Volume 41 Issue 6
Jun.  2025
Turn off MathJax
Article Contents
He Zhanchen, Shang Yuxuan, Xu Xiangping, et al. Multi-omics Mendelian randomization study on the causality between non-ionizing radiation and facial aging[J]. CHINESE JOURNAL OF BURNS AND WOUNDS, 2025, 41(6): 594-603. Doi: 10.3760/cma.j.cn501225-20240830-00320
Citation: He Zhanchen, Shang Yuxuan, Xu Xiangping, et al. Multi-omics Mendelian randomization study on the causality between non-ionizing radiation and facial aging[J]. CHINESE JOURNAL OF BURNS AND WOUNDS, 2025, 41(6): 594-603. Doi: 10.3760/cma.j.cn501225-20240830-00320

Multi-omics Mendelian randomization study on the causality between non-ionizing radiation and facial aging

doi: 10.3760/cma.j.cn501225-20240830-00320
Funds:

Scientific Research Fund of Hunan Provincial Education Department of China 24C091

Natural Science Foundation of Hunan Province of China 2024JJ7457

More Information
  •   Objective  To investigate the causality between non-ionizing radiation and facial aging, and to identify potential genes associated with facial aging.  Methods  This study employed a method of analysis based on multiple Mendelian randomization (MR). Genome-wide association study data of non-ionizing radiation (FinnGen database, n=218 281) and facial aging (UK Biobank database, n=423 999) were retrieved. Single nucleotide polymorphisms (SNPs) were used as instrumental variables, with a significance threshold (P < 5×10-6) applied and further linkage disequilibrium analysis performed to select SNPs associated with non-ionizing radiation. Two-sample MR (TSMR) analysis was conducted to assess the causality between non-ionizing radiation and facial aging, using inverse variance weighting (IVW) method as the primary analytical method and supplementing with MR-Egger regression, weighted median, weighted mode, and simple mode methods for validation. For the selected non-ionizing radiation-associated SNPs, heterogeneity was tested by Cochran Q test, horizontal pleiotropy was assessed by the MR-Egger intercept test and MR-PRESSO test, and robustness was evaluated via leave-one-out analysis. Multivariable MR (MVMR) analysis was performed to adjust for confounding factors affecting facial aging including smoking frequency, blood alcohol concentration, exercise frequency, body mass index, and systolic and diastolic blood pressure. Summary-data-based MR (SMR) analysis using expression quantitative trait loci (eQTL) data was conducted to screen candidate genes of facial aging, which were then validated by TSMR analysis. Protein quantitative trait loci (pQTL) and methylation quantitative trait loci (mQTL) data were analyzed by TSMR analysis to examine the causal role of MED1 gene with facial aging from multi-omics aspect. The genetic association of MED1 gene with facial aging was verified by colocalization analysis (posterior probability H4 > 50%).  Results  Twenty non-ionizing radiation-related SNPs that reached the significance threshold were screened out, with F values being all > 10. IVW analysis demonstrated a positive causality between non-ionizing radiation and facial aging (with odds ratio of 1.02, with 95% confidence interval of 1.01-1.02, P < 0.05). The analysis results of MR-Egger regression, weighted median, simple mode method, and weighted mode method (with odds ratios of 1.02, 1.02, 1.01, and 1.01, respectively, with 95% confidence intervals of 1.01-1.03, 1.01-1.02, 0.99-1.02, respectively, P < 0.05) were consistent with IVW method. For these 20 non-ionizing radiation-related SNPs, Cochran Q test under IVW method and MR-Egger showed no significant heterogeneity (with Q values of 23.20 and 22.59, respectively, P > 0.05); the MR-Egger intercept test (with intercept absolute value of 0.01, with standard error of 0.01, P > 0.05) and MR-PRESSO test (P > 0.05) indicated no horizontal pleiotropy. Leave-one-out analysis further confirmed that no individual SNP had a significant effect on the results. After correction of confounding factors such as systolic blood pressure, diastolic blood pressure, smoking frequency, blood alcohol concentration, body mass index, and exercise frequency, MVMR analysis showed that non-ionizing radiation remained a risk factor for facial aging (with odds ratios of 1.01, 1.01, 1.02, 1.02, 1.01, and 1.04, respectively, with 95% confidence intervals of 1.01-1.02, 1.01-1.02, 1.01-1.02, 1.01-1.02, 1.00-1.01, and 1.03-1.05, respectively, all P values < 0.05). SMR analysis identified 12 potential facial aging-related genes (SENP7, CCND1, LTBP2, IKZF3, MED1, ORMDL3, ZBTB7B, LOX, NEBL, EXOSC6, PSMA4, and EIF2B2, with odds ratios of 1.01, 1.03, 1.04, 0.99, 1.04, 1.01, 1.06, 0.88, 1.01, 0.99, 1.04, and 0.99, respectively, all P values < 0.05). Subsequent TSMR analysis retained 6 risk genes (ZBTB7B, SENP7, NEBL, MED1, PSMA4, and ORMDL3, with odds ratios of 1.04, 1.01, 1.00, 1.02, 1.03, and 1.01, respectively, with 95% confidence intervals of 1.02-1.05, 1.00-1.01, 1.00-1.01, 1.01-1.03, 1.01-1.04, and 1.00-1.01, respectively, all P values < 0.05) for facial aging and 4 protective genes (LOX, EIF2B2, EXOSC6, and IKZF3, with odds ratios of 0.92, 0.99, 0.99, and 0.99, respectively, with 95% confidence intervals of 0.90-0.94, 0.99-0.99, 0.99-1.00, and 0.99-1.00, respectively, all P values < 0.05). TSMR analysis based on pQTL data showed the MED1 protein was positively associated with facial aging (with odds ratio of 1.04, P < 0.05), which was consistent with the causal direction observed in eQTL-based SMR and TSMR analyses. TSMR analysis based on mQTL data indicated MED1 gene methylation (with probes of cg15445000 and cg03013999) had a protective effect on facial aging (with odds ratios of 0.99 and 0.99, respectively, both P values < 0.05). Colocalization analysis yielded a posterior probability H4=58.4%, suggesting that MED1 gene and facial aging likely shared the same causal genetic variant.  Conclusions  Through multi-omics MR analyses, it has confirmed that there is a causality between non-ionizing radiation and facial aging, which remained highly significant after correcting for potential confounders such as smoking frequency, blood alcohol concentration, exercise frequency, and the others. Clearly, 10 genes including SENP7, NEBL, EIF2B2, PSMA4, EXOSC6, IKZF3, ORMDL3, ZBTB7B, LOX, and MED1, particularly the MED1, may be involved in the process of facial aging.

     

  • (1) The research findings were based on publicly available large-scale genome-wide association study data, which have the advantages of large sample sizes and reduced impact to confounding factors, and is therefore more convincing compared with traditional observational studies.
    (2) Through multi-omics Mendelian randomization analysis, it was confirmed that there was a causality between non-ionizing radiation and facial aging, which remained robust after adjusting for potential confounders including smoking frequency, blood alcohol concentration, exercise frequency, body mass index, systolic blood pressure, and diastolic blood pressure. The study identified that 10 genes, i.e., SENP7, NEBL, EIF2B2, PSMA4, EXOSC6, IKZF3, ORMDL3, ZBTB7B, LOX, and MED1, which may be involved in the process of facial aging. Among them, the MED1 gene might be a potential drug target for the assessment and treatment of facial aging.
  • loading
  • [1]
    Swift A, Liew S, Weinkle S, et al. The facial aging process from the "inside out"[J]. Aesthet Surg J, 2021, 41(10): 1107-1119. DOI: 10.1093/asj/sjaa339.
    [2]
    Franco AC, Aveleira C, Cavadas C. Skin senescence: mechanisms and impact on whole-body aging[J]. Trends Mol Med, 2022, 28(2): 97-109. DOI: 10.1016/j.molmed.2021.12.003.
    [3]
    Tuieng RJ, Cartmell SH, Kirwan CC, et al. The effects of ionising and non-ionising electromagnetic radiation on extracellular matrix proteins[J]. Cells, 2021, 10(11): 3041. DOI: 10.3390/cells10113041.
    [4]
    Pittayapruek P, Meephansan J, Prapapan O, et al. Role of matrix metalloproteinases in photoaging and photocarcinogenesis[J]. Int J Mol Sci, 2016, 17(6): 868. DOI: 10.3390/ijms17060868.
    [5]
    Bang E, Kim DH, Chung HY. Protease-activated receptor 2 induces ROS-mediated inflammation through Akt-mediated NF-κB and FoxO6 modulation during skin photoaging[J]. Redox Biol, 2021, 44: 102022. DOI: 10.1016/j.redox.2021.102022.
    [6]
    Birney E. Mendelian randomization[J]. Cold Spring Harb Perspect Med, 2022, 12(4): a041302. DOI: 10.1101/cshperspect.a041302.
    [7]
    易美慧, 郭晔. 儿童核心结合因子相关急性髓细胞白血病的细胞分子遗传学异常与预后[J]. 国际输血及血液学杂志, 2019, 42(2): 52-57. DOI: 10.3760/cma.j.issn.1673-419X.2019.02.013.
    [8]
    于秋霜, 李凌勋, 陶怡娜, 等. 免疫细胞与脓毒症的因果关联: 一项基于孟德尔随机化方法的研究[J]. 中华危重病急救医学, 2024, 36(8): 821-828. DOI: 10.3760/cma.j.cn121430-20240527-00462.
    [9]
    Bourassa KJ, Moffitt TE, Ambler A, et al. Association of treatable health conditions during adolescence with accelerated aging at midlife[J]. JAMA Pediatr, 2022, 176(4): 392-399. DOI: 10.1001/jamapediatrics.2021.6417.
    [10]
    Kadunce DP, Burr R, Gress R, et al. Cigarette smoking: risk factor for premature facial wrinkling[J]. Ann Intern Med, 1991, 114(10): 840-844. DOI: 10.7326/0003-4819-114-10-840.
    [11]
    Rungratanawanich W, Qu Y, Wang X, et al. Advanced glycation end products (AGEs) and other adducts in aging-related diseases and alcohol-mediated tissue injury [J]. Exp Mol Med, 2021, 53(2): 168-188. DOI: 10.1038/s12276-021-00561-7.
    [12]
    Bencivenga L, De Souto Barreto P, Rolland Y, et al. Blood pressure variability: a potential marker of aging[J]. Ageing Res Rev, 2022, 80: 101677. DOI: 10.1016/j.arr.2022.101677.
    [13]
    Lacolley P, Regnault V, Segers P, et al. Vascular smooth muscle cells and arterial stiffening: relevance in development, aging, and disease[J]. Physiol Rev, 2017, 97(4): 1555-1617. DOI: 10.1152/physrev.00003.2017.
    [14]
    Sanderson E. Multivariable Mendelian randomization and mediation[J]. Cold Spring Harb Perspect Med, 2021, 11(2): a038984. DOI: 10.1101/cshperspect.a038984.
    [15]
    Zhou W, Liu G, Hung RJ, et al. Causal relationships between body mass index, smoking and lung cancer: univariable and multivariable Mendelian randomization[J]. Int J Cancer, 2021, 148(5): 1077-1086. DOI: 10.1002/ijc.33292.
    [16]
    Xu S, Li X, Zhang S, et al. Oxidative stress gene expression, DNA methylation, and gut microbiota interaction trigger Crohn's disease: a multi-omics Mendelian randomization study[J]. BMC Med, 2023, 21(1): 179. DOI: 10.1186/s12916-023-02878-8.
    [17]
    Yang H, Liu D, Zhao C, et al. Mendelian randomization integrating GWAS and eQTL data revealed genes pleiotropically associated with major depressive disorder [J]. Transl Psychiatry, 2021, 11(1): 225. DOI: 10.1038/s41398-021-01348-0.
    [18]
    Su WM, Gu XJ, Dou M, et al. Systematic druggable genome-wide Mendelian randomisation identifies therapeutic targets for Alzheimer's disease[J]. J Neurol Neurosurg Psychiatry, 2023, 94(11): 954-961. DOI: 10.1136/jnnp-2023-331142.
    [19]
    Ferkingstad E, Sulem P, Atlason BA, et al. Large-scale integration of the plasma proteome with genetics and disease[J]. Nat Genet, 2021, 53(12): 1712-1721. DOI: 10.1038/s41588-021-00978-w.
    [20]
    McRae AF, Marioni RE, Shah S, et al. Identification of 55, 000 replicated DNA methylation QTL[J]. Sci Rep, 2018, 8(1): 17605. DOI: 10.1038/s41598-018-35871-w.
    [21]
    Wu Y, Zeng J, Zhang F, et al. Integrative analysis of omics summary data reveals putative mechanisms underlying complex traits[J]. Nat Commun, 2018, 9(1): 918. DOI: 10.1038/s41467-018-03371-0.
    [22]
    Sreedhar A, Aguilera-Aguirre L, Singh KK. Mitochondria in skin health, aging, and disease[J]. Cell Death Dis, 2020, 11(6): 444. DOI: 10.1038/s41419-020-2649-z.
    [23]
    López-Otín C, Blasco MA, Partridge L, et al. Hallmarks of aging: an expanding universe[J]. Cell, 2023, 186(2): 243-278. DOI: 10.1016/j.cell.2022.11.001.
    [24]
    Clarke TL, Mostoslavsky R. DNA repair as a shared hallmark in cancer and ageing[J]. Mol Oncol, 2022, 16(18): 3352-3379. DOI: 10.1002/1878-0261.13285.
    [25]
    Aging Biomarker Consortium, Bao H, Cao J, et al. Biomarkers of aging[J]. Sci China Life Sci, 2023, 66(5): 893-1066. DOI: 10.1007/s11427-023-2305-0.
    [26]
    Zimmermann A, Madreiter-Sokolowski C, Stryeck S, et al. Targeting the mitochondria-proteostasis axis to delay aging [J]. Front Cell Dev Biol, 2021, 9: 656201. DOI: 10.3389/fcell.2021.656201.
    [27]
    Wilson N, Kataura T, Korsgen ME, et al. The autophagy-NAD axis in longevity and disease[J]. Trends Cell Biol, 2023, 33(9): 788-802. DOI: 10.1016/j.tcb.2023.02.004.
    [28]
    Tokarz J, Möller G, Artati A, et al. Common muscle metabolic signatures highlight arginine and lysine metabolism as potential therapeutic targets to combat unhealthy aging[J]. Int J Mol Sci, 2021, 22 (15): 7958. DOI: 10.3390/ijms22157958.
    [29]
    Weng Z, Wang Y, Ouchi T, et al. Mesenchymal stem/stromal cell senescence: hallmarks, mechanisms, and combating strategies[J]. Stem Cells Transl Med, 2022, 11(4): 356-371. DOI: 10.1093/stcltm/szac004.
    [30]
    Zheng L, He S, Wang H, et al. Targeting cellular senescence in aging and age-related diseases: challenges, considerations, and the emerging role of senolytic and senomorphic therapies[J]. Aging Dis, 2024, 15(6): 2554-2594. DOI: 10.14336/AD.2024.0206.
    [31]
    Lv H, Gao N, Zhou Q, et al. Collagen-based dissolving microneedles with flexible pedestals: a transdermal delivery system for both anti-aging and skin diseases[J]. Adv Healthc Mater, 2023, 12(21): e2203295. DOI: 10.1002/adhm.202203295.
    [32]
    Tran DK, Phuong T, Bui NL, et al. Exploring the potential of stem cell-based therapy for aesthetic and plastic surgery [J]. IEEE Rev Biomed Eng, 2023, 16: 386-402. DOI: 10.1109/RBME.2021.3134994.
    [33]
    Montoni A, George KM, Soeur J, et al. Chronic UVA1 irradiation of human dermal fibroblasts: persistence of DNA damage and validation of a cell cultured-based model of photoaging[J]. J Invest Dermatol, 2019, 139(8): 1821-1824.e3. DOI: 10.1016/j.jid.2019.02.022.
    [34]
    Cenizo V, André V, Reymermier C, et al. LOXL as a target to increase the elastin content in adult skin: a dill extract induces the LOXL gene expression[J]. Exp Dermatol, 2006, 15(8): 574-581. DOI: 10.1111/j.1600-0625.2006.00442.x.
    [35]
    Majora M, Wittkampf T, Schuermann B, et al. Functional consequences of mitochondrial DNA deletions in human skin fibroblasts: increased contractile strength in collagen lattices is due to oxidative stress-induced lysyl oxidase activity[J]. Am J Pathol, 2009, 175(3): 1019-1029. DOI: 10.2353/ajpath.2009.080832.
    [36]
    Weihermann AC, de Carvalho CM, Schuck DC, et al. Modulation of photoaging-induced cutaneous elastin: evaluation of gene and protein expression of markers related to elastogenesis under different photoexposure conditions[J]. Dermatol Ther (Heidelb), 2021, 11(6): 2043-2056. DOI: 10.1007/s13555-021-00603-y.
    [37]
    Renard E, Chadjichristos C, Kypriotou M, et al. Chondroitin sulphate decreases collagen synthesis in normal and scleroderma fibroblasts through a Smad-independent TGF-beta pathway--implication of C-Krox and Sp1[J]. J Cell Mol Med, 2008, 12(6B): 2836-2847. DOI: 10.1111/j.1582-4934.2008.00287.x.
    [38]
    Li C, Stoma S, Lotta LA, et al. Genome-wide association analysis in humans links nucleotide metabolism to leukocyte telomere length[J]. Am J Hum Genet, 2020, 106(3): 389-404. DOI: 10.1016/j.ajhg.2020.02.006.
    [39]
    Villicaña S, Castillo-Fernandez J, Hannon E, et al. Genetic impacts on DNA methylation help elucidate regulatory genomic processes[J]. Genome Biol, 2023, 24(1): 176. DOI: 10.1186/s13059-023-03011-x.
    [40]
    Zhu H, Ren S, Bitler BG, et al. SPOP E3 ubiquitin ligase adaptor promotes cellular senescence by degrading the SENP7 deSUMOylase[J]. Cell Rep, 2015, 13(6): 1183-1193. DOI: 10.1016/j.celrep.2015.09.083.
    [41]
    Wang H, Guan T, Hu R, et al. Targeting KAT7 inhibits the progression of colorectal cancer[J]. Theranostics, 2025, 15(4): 1478-1495. DOI: 10.7150/thno.106085.
    [42]
    He L, Khanal P, Morse CI, et al. Differentially methylated gene patterns between age-matched sarcopenic and non-sarcopenic women[J]. J Cachexia Sarcopenia Muscle, 2019, 10(6): 1295-1306. DOI: 10.1002/jcsm.12478.
    [43]
    Li J, Hu S, Zhang Z, et al. LASP2 is downregulated in human liver cancer and contributes to hepatoblastoma cell malignant phenotypes through MAPK/ERK pathway[J]. Biomed Pharmacother, 2020, 127: 110154. DOI: 10.1016/j.biopha.2020.110154.
    [44]
    Yomogida K, Trsan T, Sudan R, et al. The transcription factor Aiolos restrains the activation of intestinal intraepithelial lymphocytes[J]. Nat Immunol, 2024, 25(1): 77-87. DOI: 10.1038/s41590-023-01693-w.
    [45]
    Mariani JN, Mansky B, Madsen PM, et al. Repression of developmental transcription factor networks triggers aging-associated gene expression in human glial progenitor cells[J]. Nat Commun, 2024, 15(1): 3873. DOI: 10.1038/s41467-024-48118-2.
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(4)

    Article Metrics

    Article views (140) PDF downloads(6) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return