DNA Methylation as a Marker of Body Shape in Premenopausal Women

Adeline Divoux, Alexey Eroshkin, Edina Erdos, Katalin Sandor, Timothy F Osborne, Steven R Smith, Adeline Divoux, Alexey Eroshkin, Edina Erdos, Katalin Sandor, Timothy F Osborne, Steven R Smith

Abstract

Preferential accumulation of fat in the gluteo-femoral (GF) depot (pear shape) rather than in the abdominal (A) depot (apple shape), protects against the development of metabolic diseases but the underlying molecular mechanism is still unknown. Recent data, including our work, suggest that differential epigenetic marking is associated with regulation of genes attributed to distinct fat distribution. Here, we aimed to compare the genomic DNA methylation signatures between apple and pear-shaped premenopausal women. To investigate the contribution of upper and lower body fat, we used paired samples of A-FAT and GF-FAT, analyzed on the BeadChip Methylation Array and quantified the differentially methylated sites between the 2 groups of women. We found unique DNA methylation patterns within both fat depots that are significantly different depending on the body fat distribution. Around 60% of the body shape specific DNA methylation sites identified in adipose tissue are maintained ex vivo in cultured preadipocytes. As it has been reported before in other cell types, we found only a hand full of genes showing coordinated differential methylation and expression levels. Finally, we determined that more than 50% of the body shape specific DNA methylation sites could also be detected in whole blood derived DNA. These data reveal a strong DNA methylation program associated with adipose tissue distribution with the possibility that a simple blood test could be used as a predictive diagnostic indicator of young women who are at increased risk for progressing to the apple body shape with a higher risk of developing obesity related complications. Clinical Trial Registration:https://ichgcp.net/clinical-trials-registry/NCT02728635 and https://ichgcp.net/clinical-trials-registry/NCT02226640, identifiers NCT02728635 and NCT02226640.

Keywords: DNA methylation; adipose tissues; blood marker; fat distribution; metabolic risk; preadipocytes.

Conflict of interest statement

AE is employed by Prescient Metabiomics Corp. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Divoux, Eroshkin, Erdos, Sandor, Osborne and Smith.

Figures

FIGURE 1
FIGURE 1
Differentially methylated sites between apple and pear-shaped women in A-FAT and GF-FAT. (A,B) Volcano plot showing differences in DNA methylation between 10 apple and 7 pear-shaped subjects in A- FAT (A-left side), GF-FAT (A-right side), A-preadipocytes (B-left side), and GF-preadipocytes (B-right side). Each point represents a CpG site significantly differentially methylated with a β-value difference between both groups (methylation difference Apples vs. Pears) superior to 0.2. Purple color represents the sites more methylated in Apples. Blue color represents the sites more methylated in Pears. (C,D) The Venn diagrams represent the number of differential expressed genes and methylated sites between 10 apples and 7 pears women in A-preadipocytes (C) and GF-preadipocytes (D).
FIGURE 2
FIGURE 2
Shared body shape differentially methylated sites between adipose tissue, cultured preadipocytes and whole blood (A,B). The 2 Venn diagrams depict the differentially methylated sites between 10 apple and 7 pear-shaped subjects in A (A) and GF (B) depot only in adipose tissue, only in preadipocytes or in both. Purple color depicts the number of unique genes annotated to sites hypermethylated in apple-shaped women. Blue color depicts the number of unique genes annotated to sites hypermethylated in pear-shaped women. (C) Pathway Analysis of the genes nearest the body shape specific DMS in A depot (A) and GF depot (GF). Only the common genes between whole tissue and cells were used. The size of the dots represents gene count. Only pathways with p-value < 0.05 are represented. (D) The Venn diagram depicts the common body shape DMS between adipose tissue, preadipocytes and blood for the 17 subjects. A = Abdominal, GF = Gluteo-femoral, DMS = Differentially Methylated Site.
FIGURE 3
FIGURE 3
Three of 5 body shape specific DMS found in blood were validated in an independent group of women. (A) Linear regression shows positive association between waist to hip ratio and the ratio of fat accumulated in the legs relative to the total fat mass in the initial group of women. (B,C) Percentage of DNA methylation in 5 apple- vs. 5 pear-shaped women for 2 apple shaped-specific DMS (B) and 1 pear shaped-specific DMS (C). Mean ± SEM is shown. The p-value of non-parametric Mann Whitney test are reported Level of CpG sites methylation was measured by pyrosequencing in DNA isolated from total blood samples. (D) Genomic characteristics of the CpG sites showing differential level of methylation between apple- and pear-shaped women. Chr, Chromosome, DMS, Differentially Methylated Sites, FC, Fold Change.

References

    1. Al Ajlouni M., Abujbara M., Batieha A., Ajlouni K. (2015). Prevalence of lipohypertrophy and associated risk factors in insulin-treated patients with type 2 diabetes mellitus. Int. J. Endocrinol. Metabol. 13:e20776.
    1. Blüher M. (2013). Importance of estrogen receptors in adipose tissue function. Mol. Metabol. 2 130–132. 10.1016/j.molmet.2013.07.001
    1. Dahlman I., Rydén M., Brodin D., Grallert H., Strawbridge R. J., Arner P. (2016). Numerous Genes in Loci Associated With Body Fat Distribution Are Linked to Adipose Function. Diabetes 65 433–437. 10.2337/db15-0828
    1. Divoux A., Karastergiou K., Xie H., Guo W., Perera R. J., Fried S. K., et al. (2014). Identification of a novel lncRNA in gluteal adipose tissue and evidence for its positive effect on preadipocyte differentiation. Obesity 22 1781–1785. 10.1002/oby.20793
    1. Divoux A., Sandor K., Bojcsuk D., Talukder A., Li X., Balint B. L., et al. (2018). Differential open chromatin profile and transcriptomic signature define depot-specific human subcutaneous preadipocytes: primary outcomes. Clin. Epigenet. 10:148.
    1. Divoux A., Sandor K., Bojcsuk D., Yi F., Hopf M. E., Smith J. S., et al. (2020). Fat Distribution in Women Is Associated With Depot-Specific Transcriptomic Signatures and Chromatin Structure. J. Endocr. Soc. 4:bvaa042.
    1. Divoux A., Xie H., Li J. L., Karastergiou K., Perera R. J., Chang R. J., et al. (2017). MicroRNA-196 Regulates HOX Gene Expression in Human Gluteal Adipose Tissue. Obesity 25 1375–1383. 10.1002/oby.21896
    1. Ejarque M., Ceperuelo-Mallafré V., Serena C., Maymo-Masip E., Duran X., Díaz-Ramos A., et al. (2019). Adipose tissue mitochondrial dysfunction in human obesity is linked to a specific DNA methylation signature in adipose-derived stem cells. Int. J. Obes. 43 1256–1268. 10.1038/s41366-018-0219-6
    1. Farré P., Jones M. J., Meaney M. J., Emberly E., Turecki G., Kobor M. S. (2015). Concordant and discordant DNA methylation signatures of aging in human blood and brain. Epigenet. Chromat. 8:19.
    1. Federico A., Monti S. (2020). hypeR: an R package for geneset enrichment workflows. Bioinformatics 36 1307–1308. 10.1093/bioinformatics/btz700
    1. Fontaine C., Cousin W., Plaisant M., Dani C., Peraldi P. (2008). Hedgehog signaling alters adipocyte maturation of human mesenchymal stem cells. Stem Cells 26 1037–1046. 10.1634/stemcells.2007-0974
    1. Hilton C., Neville M. J., Wittemans L. B. L., Todorcevic M., Pinnick K. E., Pulit S. L., et al. (2019). MicroRNA-196a links human body fat distribution to adipose tissue extracellular matrix composition. EBioMedicine 44 467–475. 10.1016/j.ebiom.2019.05.047
    1. Huang L. O., Rauch A., Mazzaferro E., Preuss M., Carobbio S., Bayrak C. S., et al. (2021). Genome-wide discovery of genetic loci that uncouple excess adiposity from its comorbidities. Nat. Metabol. 3 228–243. 10.1038/s42255-021-00346-2
    1. Hur S. S. J., Cropley J. E., Suter C. M. (2017). Paternal epigenetic programming: evolving metabolic disease risk. J. Mol. Endocrinol. 58 R159–R168.
    1. Jones P. A. (2012). Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat. Rev. Genet. 13 484–492. 10.1038/nrg3230
    1. Kaminsky Z., Wilcox H. C., Eaton W. W., Van Eck K., Kilaru V., Jovanovic T., et al. (2015). Epigenetic and genetic variation at SKA2 predict suicidal behavior and post-traumatic stress disorder. Translat. Psychiat. 5:e627. 10.1038/tp.2015.105
    1. Karastergiou K., Fried S. K., Xie H., Lee M. J., Divoux A., Rosencrantz M. A., et al. (2013). Distinct developmental signatures of human abdominal and gluteal subcutaneous adipose tissue depots. J. Clin. Endocrinol. Metabol. 98 362–371. 10.1210/jc.2012-2953
    1. Karpe F., Pinnick K. E. (2015). Biology of upper-body and lower-body adipose tissue–link to whole-body phenotypes. Nat. Rev. Endocrinol. 11 90–100. 10.1038/nrendo.2014.185
    1. Kleiber M. L., Singh S. M. (2009). Divergence of the vertebrate sp1A/ryanodine receptor domain and SOCS box-containing (Spsb) gene family and its expression and regulation within the mouse brain. Genomics 93 358–366. 10.1016/j.ygeno.2008.11.011
    1. Klug M., Heinz S., Gebhard C., Schwarzfischer L., Krause S. W., Andreesen R., et al. (2010). Active DNA demethylation in human postmitotic cells correlates with activating histone modifications, but not transcription levels. Genome Biol. 11:R63.
    1. Kovacova Z., Tharp W. G., Liu D., Wei W., Xie H., Collins S., et al. (2016). Adipose tissue natriuretic peptide receptor expression is related to insulin sensitivity in obesity and diabetes. Obesity 24 820–828. 10.1002/oby.21418
    1. Lee J. E., Schmidt H., Lai B., Ge K. (2019). Transcriptional and Epigenomic Regulation of Adipogenesis. Mol. Cell. Biol. 39 e601–e618.
    1. Lee M. J., Wu Y., Fried S. K. A. (2012). modified protocol to maximize differentiation of human preadipocytes and improve metabolic phenotypes. Obesity 20 2334–2340. 10.1038/oby.2012.116
    1. Lim Y. C., Li J., Ni Y., Liang Q., Zhang J., Yeo G. S. H., et al. (2017). complex association between DNA methylation and gene expression in human placenta at first and third trimesters. PLoS One 12:e0181155. 10.1371/journal.pone.0181155
    1. Lotta L. A., Gulati P., Day F. R., Payne F., Ongen H., van de Bunt M., et al. (2017). Integrative genomic analysis implicates limited peripheral adipose storage capacity in the pathogenesis of human insulin resistance. Nat. Genet. 49 17–26. 10.1038/ng.3714
    1. Magnus M. C., Olsen S. F., Granstrom C., Lund-Blix N. A., Svensson J., Johannesen J., et al. (2018). Paternal and maternal obesity but not gestational weight gain is associated with type 1 diabetes. Int. J. Epidemiol. 47 417–426. 10.1093/ije/dyx266
    1. Manolopoulos K. N., Karpe F., Frayn K. N. (2010). Gluteofemoral body fat as a determinant of metabolic health. Int. J. Obes. 34 949–959. 10.1038/ijo.2009.286
    1. McGuire M. H., Herbrich S. M., Dasari S. K., Wu S. Y., Wang Y., Rupaimoole R., et al. (2019). Pan-cancer genomic analysis links 3’UTR DNA methylation with increased gene expression in T cells. EBioMedicine 43 127–137. 10.1016/j.ebiom.2019.04.045
    1. Noble A. J., Pearson J. F., Boden J. M., Horwood L. J., Gemmell N. J., Kennedy M. A., et al. (2021). validation of Illumina EPIC array system with bisulfite-based amplicon sequencing. PeerJ 9:e10762. 10.7717/peerj.10762
    1. Nono Nankam P. A., Blüher M., Kehr S., Klöting N., Krohn K., Adams K., et al. (2020). Distinct abdominal and gluteal adipose tissue transcriptome signatures are altered by exercise training in African women with obesity. Sci. Rep. 10:10240.
    1. Passaro A., Miselli M. A., Sanz J. M., Dalla Nora E., Morieri M. L., Colonna R., et al. (2017). Gene expression regional differences in human subcutaneous adipose tissue. BMC Genomics 18:202. 10.1186/s12864-017-3564-2
    1. Pinnick K. E., Nicholson G., Manolopoulos K. N., McQuaid S. E., Valet P., Frayn K. N., et al. (2014). Distinct developmental profile of lower-body adipose tissue defines resistance against obesity-associated metabolic complications. Diabetes 63 3785–3797. 10.2337/db14-0385
    1. Russo G., Landi R., Pezone A., Morano A., Zuchegna C., Romano A., et al. (2016). DNA damage and Repair Modify DNA methylation and Chromatin Domain of the Targeted Locus: Mechanism of allele methylation polymorphism. Sci. Rep. 6:33222.
    1. Ryu M. J., Kim S. J., Choi M. J., Kim Y. K., Lee M. H., Lee S. E., et al. (2013). Mitochondrial oxidative phosphorylation reserve is required for hormone- and PPARγ agonist-induced adipogenesis. Mol. Cells 35 134–141. 10.1007/s10059-012-2257-1
    1. Stephens N. A., Brouwers B., Eroshkin A. M., Yi F., Cornnell H. H., Meyer C., et al. (2018). Exercise Response Variations in Skeletal Muscle PCr Recovery Rate and Insulin Sensitivity Relate to Muscle Epigenomic Profiles in Individuals With Type 2 Diabetes. Diabet. Care 41 2245–2254. 10.2337/dc18-0296
    1. Tang Q. Q., Otto T. C., Lane M. D. (2003). Mitotic clonal expansion: a synchronous process required for adipogenesis. Proc. Natl. Acad. Sci. U S A. 100 44–49. 10.1073/pnas.0137044100
    1. Trivanović D., Drvenica I., Kukolj T., Obradović H., Okić Djordjević I., Mojsilović S., et al. (2018). Adipoinductive effect of extracellular matrix involves cytoskeleton changes and SIRT1 activity in adipose tissue stem/stromal cells. Artific. Cells Nanomed. Biotechnol. 46 S370–S382.
    1. Vague J. (1956). [Treatment of android obesity and its complications]. Algerie Med. 60 91–99.
    1. Vogel M. A. A., Jocken J. W. E., Sell H., Hoebers N., Essers Y., Rouschop K. M. A., et al. (2018). Differences in Upper and Lower Body Adipose Tissue Oxygen Tension Contribute to the Adipose Tissue Phenotype in Humans. J. Clin. Endocrinol. Metabol. 103 3688–3697. 10.1210/jc.2018-00547
    1. Yang X., Wu R., Shan W., Yu L., Xue B., Shi H. D. N. A. (2016). Methylation Biphasically Regulates 3T3-L1 Preadipocyte Differentiation. Mol. Endocrinol. 30 677–687. 10.1210/me.2015-1135

Source: PubMed

3
Tilaa