Next Article in Journal
Altered Language-Related Effective Connectivity in Patients with Benign Childhood Epilepsy with Centrotemporal Spikes
Next Article in Special Issue
Impact of Microgravity and Other Spaceflight Factors on Retina of Vertebrates and Humans In Vivo and In Vitro
Previous Article in Journal
Understanding COVID: Collaborative Government Campaign for Citizen Digital Health Literacy in the COVID-19 Pandemic
Previous Article in Special Issue
Effect of Microgravity on the Gut Microbiota Bacterial Composition in a Hindlimb Unloading Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Addressing Spaceflight Biology through the Lens of a Histologist–Embryologist

by
Paschalis Theotokis
1,
Maria Eleni Manthou
1,
Theodora-Eleftheria Deftereou
2,
Dimosthenis Miliaras
1 and
Soultana Meditskou
1,*
1
Laboratory of Histology and Embryology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
2
Laboratory of Histology and Embryology, Democritus University of Thrace, 69100 Alexandroupolis, Greece
*
Author to whom correspondence should be addressed.
Life 2023, 13(2), 588; https://doi.org/10.3390/life13020588
Submission received: 28 November 2022 / Revised: 9 February 2023 / Accepted: 14 February 2023 / Published: 20 February 2023
(This article belongs to the Special Issue The Space Environment on Human Health and Disease)

Abstract

:
Embryogenesis and fetal development are highly delicate and error-prone processes in their core physiology, let alone if stress-associated factors and conditions are involved. Space radiation and altered gravity are factors that could radically affect fertility and pregnancy and compromise a physiological organogenesis. Unfortunately, there is a dearth of information examining the effects of cosmic exposures on reproductive and proliferating outcomes with regard to mammalian embryonic development. However, explicit attention has been given to investigations exploring discrete structures and neural networks such as the vestibular system, an entity that is viewed as the sixth sense and organically controls gravity beginning with the prenatal period. The role of the gut microbiome, a newly acknowledged field of research in the space community, is also being challenged to be added in forthcoming experimental protocols. This review discusses the data that have surfaced from simulations or actual space expeditions and addresses developmental adaptations at the histological level induced by an extraterrestrial milieu.

1. Introduction

Yuri Gagarin was the first human to journey into a space nearly void of gravity. To date, more than 550 astronauts have traveled beyond the Kármán line, the one-hundred-kilometer distance that theoretically separates Earth from outer space, and National Aeronautics and Space Administration (NASA) has been in the epicenter of these expeditions. The International Space Station (ISS), successor of the Russian Mir, is the only modular low-gravity laboratory currently in low Earth orbit (LEO) that performs and disseminates openly biology-based research [1]. Although investigations in spacelike conditions, including exposure to ionizing radiation and approaching weightlessness, which is defined as microgravity and hypergravity occurring on launch and re-entry of missions, were conducted on Mir and ISS more than 20 years ago [2,3], studies from simulations in miscellaneous organisms have been published since spacecraft flights of the late 1960s [4]. Interesting data have been collected implicating tissue and organ dysfunction; however, even more questions arise and need to be addressed.
Whether the travel is in LEO or in deep space, and whether in short- or long-duration spaceflight, such experiences set in motion various homeostatic mechanisms that equip the organism with an opportunistic advantage of survival [5]. Remarkably, these responses in a newly created embryo and the subsequent organ formation are drastically different from the adaptations that are taking place in an already developed individual organism. This review recapitulates published data on various aspects of cell biology with regard to proprioception, especially in early development, dissecting the role of gravity that governs organogenesis. Among the vital systemic organs, the vestibular system with its brain projections is the one that has been investigated most thoroughly [6,7,8,9,10], and skeletal, muscular and gonad structures are close behind [11,12,13]. Additional attention has been given to the recently acknowledged gut microbiota field and how this affects astronauts and future space missions. Despite the fact that most developmental evidence is derived from animal studies, and no formal, in vivo experiments have been conducted at the human embryonic level, potential in vitro outcomes serve as a basis for the formulation of human-applied deductive arguments.

2. The Vestibular Adaptation; Mammalian’s Earliest Experience with Gravity

Human life starts in the womb of the mother, where a healthy egg and a healthy activated sperm cell fuse to form a zygote in the fallopian tube. The fertilization process lasts 24 h and is followed by cleavage with multiple rounds of cell divisions and the formation of a blastocyst, which in turn is implanted into the endometrium. After 2 weeks, the gastrulation process creates three primary germ layers, namely, ecto- meso- and endoderm, and these structures produce all tissues and organs of the human body. At this point, from the 4th to the 8th week of pregnancy, the embryo growth procedure is called the organogenetic phase, during which the main organs of all organ systems develop. The movement of a pregnant mother’s body stimulates the vestibular sense, also known as the gravity- or our “sixth”-sense [14], providing proprioception, and is actually one of the earliest to develop in a growing baby [10,15].
The vestibular system is highly established at just 5 months in pregnancy and transmits to the developing fetal brain essential sensory information while the baby is virtually floating around in the amniotic fluid. The vestibule is a derivative of the otic vesicle, the primordium of the internal ear. The otic vesicle divides into the dorsal utricular part, which gives rise to the utricle, semicircular ducts and endolymphatic duct, whereas the ventral saccular part gives rise to the saccule and cochlear duct. The utricle is responding to linear horizontal acceleration and head tilting, while with vertical acceleration a gravitational force induces hair cells in the saccule to respond [16] (Figure 1). Accordingly, angular acceleration receptors are activated in the semicircular canals. In either case, the maculae’s otolithic membrane and the cupulae’s mucopolysaccharides instruct the abutting hair cells of the neuroepithelium to send action potentials through the vestibulocochlear nerve (CN VIII), tailoring the response to the respective movement. Notably, gravity takes its toll on the vestibular system and more specifically in otolith formation within the early developmental periods [17,18,19].
During a space flight, microgravity conditions exist and embryonic otolithic function is spared [20]. Upon launch and landing, where hypergravity prevails, gravity-sensing cells are overworking. On the other hand, the angular acceleration sensors of the semicircular canals are stimulated equally in both micro- and hypergravity [9,21]. In an experiment where rat embryos were exposed to microgravitational forces prior to the formation of the vestibule up until a somewhat functional version (from gestation day 9 to day 19), authors showed that afferent projections from sensory paths to the vestibular nuclei of the hindbrain and cerebellum where altered, especially those coming from the saccule [21]. This experimental setting was tested in embryos as well as adult mice raised in hypergravity (1.5 G) and simulating long- and short-term exposures. The authors concluded that the gravitational milieu drastically affects terminal synaptic and arborization patterns [6], whereas sensory epithelium formation was not affected in rat pups exposed to microgravity postnatally [22].

3. Brain Vulnerability to Gravitational Perturbations

As described above, signals from the semicircular canals and otolith organs branch off toward a network targeting the brainstem vestibular nuclei (VN) and vestibulocerebellar nuclei, such as the fastigial nuclei (FN), contributing significantly to spatial orientation and navigation [14]. Bruce et al. report that synaptogenesis in the medial vestibular nucleus is hampered in developing rat embryos that were gestationally raised in microgravity along with decreased axonal arborization [6,21]. However, vestibular information is also heavily exchanged in other, more diverse brain areas such as the brainstem, the cerebellum and the visual system. Specifically, it has been reported that cerebellar structure and motor coordination are heavily affected by intrauterine and perinatal exposure to hypergravity in rat neonates [23,24,25,26]. Additionally, it has been shown that exposure to both hypergravity and microgravity infers disastrous effects on locomotor behavior in rats [27], whereas neuronal degeneration has been acknowledged in various brain regions [28].
The adult CNS manifests a remarkable flexibility to gravity alterations, mainly through behavioral adjustments in motor coordination, as clearly observed in the majority of astronauts during space weightlessness and their readaptation to Earth’s forces [29,30,31,32]. On the contrary, the developing fetal CNS discloses a pronounced, considerable remodeling within core functions and structures, as indicated in rat experiments, including righting reactions after prenatal exposure to microgravity [21] and equilibrium perturbations that are directly linked to structural changes in the brain [33]. Histological analysis of developing rat brain specimens during an 11-day spaceflight suggested that variations in two proteins were involved in: (i) the formation of the choroid plexus and (ii) cerebrospinal fluid production [34]. To better illuminate brain alterations caused by spaceflight, a large-radius centrifuge was developed at NASA Ames Research Center (ARC), which provides the most elaborate and ideal system to perform large-scale and long-term developmental studies [23,35]. Furthermore, in vitro methods studying neuronal differentiation, neurite extension, and synaptogenesis have also been developed to assess radiation-based neuronal impairment [36].
In addition to rats, amphibians that were exposed to microgravity during critical developmental stages displayed miscellaneous malformations in the closure of the neural tube [37]. Another important paradigm is the one provided from crickets in space, where gravity-sensing neurons were heavily influenced by microgravity [38,39]. In the developing fish nervous system, microgravity caused synaptic contact alterations in vestibular regions during critical periods of development [40,41]. Interestingly, changes in swimming behavior were discovered under microgravity [42] and also under long-term hypergravity [43,44]. Moreover, changes in peripheral nerve reflexes have been recorded in hypergravity investigations using Xenopus laevis [45]. Lastly, there is no disputing the fact that cerebellar alterations contributed to the brain’s disability; this is supported also by biochemical data at the cellular and molecular level [46,47].

4. Mesenchyme-Derived Tissue Formation Adaptability in Space-like Conditions

Physiologically, the mammalian skeleton grows from the mesenchyme derived from the mesoderm and neural crest, initially as cartilage tissue from dense mesenchyme that will create bone through osteogenesis. It has been established that space decreases bone density, based on data from astronauts returning to earth [48,49]. To explore this mechanism, in vitro conditions of bone formation in a reduced-gravity environment were induced and tested in culture [11,50]. Authors illustrated that pre-metatarsals that have already initiated chondrogenesis resulted in increased rod size and shape, compared with older pre-metatarsal tissue [50]. Chronic exposure to ionizing radiation was shown to affect mouse fetal fibroblasts [51]. Likewise, osteoblasts have been presented showing reduced cellular activity in microgravity due to an undifferentiated state [52,53]. Table 1 summarizes the majority of experiments performed with regard to specific organ systems, during embryogenesis or postnatally, based upon research availability.
The cell differentiation process seems to be the key event affected in space expeditions, as suggested by analyzing specific cell cultures of cartilage, bone and muscle postexploration or induced gravitational variations [54,55,56]. Kacena and colleagues performed in vitro hypergravity experiments and presented altered actin and fibronectin elements, modifying the osteoblast–substrate adhesion [57]. To measure the microgravity effect on bone, research teams utilized either the Rotary Cell Culture System developed by NASA or the SJ-10 satellite and found that osteoblastic differentiation of human mesenchymal stem cells (hMSC) is inhibited and that the development of adipocytic lineage phenotype is induced [58,59]. Bone metabolism perturbations with concomitant renal stone risk were declared during ISS flights [60]. Intriguingly, cultured embryonic chick bone cells flown with NASA’s mission STS-59 were also affected by spaceflight conditions, as microgravity seems to down-regulate type I collagen and osteocalcin gene expression, thereby suppressing the osteogenic phenotype [61].
With regard to myogenesis, there has been a paucity of skeletal muscle experimentation. However a growing interest has been restored lately regarding the cardiac muscle and the cardiovascular system [62,63,64,65]. The heart begins to develop at the beginning of the 4th week (first heartbeat) from mesenchymal cells of the visceral mesoderm, which rapidly proliferates, creating the heart tubes. The heart tubes are then fused and surrounded by the primordial myocardium. During the 4th and 5th week, this structure will be divided into four chambers (atria and ventricles) to resemble the normal adult heart. Initial research was conducted to measure cardiac atrial natriuretic peptide (ANP) in rat dams and fetuses developed in space (NASA’s NIH-R1 and NIH-R2 experiments) [66]. Additional experiments in microgravity studying the developing zebrafish heart showed that there are specific gene expression changes [67]. In line with the genetic screening, human Induced Pluripotent Stem Cell–Derived Cardiomyocytes (hiPSC-CMs) were RNA-sequenced and exhibited profound differences in 2635 genes [68].
This organ-based approach can be extended further to include tissues that are metabolically active, since the space environment can compromise vital biochemical reactions with the involvement of oxidative stress, autophagy and damage [69]. For instance, Blaber and colleagues showed that exposure to space conditions for 13.5 days resulted in elevated reactive oxygen species (ROS) and activated autophagy, including the proteasome system, in the mouse liver [70]. Ramifications of this exposure revealed hepatocyte senescence triggered by a buildup of oxidized proteins and profound mitochondrial dysfunction, a common denominator when the cell homeostasis is being perturbed [70,71]. Interestingly, space radiation and microgravity can have opposite effects on specific molecular pathways. Radiation, as shown with transcriptomic analysis, inhibited autophagy, thereby promoting an aged-like phenotype, while microgravity generated a proliferative phenotype [72]. Such nuances should be taken into account to create a tailored protection that safeguards the health of astronauts.
Table 1. Synopsis of the most representative developmental occurrences and relevant research conducted to address the effect of altered gravity and radiation.
Table 1. Synopsis of the most representative developmental occurrences and relevant research conducted to address the effect of altered gravity and radiation.
CategoryDevelopmental Stage; Organ and System FormationDetails for Individual ProcessesResearch Outline and Species ExperimentationBrief OutcomeReference
1Germinal stageTime from fertilization until implantation (10 days in humans)Primordial germ cells (PGCs) from embryos of Medaka fish on the IML-2 missionGerm cells formed in space were functionally normal[73]
1.1FertilizationFusion of an egg and sperm in the ampulla of the fallopian tubeMice experiments on ISS under microgravity, gene expression determined by RNA-seq.
Microgravity on an urodele amphibian on board the MIR space station
No overt defects in spermatozoa or offspring viability. No lethal consequences of microgravity in Pleurodeles fertilized eggs were observed[74,75]
1.2CleavageDivisions of the zygoteThe location of the first horizontal cleavage was investigated in Xenopus laevis under simulated weightlessnessLate tadpoles were largely indistinguishable from controls[76]
1.3BlastulationFormation of the blastocyst; trophoblast and inner cell massSea urchin larvae developed in microgravity conditions for several days from blastula stage onwardsMicrogravity did not affect the metamorphosed individuals[77]
1.4ImplantationAttachment to the endometriumPregnant laboratory rats were laparotomized and uterine implantation sites were subjected to a simulated 10-day space shuttle flightMorphologic measurements exhibited no maternal compromise and presented normal offspring development[78]
1.5Embryonic discBilaminar embryo; epiblast and hypoblastSea urchin eggs fertilized under microgravity conditions during the Swedish sounding rocket flights MASER IV and MASER VEmbryonic and larval development produced normal advanced pluteus stages[79]
2GastrulationFormation of three germ layers (histogenesis)Development from late gastrulation (blastopore formation) in simulated microgravity was appraised using Xenopus laevis embryosMicrogravity contributed to attenuation of Hoxa2 expression and increased cell division[80,81]
3Neurulation
Neural crest migration
Folding of embryo and body cavities formation
Transformation of the neural plate (from ectoderm) into the neural tube.
Primitive gut tube is formed from endoderm lining the yolk sac
Evaluation of the long-term consequences of simulated microgravity on the neural crest cells of zebrafish embryosMicrogravity at key stages of cranial neural crest cell migration resulted in stunted growth[82]
4Organ and system developmentFrom 4th to 8th week in humans;
Organogenesis continues until birth
Essential mesenchymal-derived structures such as bone, cartilage and muscle have been studied in various gravity or radiation conditions in rodents and chicksStructural components of the extracellular matrix are altered by perturbations in gravity and radiation[48,50,51,54,61,83]
4.1BloodHaematopoietic stem cells; blood islands, first appeared in the yolk sacChanges in metabolism of human space-flown lymphocyte cultures. Additional experiments include C57BL/6 mice fetal thymus organ culture after rotation in a clinostatSpaceflight increases lymphocyte apoptosis. For T cells specifically, altered gravity decreases CD4-CD8- T cell precursors[84,85]
4.2Heart and circulatory systemEndocardial tubes from vasculogenesis; primitive myocardium from visceral mesoderm. The heart starts beating at day 22 in humansUsing transgenic zebrafish researchers investigated the gene expression pattern in the live, developing heart. Heart cell cytoskeleton structures such as fibronectin, have also been analyzed in culturesSimulated micro- and hypergravity diminish heart tissue development in vitro and alters gene expression[47,67,86]
4.3Digestive tract and accessory organsDivisions of gut tube; foregut (pharyngeal apparatus, trachea and lung buds, esophagus, stomach, duodenum -proximal to the bile duct, liver, pancreas, biliary system, midgut (duodenum -distal to the bile duct, jejunum, ileum, cecum, appendix, ascending colon, proximal 2/3 of transverse colon) and hindgut (distal third part of transverse colon, descending colon, sigmoid, rectum, upper part of anal canal, urogenital sinus).No embryologic data available to date.
Human adult NASA twins study evaluating 1-year-long stay in ISS and then compared miscellaneous parameters with earth control sibling.
Animal data utilizing fecal mice samples showcasing liver transcriptome alterations; mice gut metabolome
Microgravity and radiation seem to affect the gut microbiome composition in adults; dietary regimes have been proposed[87,88,89]
4.4Respiratory system
(lung formation)
Early branching of the primitive bronchial buds; alveoli continue to develop up until early adulthoodEmbryonic mouse lung was cultured in order to investigate development and morphogenesis during orbital spaceflight (mission STS-54); rat diaphragm and intercostal muscles were also evaluated from the same missionLung rudiments continued to grow and branch in microgravity. Oxidative and antioxidant enzyme levels of respiratory muscles were different[90,91]
4.5Urogenital systemOrgans of both urinary and genital system, intermediate mesodermKnockout mice on ISSModifications in urogenital elements[92]
4.5.1Urinary systemIntermediate mesodermDevelopment of knockout mice and study in microgravity on ISSElevated concentrations of fatty acids in the mouse plasma[92]
4.5.1.1KidneysPronephros (disappears early), mesonephros and metanephrosA study investigated the contribution of Nrf2 to kidney function in mice. Suspensions of cultured primary human embryonic kidney cells were subjected to electrophoresis on Space Shuttle flight STS-8 (Challenger) missionKidneys seem to play a central role in adaptation to gravitational changes. Changes are reflected to the cultured kidney cells. Risk for stone formation in adults[92,93,94,95]
4.5.1.2Ureters and urinary bladderUreters derive from metanephros whereas bladder derives partly from cloaca; allantois does not participateNo embryologic data available to date. 24-h urine samples were collected prior to, during space flight, and following landing. Urine composition and volume were evaluated as wellIncreased risk of stone formation in astronauts[96,97,98]
4.5.2Reproductive systemWolffian (males) and Müllerian (females) ductsA clinostat was used to emulate microgravity while incubating fertilized chicken and quail eggs. PGCs were counted during early development, and the ability of quail to reproduce after being hatched in a microgravity simulator was examinedSimulated microgravity may hinder gonadial development and lower fertility based on the low number of spermatogonia[99]
4.5.2.1MaleScrotum, phallus, testesTestes from mouse embryos and prepuberal rats were cultured in simulated microgravity and then morphological characteristics and hormonal levels were evaluated. Spermatozoa from caged male mice on ISSMicrogravity disorganized the gonadal tissues (testis cords). Leydig cell survival is severely affected by simulated microgravity[74,100,101,102]
4.5.2.2FemaleLabia, clitoris, ovariesCultured ovaries from 14-day-old mice, or preantral follicles in a simulated microgravity environment. Follicle diameter and oocyte diameter were evaluated.Microgravity is detrimental to the initial stage development of mouse preantral follicles[103,104]
4.6Nervous systemNeural tube (Central Nervous System; CNS) and neural crest (Peripheral Nervous System; PNS)Experimental data using Xenopus laevis oocytes and sperm in a sounding rocket hypergravity (centrifuge) or virtual microgravity (clinostat)Amphibian eggs fertilized in microgravity (both in vivo and in vitro) exhibited abnormalities during embryonic nervous system development but were able to produce functional larvae[105]
4.6.1CNS (Brain and spinal cord)Secondary brain vesicles—1. telencephalon (future cerebral cortex and basal ganglia), 2. diencephalon (future thalamus and hypothalamus), 3. mesencephalon (future colliculi), 4. metencephalon (future pons and cerebellum), and 5. myelencephalon (future medulla);
alar plate in spinal cord will form the dorsal (posterior -sensory) horns whereas basal plate will give rise to (anterior-motor) horns
Researchers examined the effects of 2 g hypergravity for 2 weeks on gene expression of brain-derived neurotrophic factor (BDNF) and serotonin receptors in the mouse brain. Recently mouse ventral lumbar spinal cord, which is rich in motor neurons, was tested under microgravity conditions. Cerebellar and brainstem adaptations were witnessed in rodents. Cricket insects were challenged in gravity variations.
Radiation effect in the CNS was also accessed using chick embryo neural explants
Gravity changes induced differential expression of BDNF and serotonin receptors genes in specific brain regions. Microgravity also contributes to motor neuron degeneration in the spinal cord. Radiation influences neuritogenesis[6,21,36,39,106,107,108]
4.6.2PNS (Peripheral nerves and ganglia)Neural crest migrates to form e.g., dorsal root ganglia and enteric plexiSimulated weightlessness during early ontogeny of cichlid fish and Xenopus laevis.
Simulated microgravity on cranial neural crest cells in zebrafish embryos. Vestibular peripheral connections alterations in the molecular level in mice
Pronounced variations in the composition of gangliosides[45,46,82,109]
4.7Integumentary system
(skin, nails, hair)
Epidermis is derived from the ectoderm whereas dermis is derived from mesenchymeIn vitro and in vivo studies in experimental microgravity set-ups to study keratinocyte growth rate and migrationMicrogravity interferes with epidermal repair process and negatively influences skin wound healing[110]
5Other sensory organsFormed, in part, from ectodermal thickenings: the placodesThe vestibulo-ocular reflex was evaluated in rats under hypergravity conditionsA gravitational change affects the vestibular input triggering ocular reflexes[111]
5.1EyesBegin to develop from the third week from two grooves on each side of the forebrain, which form the optic vesiclesImpact of spaceflight and artificial gravity was investigated on the mouse retina with proteomic analyses and gene expression in the lens of developing zebrafishSpaceflight induces retinal apoptosis and miscellaneous lens-specific gene expression level changes[47,112,113]
5.2EarsInner, middle and outer with distinct embryological originsZebrafish eggs, anatomical and behavioral studies of embryonic rats that developed in microgravityThere is a critical period for functional inner ear development[9,41]
5.2.1Inner earDevelops from otic placodes on each side of rhombencephalon, inducing the formation of otic pits and otic vesicles.
The saccule and the cochlear duct originate from the ventral component whereas utricle and semicircular canals derive from the dorsal part
The most abundantly studied structure in many different species at the embryonic level under microgravity, hypergravity and radiation conditions. Otolith-deficient mutant mice were utilized. Vestibular development was investigated in zebrafish. Additionally, swordtail fish flown in space on STS-89 and STS-90 and birds flown as embryos for 5 days in earth orbit (STS-29).Gravity alterations drastically affect the vestibular system and more specifically otolith formation.
Fish otoliths reared in space were significantly larger than controls.
However, no major gross morphology alterations were observed in birds.
[6,9,17,20,21,22,41,42]
5.2.2Middle earTympanic cavity and eustachian tube are generated from the 1st pharyngeal pouch while ossicles (malleus, incus, and stapes) derive from the cartilages of the 1st and 2nd pharyngeal archesNo available experimental data
5.2.3Outer earExternal auditory meatus (ear canal) is derived from the first pharyngeal cleft and auricle (pinna) from mesenchyme of 1st and 2nd pharyngeal archesNo available experimental data

5. Gut Microbiome, Cosmonauts and Space Travel as an Emerging, Indispensable Trifecta

During the 3rd to 4th week of gestation, beneath the neural tube a second tube is being formed (primitive gut tube), and as cephalocaudal folding of the embryo occurs, anterior and posterior endodermal elements of the yolk sac give rise to foregut and hindgut, respectively. Later on, membranes that prevent the connection with the amniotic fluid (oropharyngeal and cloacal membranes) break down to form the mouth and the anus. The fetus presumably develops with a relatively sterile gut [114], devoid of microbiota (i.e., bacteria, viruses, and fungi) and their microbiome, which is the collection of genomes from all the microorganisms [115]. However, recent microbiota studies [116,117,118,119,120,121] reported a physiological bacterial presence along with metabolites that were produced either directly by the microbiota in utero or by transplacental trafficking through the mother’s bloodstream [122,123]. In addition, maternal cervicovaginal microbes that populate the birth canal can be passed on naturally to the delivered baby [124], whereas breastfeeding provides a constant supply of maternal microbiota after birth [125].
Among different body sites, the intestinal microbiota seems to be the most important. Research is being conducted on the ISS in addition to that simulated on earth, pertaining to the astronauts’ healthy gut [126]. To achieve this, space dietary regimes have been proposed [127], although astronauts’ caloric intake can deviate from baseline to as low as 70–80% during the first days of flight due to space motion sickness, a set of symptoms that impair operational performance and appear upon initial encounter with microgravity [128]. More than 90% of the human gut microbiota is represented by Firmicutes and Bacteroidetes (or Bacteroidota) phyla [129]. Firmicutes are heavily associated with metabolism and the nutritional status via short-chain fatty acid (SCFA) synthesis, including acetate, propionate, butyric acid and caproic acid [130], whereas Bacteroidetes are more involved in immunomodulation. It has also been suggested that the predominant gut bacteria found in a developing fetus are Bacteriodetes rather than Firmicutes, and this ratio is reversed as we grow older [131].
Due to the novelty of this research niche, microbiome-related embryological data are almost nonexistent at this time. Nevertheless, space-related factors have been shown to alter the microbiome’s profile [88,132]. Besides one or two studies conducted in the late 1970s, significant experiments started in 2015 with 16S rRNA gene amplicon sequencing to study the human host microbiome [133]. More studies followed over the next years, with the hallmark MARS500 study analyzing human fecal samples before spaceflight and continuing for 6 months [134], as well as the Hawai’i Space Exploration Analog and Simulation (Hi-SEAS) simulations. Probably the largest microbiota- and microbiome-based experimentation is the “NASA Twins Study” sequencing genomic DNA [87]. The authors declared that while gut microbiota composition and function were altered during spaceflight, microbiome heterogeneity remained unchanged. Mice experiments also started the same year in a simulation environment [135] and on the ISS [88]. More ISS investigations followed [132,136,137,138,139], and more are on the way as we progress toward the challenging Mars travel set for the late 2030s or early 2040s.

6. Fertility and Reproduction Repercussions

Last but not least, the endocrine system and, more important, the gonadal axis of it is listed among the systems affected by altered gravity and radiation [140,141]. In plain terms, the levels of radiation on the ISS are 100 times higher than those naturally occurring on Earth [142]. Therefore, exposure to ionizing radiation poses an inherent risk to astronauts who engage in space missions [143], suggesting even transgenerational inheritance of mutations to offspring [144]. There are miscellaneous effects on sex and gender [145] that mostly affect females, potentially through cumulative oxidative stress [146] or depletion of cellular reserves leading to ovarian shrinkage [13]. Additionally, gynecological issues include hormonal menstrual suppression [147,148]. In contrast, spermatozoa are radio-resistant, allowing the restoration of spermatogenesis [149]. Nevertheless, long-term exposure to microgravity leads to buffered serum testosterone levels [150] and atrophy of seminiferous tubules [151].
To assess the effects of radiation on mammalian germ cells, mice male gametes were kept for several months on the ISS and then tested for their ability to fertilize healthy oocytes on Earth. Researchers showed that although there was profound DNA damage to the spermatozoa, this did not affect the procreation and rearing of progeny [142]. More experiments were carried out in LEO with invertebrates, fish, reptiles and amphibians [75,152,153]. Collectively, these studies investigated the fertilization profile from the zygote and blastocyst level up to the delivery outcome. For sea urchin larvae, no abnormalities were conferred [79], whereas increased thickness on the top of the blastocyst in Xenopus laevis was recorded, although normal larvae were produced [152]. Medaka killifish were the first vertebrates to successfully mate in space during a 15-day mission, resulting in the production of 43 eggs, 8 of which hatched in space and the remaining after landing on earth [73]. All in all, as seen in rats, several facets of pregnancy, parturition and early mammalian developmental milestones can be achieved while gravity is perturbed [154,155]; however, species complexity is always a catalytic factor.

7. Conclusions—Perspectives

This literature review summarizes the data provided from experiments in low Earth orbit (such as from the ISS) or from expeditions in deeper space, categorized on the basis of tissue and organ morphogenesis and respective alterations. Many of the studies refer to the very early embryonic stage and explore the adaptability of gonads, fertility and how the zygote comes into creation, whereas others study the adaptability of already formed organs achieving homeostasis and how they react to microgravity or cosmic radiation. The latter is critically applicable to astronauts and advances our knowledge for deep-space exploration [156,157]. Through this review we also delineated the gaps in current knowledge, especially in fields that are newly unveiled, such as the influence of gut microbiome on successful space missions. From a translational viewpoint, along with the rise of “-omics” techniques, advanced research tools are highlighted and open up new horizons in quick, accurate and efficient tests with practical applications for monitoring in real time the genome, transcriptome or proteome of a recruited astronaut.
However, someone can argue that limitations accompany the narrative of this trajectory. Ethical reasons do not allow extensive manipulations, currently prohibited in humans, and even if this were permitted it would literally take years of monitoring to identify the experimental effect on any tissue or organ system. Additionally, one can premise that most of the experiments are performed on larvae, crickets or amphibians that do not emulate human biology, per se. As highlighted in the current review, these model organisms have been widely employed by NASA to assess biological risks associated with space travel (e.g., the vestibular system’s adaptation performed on STS-40 (Spacelab-1), STS-58 (Spacelab-2), and STS-90 (Neurolab)) and bone formation, to capitalize on human exploration and benefit society on Earth [158]. Additionally, space medicine may sound utopian, but miniaturized organoids on chips testing drugs [159] and cancer studies have been designed under space conditions [160] already, postulating promising results. This is a starting point nonetheless, justifying more investigations if we ever want to succeed in interstellar exploration and colonization.

Author Contributions

P.T., writing—original draft preparation; S.M., conceptualization; M.E.M., T.-E.D. and D.M., review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

NASANational Aeronautics and Space Administration
ISSInternational Space Station
LEOLow Earth Orbit
hMSCshuman Mesenchymal Stem Cells
ANPAtrial Natriuretic Peptide
hiPSC-CMhuman induced Pluripotent Stem Cell–Derived Cardiomyocytes
Hi-SEASHawaii Space Exploration Analog and Simulation
PGCsPrimordial Germ Cells
SCFAShort-chain fatty acids
ROSReactive oxygen species
BDNFBrain-derived neurotrophic factor

References

  1. Warren, L.E. International Space Station Open-Source Data. Patterns 2020, 1, 100172. [Google Scholar] [CrossRef]
  2. Schlagheck, R.A.; Trach, B.L. Microgravity research results and experiences from the NASA/MIR space station program. Acta Astronaut. 2003, 53, 983–996. [Google Scholar] [CrossRef]
  3. Thirsk, R.; Kuipers, A.; Mukai, C.; Williams, D. The space-flight environment: The International Space Station and beyond. CMAJ 2009, 180, 1216–1220. [Google Scholar] [CrossRef] [Green Version]
  4. Edwards, B.F. Weightlessness experiments on Biosatellite II. Life Sci. Space Res. 1969, 7, 84–92. [Google Scholar]
  5. Stella, A.B.; Ajčević, M.; Furlanis, G.; Manganotti, P. Neurophysiological adaptations to spaceflight and simulated microgravity. Clin. Neurophysiol. 2021, 132, 498–504. [Google Scholar] [CrossRef] [PubMed]
  6. Bruce, L. Adaptations of the vestibular system to short and long-term exposures to altered gravity. Adv. Space Res. 2003, 32, 1533–1539. [Google Scholar] [CrossRef] [PubMed]
  7. Van Ombergen, A.; Demertzi, A.; Tomilovskaya, E.; Jeurissen, B.; Sijbers, J.; Kozlovskaya, I.B.; Parizel, P.M.; Van de Heyning, P.H.; Sunaert, S.; Laureys, S.; et al. The effect of spaceflight and microgravity on the human brain. J. Neurol. 2017, 264 (Suppl. 1), 18–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Carriot, J.; Mackrous, I.; Cullen, K.E. Challenges to the Vestibular System in Space: How the Brain Responds and Adapts to Microgravity. Front. Neural Circuits 2021, 15, 760313. [Google Scholar] [CrossRef] [PubMed]
  9. Ronca, A.E.; Fritzsch, B.; Alberts, J.R.; Bruce, L.L. Effects of microgravity on vestibular development and function in rats: Genetics and environment. Korean J. Biol. Sci. 2000, 4, 215–221. [Google Scholar] [CrossRef] [PubMed]
  10. Jamon, M. The development of vestibular system and related functions in mammals: Impact of gravity. Front. Integr. Neurosci. 2014, 8, 11. [Google Scholar] [CrossRef] [Green Version]
  11. Klein-Nulend, J.; Bacabac, R.G.; Veldhuijzen, J.P.; Van Loon, J.J.W.A. Microgravity and bone cell mechanosensitivity. Adv. Space Res. 2003, 32, 1551–1559. [Google Scholar] [CrossRef] [PubMed]
  12. Fitts, R.H.; Riley, D.R.; Widrick, J.J. Functional and structural adaptations of skeletal muscle to microgravity. J. Exp. Biol. 2001, 204, 3201–3208. [Google Scholar] [CrossRef] [PubMed]
  13. Mishra, B.; Luderer, U. Reproductive hazards of space travel in women and men. Nat. Rev. Endocrinol. 2019, 15, 713–730. [Google Scholar] [CrossRef] [PubMed]
  14. Angelaki, D.E.; Cullen, K.E. Vestibular System: The Many Facets of a Multimodal Sense. Annu. Rev. Neurosci. 2008, 31, 125–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Mackowetzky, K.; Yoon, K.H.; Mackowetzky, E.J.; Waskiewicz, A.J. Development and evolution of the vestibular apparatuses of the inner ear. J. Anat. 2021, 239, 801–828. [Google Scholar] [CrossRef] [PubMed]
  16. Beisel, K.W.; Wang-Lundberg, Y.; Maklad, A.; Fritzsch, B. Development and evolution of the vestibular sensory apparatus of the mammalian ear. J. Vestib. Res. 2005, 15, 225–241. [Google Scholar] [CrossRef]
  17. Wiederhold, M.L.; Harrison, J.L.; Gao, W. A critical period for gravitational effects on otolith formation. J. Vestib. Res. 2003, 13, 205–214. [Google Scholar] [CrossRef]
  18. Wiederhold, M.L.; Harrison, J.L.; Parker, K.; Nomura, H. Otoliths developed in microgravity. J. Gravit. Physiol. J. Int. Soc. Gravit. Physiol. 2000, 7, 39–42. [Google Scholar]
  19. Ronca, A.; Fritzsch, B.; Bruce, L.L.; Alberts, J.R. Orbital spaceflight during pregnancy shapes function of mammalian vestibular system. Behav. Neurosci. 2008, 122, 224–232. [Google Scholar] [CrossRef] [Green Version]
  20. Jones, T.A.; Fermin, C.; Hester, P.Y.; Vellinger, J. Effects of Microgravity on Vestibular Ontogeny: Direct Physiological and Anatomical Measurements following Space Flight (STS-29). Acta Vet. 1993, 62, S35–S42. [Google Scholar] [CrossRef]
  21. Bruce, L.L.; Fritzsch, B. The development of vestibular connections in rat embryos in microgravity. J. Gravit. Physiol. J. Int. Soc. Gravit. Physiol. 1997, 4, 59–62. [Google Scholar]
  22. Demêmes, D.; Dechesne, C.J.; Venteo, S.; Gaven, F.; Raymond, J. Development of the rat efferent vestibular system on the ground and in microgravity. Dev. Brain Res. 2001, 128, 35–44. [Google Scholar] [CrossRef] [PubMed]
  23. Nguon, K.; Ladd, B.; Sajdel-Sulkowska, E. Exposure to altered gravity during specific developmental periods differentially affects growth, development, the cerebellum and motor functions in male and female rats. Adv. Space Res. 2006, 38, 1138–1147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Sajdel-Sulkowska, E.M.; Li, G.-H.; Ronca, A.E.; Baer, L.A.; Sulkowski, G.M.; Koibuchi, N.; Wade, C.E. Effects of hypergravity exposure on the developing central nervous system: Possible involvement of thyroid hormone. Exp. Biol. Med. 2001, 226, 790–798. [Google Scholar] [CrossRef] [PubMed]
  25. Nguon, K.; Li, G.-H.; Sajdel-Sulkowska, E. CNS development under altered gravity: Cerebellar glial and neuronal protein expression in rat neonates exposed to hypergravity. Adv. Space Res. 2004, 33, 1375–1380. [Google Scholar] [CrossRef]
  26. Sajdel-Sulkowska, E.M.; Nguon, K.; Sulkowski, Z.L.; Rosen, G.D.; Baxter, M.G. Purkinje cell loss accompanies motor impairment in rats developing at altered gravity. Neuroreport 2005, 16, 2037–2040. [Google Scholar] [CrossRef]
  27. Bouët, V.; Wubbels, R.; de Jong, H.; Gramsbergen, A. Behavioural consequences of hypergravity in developing rats. Dev. Brain Res. 2004, 153, 69–78. [Google Scholar] [CrossRef]
  28. Sarkar, P.; Sarkar, S.; Ramesh, V.; Hayes, B.E.; Thomas, R.L.; Wilson, B.L.; Kim, H.; Barnes, S.; Kulkarni, A.; Pellis, N.; et al. Proteomic Analysis of Mice Hippocampus in Simulated Microgravity Environment. J. Proteome Res. 2006, 5, 548–553. [Google Scholar] [CrossRef] [Green Version]
  29. Newberg, A.B. Changes in the central nervous system and their clinical correlates during long-term spaceflight. Aviat. Space Environ. Med. 1994, 65, 562–572. [Google Scholar]
  30. Tafforin, C. Relationships between orientation, movement and posture in weightlessness: Preliminary ethological observations. Acta Astronaut. 1990, 21, 271–280. [Google Scholar] [CrossRef]
  31. Yates, B.; Holmes, M.; Jian, B. Plastic changes in processing of graviceptive signals during spaceflight potentially contribute to postflight orthostatic intolerance. J. Vestib. Res. 2003, 13, 395–404. [Google Scholar] [CrossRef] [PubMed]
  32. Reschke, M.F.; Bloomberg, J.J.; Harm, D.L.; Paloski, W.H.; Layne, C.; McDonald, V. Posture, locomotion, spatial orientation, and motion sickness as a function of space flight. Brain Res. Rev. 1998, 28, 102–117. [Google Scholar] [CrossRef] [PubMed]
  33. Shipov, A.A.; Aizikov, G.S. Physical state, equilibrium function and vestibulo- motor reflexes in the rats after space flights on board biosatellites of Cosmos series. Physiologist 1992, 35 (Suppl. 1), S206–S207. [Google Scholar] [PubMed]
  34. Mani-Ponset, L.; Masseguin, C.; Davet, J.; Herbuté, S.; Maurel, D.; Ghandour, M.; Reiss-Bubenheim, D.; Güell, A.; Gabrion, J. Effects of an 11-day spaceflight on the choroid plexus of developing rats. Dev. Brain Res. 1997, 99, 187–200. [Google Scholar] [CrossRef] [PubMed]
  35. Johnson, C.C. The Biological Flight Research Facility. Microgravity Q. MGQ 1992, 2, 115–121. [Google Scholar] [PubMed]
  36. Vazquez, M.; Broglio, T.; Worgul, B.; Benton, E. Neuritogenesis: A model for space radiation effects on the central nervous system. Adv. Space Res. 1994, 14, 467–474. [Google Scholar] [CrossRef] [PubMed]
  37. Gualandris-Parisot, L.; Husson, D.; Foulquier, F.; Kan, P.; Davet, J.; Aimar, C.; Dournon, C.; Duprat, A. Pleurodeles waltl, amphibian, Urodele, is a suitable biological model for embryological and physiological space experiments on a vertebrate. Adv. Space Res. 2001, 28, 569–578. [Google Scholar] [CrossRef] [PubMed]
  38. Horn, E.; Agricola, H.; Böser, S.; Förster, S.; Kämper, G.; Riewe, P.; Sebastian, C. Crickets in space: Morphological, physiological and behavioral alterations induced by space flight and hypergravity. Adv. Space Res. 2002, 30, 819–828. [Google Scholar] [CrossRef]
  39. Horn, E.; Böser, S.; Förster, S.; Riewe, P.; Sebastian, C.; Agricola, H. Crickets in space. Acta Astronaut. 2001, 49, 345–363. [Google Scholar] [CrossRef]
  40. Anken, R.; Ibsch, M.; Rahmann, H. Microgravity (STS-90 Neurolab-Mission) influences synapse formation in a vestibular nucleus of fish brain. Adv. Space Res. 2002, 30, 843–847. [Google Scholar] [CrossRef]
  41. Moorman, S.J.; Cordova, R.; Davies, S.A. A critical period for functional vestibular development in zebrafish. Dev. Dyn. 2002, 223, 285–291. [Google Scholar] [CrossRef] [PubMed]
  42. Ijiri, K.; Mizuno, R.; Eguchi, H. Use of an otolith-deficient mutant in studies of fish behavior in microgravity. Adv. Space Res. 2003, 32, 1501–1512. [Google Scholar] [CrossRef] [PubMed]
  43. Anken, R.H.; Kappel, T.; Rahmann, H. Morphometry of fish inner ear otoliths after development at 3g hypergravity. Acta Otolaryngol. 1998, 118, 534–539. [Google Scholar] [PubMed]
  44. Rahmann, H.; Hilbig, R.; Flemming, J.; Slenzka, K. Influence of long-term altered gravity on the swimming performance of developing cichlid fish: Including results from the 2nd German Spacelab Mission D-2. Adv. Space Res. 1996, 17, 121–124. [Google Scholar] [CrossRef] [PubMed]
  45. Sebastian, C.; Pfau, K.; Horn, E. An age-dependent sensitivity of the roll-induced vestibuloocular reflex to hypergravity exposure of several days in an amphibian (Xenopus laevis). Acta Astronaut. 1998, 42, 419–430. [Google Scholar] [CrossRef] [PubMed]
  46. Fritzsch, B. Molecular developmental neurobiology of formation, guidance and survival of primary vestibular neurons. Adv. Space Res. 2003, 32, 1495–1500. [Google Scholar] [CrossRef]
  47. Shimada, N.; Sokunbi, G.; Moorman, S.J. Changes in gravitational force affect gene expression in developing organ systems at different developmental times. BMC Dev. Biol. 2005, 5, 10. [Google Scholar] [CrossRef] [Green Version]
  48. Zérath, E. Effects of microgravity on bone and calcium homeostasis. Adv. Space Res. 1998, 21, 1049–1058. [Google Scholar] [CrossRef]
  49. Burger, E.; Klein-Nulend, J. Microgravity and Bone Cell Mechanosensitivity. Bone 1998, 22 (Suppl. 5), 127S–130S. [Google Scholar] [CrossRef]
  50. Klement, B.J.; Spooner, B.S. Pre-metatarsal skeletal development in tissue culture at unit- and microgravity. J. Exp. Zool. 1994, 269, 230–241. [Google Scholar] [CrossRef]
  51. Beck, M.; Moreels, M.; Quintens, R.; Abou-El-Ardat, K.; El-Saghire, H.; Tabury, K.; Michaux, A.; Janssen, A.; Neefs, M.; VAN Oostveldt, P.; et al. Chronic exposure to simulated space conditions predominantly affects cytoskeleton remodeling and oxidative stress response in mouse fetal fibroblasts. Int. J. Mol. Med. 2014, 34, 606–615. [Google Scholar] [CrossRef] [Green Version]
  52. Carmeliet, G.; Nys, G.; Bouillon, R. Microgravity Reduces the Differentiation of Human Osteoblastic MG-63 Cells. J. Bone Miner. Res. 1997, 12, 786–794. [Google Scholar] [CrossRef]
  53. Cazzaniga, A.; Maier, J.A.; Castiglioni, S. Impact of simulated microgravity on human bone stem cells: New hints for space medicine. Biochem. Biophys. Res. Commun. 2016, 473, 181–186. [Google Scholar] [CrossRef]
  54. Crawford-Young, S.J. Effects of microgravity on cell cytoskeleton and embryogenesis. Int. J. Dev. Biol. 2006, 50, 183–191. [Google Scholar] [CrossRef]
  55. Doty, S.B.; Stiner, D.; Telford, W.G. The effect of spaceflight on cartilage cell cycle and differentiation. J. Gravit. Physiol. J. Int. Soc. Gravit. Physiol. 1999, 6, 89–90. [Google Scholar]
  56. Nakaji-Hirabayashi, T.; Matsumura, K.; Ishihara, R.; Ishiguro, T.; Nasu, H.; Kanno, M.; Ichida, S.; Hatashima, T. Enhanced proliferation and differentiation of human mesenchymal stem cells in the gravity-controlled environment. Artif. Organs 2022, 46, 1760–1770. [Google Scholar] [CrossRef] [PubMed]
  57. Kacena, M.A.; Todd, P.; Gerstenfeld, L.C.; Landis, W.J. Experiments with osteoblasts cultured under hypergravity conditions. Microgravity Sci. Technol. 2004, 15, 28–34. [Google Scholar] [CrossRef] [PubMed]
  58. Zayzafoon, M.; Gathings, W.E.; McDonald, J.M. Modeled Microgravity Inhibits Osteogenic Differentiation of Human Mesenchymal Stem Cells and Increases Adipogenesis. Endocrinology 2004, 145, 2421–2432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Zhang, C.; Li, L.; Jiang, Y.; Wang, C.; Geng, B.; Wang, Y.; Chen, J.; Liu, F.; Qiu, P.; Zhai, G.; et al. Space microgravity drives transdifferentiation of human bone marrow-derived mesenchymal stem cells from osteogenesis to adipogenesis. FASEB J. 2018, 32, 4444–4458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Smith, S.M.; Heer, M.; Shackelford, L.C.; Sibonga, J.D.; Spatz, J.; Pietrzyk, R.A.; Hudson, E.K.; Zwart, S.R. Bone metabolism and renal stone risk during International Space Station missions. Bone 2015, 81, 712–720. [Google Scholar] [CrossRef] [PubMed]
  61. Landis, W.J.; Hodgens, K.J.; Block, D.; Toma, C.D.; Gerstenfeld, L.C. Spaceflight Effects on Cultured Embryonic Chick Bone Cells. J. Bone Miner. Res. 2000, 15, 1099–1112. [Google Scholar] [CrossRef] [PubMed]
  62. Baran, R.; Marchal, S.; Campos, S.G.; Rehnberg, E.; Tabury, K.; Baselet, B.; Wehland, M.; Grimm, D.; Baatout, S. The Cardiovascular System in Space: Focus on In Vivo and In Vitro Studies. Biomedicines 2021, 10, 59. [Google Scholar] [CrossRef] [PubMed]
  63. Vernice, N.A.; Meydan, C.; Afshinnekoo, E.; Mason, C.E. Long-term spaceflight and the cardiovascular system. Precis. Clin. Med. 2020, 3, 284–291. [Google Scholar] [CrossRef] [PubMed]
  64. Fu, Q.; Shibata, S.; Hastings, J.L.; Platts, S.H.; Hamilton, D.M.; Bungo, M.W.; Stenger, M.B.; Ribeiro, C.; Adams-Huet, B.; Levine, B.D. Impact of Prolonged Spaceflight on Orthostatic Tolerance During Ambulation and Blood Pressure Profiles in Astronauts. Circulation 2019, 140, 729–738. [Google Scholar] [CrossRef]
  65. Ly, V.; Velichala, S.R.; Hargens, A.R. Cardiovascular, Lymphatic, and Ocular Health in Space. Life 2022, 12, 268. [Google Scholar] [CrossRef] [PubMed]
  66. Davet, J.; Fagette, S.; Mani-Ponset, L.; Bayard, B.; Dumars, P.; Reiss-Bubenheim, D.; Güell, A.; Gharib, C.; Gabrion, J. Cardiac atrial natriuretic peptide (ANP) in rat dams and fetuses developed in space (NIH-R1 and NIH-R2 experiments). Life Sci. 1999, 64, 1533–1541. [Google Scholar] [CrossRef]
  67. Gillette-Ferguson, I.; Ferguson, D.; Poss, K.; Moorman, S. Changes in gravitational force induce alterations in gene expression that can be monitored in the live, developing zebrafish heart. Adv. Space Res. 2003, 32, 1641–1646. [Google Scholar] [CrossRef]
  68. Wnorowski, A.; Sharma, A.; Chen, H.; Wu, H.; Shao, N.-Y.; Sayed, N.; Liu, C.; Countryman, S.; Stodieck, L.S.; Rubins, K.H.; et al. Effects of Spaceflight on Human Induced Pluripotent Stem Cell-Derived Cardiomyocyte Structure and Function. Stem Cell Rep. 2019, 13, 960–969. [Google Scholar] [CrossRef] [Green Version]
  69. Goodwin, T.J.; Christofidou-Solomidou, M. Oxidative Stress and Space Biology: An Organ-Based Approach. Int. J. Mol. Sci. 2018, 19, 959. [Google Scholar] [CrossRef] [Green Version]
  70. Blaber, E.A.; Pecaut, M.J.; Jonscher, K.R. Spaceflight Activates Autophagy Programs and the Proteasome in Mouse Liver. Int. J. Mol. Sci. 2017, 18, 2062. [Google Scholar] [CrossRef]
  71. Nguyen, H.P.; Tran, P.H.; Kim, K.-S.; Yang, S.-G. The effects of real and simulated microgravity on cellular mitochondrial function. NPJ Microgravity 2021, 7, 44. [Google Scholar] [CrossRef] [PubMed]
  72. Barravecchia, I.; De Cesari, C.; Forcato, M.; Scebba, F.; Pyankova, O.V.; Bridger, J.M.; Foster, H.A.; Signore, G.; Borghini, A.; Andreassi, M.; et al. Microgravity and space radiation inhibit autophagy in human capillary endothelial cells, through either opposite or synergistic effects on specific molecular pathways. Cell. Mol. Life Sci. 2021, 79, 28. [Google Scholar] [CrossRef] [PubMed]
  73. Ijiri, K. Development of space-fertilized eggs and formation of primordial germ cells in the embryos of medaka fish. Adv. Space Res. 1998, 21, 1155–1158. [Google Scholar] [CrossRef] [PubMed]
  74. Matsumura, T.; Noda, T.; Muratani, M.; Okada, R.; Yamane, M.; Isotani, A.; Kudo, T.; Takahashi, S.; Ikawa, M. Male mice, caged in the International Space Station for 35 days, sire healthy offspring. Sci. Rep. 2019, 9, 13733. [Google Scholar] [CrossRef] [Green Version]
  75. Aimar, C.; Bautz, A.; Durand, D.; Membre, H.; Chardard, D.; Gualandris-Parisot, L.; Husson, D.; Dournon, C. Microgravity and Hypergravity Effects on Fertilization of the Salamander Pleurodeles waltl (Urodele Amphibian). Biol. Reprod. 2000, 63, 551–558. [Google Scholar] [CrossRef]
  76. Neff, A.W.; Yokota, H.; Chung, H.-M.; Wakahara, M.; Malacinski, G.M. Early Amphibian (Anuran) Morphogenesis Is Sensitive to Novel Gravitational Fields. Dev. Biol. 1993, 155, 270–274. [Google Scholar] [CrossRef]
  77. Marthy, H.; Gasset, G.; Tixador, R.; Eche, B.; Schatt, P.; Dessommes, A.; Marthy, U.; Bacchieri, R. Skeletogenesis in sea urchin larvae under modified gravity conditions. Adv. Space Res. 1998, 21, 1151–1154. [Google Scholar] [CrossRef]
  78. Alberts, J.R.; Burden, H.W.; Hawes, N.; Ronca, A.E. Sampling of prenatal and postnatal offspring from individual rat dams enhances animal use without compromising development. Contemp. Top. Lab. Anim. Sci. 1996, 35, 61–65. [Google Scholar]
  79. Marthy, H.-J.; Schatt, P.; Santella, L. Fertilization of sea urchin eggs in space and subsequent development under normal conditions. Adv. Space Res. 1994, 14, 197–208. [Google Scholar] [CrossRef]
  80. Olson, W.M.; Wiens, D.J.; Gaul, T.L.; Rodriguez, M.; Hauptmeier, C.L. Xenopus development from late gastrulation to feeding tadpole in simulated microgravity. Int. J. Dev. Biol. 2010, 54, 167–174. [Google Scholar] [CrossRef] [Green Version]
  81. Komazaki, S. Experimental analysis of gravitational effects on amphibian gastrulation. Biol. Sci. Space 2002, 16, 41–46. [Google Scholar] [CrossRef] [Green Version]
  82. Edsall, S.C.; Franz-Odendaal, T.A. An Assessment of the Long-Term Effects of Simulated Microgravity on Cranial Neural Crest Cells in Zebrafish Embryos with a Focus on the Adult Skeleton. PLoS ONE 2014, 9, e89296. [Google Scholar] [CrossRef] [PubMed]
  83. Hardman, P.; Spooner, B.S. Collagen in Organ Development. Trans. Kans. Acad. Sci. 1992, 95, 29. [Google Scholar] [CrossRef]
  84. Lewis, M.L.; Reynolds, J.L.; Cubano, L.A.; Hatton, J.P.; Lawless, B.D.; Piepmeier, E.H. Spaceflight alters microtubules and increases apoptosis in human lymphocytes (Jurkat). FASEB J. 1998, 12, 1007–1018. [Google Scholar] [CrossRef] [PubMed]
  85. Woods, C.C.; Banks, K.E.; Gruener, R.; DeLuca, D. Loss of T cell precursors after spaceflight and exposure to vector-averaged gravity. FASEB J. 2003, 17, 1526–1528. [Google Scholar] [CrossRef]
  86. Lwigale, P.Y.; Thurmond, J.E.; Norton, W.N.; Spooner, B.S.; Wiens, D.J. Simulated Microgravity and Hypergravity Attenuate Heart Tissue Development in Explant Culture. Cells Tissues Organs 2000, 167, 171–183. [Google Scholar] [CrossRef] [PubMed]
  87. Garrett-Bakelman, F.E.; Darshi, M.; Green, S.J.; Gur, R.C.; Lin, L.; Macias, B.R.; McKenna, M.J.; Meydan, C.; Mishra, T.; Nasrini, J.; et al. The NASA Twins Study: A multidimensional analysis of a year-long human spaceflight. Science 2019, 364, eaau8650. [Google Scholar] [CrossRef]
  88. Jiang, P.; Green, S.J.; Chlipala, G.E.; Turek, F.W.; Vitaterna, M.H. Reproducible changes in the gut microbiome suggest a shift in microbial and host metabolism during spaceflight. Microbiome 2019, 7, 113. [Google Scholar] [CrossRef]
  89. Casero, D.; Gill, K.; Sridharan, V.; Koturbash, I.; Nelson, G.; Hauer-Jensen, M.; Boerma, M.; Braun, J.; Cheema, A.K. Space-type radiation induces multimodal responses in the mouse gut microbiome and metabolome. Microbiome 2017, 5, 105. [Google Scholar] [CrossRef]
  90. Spooner, B.S.; Hardman, P.; Paulsen, A. Gravity in mammalian organ development: Differentiation of cultured lung and pancreas rudiments during spaceflight. J. Exp. Zool. 1994, 269, 212–222. [Google Scholar] [CrossRef]
  91. Lee, M.D.; Tuttle, R.; Girten, B. Effect of spaceflight on oxidative and antioxidant enzyme activity in rat diaphragm and intercostal muscles. J. Gravit. Physiol. J. Int. Soc. Gravit. Physiol. 1995, 2, 68–69. [Google Scholar]
  92. Suzuki, N.; Iwamura, Y.; Nakai, T.; Kato, K.; Otsuki, A.; Uruno, A.; Saigusa, D.; Taguchi, K.; Suzuki, M.; Shimizu, R.; et al. Gene expression changes related to bone mineralization, blood pressure and lipid metabolism in mouse kidneys after space travel. Kidney Int. 2022, 101, 92–105. [Google Scholar] [CrossRef] [PubMed]
  93. Morrison, D.R.; Lewis, M.L.; Barlow, G.H.; Todd, P.; Kunze, M.E.; Sarnoff, B.E.; Li, Z. Properties of electrophoretic fractions of human embryonic kidney cells separated on Space Shuttle flight STS-8. Adv. Space Res. 1984, 4, 77–79. [Google Scholar] [CrossRef] [PubMed]
  94. Stewart, R.M.; Todd, P.; Cole, K.D.; Morrison, D.R. Further analyses of human kidney cell populations separated on the space shuttle. Adv. Space Res. 1992, 12, 223–229. [Google Scholar] [CrossRef]
  95. Liakopoulos, V.; Leivaditis, K.; Eleftheriadis, T.; Dombros, N. The kidney in space. Int. Urol. Nephrol. 2012, 44, 1893–1901. [Google Scholar] [CrossRef]
  96. Whitson, P.A.; Pietrzyk, R.A.; Pak, C.Y. Renal stone risk assessment during space shuttle flights. J. Urol. 1997, 158, 2305–2310. [Google Scholar] [CrossRef] [Green Version]
  97. Whitson, P.A.; Pietrzyk, R.A.; Sams, C.F. Urine volume and its effects on renal stone risk in astronauts. Aviat. Space Environ. Med. 2001, 72, 368–372. [Google Scholar]
  98. Pietrzyk, R.A.; Jones, J.A.; Sams, C.F.; Whitson, P.A. Renal stone formation among astronauts. Aviat. Space Environ. Med. 2007, 78 (Suppl. 4), 9–13. [Google Scholar]
  99. Li, Z.; Song, Y.; Ma, Y.; Wei, H.; Liu, C.; Huang, J.; Wang, N.; Sha, J.; Sakurai, F. Influence of simulated microgravity on avian primordial germ cell migration and reproductive capacity. J. Exp. Zool. 2002, 292, 672–676. [Google Scholar] [CrossRef]
  100. Nowacki, D.; Klinger, F.G.; Mazur, G.; De Felici, M. Effect of Culture in Simulated Microgravity on the Development of Mouse Embryonic Testes. Adv. Clin. Exp. Med. 2015, 24, 769–774. [Google Scholar] [CrossRef] [Green Version]
  101. Ricci, G.; Catizone, A.; Esposito, R.; Galdieri, M. Microgravity effect on testicular functions. J. Gravit. Physiol. J. Int. Soc. Gravit. Physiol. 2004, 11, 61–62. [Google Scholar]
  102. Ricci, G.; Esposito, R.; Catizone, A.; Galdieri, M. Direct effects of microgravity on testicular function: Analysis of hystological, molecular and physiologic parameters. J. Endocrinol. Investig. 2008, 31, 229–237. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, S.; Zheng, D.; Wu, Y.; Lin, W.; Chen, Z.; Meng, L.; Liu, J.; Zhou, Y. Simulated Microgravity Using a Rotary Culture System Compromises the In Vitro Development of Mouse Preantral Follicles. PLoS ONE 2016, 11, e0151062. [Google Scholar] [CrossRef] [Green Version]
  104. Zhang, S.; Wu, Y.; Weng, Y.; Xu, Z.; Chen, W.; Zheng, D.; Lin, W.; Liu, J.; Zhou, Y. In Vitro Growth of Mouse Preantral Follicles Under Simulated Microgravity. J. Vis. Exp. 2017, 130, e55641. [Google Scholar]
  105. Duprat, A.M.; Husson, D.; Gualandris-Parisot, L. Does gravity influence the early stages of the development of the nervous system in an amphibian? Brain Res. Rev. 1998, 28, 19–24. [Google Scholar] [CrossRef] [PubMed]
  106. Ishikawa, C.; Li, H.; Ogura, R.; Yoshimura, Y.; Kudo, T.; Shirakawa, M.; Shiba, D.; Takahashi, S.; Morita, H.; Shiga, T. Effects of gravity changes on gene expression of BDNF and serotonin receptors in the mouse brain. PLoS ONE 2017, 12, e0177833. [Google Scholar] [CrossRef] [Green Version]
  107. Yoshikawa, M.; Ishikawa, C.; Li, H.; Kudo, T.; Shiba, D.; Shirakawa, M.; Muratani, M.; Takahashi, S.; Aizawa, S.; Shiga, T. Comparing effects of microgravity and amyotrophic lateral sclerosis in the mouse ventral lumbar spinal cord. Mol. Cell. Neurosci. 2022, 121, 103745. [Google Scholar] [CrossRef] [PubMed]
  108. Olenev, S.N.; Danilov, A.R.; Kryuchkova, T.A.; Sorokina, L.M.; Krasnov, I.B. Effect of weightlessness on some indices of brain development (results of residence of pregnant rats aboard the Cosmos-1514 biosatellite and investigation of subsequent development of their offspring on earth). Neurosci. Behav. Physiol. 1989, 19, 191–195. [Google Scholar] [CrossRef] [PubMed]
  109. Slenzka, K.; Appel, R.; Hilbig, R.; Kappel, T.; Vetter, S.; Freischütz, B.; Rahmann, H. Behavioural and biochemical investigations of the influence of altered gravity on the CNS of aquatic vertebrates during ontogeny. Adv. Space Res. 1994, 14, 309–312. [Google Scholar] [CrossRef]
  110. Bacci, S.; Bani, D. The Epidermis in Microgravity and Unloading Conditions and Their Effects on Wound Healing. Front. Bioeng. Biotechnol. 2022, 10, 666434. [Google Scholar] [CrossRef]
  111. Wubbels, R.J.; De Jong, H.A. The vestibulo-ocular reflex of hypergravity rats. J. Gravit. Physiol. J. Int. Soc. Gravit. Physiol. 2001, 8, 113–114. [Google Scholar]
  112. Mao, X.W.; Byrum, S.; Nishiyama, N.C.; Pecaut, M.J.; Sridharan, V.; Boerma, M.; Tackett, A.J.; Shiba, D.; Shirakawa, M.; Takahashi, S.; et al. Impact of Spaceflight and Artificial Gravity on the Mouse Retina: Biochemical and Proteomic Analysis. Int. J. Mol. Sci. 2018, 19, 2546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Shimada, N.; Moorman, S.J. Changes in gravitational force cause changes in gene expression in the lens of developing zebrafish. Dev. Dyn. 2006, 235, 2686–2694. [Google Scholar] [CrossRef]
  114. Miko, E.; Csaszar, A.; Bodis, J.; Kovacs, K. The Maternal-Fetal Gut Microbiota Axis: Physiological Changes, Dietary Influence, and Modulation Possibilities. Life 2022, 12, 424. [Google Scholar] [CrossRef]
  115. Ursell, L.K.; Metcalf, J.L.; Parfrey, L.W.; Knight, R. Defining the human microbiome. Nutr. Rev. 2012, 70 (Suppl. 1), S38–S44. [Google Scholar] [CrossRef] [Green Version]
  116. Stinson, L.F.; Boyce, M.C.; Payne, M.S.; Keelan, J.A. The Not-so-Sterile Womb: Evidence That the Human Fetus Is Exposed to Bacteria Prior to Birth. Front. Microbiol. 2019, 10, 1124. [Google Scholar] [CrossRef] [Green Version]
  117. Perez-Munoz, M.E.; Arrieta, M.C.; Ramer-Tait, A.E.; Walter, J. A critical assessment of the “sterile womb” and “in utero colonization” hypotheses: Implications for research on the pioneer infant microbiome. Microbiome 2017, 5, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Collado, M.C.; Rautava, S.; Aakko, J.; Isolauri, E.; Salminen, S. Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Sci. Rep. 2016, 6, 23129. [Google Scholar] [CrossRef] [Green Version]
  119. Neu, J. The microbiome during pregnancy and early postnatal life. Semin. Fetal Neonatal Med. 2016, 21, 373–379. [Google Scholar] [CrossRef] [PubMed]
  120. DiGiulio, D.B. Diversity of microbes in amniotic fluid. Semin. Fetal Neonatal Med. 2012, 17, 2–11. [Google Scholar] [CrossRef] [PubMed]
  121. Mishra, A.; Lai, G.C.; Yao, L.J.; Aung, T.T.; Shental, N.; Rotter-Maskowitz, A.; Shepherdson, E.; Singh, G.S.N.; Pai, R.; Shant, A.; et al. Microbial exposure during early human development primes fetal immune cells. Cell 2021, 184, 3394–3409. [Google Scholar] [CrossRef] [PubMed]
  122. Jašarević, E.; Bale, T.L. Prenatal and postnatal contributions of the maternal microbiome on offspring programming. Front. Neuroendocr. 2019, 55, 100797. [Google Scholar] [CrossRef]
  123. Stinson, L.F.; Payne, M.S.; Keelan, J.A. Planting the seed: Origins, composition, and postnatal health significance of the fetal gastrointestinal microbiota. Crit. Rev. Microbiol. 2017, 43, 352–369. [Google Scholar] [CrossRef] [PubMed]
  124. McDonald, B.; McCoy, K.D. Maternal microbiota in pregnancy and early life. Science 2019, 365, 984–985. [Google Scholar] [CrossRef]
  125. van den Elsen, L.W.J.; Garssen, J.; Burcelin, R.; Verhasselt, V. Shaping the Gut Microbiota by Breastfeeding: The Gateway to Allergy Prevention? Front. Pediatr. 2019, 7, 47. [Google Scholar] [CrossRef] [PubMed]
  126. Kuehnast, T.; Abbott, C.; Pausan, M.R.; Pearce, D.A.; Moissl-Eichinger, C.; Mahnert, A. The crewed journey to Mars and its implications for the human microbiome. Microbiome 2022, 10, 26. [Google Scholar] [CrossRef]
  127. Douglas, G.L.; Zwart, S.R.; Smith, S.M. Space Food for Thought: Challenges and Considerations for Food and Nutrition on Exploration Missions. J. Nutr. 2020, 150, 2242–2244. [Google Scholar] [CrossRef]
  128. Lackner, J.R.; Dizio, P. Space motion sickness. Exp. Brain Res. 2006, 175, 377–399. [Google Scholar] [CrossRef]
  129. Shreiner, A.B.; Kao, J.Y.; Young, V.B. The gut microbiome in health and in disease. Curr. Opin. Gastroenterol. 2015, 31, 69–75. [Google Scholar] [CrossRef]
  130. Ziętek, M.; Celewicz, Z.; Szczuko, M. Short-Chain Fatty Acids, Maternal Microbiota and Metabolism in Pregnancy. Nutrients 2021, 13, 1244. [Google Scholar] [CrossRef]
  131. Conlon, M.A.; Bird, A.R. The impact of diet and lifestyle on gut microbiota and human health. Nutrients 2014, 7, 17–44. [Google Scholar] [CrossRef] [PubMed]
  132. Liu, Z.; Luo, G.; Du, R.; Sun, W.; Li, J.; Lan, H.; Chen, P.; Yuan, X.; Cao, D.; Li, Y.; et al. Effects of spaceflight on the composition and function of the human gut microbiota. Gut Microbes 2020, 11, 807–819. [Google Scholar] [CrossRef] [PubMed]
  133. Ritchie, L.E.; Taddeo, S.S.; Weeks, B.R.; Lima, F.; Bloomfield, S.A.; Azcarate-Peril, M.A.; Zwart, S.R.; Smith, S.M.; Turner, N.D. Space Environmental Factor Impacts upon Murine Colon Microbiota and Mucosal Homeostasis. PLoS ONE 2015, 10, e0125792. [Google Scholar] [CrossRef]
  134. Turroni, S.; Rampelli, S.; Biagi, E.; Consolandi, C.; Severgnini, M.; Peano, C.; Quercia, S.; Soverini, M.; Carbonero, F.G.; Bianconi, G.; et al. Temporal dynamics of the gut microbiota in people sharing a confined environment, a 520-day ground-based space simulation, MARS500. Microbiome 2017, 5, 39. [Google Scholar] [CrossRef] [Green Version]
  135. Alauzet, C.; Cunat, L.; Wack, M.; Lozniewski, A.; Busby, H.; Agrinier, N.; Cailliez-Grimal, C.; Frippiat, J.-P. Hypergravity disrupts murine intestinal microbiota. Sci. Rep. 2019, 9, 9410. [Google Scholar] [CrossRef] [Green Version]
  136. Voorhies, A.A.; Ott, C.M.; Mehta, S.; Pierson, D.L.; Crucian, B.E.; Feiveson, A.; Oubre, C.M.; Torralba, M.; Moncera, K.; Zhang, Y.; et al. Study of the impact of long-duration space missions at the International Space Station on the astronaut microbiome. Sci. Rep. 2019, 9, 9911. [Google Scholar] [CrossRef] [Green Version]
  137. Avila-Herrera, A.; Thissen, J.; Urbaniak, C.; Be, N.A.; Smith, D.J.; Karouia, F.; Mehta, S.; Venkateswaran, K.; Jaing, C. Crewmember microbiome may influence microbial composition of ISS habitable surfaces. PLoS ONE 2020, 15, e0231838. [Google Scholar] [CrossRef] [PubMed]
  138. Morrison, M.D.; Thissen, J.B.; Karouia, F.; Mehta, S.; Urbaniak, C.; Venkateswaran, K.; Smith, D.J.; Jaing, C. Investigation of Spaceflight Induced Changes to Astronaut Microbiomes. Front. Microbiol. 2021, 12, 659179. [Google Scholar] [CrossRef] [PubMed]
  139. Urbaniak, C.; Lorenzi, H.; Thissen, J.; Jaing, C.; Crucian, B.; Sams, C.; Pierson, D.; Venkateswaran, K.; Mehta, S. The influence of spaceflight on the astronaut salivary microbiome and the search for a microbiome biomarker for viral reactivation. Microbiome 2020, 8, 56. [Google Scholar] [CrossRef] [PubMed]
  140. Strollo, F.; Strollo, G.; More, M.; Bollanti, L.; Ciarmatori, A.; Longo, E.; Quintiliani, R.; Mambro, A.; Mangrossa, N.; Ferretti, C. Hormonal adaptation to real and simulated microgravity. J. Gravit. Physiol. J. Int. Soc. Gravit. Physiol. 1998, 5, 89–92. [Google Scholar]
  141. Strollo, F. Chapter 4 Hormonal Changes in Humans During Spaceflight. Adv. Space Biol. Med. 1999, 7, 99–129. [Google Scholar] [CrossRef]
  142. Wakayama, S.; Kamada, Y.; Yamanaka, K.; Kohda, T.; Suzuki, H.; Shimazu, T.; Tada, M.N.; Osada, I.; Nagamatsu, A.; Kamimura, S.; et al. Healthy offspring from freeze-dried mouse spermatozoa held on the International Space Station for 9 months. Proc. Natl. Acad. Sci. USA 2017, 114, 5988–5993. [Google Scholar] [CrossRef] [Green Version]
  143. Rouchka, E.C.; Flight, R.M.; Fasciotto, B.H.; Estrada, R.; Eaton, J.W.; Patibandla, P.K.; Waigel, S.J.; Li, D.; Kirtley, J.K.; Sethu, P.; et al. Transcriptional profile of immediate response to ionizing radiation exposure. Genom. Data 2016, 7, 82–85. [Google Scholar] [CrossRef] [Green Version]
  144. Dubrova, Y.E.; Plumb, M.; Gutierrez, B.; Boulton, E.; Jeffreys, A.J. Transgenerational mutation by radiation. Nature 2000, 405, 37. [Google Scholar] [CrossRef]
  145. Ronca, A.E.; Baker, E.S.; Bavendam, T.G.; Beck, K.D.; Miller, V.M.; Tash, J.S.; Jenkins, M. Effects of sex and gender on adaptations to space: Reproductive health. J. Women’s Health 2014, 23, 967–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Steller, J.G.; Alberts, J.R.; Ronca, A.E. Oxidative Stress as Cause, Consequence, or Biomarker of Altered Female Reproduction and Development in the Space Environment. Int. J. Mol. Sci. 2018, 19, 3729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Jennings, R.T.; Baker, E.S. Gynecological and Reproductive Issues for Women in Space: A Review. Obstet. Gynecol. Surv. 2000, 55, 109–116. [Google Scholar] [CrossRef] [Green Version]
  148. Jain, V.; Wotring, V.E. Medically induced amenorrhea in female astronauts. NPJ Microgravity 2016, 2, 16008. [Google Scholar] [CrossRef] [Green Version]
  149. Strollo, F.; Riondino, G.; Harris, B.; Strollo, G.; Casarosa, E.; Mangrossa, N.; Ferretti, C.; Luisi, M. The effect of microgravity on testicular androgen secretion. Aviat. Space Environ. Med. 1998, 69, 133–136. [Google Scholar] [PubMed]
  150. Smith, S.M.; Heer, M.; Wang, Z.; Huntoon, C.L.; Zwart, S.R. Long-Duration Space Flight and Bed Rest Effects on Testosterone and Other Steroids. J. Clin. Endocrinol. Metab. 2012, 97, 270–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Masini, M.A.; Albi, E.; Barmo, C.; Bonfiglio, T.; Bruni, L.; Canesi, L.; Cataldi, S.; Curcio, F.; D’Amora, M.; Ferri, I.; et al. The Impact of Long-Term Exposure to Space Environment on Adult Mammalian Organisms: A Study on Mouse Thyroid and Testis. PLoS ONE 2012, 7, e35418. [Google Scholar] [CrossRef] [Green Version]
  152. Souza, K.A.; Black, S.D.; Wassersug, R.J. Amphibian development in the virtual absence of gravity. Proc. Natl. Acad. Sci. USA 1995, 92, 1975–1978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Gulimova, V.; Proshchina, A.; Kharlamova, A.; Krivova, Y.; Barabanov, V.; Berdiev, R.; Asadchikov, V.; Buzmakov, A.; Zolotov, D.; Saveliev, S. Reptiles in Space Missions: Results and Perspectives. Int. J. Mol. Sci. 2019, 20, 3019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Ronca, A.E. Studies toward birth and early mammalian development in space. Adv. Space Res. 2003, 32, 1483–1490. [Google Scholar] [CrossRef] [PubMed]
  155. Wong, A.M.; De Santis, M. Rat gestation during space flight: Outcomes for dams and their offspring born after return to earth. Integr. Psychol. Behav. Sci. 1997, 32, 322–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Roberts, D.R.; Albrecht, M.H.; Collins, H.R.; Asemani, D.; Chatterjee, A.R.; Spampinato, M.V.; Zhu, X.; Chimowitz, M.I.; Antonucci, M.U. Effects of Spaceflight on Astronaut Brain Structure as Indicated on MRI. N. Engl. J. Med. 2017, 377, 1746–1753. [Google Scholar] [CrossRef]
  157. Afshinnekoo, E.; Scott, R.T.; MacKay, M.J.; Pariset, E.; Cekanaviciute, E.; Barker, R.; Gilroy, S.; Hassane, D.; Smith, S.M.; Zwart, S.R.; et al. Fundamental Biological Features of Spaceflight: Advancing the Field to Enable Deep-Space Exploration. Cell 2021, 184, 6002. [Google Scholar] [CrossRef]
  158. Reynolds, R.J.; Scott, R.T.; Turner, R.T.; Iwaniec, U.T.; Bouxsein, M.L.; Sanders, L.M.; Antonsen, E.L. Validating Causal Diagrams of Human Health Risks for Spaceflight: An Example Using Bone Data from Rodents. Biomedicines 2022, 10, 2187. [Google Scholar] [CrossRef]
  159. Yeung, C.K.; Koenig, P.; Countryman, S.; Thummel, K.E.; Himmelfarb, J.; Kelly, E.J. Tissue Chips in Space—Challenges and Opportunities. Clin. Transl. Sci. 2020, 13, 8–10. [Google Scholar] [CrossRef]
  160. Cortés-Sánchez, J.L.; Callant, J.; Krüger, M.; Sahana, J.; Kraus, A.; Baselet, B.; Infanger, M.; Baatout, S.; Grimm, D. Cancer Studies under Space Conditions: Finding Answers Abroad. Biomedicines 2021, 10, 25. [Google Scholar] [CrossRef]
Figure 1. Otolith organs of the vestibular system. The vestibular system is responsible for the body’s equilibrium. It maintains balance and provides awareness of the body’s spatial orientation. The sensory part is located in the inner ear. The two otolithic organs are two patches of hair cells, oriented nearly perpendicular to each other. The saccule rests vertically, whereas the utricle hangs horizontally. The stereocilia of these cells are embedded in a gel-like layer, sprinkled with calcium carbonate crystals called otoconia. The crystals add weight to the layer, pulling it down with gravity. These sensory organs detect not the motion itself, but changes in the rate of motion, specifically acceleration or deceleration (This figure was generated within biorender.com).
Figure 1. Otolith organs of the vestibular system. The vestibular system is responsible for the body’s equilibrium. It maintains balance and provides awareness of the body’s spatial orientation. The sensory part is located in the inner ear. The two otolithic organs are two patches of hair cells, oriented nearly perpendicular to each other. The saccule rests vertically, whereas the utricle hangs horizontally. The stereocilia of these cells are embedded in a gel-like layer, sprinkled with calcium carbonate crystals called otoconia. The crystals add weight to the layer, pulling it down with gravity. These sensory organs detect not the motion itself, but changes in the rate of motion, specifically acceleration or deceleration (This figure was generated within biorender.com).
Life 13 00588 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Theotokis, P.; Manthou, M.E.; Deftereou, T.-E.; Miliaras, D.; Meditskou, S. Addressing Spaceflight Biology through the Lens of a Histologist–Embryologist. Life 2023, 13, 588. https://doi.org/10.3390/life13020588

AMA Style

Theotokis P, Manthou ME, Deftereou T-E, Miliaras D, Meditskou S. Addressing Spaceflight Biology through the Lens of a Histologist–Embryologist. Life. 2023; 13(2):588. https://doi.org/10.3390/life13020588

Chicago/Turabian Style

Theotokis, Paschalis, Maria Eleni Manthou, Theodora-Eleftheria Deftereou, Dimosthenis Miliaras, and Soultana Meditskou. 2023. "Addressing Spaceflight Biology through the Lens of a Histologist–Embryologist" Life 13, no. 2: 588. https://doi.org/10.3390/life13020588

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop