Effects of Age and Estrogen on Skeletal Gene Expression in Humans as Assessed by RNA Sequencing

Joshua N Farr, Matthew M Roforth, Koji Fujita, Kristy M Nicks, Julie M Cunningham, Elizabeth J Atkinson, Terry M Therneau, Louise K McCready, James M Peterson, Matthew T Drake, David G Monroe, Sundeep Khosla, Joshua N Farr, Matthew M Roforth, Koji Fujita, Kristy M Nicks, Julie M Cunningham, Elizabeth J Atkinson, Terry M Therneau, Louise K McCready, James M Peterson, Matthew T Drake, David G Monroe, Sundeep Khosla

Abstract

Precise delineation of the specific genes and pathways altered with aging and estrogen (E) therapy may lead to new skeletal biomarkers and the development of novel bone therapeutics. Previous human bone studies, however, have been limited by only examining pre-specified genes and pathways. High-throughput RNA sequencing (RNAseq), on the other hand, offers an unbiased approach to examine the entire transcriptome. Here we present an RNAseq analysis of human bone samples, obtained from iliac crest needle biopsies, to yield the first in vivo interrogation of all genes and pathways that may be altered in bone with aging and E therapy in humans. 58 healthy women were studied, including 19 young women (mean age ± SD, 30.3 ± 5.4 years), 19 old women (73.1 ± 6.6 years), and 20 old women treated with 3 weeks of E therapy (70.5 ± 5.2 years). Using generally accepted criteria (false discovery rate [q] < 0.10), aging altered a total of 678 genes and 12 pathways, including a subset known to regulate bone metabolism (e.g., Notch). Interestingly, the LEF1 transcription factor, which is a classical downstream target of the Wnt/β-catenin signaling pathway, was significantly downregulated in the bones from the old versus young women; consistent with this, LEF1 binding sites were significantly enriched in the promoter regions of the differentially expressed genes in the old versus young women, suggesting that aging was associated with alterations in Wnt signaling in bone. Further, of the 21 unique genes altered in bone by E therapy, the expression of INHBB (encoding for the inhibin, beta B polypeptide), which decreased with aging (by 0.6-fold), was restored to young adult levels in response to E therapy. In conclusion, our data demonstrate that aging alters a substantial portion of the skeletal transcriptome, whereas E therapy appears to have significant, albeit less wide-ranging effects. These data provide a valuable resource for the potential identification of novel biomarkers associated with age-related bone loss and also highlight potential pathways that could be targeted to treat osteoporosis.

Trial registration: ClinicalTrials.gov NCT02349113.

Conflict of interest statement

Competing Interests: This study was supported, in part, by an investigator-initiated grant from Merck. There are no other relevant declarations. This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Flow diagram for enrollment and…
Fig 1. Flow diagram for enrollment and allocation of the subjects in the E arm of the study.
Fig 2. (A) Notch pathway and (B)…
Fig 2. (A) Notch pathway and (B) Wnt/β-catenin pathway genes are altered with aging in human bone biopsies.
Genes in the Notch pathway and Wnt/β-catenin pathway are significantly altered in old relative to young women revealed by RNAseq based on pathway analysis using the Ingenuity Pathway Analysis software (see Statistical analyses). Values are presented as median fold changes (95% CIs) for old relative to young subjects. †p < 0.01; ‡p < 0.001.
Fig 3. Networks derived from age-related differentially…
Fig 3. Networks derived from age-related differentially expressed genes (DEGs) in bone.
(A) Top 10 scoring networks and associated network functions derived from the 678 DEGs altered (median count ≥ 10, q < 0.10) in the young versus old dataset determined using the Ingenuity Pathway Analysis software (see Statistical Analysis). (B) Network of cell morphology, hematological system development and function, and protein synthesis (score = 33); key molecules implicated in bone metabolism include LEF1 and CDKN1A (p21). (C) Network of cardiovascular system development and function, cellular development, and organismal development (score = 31); key molecules implicated in bone metabolism include Notch signaling components (NOTCH3, NOTCH4, HEY1, JAG2, and DLL4) and SEMA3A. Green indicates downregulated genes, and red indicates upregulated genes.
Fig 4. Effects of age and estrogen…
Fig 4. Effects of age and estrogen (E) on serum sclerostin levels.
Serum sclerostin levels in the young versus old subjects (A) and in the old versus E-treated subjects (B) by the Biomedica and Meso Scale Discovery (MSD) assays. Data are mean ± SEM; note the difference in scales for the two sclerostin assays. *p < 0.05; †p < 0.01.
Fig 5. Effects of age and estrogen…
Fig 5. Effects of age and estrogen (E) on gene expression of INHBA and INHBB.
Bone INHBB and INHBA gene expression levels by RNAseq in the old relative to young women (A) and in the E-treated relative to untreated old women (B). Data are shown as median fold changes (95% CIs). †p < 0.01; ‡p < 0.001.

References

    1. Khosla S (2013) Pathogenesis of age-related bone loss in humans. J Gerontol A Biol Sci Med Sci 68: 1226–1235. 10.1093/gerona/gls163
    1. Lips P, Courpron P, Meunier PJ (1978) Mean wall thickness of trabecular bone packets in the human iliac crest: changes with age. Calcif Tissue Res 26: 13–17.
    1. Fatayerji D, Eastell R (1999) Age-related changes in bone turnover in men. J Bone Miner Res 14: 1203–1210.
    1. Khosla S, Melton LJ 3rd, Atkinson EJ, O'Fallon WM, Klee GG, Riggs BL (1998) Relationship of serum sex steroid levels and bone turnover markers with bone mineral density in men and women: A key role for bioavailable estrogen. J Clin Endocrinol Metab 83: 2266–2274.
    1. Riggs BL, Khosla S, Melton LJ 3rd (2002) Sex steroids and the construction and conservation of the adult skeleton. Endocr Rev 23: 279–302.
    1. Roforth MM, Fujita K, McGregor UI, Kirmani S, McCready LK, Peterson JM, et al. (2014) Effects of age on bone mRNA levels of sclerostin and other genes relevant to bone metabolism in humans. Bone 59: 1–6. 10.1016/j.bone.2013.10.019
    1. Fujita K, Roforth MM, Demaray S, McGregor U, Kirmani S, McCready LK, et al. (2014) Effects of estrogen on bone mRNA levels of sclerostin and other genes relevant to bone metabolism in postmenopausal women. J Clin Endocrinol Metab 99: E81–88. 10.1210/jc.2013-3249
    1. Fujita K, Roforth MM, Atkinson EJ, Peterson JM, Drake MT, McCready LK, et al. (2014) Isolation and characterization of human osteoblasts from needle biopsies without in vitro culture. Osteoporos Int 25: 887–895. 10.1007/s00198-013-2529-9
    1. Radonic A, Thulke S, Mackay IM, Landt O, Siegert W, Nitsche A (2004) Guideline to reference gene selection for quantitative real-time PCR. Biochem Biophys Res Commun 313: 856–862.
    1. Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, et al. (2002) Accurate normalization of real-time quantitative RT-PCR data by geometeric averaging of multiple internal control genes. Genome Biol 3: research00340031-0030-0034.0011.
    1. Zhao S, Fernald RD (2005) Comprehensive algorithm for quantitative real-time polymerase chain reaction. J Comput Biol 12: 1047–1064.
    1. Levey AS, Coresh J, Greene T, Stevens LA, Zhang YL, Hendriksen S, et al. (2006) Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. Ann Intern Med 145: 247–254.
    1. van Lierop AH, Witteveen JE, Hamdy NA, Papapoulos SE (2010) Patients with primary hyperparathyroidism have lower circulating sclerostin levels than euparathyroid controls. Eur J Endocrinol 163: 833–837. 10.1530/EJE-10-0699
    1. van Lierop AH, Hamdy NAT, Hamersma H, van Bezooijen RL, Power J, Loveridge N, et al. (2011) Patients with sclerosteosis and disease carriers: human models of the effect of sclerostin on bone turnover. J Bone Miner Res 26: 2804–2811. 10.1002/jbmr.474
    1. Trapnell C, Pachter L, Salzberg SL (2009) TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25: 1105–1111. 10.1093/bioinformatics/btp120
    1. Langmead B, Trapnell C, Pop M, Salzberg SL (2009) Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol 10: R25 10.1186/gb-2009-10-3-r25
    1. Wang L, Wang S, Li W (2012) RSeQC: quality control of RNA-seq experiments. Bioinformatics 28: 2184–2185. 10.1093/bioinformatics/bts356
    1. Hansen KD, Irizarry RA (2012) Removing technical variability in RNA-seq data using conditional quantile normalization. Biostatistics 13: 204–216. 10.1093/biostatistics/kxr054
    1. Robinson MD, McCarthy DJ, Smyth GK (2010) edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26: 139–140. 10.1093/bioinformatics/btp616
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. (2005) Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA 102: 15545–15550.
    1. Xie X, Lu J, Kulbokas EJ, Golub TR, Mootha V, Lindblad-Toh K, et al. (2005) Systematic discovery of regulatory motifs in human promoters and 3' UTRs by comparison of several mammals. Nature 434: 338–345.
    1. Giese K, Amsterdam A, Grosschedl R (1991) DNA-binding properties of the HMG domain of the lymphoid-specific transcriptional regulator LEF-1. Gene Dev 5: 2567–2578.
    1. Love JJ, Li X, Case DA, Giese K, Grosschedl R, Wright PE (1995) Structural basis for DNA bending by the architectural transcription factor LEF-1. Nature 376: 791–795.
    1. Baker DJ, Weaver RL, van Deursen JM (2013) p21 both attenuates and drives senescence and aging in BubR1 progeriod mice. Cell Rep 3: 1164–1174. 10.1016/j.celrep.2013.03.028
    1. Fukuda T, Takeda S, Xu R, Ochi H, Sunamura S, Sato T, et al. (2013) Sema3A regulates bone-mass accrual through sensory innervations. Nature 497: 490–493. 10.1038/nature12115
    1. Hayashi H, Abdollah S, Qiu Y, Cai J, Xu YY, Grinnell BW, et al. (1997) The MAD-related protein Smad7 associates with the TGFbeta receptor and functions as an antagonist of TGFbeta signaling. Cell 89: 1165–1173.
    1. Hamada Y, Kadokawa Y, Okabe M, Ikawa M, Coleman JR, Tsujimoto Y (1999) Mutation in ankyrin repeats of the mouse Notch2 gene induces early embryonic lethality. Development 126: 3415–3424.
    1. Gale NW, Dominguez MG, Noguera I, Pan L, Hughes V, Valenzuela DM, et al. (2004) Haploinsufficiency of delta-like 4 ligand results in embryonic lethality due to major defects in arterial and vascular development. Proc Natl Acad Sci U S A 101: 15949–15954.
    1. Xue Y, Gao X, Lindsell CE, Norton CR, Chang B, Hicks C, et al. (1999) Embryonic lethality and vascular defects in mice lacking the Notch ligand Jagged1. Hum Mol Genet 8: 723–730.
    1. Krebs LT, Xue Y, Norton CR, Shutter JR, Maguire M, Sundberg JP, et al. (2000) Notch signaling is essential for vascular morphogenesis in mice. Genes Dev 14: 1343–1352.
    1. Swiatek PJ, Lindsell CE, del Amo FF, Weinmaster G, Gridley T (1994) Notch1 is essential for postimplantation development in mice. Genes Dev 8: 707–719.
    1. Domenga V, Fardoux P, Lacombe P, Monet M, Maciazek J, Krebs LT, et al. (2004) Notch3 is required for arterial identity and maturation of vascular smooth muscle cells. Genes Dev 18: 2730–2735.
    1. Hrabe de Angelis M, McIntyre J 2nd, Gossler A (1997) Maintenance of somite borders in mice requires the Delta homologue DII1. Nature 386: 717–721.
    1. Jiang R, Lan Y, Chapman HD, Shawber C, Norton CR, Serreze DV, et al. (1998) Defects in limb, craniofacial, and thymic development in Jagged2 mutant mice. Genes Dev 12: 1046–1057.
    1. Zamurovic N, Cappellen D, Rohner D, Susa M (2004) Coordinated activation of notch, Wnt, and transforming growth factor-beta signaling pathways in bone morphogenic protein 2-induced osteogenesis. Notch target gene Hey1 inhibits mineralization and Runx2 transcriptional activity. J Biol Chem 279: 37704–37715.
    1. Garg V, Muth AN, Ransom JF, Schluterman MK, Barnes R, King IN, et al. (2005) Mutations in NOTCH1 cause aortic valve disease. Nature 437: 270–274.
    1. Hilton MJ, Tu X, Wu X, Bai S, Zhao H, Kobayashi T, et al. (2008) Notch signaling maintains bone marrow mesenchymal progenitors by suppressing osteoblast differentiation. Nat Med 14: 306–314. 10.1038/nm1716
    1. Salie R, Kneissel M, Vukevic M, Zamurovic N, Kramer I, Evans G, et al. (2010) Ubiquitous overexpression of Hey1 transcription factor leads to osteopenia and chondrocyte hypertrophy in bone. Bone 46: 680–694. 10.1016/j.bone.2009.10.022
    1. Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S, Reginato AM, et al. (2001) LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell 107: 513–523.
    1. Little RD, Carulli JP, Del Mastro RG, Dupuis J, Osborne M, Folz C, et al. (2002) A mutation in the LDL receptor-related protein 5 gene results in the autosomal dominant high-bone-mass trait. Am J Hum Genet 70: 11–19.
    1. Boyden LM, Mao J, Belsky J, Mitzner L, Farhi A, Mitnick MA, et al. (2002) High bone density due to a mutation in LDL-receptor-related protein 5. N Engl J Med 346: 1513–1521.
    1. Brunkow ME, Gardner JC, Van Ness J, Paeper BW, Kovacevich BR, Proll S, et al. (2001) Bone dysplasia sclerosteosis results from loss of the SOST gene product, a novel cystine knot-containing protein. Am J Hum Genet 68: 577–589.
    1. Balemans W, Patel N, Ebeling M, Van Hul E, Wuyts W, Lacza C, et al. (2002) Identification of a 52 kb deletion downstream of the SOST gene in patients with van Buchem disease. J Med Genet 39: 91–97.
    1. Loots GG, Kneissel M, Keller H, Baptist M, Chang J, Collette NM, et al. (2005) Genomic deletion of a long-range bone enhancer misregulates sclerostin in Van Buchem disease. Genome Res 15: 928–935.
    1. Baron R, Kneissel M (2013) WNT signaling in bone homeostasis and disease: from human mutations to treatments. Nat Med 19: 179–192. 10.1038/nm.3074
    1. Chim CS, Pang R, Fung TK, Choi CL, Liang R (2007) Epigenetic dysregulation of Wnt signaling pathway in multiple myeloma. Leukemia 21: 2527–2536.
    1. Khosla S, Westendorf JJ, Oursler MJ (2008) Building bone to reverse osteoporosis and repair fractures. J Clin Invest 118: 421–428. 10.1172/JCI33612
    1. Modder UI, Hoey KA, Amin S, McCready LK, Achenbach SJ, Riggs BL, et al. (2011) Relation of age, gender, and bone mass to circulating sclerostin levels in women and men. J Bone Miner Res 26: 373–379. 10.1002/jbmr.217
    1. Clarke BL, Drake MT (2013) Clinical utility of serum sclerostin measurements. BoneKEy Reports 2: 361 10.1038/bonekey.2013.95
    1. Durosier C, van Lierop A, Ferrari S, Chevalley T, Papapoulos S, Rizzoli R (2013) Association of circulating sclerostin with bone mineral mass, microstructure, and turnover biochemical markers in healthy elderly men and women. J Clin Endocrinol Metab 98: 3873–3883. 10.1210/jc.2013-2113
    1. Modder UIL, Clowes JA, Hoey K, Peterson JM, McCready L, Oursler MJ, et al. (2011) Regulation of circulating sclerostin levels by sex steroids in women and men. J Bone Miner Res 26: 27–34. 10.1002/jbmr.128
    1. Yi P, Park JS, Melton DA (2013) Betatrophin: a hormone that controls pancreatic beta cell proliferation. Cell 153: 747–758. 10.1016/j.cell.2013.04.008
    1. Tseng YH, Ke PY, Liao CJ, Wu SM, Chi HC, Tsai CY, et al. (2014) Chromosome 19 open reading frame 80 is upregulated by thyroid hormone and modulates autophagy and lipid metabolism. Autophagy 10: 20–31. 10.4161/auto.26126
    1. Carmon KS, Gong X, Lin Q, Thomas A, Liu Q (2011) R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/beta-catenin signaling. Proc Natl Acad Sci U S A 108: 11452–11457. 10.1073/pnas.1106083108
    1. Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spiegelman BM (1998) A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell 92: 829–839.
    1. St-Pierre J, Drori S, Uldry M, Silvaggi JM, Rhee J, Jager S, et al. (2006) Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell 127: 397–408.
    1. Liu C, Li S, Liu T, Borjigin J, Lin JD (2007) Transcriptional coactivator PGC-1alpha integrates the mammalian clock and energy metabolism. Nature 447: 477–481.
    1. Makanji Y, Zhu J, Mishra R, Holmquist C, Wong WP, Schwartz NB, et al. (2014) Inhibin at 90: from discovery to clinical application, a historical review. Endoc Rev 35: 747–794.
    1. Lotinun S, Pearsall RS, Horne WC, Baron R (2012) Activin receptor signaling: a potential therapeutic target for osteoporosis. Curr Mol Pharmacol 5: 195–204.
    1. Nicks KM, Perrien DS, Akel NS, Suva LJ, Gaddy D (2009) Regulation of osteoblastogenesis and osteoclastogenesis by the other reproductive hormones, Activin and Inhibin. Mol Cell Endocrinol 310: 11–20. 10.1016/j.mce.2009.07.001
    1. Pearsall RS, Canalis E, Cornwall-Brady M, Underwood KW, Haigis B, Ucran J, et al. (2008) A soluble activin type IIA receptor induces bone formation and improves skeletal integrity. Proc Natl Acad Sci USA 105: 7082–7087. 10.1073/pnas.0711263105
    1. Lotinun S, Pearsall RS, Davies MV, Marvell TH, Monnell TE, Ucran J, et al. (2010) A soluble activin receptor Type IIA fusion protein (ACE-011) increases bone mass via a dual anabolic-antiresorptive effect in Cynomolgus monkeys. Bone 46: 1082–1088. 10.1016/j.bone.2010.01.370
    1. Fajardo RJ, Manoharan RK, Pearsall RS, Davies MV, Marvell T, Monnell TE, et al. (2010) Treatment with a soluble receptor for activin improves bone mass and structure in the axial and appendicular skeleton of female cynomolgus macaques (Macaca fascicularis). Bone 46: 64–71. 10.1016/j.bone.2009.09.018
    1. Ruckle J, Jacobs M, Kramer W, Pearsall AE, Kumar R, Underwood KW, et al. (2009) Single-dose, randomized, double-blind, placebo-controlled study of ACE-011 (ActRIIA-IgG1) in postmenopausal women. J Bone Miner Res 24: 744–752. 10.1359/jbmr.081208
    1. Ayturk UM, Jacobsen CM, Christodoulou DC, Gorham J, Seidman JG, Seidman CE, et al. (2013) An RNA-seq protocol to identify mRNA expression changes in mouse diaphyseal bone: applications in mice with bone property altering Lrp5 mutations. J Bone Miner Res 28: 2081–2093. 10.1002/jbmr.1946
    1. McClung MR, Grauer A, Boonen S, Bolognese MA, Brown JP, Diez-Perez A, et al. (2014) Romosozumab in postmenopausal women with low bone mineral density. N Engl J Med 370: 412–420. 10.1056/NEJMoa1305224
    1. Tu X, Delgado-Calle J, Condon KW, Maycas M, Zhang H, Carlesso N, et al. (2015) Osteocytes mediate the anabolic actions of canonical Wnt/β-catenin signaling in bone. Proc Natl Acad Sci USA 112: E478–E486. 10.1073/pnas.1409857112

Source: PubMed

3
S'abonner