2,958
Views
15
CrossRef citations to date
0
Altmetric
State Of The Art Reviews

Advance of autophagy in chronic kidney diseases

, &
Pages 306-313 | Received 07 Sep 2016, Accepted 15 Dec 2016, Published online: 13 Jan 2017

Abstract

Autophagy, a highly conserved mechanism for cell survival, emerges as an important pathway in many biological processes and diseases conditions. Studies of cultured renal cells, human kidney tissues and experimental animal models implicate that autophagy regulation is the critical aspects in chronic kidney diseases (CKD). Here, we summarize the current studies on the role of autophagy in CKD. Unveiling the precise regulation mechanism of autophagy in CKD is essential for developing potential prevention, diagnostic and therapeutic targets of these sticky clinical challenges.

1. Autophagy

Autophagy is a tightly precise system in which the endogenous aggregates cellular protein and damaged organelles are removed via the lysosomal pathway. Except for eliminating materials, autophagy also serves as a highly efficient recycling system that produces new components and energy for cellular renovation and balance of homeostasis. Depend on different delivering mechanism of cargo to the lysosome, autophagy is classified at least into three types: microautophagy, macroautophagy, and chaperone-mediated autophagy (CMA).

1.1. Macroautophagy

Evolutionarily-conserved macroautophagy activated in response to stressful condition is a lysosomal-mediated intracellular degradation process.Citation1,Citation2 The process starts with the formation of a structure on the endoplasmic reticulum (ER), called the omegasome. Then, it expands into a phagophore or isolation membrane to assemble into the precursor of the autophagosome. During phagophore elongation, cytoplasmic components are assembled within this structure. Next, it forms a closed double-membrane vesicle, called an autophagosome. Last, amphisomes fuses with acidic lysosomes, forming autolysosomes, where the entrapped contents are degraded.Citation3 This process is not only the fundamental in disposing of aged and aggregated proteins, but also is the known mechanism for ridding of damaged organelles under normal conditions as well as stress conditions.Citation4–8 In states of normal physiology, there is an active autophagic process working in all cell types of the kidney and is especially prominent in podocytes to protect cellular integrity against the stresses, which these cells encounter at the filtration barrier.Citation9 Meanwhile, autophagy is also participated in various pathological processes of renal diseases. What’s more, macroautophagy level declines with age, leading to cellular age-related waste accumulation, a cause of the progression of ageing process.Citation10

1.2. Microautophagy

Microautophagy is a nonselective type of autophagy. It involves engulfment of cytoplasmic cargo by direct invagination of the lysosomal membranes into autophagic tubes, which mediate both invagination and vesicle scission into the lumen. Although microautophagy is constitutive, it can be induced by starvation and rapamycin.Citation11 Microautophagic invagination and budding regulate lysosomal size by consuming superabundant membrane formed during macroautophagy.Citation12 However, the exact functions of microautophagy in mammalian cells are not yet fully understood.Citation13 It has been suggested that microautophagy is functions in the maintenance of organellar size, membrane composition, cell survival under nitrogen restriction, and the transition pathway from starvation-induced growth arrest to logarithmic growth.Citation14

1.3. Chaperone-mediated autophagy

CMA is a type of selective autophagy, so far only identified in mammalian cells. Different from other types of autophagy, its substrates, mainly cytosolic proteins, enter the lysosome.Citation15 Proteins degraded by CMA are identified one-by-one by a cytosolic chaperone utilizing heat shock chaperone of 70 kDa (hsc70), which recognizes the pentapeptide KFERQ motif upon misfolding of protein complexes. This hsc70-protein complex binds to a lysosomal receptor complex and translocate into the lysosome.Citation16–18 Thus, three events—(a) individual identification of single proteins, (b) unfolding before degradation, and (c) translocation inside the lysosomes—constitute the typical signature of CMA.

2. Types of autophagy involved in chronic kidney diseases (CKD)

The kidney is important for healthy living. It regulates the body fluids and blood pressure, excretes waste products and produces red blood cells.Citation19 Human kidney receives nearly 25% of cardiac output and consumes 7% of daily energy expenditure.Citation20 CKD, characterized by a glomerular filtration rate below 60 mL per minute for over 3 months,Citation21 is the end stage of various kidney diseases, leading to poor health outcomes and very high health care costs. Fibrosis is responsible for chronic progressive kidney failure, and the prevalence of CKD is increasing worldwide.Citation22,Citation23 Other mechanisms, such as oxidative stress,Citation24 inflammation,Citation25,Citation26 mitochondrial damage,Citation27,Citation28 ER stress,Citation29–31 and autophagy,Citation32,Citation33 are also involved in the progress of pathophysiology of CKD.

Autophagy is associated with the pathogenesis of a number of diseases. However, little is known about the relationship between autophagy and kidney diseases, especially autophagy and CKD. Until currently, emerging body of evidences suggest that autophagy plays a crucial role in chronic kidney diseases.

2.1. Macroautophagy and CKD

Diabetic nephropathy (DN) refers to CKD initiated by diabetes mellitus. There is a growing evidence that autophagy or mitophagy is altered in DN. An early study in diabetic rats detected an association between decreased autophagic activity and densities of autophagic vacuoles in proximal tubular epithelial cells.Citation34 Accordingly, a similar study suggested that regulation of blood glucose with insulin treatment led to the normalization of the density of autophagic vesicles, suggesting that this phenomenon was glucose-mediated.Citation35

2.2. Endoplasmic reticulum autophagy and CKD

Autophagy eliminates ER filled with unfolded proteins, which is called ER-phagy. Bernales et al. discovered and first named “endoplasmic reticulum autophagy (ER-phagy)” during their unfolded protein response (UPR) study.Citation36 This ER-phagy can maintain homeostasis of ER. ER-phagy seems to be cytoprotective against various folding diseases, including neurodegenerative diseases and diabetes.

ER is one of the multiple membrane sources for autophagosome biogenesis.Citation37 During prolonged ER stress, nonselective macro-autophagy can be induced,Citation38 and was suggested to serve as a backup for ER-associated degradation.Citation39,Citation40 Most notably, ER-phagy is also induced by ER stress,Citation36 which does not require the macroautophagy machinery components.Citation41 Thus, ER-phagy can maintain homeostasis of ER. ER-phagy seems to be cytoprotective against various folding diseases, including neurodegenerative diseases and diabetes.

2.3. CMA and CKD

In CMA, proteins containing KFERQ motif are selectively recognized by the cytosolic chaperone, hsc70. And in turn, the hsc70/substrate protein complex binds to the lysosomal membrane via lysosome-associated membrane protein 2A (LAMP-2A), leading to unfolding of the substrate and translocation inside the lysosome for subsequent degradation.Citation42

Up to now, the role of CMA in the kidney has not been systematically studied. Sooparb et al. found that if CMA was suppressed, PAX2 expression increased in cultured cells. PAX2 was an important factor in kidney development, indicating the possibility that CMA and PAX2 could play roles in epithelial cell growth.Citation43 In vivo, Sooparb et al. reported the abundance of proteins in diabetic kidneys containing the CMA KFERQ signal motif and individual KFERQ-containing proteins (e.g., PKM2, GAPDH, and PAX2) were more abundant in diabetic rats. LAMP2A and HSPA8 decrease in cortical lysosomes from diabetic vs. control rats, indicating the possibility that CMA might be increased in diabetic kidneys.Citation43

3. Autophagy in different cell types of kidney

3.1. Glomerular mesangial cells

Glomerular mesangial cells are specialized contractile pericytes, which locate in the centrilobular region called the mesangium and together with adjacent glomerular capillary endothelial cells and podocytes to regulate glomerular filtration. They are major targets of various glomerular diseases, such as IgA nephropathy and DN. In response to injury and progressive kidney diseases, mesangial cells proliferate and produce excessive extracellular matrix (ECM), leading to the development of glomerulosclerosis and kidney fibrosis, which is closely associated with the development and progression of CKD.

The role of autophagy in mesangial cells is not well recognized until recently. Wang et al. reported that autophagy was induced in mesangial cells following exposure to the heavy metal cadmium, an environmental toxin which accumulated in the kidneys and caused nephrotoxicity.Citation44

Under the condition of serum deprivation, autophagy was induced by transforming growth factor-β1(TGF-β1) in mesangial cells, and autophagy-enhanced survival by preventing mesangial cells from undergoing apoptosis.Citation45 The balance between synthesis and degradation of mesangial matrix is crucial progress in CKD. Studies unveiled a novel role of autophagy in negatively regulating the matrix production in mesangial cells by promoting the degradation of intracellular collagen I(Col-I), confirming that Col-I and aggregated, insoluble procollagen I undergo intracellular degradation through autophagy.Citation45 Consistent with this, Kim et al. also found increase of Col-I deposition in the kidney in UUO model of Beclin 1± mice and mesangial cells with reduced expression of Beclin 1. Then, their further studies unveiled that the activators of autophagy reduces protein levels of Col-I induced by TGF-β1 in mesangial cells.Citation46

3.2. Podocytes

Podocytes, highly differentiated epithelial cells lining the outer aspect of the glomerular basement membrane, is one of the important components of the filtration barrier. Podocyte injury is associated with marked proteinuria and decrease of glomerular filtration rate. Additionally, terminally differentiated podocytes are vulnerable to varity of injury, and the loss of podocytes is considered a key character of glomerular disease. Studies had confirmed that glomerular podocytes exhibited significant levels of autophagosomes even under basal conditions.Citation9,Citation47

Aldosterone (Aldo) is involved in the development and progression of CKD. Study found Aldo-induced podocyte apoptosis, autophagy, ROS generation, and downregulation of nephrin protein in a time-dependent manner.Citation27 Accordingly, Yuan at al detected Aldo-induced ER stress and podocyte autophagy both in vivo and in vitro, and blockade of ER stress significantly reduced aldosterone-induced podocyte injury.Citation48

Podocyte-specific deletion of the Atg5 gene resulted in proteinuria, loss of podocytes, and aging-related glomerulosclerosis indicating the critical importance of autophagy for glomerular maintenance.Citation9 Autophagy-specific Atg5 or Atg7conditional knockout mice exhibited enhanced vacuolization in podocytes and tubularcells and ultimately resulted in FSGS and organ failure.Citation33 A study comparing renal biopsies from patients with minimal change disease and patients with focal segmental glomerulosclerosis (FSGS), showed higher levels of Beclin 1-mediated autophagic activity in minimal change disease patients than those from FSGS patients, suggesting decreasing levels of autophagy might lead to progress to FSGS.Citation49

3.3. Tubular epithelial cells

Renal tubular epithelial cells (RTECs) are key targets in acute kidney injury (AKI) and CKD, and histological studies have indicated that renal function correlates better with tubular and interstitial changes than glomerular changes. Renal tubulointerstitial fibrosis is the hallmark of progressive CKD and end-stage renal diseases (ESRD) accompanied by tubular degeneration and atrophy.

Cyclosporine is a potent immunosuppressive drug widely used in preventing transplant rejection and treating autoimmune diseases, but its long-term use leads to the development of a chronic nephrotoxicity characterized by tubular atrophy, interstitial fibrosis, glomerulosclerosis, and impaired renal function. Cyclosporine induces ER stress, which is a potent stimulus for autophagy activation. Autophagy inhibition during cyclosporine treatment with beclin1 siRNA significantly increases tubular cell death.Citation50 Thus, induction of autophagy is protective against cyclosporine-induced tubular cell death.Citation50

TGF-β1 is known as the most potent profibrotic cytokine known. Studies in cultured human renal proximal tubular epithelial cells demonstrated that TGF-β1 induced upregulation of autophagy-related genes, Atg5, Atg7, and Beclin 1, and accumulation of autophagosomes in a time- and dose-dependent manner.Citation51 Furthermore, TGF-β1 activated autophagy through the generation of ROS, and promoted apoptosis in tubular cells.Citation51

Induction of autophagy was reported in a transgenic mouse model with tertracycline-controlled overexpression of TGF-β1 in RTECs.Citation52 The ureteral obstruction (UUO) model is a well-established model of progressive renal interstitial fibrosis. Increased autophagy with apoptosis and necrosis in tubules was also demonstrated in the renal fibrosis model, induced by UUO.Citation53,Citation54 What’s more, Yan Ding et al. found autophagy is induced primarily in RTECs of obstructed kidneys after UUO, and further study in UUO model of LC3 null (LC3−/−) mice and beclin 1± mice unveiled that deficiency of autophagic proteins lc3 and beclin1 results in increased collagen deposition in obstructed kidneys.Citation32

3.4. Endothelial cells

The glomerular endothelial cells are important components of the glomerular filtration unit, together with the podocytes and mesangial cells to their contribution to glomerular barrier function. Renal endothelial dysfunction, therefore, leads to kidney dysfunction and may contribute to progression of CKD and renal fibrosis. In this regard, the renal endothelium has garnered much interest, however, its role in the initiation and the progression of renal fibrosis remains not well understood. Akin to EMT in tubular epithelial cells, the concept that endothelial cells may also acquire functional and structural characteristics of mesenchymal cells, called endothelial–mesenchymal transition (EndoMT), following tissue injury.Citation55

EndMT plays a significant role in kidney fibrosis.Citation56 Studies showed that hyperphosphatemia induced endothelial autophagy, possibly through the inhibition of the Akt/mTOR signaling pathway, which might play a protective role against high Pi-induced apoptosis in cultured human microvascular endothelial cell. What’s more, rat model of CKD renal tissue revealed that hyperphosphatemia was associated with increased endothelial LC3 staining.Citation57

4. Autophagy pathways in CKD

Recent studies revealed dysregulated autophagy characterized by fibrosis in various tissues, including cardiac fibrosis, liver fibrosis, and idiopathic pulmonary fibrosis.Citation58 Here, we review several autophagy signaling pathways in CKD.

4.1. Phosphoinositide 3-kinase/protein kinase B (PI3K/akt) signal pathway

Phosphoinositide 3-kinase/protein kinase B(PI3K/Akt) has vital significance in autophagy.Citation59 DN is one of the major causes of ESRD, and the incidence of DN is increasing worldwide. Li et al demonstrated that high-glucose (HG) induced upregulation of (Pro)renin receptor played an important role in the reduction of autophagy and enhanced apoptosis in podocytes. And then, they further unveiled that the PI3K/Akt/mTOR/ULK1 signaling pathway mediated this effects.Citation60 Consistent with their study, Lu et al found the mesangial cells exposed to HG showed upregulated miRNA-21 expression, downregulated PTEN protein and mRNA expression, upregulated p85PI3K, pAkt, pmTOR, p62/SQSTMI, and Col-I expression and downregulated LC3II expression. LY294002, inhibitor of PI3K, inhibited HG-induced mesangial cell hypertrophy and proliferation, downregulated p85PI3K, pAkt, pmTOR, p62/SQSTMI, and Col-I expression and upregulated LC3II expression.Citation61

4.2. Rapamycin (mTOR) signal pathway

High evolutionarily conserved mechanistic target of rapamycin (mTOR) is a known pathway negatively regulates autophagy.Citation62,Citation63 Study suggested the induction of autophagy preceded tubular apoptosis and interstitial fibrosis and peaked after 3 days of UUO in the obstructed kidney of rats, and inhibition of autophagy with 3-MA enhanced tubular cell apoptosis and interstitial fibrosis.Citation64 The expression of Akt and mTOR was decreased over the first 3 days in the obstructed kidney after UUO, implying that Akt-mTOR signaling might regulate activation of autophagy in the UUO model.Citation64 These studies indicate that autophagy has a beneficial role in alleviating tubular damage and kidney fibrosis. Meanwhile, current study reported high Pi-induced autophagy activity in endothelial cells possibly through inhibiting the Akt/mTOR signaling pathway.Citation57

4.3. TGF-β signal pathway

In progressive kidney diseases, fibrosis represents the common pathway to ESRD, regardless of the initial causes. TGF-β1 is the most potent profibrogenic factor for the development and progress of renal fibrosis.Citation65 Studies showed that TGF-β1-activated autophagy via ROS, and autophagy promoted apoptosis in tubular cells.Citation51 Ding et al. revealed that TGF-β1 upregulated LC3 through the Akt pathway.Citation45 Kim et al. confirmed that TGF-β1-induced autophagy in mesangial cells by the TAK1-MKK3-p38 signaling pathway and concluded that the dual functions of TGF-β1, as both an inducer of Col-1 synthesis and an inducer of autophagy and Col-1 degradation underscore the pleiotropic function of TGF-β1.Citation46 Report also found that the induction of autophagy in RTECs protected against apoptosis and promoted TGF-β1 degradation, thereby reducing TGF-β1 secretion could prevent the development of renal interstitial fibrosis.Citation32

Induction of autophagy was reported in a transgenic mouse model with tertracycline-controlled overexpression of TGF-β1 in RTECs.Citation52 Tubular overexpression of TGF-β1-induced dedifferentiation and decomposition of tubular cells by autophagy and tubule interstitial fibrosis, but not epithelial-to-mesenchymal transition, and no sign of apoptosis, implying that autophagy might mediate tubular cell death.Citation52

5. The advantages and disadvantages of autophagy in CKD

The role of autophagy in progression of CKD is a controversial issue. On the one hand, autophagy may contribute to the degradation of Col-I and active TGF-β1 to inhibit kidney fibrosis. Wan-young Kim showed inhibition of autophagy by 3-MA enhanced tubular cell apoptosis and tubulointerstitial fibrosis in the obstructed kidney after UUO, implying that autophagy played a protective role in obstructed rat kidney.Citation64 Study also showed that kidneys from mice deficient in autophagic protein Beclin 1 or knockdown by specific siRNA in primary mouse mesangial cells exhibited profibrotic phenotype with increased Col-I deposition, whereas treatment with inducers of autophagy resulted in decreased Col-I induced by TGF-β1 in primary mouse mesangial cells.Citation46 According to these, LC3−/− mice resulted in increased collagen deposition and increased mature profibrotic factor TGF-β1 levels in obstructed kidneys and Beclin 1± mice also displayed increased collagen deposition in the obstructed kidneys after UUOCitation32. Genetic ablation of autophagy by proximal tubular epithelial cell-specific deletion of Atg5 also explored deposition of Col-I.Citation66

On the other hand, autophagy has been suggested to induce tubular atrophy and decomposition to promote fibrosis. Some recent studies had consistently demonstrated that UUO-induced autophagy in proximal tubules (14 days of duration) may act in concert with apoptosis to induce tubular atrophy and nephron loss that was associated with the progression of interstitial fibrosis.Citation53,Citation54,Citation67 Dong et al. found detected persistent autophagy in kidney proximal tubules of UUO mice. UUO-associated fibrosis—as indicated by the expression of ACTA2/a-smooth muscle actin, vimentin, fibronectin1, and collagen fibrils—was suppressed by pharmacological inhibitors of autophagy and also by kidney proximal tubule-specific knockout of autophagy-related 7 (PT-Atg7 KO) mice.Citation68

6. Prospective

CKD is a complex pathological process. It is not only a decline of glomerular filtration rate, but also is accompanied by a variety of metabolic changes: abnormal lipid metabolism, calcium and phosphorus metabolism disorder, water and salt metabolism unbalance, reduction of erythropoietin production. Furthermore, CKD is the key to progress to ESRD, which can lead to distant organs damage, such as heart, lung, liver, and so on. Various metabolic factors and different organs influence each other, leading the injury of CKD to moving forward, finally resulting in high mortality. Therefore, study of the occurrence and development mechanism of CKD has a very important clinical significance to prevent and the treatment of ESRD.

Autophagy involves in the whole process of the development of CKD. But past studies mainly focused on macroautophagy, while mitoautophagy, lipidautophagy, ER-phagy, and CMA are largely studied. Recent studies show that plasmid metabolism, ER stress and UPR play a very important role in the pathogenesis of CKD. Therefore, future studies should use transgene technology to investigate other types of autophagy and their precise mechanism in CKD.

The kidney contains numerous types of local cells. Recent studies mainly pay attention to podocytes, tubular epithelial cells, and mesangial cells, other cell types remains unexplored. However, the kidney is consisted by glomerulus, which contains millions of vascular endothelial cells. Damage of endothelial cells has much to do with renal function. Current researches has found that autophagy in endothelial cells is closely related cardiovascular diseases. Just because of this, as a major component of the glomerular, autophagy in endothelial cells could possibly plays a critical role in the CKD, and more attention should be paid to this issue.

Every coin has two sides. The pros and cons of autophagy attract growing attention recently. Autophagy can act as a mechanism of cell survival, but it can also lead to cell death in response to persistent stress. Depending on the cell or tissue types and pathological settings, autophagy can be profibrotic or antifibrotic in different conditions. Meanwhile, the etiology of CKD is very complex, as well as its pathological changes. Therefore, to find the common aspects of autophagy in occurrence and development of CKD, and different aspects of autophagy in CKD has vital significant clinical importance.

Taken together, these studies implicate that autophagy plays a vital role in the development and progression in CKD, and unveiling the precise regulation mechanism that promises a new preventive therapeutic target to mitigate the pathogenesis of CKD.

Disclosure statement

There are no conflicts of interest to declare. The work has not already been published and has not been submitted simultaneously to any other journal.

Additional information

Funding

This work was supported by National Natural Science Foundation of China [grant no. 81325004, 81270797, 81530023, 81570663].

References

  • Sureshbabu A, Ryter SW, Choi ME. Oxidative stress and autophagy: Crucial modulators of kidney injury. Redox Biol. 2015;4:208–214.
  • Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med. 2013;368:651–662.
  • Ravikumar B, Sarkar S, Davies JE, et al. Regulation of mammalian autophagy in physiology and pathophysiology. Physiol Rev. 2010;90:1383–1435.
  • Gustafsson AB, Gottlieb RA. Recycle or die: The role of autophagy in cardioprotection. J Mol Cell Cardiol. 2008;44:654–661.
  • Kundu M, Thompson CB. Autophagy: Basic principles and relevance to disease. Ann Rev Pathol. 2008;3:427–455.
  • Levine B, Kroemer G. Autophagy in aging, disease and death: The true identity of a cell death impostor. Cell Death Differ. 2009;16:1–2.
  • Mizushima N, Levine B, Cuervo AM, Klionsky DJ. Autophagy fights disease through cellular self-digestion. Nature. 2008;451:1069–1075.
  • Pallet N, Anglicheau D. Autophagy: A protective mechanism against nephrotoxicant-induced renal injury. Kidney Int. 2009;75:118–119.
  • Hartleben B, Godel M, Meyer-Schwesinger C, et al. Autophagy influences glomerular disease susceptibility and maintains podocyte homeostasis in aging mice. J Clin Invest. 2010;120:1084–1096.
  • Huber TB, Edelstein CL, Hartleben B, et al. Emerging role of autophagy in kidney function, diseases and aging. Autophagy. 2012;8:1009–1031.
  • Dubouloz F, Deloche O, Wanke V, Cameroni E, De Virgilio C. The TOR and EGO protein complexes orchestrate microautophagy in yeast. Mol Cell. 2005;19:15–26.
  • Todde V, Veenhuis M, van der Klei IJ. Autophagy: Principles and significance in health and disease. Biochim Biophys Acta. 2009;1792:3–13.
  • Li WW, Li J, Bao JK. Microautophagy: Lesser-known self-eating. Cell Mol Life Sci.2012;69:1125–1136.
  • Roberts P, Moshitch-Moshkovitz S, Kvam E, O'Toole E, Winey M, Goldfarb DS. Piecemeal microautophagy of nucleus in Saccharomyces cerevisiae. Mol Biol Cell. 2003;14:129–141.
  • Cuervo AM. Chaperone-mediated autophagy: Dice's 'wild' idea about lysosomal selectivity. Nat Rev Mol Cell Biol. 2011;12:535–541.
  • Arias E, Cuervo AM. Chaperone-mediated autophagy in protein quality control. Curr Opin Cell Biol. 2011;23:184–189.
  • Franch HA. Kidney growth during catabolic illness: What it does not destroy makes it grow stronger. J Ren Nutr. 2007;17:167–172.
  • Sooparb S, Price SR, Shaoguang J, Franch HA. Suppression of chaperone-mediated autophagy in the renal cortex during acute diabetes mellitus. Kidney Int. 2004;65:2135–2144.
  • Guyton AC. Blood pressure control-special role of the kidneys and body fluids. Science. 1991;252:1813–1816.
  • Macdonald JH, Fearn L, Jibani M, Marcora SM. Exertional fatigue in patients with CKD. Am J Kidney Dis. 2012; 60:930–939.
  • Bruck K, Jager KJ, Dounousi E, et al. Methodology used in studies reporting chronic kidney disease prevalence: a systematic literature review. Nephrology, Dialysis, Transplantation. 2015; 30:iv6–i16.
  • Coresh J, Astor BC, Greene T, Eknoyan G, Levey AS. Prevalence of chronic kidney disease and decreased kidney function in the adult US population: Third National Health and Nutrition Examination Survey. Am J Kidney Dis. 2003; 41:1–12.
  • Lameire N, Jager K, Van Biesen W, de Bacquer D, Vanholder R. Chronic kidney disease: A European perspective. Kidney Int Suppl. 2005;(99)S30–S38.
  • Modaresi A, Nafar M, Sahraei Z. Oxidative stress in chronic kidney disease. Iran J Kidney Dis. 2015;9:165–179.
  • Kovesdy CP. How can erythropoeitin-stimulating agent use be reduced in chronic dialysis patients?: Can reduction of inflammation improve ESA dose response? Semin Dial. 2013;26:540–542.
  • Kalantar-Zadeh K. Recent advances in understanding the malnutrition-inflammation-cachexia syndrome in chronic kidney disease patients: What is next? Semin Dial. 2005;18:365–369.
  • Bai M, Che R, Zhang Y, et al. Reactive oxygen species-initiated autophagy opposes aldosterone-induced podocyte injury. Am J Physiol Renal Physiol. 2016. [Epub ahead of print]. doi: 10.1152/ajprenal.00409.2015.
  • Xu J, Guo Z, Bai Y, et al. Single nucleotide polymorphisms in the D-loop region of mitochondrial DNA is associated with the kidney survival time in chronic kidney disease patients. Renal Failure. 2015;37:108–112.
  • Zhuang A, Forbes JM. Stress in the kidney is the road to pERdition: Is endoplasmic reticulum stress a pathogenic mediator of diabetic nephropathy? J Endocrinol. 2014;222:R97–111.
  • Cao Y, Hao Y, Li H, et al. Role of endoplasmic reticulum stress in apoptosis of differentiated mouse podocytes induced by high glucose. Int J Mol Med. 2014; 33:809–816.
  • Liu J, Huang K, Cai GY, et al. Receptor for advanced glycation end-products promotes premature senescence of proximal tubular epithelial cells via activation of endoplasmic reticulum stress-dependent p21 signaling. Cell Signal. 2014;26:110–121.
  • Ding Y, Kim S, Lee SY, Koo JK, Wang Z, Choi ME. Autophagy regulates TGF-beta expression and suppresses kidney fibrosis induced by unilateral ureteral obstruction. J Am Soc Nephrol. 2014; 25:2835–2846.
  • Kawakami T, Gomez IG, Ren S, et al. Deficient autophagy results in mitochondrial dysfunction and FSGS. J Am Soc Nephrol. 2015; 26:1040–1052.
  • Barbosa Junior Ade A, Zhou H, Hultenschmidt D, Totovic V, Jurilj N, Pfeifer U. Inhibition of cellular autophagy in proximal tubular cells of the kidney in streptozotocin-diabetic and uninephrectomized rats. Virchows Archiv B. 1992;61:359–366.
  • Han K, Lehringer-Polzin M, Zhou H, Pfeifer U. Cellular autophagy in proximal tubules of early diabetic rats following insulin treatment and islet transplantation. Virchows Archiv B. 1992;61:6): 367–73.
  • Bernales S, Schuck S, Walter P. ER-phagy: selective autophagy of the endoplasmic reticulum. Autophagy. 2007;3:285–287.
  • Lamb CA, Yoshimori T, Tooze SA. The autophagosome: Origins unknown, biogenesis complex. Nat Rev Mol Cell Biol. 2013;14:759–774.
  • Yorimitsu T, Klionsky DJ. Endoplasmic reticulum stress: A new pathway to induce autophagy. Autophagy. 2007;3:160–162.
  • Vembar SS, Brodsky JL. One step at a time: Endoplasmic reticulum-associated degradation. Nat Rev Mol Cell Biol. 2008;9:944–957.
  • Houck SA, Ren HY, Madden VJ, et al. Quality control autophagy degrades soluble ERAD-resistant conformers of the misfolded membrane protein GnRHR. Mol Cell. 2014;54:166–179.
  • Schuck S, Gallagher CM, Walter P. ER-phagy mediates selective degradation of endoplasmic reticulum independently of the core autophagy machinery. J Cell Sci. 2014;127:4078–4088.
  • Kaushik S, Cuervo AM. Chaperone-mediated autophagy: A unique way to enter the lysosome world. Trends Cell Biol. 2012;22:407–417.
  • Han K, Zhou H, Pfeifer U. Inhibition and restimulation by insulin of cellular autophagy in distal tubular cells of the kidney in early diabetic rats. Kidney Blood Press Res. 1997;20:258–263.
  • Wang SH, Shih YL, Ko WC, Wei YH, Shih CM. Cadmium-induced autophagy and apoptosis are mediated by a calcium signaling pathway. Cell Mol Life Sci. 2008;65:3640–3652.
  • Ding Y, Kim JK, Kim SI, et al. TGF-{beta}1 protects against mesangial cell apoptosis via induction of autophagy. J Biol Chem. 2010;285:37909–37919.
  • Kim SI, Na HJ, Ding Y, Wang Z, Lee SJ, Choi ME. Autophagy promotes intracellular degradation of type I collagen induced by transforming growth factor (TGF)-beta1. J Biol Chem. 2012; 287:11677–11688.
  • Mizushima N, Yamamoto A, Matsui M, Yoshimori T, Ohsumi Y. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol Biol Cell. 2004;15:1101–1111.
  • Yuan Y, Xu X, Zhao C, et al. The roles of oxidative stress, endoplasmic reticulum stress, and autophagy in aldosterone/mineralocorticoid receptor-induced podocyte injury. Lab Invest. 2015; 95:1374–1386.
  • Zeng C, Fan Y, Wu J, et al. Podocyte autophagic activity plays a protective role in renal injury and delays the progression of podocytopathies. J Pathol. 2014;234:203–213.
  • Pallet N, Bouvier N, Legendre C, et al. Autophagy protects renal tubular cells against cyclosporine toxicity. Autophagy. 2008; 4:783–791.
  • Xu Y, Yang S, Huang J, Ruan S, Zheng Z, Lin J. Tgf-beta1 induces autophagy and promotes apoptosis in renal tubular epithelial cells. Int J Mol Med. 2012; 29:781–790.
  • Koesters R, Kaissling B, Lehir M, et al. Tubular overexpression of transforming growth factor-beta1 induces autophagy and fibrosis but not mesenchymal transition of renal epithelial cells. Am J Pathol. 2010;177:632–643.
  • Li L, Zepeda-Orozco D, Black R, Lin F. Autophagy is a component of epithelial cell fate in obstructive uropathy. Am J Pathol. 2010;176:1767–1778.
  • Xu Y, Ruan S, Wu X, Chen H, Zheng K, Fu B. Autophagy and apoptosis in tubular cells following unilateral ureteral obstruction are associated with mitochondrial oxidative stress. Int J Mol Med. 2013; 31:628–636.
  • Guerrot D, Dussaule JC, Kavvadas P, Boffa JJ, Chadjichristos CE, Chatziantoniou C. Progression of renal fibrosis: The underestimated role of endothelial alterations. Fibrogenesis Tissue Repair 2012;5:S15.
  • Zeisberg EM, Potenta SE, Sugimoto H, Zeisberg M, Kalluri R. Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J Am Soc Nephrol.2008; 19:2282–2287.
  • Hsu YJ, Hsu SC, Huang SM, et al. Hyperphosphatemia induces protective autophagy in endothelial cells through the inhibition of Akt/mTOR signaling. J Vasc Surg. 2015;62:210–221 e2.
  • Del Principe D, Lista P, Malorni W, Giammarioli AM. Fibroblast autophagy in fibrotic disorders. J Pathol. 2013;229:208–220.
  • Zhang Q, Zhu H, Xu X, Li L, Tan H, Cai X. Inactivated Sendai virus induces apoptosis and autophagy via the PI3K/Akt/mTOR/p70S6K pathway in human non-small cell lung cancer cells. Biochem Biophys Res Commun. 2015;465:64–70.
  • Li C, Siragy HM. (Pro)renin receptor regulates autophagy and apoptosis in podocytes exposed to high glucose. Am J Physiol Endocrinol Metab. 2015;309:E302–E310.
  • Lu X, Fan Q, Xu L, et al. Ursolic acid attenuates diabetic mesangial cell injury through the up-regulation of autophagy via miRNA-21/PTEN/Akt/mTOR suppression. PLoS One. 2015;10:e0117400.
  • Dennis MD, Baum JI, Kimball SR, Jefferson LS. Mechanisms involved in the coordinate regulation of mTORC1 by insulin and amino acids. J Biol Chem. 2011;286:8287–8296.
  • Inoki K. mTOR signaling in autophagy regulation in the kidney. Semin Nephrol.2014;34:2–8.
  • Kim WY, Nam SA, Song HC, et al. The role of autophagy in unilateral ureteral obstruction rat model. Nephrology (Carlton). 2012;17:148–159.
  • Lee SY, Kim SI, Choi ME. Therapeutic targets for treating fibrotic kidney diseases. Transl Res. 2015; 165:512–530.
  • Li H, Peng X, Wang Y, et al. Atg5-mediated autophagy deficiency in proximal tubules promotes cell cycle G2/M arrest and renal fibrosis. Autophagy. 2016;12:1472–1486.
  • Forbes MS, Thornhill BA, Chevalier RL. Proximal tubular injury and rapid formation of atubular glomeruli in mice with unilateral ureteral obstruction: A new look at an old model. Am J Physiol Renal Physiol. 2011;301:F110–F117.
  • Livingston MJ, Ding HF, Huang S, Hill JA, Yin XM, Dong Z. Persistent activation of autophagy in kidney tubular cells promotes renal interstitial fibrosis during unilateral ureteral obstruction. Autophagy. 2016;12:976–998.