Identification of Senescent Cells in the Bone Microenvironment

Oxford University Press (OUP) - Tập 31 Số 11 - Trang 1920-1929 - 2016
Joshua N. Farr1, Daniel G. Fraser1, Haitao Wang2, Katharina Jähn3, Mikołaj Ogrodnik1, Megan Weivoda1, Matthew T. Drake1, Tamar Tchkonia1, Nathan K. LeBrasseur1, James L. Kirkland1, Lynda F. Bonewald3, Robert J. Pignolo2, David G. Monroe1, Sundeep Khosla1
1Robert and Arlene Kogod Center on Aging and Endocrine Research Unit, Mayo Clinic College of Medicine, Rochester, MN, USA
2Departments of Orthopaedic Surgery and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
3Department of Oral Biology, University of Missouri-Kansas City, School of Dentistry, Kansas City, MO, USA

Tóm tắt

ABSTRACT Cellular senescence is a fundamental mechanism by which cells remain metabolically active yet cease dividing and undergo distinct phenotypic alterations, including upregulation of p16Ink4a, profound secretome changes, telomere shortening, and decondensation of pericentromeric satellite DNA. Because senescent cells accumulate in multiple tissues with aging, these cells and the dysfunctional factors they secrete, termed the senescence-associated secretory phenotype (SASP), are increasingly recognized as promising therapeutic targets to prevent age-related degenerative pathologies, including osteoporosis. However, the cell type(s) within the bone microenvironment that undergoes senescence with aging in vivo has remained poorly understood, largely because previous studies have focused on senescence in cultured cells. Thus in young (age 6 months) and old (age 24 months) mice, we measured senescence and SASP markers in vivo in highly enriched cell populations, all rapidly isolated from bone/marrow without in vitro culture. In both females and males, p16Ink4a expression by real-time quantitative polymerase chain reaction (rt-qPCR) was significantly higher with aging in B cells, T cells, myeloid cells, osteoblast progenitors, osteoblasts, and osteocytes. Further, in vivo quantification of senescence-associated distension of satellites (SADS), ie, large-scale unraveling of pericentromeric satellite DNA, revealed significantly more senescent osteocytes in old compared with young bone cortices (11% versus 2%, p < 0.001). In addition, primary osteocytes from old mice had sixfold more (p < 0.001) telomere dysfunction-induced foci (TIFs) than osteocytes from young mice. Corresponding with the age-associated accumulation of senescent osteocytes was significantly higher expression of multiple SASP markers in osteocytes from old versus young mice, several of which also showed dramatic age-associated upregulation in myeloid cells. These data show that with aging, a subset of cells of various lineages within the bone microenvironment become senescent, although senescent myeloid cells and senescent osteocytes predominantly develop the SASP. Given the critical roles of osteocytes in orchestrating bone remodeling, our findings suggest that senescent osteocytes and their SASP may contribute to age-related bone loss. © 2016 American Society for Bone and Mineral Research

Từ khóa


Tài liệu tham khảo

Goldman, 2013, Substantial health and economic returns from delayed aging may warrant a new focus for medical research, Health Aff (Millwood), 32, 1698, 10.1377/hlthaff.2013.0052

Tchkonia, 2013, Cellular senescence and the senescent secretory phenotype: therapeutic opportunities, J Clin Invest, 123, 966, 10.1172/JCI64098

van Deursen, 2014, The role of senescent cells in ageing, Nature, 509, 439, 10.1038/nature13193

Childs, 2015, Cellular senescence in aging and age-related disease: from mechanisms to therapy, Nat Med, 21, 1424, 10.1038/nm.4000

Swanson, 2013, Higher-order unfolding of satellite heterochromatin is a consistent and early event in cell senescence, J Cell Biol, 203, 929, 10.1083/jcb.201306073

von Zglinicki, 2005, Human cell senescence as a DNA damage response, Mech Ageing Dev, 126, 111, 10.1016/j.mad.2004.09.034

Ziegler, 2015, Mitochondrial effectors of cellular senescence: beyond the free radical theory of aging, Aging Cell, 14, 1, 10.1111/acel.12287

Wang, 1995, Senescent human fibroblasts resist programmed cell death, and failure to suppress bcl2 is involved, Cancer Res, 55, 2284

Coppé, 2008, Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor, PLoS Biol, 6, 2853, 10.1371/journal.pbio.0060301

Acosta, 2013, A complex secretory program orchestrated by the inflammasomes controls paracrine senescence, Nat Cell Biol, 15, 978, 10.1038/ncb2784

Coppé, 2010, The senescence-associated secretory phenotype: the dark side of tumor suppression, Annu Rev Pathol, 5, 99, 10.1146/annurev-pathol-121808-102144

Nelson, 2012, A senescent cell bystander effect: senescence-induced senescence, Aging Cell, 11, 345, 10.1111/j.1474-9726.2012.00795.x

Herbig, 2006, Cellular senescence in aging primates, Science, 311, 1257, 10.1126/science.1122446

Baker, 2011, Clearance of p16Ink4a-positive senescent cells delay aging-associated disorders, Nature, 479, 232, 10.1038/nature10600

Baker, 2016, Naturally occurring p16Ink4a-positive cells shorten healthy lifespan, Nature, 530, 184, 10.1038/nature16932

Zhu, 2015, The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs, Aging Cell, 14, 644, 10.1111/acel.12344

Kassem, 2011, Senescence-associated intrinsic mechanisms of osteoblast dysfunctions, Aging Cell, 10, 191, 10.1111/j.1474-9726.2011.00669.x

Marie, 2014, Bone cell senescence: mechanisms and perspectives, J Bone Miner Res, 29, 1311, 10.1002/jbmr.2190

Chen, 2013, DNA damage drives accelerated bone aging via an NF-kB-dependent mechanism, J Bone Miner Res, 28, 1214, 10.1002/jbmr.1851

Krishnamurthy, 2004, Ink4a/Arf expression is a biomarker of aging, J Clin Invest, 114, 1299, 10.1172/JCI22475

Burd, 2013, Monitoring tumorigenesis and senescence in vivo with a p16(INK4a) luciferase model, Cell, 152, 340, 10.1016/j.cell.2012.12.010

Waaijer, 2012, The number of p16INK4a positive cells in human skin reflects biological age, Aging Cell, 11, 722, 10.1111/j.1474-9726.2012.00837.x

Beauséjour, 2003, Reversal of human cellular senescence: roles of the p53 and p16 pathways, EMBO J, 22, 4212, 10.1093/emboj/cdg417

Baker, 2013, p21 both attenuates and drives senescence and aging in BubR1 progeroid mice, Cell Rep, 3, 1164, 10.1016/j.celrep.2013.03.028

Itoh, 2009, A novel purification method for multipotential skeletal stem cells, J Cell Biochem, 108, 368, 10.1002/jcb.22262

Syed, 2010, Effects of chronic estrogen treatment on modulating age-related bone loss in female mice, J Bone Miner Res, 25, 2438, 10.1002/jbmr.129

Zhou, 2014, Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow, Cell Stem Cell, 15, 154, 10.1016/j.stem.2014.06.008

Fujita, 2014, Isolation and characterization of human osteoblasts from needle biopsies without in vitro culture, Osteoporos Int, 25, 887, 10.1007/s00198-013-2529-9

Qing, 2012, Demonstration of osteocytic perilacunar/canalicular remodeling in mice during lactation, J Bone Miner Res, 27, 1018, 10.1002/jbmr.1567

Farr, 2015, Effects of age and estrogen on skeletal gene expression in humans as assessed by RNA sequencing, PLoS One, 10, e0138347, 10.1371/journal.pone.0138347

Subramanian, 2005, Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide experssion profiles, Proc Natl Acad Sci U S A, 102, 15545, 10.1073/pnas.0506580102

Efron, 2007, On testing the significance of sets of genes, Ann Appl Statist, 1, 107, 10.1214/07-AOAS101

Paic, 2009, Identification of differentially expressed genes between osteoblasts and osteocytes, Bone, 45, 682, 10.1016/j.bone.2009.06.010

Nioi, 2015, Transcriptional profiling of laser capture microdissected subpopulations of the osteoblast lineage provides insight into the early response to sclerostin antibody in rats, J Bone Miner Res, 30, 1457, 10.1002/jbmr.2482

Kang, 2015, The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4, Science, 349, aaa5612, 10.1126/science.aaa5612

Garcia-Prat, 2016, Autophagy maintains stemness by preventing senescence, Nature, 529, 37, 10.1038/nature16187

Komatsu, 2005, Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice, J Cell Biol, 169, 425, 10.1083/jcb.200412022

Kabeya, 2000, LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing, EMBO J, 19, 5720, 10.1093/emboj/19.21.5720

Stern, 2012, Isolation and culture of primary osteocytes from the long bones of skeletally mature and aged mice, Biotechniques, 52, 361, 10.2144/0000113876

Kenyon, 2007, The role of DNA damage repair in aging of adult stem cells, Nucleic Acids Res, 35, 7557, 10.1093/nar/gkm1064

Jurk, 2012, Postmitotic neurons develop a p21-dependent senescence-like phenotype driven by a DNA damage response, Aging Cell, 11, 996, 10.1111/j.1474-9726.2012.00870.x

Jurk, 2014, Chronic inflammation induces telomere dysfunction and accelerates ageing in mice, Nat Commun, 2, 4172, 10.1038/ncomms5172

Minamino, 2009, A crucial role for adipose tissue p53 in the regulation of insulin resistance, Nat Med, 15, 1082, 10.1038/nm.2014

Dallas, 2013, The osteocyte: an endocrine cell . . . and more, Endocr Rev, 34, 658, 10.1210/er.2012-1026

Salminen, 2012, Emerging role of NF-kappaB signaling in the induction of senescence-associated secretory phenotype (SASP), Cell Signal, 24, 835, 10.1016/j.cellsig.2011.12.006

Nakashima, 2011, Evidence for osteocyte regulation of bone homeostasis through RANKL expression, Nat Med, 17, 1231, 10.1038/nm.2452

Xiong, 2011, Matrix-embedded cells control osteoclast formation, Nat Med, 17, 1235, 10.1038/nm.2448

Lu, 2007, DMP1-targeted Cre expression in odontoblasts and osteocytes, J Dent Res, 86, 320, 10.1177/154405910708600404

Onal, 2013, Suppression of autophagy in osteocytes mimics skeletal aging, J Biol Chem, 288, 17432, 10.1074/jbc.M112.444190