Primary proximal tubule injury leads to epithelial cell cycle arrest, fibrosis, vascular rarefaction, and glomerulosclerosis

Joseph V Bonventre, Joseph V Bonventre

Abstract

Tubular injury has a major etiological role in fibrosis. For many years, this relationship has been dominated by the perception that epithelial cells are transformed into myofibroblasts that proliferate and generate fibrotic matrix-the so-called epithelial-to-mesenchymal transition. Here we focus on mechanisms by which injury to the tubule results in fibrosis because of paracrine mechanisms. Specific injury to the proximal tubule results in inflammation, reversible injury, and adaptive repair if the insult is mild, self-limited in time, and occurs in a background of a normal kidney. Repeated injury, in contrast, leads to maladaptive repair with sustained tubule injury, chronic inflammation, proliferation of interstitial myofibroblasts, vascular rarefaction, interstitial fibrosis, and glomerular sclerosis. During the maladaptive repair process after the renal insult, many tubular cells become arrested in the G2/M phase of the cell cycle. This results in activation of the DNA repair response with the resultant synthesis and secretion of pro-fibrotic factors. Pharmacologic interventions that enhance the movement through G2/M or facilitate apoptosis of cells that otherwise would be blocked in G2/M may reduce the development of fibrosis after kidney injury and reduce the progression of chronic kidney disease.

Keywords: G2/M arrest; acute kidney injury; apoptosis; chronic kidney disease; mitotic stress.

Figures

Figure 1
Figure 1
Adaptive and maladaptive repair after injury. After mild injury self-limited over time and to a kidney that has not sustained prior insults of significance, damaged proximal tubule cells are replaced by surviving proximal tubule cells that dedifferentiate, proliferate, and restore the normal epithelium. By contrast, if the injury is more severe, repeated, or to a kidney that is abnormal at baseline, then the cells that are stimulated to proliferate cannot complete the cell cycle efficiently and often are arrested in G2/M. Apoptosis of these cells may be blocked so that they continue to survive. They then turn on signaling pathways involving Jun N-terminal kinase (JNK), and activate pro-fibrotic gene transcription. This sets off a sequence of events resulting in vascular rarefaction, myofibroblast proliferation, and deposition of collagens and other matrix components in the interstitium.
Figure 2
Figure 2
In vivo cell cycle analysis in acute kidney injury (AKI). Proximal tubule cell cycle distribution (G1, S, and G2/M) of cells in the cell cycle over 42 days after moderate ischemia reperfusion injury (IRI; top left), unilateral ischemia/reperfusion (bottom left), and after administration of aristolochic acid (bottom right; n=3 mice of each time point in each group). AAN, aristolochic acid nephropathy. (Modified from Yang et al.)

References

    1. Kaissling B, Lehir M, Kriz W. Renal epithelial injury and fibrosis. Biochim Biophys Acta. 2013;1832:931–939.
    1. Venkatachalam MA, Griffin KA, Lan R, et al. Acute kidney injury: a springboard for progression in chronic kidney disease. Am J Physiol Renal Physiol. 2010;298:F1078–F1094.
    1. Grgic I, Campanholle G, Bijol V, et al. Targeted proximal tubule injury triggers interstitial fibrosis and glomerulosclerosis. Kidney Int. 2012;82:172–183.
    1. Buch T, Heppner FL, Tertilt C, et al. A Cre-inducible diphtheria toxin receptor mediates cell lineage ablation after toxin administration. Nat Methods. 2005;2:419–426.
    1. Chevalier RL, Forbes MS. Generation and evolution of atubular glomeruli in the progression of renal disorders. J Am Soc Nephrol. 2008;19:197–206.
    1. Beeuwkes R, III, Bonventre JV. Tubular organization and vascular-tubular relations in the dog kidney. Am J Physiol. 1975;229:695–713.
    1. Duffield JS, Park KM, Hsiao LL, et al. Restoration of tubular epithelial cells during repair of the postischemic kidney occurs independently of bone marrow-derived stem cells. J Clin Invest. 2005;115:1743–1755.
    1. Humphreys BD, Valerius MT, Kobayashi A, et al. Intrinsic epithelial cells repair the kidney after injury. Cell Stem Cell. 2008;2:284–291.
    1. Humphreys BD, Czerniak S, Dirocco DP, et al. Repair of injured proximal tubule does not involve specialized progenitors. Proc Natl Acad Sci USA. 2011;108:9226–9231.
    1. Humphreys BD, Lin SL, Kobayashi A, et al. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol. 2010;176:85–97.
    1. Beausejour CM, Krtolica A, Galimi F, et al. Reversal of human cellular senescence: roles of the p53 and p16 pathways. EMBO J. 2003;22:4212–4222.
    1. Rodier F, Campisi J, Bhaumik D. Two faces of p53: aging and tumor suppression. Nucleic Acids Res. 2007;35:7475–7484.
    1. Rodier F, Campisi J. Four faces of cellular senescence. J Cell Biol. 2011;192:547–556.
    1. Yang L, Besschetnova TY, Brooks CR, et al. Epithelial cell cycle arrest in G2/M mediates kidney fibrosis after injury. Nat Med. 2010;16:535–543.
    1. Tang J, Liu N, Tolbert E, et al. Sustained activation of EGFR triggers renal fibrogenesis after acute kidney injury. Am J Pathol. 2013;183:160–172.
    1. Cianciolo Cosentino C, Skrypnyk NI, Brilli LL, et al. Histone deacetylase inhibitor enhances recovery after AKI. J Am Soc Nephrol. 2013;24:943–953.
    1. Wu CF, Chiang WC, Lai CF, et al. Transforming growth factor beta-1 stimulates profibrotic epithelial signaling to activate pericyte-myofibroblast transition in obstructive kidney fibrosis. Am J Pathol. 2013;182:118–131.
    1. Jenkins RH, Davies LC, Taylor PR, et al. miR-192 induces G/M growth arrest in aristolochic acid nephropathy. Am J Pathol. 2014;184:996–1009.
    1. Qi W, Chen X, Poronnik P, et al. Transforming growth factor-beta/connective tissue growth factor axis in the kidney. Int J Biochem Cell Biol. 2008;40:9–13.
    1. Megyesi J, Udvarhelyi N, Safirstein RL, et al. The p53-independent activation of transcription of p21 WAF1/CIP1/SDI1 after acute renal failure. Am J Physiol. 1996;271:F1211–F1216.
    1. Megyesi J, Andrade L, Vieira JM, Jr., et al. Positive effect of the induction of p21WAF1/CIP1 on the course of ischemic acute renal failure. Kidney Int. 2001;60:2164–2172.
    1. Megyesi J, Price PM, Tamayo E, et al. The lack of a functional p21(WAF1/CIP1) gene ameliorates progression to chronic renal failure. Proc Natl Acad Sci USA. 1999;96:10830–10835.
    1. Molitoris BA, Dagher PC, Sandoval RM, et al. siRNA targeted to p53 attenuates ischemic and cisplatin-induced acute kidney injury. J Am Soc Nephrol. 2009;20:1754–1764.
    1. Park SK, Kang MJ, Kim W, et al. Renal tubule regeneration after ischemic injury is coupled to the up-regulation and activation of cyclins and cyclin dependent kinases. Kidney Int. 1997;52:706–714.
    1. Cimprich KA, Cortez D. ATR: an essential regulator of genome integrity. Nat Rev Mol Cell Biol. 2008;9:616–627.
    1. Zahedi K, Revelo MP, Barone S, et al. Stathmin-deficient mice develop fibrosis and show delayed recovery from ischemic-reperfusion injury. Am J Physiol Renal Physiol. 2006;290:F1559–F1567.
    1. Baker DJ, Wijshake T, Tchkonia T, et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011;479:232–236.
    1. Kashani K, Al-Khafaji A, Ardiles T, et al. Discovery and validation of cell cycle arrest biomarkers in human acute kidney injury. Crit Care. 2013;17:R25.
    1. Benatar T, Yang W, Amemiya Y, et al. IGFBP7 reduces breast tumor growth by induction of senescence and apoptosis pathways. Breast Cancer Res Treat. 2012;133:563–573.
    1. Seo DW, Li H, Guedez L, et al. TIMP-2 mediated inhibition of angiogenesis: an MMP-independent mechanism. Cell. 2003;114:171–180.
    1. Wang Z, Famulski K, Lee J, et al. TIMP2 and TIMP3 have divergent roles in early renal tubulointerstitial injury. Kidney Int. 2014;85:82–93.

Source: PubMed

3
購読する