Autophagy: A new treatment strategy for MSC-based therapy in acute kidney injury (Review)

  • Authors:
    • Haoyuan Jia
    • Yongmin Yan
    • Zhaofeng Liang
    • Nitin Tandra
    • Bin Zhang
    • Juanjuan Wang
    • Wenrong Xu
    • Hui Qian
  • View Affiliations

  • Published online on: December 19, 2017     https://doi.org/10.3892/mmr.2017.8311
  • Pages: 3439-3447
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Acute kidney injury (AKI) is a common and serious medical condition associated with poor health outcomes. Autophagy is a conserved multistep pathway that serves a major role in many biological processes and diseases. Recent studies have demonstrated that autophagy is induced in proximal tubular cells during AKI. Autophagy serves a pro‑survival or pro‑death role under certain conditions. Furthermore, mesenchymal stem cells (MSCs) have therapeutic potential in the repair of renal injury. This review summarizes the recent progress on the role of autophagy in AKI and MSCs‑based therapy for AKI. Further research is expected to prevent and treat acute kidney injury.

Introduction

Autophagy is imperative in normal and pathological conditions, including inflammation, adaptation to stress, aging, immunity, metabolic and neurodegenerative disorders, and cancer (17). Both in vivo animal models and in vitro cell culture studies suggest that autophagy serves a role in the maintenance of renal function under pathological conditions including nephrotoxic drugs treatment or ischemia/reperfusion (I/R) injury (8,9).

Acute kidney injury (AKI) is characterized by tubular cell injury and death that leads to a rapid and progressive loss of renal function, including declined glomerular filtration, accumulation of nitrogenous wastes and imbalance of water, electrolytes and acid-base reactions. AKI is a common disease and constitutes a risk factor for chronic kidney disease (10). The incidence of AKI during an ICU stay ranges from 22 to 67% (11). Although there is certain progress in basic research and clinical application, AKI with an increasing morbidity and mortality rate and few prevention and treatment approaches, remains a common and serious clinical condition in hospitalized patients (11).

Autophagy in disease

Autophagy is classified into three subtypes: macroautophagy, chaperone-mediated autophagy, and microautophagy (12). Macroautophagy, generally referred to as autophagy, is well-studied and is the focus of this review. Autophagy is an ‘auto-eating’ process in a cell where intracellular organelles, proteins, and other macromolecules are sequestered into autophagic vesicles (known as autophagosomes) and then degraded by the hydrolases of lysosomes (13). The formation of autophagosomes involves the following steps: Induction, nucleation and elongation that form a complete isolation membrane. Subsequently, double-membrane autophagosomes with sequestered materials inside fuses with the lysosome and form autolysosomes (Fig. 1). Cytoplasmic constituents are degraded by the lysosomal hydrolases for cyclic utilization (14). The initiation step of autophagosome formation involved the ULK1/2-Atg13-FIP200 complex, which is required for the phagophore membranes. The phagophore membrane PI3K/Vps34 complex containing Vps34, Vps15, Beclin-1 and Atg14 L is recruited for nucleation. Autophagy-related gene 9 (Atg 9) and vacuole membrane protein 1 (VMP1) are necessary for membrane expansion. Atg1-Atg13-Atg17 complex promote Atg9 cycling between the phagophore assembly site (PAS) and non-PAS compartments and aided in localization of Atg9 to the PAS (15,16). Indeed, Atg12-Atg5-Atg16 complex and Atg8/light chain 3-II are two ubiquitin-like conjugation systems that are involved in the elongation and expansion steps during autophagosome formation (17). Mi et al (18) demonstrated that actin served a role in autophagosome formation. They demonstrated that polymerized actin puncta were co-localized with DFCP1 [a PtdIns(3)P-binding ER protein that marks omegasomes] and LC3-positive puncta. PtdIns(3)P promoted actin network formation inside membranes, thereby branched actin networks are essential for autophagosomal membrane shaping.

Recent studies have documented the key role of autophagy in normal physiological conditions as well as in pathogenesis conditions. In physiological processes, a low level of basal autophagy occurs to maintain cellular homeostasis. In pathological conditions, cellular stress including hypoxia, cell starvation, oxidant injury, genotoxic agents, and other damaging factors contribute to the induction of autophagy (19,20).

Autophagy has both pro-survival and pro-death functions in gastric cancer cells (21). Autophagy is upregulated in cancer cells to provide nutritive material for cell survival (22). Pla et al (23) demonstrated that autophagy decreased ethanol toxicity in mouse neurons. Autophagy and autophagic flux decreased in aging hearts, stimulation of autophagy alleviated aging-associated pathology in the heart (24). Furthermore, autophagy is considered to play a renoprotective role in kidney injury (25).

Prevention and conventional treatment of AKI

AKI is mainly caused by I/R injury and nephrotoxic drugs which impair renal function Adequate hydration control, maintenance of arterial pressure, pre-emptive use of antioxidants may prevent AKI; few treatments could change the development of AKI (26). Previous studies have demonstrated autophagy serves a role in AKI (2733).

Autophagy in ischemia reperfusion injury

I/R injury is a leading cause of AKI, which is frequently associated with many clinical conditions but lacks effective therapies (27). Recent studies have reported that autophagy is activated in renal I/R injury in vitro and in vivo models irrespective of the genetic or pharmacological impairment of autophagy (2735).

In vivo renal I/R injury patterns, pretreated with 3-methyladenine (3-MA, an autophagy inhibitor) or pretreated with rapamycin (an autophagy activator), demonstrated that autophagy served a renoprotective effect by inhibiting apoptosis (27,28) or by decreasing pro-inflammatory cytokines (29). Similar results were reported by Jiang et al (30), indicated that inhibition of autophagy by chloroquine aggravated AKI, whereas the upregulation of autophagy by rapamycin improved lost renal function, further indicating the protective role of autophagy in AKI. In C57BL/6 mice, renal I/R injury is aggravated by the suppression of autophagy using chloroquine and 3-MA (31). Endoplasmic reticulum stress inducer tunicamycin, caused significant Grp78 expression and ameliorating renal I/R injury in vivo by inducing autophagy (32). It was also found that autophagy protects kidneys from I/R injury in vivo, but the exact mechanisms remain to be elucidated.

Pharmacological inhibitors or inducers of autophagy have non-specific effects and therefore previous studies demonstrated the effect of autophagy during renal I/R by using conditional kidney proximal tubule-specific Atg5- or Atg7-knockout mice (31,33). Kimura et al (33) demonstrated that I/R injury increases proximal tubule cell apoptosis in kidney proximal tubule-specific Atg5 mice compared with the wild-type mice. It is also demonstrated that autophagy serves a critical role in maintaining tubular cell integrity during stress conditions (34).

Jiang et al (30) revealed that autophagy was induced earlier than apoptosis in response to hypoxia, in GFP-LC3-transfected tubular cells. Blockade of autophagy by 3-methyladenine or small-interfering RNA knockdown of Beclin-1 and Atg5 (two essential autophagic genes) lead to hypoxia-induced apoptosis in vitro (31). Transfection of small interfering RNAs for heat shock protein1 have been shown to inhibit cell autophagy and increased apoptosis in GFP-LC3-transfected NRK-52e cells during H2O2 treatment (34,35). Besides rat tubular cells, Wang et al (36) demonstrated that adenosine monophosphate-activated kinase (AMPK) protected pig renal tubular cells (LLC-PK1) from I/R injury by regulating mTOR pathway in vitro.

On the other hand, excessive stimulation of autophagy has been described to exacerbate I/R injury in the kidney (37). Isaka et al (38) used LC3-GFP transgenic mice and demonstrated that I/R injury promoted the formation of LC3-GFP dots. By contrast, increased B-cell lymphoma (Bcl)-2 protein protected tubular epithelial cells from I/R injury by suppressing autophagy and inhibiting apoptosis (38). Furthermore, overexpression of Bcl-xL and Bcl-2 eliminate both apoptosis and autophagy (39). Suzuki et al (40) used HK-2 cells subjected to hypoxia or activation of reactive oxygen species and demonstrated that compared to normoxic conditions LC3-labeled autophagic vacuoles slightly increased and lysosome-associated membrane protein2-labeled lysosomes markedly increased following 48 h of hypoxia. Using lysosomal protease inhibitors autophagosomes increased significantly under hypoxia, suggesting that hypoxia highly induces autophagic generation and degradation. Moreover, HK2 cells with Atg7 deletion significantly inhibit H2O2-induced cell death (40). These results indicate that autophagy may contribute to cell death during renal ischemia-reperfusion injury. In another study it was demonstrated that kidneys of GFP-LC3 transgenic mice that were subjected to 48 h of cold ischemia in the presence of lysosomal inhibitor bafilomycin A1, the number of apoptotic cells were significantly reduced (41) suggesting that autophagy may serve a role in I/R injury.

Non-coding RNAs, including lncRNA and microRNAs, exhibit an effect on regulating autophagy during I/R injury (4244). MicroRNAs serve a significant effect in autophagy during kidney I/R injury both in vivo and in vitro (43,44). Liu et al (43) demonstrated that miR-34a reduced the autophagic activity and caused injury in I/R tubular epithelial cells via a direct binding to the Atg4B 3′-untranslated region. In addition, miR-21 mimics directly targeted Rab11a 3′-UTR reducing beclin-1, LC3-II expression and increased p62 expression in the I/R model. Overexpression of Rab11a weakened the effect of miR-21 mimics and I/R on cell apoptosis (44). In a rat I/R model, pre-treatment with miR-21 inhibitor injection ameliorated the renal injury, and increased the expression of LC3-II and beclin-1 (44). The role of autophagy in I/R damage depends on the injury situation such as the duration of hypoxia, ischemia and reperfusion, and the effect of autophagy in different studies is shown in Table I.

Table I.

Overview of different studies of autophagy in renal I/R injury.

Table I.

Overview of different studies of autophagy in renal I/R injury.

Cause of AKIModelAutophagy modulation methodRole of autophagy(Refs.)
Ischemia 45 minMale3MA RapaProtective(27)
Reperfusion 24 hWistar rats
Hypoxia 24 hTCMK-1 cells3MA RapaProtective(28)
Reoxygenation 2–24 h
Ischemia 30 minMale
reperfusion 0 h-7 dBalB/c mice
Ischemia 30 minMale SD rats3MA RapaProtective(29)
Reperfusion 3, 12, 24 h
Hypoxic 24 hRPTC cellsGFP-LC3 3MAProtective(30)
ATG5 Beclin shRNA
Ischemia 30 minC57BL/6 mice3MA CQ
Reperfusion 0, 6, 24, 48 h
Ischemia 25 minC57BL/6 miceCQ RapaProtective(31)
reperfusion 24 h ATG7 KO
H2O2 200 Μm 4 hLLC-PK1 cellsBeclin ATG5 siRNAProtective(32)
GFP-LC3 3MA CQ
wortmannin tunicamycin
Ischemia 35 minC57BL/6 miceATG5 KOProtective(33)
Reperfusion 3, 6, 12, 24, 36, 48 h GFP-LC3 transgenic
H2O2 200, 400,NRK-52E cellsHSPB1 siRNAProtective(35)
600 mM 4 h LC3 siRNA
Rapa BAF
Ischemia 30 minMale SD rats
Reperfusion 0–72 h
Antimycin A andNRK-52E cellsGFP-LC3Protective(36)
2-deoxyglucose 1.5 hLLC-PK1 cellsshRNA AMPK 3MA Rapa
reperfusion 24 h
Ischemia 45 minMiceLC3-GFP transgenicDetrimental(38)
Reperfusion 96 h Bcl-2/LC3-GFP transgenic
Hypoxia 6–24 hHK-23MA E64d ATG7 siRNAProtective(40)
H2O2 500 µM 8–12 h pepstatin A
Ischemia 30 minMale
Reperfusion 0–48 hC57BL/6 mice
Cold ischemia 48 hMiceGFP-LC3 transgenicDetrimental(41)
Ischemia 35 minMaleProtective(43)
Reperfusion 1, 3, 7 dC57BL/6 mice
Hypoxia 24 hNRK-52E cellsRab11a siRNAProtective(44)
Reoxygenation 24h pcDNA3.1-Rab11a

[i] RPTC, renal proximal tubular cell; Rapa, rapamycin; CQ, chloroquine; BAF, bafilomycin A1; 3MA, 3-methyladenine.

Autophagy in drug-induced acute kidney injury

Cisplatin is a widely used chemotherapeutic drug, with major side effects in kidneys, leading to cell death and tissue damage (45,46). Several studies demonstrated that cisplatin is capable of activating autophagy both in vitro and in vivo AKI models (4751).

Depending on the experimental conditions, autophagy could act as a mechanism of cytoprotection (47,48). Cisplatin induced an increase of LC3-II expression that is further enhanced by emodin treatment in NRK-52E cells (49). Autophagy activity is increased after 6 h of cisplatin treatment and began a gradual decrease from 12 h in proximal tubular cells (RPTCs), which was transiently transfected with GFP-LC3, indicating that autophagy is induced before apoptosis in RPTCs during cisplatin treatment (50). Consistently, using mRFP-LC3-transfected NRK-52E cells it was demonstrated that emodin increased the formation of mRFP-LC3 dots and decreased apoptosis during cisplatin treatment (51). Inhibition of autophagy with either 3-MA or bafilomycin A1 elevated renal proximal tubular cell apoptosis and abolished the protective effects of emodin during cisplatin treatment (50,51), suggesting a renoprotective role of autophagy in cisplatin-induced AKI. HEK cells with Beclin-1 knockdown prevent the formation of GFP-LC3 dots during cisplatin treatment and sensitize cells to cisplatin-induced apoptosis (50). By contrast, overexpressed beclin-1 and Atg5 in LLC-PK1 cells inhibit caspase activation and protecte tubular cells from cisplatin toxicity (51). This further supports that autophagy serves a protective role against cisplatin injury to proximal tubular epithelial cells.

Furthermore, further in vivo studies have affirmed that autophagy serves the same cytoprotective role as in the in vitro findings. An in vivo model autophagy was induced in proximal kidney tubules during cisplatin treatment in C57BL/6 mice (50). Autophagic vesicles were increased in C57BL/6 mice after cisplatin treatment, as detected by electron microscopy (50). Activation of autophagy by rapamycin ameliorated cisplatin-induced AKI, whereas the inhibition of autophagy by chloroquine aggravated cisplatin-induced AKI in C57BL/6 mice (31,51). In addition, proximal tubule-specific Atg7-deficient mice, which were more sensitive to cisplatin-induced injury compared with the wild-type mice (31).

The activity of autophagy is crucial to protect kidney against chemotherapeutics injury (49). The underlying mechanism of how autophagy protects the kidney from the damage induced by cisplatin is not clearly understood (31). P53 is reported to participate in cisplatin-induced renal cell apoptosis in in vitro and in vivo models (5258). Feng et al (59) suggested that p53 regulates autophagy. Periyasamy-Thandavan et al (50) demonstrated that pifithrin-α, a pharmacological inhibitor of p53, inhibited autophagosome formation during cisplatin treatment in RPTCs. Increased p53 and JNK activation aggravate cisplatin-induced proximal tubular cells apoptosis in PT-Atg7-KO mice (31).

mTORC1 is a critical serine-threonine kinase in the autophagy regulation pathway that negatively regulates autophagy activity (49). However, AMPK inhibited the activity of mTORC1, a AMPK inhibitor compound C significantly suppressed emodin-induced AMPK phosphorylation and LC3 conversion, consequently inhibited the autophagic activity and increased the cisplatin-induced proximal tubular cells apoptosis (49). Cells transfected with AMPK small interference (si)RNA were sensitive to cisplatin-induced AKI (60). NAD(P)H: quinone oxidoreductase 1 (NQO1) knockout enhances autophagy in ACHN cells and mice during cisplatin treatment by the AMPK/mTOR signaling pathway (61).

Caspase inhibitor zVAD-fmk prevented the degradation of Atg5, Atg12, and beclin-1 thereby increasing GFP-LC3-II dots in LLC-PK1 during cisplatin treatment. However, autophagosome formation and p62 expression were not significantly increased in the presence of zVAD-fmk and chloroquine, demonstrating that zVAD-fmk impaired autophagic flux by blocking the autophagosome clearance (62). zVAD-fmk suppressed lysosomal function and impaired autophagic flux by inhibiting lysosomal cathepsins (63) and calpains (64,65). A similar result is obtained zVAD-fmk prevented beclin-1 cleavage to impair autophagic flux and increase cisplatin-induced cellular injury in a mouse model (62). There is a connection between apoptosis and autophagy, where a signaling pathway that regulates autophagy can also regulate apoptosis (17). Therfore, 10 µM cisplatin induced autophagy to maintain cell homeostasis, whereas 50 µM cisplatin induced cell apoptosis in NRK-52E cells. However, Rovetta et al (66) demonstrated that ER signaling pathway regulated the balance between autophagy and apoptosis induced by cisplatin. Autophagy mostly served a renoprotective action in cisplatin-induced AKI (Table II).

Table II.

Overview of different studies of autophagy in renal drugs injury.

Table II.

Overview of different studies of autophagy in renal drugs injury.

Cause of AKIModelAutophagy modulation methodRole of autophagy(Refs.)
Cisplatin 10–50 µM 24 hNRK-52E cellsRapa BAFProtective(49)
compound C
Cisplatin 20 µM 0–16 hRPTCBeclin shRNA Bcl-2 transfectionProtective(50)
30 mg/kg 0–3 dC57BL/6 mice
Cisplatin 10 µg/ml 48 hPrimary kidney cellsCompound CProtective(59)
25 mg/kg 48 hC57BL/6 miceAMPK siRNA
Cisplatin 16 mg/kg 3 dMale Wistar ratsCQ RapaProtective(60)
Cisplatin 20 µM 24 hACHN cells3MA CQ RapaProtective(61)
18 mg/kgC57BL/6 miceCompound C NQO1-KO
Cyclosporine A 20 µMHK-2BAFProtective(69)
TMBIM6 KO mice

[i] RPTC, renal proximal tubular cell; Rapa, rapamycin; CQ, chloroquine; BAF, bafilomycin A1; 3MA, 3-methyladenine; NQO1, quinone oxidoreductase 1.

Cyclosporine A (CsA) is an immune-suppressor used in renal allograft transplantation (67). Previous studies demonstrated that CsA induced the accumulation of autophagosomes and inhibited autophagic clearance (68). Yadav et al (69) demonstrated that expression of LC3-II was increased and SQSTM1 accumulation was decreased following CsA treatment in transmembrane BAX inhibitor motif containing 6 (TMBIM6)-expressing HK-2 cells compared to NC cells, suggesting that the autophagic flux was standard in TMBIM6 cells. A similar result was obtained in vivo, in CsA-treated TMBIM6−/− mice where autophagosome formation was increased and the formation and activity of lysosome were decreased (69). The mechanism of TMBIM6-induced autophagy was that it activated PRKAA and suppressed mTORC1 in CsA-treated HK-2 cells. This further demonstrated that TMBIM6 reversed the impaired autophagic flux by stimulating lysosome biogenesis through TFEB activation.

Autophagy in sepsis renal injury

Sepsis-related AKI is an important clinical issue occurring during severe infection including cecal ligation and puncture (CLP) and LPS (70,71). The effect of autophagy in septic AKI remains unclear, compared with the genetic evidence in I/R and cisplatin models (17). Lipopolysaccharide (LPS)-induced accumulation of LC3-II in HPT1 cells derived from human proximal tubular epithelial cells (PTEC) (72). Preincubation of rapamycin decreased tumor necrosis factor (TNF) and induces NRK-52E cell death, whereas the knockdown of Atg7 exaggerated TNF-induced DNA fragmentation (73). This demonstrated that autophagy serves a cytoprotective role in the sepsis-induced AKI. Similar results were found in vivo, LPS induces the accumulation of LC3-positive puncta in GFP-LC3 transgenic mice (74). CLP-induced autophagy was time-dependent in rat, LC3-II was elevated transiently at 3 h but declined at 9 h until 18 h after CLP, renal dysfunction occurred at 9 and 18 h after CLP (73), indicating that autophagy occurred prior to apoptosis and the decline of autophagy aggravated TNF-α-induced cell death. Chloroquine, an inhibitor of lysosomes also aggravates LPS-induced AKI in C57BL/6 mice (74). Tubular injury was more severe in ATG7KO mice than in controls and the expression of IL-6 was significantly increased in ATG7KO kidneys compared with controls following LPS treatment (73), indicating that autophagy protected kidneys from CLP and LPS-induced sepsis injury. Leventhal et al (72) reported that LPS-induced autophagy in RTEC was TLR4-dependent, there were no differences in LC3-II accumulation in C57Bl\10ScN mice, which was no functional TLR4 compared with saline-injected mice. Autophagy was decreased in adult mice during LPS-induced AKI, administration of the mTOR inhibitor temsirolimus increased autophagy and improved renal function in adult mice during LPS-induced AKI (75). In young mice (8 weeks of age), the inhibitor of VPS34 damaged renal function after LPS treatment (75). These results suggest that autophagy serves a renoprotective role irrespective of age (young or aged mice) during LPS treatment (Table III).

Table III.

Overview of different studies of autophagy in renal sepsis injury.

Table III.

Overview of different studies of autophagy in renal sepsis injury.

Cause of AKIModelAutophagy modulation methodRole of autophagy(Refs.)
LPS 1 mg/ml 24 hRTECBafilomycin A1Protective(72)
LPS 15 mg/kg 48 hMale C57BL/6 miceATG7 KO
TNF-α 2 ng/µl 24 hNRK-52E cellssiRNAs Atg7 RapaProtective(73)
CLP 3, 6, 9, 18 hmale SD rats
LPS 10 mg/kg 24 hMale C57BL/6 miceCQ PT-Atg7-KOProtective(74)
RFP-EGFP-LC3
LPS 1.5 mg/kg 48 hMale C57BL/6 micemTOR inhibitorProtective(75)
activator of AMPK
siRNA VPS34

[i] RTEC, renal tubular epithelial cell; Rapa, rapamycin; CQ, chloroquine.

MSCs and extracellular vesicles therapy in AKI

Mesenchymal stem cells (MSCs) are multipotent stem cells isolated from different tissues including bone marrow, umbilical cord, adipose tissue or muscle (76). These cells have self-renewal and multiple differentiation potential including adipocytes, chondrocytes and osteocytes (77). A large number of studies have documented the potential therapeutic effects of MSC, including cardiopathy (78), hepatic diseases (79), and renal injury (80). The therapeutic potential of MSCs includes the anti-inflammatory, antioxidant, anti-fibrotic, anti-apoptotic, pro-angiogenic, stimulation of endogenous progenitor cells, and promotion of cellular reprogramming (7880). Exosomes derived from MSC have beneficial effects in distinct models of injury. Our previous studies suggested that exosomes derived from human umbilical cord MSCs alleviated liver fibrosis (81), cutaneous wound healing (82), and acute myocardial ischemic injury (83).

MSCs are recruited to injured tissues and release certain cytokines and growth factors, such as insulin-like, hepatocyte and vascular endothelial growth factors, which could activate endogenous cellular repair programs contributing to the growth and survival of endothelial and epithelial tubular cells thus promoting renal angiogenesis and regeneration (84,85). Bruno et al (86) demonstrated that MSC-derived microvesicles express MSC markers and transferr cellular materials to neighboring cells, including RNA and proteins, consequently promoting cell proliferation and inhibited apoptosis.

Human MSCs repair HK2 cell after ischemia injury by stimulating normal reactive oxygen species handling, anti-apoptotic activity, energy production, protein synthesis, cytoskeleton organization and cell proliferation (87). In I/R rats, MSCs repair kidney by anti-inflammatory, anti-apoptotic and by enhancing the repair of peritubular capillaries and tubular epithelial cells (88). Furthermore, extracellular vesicles derived from MSCs alleviate kidney injury through anti-oxidation by strengthening Nrf2/ARE activation in I/R rats (89). BM-MSCs ameliorate cisplatin-induced AKI by increasing Foxp3+ T-regulatory cell infiltration in a monkey model (90). Human adipose-derived mesenchymal stem cells protect against cisplatin-induced AKI via anti-apoptotic pathways (91). In a previous study, it demonstrated that human umbilical cord mesenchymal stem cells-derived exosomes decrease cisplatin-induced renal oxidative stress and apoptosis in NRK-52E cells and in rats (92), indicating that MSCs and microvesicles released by MSCs may improve renal injury in AKI.

The role of autophagy in MSCs cell-based therapies

Autophagy plays a critical role in MSCs-based therapy of tissues injury. Hypoxia and heat shock pretreatment enhance survival in BM-MSCs and increase the therapeutic potential of the stem cell by increasing autophagy, providing a novel strategy to improve MSC-based therapies (93,94).

Bone marrow (BM)-MSCs alleviate chronic high glucose-induced injury in INS-1 cells (95) and reduce the severity of lung I/R injury in human pulmonary microvascular endothelial cells (HPMVECs) (96) by enhanced autophagic activity. Similar results are presented in vivo, where BM-MSCs promote the recovery of pancreatic damage in T2D rats (95) and reduce the severity of lung I/R injury in C57BL/6J mice (96). Shin et al (97) demonstrated that MSCs significantly enhance autophagy and the clearance of amyloid-β in Alzheimer disease models to increase neuronal survival against Aβ toxicity. In a Parkinsonian model, BM-MSCs increase the cell viability and reduce α-synuclein by upregulating autophagolysosome formation (98). Tonsil-derived MSCs ameliorate liver fibrosis via the downregulation of TGF-β and autophagy activation (99). These findings support the protective effect of autophagy in MSCs, thereby repairing the pancreas, lung, nervous diseases. In our previous studies, it was demonstrated that human umbilical cord mesenchymal stem cell-derived exosomes prevent cisplatin-induced AKI by autophagy (100) which providing a novel strategy of MSC-based therapy for AKI.

Conclusions

In conclusion, autophagy is induced in kidneys in response to AKI, autophagy in AKI is multifaceted and complex, and it can protect against kidney injury or promote cell death depending on experimental conditions (17). The activity of autophagy is time-dependent. In the early stages of renal injury, autophagy contributes to cell survival, whereas in later stages, autophagy may activate apoptosis signaling pathways sequentially promoting cell death and renal function injury, but the mechanism remains unknown. Autophagy and apoptosis share similar regulators and are mutually inhibitory (101). Many factors that activate autophagy also activate cell apoptosis, which usually precedes cell apoptosis (17). Several critical signaling pathways positively regulate autophagy and apoptosis, such as the tumor suppressor p53, bcl-2 family, death-associated protein kinases and c-Jun N terminal kinases (101). In the model of cisplatin-induced nephrotoxicity, PRKCD suppressed autophagy and promoted renal cell death by AKT/mTOR signal pathway (102). The possible protective mechanism of autophagy in AKI is summarized in Fig. 2. Autophagic flux is frequently defined as a measure of autophagic degradation activity. Continuous observation autophagy is imperative, and to assess the efficiency of the autophagic flux is necessarily in measuring autophagic degradation activity. However, the molecules participating in the autophagosome formation and the regulating mechanism are not known. The signaling pathways, which have been reported in AKI models, such as AMPK and mTOR, are critical in inducing and regulating autophagy but these are also poorly understood.

Pharmacological inhibitors (3-MA or bafilomycin A or wortmannin) or inducers (Rapa) of autophagy are non-specific for autophagy (17). It is necessary to find autophagy inducers in proximal tubules and examine their effects in models of AKI. A comprehensive understanding of autophagy may improve renal function and prevent AKI progression.

MSC-based therapies have been extensively studied as a potential treatment for several diseases (7880). However, our understanding of the regulatory mechanisms of MSCs or MSC-derived vesicles in kidney disease, remain to be fully elucidated. Autophagy serves a key role in MSCs-based therapy, but effects of autophagy in AKI, which are repaired by MSCs are not very clear. Therefore, elucidating its role may provide a novel approach towards the therapy of AKI.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (grant no. 81272481), the Major Research Plan of Jiangsu Higher Education (grant no. 15KJA320001) and a project funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions.

References

1 

Qian M, Fang X and Wang X: Autophagy and inflammation. Clin Transl Med. 6:242017. View Article : Google Scholar : PubMed/NCBI

2 

Tomiyama R, Takakura K, Takatou S, Le TM, Nishiuchi T, Nakamura Y, Konishi T, Matsugo S and Hori O: 3,4-dihydroxybenzalacetone and caffeic acid phenethyl ester induce preconditioning ER stress and autophagy in SH-SY5Y cells. J Cell Physiol. 233:1671–1684. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Antikainen H, Driscoll M, Haspel G and Dobrowolski R: TOR-mediated regulation of metabolism in aging. Aging Cell. 16:1219–1233. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Germic N, Stojkov D, Oberson K, Yousefi S and Simon HU: Neither eosinophils nor neutrophils require ATG5-dependent autophagy for extracellular DNA trap formation. Immunology. 152:517–525. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Long M, Li X, Li L, Dodson M, Zhang DD and Zheng H: Multifunctional p62 effects underlie diverse metabolic diseases. Trends Endocrinol Metab. 28:818–830. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Hu ZY, Chen B, Zhang JP and Ma YY: Up-regulation of autophagy-related gene 5 (ATG5) protects dopaminergic neurons in a zebrafish model of Parkinson's disease. J Biol Chem. 292:18062–18074. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Choi HS, Jeong EH, Lee TG, Kim SY, Kim HR and Kim CH: Autophagy inhibition with monensin enhances cell cycle arrest and apoptosis induced by mTOR or epidermal growth factor receptor inhibitors in lung cancer cells. Tuberc Respir Dis (Seoul). 75:9–17. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Wang Z and Choi ME: Autophagy in kidney health and disease. Antioxid Redox Signal. 20:519–537. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Huber TB, Edelstein CL, Hartleben B, Inoki K, Jiang M, Koya D, Kume S, Lieberthal W, Pallet N, Quiroga A, et al: Emerging role of autophagy in kidney function, diseases and aging. Autophagy. 8:1009–1031. 2012. View Article : Google Scholar : PubMed/NCBI

10 

He L, Wei Q, Liu J, Yi M, Liu Y, Liu H, Sun L, Peng Y, Liu F, Venkatachalam MA and Dong Z: AKI on CKD: Heightened injury, suppressed repair and the underlying mechanisms. Kidney Int. 92:1071–1083. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Trongtrakul K, Sawawiboon C, Wang AY, Chitsomkasem A, Limphunudom P, Kurathong S, Prommoon S, Trakarnvanich T and Srisawat N: Acute kidney injury in critically Ill surgical patients: Epidemiology, risk factors and outcomes. Nephrology (Carlton). 2017. View Article : Google Scholar : PubMed/NCBI

12 

Deng X, Xie Y and Zhang A: Advance of autophagy in chronic kidney diseases. Ren Fail. 39:306–313. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Levine B and Klionsky DJ: Development by self-digestion: Molecular mechanisms and biological functions of autophagy. Dev Cell. 6:463–477. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Mizushima N and Komatsu M: Autophagy: Renovation of cells and tissues. Cell. 147:728–741. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Ravikumar B, Sarkar S, Davies JE, Futter M, Garcia-Arencibia M, Green-Thompson ZW, Jimenez-Sanchez M, Korolchuk VI, Lichtenberg M, Luo S, et al: Regulation of mammalian autophagy in physiology and pathophysiology. Physiol Rev. 90:1383–1435. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Sekito T, Kawamata T, Ichikawa R, Suzuki K and Ohsumi Y: Atg17 recruits Atg9 to organize the pre-autophagosomal structure. Genes Cells. 14:525–538. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Livingston MJ and Dong Z: Autophagy in acute kidney injury. Semin Nephrol. 34:17–26. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Mi N, Chen Y, Wang S, Chen M, Zhao M, Yang G, Ma M, Su Q, Luo S, Shi J, et al: CapZ regulates autophagosomal membrane shaping by promoting actin assembly inside the isolation membrane. Nat Cell Biol. 17:1112–1123. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Sureshbabu A, Ryter SW and Choi ME: Oxidative stress and autophagy: Crucial modulators of kidney injury. Redox Biol. 4:208–214. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Ravanan P, Srikumar IF and Talwar P: Autophagy: The spotlight for cellular stress responses. Life Sci. 188:53–67. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Tang JC, Feng YL, Liang X and Cai XJ: Autophagy in 5-fluorouracil therapy in gastrointestinal cancer: Trends and challenges. Chin Med J (Engl). 129:456–463. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, Mukherjee C, Shi Y, Gélinas C, Fan Y, et al: Autophagy promotes tumor cell survival and restricts necrosis, inflammation and tumorigenesis. Cancer Cell. 10:51–64. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Pla A, Pascual M and Guerri C: Autophagy constitutes a protective mechanism against ethanol toxicity in mouse astrocytes and neurons. PLoS One. 11:e01530972016. View Article : Google Scholar : PubMed/NCBI

24 

Shirakabe A, Ikeda Y, Sciarretta S, Zablocki DK and Sadoshima J: Aging and autophagy in the heart. Circ Res. 118:1563–1576. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Wang XY, Yang H, Wang MG, Yang DB, Wang ZY and Wang L: Trehalose protects against cadmium-induced cytotoxicity in primary rat proximal tubular cells via inhibiting apoptosis and restoring autophagic flux. Cell Death Dis. 8:e30992017. View Article : Google Scholar : PubMed/NCBI

26 

de Almeida DC, Donizetti-Oliveira C, Barbosa-Costa P, Origassa CS and Câmara NO: In search of mechanisms associated with mesenchymal stem cell-based therapies for acute kidney injury. Clin Biochem Rev. 34:131–144. 2013.PubMed/NCBI

27 

Zhang YL, Zhang J, Cui LY and Yang S: Autophagy activation attenuates renal ischemia-reperfusion injury in rats. Exp Biol Med (Maywood). 240:1590–1598. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Guan X, Qian Y, Shen Y, Zhang L, Du Y, Dai H, Qian J and Yan Y: Autophagy protects renal tubular cells against ischemia/reperfusion injury in a time-dependent manner. Cell Physiol Biochem. 36:285–298. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Ling H, Chen H, Wei M, Meng X, Yu Y and Xie K: The effect of autophagy on inflammation cytokines in renal ischemia/reperfusion injury. Inflammation. 39:347–356. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Jiang M, Liu K, Luo J and Dong Z: Autophagy is a renoprotective mechanism during in vitro hypoxia and in vivo ischemia-reperfusion injury. Am J Pathol. 176:1181–1192. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Jiang M, Wei Q, Dong G, Komatsu M, Su Y and Dong Z: Autophagy in proximal tubules protects against acute kidney injury. Kidney Int. 82:1271–1283. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Chandrika BB, Yang C, Ou Y, Feng X, Muhoza D, Holmes AF, Theus S, Deshmukh S, Haun RS and Kaushal GP: Endoplasmic reticulum stress-induced autophagy provides cytoprotection from chemical hypoxia and oxidant injury and ameliorates renal ischemia-reperfusion injury. PLoS One. 10:e01400252015. View Article : Google Scholar : PubMed/NCBI

33 

Kimura T, Takabatake Y, Takahashi A, Kaimori JY, Matsui I, Namba T, Kitamura H, Niimura F, Matsusaka T, Soga T, et al: Autophagy protects the proximal tubule from degeneration and acute ischemic injury. J Am Soc Nephrol. 22:902–913. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Liu S, Hartleben B, Kretz O, Wiech T, Igarashi P, Mizushima N, Walz G and Huber TB: Autophagy plays a critical role in kidney tubule maintenance, aging and ischemia-reperfusion injury. Autophagy. 8:826–837. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Matsumoto T, Urushido M, Ide H, Ishihara M, Hamada-Ode K, Shimamura Y, Ogata K, Inoue K, Taniguchi Y, Taguchi T, et al: Small heat shock protein beta-1 (HSPB1) is upregulated and regulates autophagy and apoptosis of renal tubular cells in acute kidney injury. PLoS One. 10:e01262292015. View Article : Google Scholar : PubMed/NCBI

36 

Wang LT, Chen BL, Wu CT, Huang KH, Chiang CK and Hwa Liu S: Protective role of AMP-activated protein kinase-evoked autophagy on an in vitro model of ischemia/reperfusion-induced renal tubular cell injury. PLoS One. 8:e798142013. View Article : Google Scholar : PubMed/NCBI

37 

Decuypere JP, Ceulemans LJ, Agostinis P, Monbaliu D, Naesens M, Pirenne J and Jochmans I: Autophagy and the kidney: Implications for ischemia-reperfusion injury and therapy. Am J Kidney Dis. 66:699–709. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Isaka Y, Suzuki C, Abe T, Okumi M, Ichimaru N, Imamura R, Kakuta Y, Matsui I, Takabatake Y, Rakugi H, et al: Bcl-2 protects tubular epithelial cells from ischemia/reperfusion injury by dual mechanisms. Transplant Proc. 41:pp. 52–54. 2009; View Article : Google Scholar : PubMed/NCBI

39 

Chien CT, Shyue SK and Lai MK: Bcl-xL augmentation potentially reduces ischemia/reperfusion induced proximal and distal tubular apoptosis and autophagy. Transplantation. 84:1183–1190. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Suzuki C, Isaka Y, Takabatake Y, Tanaka H, Koike M, Shibata M, Uchiyama Y, Takahara S and Imai E: Participation of autophagy in renal ischemia/reperfusion injury. Biochem Biophys Res Commun. 368:100–106. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Turkmen K, Martin J, Akcay A, Nguyen Q, Ravichandran K, Faubel S, Pacic A, Ljubanović D, Edelstein CL and Jani A: Apoptosis and autophagy in cold preservation ischemia. Transplantation. 91:1192–1197. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Yu SY, Dong B, Zhou SH and Tang L: LncRNA MALAT1: A potential regulator of autophagy in myocardial ischemia-reperfusion injury. Int J Cardiol. 247:252017. View Article : Google Scholar : PubMed/NCBI

43 

Liu XJ, Hong Q, Wang Z, Yu YY, Zou X and Xu LH: MicroRNA-34a suppresses autophagy in tubular epithelial cells in acute kidney injury. Am J Nephrol. 42:168–175. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Liu X, Hong Q, Wang Z, Yu Y, Zou X and Xu L: MiR-21 inhibits autophagy by targeting Rab11a in renal ischemia/reperfusion. Exp Cell Res. 338:64–69. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Arany I and Safirstein RL: Cisplatin nephrotoxicity. Semin Nephrol. 23:460–464. 2003. View Article : Google Scholar : PubMed/NCBI

46 

Pabla N and Dong Z: Cisplatin nephrotoxicity: Mechanisms and renoprotective strategies. Kidney Int. 73:994–1007. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Levine B and Yuan J: Autophagy in cell death: An innocent convict? J Clin Invest. 115:2679–2688. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Maiuri MC, Zalckvar E, Kimchi A and Kroemer G: Self-eating and self-killing: Crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol. 8:741–752. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Liu H, Gu LB, Tu Y, Hu H, Huang YR and Sun W: Emodin ameliorates cisplatin-induced apoptosis of rat renal tubular cells in vitro by activating autophagy. Acta Pharmacol Sin. 37:235–245. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Periyasamy-Thandavan S, Jiang M, Wei Q, Smith R, Yin XM and Dong Z: Autophagy is cytoprotective during cisplatin injury of renal proximal tubular cells. Kidney Int. 74:631–640. 2008. View Article : Google Scholar : PubMed/NCBI

51 

Fang B and Xiao H: Rapamycin alleviates cisplatin-induced ototoxicity in vivo. Biochem Biophys Res Commun. 448:443–447. 2014. View Article : Google Scholar : PubMed/NCBI

52 

Sprowl JA, Lancaster CS, Pabla N, Hermann E, Kosloske AM, Gibson AA, Li L, Zeeh D, Schlatter E, Janke LJ, et al: Cisplatin-induced renal injury is independently mediated by OCT2 and p53. Clin Cancer Res. 20:4026–4035. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Cummings BS and Schnellmann RG: Cisplatin-induced renal cell apoptosis: Caspase 3-dependent and -independent pathways. J Pharmacol Exp Ther. 302:8–17. 2002. View Article : Google Scholar : PubMed/NCBI

54 

Jiang M, Wei Q, Wang J, Du Q, Yu J, Zhang L and Dong Z: Regulation of PUMA-alpha by p53 in cisplatin-induced renal cell apoptosis. Oncogene. 25:4056–4066. 2006. View Article : Google Scholar : PubMed/NCBI

55 

Jiang M, Yi X, Hsu S, Wang CY and Dong Z: Role of p53 in cisplatin-induced tubular cell apoptosis: Dependence on p53 transcriptional activity. Am J Physiol Renal Physiol. 287:F1140–F1147. 2004. View Article : Google Scholar : PubMed/NCBI

56 

Seth R, Yang C, Kaushal V, Shah SV and Kaushal GP: p53-Dependent caspase-2 activation in mitochondrial release of apoptosis-inducing factor and its role in renal tubular epithelial cell injury. J Biol Chem. 280:31230–31239. 2005. View Article : Google Scholar : PubMed/NCBI

57 

Wei Q, Dong G, Franklin J and Dong Z: The pathological role of Bax in cisplatin nephrotoxicity. Kidney Int. 72:53–62. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Wei Q, Dong G, Yang T, Megyesi J, Price PM and Dong Z: Activation and involvement of p53 in cisplatin-induced nephrotoxicity. Am J Physiol Renal Physiol. 293:F1282–F1291. 2007. View Article : Google Scholar : PubMed/NCBI

59 

Feng Z, Zhang H, Levine AJ and Jin S: The coordinate regulation of the p53 and mTOR pathways in cells. Proc Natl Acad Sci USA. 102:pp. 8204–8209. 2005; View Article : Google Scholar : PubMed/NCBI

60 

Wei L, Chen W, Zou Y, Huang H, Pan B, Jin S, Huang R, Nie S and Kong G: AMP-activated protein kinase regulates autophagic protection against cisplatin-induced tissue injury in the kidney. Genet Mol Res. 14:12006–12015. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Kim TW, Kim YJ, Kim HT, Park SR, Lee MY, Park YD, Lee CH and Jung JY: NQO1 deficiency leads enhanced autophagy in cisplatin-induced acute kidney injury through the AMPK/TSC2/mTOR signaling pathway. Antioxid Redox Signal. 24:867–883. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Herzog C, Yang C, Holmes A and Kaushal GP: zVAD-fmk prevents cisplatin-induced cleavage of autophagy proteins but impairs autophagic flux and worsens renal function. Am J Physiol Renal Physiol. 303:F1239–F1250. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Scaringi L, Cornacchione P, Ayroldi E, Corazzi L, Capodicasa E, Rossi R and Marconi P: Omeprazole induces apoptosis in jurkat cells. Int J Immunopathol Pharmacol. 17:331–342. 2004. View Article : Google Scholar : PubMed/NCBI

64 

Bizat N, Galas MC, Jacquard C, Boyer F, Hermel JM, Schiffmann SN, Hantraye P, Blum D and Brouillet E: Neuroprotective effect of zVAD against the neurotoxin 3-nitropropionic acid involves inhibition of calpain. Neuropharmacology. 49:695–702. 2005. View Article : Google Scholar : PubMed/NCBI

65 

Madden DT, Egger L and Bredesen DE: A calpain-like protease inhibits autophagic cell death. Autophagy. 3:519–522. 2007. View Article : Google Scholar : PubMed/NCBI

66 

Rovetta F, Stacchiotti A, Consiglio A, Cadei M, Grigolato PG, Lavazza A, Rezzani R and Aleo MF: ER signaling regulation drives the switch between autophagy and apoptosis in NRK-52E cells exposed to cisplatin. Exp Cell Res. 318:238–250. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Xu QX, Qiu XY, Jiao Z, Zhang M and Zhong MK: FOXP3 rs3761549 polymorphism predicts long-term renal allograft function in patients receiving cyclosporine-based immunosuppressive regimen. Gene. 2017.(Epub ahead of print).

68 

Lim SW, Hyoung BJ, Piao SG, Doh KC, Chung BH and Yang CW: Chronic cyclosporine nephropathy is characterized by excessive autophagosome formation and decreased autophagic clearance. Transplantation. 94:218–225. 2012. View Article : Google Scholar : PubMed/NCBI

69 

Yadav RK, Lee GH, Lee HY, Li B, Jung HE, Rashid HO, Choi MK, Yadav BK, Kim WH, Kim KW, et al: TMBIM6 (transmembrane BAX inhibitor motif containing 6) enhances autophagy and reduces renal dysfunction in a Cyclosporine A-induced nephrotoxicity model. Autophagy. 11:1760–1774. 2015. View Article : Google Scholar : PubMed/NCBI

70 

Yegenaga I, Tuglular S, Ari E, Etiler N, Baykara N, Torlak S, Acar S, Akbas T, Toker K and Solak ZM: Evaluation of sepsis/systemic inflammatory response syndrome, acute kidney injury and RIFLE criteria in two tertiary hospital intensive care units in Turkey. Nephron Clin Pract. 115:c276–c282. 2010. View Article : Google Scholar : PubMed/NCBI

71 

Schrier RW and Wang W: Acute renal failure and sepsis. N Engl J Med. 351:159–169. 2004. View Article : Google Scholar : PubMed/NCBI

72 

Leventhal JS, Ni J, Osmond M, Lee K, Gusella GL, Salem F and Ross MJ: Autophagy limits endotoxemic acute kidney injury and alters renal tubular epithelial cell cytokine expression. PLoS One. 11:e01500012016. View Article : Google Scholar : PubMed/NCBI

73 

Hsiao HW, Tsai KL, Wang LF, Chen YH, Chiang PC, Chuang SM and Hsu C: The decline of autophagy contributes to proximal tubular dysfunction during sepsis. Shock. 37:289–296. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Mei S, Livingston M, Hao J, Li L, Mei C and Dong Z: Autophagy is activated to protect against endotoxic acute kidney injury. Sci Rep. 6:221712016. View Article : Google Scholar : PubMed/NCBI

75 

Howell GM, Gomez H, Collage RD, Loughran P, Zhang X, Escobar DA, Billiar TR, Zuckerbraun BS and Rosengart MR: Augmenting autophagy to treat acute kidney injury during endotoxemia in mice. PLoS One. 8:e695202013. View Article : Google Scholar : PubMed/NCBI

76 

Kern S, Eichler H, Stoeve J, Klüter H and Bieback K: Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells. 24:1294–1301. 2006. View Article : Google Scholar : PubMed/NCBI

77 

Caplan AI: Mesenchymal stem cells. J Orthop Res. 9:641–650. 1991. View Article : Google Scholar : PubMed/NCBI

78 

Golpanian S, Wolf A, Hatzistergos KE and Hare JM: Rebuilding the damaged heart: Mesenchymal stem cells, cell-based therapy and engineered heart tissue. Physiol Rev. 96:1127–1168. 2016. View Article : Google Scholar : PubMed/NCBI

79 

Gazdic M, Arsenijevic A, Markovic BS, Volarevic A, Dimova I, Djonov V, Arsenijevic N, Stojkovic M and Volarevic V: Mesenchymal stem cell-dependent modulation of liver diseases. Int J Biol Sci. 13:1109–1117. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Chen C and Hou J: Mesenchymal stem cell-based therapy in kidney transplantation. Stem Cell Res Ther. 7:162016. View Article : Google Scholar : PubMed/NCBI

81 

Li T, Yan Y, Wang B, Qian H, Zhang X, Shen L, Wang M, Zhou Y, Zhu W, Li W and Xu W: Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev. 22:845–854. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Zhang B, Wang M, Gong A, Zhang X, Wu X, Zhu Y, Shi H, Wu L, Zhu W, Qian H and Xu W: HucMSC-exosome mediated-Wnt4 signaling is required for cutaneous wound healing. Stem Cells. 33:2158–2168. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Zhao Y, Sun X, Cao W, Ma J, Sun L, Qian H, Zhu W and Xu W: Exosomes derived from human umbilical cord mesenchymal stem cells relieve acute myocardial ischemic injury. Stem Cells Int. 2015:7616432015. View Article : Google Scholar : PubMed/NCBI

84 

Meirelles Lda S, Fontes AM, Covas DT and Caplan AI: Mechanisms involved in the therapeutic properties of mesenchymal stem cells. Cytokine Growth Factor Rev. 20:419–427. 2009. View Article : Google Scholar : PubMed/NCBI

85 

Tögel F, Zhang P, Hu Z and Westenfelder C: VEGF is a mediator of the renoprotective effects of multipotent marrow stromal cells in acute kidney injury. J Cell Mol Med. 13:2109–2114. 2009. View Article : Google Scholar : PubMed/NCBI

86 

Bruno S, Grange C, Collino F, Deregibus MC, Cantaluppi V, Biancone L, Tetta C and Camussi G: Microvesicles derived from mesenchymal stem cells enhance survival in a lethal model of acute kidney injury. PLoS One. 7:e331152012. View Article : Google Scholar : PubMed/NCBI

87 

da Costa MR, Pizzatti L, Lindoso RS, Sant'Anna JF, DuRocher B, Abdelhay E and Vieyra A: Mechanisms of kidney repair by human mesenchymal stromal cells after ischemia: A comprehensive view using label-free MS(E). Proteomics. 14:1480–1493. 2014. View Article : Google Scholar : PubMed/NCBI

88 

Xing L, Cui R, Peng L, Ma J, Chen X, Xie RJ and Li B: Mesenchymal stem cells, not conditioned medium, contribute to kidney repair after ischemia-reperfusion injury. Stem Cell Res Ther. 5:1012014. View Article : Google Scholar : PubMed/NCBI

89 

Zhang G, Zou X, Huang Y, Wang F, Miao S, Liu G, Chen M and Zhu Y: Mesenchymal stromal cell-derived extracellular vesicles protect against acute kidney injury through anti-oxidation by enhancing Nrf2/ARE activation in rats. Kidney Blood Press Res. 41:119–128. 2016. View Article : Google Scholar : PubMed/NCBI

90 

Moghadasali R, Azarnia M, Hajinasrollah M, Arghani H, Nassiri SM, Molazem M, Vosough A, Mohitmafi S, Najarasl M, Ajdari Z, et al: Intra-renal arterial injection of autologous bone marrow mesenchymal stromal cells ameliorates cisplatin-induced acute kidney injury in a rhesus Macaque mulatta monkey model. Cytotherapy. 16:734–749. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Zhou Y, Xu H, Xu W, Wang B, Wu H, Tao Y, Zhang B, Wang M, Mao F, Yan Y, et al: Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin-induced renal oxidative stress and apoptosis in vivo and in vitro. Stem Cell Res Ther. 4:342013. View Article : Google Scholar : PubMed/NCBI

92 

Yao W, Hu Q, Ma Y, Xiong W, Wu T, Cao J and Wu D: Human adipose-derived mesenchymal stem cells repair cisplatin-induced acute kidney injury through antiapoptotic pathways. Exp Ther Med. 10:468–476. 2015. View Article : Google Scholar : PubMed/NCBI

93 

Zhang W, Liu L, Huo Y, Yang Y and Wang Y: Hypoxia-pretreated human MSCs attenuate acute kidney injury through enhanced angiogenic and antioxidative capacities. Biomed Res Int. 2014:4624722014.PubMed/NCBI

94 

Qiao PF, Yao L, Zhang XC, Li GD and Wu DQ: Heat shock pretreatment improves stem cell repair following ischemia-reperfusion injury via autophagy. World J Gastroenterol. 21:12822–12834. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Zhao K, Hao H, Liu J, Tong C, Cheng Y, Xie Z, Zang L, Mu Y and Han W: Bone marrow-derived mesenchymal stem cells ameliorate chronic high glucose-induced β-cell injury through modulation of autophagy. Cell Death Dis. 6:e18852015. View Article : Google Scholar : PubMed/NCBI

96 

Li J, Zhou J, Zhang D, Song Y, She J and Bai C: Bone marrow-derived mesenchymal stem cells enhance autophagy via PI3K/AKT signaling to reduce the severity of ischaemia/reperfusion-induced lung injury. J Cell Mol Med. 19:2341–2351. 2015. View Article : Google Scholar : PubMed/NCBI

97 

Shin JY, Park HJ, Kim HN, Oh SH, Bae JS, Ha HJ and Lee PH: Mesenchymal stem cells enhance autophagy and increase β-amyloid clearance in Alzheimer disease models. Autophagy. 10:32–44. 2014. View Article : Google Scholar : PubMed/NCBI

98 

Park HJ, Shin JY, Kim HN, Oh SH and Lee PH: Neuroprotective effects of mesenchymal stem cells through autophagy modulation in a parkinsonian model. Neurobiol Aging. 35:1920–1928. 2014. View Article : Google Scholar : PubMed/NCBI

99 

Park M, Kim YH, Woo SY, Lee HJ, Yu Y, Kim HS, Park YS, Jo I, Park JW, Jung SC, et al: Tonsil-derived mesenchymal stem cells ameliorate CCl4-induced liver fibrosis in mice via autophagy activation. Sci Rep. 5:86162015. View Article : Google Scholar : PubMed/NCBI

100 

Wang B, Jia H, Zhang B, Wang J, Ji C, Zhu X, Yan Y, Yin L, Yu J, Qian H and Xu W: Pre-incubated with hucMSC-Exosomes prevent cisplatin-induced nephrotoxicity by activating autophagy. Stem Cell Res Ther. 8:752017. View Article : Google Scholar : PubMed/NCBI

101 

Fougeray S and Pallet N: Mechanisms and biological functions of autophagy in diseased and ageing kidneys. Nat Rev Nephrol. 11:34–45. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Zhang D, Xu X and Dong Z: PRKCD/PKCσ contributes to nephrotoxicity during cisplatin chemotherapy by suppressing autophagy. Autophagy. 13:631–632. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2018
Volume 17 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jia H, Yan Y, Liang Z, Tandra N, Zhang B, Wang J, Xu W and Qian H: Autophagy: A new treatment strategy for MSC-based therapy in acute kidney injury (Review). Mol Med Rep 17: 3439-3447, 2018
APA
Jia, H., Yan, Y., Liang, Z., Tandra, N., Zhang, B., Wang, J. ... Qian, H. (2018). Autophagy: A new treatment strategy for MSC-based therapy in acute kidney injury (Review). Molecular Medicine Reports, 17, 3439-3447. https://doi.org/10.3892/mmr.2017.8311
MLA
Jia, H., Yan, Y., Liang, Z., Tandra, N., Zhang, B., Wang, J., Xu, W., Qian, H."Autophagy: A new treatment strategy for MSC-based therapy in acute kidney injury (Review)". Molecular Medicine Reports 17.3 (2018): 3439-3447.
Chicago
Jia, H., Yan, Y., Liang, Z., Tandra, N., Zhang, B., Wang, J., Xu, W., Qian, H."Autophagy: A new treatment strategy for MSC-based therapy in acute kidney injury (Review)". Molecular Medicine Reports 17, no. 3 (2018): 3439-3447. https://doi.org/10.3892/mmr.2017.8311