Age-related inflammation triggers skeletal stem/progenitor cell dysfunction

Significance As we age, our capacity for tissue repair and regeneration in response to injury declines. Accordingly, bone repair is delayed and impaired in older patients. At the cornerstone of bone healing is the skeletal stem/progenitor cell (SSPC), whose function and number diminishes with age. However, the mechanisms driving this decline remain unclear. Here, we identify age-associated inflammation (“inflamm-aging”) as the main culprit of SSPC dysfunction and provide support for a central role of NF-κB as a mediator of inflamm-aging. Our results show that modification of the inflammatory environment may be a translational approach to functionally rejuvenate the aged SSPC, thereby improving the regenerative capacity of the aged skeleton. Aging is associated with impaired tissue regeneration. Stem cell number and function have been identified as potential culprits. We first demonstrate a direct correlation between stem cell number and time to bone fracture union in a human patient cohort. We then devised an animal model recapitulating this age-associated decline in bone healing and identified increased cellular senescence caused by a systemic and local proinflammatory environment as the major contributor to the decline in skeletal stem/progenitor cell (SSPC) number and function. Decoupling age-associated systemic inflammation from chronological aging by using transgenic Nfkb1KO mice, we determined that the elevated inflammatory environment, and not chronological age, was responsible for the decrease in SSPC number and function. By using a pharmacological approach inhibiting NF-κB activation, we demonstrate a functional rejuvenation of aged SSPCs with decreased senescence, increased SSPC number, and increased osteogenic function. Unbiased, whole-genome RNA sequencing confirmed the reversal of the aging phenotype. Finally, in an ectopic model of bone healing, we demonstrate a functional restoration of regenerative potential in aged SSPCs. These data identify aging-associated inflammation as the cause of SSPC dysfunction and provide mechanistic insights into its reversal.

[1]  C. Lawless,et al.  A senescent cell bystander effect: senescence-induced senescence , 2012, Aging cell.

[2]  Susan J Wilson,et al.  Nuclear Factor-(cid:2) B1 (p50) Limits the Inflammatory and Fibrogenic Responses to Chronic Injury , 2022 .

[3]  J Denton,et al.  Adipocytic proportion of bone marrow is inversely related to bone formation in osteoporosis , 2002, Journal of clinical pathology.

[4]  C. Beck,et al.  Aging periosteal progenitor cells have reduced regenerative responsiveness to bone injury and to the anabolic actions of PTH 1-34 treatment. , 2014, Bone.

[5]  C. Franceschi,et al.  Inflamm‐aging: An Evolutionary Perspective on Immunosenescence , 2000 .

[6]  R. Gaynor,et al.  Therapeutic potential of inhibition of the NF-kappaB pathway in the treatment of inflammation and cancer. , 2001, The Journal of clinical investigation.

[7]  R. Gaynor,et al.  The anti-inflammatory agents aspirin and salicylate inhibit the activity of I(kappa)B kinase-beta. , 1998, Nature.

[8]  D. Baker,et al.  p21 both attenuates and drives senescence and aging in BubR1 progeroid mice. , 2013, Cell reports.

[9]  M. Serrano,et al.  Tumor suppressors and oncogenes in cellular senescence☆ , 2000, Experimental Gerontology.

[10]  D. Baltimore,et al.  Physiological functions for brain NF-kappaB. , 2005, Trends in neurosciences.

[11]  A. Wagers,et al.  Stem cell aging: mechanisms, regulators and therapeutic opportunities , 2014, Nature Medicine.

[12]  N. LeBrasseur,et al.  Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders , 2011, Nature.

[13]  A. Tosteson,et al.  Incidence and Economic Burden of Osteoporosis‐Related Fractures in the United States, 2005–2025 , 2007, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[14]  O. Kennedy,et al.  The Selective Serotonin Reuptake Inhibitor Fluoxetine Directly Inhibits Osteoblast Differentiation and Mineralization During Fracture Healing in Mice , 2017, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[15]  A. Scialdone,et al.  Adipocyte Accumulation in the Bone Marrow during Obesity and Aging Impairs Stem Cell-Based Hematopoietic and Bone Regeneration , 2017, Cell stem cell.

[16]  S. Morrison,et al.  Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. , 2014, Cell stem cell.

[17]  D. Sahoo,et al.  Identification and Specification of the Mouse Skeletal Stem Cell , 2015, Cell.

[18]  M. Almeida,et al.  DNA damage and senescence in osteoprogenitors expressing Osx1 may cause their decrease with age , 2017, Aging cell.

[19]  C Roskelley,et al.  A biomarker that identifies senescent human cells in culture and in aging skin in vivo. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[20]  G. Topulos,et al.  Nuclear factor-kappaB p50 limits inflammation and prevents lung injury during Escherichia coli pneumonia. , 2003, American journal of respiratory and critical care medicine.

[21]  N. Endo,et al.  Number of osteoprogenitor cells in human bone marrow markedly decreases after skeletal maturation , 1999, Journal of Bone and Mineral Metabolism.

[22]  M. Kassem,et al.  New factors controlling the balance between osteoblastogenesis and adipogenesis. , 2012, Bone.

[23]  C. Franceschi,et al.  Inflammaging and ‘Garb-aging’ , 2017, Trends in Endocrinology & Metabolism.

[24]  D. Baltimore,et al.  Physiological functions for brain NF-κB , 2005, Trends in Neurosciences.

[25]  R. Amann,et al.  Anti-inflammatory effects of aspirin and sodium salicylate. , 2002, European journal of pharmacology.

[26]  S. Gerondakis,et al.  Unravelling the complexities of the NF-κB signalling pathway using mouse knockout and transgenic models , 2006, Oncogene.

[27]  H. Ouyang,et al.  DNA damage drives accelerated bone aging via an NF‐κB–dependent mechanism , 2013, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[28]  T. Suuronen,et al.  Activation of innate immunity system during aging: NF-kB signaling is the molecular culprit of inflamm-aging , 2008, Ageing Research Reviews.

[29]  Laura C. Greaves,et al.  Chronic inflammation induces telomere dysfunction and accelerates ageing in mice , 2014, Nature Communications.

[30]  R. Gaynor,et al.  The anti-inflammatory agents aspirin and salicylate inhibit the activity of IκB kinase-β , 1998, Nature.

[31]  F. Luscinskas,et al.  Salicylates inhibit I kappa B-alpha phosphorylation, endothelial-leukocyte adhesion molecule expression, and neutrophil transmigration. , 1996, Journal of immunology.

[32]  Ankush Gosain,et al.  Aging and Wound Healing , 2004, World Journal of Surgery.

[33]  P. Wehrwein Stem cells: Repeat to fade , 2012, Nature.

[34]  Moshe Arditi,et al.  Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. , 2012, Immunity.

[35]  M. Pozzobon,et al.  Low-affinity Nerve Growth Factor Receptor (CD271) Heterogeneous Expression in Adult and Fetal Mesenchymal Stromal Cells , 2018, Scientific Reports.

[36]  G. Hannon,et al.  A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4 , 1993, Nature.

[37]  J. Campisi Cellular senescence: putting the paradoxes in perspective. , 2011, Current opinion in genetics & development.

[38]  H. Qian,et al.  Primary Mesenchymal Stem and Progenitor Cells from Bone Marrow Lack Expression of CD44 Protein* , 2012, The Journal of Biological Chemistry.

[39]  L. Kanz,et al.  Novel Markers for the Prospective Isolation of Human MSC , 2007, Annals of the New York Academy of Sciences.

[40]  Thomas A Rando,et al.  Aging, Stem Cells and Tissue Regeneration: Lessons from Muscle , 2005, Cell cycle.

[41]  J. Elisseeff,et al.  Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment , 2017, Nature Medicine.

[42]  N. Sharpless,et al.  Forging a signature of in vivo senescence , 2015, Nature Reviews Cancer.

[43]  J. Campisi Aging, cellular senescence, and cancer. , 2013, Annual review of physiology.

[44]  I. Vega-Naredo,et al.  Differential inflammatory responses in aging and disease: TNF-alpha and IL-6 as possible biomarkers. , 2010, Free radical biology & medicine.

[45]  B. Alman,et al.  Exposure to a youthful circulaton rejuvenates bone repair through modulation of β-catenin , 2015, Nature Communications.

[46]  M. V. D. van der Meulen,et al.  A method for isolating high quality RNA from mouse cortical and cancellous bone. , 2014, Bone.

[47]  J. Campisi,et al.  Cellular senescence: when bad things happen to good cells , 2007, Nature Reviews Molecular Cell Biology.

[48]  C. Colnot,et al.  Renal capsule transplantations to assay skeletal angiogenesis. , 2014, Methods in molecular biology.

[49]  R. Henschler,et al.  CD271 antigen defines a subset of multipotent stromal cells with immunosuppressive and lymphohematopoietic engraftment-promoting properties , 2010, Haematologica.

[50]  E. Schwarz,et al.  Cyclooxygenase-2 regulates mesenchymal cell differentiation into the osteoblast lineage and is critically involved in bone repair. , 2002, The Journal of clinical investigation.