1. Introduction
Understanding the effects of space travel on human health is essential, as space missions pose unique risks and challenges. This knowledge is critical not only for safeguarding astronauts but also for its broader implications, offering insights into conditions such as aging, osteoporosis, and cardiovascular diseases. The environmental stressors encountered in space—particularly radiation and microgravity—can trigger various health issues, potentially intensifying as the mission duration increases. Addressing these factors is crucial to enabling safer long-term space exploration and advancing our understanding of human physiology.
NASA identifies five main risks in spaceflight: microgravity, radiation, isolation and confinement, hostile environments, and distance from Earth [
1,
2]. Among these, microgravity presents substantial physiological challenges, impacting the body across three primary phases: initial adaptation, prolonged exposure, and readaptation to Earth’s gravity [
3]. Key physiological changes include fluid redistribution, plasma volume reduction, and decreased hemoglobin levels, collectively leading to a condition termed space anemia [
4]. Microgravity also adversely affects cardiovascular health, bone density, and muscle mass, with fluid shifts contributing to sensory changes, such as altered olfaction [
5]. Additionally, structural changes in the brain’s gray matter have been observed, underscoring the extensive neurological impact of microgravity [
6,
7].
Space radiation, though not the primary focus, is another critical concern, damaging DNA and increasing the risk of carcinogenesis [
8]. Astronauts face chronic radiation exposure from galactic cosmic rays (GCRs), which differ significantly from Earth’s radiation, causing oxidative damage and genetic mutations [
9,
10,
11]. NASA has increased the career radiation dose limit to 600 mSV, a threshold that missions to Mars are projected to exceed substantially [
9,
12].
The isolation and confinement of space, combined with the lack of a circadian rhythm, present risks to mental health and sleep, potentially affecting mission performance [
1]. Environmental conditions during spaceflight must be closely controlled to mimic Earth, but challenges such as communication delays and limited resources heighten stress and suppress immune function, further complicating long-term space missions [
1,
3,
5,
13,
14].
Space travel presents unique health challenges, making personalized strategies essential, especially as astronaut diversity grows. The NASA Twin Study demonstrated long-lasting physiological changes, emphasizing the need for tailored approaches [
13]. Bioinformatics and omics technologies—genomics, proteomics, and metabolomics—enable the development of unique health profiles and personalized medical plans for astronauts, which are critical due to limited crew numbers and resource constraints [
15]. Genomics provides insight into disease predispositions but lacks adaptability to changing conditions. Transcriptomics, in contrast, captures real-time gene expression changes in response to space-related stressors, aiding in the development of personalized space travel approaches. This research focuses on microarray technology, which, though limited to known genes, is cost-effective and well suited to analyzing gene expression changes due to spaceflight [
16]. Identifying differentially expressed genes between ground-based controls and space samples can uncover the molecular mechanisms underlying astronauts’ adaptation to space, which is critical for long-term space missions and personalized medicine advancements.
Understanding these mechanisms is key to developing effective countermeasures. Space-related factors disrupt the immune system, increasing susceptibility to infections. While current strategies are generalized, precision medicine offers a means of delivering individualized immune countermeasures by targeting the specific molecular changes induced by space conditions [
17].
Numerous studies have analyzed astronauts’ transcriptomic data to identify differentially expressed genes (DEGs) between pre-flight and during-flight states, focusing on changes in gene expression and protein function due to space conditions. Sakharkar and Yang [
18] examined gene expression changes in astronauts’ hair follicles across different stages of spaceflight using microarray data, identifying the over-expressed genes
LIMCH1 and
IER3, which are associated with cancer growth and progression, and the under-expressed genes
NFATC1, linked to osteoporosis, and
KIDINS220, which potentially leads to neurological and cardiovascular issues. Hwang et al. [
19] analyzed cardiac progenitor spheres exposed to long-term microgravity using RNA-seq data, and found the over-expression of gene
CCBE1, which is crucial for heart development, and the under-expression of gene
CD24, which is a marker for pluripotent stem cells. This research expands on these findings by comparing DEGs from various datasets under microgravity conditions to identify novel molecular targets for therapeutic intervention.
Given the limited opportunities for direct human space travel, collecting comprehensive and continuous astronaut data presents a significant challenge. Thus, human cell lines are utilized to compare molecular responses to existing knowledge, providing a more robust analysis of how microgravity affects cellular and molecular pathways. The primary objective of this research is to analyze transcriptomics data from human cell lines exposed to microgravity to identify microgravity’s effects on cellular processes. Specifically, this study focuses on DLD-1 (solid tumor cells), MOLT-4 (hematological tumor cells), Jurkat T lymphocytic, and myelomonocytic U937 cells. DLD-1 and MOLT-4 were exposed to simulated microgravity, while Jurkat T cells and U937 cells were subjected to real microgravity during sounding rocket and parabolic flights.
This research investigates the effects of microgravity on gene expression and explores potential therapeutic interventions for damage caused by spaceflight. The study focuses on identifying which genes are differentially expressed in response to microgravity, determining which biological pathways are significantly affected and assessing whether drugs can be identified to reverse these gene expression changes.
Using a bioinformatics workflow, the research began by gathering data from NASA’s GeneLab repository, specifically targeting datasets with microgravity conditions. Differential expression analysis identified significantly altered genes, and Venn diagram analysis uncovered common DEGs across the datasets. Gene enrichment analysis provided insights into the biological significance of these genes, while drug repurposing analysis proposed potential therapeutic candidates. Finally, gene target enrichment and pathway comparisons highlighted shared biological processes between DEGs and drug gene targets, bridging spaceflight-induced gene expression changes with possible pharmacological interventions.
3. Discussion
This study investigated the impact of microgravity on gene expression in human cell lines, revealing extensive changes in gene expression and biological pathways across five comparisons from various transcriptomic datasets. The cells analyzed varied in type, exposure duration, and the microgravity conditions they experienced, a diversity intentionally chosen to capture a broader spectrum of cellular responses and enhance the generalizability of our findings.
As previously mentioned in the results, simulated microgravity induced a balanced response in the DLD-1 and MOLT-4 cells, while real microgravity exposure revealed the increasing activation of gene expression over time in Jurkat cells and significant downregulation in U937 cells treated with SKF96365. Similar responses have been reported in previous studies, such as the work by Vidyasekar et al. [
25], where microgravity was shown to influence cancer cell behavior through alterations in gene expression profiles. In contrast, Jurkat T lymphocytic cells exposed to real microgravity demonstrated a substantial number of differentially expressed genes (DEGs) that increased with longer exposure times. This increase in gene expression changes over time suggests the stronger activation of transcriptional processes under prolonged microgravity. Similar findings were reported in Cora S. Thiel et al.’s study [
26], where T cells exhibited rapid transcriptional changes under altered gravity conditions, highlighting a time-dependent transcriptional response induced by microgravity.
This study identified 11 DEGs that were consistently affected across the conditions tested. Among the under-expressed genes,
DNPH1 plays a critical role in nucleotide metabolic processes required for DNA replication, repair, RNA synthesis, and cellular energy homeostasis, all of which are essential for cell division and the balance between cell growth and death, with implications for cancer aggressiveness and metastasis [
25]. Similarly,
EXOSC5, which is involved in RNA degradation and processing, is essential for RNA quality control and immune responses. Microgravity exposure has been shown to disrupt RNA polymerase II binding to DNA, affecting gene transcription and potentially contributing to immune dysfunction [
27]. Furthermore,
L3MBTL2, involved in chromatin modification and DNA damage repair, was also downregulated in our study, which is consistent with literature showing increased sensitivity to ionizing radiation in its absence [
28,
29]. Moreover, the downregulation of
LGALS3BP, which plays a role in the immune response and cell adhesion, aligns with existing studies demonstrating that spaceflight weakens immunity [
8,
17,
30,
31]. It has been also reported that the loss of cell adhesion in various tissues results in adverse effects on cardiovascular health, immune function, bone density, and muscle strength, while potentially enhancing cancer cell apoptosis and contributing to the progression of certain forms of metastasis [
32].
The upregulation of
CSNK2A2 and
HIPK1 suggests increased apoptotic signaling, p53 regulation, and protein phosphorylation. Microgravity accelerates T cell apoptosis, weakening immune function and increasing infection risks for astronauts [
33]. The heightened oxidative stress in microgravity worsens cellular damage, highlighting the need for immune protection during space missions [
34].
HIPK1 also plays a role in the cellular stress response, reducing TNF-α and inflammation, suggesting protection against inflammation and metabolic disturbances [
35]. Upregulation of
CSGALNACT2 has been linked to reduced inflammation, possibly related to microgravity-induced immune suppression [
36].
PHF21A, involved in histone deacetylation, affects gene expression, cellular stress responses, and can contribute to neuroinflammation, impaired memory, and tumor formation [
37,
38,
39,
40].
RAP1A, involved in signal transduction and MAPK pathways, is linked to oncogenesis, and its activation increases under microgravity [
41,
42].
MBNL2 regulates RNA splicing and shows variable responses to microgravity exposure. This variability suggests that prolonged microgravity exacerbates gene dysregulation, though genes like
EXOSC5,
L3MBTL2, and
MBNL2 showed no expected increase in dysregulation. This may indicate that RNA processing and gene regulation are better adapted to microgravity [
13,
43,
44]. Cora S. Thiel hypothesized that gravitational forces influence chromatin structure, maintaining gene expression homeostasis [
44]. A summary of the functions and relevance to space biology of the 11 common DEGs is presented in
Table 8.
As noted earlier, there is limited literature on the DEGs identified in this study in relation to microgravity. One exception is
DNPH1, which is under-expressed in Jurkat T lymphocytes and linked to cellular proliferation and transformation as a putative oncogene involved in c-Myc-mediated transformation [
45]. Many microgravity-affected DEGs are related to cancer. For instance,
PHF21A, over-expressed in our study, is implicated in colorectal cancer, enhancing lymphangiogenesis and immune cell infiltration associated with metastasis [
46].
PHF21A disruption affects proliferation and lineage commitment, leading to disrupted cell renewal [
47].
RAP1A, also over-expressed, is linked to esophageal squamous cell carcinoma, enhancing metastasis through increased cell migration and invasion [
48].
EXOSC5, recognized as a tumor suppressor, has been downregulated in various cancers, including renal cell carcinoma, and plays a critical role in signaling pathways like JAK-STAT, MAPK, and WNT, which are crucial for cancer progression [
49].
LGALS3BP and
CSNK2A2 are associated with the NF-κB pathway, which is crucial in tumor progression.
LGALS3BP is suspected to be a tumor suppressor due to its under-expression in prostate cancer, while
CSNK2A2 over-expression promotes tumor progression, particularly in hepatocellular carcinoma [
50].
MBNL2, typically tumor-suppressive, promotes cancer when depleted but, in some contexts or stages of cancer, contributes to cancer development [
51]; it plays a key role in cardiac fibrosis, especially in the aging heart, promoting fibroblast senescence and fibrosis [
52]. Lastly, the role of under-expressed
SPRYD4 is less defined, though its over-expression in cholangiocarcinoma leads to cell cycle arrest and apoptosis, while knockdown of this gene causes the opposite effect, suggesting a role in cell cycle regulation and apoptosis [
53]. This complex relationship between microgravity-induced gene expression changes and cancer pathways highlights potential implications for astronaut health. Understanding these pathways could help develop countermeasures for long-term space missions.
In this study, in silico drug repurposing identified daunorubicin, vorinostat, and fludarabine as potential modulators for DEGs. Daunorubicin, a chemotherapeutic, intercalates DNA and inhibits topoisomerase II, disrupting DNA replication and transcription [
54]. However, its use in space presents significant challenges. While initial studies suggested daunorubicin as a candidate for mitigating muscle atrophy in space [
55], further research highlighted its potential risks. Microgravity has been shown to alter the drug’s efficacy, leading to increased migration of cancer cells; as well as being a potent chemotherapeutic agent, it possesses significant side effects, including cardiotoxicity, myelosuppression, hair loss, nausea, and vomiting [
56]. This paradoxical effect underscores the need for comprehensive space-based drug testing and potential adjustments in drug dosing regimens to ensure its safety and effectiveness for astronauts. Vorinostat, an HDAC inhibitor, modifies chromatin and gene expression [
57]. Its mechanism involves histone acetylation changes, impacting cancer-related pathways such as those regulated by
EXOSC5 [
58]. Moreover, vorinostat’s dual role as a radioprotector and radiosensitizer [
39] suggests an application for managing radiation and cancer in space, particularly with microgravity-induced epigenetic changes [
59]. Fludarabine has been shown to have synergistic effects when combined with other cancer therapeutics, including HDAC inhibitors [
39]. This synergy could be particularly valuable in the context of space travel, where the disruption of cellular pathways by microgravity mirrors some aspects of cancer pathology [
60]. The potential for fludarabine to enhance the efficacy of other treatments, such as vorinostat, makes it a candidate worth further exploration for spaceflight applications.
These drugs, though designed for cancer, may overlap with pathways altered by spaceflight, like immune function and cellular repair. Their interaction with space-induced cellular changes highlights the need for further in vitro and in vivo testing under microgravity to ensure their safety and efficacy for astronauts.
This study’s limitations include reliance on specific cell lines, short-duration microgravity exposure, and simulated conditions that may not fully replicate long-term spaceflight effects. Differences between simulated and real microgravity could cause discrepancies in gene expression results. Gene expression is context-dependent, influenced by factors like cell type and exposure duration. Dysregulated gene effects were not validated experimentally and, while in silico analysis suggested potential drugs to combat these effects, their safety in space, particularly with radiation exposure and altered immune function, remains uncertain.