Cellular Senescence in the Kidney

Marie-Helena Docherty, Eoin D O'Sullivan, Joseph V Bonventre, David A Ferenbach, Marie-Helena Docherty, Eoin D O'Sullivan, Joseph V Bonventre, David A Ferenbach

Abstract

Senescent cells have undergone permanent growth arrest, adopt an altered secretory phenotype, and accumulate in the kidney and other organs with ageing and injury. Senescence has diverse physiologic roles and experimental studies support its importance in nephrogenesis, successful tissue repair, and in opposing malignant transformation. However, recent murine studies have shown that depletion of chronically senescent cells extends healthy lifespan and delays age-associated disease-implicating senescence and the senescence-associated secretory phenotype as drivers of organ dysfunction. Great interest is therefore focused on the manipulation of senescence as a novel therapeutic target in kidney disease. In this review, we examine current knowledge and areas of ongoing uncertainty regarding senescence in the human kidney and experimental models. We summarize evidence supporting the role of senescence in normal kidney development and homeostasis but also senescence-induced maladaptive repair, renal fibrosis, and transplant failure. Recent studies using senescent cell manipulation and depletion as novel therapies to treat renal disease are discussed, and we explore unanswered questions for future research.

Keywords: acute renal failure; chronic kidney disease; fibrosis; kidney development; progression of renal failure.

Copyright © 2019 by the American Society of Nephrology.

Figures

Figure 1.
Figure 1.
Pathways to cellular senescence in eukaryotic cells. Multiple discrete cellular insults act via distinct signaling mechanisms to induce cell-cycle arrest in the kidney at either the G1/S (via inhibition of cdk2 and/or cdk4/6) or G2/M checkpoints (via Chk1/2 activation or cdc2/25c inhibition). Inactivation of oncogenes and spindle/epigenetic/nucleolar stress trigger activation of the cyclin-dependent kinase inhibitor p16ink4a. Telomere shortening, DNA damage, mitogen or oncogene activation, and hypoxia/reoxygenation also result in G1/S cell-cycle arrest, via a pathway dependent on p53 and p21cip1 activation. In contrast to this, developmental senescence appears to induce p21cip1 by pathways mediated by TGFβ/PI3K and independent of p53. ATM/ATR/ARF, Ataxia–Telangiectasia Mutated/Ataxia Telangiectasia and Rad3-related protein/p14 Alternate Reading Frame (human).
Figure 2.
Figure 2.
Renal disease increases renal expression of the cyclin-dependent kinase inhibitor p16ink4a. (A) Normal kidney biopsy specimen from a 42-year-old living related donor with p16ink4a staining in some tubular epithelial cell nuclei. (B) Membranous nephropathy from a 64-year-old man demonstrating cytoplasmic and nuclear p16ink4a in tubular epithelium and interstitial nuclei (arrows). (C) FSGS biopsy specimen from a 48-year-old man, showing increased p16ink4a staining in some tubules and interstitial cell nuclei (arrows). (D) Grade 5 IgA nephropathy, with p16ink4a staining demonstrating increased cytoplasmic and nuclear staining. (E) Implantation biopsy specimen from a 45-year-old donor with expected low levels of p16ink4a-positive staining. (F) Repeat biopsy specimen of the same kidney 7 years post-transplantation demonstrating intense nuclear and cytoplasmic staining (note that the anti-p16ink4a [F12] antibody from Santa Cruz Biotechnology used in all of these images is no longer available).
Figure 3.
Figure 3.
The potential tissue effects of pathologic growth arrest in maladaptive repair. This flow chart illustrates the putative steps required for repair after AKI. In the case of adaptive repair, acute senescence is induced but then cleared in a tightly controlled manner, with normal tissue structure being restored by proliferation of resident cells. In maladaptive repair, it is recognized that there is an accumulation of both tissue fibrosis and secondary senescent cells. Recent studies indicate that pharmacologic/genetic targeting of senescent cells may protect against progressive fibrosis in murine models of aging.,
Figure 4.
Figure 4.
Current and potential future interventions to target growth-arrested cells in the kidney in vivo. Multiple points in the generation of senescent cells are under investigation as potential windows to alter their accumulation and/or pathogenic effects. Interventions are being trialed to prevent senescent cell formation (e.g., weight loss, exercise), promote senescent cell apoptosis (e.g., FOXO4-DRI, ABT-263), inhibit SASP release (e.g., sirolimus, metformin), or utilize the metabolic activity of senescent cells to activate compounds (e.g., galacto-oligosaccharide–conjugated drugs). shRNA, short hairpin RNA.
Figure 5.
Figure 5.
Unanswered questions about the roles and significance of senescent cells in the kidney. Several key questions regarding the role of the senescent cells in the kidney require ongoing research, with the goal of (1) quantifying senescent cell load noninvasively, (2) understanding the need for senescent cells in renal repair after injury, (3) determining the factors preventing senescent cell clearance from the kidney, and (4) testing the efficacy of senescence-modifying interventions in man.

References

    1. Centers for Disease Control and Prevention: The state of aging and health in America, 2013. Atlanta, GA: Centers for Disease Control and Prevention, US Dept of Health and Human Services; 2013.
    1. O’Sullivan ED, Hughes J, Ferenbach DA: Renal aging: Causes and consequences. J Am Soc Nephrol 28: 407–420, 2017
    1. Ferenbach DA, Bonventre JV: Mechanisms of maladaptive repair after AKI leading to accelerated kidney ageing and CKD. Nat Rev Nephrol 11: 264–276, 2015
    1. Hayflick L, Moorhead PS: The serial cultivation of human diploid cell strains. Exp Cell Res 25: 585–621, 1961
    1. Simons MJ: Questioning causal involvement of telomeres in aging. Ageing Res Rev 24[Pt B]: 191–196, 2015
    1. Melk A: Senescence of renal cells: Molecular basis and clinical implications. Nephrol Dial Transplant 18: 2474–2478, 2003
    1. Halloran PF, Melk A, Barth C: Rethinking chronic allograft nephropathy: The concept of accelerated senescence. J Am Soc Nephrol 10: 167–181, 1999
    1. Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, et al. : Senolytics improve physical function and increase lifespan in old age. Nat Med 24: 1246–1256, 2018
    1. Baker DJ, Perez-Terzic C, Jin F, Pitel KS, Niederländer NJ, Jeganathan K, et al. : Opposing roles for p16Ink4a and p19Arf in senescence and ageing caused by BubR1 insufficiency. Nat Cell Biol 10: 825–836, 2008
    1. Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, et al. : Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479: 232–236, 2011
    1. Campisi J: Aging, cellular senescence, and cancer. Annu Rev Physiol 75: 685–705, 2013
    1. Childs BG, Durik M, Baker DJ, van Deursen JM: Cellular senescence in aging and age-related disease: From mechanisms to therapy. Nat Med 21: 1424–1435, 2015
    1. Muñoz-Espín D, Cañamero M, Maraver A, Gómez-López G, Contreras J, Murillo-Cuesta S, et al. : Programmed cell senescence during mammalian embryonic development. Cell 155: 1104–1118, 2013
    1. van Deursen JM: The role of senescent cells in ageing. Nature 509: 439–446, 2014
    1. d’Adda di Fagagna F, Reaper PM, Clay-Farrace L, Fiegler H, Carr P, Von Zglinicki T, et al. : A DNA damage checkpoint response in telomere-initiated senescence. Nature 426: 194–198, 2003
    1. Tchkonia T, Zhu Y, van Deursen J, Campisi J, Kirkland JL: Cellular senescence and the senescent secretory phenotype: Therapeutic opportunities. J Clin Invest 123: 966–972, 2013
    1. Campisi J, d’Adda di Fagagna F: Cellular senescence: When bad things happen to good cells. Nat Rev Mol Cell Biol 8: 729–740, 2007
    1. Megyesi J, Andrade L, Vieira JM Jr, Safirstein RL, Price PM: Positive effect of the induction of p21WAF1/CIP1 on the course of ischemic acute renal failure. Kidney Int 60: 2164–2172, 2001
    1. Lee DH, Wolstein JM, Pudasaini B, Plotkin M: INK4a deletion results in improved kidney regeneration and decreased capillary rarefaction after ischemia-reperfusion injury. Am J Physiol Renal Physiol 302: F183–F191, 2012
    1. Yang L, Besschetnova TY, Brooks CR, Shah JV, Bonventre JV: Epithelial cell cycle arrest in G2/M mediates kidney fibrosis after injury. Nat Med 16: 535–543, p following 143, 2010.
    1. Luo Q, Guo H, Kuang P, Cui H, Deng H, Liu H, et al.: Sodium fluoride arrests renal G2/M phase cell-cycle progression by activating ATM-Chk2-P53/Cdc25C signaling pathway in mice. Cell Physiol Biochem 51: 2421–2433, 2018.
    1. Jenkins RH, Davies LC, Taylor PR, Akiyama H, Cumbes B, Beltrami C, et al. : miR-192 induces G2/M growth arrest in aristolochic acid nephropathy. Am J Pathol 184: 996–1009, 2014
    1. Serrano M, Blasco MA: Putting the stress on senescence. Curr Opin Cell Biol 13: 748–753, 2001
    1. Demaria M, Ohtani N, Youssef SA, Rodier F, Toussaint W, Mitchell JR, et al. : An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev Cell 31: 722–733, 2014
    1. Burd CE, Sorrentino JA, Clark KS, Darr DB, Krishnamurthy J, Deal AM, et al. : Monitoring tumorigenesis and senescence in vivo with a p16(INK4a)-luciferase model. Cell 152: 340–351, 2013
    1. Baker DJ, Childs BG, Durik M, Wijers ME, Sieben CJ, Zhong J, et al. : Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature 530: 184–189, 2016
    1. Coppé JP, Patil CK, Rodier F, Sun Y, Muñoz DP, Goldstein J, et al. : Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol 6: 2853–2868, 2008
    1. Parrinello S, Coppe JP, Krtolica A, Campisi J: Stromal-epithelial interactions in aging and cancer: Senescent fibroblasts alter epithelial cell differentiation. J Cell Sci 118: 485–496, 2005
    1. Acosta JC, O’Loghlen A, Banito A, Guijarro MV, Augert A, Raguz S, et al. : Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell 133: 1006–1018, 2008
    1. Binet R, Ythier D, Robles AI, Collado M, Larrieu D, Fonti C, et al. : WNT16B is a new marker of cellular senescence that regulates p53 activity and the phosphoinositide 3-kinase/AKT pathway. Cancer Res 69: 9183–9191, 2009
    1. Takahashi A, Loo TM, Okada R, Kamachi F, Watanabe Y, Wakita M, et al. : Downregulation of cytoplasmic DNases is implicated in cytoplasmic DNA accumulation and SASP in senescent cells. Nat Commun 9: 1249, 2018
    1. Kuilman T, Michaloglou C, Vredeveld LC, Douma S, van Doorn R, Desmet CJ, et al. : Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell 133: 1019–1031, 2008
    1. Freund A, Orjalo AV, Desprez PY, Campisi J: Inflammatory networks during cellular senescence: Causes and consequences. Trends Mol Med 16: 238–246, 2010
    1. Jun JI, Lau LF: The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat Cell Biol 12: 676–685, 2010
    1. Kang TW, Yevsa T, Woller N, Hoenicke L, Wuestefeld T, Dauch D, et al. : Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature 479: 547–551, 2011
    1. Salminen A, Kauppinen A, Kaarniranta K: Emerging role of NF-κB signaling in the induction of senescence-associated secretory phenotype (SASP). Cell Signal 24: 835–845, 2012
    1. Kuźnar-Kamińska B, Mikuła-Pietrasik J, Witucka A, Romaniuk A, Konieczna N, Rubiś B, et al. : Serum from patients with chronic obstructive pulmonary disease induces senescence-related phenotype in bronchial epithelial cells. Sci Rep 8: 12940, 2018
    1. Davaapil H, Brockes JP, Yun MH: Conserved and novel functions of programmed cellular senescence during vertebrate development. Development 144: 106–114, 2017
    1. Storer M, Mas A, Robert-Moreno A, Pecoraro M, Ortells MC, Di Giacomo V, et al. : Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell 155: 1119–1130, 2013
    1. Krizhanovsky V, Yon M, Dickins RA, Hearn S, Simon J, Miething C, et al. : Senescence of activated stellate cells limits liver fibrosis. Cell 134: 657–667, 2008
    1. Wolstein JM, Lee DH, Michaud J, Buot V, Stefanchik B, Plotkin MD: INK4a knockout mice exhibit increased fibrosis under normal conditions and in response to unilateral ureteral obstruction. Am J Physiol Renal Physiol 299: F1486–F1495, 2010
    1. Macher-Goeppinger S, Bermejo JL, Schirmacher P, Pahernik S, Hohenfellner M, Roth W: Senescence-associated protein p400 is a prognostic marker in renal cell carcinoma. Oncol Rep 30: 2245–2253, 2013
    1. Shen Y, Yu D, Qi P, Wang X, Guo X, Zhang A: Calcitriol induces cell senescence of kidney cancer through JMJD3 mediated histone demethylation. Oncotarget 8: 100187–100195, 2017
    1. Martín-Caballero J, Flores JM, García-Palencia P, Serrano M: Tumor susceptibility of p21(Waf1/Cip1)-deficient mice. Cancer Res 61: 6234–6238, 2001
    1. Kamijo T, Zindy F, Roussel MF, Quelle DE, Downing JR, Ashmun RA, et al. : Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19ARF. Cell 91: 649–659, 1997
    1. Melk A, Schmidt BM, Vongwiwatana A, Rayner DC, Halloran PF: Increased expression of senescence-associated cell cycle inhibitor p16INK4a in deteriorating renal transplants and diseased native kidney. Am J Transplant 5: 1375–1382, 2005
    1. Sis B, Tasanarong A, Khoshjou F, Dadras F, Solez K, Halloran PF: Accelerated expression of senescence associated cell cycle inhibitor p16INK4A in kidneys with glomerular disease. Kidney Int 71: 218–226, 2007
    1. Liu J, Yang JR, He YN, Cai GY, Zhang JG, Lin LR, et al. : Accelerated senescence of renal tubular epithelial cells is associated with disease progression of patients with immunoglobulin A (IgA) nephropathy. Transl Res 159: 454–463, 2012
    1. Jin H, Zhang Y, Ding Q, Wang SS, Rastogi P, Dai DF, et al. : Epithelial innate immunity mediates tubular cell senescence after kidney injury. JCI Insight 4, 2019
    1. Szeto CC, Poon PYK, Lai FMM, Chow KM, Szeto CYK, Li PKT: Chromosomal telomere shortening of kidney cells in IgA nephropathy by the measurement of DNA in urinary sediment. Clin Nephrol 64: 337–342, 2005
    1. Cippà PE, Sun B, Liu J, Chen L, Naesens M, McMahon AP: Transcriptional trajectories of human kidney injury progression. JCI Insight 3, 2018
    1. Verzola D, Gandolfo MT, Gaetani G, Ferraris A, Mangerini R, Ferrario F, et al. : Accelerated senescence in the kidneys of patients with type 2 diabetic nephropathy. Am J Physiol Renal Physiol 295: F1563–F1573, 2008
    1. Al-Douahji M, Brugarolas J, Brown PA, Stehman-Breen CO, Alpers CE, Shankland SJ: The cyclin kinase inhibitor p21WAF1/CIP1 is required for glomerular hypertrophy in experimental diabetic nephropathy. Kidney Int 56: 1691–1699, 1999
    1. Wolf G, Schanze A, Stahl RA, Shankland SJ, Amann K: p27(Kip1) Knockout mice are protected from diabetic nephropathy: Evidence for p27(Kip1) haplotype insufficiency. Kidney Int 68: 1583–1589, 2005
    1. Park JY, Schutzer WE, Lindsley JN, Bagby SP, Oyama TT, Anderson S, et al. : p21 is decreased in polycystic kidney disease and leads to increased epithelial cell cycle progression: Roscovitine augments p21 levels. BMC Nephrol 8: 12, 2007
    1. Park JY, Park SH, Weiss RH: Disparate effects of roscovitine on renal tubular epithelial cell apoptosis and senescence: Implications for autosomal dominant polycystic kidney disease. Am J Nephrol 29: 509–515, 2009
    1. Ferlicot S, Durrbach A, Bâ N, Desvaux D, Bedossa P, Paradis V: The role of replicative senescence in chronic allograft nephropathy. Hum Pathol 34: 924–928, 2003
    1. Melk A, Schmidt BM, Braun H, Vongwiwatana A, Urmson J, Zhu LF, et al. : Effects of donor age and cell senescence on kidney allograft survival. Am J Transplant 9: 114–123, 2009
    1. McGlynn LM, Stevenson K, Lamb K, Zino S, Brown M, Prina A, et al. : Cellular senescence in pretransplant renal biopsies predicts postoperative organ function. Aging Cell 8: 45–51, 2009
    1. Hoffmann U, Neudörfl C, Daemen K, Keil J, Stevanovic-Meyer M, Lehner F, et al. : NK cells of kidney transplant recipients display an activated phenotype that is influenced by immunosuppression and pathological staging. PLoS One 10: e0132484, 2015
    1. Sagiv A, Krizhanovsky V: Immunosurveillance of senescent cells: The bright side of the senescence program. Biogerontology 14: 617–628, 2013
    1. Chkhotua AB, Gabusi E, Altimari A, D’Errico A, Yakubovich M, Vienken J, et al. : Increased expression of p16(INK4a) and p27(Kip1) cyclin-dependent kinase inhibitor genes in aging human kidney and chronic allograft nephropathy. Am J Kidney Dis 41: 1303–1313, 2003
    1. Melk A, Halloran PF: Cell senescence and its implications for nephrology. J Am Soc Nephrol 12: 385–393, 2001
    1. Oberhuber R, Ge X, Tullius SG: Donor age-specific injury and immune responses. Am J Transplant 12: 38–42, 2012
    1. Lim WH, Clayton P, Wong G, Campbell SB, Cohney S, Russ GR, et al. : Outcomes of kidney transplantation from older living donors. Transplantation 95: 106–113, 2013
    1. Braun H, Schmidt BM, Raiss M, Baisantry A, Mircea-Constantin D, Wang S, et al. : Cellular senescence limits regenerative capacity and allograft survival. J Am Soc Nephrol 23: 1467–1473, 2012
    1. Brighton PJ, Maruyama Y, Fishwick K, Vrljicak P, Tewary S, Fujihara R, et al. : Clearance of senescent decidual cells by uterine natural killer cells in cycling human endometrium. ELife 6, 2017
    1. Egashira M, Hirota Y, Shimizu-Hirota R, Saito-Fujita T, Haraguchi H, Matsumoto L, et al. : F4/80+ macrophages contribute to clearance of senescent cells in the mouse postpartum uterus. Endocrinology 158: 2344–2353, 2017
    1. Jeon OH, Kim C, Laberge RM, Demaria M, Rathod S, Vasserot AP, et al. : Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat Med 23: 775–781, 2017
    1. Chang J, Wang Y, Shao L, Laberge RM, Demaria M, Campisi J, et al. : Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat Med 22: 78–83, 2016
    1. Baar MP, Brandt RMC, Putavet DA, Klein JDD, Derks KWJ, Bourgeois BRM, et al. : Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell 169: 132–147.e16, 2017
    1. Schafer MJ, White TA, Evans G, Tonne JM, Verzosa GC, Stout MB, et al. : Exercise prevents diet-induced cellular senescence in adipose tissue. Diabetes 65: 1606–1615, 2016
    1. Werner C, Fürster T, Widmann T, Pöss J, Roggia C, Hanhoun M, et al. : Physical exercise prevents cellular senescence in circulating leukocytes and in the vessel wall. Circulation 120: 2438–2447, 2009
    1. List EO, Jensen E, Kowalski J, Buchman M, Berryman DE, Kopchick JJ: Diet-induced weight loss is sufficient to reduce senescent cell number in white adipose tissue of weight-cycled mice. Nutr Healthy Aging 4: 95–99, 2016
    1. Chen P, Luo X, Che Z, Zhang W, Liu F, Hou D, et al.: Targeting of the C-Jun/BCL-XL/P21 axis accelerates the switch from senescence to apoptosis upon ROC1 knockdown in gastric cancer cells. Cell Physiol Biochem 48: 1123–1138, 2018.
    1. Zhu Y, Tchkonia T, Fuhrmann-Stroissnigg H, Dai HM, Ling YY, Stout MB, et al. : Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell 15: 428–435, 2016
    1. Rudin CM, Hann CL, Garon EB, Ribeiro de Oliveira M, Bonomi PD, Camidge DR, et al.: Phase II study of single-agent navitoclax (ABT-263) and biomarker correlates in patients with relapsed small cell lung cancer. Clin Cancer Res 18: 3163–3169, 2012.
    1. Gandhi L, Camidge DR, Ribeiro de Oliveira M, Bonomi P, Gandara D, Khaira D, et al. : Phase I study of Navitoclax (ABT-263), a novel Bcl-2 family inhibitor, in patients with small-cell lung cancer and other solid tumors. J Clin Oncol 29: 909–916, 2011
    1. Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, et al. : The Achilles’ heel of senescent cells: From transcriptome to senolytic drugs. Aging Cell 14: 644–658, 2015
    1. Tchkonia T, Kirkland JL: Aging, cell senescence, and chronic disease: Emerging therapeutic strategies. JAMA 320: 1319–1320, 2018
    1. Georgilis A, Klotz S, Hanley CJ, Herranz N, Weirich B, Morancho B, et al.: PTBP1-mediated alternative splicing regulates the inflammatory secretome and the pro-tumorigenic effects of senescent cells. Cancer Cell 34: 85–102.e9, 2018.
    1. Muñoz-Espín D, Rovira M, Galiana I, Giménez C, Lozano-Torres B, Paez-Ribes M, et al. : A versatile drug delivery system targeting senescent cells. EMBO Mol Med 10: 10, 2018
    1. Kennedy BK, Lamming DW: The mechanistic target of rapamycin: The grand conducTOR of metabolism and aging. Cell Metab 23: 990–1003, 2016
    1. Miwa S, Jow H, Baty K, Johnson A, Czapiewski R, Saretzki G, et al. : Low abundance of the matrix arm of complex I in mitochondria predicts longevity in mice. Nat Commun 5: 3837, 2014
    1. Piskovatska V, Stefanyshyn N, Storey KB, Vaiserman AM, Lushchak O: Metformin as a geroprotector: Experimental and clinical evidence. Biogerontology 20: 33–48, 2019
    1. Antonioli E, Torres N, Ferretti M, Piccinato CA, Sertie AL: Individual response to mTOR inhibition in delaying replicative senescence of mesenchymal stromal cells. PLoS One 14: e0204784, 2019
    1. Wang R, Yu Z, Sunchu B, Shoaf J, Dang I, Zhao S, et al. : Rapamycin inhibits the secretory phenotype of senescent cells by a Nrf2-independent mechanism. Aging Cell 16: 564–574, 2017
    1. Lesniewski LA, Seals DR, Walker AE, Henson GD, Blimline MW, Trott DW, et al. : Dietary rapamycin supplementation reverses age-related vascular dysfunction and oxidative stress, while modulating nutrient-sensing, cell cycle, and senescence pathways. Aging Cell 16: 17–26, 2017
    1. Gu Z, Tan W, Ji J, Feng G, Meng Y, Da Z, et al. : Rapamycin reverses the senescent phenotype and improves immunoregulation of mesenchymal stem cells from MRL/lpr mice and systemic lupus erythematosus patients through inhibition of the mTOR signaling pathway. Aging (Albany NY) 8: 1102–1114, 2016
    1. Tilstra JS, Robinson AR, Wang J, Gregg SQ, Clauson CL, Reay DP, et al. : NF-κB inhibition delays DNA damage-induced senescence and aging in mice. J Clin Invest 122: 2601–2612, 2012
    1. Xu M, Tchkonia T, Ding H, Ogrodnik M, Lubbers ER, Pirtskhalava T, et al. : JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc Natl Acad Sci USA 112: E6301–E6310, 2015
    1. Iwasa H, Han J, Ishikawa F: Mitogen-activated protein kinase p38 defines the common senescence-signalling pathway. Genes Cells 8: 131–144, 2003
    1. Chien Y, Scuoppo C, Wang X, Fang X, Balgley B, Bolden JE, et al. : Control of the senescence-associated secretory phenotype by NF-κB promotes senescence and enhances chemosensitivity. Genes Dev 25: 2125–2136, 2011
    1. Yang HC, Rossini M, Ma LJ, Zuo Y, Ma J, Fogo AB: Cells derived from young bone marrow alleviate renal aging. J Am Soc Nephrol 22: 2028–2036, 2011

Source: PubMed

3
Abonnieren