Abstract
The extreme space environment accelerates aging and compromises human health. NASA has named five main hazards in space, including gravity changes. However, the contribution of each factor to the overall impact on biomolecular and cellular processes is not always clear. We aimed to explore the effects of microgravity on the transcriptomes of healthy volunteers, with a focus on gene expression in p53 pathways. Ten healthy men were exposed to dry immersion simulated microgravity (DI-SMG) for three weeks and blood samples were collected at five timepoints before, during and after the course of DI-SMG. T cells were purified from the peripheral blood samples and total RNA was isolated and sequenced followed by a bioinformatics analysis of the volunteers’ global transcriptomes. A differential expression of p53 network genes was observed. The expression of 30 genes involved in the p53 gene network was affected during a 3-week course of DI-SMG including classic p53 downstream target genes involved in cellular senescence: GADD45, p21, PUMA, IGF1 and other target genes. For the first time, the p53-associated cell signaling pathways and gene networks in human T cells were reported to be affected in vivo by DI-SMG. It is evident that the relatively mild effects of simulated weightlessness on the human body are sufficient to activate these pathways. Identified transcriptomic changes point toward a potential molecular overlap with aging and cellular senescence. These findings could contribute to a broader research landscape that may lead to the discovery of a new class of drugs—MG-senolytics.
Keywords:
aging; microgravity; dry immersion; gene expression; gene targets; p53 network; transcriptome 1. Introduction
The physiological changes caused by spaceflight are similar to those that occur with aging [,]. NASA’s Human Research Program has identified five hazards that astronauts will face in space. These include space radiation, isolation and confinement, distance from Earth, gravity (and the lack of it), and closed or hostile environments []. The use of a DI-SMG system is a well-established, ground-based approach to the modelling of orbital space microgravity in vivo. It provides investigators with unique data on the physiological effects induced by DI-SMG []. The knowledge gained from those investigations is important for developing countermeasures against undesirable effects caused by the space environment and aging []. The well-studied physiological impacts of microgravity on human organ systems still lack precise and thorough complements at the level of cellular effects and biomolecular response mechanisms to weightlessness.
The most commonly mutated gene in cancer [], the TP53 tumor suppressor gene, which encodes transcription factor p53, has been called the “guardian of the genome” by one of its discoverers, Professor Sir David Lane [], for its cell protective function under conditions of DNA damage stress. The p53 gene network compiles multifunctional biomolecular players, including target genes downstream of p53 [,,,,,,,]. The p53 gene network responds to a variety of environmental and internal signals and comprises several crucial pathways including DNA repair, the regulation of cell cycle and the induction of cell cycle arrest, apoptosis and cellular senescence. These pathways are vital for maintaining genome integrity and cellular homeostasis. p53 and several members of the p53 gene network are considered to be involved in aging processes or mediating longevity [,,].
A four-fold increase was reported in the content of p53 in the skin cells of rats in space on the STS-58 mission (Columbia) []. Furthermore, microgravity effects on the p53 pathway in vitro were registered in mouse sperm cells [], cultured human lymphoblastoid cell lines [,], in macrophages [] and in human soleus muscle after 3-day dry immersion []. In human lymphocytes from two healthy donors, exposure to the spaceflight environment aboard the ISS for 48 h resulted in an increase in TP53 mRNA levels, an effect attributed to the combinatory action of space radiation and microgravity [].
Objective
In a DI-SMG study conducted on ten healthy volunteers, we aimed to investigate whether simulated microgravity affects the expression of aging-related genes in the p53 gene network in human peripheral blood T cells in vivo.
2. Results and Discussion
2.1. RNA-Seq Data Output Summary
The DI-SMG study was conducted and global transcriptome profiling via RNA-seq was performed on T cells as described below (see Section 3. Materials and Methods). RNA-seq analysis yielded expression data for 58,676 transcripts, including 17,760 known genes and 38,430 transcripts associated with uncharacterized gene products. This dataset has been deposited in the NCBI Gene Expression Omnibus (GEO) repository under accession number GSE301964. Subsequent analysis focused specifically on interrogating the p53 gene network.
2.2. TP53 Tumor Suppressor Gene Expression
The transcript level of the TP53 tumor suppressor gene varied among all 10 volunteers in the study. At different timepoints of the experiment, TP53 was up-regulated in 9 of 10 volunteers compared to background levels (timepoint “−7 day”, Figure 1), and the dynamics of its expression varied between individuals. Seven volunteers (## 1–5, 7, and 8) demonstrated an increase in TP53 transcript levels at 7 days in SMG and then showed variable expression with an overall upward trend. Volunteer #6 showed a slight decrease in TP53 transcript levels during the SMG course and then an up-regulation above the background (BG) level at the 28-day timepoint after the SMG course. Volunteer #9 showed an overall down-regulation of the TP53 transcript level with a decline in its expression at 7 days and 14 days in SMG, and then its level was partially recovered at the 21-day timepoint. Volunteer #10 also showed a decrease in TP53 expression at 7 days and 14 days in SMG, but it was increased on day 21 of SMG, and the expression level increased further after the SMG course on day 28 (Figure 2).
Figure 1.
The setup of the simulated microgravity study in the dry immersion system. (A). The dry immersion facility at the Institute of Biomedical Problems (B). The time course and flow chart of the DI-SMG study. Time course timepoints: before (−7 day or BG, background), during (7 day; 14 day; 21 day) and after (28 day) 3 weeks of DI-SMG.
Figure 2.
TP53 gene differential expression in the DI-SMG study. A general trend towards an increased expression of the tumor suppressor gene TP53 was observed in T cell samples from 9 of 10 volunteers. Time course timepoints: before (−7 day or BG, background), during (7 day; 14 day; 21 day) and after (28 day) 3 weeks of DI-SMG. The parameters shown are (A) non-normalized expression values, Log2 fold change; (B) normalized fold change. # is the number of volunteers.
The statistical analysis reveals a statistically significant upward trend in TP53 gene expression over the course of the study (Figure S1). This observation is supported by a mean slope coefficient of 1.22 ± 0.88 with a 95% confidence interval that does not include zero
2.3. p53 Gene Network
The expression profile of the p53 gene network was significantly altered in response to the 3-week DI-SMG intervention. The expression profile of the complete gene set (n = 75) from the KEGG p53 signaling pathway (hsa04115) was evaluated. Notably, a subset of 30 genes (Table 1) showed differential expression in all, or nearly all, of the ten volunteers. This gene set included central apoptosis and senescence regulators such as GADD45, p21, PUMA, NOXA, and p19ARF, highlighting the pathway’s central role in the physiological response to simulated microgravity.
In relation to the regulation of the cell cycle, the gene expression of three B cyclins (CCNB1, CCNB2, and CCNB3) and two cyclin-dependent kinase inhibitors (CDKN1A and CDKN1B) in p53 networks exhibited dynamic changes under DI-SMG conditions in all ten subjects.
Growth arrest and DNA damage-inducible alpha, beta and gamma (GADD45A, GADD45B, and GADD45G) playing roles in cell cycle arrest, DNA repair, apoptosis, innate immunity, genomic stability, and senescence showed coordinated differential expression during the DI SMG study in all ten volunteers (Figure 3).
Figure 3.
A heatmap of p53 gene network expression in the DI SMG study. The expression of the genes involved in the p53 gene network in ten volunteers’ T cells at five timepoints: before (BG, background or −7 day), during (7 day; 14 day; 21 day) and after (28 day) 3 weeks of DI-SMG. The log2 normalized count matrix RNA-seq read values are shown.
KEGG pathway enrichment analysis indicated that DI-SMG exposure significantly affected the p53 signaling pathway hsa04115 (Supplementary File S1).
2.4. Aging-Associated Genes in p53 Gene Network and Interacting Pathways
The differential expression of p53 network genes linked to aging and cellular senescence was recorded in the SMG study. The expression of dominant active p53 leads to the constitutive expression of downstream target genes and results in premature aging []. There were varying changes in the TP53 transcript level observed during the experiment as described above (Section 2.2, Figure 2).
The cell cycle cyclin-dependent kinase inhibitor 1A (CDKN1A) interacts with cyclin-dependent kinases CDK2 and CDK4 and blocks their progression through the cell cycle. BH domain Bcl-2-associated protein X (Bax) and p53 up-regulated modulator of apoptosis (PUMA) are potent inducers of apoptosis. Cellular senescence and apoptosis prevent tumorigenesis. Also, both pathways have the potential to deplete stem and progenitor cell pools that leads to impaired tissue renewal, ultimately impairing organ homeostasis—a hallmark of aging []. Genes encoding cyclin-dependent kinase inhibitors CDKN1A and CDKN1B, Bax, PUMA, and cyclin-dependent kinases CDK4 and CDK6 demonstrated differential expression during the DI SMG experiment in all ten volunteers. Sestrins are conserved stress-inducible anti-aging genes []. The expression of genes encoding sestrins was changed during DI SMG in all or almost all ten volunteers (Figure 3, Table 1).
In addition, p53 affects the insulin-like growth factor 1 (IGF1) signaling pathway, which is a key determinant of aging and longevity []. Reduced IGF1 signaling is associated with extended lifespan in the highly conserved pathway among different organisms from nematodes to mammals []. IGFBP-3 plays an important role in senescence as an aging marker. It binds to IGF1, which regulates growth, survival, and aging. IGFBP-3 up-regulates the PI3K/Akt/mTOR signaling pathway during cell aging []. Also, p53 was elevated in IGFBP3 gene KO cells when compared to normal cells []. The expression of both genes: IGF1 and IGFBP3 was changed during the DI SMG study in 5 out of 10 and in all 10 volunteers, respectively (Figure 3, Table 1).
Table 1.
p53 pathway genes differentially expressed in 10 volunteers’ T cells during 3-week DI-SMG study.
Table 1.
p53 pathway genes differentially expressed in 10 volunteers’ T cells during 3-week DI-SMG study.
| # | NCBI Gene ID | Gene Symbol | Timepoint/ log2FC/ q-Value | Gene Name | Cite |
|---|---|---|---|---|---|
| 1 | 317 | APAF1 | 14 day/0.9944/0.0115 | apoptotic peptidase activating factor 1 | [] |
| 2 | 581 | BAX | 7 day/0.6738/0.0464 | BCL2-associated X, apoptosis regulator | [] |
| 3 | 27113 | BBC3 | 7 day/1.0363/0.0245 | BCL2 binding component 3 | [] |
| 4 | 891 | CCNB1 | 21 day/−1.1572/0.0027 | cyclin B1 | [] |
| 5 | 9133 | CCNB2 | 21 day/−2.1904/0.0038 | cyclin B2 | [] |
| 6 | 85417 | CCNB3 | 7 day/−2.3464/0.0014 | cyclin B3 | [] |
| 7 | 3732 | CD82 | 14 day/0.6182/0.0440 | CD82 molecule | [] |
| 8 | 983 | CDK1 | 14 day/−1.6466/0.0474 | cyclin-dependent kinase 1 | [] |
| 9 | 1019 | CDK4 | 7 day/1.1546/0.0060 | cyclin-dependent kinase 4 | [] |
| 10 | 1021 | CDK6 | 14 day/−1.355/3.34 × 10−6 | cyclin-dependent kinase 6 | [] |
| 11 | 1026 | CDKN1A | 7 day/−5.2107/2.27 × 10−32 | cyclin-dependent kinase inhibitor 1A | [] |
| 12 | 1026 | CDKN2A | 21 day/1.8168/0.0341 | cyclin-dependent kinase inhibitor 2A | [] |
| 13 | 54205 | CYCS | 7 day/−1.4602/3.51 × 10−8 | cytochrome c, somatic | [] |
| 14 | 1647 | GADD45A | 7 day/−3.9401/2.21 × 10−37 | growth arrest and DNA damage-inducible alpha | [] |
| 15 | 4616 | GADD45B | 7 day/−2.3869/3.54 × 10−32 | growth arrest and DNA damage-inducible beta | [] |
| 16 | 10912 | GADD45G | 28 day/−3.1241/4.80 × 10−3 | growth arrest and DNA damage-inducible gamma | [] |
| 17 | 51512 | GTSE1 | 28 day/−1.9802/1 × 10−5 | G2 and S-phase expressed 1 | [] |
| 18 | 3479 | IGF1 | 28 day/1.0213/0.0041 | insulin-like growth factor 1 | [,] |
| 19 | 3486 | IGFBP3 | 7 day/0.8320/0.0199 | insulin-like growth factor binding protein 3 | [] |
| 20 | 4194 | MDM4 | 7 day/−0.7708/0.0022 | MDM4 regulator of p53 | [] |
| 21 | 5366 | PMAIP1 | 7 day/−2.4366/2.24 × 10−11 | phorbol-12-myristate-13-acetate-induced protein 1 | [] |
| 22 | 27244 | SESN1 | 14 day/0.9173/0.0069 | sestrin 1 | [] |
| 23 | 2810 | SFN | 7 day/−2.5663/4.25 × 10−5 | stratifin | [] |
| 24 | 6477 | SIAH1 | 7 day/−2.134/0.0121 | siah E3 ubiquitin protein ligase 1 | [] |
| 25 | 55240 | STEAP3 | 21 day/−3.4598/0.0452 | STEAP3 metalloreductase | [] |
| 26 | 7057 | THBS1 | 7 day/−5.8423/4.97 × 10−13 | thrombospondin 1 | [] |
| 27 | 7157 | TP53 | 21 day/1.1392/0.00134 | tumor protein p53 | [,] |
| 28 | 63970 | TP53AIP1 | 28 day/0.89438/0.0434 | tp53-regulated apoptosis-inducing protein 1 | [] |
| 29 | 9540 | TP53I3 | 14 day/1.2081/0.0103 | tumor protein p53-inducible protein 3 | [] |
| 30 | 7161 | TP73 | 14 day/−2.3214/0.0387 | tumor protein p73 | [] |
A number of key genes and encoded proteins, previously shown to be linked to aging, longevity and cellular senescence, are associated with p53 network genes and corresponding proteins as indicated by STRING (Figure 4). Among them are important regulators of cell signaling pathways: proto-oncogenes Myc [] and Jun []; members of the NF-kB network (NFKB1, NFKB2, and NFKBIA) [,]; hypoxia-inducible factor 1-alpha (HIF1A) []; matrix metalloproteinase-9 (MMP9) []; and forkhead box P1 (FOXP1) [].
Figure 4.
The protein–protein interaction (PPI) network of the p53 signaling pathway. The PPI network was generated using the STRING database, with the input gene set comprising molecular components annotated in the KEGG p53 signaling pathway (hsa04115). The network illustrates the complex functional relationships between the components of this key pathway, which was significantly enriched in the DI-SMG study, and key regulators in interconnecting signaling pathways. Known (blue and purple edges) and predicted (green edges) interactions are shown.
2.5. Limitations of the Study and Future Perspectives
This study is subject to several limitations. Firstly, the cohort consisted of a small sample size (n = 10) of young, healthy male volunteers. Consequently, the findings may lack generalizability to female, older, or clinically compromised populations. Secondly, while the three-week DI-SMG protocol effectively models short-duration spaceflight, it may not fully recapitulate the physiological adaptations associated with longer mission or cumulative exposures. Finally, although microgravity was the variable of interest, the potential confounding influences of stress, physical inactivity, and confinement cannot be entirely discounted. To isolate the specific effects of microgravity simulation, future investigations should incorporate a parallel control group, such as subjects undergoing horizontal bed rest without SMG intervention. The transcriptomic changes identified here suggest a potential association with senescence. The direct measurement of senescence hallmarks, such as the secretion of SASP factors and the assessment of telomere attrition, will be critical for confirming the functional onset of senescence in future studies.
3. Materials and Methods
3.1. Dry Immersion Study Ethical Approval
All participated volunteers provided informed consent for the use and sharing of their fully anonymized data, according to the Helsinki Code of Medical Ethics for human samples. The Biomedical Ethics Committee of the IBMP RAS and Section of Physiology of the Bioethics Committee of the UNESCO National Bioethics Commission approved this study (Meeting No. 483 took place on 3 August 2018).
3.2. Dry Immersion Experiment Setup
An experiment with three weeks of exposure to DI-SMG without any countermeasures was performed at the Institute of Biomedical Problems (Figure 1A) during a period of 8 months from September 2018 to April 2019 with the participation of ten healthy men aged from 24 to 32 years as described [].
3.3. Peripheral Blood Sample Collection
In total, five samples of 10 mL peripheral blood were collected in sodium heparin tubes from each volunteer at five timepoints during the time course of the DI SMG study: at 7 days prior to Day 0 of DI (timepoint “−7 day”; background, BG), after 7 days, 14 days and 21 days in DI SMG, and on the 7th day after DI SMG (timepoint “28 days”) (Figure 1B).
3.4. CD3+ T Cell Isolation
Peripheral blood mononuclear cells (PBMCs) were obtained using the Ficoll density gradient centrifugation isolation protocol optimized for Ficoll-Paque™ PLUS (Cytiva, Uppsala, Sweden). Human T cells were enriched by the positive CD3 cell selection kit (EasySep™ Human CD3 Positive Selection Kit II, Stem Cell Technology, Vancouver, British Columbia, Canada) with a purity of about 95%.
3.5. RNA Extraction
Total RNA was purified from sorted cells using the RNeasy kit and treated with the RNAse-free DNAse kit (##74104; 79254 QIAGEN, Hilden, Germany). The total RNA concentration was measured (NanoDrop2000, Thermo Scientific, Wilmington, DE, USA) and RNA integrity and purity were evaluated by gel electrophoresis analysis (1 × TAE 1% UltraPureAgarose, #16500500 Invitrogen, Waltham, MA, USA). Gel images were created with the ImageQuant LAS 4000 LAS4000 Image system using ImageQuant LAS 4000 Control Software (GE Healthcare, Freiburg, Germany) as described []. All selected isolated RNA samples were subjected to and passed both the internal lab quality control (QC) and QC performed by a sequencing company (Novogene, Hongkong).
3.6. RNA-Seq and Bioinformatic Analysis
Sequencing libraries were constructed from ribodepleted RNA using a stranded protocol. RNA sequencing and data quality control were performed with the Illumina HiSeq-PE150 Platform using the HiSeq 2500 Sequencing System at Novogene (Hongkong). Sequences were mapped to the reference genome with Tophat, v.2.0.12. Reads were aligned to the human reference genome assembly in December 2013 (GRCh38/hg38). The quality of the resulting RNA-seq data was assessed, with key metrics summarized in Supplementary Table S1. All bioinformatic software applications, their versions and statistical parameters are listed in Supplementary Table S2. The method, including software and statistical parameters, for differential gene expression analysis is detailed in Supplementary Table S3. In gene expression analysis by Novogene (Hongkong), H-cluster, K-means, and SOM were used to cluster log2 (ratios). Genes with an adjusted p-value (q-value) of <0.05 and an absolute log2 fold change of >1 were considered significantly differentially expressed. The gene expression heatmap was composed in NASQAR v1.0 (New York University (NYU) Center for Health Informatics and Bioinformatics, New York, NY, USA) using log2 normalized count matrix RNA-seq reads. The functional classification of refined data for peak-related genes was performed using NCBI Gene resource www.ncbi.nlm.nih.gov (accessed on 22 June 2025) (Bethesda, MD, USA), as described previously [,,].
3.7. Statistical Analysis
A statistical analysis was performed to highlight the p53 gene expression trend over time course of the DI-SMG study. A linear trendline was calculated for each subject, and the resulting slope coefficients were analyzed using Excel (Microsoft 365 version (Microsoft Corporation, Redmond, WA, USA). The statistical inference of a significant upward trend was based on the statistical property that the 95% confidence interval around the mean slope was greater than zero.
3.8. Protein–Protein Interaction (PPI) Network
A network of protein–protein interactions (PPIs) was constructed using STRING (v12.0, Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland; string-db.org accessed on 19 September 2023) with all active evidence channels enabled, including Experiments, Databases, Co-expression, Neighborhood, Gene Fusion, Co-occurrence, and Textmining, and interactions were filtered to include only those with a high confidence score of ≥0.700; subsequently, KEGG pathway enrichment analysis was performed, with statistical significance defined by a corrected p-value of <0.05 using the Bonferroni method. The PPI network was generated with the input gene set comprising molecular components annotated in the KEGG p53 signaling pathway (hsa04115). The combined confidence scores, which integrated evidence from multiple channels, were calculated by STRING using a default phylogenetically curated collection of genomes as the statistical background.
4. Conclusions
The principle of design, uniqueness and robustness of the dry immersion system allows for original research to be carried out in simulated microgravity conditions close to orbital ones. T cell response to simulated microgravity includes the differential expression of p53 network aging-associated genes as well as interconnections with key regulators in interacting pathways. This finding opens an interesting opportunity to explore new protective strategies, which could, in time, provide insights into the future development of senolytics.
Supplementary Materials
The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms262211140/s1.
Author Contributions
Investigation, formal analysis, validation, writing—original draft preparation and visualization, N.V.K.; writing—review and editing, N.V.K. and E.T.; investigation, formal analysis, validation, D.D.V. and A.A.K.; conceptualization, project administration and funding acquisition, supervision, N.V.K., E.T. and M.L. All authors have read and agreed to the published version of the manuscript.
Funding
This study was supported by ASPIRE, the technology program management pillar of Abu Dhabi’s Advanced Technology Research Council (ATRC), via the ASPIRE Precision Medicine Research Institute Abu Dhabi (ASPIREPMRIAD) award grant number VRI-20-10.
Institutional Review Board Statement
All participating volunteers signed a written consent form according to the Helsinki Code of Medical Ethics for human samples. The Biomedical Ethics Committee of the IBMP RAS and Section of Physiology of the Bioethics Committee of the UNESCO National Bioethics Commission approved this study (Meeting No. 483 took place on 3 August 2018).
Data Availability Statement
The data that support the findings of this study are currently available in the NCBI GEO public repository for the editorial board and manuscript reviewers only. https://www.ncbi.nlm.nih.gov/geo/ [Gene Expression Omnibus] [https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE301964 (accessed on 22 June 2025)] [GSE301964].
Acknowledgments
The authors thank colleagues at New York University Abu Dhabi, the Karolinska Institute, and the Institute of Biomedical Problem for their assistance and lively discussions. We give special thanks to Sergey A. Ponomarev, Head of the Laboratory of Immune System Physiology at the IBMP RAS, for his invaluable help in implementing this project.
Conflicts of Interest
The authors declare no conflicts of interest.
Abbreviations
| BG | background |
| DI | dry immersion |
| FPKM | fragments per kilobase of transcript per million mapped fragments |
| MG | microgravity |
| PBMC | peripheral blood mononuclear cells |
| PPI | protein–protein interactions |
| QC | quality control |
| SMG | simulated microgravity |
References
- Goswami, N.; van Loon, J.J.; Roessler, A.; Blaber, A.P.; White, O. Gravitational physiology, aging and medicine. Front. Physiol. 2019, 10, 1338. [Google Scholar] [CrossRef] [PubMed]
- Kuznetsov, N.V.; Statsenko, Y.; Ljubisavljevic, M. An Update on Neuroaging on Earth and in Spaceflight. Int. J. Mol. Sci. 2025, 26, 1738. [Google Scholar] [CrossRef] [PubMed]
- 5 Hazards of Human Spaceflight. Available online: https://www.nasa.gov/hrp/hazards/ (accessed on 30 December 2024).
- Tomilovskaya, E.; Shigueva, T.; Sayenko, D.; Rukavishnikov, I.; Kozlovskaya, I. Dry immersion as a ground-based model of microgravity physiological effects. Front. Physiol. 2019, 10, 284. [Google Scholar] [CrossRef] [PubMed]
- Petitjean, A.; Mathe, E.; Kato, S.; Ishioka, C.; Tavtigian, S.V.; Hainaut, P.; Olivier, M. Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: Lessons from recent developments in the IARC TP53 database. Hum. Mutat. 2007, 28, 622–629. [Google Scholar] [CrossRef]
- Lane, D.P. p53, guardian of the genome. Nature 1992, 358, 6381. [Google Scholar] [CrossRef]
- Zaĭchuk, T.A.; Kuznetsov, N.V.; Osovskaia, V.S.; Kopnin, B.P.; Chumakov, P.M. Specific DNA-binding properties of oncoprotein p53 in human tumor cells. Dokl. Akad. Nauk. 1993, 330, 379–382. [Google Scholar]
- Zaichuk, T.A.; Kuznetsov, N.V.; Ossovskaya, V.S.; Kopnin, B.J.; Chumakov, P.M. Specific DNA-binding properties of p53 oncoprotein from human tumor cells. Proc. Russ. Acad. Sci. 1993, 330, 386–389. [Google Scholar]
- Kuznetsov, N.V. Analysis of DNA-Binding Properties of Protein P53. 1995. Available online: www.dslib.net/molekula/analiz-dnk-svjazyvajuwih-svojstv-belka-r53.html (accessed on 30 December 2024).
- Kondratov, R.V.; Kuznetsov, N.V.; Pugacheva, E.N.; Almazov, V.P.; Prasolov, V.S.; Kopnin, B.P.; Chumakov, P.M. Functional heterogeneity of p53-responsive elements. Mol. Biol. 1996, 30, 613–620. [Google Scholar]
- Kondratov, R.V.; Pugacheva, E.N.; Kuznetsov, N.V.; Prasolov, V.S.; Kopnin, B.P.; Chumakov, P.M. The human adenosine deaminase gene contains a p53-responsive element. Dokl. Akad. Nauk. 1996, 346, 260–262. [Google Scholar]
- Chumakov, P.M.; Pugacheva, E.N.; Ossovskaya, V.S.; Kondratov, R.V.; Kuznetsov, N.V.; Ivanov, A.V.; Prassolov, V.S.; Mazo, I.A.; Gudkov, A.V.; Kopnin, B.P. Functions of the p53 involved in control of malignant transformation. In Cancer and the Cell Cycle; ISREC Press: Lausanne, Switzerland, 1996. [Google Scholar]
- Kondratov, R.V.; Kuznetsov, N.V.; Pugacheva, E.N.; Almazov, V.P.; Prasolov, V.S.; Chumakov, P.M.; Kopnin, B.P. Functional. Mol. Biol. 1996, 30 (Suppl. 2), 363–367. [Google Scholar]
- Chumakov, P.M. Versatile functions of p53 protein in multicellular organisms. Biochemistry 2007, 72, 1399–1421. [Google Scholar] [CrossRef]
- Tyner, S.D.; Venkatachalam, S.; Choi, J.; Jones, S.; Ghebranious, N.; Igelmann, H.; Lu, X.; Soron, G.; Cooper, B.; Brayton, C.; et al. p53 mutant mice that display early ageing-associated phenotypes. Nature 2002, 415, 45–53. [Google Scholar] [CrossRef]
- Maier, B.; Gluba, W.; Bernier, B.; Turner, T.; Mohammad, K.; Guise, T.; Sutherland, A.; Thorner, M.; Scrable, H. Modulation of mammalian life span by the short isoform of p53. Genes Dev. 2004, 18, 306–319. [Google Scholar] [CrossRef] [PubMed]
- Reinhardt, H.C.; Schumacher, B. The p53 network: Cellular and systemic DNA damage responses in aging and cancer. Trends Genet. 2012, 28, 128–136. [Google Scholar] [CrossRef] [PubMed]
- Ohnishi, T.; Takahashi, A.; Wang, X.; Ohnishi, K.; Ohira, Y.; Nagaoka, S. Accumulation of a tumor suppressor p53 protein in rat muscle during a space flight. Mutat. Res. Fundam. Mol. Mech. Mutagen. 1999, 430, 271–274. [Google Scholar] [CrossRef] [PubMed]
- Li, H.Y.; Zhang, H.; Miao, G.Y.; Yi, X.I.; Chao, S.U.; Di, C.X.; Yang, L.I.; Liu, Y.Y.; Zhang, X.; Fei, X.; et al. Simulated microgravity conditions and carbon ion irradiation induce spermatogenic cell apoptosis and sperm DNA damage. Biomed. Environ. Sci. 2013, 26, 726–734. [Google Scholar]
- Takahashi, A.; Suzuki, H.; Omori, K.; Seki, M.; Hashizume, T.; Shimazu, T.; Ishioka, N.; Ohnishi, T. The expression of p53-regulated genes in human cultured lymphoblastoid TSCE5 and WTK1 cell lines during spaceflight. Int. J. Radiat. Biol. 2010, 86, 669–681. [Google Scholar] [CrossRef]
- Takahashi, A.; Suzuki, H.; Omori, K.; Seki, M.; Hashizume, T.; Shimazu, T.; Ishioka, N.; Ohnishi, T. Expression of p53-regulated proteins in human cultured lymphoblastoid TSCE5 and WTK1 cell lines during spaceflight. J. Radiat. Res. 2012, 53, 168–175. [Google Scholar] [CrossRef]
- Shi, L.; Tian, H.; Wang, P.; Li, L.; Zhang, Z.; Zhang, J.; Zhao, Y. Spaceflight and simulated microgravity suppresses macrophage development via altered RAS/ERK/NFκB and metabolic pathways. Cell. Mol. Immunol. 2021, 18, 1489–1502. [Google Scholar] [CrossRef]
- Popov, D.V.; Makhnovskii, P.A.; Zgoda, V.G.; Gazizova, G.R.; Vepkhvadze, T.F.; Lednev, E.M.; Motanova, E.S.; Lysenko, E.A.; Orlov, O.I.; Tomilovskaya, E.S. Rapid changes in transcriptomic profile and mitochondrial function in human soleus muscle after 3-day dry immersion. J. Appl. Physiol. 2023, 134, 1256–1264. [Google Scholar] [CrossRef]
- Battista, N.; Meloni, M.A.; Bari, M.; Mastrangelo, N.; Galleri, G.; Rapino, C.; Dainese, E.; Agro, A.F.; Pippia, P.; Maccarrone, M. 5-Lipoxygenase-dependent apoptosis of human lymphocytes in the International Space Station: Data from the ROALD experiment. FASEB J. 2012, 26, 1791–1798. [Google Scholar] [CrossRef]
- Rai, N.; Dey, S. Protective response of Sestrin under stressful conditions in aging. Ageing Res. Rev. 2020, 64, 101186. [Google Scholar] [CrossRef]
- Mathew, R.; Pal Bhadra, M.; Bhadra, U. Insulin/insulin-like growth factor-1 signalling (IIS) based regulation of lifespan across species. Biogerontology 2017, 18, 35–53. [Google Scholar] [CrossRef] [PubMed]
- Hong, S.; Kim, M.M. IGFBP-3 plays an important role in senescence as an aging marker. Environ. Toxicol. Pharmacol. 2018, 59, 138–145. [Google Scholar] [CrossRef] [PubMed]
- Eom, S.Y.; Kim, M.M. The effect of IGFBP3 gene knockout by the CRISPR/Cas9 system on the IGF-1 pathway in murine cells. Arch. Gerontol. Geriatr. 2024, 125, 105484. [Google Scholar] [CrossRef]
- Robles, A.I.; Bemmels, N.A.; Foraker, A.B.; Harris, C.C. APAF-1 is a transcriptional target of p53 in DNA damage-induced apoptosis. Cancer Res. 2001, 61, 6660–6664. [Google Scholar]
- Qian, S.; Wei, Z.; Yang, W.; Huang, J.; Yang, Y.; Wang, J. The role of BCL-2 family proteins in regulating apoptosis and cancer therapy. Front. Oncol. 2022, 12, 985363. [Google Scholar] [CrossRef]
- Hikisz, P.; Kiliańska, Z. PUMA, a critical mediator of cell death—One decade on from its discovery. Cell. Mol. Biol. Lett. 2012, 17, 646–669. [Google Scholar] [CrossRef]
- Moore, J.D.; Kirk, J.A.; Hunt, T. Unmasking the S-phase-promoting potential of cyclin B1. Science 2003, 300, 987–990. [Google Scholar] [CrossRef]
- Jackman, M.; Firth, M.; Pines, J. Human cyclins B1 and B2 are localized to strikingly different structures: B1 to microtubules, B2 primarily to the Golgi apparatus. EMBO J. 1995, 14, 1646–1654. [Google Scholar] [CrossRef] [PubMed]
- Lozano, J.C.; Perret, E.; Schatt, P.; Arnould, C.; Peaucellier, G.; Picard, A. Molecular cloning, gene localization, and structure of human cyclin B3. Biochem. Biophys. Res. Commun. 2002, 291, 406–413. [Google Scholar] [CrossRef] [PubMed]
- Christgen, M.; Bruchhardt, H.; Ballmaier, M.; Krech, T.; Länger, F.; Kreipe, H.; Lehmann, U. KAI1/CD82 is a novel target of estrogen receptor-mediated gene repression and downregulated in primary human breast cancer. Int. J. Cancer 2008, 123, 2239–2246. [Google Scholar] [CrossRef] [PubMed]
- Kipreos, E.T.; Wang, J.Y. Differential phosphorylation of c-Abl in cell cycle determined by cdc 2 kinase and phosphatase activity. Science 1990, 248, 217–220. [Google Scholar] [CrossRef]
- Ziegler, D.V.; Parashar, K.; Fajas, L. Beyond cell cycle regulation: The pleiotropic function of cdk4 in cancer. In Seminars in Cancer Biology; Academic Press: Cambridge, MA, USA, 2024; Volume 98, pp. 51–63. [Google Scholar]
- Nagasawa, M.; Melamed, I.; Kupfer, A.; Gelfand, E.W.; Lucas, J.J. Rapid nuclear translocation and increased activity of cyclin-dependent kinase 6 after T cell activation. J. Immunol. 1997, 158, 5146–5154. [Google Scholar] [CrossRef]
- Harper, J.W.; Adami, G.R.; Wei, N.; Keyomarsi, K.; Elledge, S.J. The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell 1993, 75, 805–816. [Google Scholar] [CrossRef]
- Wu, Y.; Jiang, D.; Liu, Q.; Yan, S.; Liu, X.; Wu, T.; Sun, W.; Li, G. Cathepsin L induces cellular senescence by upregulating CUX1 and p16INK4a. Aging 2024, 16, 10749. [Google Scholar] [CrossRef]
- Chapman, J.; Fielder, E.; Passos, J.F. Mitochondrial dysfunction and cell senescence: Deciphering a complex relationship. FEBS Lett. 2019, 593, 1566–1579. [Google Scholar] [CrossRef]
- Smith, M.L.; Chen, I.T.; Zhan, Q.; Bae, I.; Chen, C.Y.; Gilmer, T.M.; Kastan, M.B.; O’Connor, P.M.; Fornace, A.J. Interaction of the p53-regulated protein Gadd45 with proliferating cell nuclear antigen. Science 1994, 266, 1376–1380. [Google Scholar] [CrossRef]
- Verzella, D.; Bennett, J.; Fischietti, M.; Thotakura, A.K.; Recordati, C.; Pasqualini, F.; Capece, D.; Vecchiotti, D.; d’Andrea, D.; Di Francesco, B.; et al. GADD45β loss ablates innate immunosuppression in cancer. Cancer Res. 2018, 78, 1275–1292. [Google Scholar] [CrossRef]
- Zhang, P.; You, N.; Ding, Y.; Zhu, W.; Wang, N.; Xie, Y.; Huang, W.; Ren, Q.; Qin, T.; Fu, R.; et al. Gadd45g insufficiency drives the pathogenesis of myeloproliferative neoplasms. Nat. Commun. 2024, 15, 2989. [Google Scholar] [CrossRef]
- Lin, F.; Xie, Y.J.; Zhang, X.K.; Huang, T.J.; Xu, H.F.; Mei, Y.; Liang, H.; Hu, H.; Lin, S.T.; Luo, F.F.; et al. GTSE1 is involved in breast cancer progression in p53 mutation-dependent manner. J. Exp. Clin. Cancer Res. 2019, 38, 1–6. [Google Scholar] [CrossRef]
- Khan, M.Z.; Zugaza, J.L.; Aleman, I.T. The signaling landscape of insulin-like growth factor 1. J. Biol. Chem. 2024, 3, 108047. [Google Scholar] [CrossRef] [PubMed]
- Conover, C.A.; Oxvig, C. The Insulin-like Growth Factor System and Aging. Endocr. Rev. 2024, 17, bnae029. [Google Scholar]
- Almeida, O.P.; Hankey, G.J.; Yeap, B.B.; Paul Chubb, S.A.; Gollege, J.; Flicker, L. Risk of prevalent and incident dementia associated with insulin-like growth factor and insulin-like growth factor-binding protein 3. Mol. Psychiatry 2018, 23, 1825–1829. [Google Scholar] [CrossRef] [PubMed]
- Francoz, S.; Froment, P.; Bogaerts, S.; De Clercq, S.; Maetens, M.; Doumont, G.; Bellefroid, E.; Marine, J.C. Mdm4 and Mdm2 cooperate to inhibit p53 activity in proliferating and quiescent cells in vivo. Proc. Natl. Acad. Sci. USA 2006, 103, 3232–3237. [Google Scholar] [CrossRef]
- Morsi, R.Z.; Hage-Sleiman, R.; Kobeissy, H.; Dbaibo, G. Noxa: Role in cancer pathogenesis and treatment. Curr. Cancer Drug Targets 2018, 18, 914–928. [Google Scholar] [CrossRef]
- Budanov, A.V.; Sablina, A.A.; Feinstein, E.; Koonin, E.V.; Chumakov, P.M. Regeneration of peroxiredoxins by p53-regulated sestrins, homologs of bacterial AhpD. Science 2004, 304, 596–600. [Google Scholar] [CrossRef]
- Kim, Y.; Shiba-Ishii, A.; Nakagawa, T.; Iemura, S.I.; Natsume, T.; Nakano, N.; Matsuoka, R.; Sakashita, S.; Lee, S.; Kawaguchi, A.; et al. Stratifin regulates stabilization of receptor tyrosine kinases via interaction with ubiquitin-specific protease 8 in lung adenocarcinoma. Oncogene 2018, 37, 5387–5402. [Google Scholar] [CrossRef]
- Fujita, K.; Horikawa, I.; Mondal, A.M.; Jenkins, L.M.; Appella, E.; Vojtesek, B.; Bourdon, J.C.; Lane, D.P.; Harris, C.C. Positive feedback between p53 and TRF2 during telomere-damage signalling and cellular senescence. Nat. Cell Biol. 2010, 12, 1205–1212. [Google Scholar] [CrossRef]
- Wang, B.; Han, J.; Elisseeff, J.H.; Demaria, M. The senescence-associated secretory phenotype and its physiological and pathological implications. Nat. Rev. Mol. Cell Biol. 2024, 25, 958–978. [Google Scholar] [CrossRef] [PubMed]
- LeBlanc, A.J.; Kelm, N.Q. Thrombospondin-1, free radicals, and the coronary microcirculation: The aging conundrum. Antioxid. Redox Signal. 2017, 27, 785–801. [Google Scholar] [CrossRef] [PubMed]
- Lane, D.; Levine, A. p53 Research: The past thirty years and the next thirty years. Cold Spring Harb. Perspect. Biol. 2010, 2, a000893. [Google Scholar] [CrossRef] [PubMed]
- Fischer, M. Gene regulation by the tumor suppressor p53–The omics era. Biochim. Biophys. Acta (BBA) Rev. Cancer 2024, 11, 189111. [Google Scholar] [CrossRef]
- Liu, S.; Xu, T.; Chen, X.; Tang, L.; Li, L.; Zhang, L.; Yang, Y.; Huang, J. TP53AIP1 induce autophagy via the AKT/mTOR signaling pathway in the breast cancer cells. Cancer Biol. Ther. 2024, 25, 2398297. [Google Scholar] [CrossRef]
- Chaudhry, S.R.; Lopes, J.; Levin, N.K.; Kalpage, H.; Tainsky, M.A. Germline mutations in apoptosis pathway genes in ovarian cancer; the functional role of a TP53I3 (PIG3) variant in ROS production and DNA repair. Cell Death Discov. 2021, 7, 62. [Google Scholar] [CrossRef]
- Lopriore, P.; Capitanio, N.; Panatta, E.; Di Daniele, N.; Gambacurta, A.; Melino, G.; Amelio, I. TAp73 regulates ATP7A: Possible implications for ageing-related diseases. Aging 2018, 10, 3745. [Google Scholar] [CrossRef]
- Prochownik, E.V.; Wang, H. Lessons in aging from Myc knockout mouse models. Front. Cell Dev. Biol. 2023, 11, 1244321. [Google Scholar] [CrossRef]
- Yu, X.; Wang, Y.; Song, Y.; Gao, X.; Deng, H. AP-1 is a regulatory transcription factor of inflammaging in the murine kidney and liver. Aging Cell 2023, 22, e13858. [Google Scholar] [CrossRef]
- Cai, D.; Han, J.D. Aging-associated lncRNAs are evolutionarily conserved and participate in NFκB signaling. Nat. Aging 2021, 1, 438–453. [Google Scholar] [CrossRef]
- Haga, M.; Okada, M. Systems approaches to investigate the role of NF-κB signaling in aging. Biochem. J. 2022, 479, 161–183. [Google Scholar] [CrossRef] [PubMed]
- Alique, M.; Sánchez-López, E.; Bodega, G.; Giannarelli, C.; Carracedo, J.; Ramírez, R. Hypoxia-inducible factor-1α: The master regulator of endothelial cell senescence in vascular aging. Cells 2020, 9, 195. [Google Scholar] [CrossRef] [PubMed]
- Cancemi, P.; Aiello, A.; Accardi, G.; Caldarella, R.; Candore, G.; Caruso, C.; Ciaccio, M.; Cristaldi, L.; Di Gaudio, F.; Siino, V.; et al. The role of matrix metalloproteinases (MMP-2 and MMP-9) in ageing and longevity: Focus on sicilian long-living individuals (LLIs). Mediat. Inflamm. 2020, 2020, 8635158. [Google Scholar] [CrossRef] [PubMed]
- Wu, M.; Tang, W.; Chen, Y.; Xue, L.; Dai, J.; Li, Y.; Zhu, X.; Wu, C.; Xiong, J.; Zhang, J.; et al. Spatiotemporal transcriptomic changes of human ovarian aging and the regulatory role of FOXP1. Nat. Aging 2024, 4, 527–545. [Google Scholar] [CrossRef]
- Kutko, O.V.; Rykova, M.P.; Antropova, E.N.; Kalinin, S.A.; Shulgina, S.M.; Sadova, A.A.; Orlova, K.D.; Kiseleva, D.D.; Smarov, V.A.; Vassilieva, G.Y.; et al. Effect of 21-day dry immersion on the production of cytokines involved in the regulation of bone metabolism by T cells. Hum. Physiol. 2020, 46, 787–791. [Google Scholar] [CrossRef]
- Kuznetsov, N.V.; Almuzzaini, B.; Kritikou, J.S.; Baptista, M.A.P.; Oliveira, M.M.S.; Keszei, M.; Snapper, S.B.; Percipalle, P.; Westerberg, L.S. Nuclear Wiskott-Aldrich syndrome protein co-regulates T cell factor 1-mediated transcription in T cells. Genome Med. 2017, 9, 91. [Google Scholar] [CrossRef]
- Kouznetsov, N.V. Cell responses to simulated microgravity and hydrodynamic stress can be distinguished by using comparative transcriptomic analysis. bioRxiv 2021. [Google Scholar] [CrossRef]
- Kuznetsov, N.V. Expression of the p53 Network Aging-associated Genes “shaken, not stirred” in a 3-Week Microgravity Simulation Human Study. bioRxiv 2025. [Google Scholar] [CrossRef]
- Kouznetsov, N.V. Cell Responses to Simulated Microgravity and Hydrodynamic Stress Can Be Distinguished by Comparative Transcriptomics. Int. J. Transl. Med. 2022, 2, 364–386. [Google Scholar] [CrossRef]
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).