Autophagy plays a critical role in kidney tubule maintenance, aging and ischemia-reperfusion injury

Autophagy is responsible for the degradation of protein aggregates and damaged organelles. Several studies have reported increased autophagic activity in tubular cells after kidney injury. Here, we examine the role of tubular cell autophagy in vivo under both physiological conditions and stress using two different tubular-specific Atg5-knockout mouse models. While Atg5 deletion in distal tubule cells does not cause a significant alteration in kidney function, deleting Atg5 in both distal and proximal tubule cells results in impaired kidney function. Already under physiological conditions, Atg5-null tubule cells display a significant accumulation of p62 and oxidative stress markers. Strikingly, tubular cell Atg5-deficiency dramatically sensitizes the kidneys to ischemic injury, resulting in impaired kidney function, accumulation of damaged mitochondria as well as increased tubular cell apoptosis and proliferation, highlighting the critical role that autophagy plays in maintaining tubular cell integrity during stress conditions.

[1]  C. Edelstein,et al.  Apoptosis and Autophagy in Cold Preservation Ischemia , 2011, Transplantation.

[2]  T. Soga,et al.  Autophagy protects the proximal tubule from degeneration and acute ischemic injury. , 2011, Journal of the American Society of Nephrology : JASN.

[3]  B. Molitoris,et al.  Pathophysiology of ischemic acute kidney injury , 2011, Nature Reviews Nephrology.

[4]  A. Cuervo,et al.  Integration of clearance mechanisms: the proteasome and autophagy. , 2010, Cold Spring Harbor perspectives in biology.

[5]  M. Djavaheri-Mergny,et al.  Autophagosome maturation is impaired in Fabry disease , 2010, Autophagy.

[6]  D. Rubinsztein,et al.  Mechanisms of cross‐talk between the ubiquitin‐proteasome and autophagy‐lysosome systems , 2010, FEBS letters.

[7]  M. Komatsu,et al.  Physiological significance of selective degradation of p62 by autophagy , 2010, FEBS letters.

[8]  Ling Li,et al.  Autophagy is a component of epithelial cell fate in obstructive uropathy. , 2010, The American journal of pathology.

[9]  C. Cohen,et al.  Autophagy influences glomerular disease susceptibility and maintains podocyte homeostasis in aging mice. , 2010, The Journal of clinical investigation.

[10]  Z. Dong,et al.  Autophagy is a renoprotective mechanism during in vitro hypoxia and in vivo ischemia-reperfusion injury. , 2010, The American journal of pathology.

[11]  N. Mizushima,et al.  Methods in Mammalian Autophagy Research , 2010, Cell.

[12]  M. Sarwal,et al.  Mitochondrial autophagy promotes cellular injury in nephropathic cystinosis. , 2010, Journal of the American Society of Nephrology : JASN.

[13]  H. Kuwana,et al.  Cisplatin-induced macroautophagy occurs prior to apoptosis in proximal tubules in vivo , 2010, Clinical and Experimental Nephrology.

[14]  D. Metzger,et al.  Autophagy is required to maintain muscle mass. , 2009, Cell metabolism.

[15]  Ivan Dikic,et al.  NBR1 co-operates with p62 in selective autophagy of ubiquitinated targets , 2009, Autophagy.

[16]  Kun Wook Chung,et al.  Loss of autophagy diminishes pancreatic beta cell mass and function with resultant hyperglycemia. , 2008, Cell metabolism.

[17]  Masaaki Komatsu,et al.  Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet. , 2008, Cell metabolism.

[18]  Xiao-Ming Yin,et al.  Autophagy is cytoprotective during cisplatin injury of renal proximal tubular cells. , 2008, Kidney international.

[19]  Oliver Greiner,et al.  An efficient and versatile system for acute and chronic modulation of renal tubular function in transgenic mice , 2008, Nature Medicine.

[20]  Sudhir V. Shah,et al.  Autophagy is associated with apoptosis in cisplatin injury to renal tubular epithelial cells. , 2008, American journal of physiology. Renal physiology.

[21]  M. Koike,et al.  Participation of autophagy in renal ischemia/reperfusion injury. , 2008, Biochemical and biophysical research communications.

[22]  Daniel J. Klionsky,et al.  Autophagy fights disease through cellular self-digestion , 2008, Nature.

[23]  Masaaki Komatsu,et al.  Homeostatic Levels of p62 Control Cytoplasmic Inclusion Body Formation in Autophagy-Deficient Mice , 2007, Cell.

[24]  Masahiko Watanabe,et al.  Aberrant Membranes and Double-Membrane Structures Accumulate in the Axons of Atg5-Null Purkinje Cells before Neuronal Death , 2007, Autophagy.

[25]  L. Luo,et al.  A global double‐fluorescent Cre reporter mouse , 2007, Genesis.

[26]  G. Bjørkøy,et al.  p62/SQSTM1 Binds Directly to Atg8/LC3 to Facilitate Degradation of Ubiquitinated Protein Aggregates by Autophagy* , 2007, Journal of Biological Chemistry.

[27]  Yasushi Matsumura,et al.  The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress , 2007, Nature Medicine.

[28]  Masaaki Komatsu,et al.  Loss of autophagy in the central nervous system causes neurodegeneration in mice , 2006, Nature.

[29]  Hideyuki Okano,et al.  Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice , 2006, Nature.

[30]  Masaaki Komatsu,et al.  Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice , 2005, The Journal of cell biology.

[31]  Takeshi Tokuhisa,et al.  The role of autophagy during the early neonatal starvation period , 2004, Nature.

[32]  A. Schwarz,et al.  Long-term effects of acute ischemia and reperfusion injury. , 2004, Kidney international.

[33]  K. Mori,et al.  Neutrophil Gelatinase-Associated Lipocalin: A Novel Early Urinary Biomarker for Cisplatin Nephrotoxicity , 2004, American Journal of Nephrology.

[34]  M. Matsui,et al.  In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. , 2003, Molecular biology of the cell.

[35]  M. Mitsnefes,et al.  Identification of neutrophil gelatinase-associated lipocalin as a novel early urinary biomarker for ischemic renal injury. , 2003, Journal of the American Society of Nephrology : JASN.

[36]  P. Igarashi,et al.  Epithelial-specific Cre/lox recombination in the developing kidney and genitourinary tract. , 2002, Journal of the American Society of Nephrology : JASN.