsin Card Sorting Test. Brain Cogn. 2008;67(3):247–253. [DOI] [PubMed] [Google Scholar]
- 167.Sawada Y, Nishio Y, Suzuki K, et al. Attentional set-shifting deficit in Parkinson’s disease is associated with prefrontal dysfunction: an FDG-PET study. PloS one. 2012;7(6):e38498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Monchi O, Petrides M, Doyon J, Postuma RB, Worsley K, Dagher A. Neural bases of set-shifting deficits in Parkinson’s disease. J Neurosci. 2004;24(3):702–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Yerys BE, Wallace GL, Harrison B, Celano MJ, Giedd JN, Kenworthy LE. Set-shifting in children with autism spectrum disorders: reversal shifting deficits on the Intradimensional/Extradimensional Shift Test correlate with repetitive behaviors. Autism. 2009;13(5):523–538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Birrell JM, Brown VJ. Medial frontal cortex mediates perceptual attentional set shifting in the rat. J Neurosci. 2000;20(11):4320–4324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Jewell JS, Duncan VD, Fesshaye A, Tondin A, Macadat E, Britten RA. Exposure to </=15 cGy of 600 MeV/n (56)Fe Particles Impairs Rule Acquisition but not Long-Term Memory in the Attentional Set-Shifting Assay. Radiat Res. 2018;190(6):565–575. [DOI] [PubMed] [Google Scholar]
- 172.Britten RA, Duncan VD, Fesshaye AS, Wellman LL, Fallgren CM, Sanford LD. Sleep fragmentation exacerbates executive function impairments induced by protracted low dose rate neutron exposure. Int J Radiat Biol. 2019:1–11. [DOI] [PubMed] [Google Scholar]
- 173.Britten RA, Miller VD, Hadley MM, Jewell JS, Macadat E. Performance in hippocampus- and PFC-dependent cognitive domains are not concomitantly impaired in rats exposed to 20cGy of 1GeV/n (56)Fe particles. Life Sci Space Res (Amst). 2016;10:17–22. [DOI] [PubMed] [Google Scholar]
- 174.Acharya MM, Baddour AA, Kawashita T, et al. Epigenetic determinants of space radiation-induced cognitive dysfunction. Sci Rep. 2017;7:42885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Bellone JA, Gifford PS, Nishiyama NC, Hartman RE, Mao XW. Long-term effects of simulated microgravity and/or chronic exposure to low-dose gamma radiation on behavior and blood-brain barrier integrity. NPJ Microgravity. 2016;2:16019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Sicard G, Royet JP, Jourdan F. A comparative study of 2-deoxyglucose patterns of glomerular activation in the olfactory bulbs of C57 BL/6J and AKR/J mice. Brain Res. 1989;481(2):325–334. [DOI] [PubMed] [Google Scholar]
- 177.Wu X, Li D, Liu J, et al. Dammarane Sapogenins Ameliorates Neurocognitive Functional Impairment Induced by Simulated Long-Duration Spaceflight. Front Pharmacol. 2017;8:315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Sprugnoli G, Cagle YD, Santarnecchi E. Microgravity and Cosmic Radiations During Space Exploration as a Window Into Neurodegeneration on Earth. JAMA Neurol. 2020;77(2):157–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Stojanoski B, Benoit A, Van Den Berg N, et al. Sustained vigilance is negatively affected by mild and acute sleep loss reflected by reduced capacity for decision making, motor preparation, and execution. Sleep. 2019;42(1). [DOI] [PubMed] [Google Scholar]
- 180.Nair D, Zhang SX, Ramesh V, et al. Sleep fragmentation induces cognitive deficits via nicotinamide adenine dinucleotide phosphate oxidase-dependent pathways in mouse. Am J Respir Crit Care Med. 2011;184(11):1305–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.McCoy JG, Tartar JL, Bebis AC, et al. Experimental sleep fragmentation impairs attentional set-shifting in rats. Sleep. 2007;30(1):52–60. [DOI] [PubMed] [Google Scholar]
- 182.Martin SE, Engleman HM, Deary IJ, Douglas NJ. The effect of sleep fragmentation on daytime function. Am J Respir Crit Care Med. 1996;153(4 Pt 1):1328–1332. [DOI] [PubMed] [Google Scholar]
- 183.Britten RA, Fesshaye AS, Duncan VD, Wellman LL, Sanford LD. Sleep Fragmentation Exacerbates Executive Function Impairments Induced by Low Doses of Si Ions. Radiat Res. 2020;194(2):116–123. [DOI] [PubMed] [Google Scholar]
- 184.Lee SMC, Ribeiro LC, Martin DS, et al. Arterial structure and function during and after long-duration spaceflight. J Appl Physiol (1985). 2020;129(1):108–123. [DOI] [PubMed] [Google Scholar]
- 185.Mao XW, Byrum S, Nishiyama NC, et al. Impact of Spaceflight and Artificial Gravity on the Mouse Retina: Biochemical and Proteomic Analysis. Int J Mol Sci. 2018;19(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Mao XW, Nishiyama NC, Byrum SD, et al. Spaceflight induces oxidative damage to blood-brain barrier integrity in a mouse model. Faseb J. 2020;34(11):15516–15530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Sofronova SI, Tarasova OS, Gaynullina D, et al. Spaceflight on the Bion-M1 biosatellite alters cerebral artery vasomotor and mechanical properties in mice. J Appl Physiol (1985). 2015;118(7):830–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Hatton DC, Yue Q, Chapman J, et al. Blood pressure and mesenteric resistance arterial function after spaceflight. J Appl Physiol (1985). 2002;92(1):13–17. [DOI] [PubMed] [Google Scholar]
- 189.Prisby RD, Wilkerson MK, Sokoya EM, Bryan RM Jr., Wilson E, Delp MD. Endothelium-dependent vasodilation of cerebral arteries is altered with simulated microgravity through nitric oxide synthase and EDHF mechanisms. J Appl Physiol (1985). 2006;101(1):348–353. [DOI] [PubMed] [Google Scholar]
- 190.Zhang R, Jia G, Bao J, et al. Increased vascular cell adhesion molecule-1 was associated with impaired endothelium-dependent relaxation of cerebral and carotid arteries in simulated microgravity rats. J Physiol Sci. 2008;58(1):67–73. [DOI] [PubMed] [Google Scholar]
- 191.Ade CJ, Broxterman RM, Charvat JM, Barstow TJ. Incidence Rate of Cardiovascular Disease End Points in the National Aeronautics and Space Administration Astronaut Corps. J Am Heart Assoc. 2017;6(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Kashcheev VV, Chekin SY, Karpenko SV, et al. Radiation Risk of Cardiovascular Diseases in the Cohort of Russian Emergency Workers of the Chernobyl Accident. Health Phys. 2017;113(1):23–29. [DOI] [PubMed] [Google Scholar]
- 194.Little MP, Azizova TV, Bazyka D, et al. Systematic review and meta-analysis of circulatory disease from exposure to low-level ionizing radiation and estimates of potential population mortality risks. Environ Health Perspect. 2012;120(11):1503–1511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Hughson RL, Helm A, Durante M. Heart in space: effect of the extraterrestrial environment on the cardiovascular system. Nat Rev Cardiol. 2018;15(3):167–180. [DOI] [PubMed] [Google Scholar]
- 196.Soucy KG, Lim HK, Kim JH, et al. HZE (5)(6)Fe-ion irradiation induces endothelial dysfunction in rat aorta: role of xanthine oxidase. Radiat Res. 2011;176(4):474–485. [DOI] [PubMed] [Google Scholar]
- 197.Soucy KG, Lim HK, Attarzadeh DO, et al. Dietary inhibition of xanthine oxidase attenuates radiation-induced endothelial dysfunction in rat aorta. J Appl Physiol (1985). 2010;108(5):1250–1258. [DOI] [PubMed] [Google Scholar]
- 198.Grabham P, Sharma P, Bigelow A, Geard C. Two distinct types of the inhibition of vasculogenesis by different species of charged particles. Vasc Cell. 2013;5(1):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Mao XW, Favre CJ, Fike JR, et al. High-LET radiation-induced response of microvessels in the Hippocampus. Radiat Res. 2010;173(4):486–493. [DOI] [PubMed] [Google Scholar]
- 200.Yu T, Parks BW, Yu S, et al. Iron-ion radiation accelerates atherosclerosis in apolipoprotein E-deficient mice. Radiat Res. 2011;175(6):766–773. [DOI] [PubMed] [Google Scholar]
- 201.Koturbash I, Miousse IR, Sridharan V, et al. Radiation-induced changes in DNA methylation of repetitive elements in the mouse heart. Mutat Res. 2016;787:43–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Sasi SP, Yan X, Zuriaga-Herrero M, et al. Different Sequences of Fractionated Low-Dose Proton and Single Iron-Radiation-Induced Divergent Biological Responses in the Heart. Radiat Res. 2017;188(2):191–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Marshall-Goebel K, Laurie SS, Alferova IV, et al. Assessment of Jugular Venous Blood Flow Stasis and Thrombosis During Spaceflight. JAMA Netw Open. 2019;2(11):e1915011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Aunon-Chancellor SM, Pattarini JM, Moll S, Sargsyan A. Venous Thrombosis during Spaceflight. N Engl J Med. 2020;382(1):89–90. [DOI] [PubMed] [Google Scholar]
- 205.Blaber EA, Dvorochkin N, Torres ML, et al. Mechanical unloading of bone in microgravity reduces mesenchymal and hematopoietic stem cell-mediated tissue regeneration. Stem Cell Res. 2014;13(2):181–201. [DOI] [PubMed] [Google Scholar]
- 206.Ambrosi TH, Scialdone A, Graja A, et al. Adipocyte Accumulation in the Bone Marrow during Obesity and Aging Impairs Stem Cell-Based Hematopoietic and Bone Regeneration. Cell Stem Cell. 2017;20(6):771–784.e776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Valenti MT, Dalle Carbonare L, Dorelli G, Mottes M. Effects of physical exercise on the prevention of stem cells senescence. Stem Cell Rev Rep. 2020;16(1):33–40. [DOI] [PubMed] [Google Scholar]
- 208.DeCarolis NA, Kirby ED, Wyss-Coray T, Palmer TD. The Role of the Microenvironmental Niche in Declining Stem-Cell Functions Associated with Biological Aging. Cold Spring Harb Perspect Med. 2015;5(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Brown PT, Handorf AM, Jeon WB, Li WJ. Stem cell-based tissue engineering approaches for musculoskeletal regeneration. Curr Pharm Des. 2013;19(19):3429–3445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Karunagaran D, Joseph J, Kumar TR. Cell growth regulation. Adv Exp Med Biol. 2007;595:245–268. [DOI] [PubMed] [Google Scholar]
- 211.Shinde V, Brungs S, Henry M, et al. Simulated Microgravity Modulates Differentiation Processes of Embryonic Stem Cells. Cell Physiol Biochem. 2016;38(4):1483–1499. [DOI] [PubMed] [Google Scholar]
- 212.Blaber EA, Finkelstein H, Dvorochkin N, et al. Microgravity Reduces the Differentiation and Regenerative Potential of Embryonic Stem Cells. Stem Cells Dev. 2015;24(22):2605–2621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Lei X, Cao Y, Zhang Y, et al. Effect of microgravity on proliferation and differentiation of embryonic stem cells in an automated culturing system during the TZ-1 space mission. Cell Prolif. 2018;51(5):e12466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Acharya A, Brungs S, Henry M, et al. Modulation of Differentiation Processes in Murine Embryonic Stem Cells Exposed to Parabolic Flight-Induced Acute Hypergravity and Microgravity. Stem Cells Dev. 2018;27(12):838–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Baio J, Martinez AF, Silva I, et al. Cardiovascular progenitor cells cultured aboard the International Space Station exhibit altered developmental and functional properties. NPJ Microgravity. 2018;4:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Baio J, Martinez AF, Bailey L, Hasaniya N, Pecaut MJ, Kearns-Jonker M. Spaceflight Activates Protein Kinase C Alpha Signaling and Modifies the Developmental Stage of Human Neonatal Cardiovascular Progenitor Cells. Stem Cells Dev. 2018;27(12):805–818. [DOI] [PubMed] [Google Scholar]
- 217.Camberos V, Baio J, Bailey L, Hasaniya N, Lopez LV, Kearns-Jonker M. Effects of Spaceflight and Simulated Microgravity on YAP1 Expression in Cardiovascular Progenitors: Implications for Cell-Based Repair. Int J Mol Sci. 2019;20(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.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] [PMC free article] [PubMed] [Google Scholar]
- 219.Touchstone H, Bryd R, Loisate S, et al. Recovery of stem cell proliferation by low intensity vibration under simulated microgravity requires LINC complex. NPJ Microgravity. 2019;5:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Merzlikina NV, Buravkova LB, Romanov YA. The primary effects of clinorotation on cultured human mesenchymal stem cells. J Gravit Physiol. 2004;11(2):P193–194. [PubMed] [Google Scholar]
- 221.Gershovich JG, Buravkova LB. Morphofunctional status and osteogenic differentiation potential of human mesenchymal stromal precursor cells during in vitro modeling of microgravity effects. Bull Exp Biol Med. 2007;144(4):608–613. [DOI] [PubMed] [Google Scholar]
- 222.Yuge L, Kajiume T, Tahara H, et al. Microgravity potentiates stem cell proliferation while sustaining the capability of differentiation. Stem Cells Dev. 2006;15(6):921–929. [DOI] [PubMed] [Google Scholar]
- 223.Dai ZQ, Wang R, Ling SK, Wan YM, Li YH. Simulated microgravity inhibits the proliferation and osteogenesis of rat bone marrow mesenchymal stem cells. Cell Prolif. 2007;40(5):671–684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Monticone M, Liu Y, Pujic N, Cancedda R. Activation of nervous system development genes in bone marrow derived mesenchymal stem cells following spaceflight exposure. J Cell Biochem. 2010;111(2):442–452. [DOI] [PubMed] [Google Scholar]
- 225.Hu Z, Wang Y, Sun Z, et al. miRNA-132–3p inhibits osteoblast differentiation by targeting Ep300 in simulated microgravity. Sci Rep. 2015;5:18655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Markina E, Andreeva E, Andrianova I, Sotnezova E, Buravkova L. Stromal and Hematopoietic Progenitors from C57/BI/6N Murine Bone Marrow After 30-Day “BION-M1” Spaceflight. Stem Cells Dev. 2018;27(18):1268–1277. [DOI] [PubMed] [Google Scholar]
- 227.Blaber EA, Dvorochkin N, Lee C, et al. Microgravity induces pelvic bone loss through osteoclastic activity, osteocytic osteolysis, and osteoblastic cell cycle inhibition by CDKN1a/p21. PLoS One. 2013;8(4):e61372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Zhang C, Li L, Jiang Y, et al. Space microgravity drives transdifferentiation of human bone marrow-derived mesenchymal stem cells from osteogenesis to adipogenesis. FASEB J. 2018;32(8):4444–4458. [DOI] [PubMed] [Google Scholar]
- 229.Meyers VE, Zayzafoon M, Douglas JT, McDonald JM. RhoA and cytoskeletal disruption mediate reduced osteoblastogenesis and enhanced adipogenesis of human mesenchymal stem cells in modeled microgravity. J Bone Miner Res. 2005;20(10):1858–1866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Pecaut MJ, Mao XW, Bellinger DL, et al. Is spaceflight-induced immune dysfunction linked to systemic changes in metabolism? PLoS One. 2017;12(5):e0174174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Gridley DS, Slater JM, Luo-Owen X, et al. Spaceflight effects on T lymphocyte distribution, function and gene expression. J Appl Physiol (1985). 2009;106(1):194–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Crucian B, Stowe R, Quiriarte H, Pierson D, Sams C. Monocyte phenotype and cytokine production profiles are dysregulated by short-duration spaceflight. Aviat Space Environ Med. 2011;82(9):857–862. [DOI] [PubMed] [Google Scholar]
- 233.Sonnenfeld G The immune system in space, including Earth-based benefits of space-based research. Curr Pharm Biotechnol. 2005;6(4):343–349. [DOI] [PubMed] [Google Scholar]
- 234.Cao D, Song J, Ling S, et al. Hematopoietic stem cells and lineage cells undergo dynamic alterations under microgravity and recovery conditions. FASEB J. 2019;33(6):6904–6918. [DOI] [PubMed] [Google Scholar]
- 235.Benjamin CL, Stowe RP, St John L, et al. Decreases in thymopoiesis of astronauts returning from space flight. JCI Insight. 2016;1(12):e88787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Davis TA, Wiesmann W, Kidwell W, et al. Effect of spaceflight on human stem cell hematopoiesis: suppression of erythropoiesis and myelopoiesis. J Leukoc Biol. 1996;60(1):69–76. [DOI] [PubMed] [Google Scholar]
- 237.Barrila J, Ott CM, LeBlanc C, et al. Spaceflight modulates gene expression in the whole blood of astronauts. NPJ Microgravity. 2016;2:16039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Sotnezova EV, Markina EA, Andreeva ER, Buravkova LB. Myeloid Precursors in the Bone Marrow of Mice after a 30-Day Space Mission on a Bion-M1 Biosatellite. Bull Exp Biol Med. 2017;162(4):496–500. [DOI] [PubMed] [Google Scholar]
- 239.Low EK, Brudvik E, Kuhlman B, Wilson PF, Almeida-Porada G, Porada CD. Microgravity Impairs DNA Damage Repair in Human Hematopoietic Stem/Progenitor Cells and Inhibits Their Differentiation into Dendritic Cells. Stem Cells Dev. 2018;27(18):1257–1267. [DOI] [PubMed] [Google Scholar]
- 240.Mattei C, Alshawaf A, D’Abaco G, Nayagam B, Dottori M. Generation of Neural Organoids from Human Embryonic Stem Cells Using the Rotary Cell Culture System: Effects of Microgravity on Neural Progenitor Cell Fate. Stem Cells Dev. 2018;27(12):848–857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Jha R, Wu Q, Singh M, et al. Simulated Microgravity and 3D Culture Enhance Induction, Viability, Proliferation and Differentiation of Cardiac Progenitors from Human Pluripotent Stem Cells. Sci Rep. 2016;6:30956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Xue L, Li Y, Chen J. Duration of simulated microgravity affects the differentiation of mesenchymal stem cells. Mol Med Rep. 2017;15(5):3011–3018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Almeida-Porada G, Rodman C, Kuhlman B, 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(18):1237–1256. [DOI] [PubMed] [Google Scholar]
- 244.Rodman C, Almeida-Porada G, George SK, 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(6):1398–1407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Encinas JM, Vazquez ME, Switzer RC, et al. Quiescent adult neural stem cells are exceptionally sensitive to cosmic radiation. Exp Neurol. 2008;210(1):274–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Suman S, Rodriguez OC, Winters TA, Fornace AJ, Albanese C, Datta K. Therapeutic and space radiation exposure of mouse brain causes impaired DNA repair response and premature senescence by chronic oxidant production. Aging (Albany NY). 2013;5(8):607–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Cekanaviciute E, Rosi S, Costes SV. Central Nervous System Responses to Simulated Galactic Cosmic Rays. Int J Mol Sci. 2018;19(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.McConnell AM, Konda B, Kirsch DG, Stripp BR. Distal airway epithelial progenitor cells are radiosensitive to High-LET radiation. Sci Rep. 2016;6:33455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.An L, Li Y, Fan Y, et al. The Trends in Global Gene Expression in Mouse Embryonic Stem Cells During Spaceflight. Front Genet. 2019;10:768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Mao XW, Nishiyama NC, Pecaut MJ, et al. Simulated Microgravity and Low-Dose/Low-Dose-Rate Radiation Induces Oxidative Damage in the Mouse Brain. Radiat Res. 2016;185(6):647–657. [DOI] [PubMed] [Google Scholar]
- 251.Shanmugarajan S, Zhang Y, Moreno-Villanueva M, et al. Combined Effects of Simulated Microgravity and Radiation Exposure on Osteoclast Cell Fusion. Int J Mol Sci. 2017;18(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Kondo H, Limoli C, Searby ND, et al. Shared oxidative pathways in response to gravity-dependent loading and gamma-irradiation of bone marrow-derived skeletal cell progenitors. Radiats Biol Radioecol. 2007;47(3):281–285. [PubMed] [Google Scholar]
- 253.Manti L, Durante M, Cirrone GA, et al. Modelled microgravity does not modify the yield of chromosome aberrations induced by high-energy protons in human lymphocytes. Int J Radiat Biol. 2005;81(2):147–155. [DOI] [PubMed] [Google Scholar]
- 254.Yan Y, Zhang K, Zhou G, Hu W. MicroRNAs Responding to Space Radiation. Int J Mol Sci. 2020;21(18). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Girardi C, De Pittà C, Casara S, et al. Analysis of miRNA and mRNA expression profiles highlights alterations in ionizing radiation response of human lymphocytes under modeled microgravity. PLoS One. 2012;7(2):e31293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.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. [DOI] [PubMed] [Google Scholar]
- 257.Blaber E, Sato K, Almeida EA. Stem cell health and tissue regeneration in microgravity. Stem Cells Dev. 2014;23 Suppl 1:73–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Grimm D, Egli M, Krüger M, et al. Tissue Engineering Under Microgravity Conditions-Use of Stem Cells and Specialized Cells. Stem Cells Dev. 2018;27(12):787–804. [DOI] [PubMed] [Google Scholar]
- 259.Otsuka T, Imura T, Nakagawa K, et al. Simulated Microgravity Culture Enhances the Neuroprotective Effects of Human Cranial Bone-Derived Mesenchymal Stem Cells in Traumatic Brain Injury. Stem Cells Dev. 2018;27(18):1287–1297. [DOI] [PubMed] [Google Scholar]
- 260.Imura T, Nakagawa K, Kawahara Y, Yuge L. Stem Cell Culture in Microgravity and Its Application in Cell-Based Therapy. Stem Cells Dev. 2018;27(18):1298–1302. [DOI] [PubMed] [Google Scholar]
- 261.Blaber EA. Special Issue: Stem Cells, Radiation, and Microgravity. Stem Cells Dev. 2018;27(18):1227–1229. [DOI] [PubMed] [Google Scholar]
- 262.Blaber EA, Parker GC. Special Issue: Stem Cells and Microgravity. Stem Cells Dev. 2018;27(12):783–786. [DOI] [PubMed] [Google Scholar]
- 263.Chen Z, Luo Q, Lin C, Song G. Simulated microgravity inhibits osteogenic differentiation of mesenchymal stem cells through down regulating the transcriptional co-activator TAZ. Biochem Biophys Res Commun. 2015;468(1–2):21–26. [DOI] [PubMed] [Google Scholar]
- 264.Mao X, Chen Z, Luo Q, Zhang B, Song G. Simulated microgravity inhibits the migration of mesenchymal stem cells by remodeling actin cytoskeleton and increasing cell stiffness. Cytotechnology. 2016;68(6):2235–2243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Zarrinpour V, Hajebrahimi Z, Jafarinia M. Expression pattern of neurotrophins and their receptors during neuronal differentiation of adipose-derived stem cells in simulated microgravity condition. Iran J Basic Med Sci. 2017;20(2):178–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Wilkerson MK, Lesniewski LA, Golding EM, et al. Simulated microgravity enhances cerebral artery vasoconstriction and vascular resistance through endothelial nitric oxide mechanism. Am J Physiol Heart Circ Physiol. 2005;288(4):H1652–1661. [DOI] [PubMed] [Google Scholar]
- 267.Colleran PN, Wilkerson MK, Bloomfield SA, Suva LJ, Turner RT, Delp MD. Alterations in skeletal perfusion with simulated microgravity: a possible mechanism for bone remodeling. J Appl Physiol (1985). 2000;89(3):1046–1054. [DOI] [PubMed] [Google Scholar]
- 268.Geary GG, Krause DN, Purdy RE, Duckles SP. Simulated microgravity increases myogenic tone in rat cerebral arteries. J Appl Physiol (1985). 1998;85(5):1615–1621. [DOI] [PubMed] [Google Scholar]
- 269.Davet J, Clavel B, Datas L, et al. Choroidal readaptation to gravity in rats after spaceflight and head-down tilt. J Appl Physiol (1985). 1998;84(1):19–29. [DOI] [PubMed] [Google Scholar]
- 270.Gabrion J, Herbute S, Oliver J, et al. Choroidal responses in microgravity. (SLS-1, SLS-2 and hindlimb-suspension experiments). Acta Astronaut. 1995;36(8–12):439–448. [DOI] [PubMed] [Google Scholar]
- 271.Campagne DM. Stress and perceived social isolation (loneliness). Arch Gerontol Geriatr. 2019;82:192–199. [DOI] [PubMed] [Google Scholar]
- 272.Mumtaz F, Khan MI, Zubair M, Dehpour AR. Neurobiology and consequences of social isolation stress in animal model-A comprehensive review. Biomed Pharmacother. 2018;105:1205–1222. [DOI] [PubMed] [Google Scholar]
- 273.Mason G, Wilson D, Hampton C, Wurbel H. Non-invasively assessing disturbance and stress in laboratory rats by scoring chromodacryorrhoea. Altern Lab Anim. 2004;32 Suppl 1A:153–159. [DOI] [PubMed] [Google Scholar]
- 274.Horie K, Kudo T, Yoshinaga R, 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):745–750. [DOI] [PubMed] [Google Scholar]
- 275.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(37):14777–14782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.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):465–471. [DOI] [PubMed] [Google Scholar]
- 277.Iwaniec UT, Philbrick KA, Wong CP, et al. Room temperature housing results in premature cancellous bone loss in growing female mice: implications for the mouse as a preclinical model for age-related bone loss. Osteoporos Int. 2016;27(10):3091–3101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Luan HQ, Sun LW, Huang YF, et al. Use of micro-computed tomography to evaluate the effects of exercise on preventing the degeneration of articular cartilage in tail-suspended rats. Life Sci Space Res (Amst). 2015;6:15–20. [DOI] [PubMed] [Google Scholar]
- 279.Djordjevic J, Djordjevic A, Adzic M, Radojcic MB. Effects of chronic social isolation on Wistar rat behavior and brain plasticity markers. Neuropsychobiology. 2012;66(2):112–119. [DOI] [PubMed] [Google Scholar]
- 280.McLean S, Grayson B, Harris M, Protheroe C, Woolley M, Neill J. Isolation rearing impairs novel object recognition and attentional set shifting performance in female rats. J Psychopharmacol. 2010;24(1):57–63. [DOI] [PubMed] [Google Scholar]
- 281.Jessop JJ, Bayer BM. Time-dependent effects of isolation on lymphocyte and adrenocortical activity. J Neuroimmunol. 1989;23(2):143–147. [DOI] [PubMed] [Google Scholar]
- 282.Ros-Simo C, Valverde O. Early-life social experiences in mice affect emotional behaviour and hypothalamic-pituitary-adrenal axis function. Pharmacol Biochem Behav. 2012;102(3):434–441. [DOI] [PubMed] [Google Scholar]
- 283.Shetty RA, Sadananda M. Brief Social Isolation in the Adolescent Wistar-Kyoto Rat Model of Endogenous Depression Alters Corticosterone and Regional Monoamine Concentrations. Neurochem Res. 2017;42(5):1470–1477. [DOI] [PubMed] [Google Scholar]
- 284.Bellavance MA, Rivest S. The HPA - Immune Axis and the Immunomodulatory Actions of Glucocorticoids in the Brain. Front Immunol. 2014;5:136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.Krugel U, Fischer J, Bauer K, Sack U, Himmerich H. The impact of social isolation on immunological parameters in rats. Arch Toxicol. 2014;88(3):853–855. [DOI] [PubMed] [Google Scholar]
- 286.Wu W, Yamaura T, Murakami K, et al. Social isolation stress enhanced liver metastasis of murine colon 26-L5 carcinoma cells by suppressing immune responses in mice. Life Sci. 2000;66(19):1827–1838. [DOI] [PubMed] [Google Scholar]
- 287.Wang L, Cao M, Pu T, Huang H, Marshall C, Xiao M. Enriched Physical Environment Attenuates Spatial and Social Memory Impairments of Aged Socially Isolated Mice. Int J Neuropsychopharmacol. 2018;21(12):1114–1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Pereda-Perez I, Popovic N, Otalora BB, et al. Long-term social isolation in the adulthood results in CA1 shrinkage and cognitive impairment. Neurobiol Learn Mem. 2013;106:31–39. [DOI] [PubMed] [Google Scholar]
- 289.Gong WG, Wang YJ, Zhou H, et al. Citalopram Ameliorates Synaptic Plasticity Deficits in Different Cognition-Associated Brain Regions Induced by Social Isolation in Middle-Aged Rats. Mol Neurobiol. 2017;54(3):1927–1938. [DOI] [PubMed] [Google Scholar]
- 290.Smith BM, Yao X, Chen KS, Kirby ED. A Larger Social Network Enhances Novel Object Location Memory and Reduces Hippocampal Microgliosis in Aged Mice. Front Aging Neurosci. 2018;10:142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291.Filipovic D, Stanisavljevic A, Jasnic N, et al. Chronic Treatment with Fluoxetine or Clozapine of Socially Isolated Rats Prevents Subsector-Specific Reduction of Parvalbumin Immunoreactive Cells in the Hippocampus. Neuroscience. 2018;371:384–394. [DOI] [PubMed] [Google Scholar]
- 292.Todorovic N, Filipovic D. The antidepressant- and anxiolytic-like effects of fluoxetine and clozapine in chronically isolated rats involve inhibition of hippocampal TNF-alpha. Pharmacol Biochem Behav. 2017;163:57–65. [DOI] [PubMed] [Google Scholar]
- 293.Zhou Y, Ni H, Li M, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Doty SB, Vico L, Wronski T, Morey-Holton E. Use of animal models to study skeletal effects of space flight. Adv Space Biol Med. 2005;10:209–224. [DOI] [PubMed] [Google Scholar]
- 295.Musacchia XJ, Fagette S. Weightlessness simulations for cardiovascular and muscle systems: validity of rat models. Journal of gravitational physiology : a journal of the International Society for Gravitational Physiology. 1997;4(3):49–59. [PubMed] [Google Scholar]