mTOR signaling pathway

mTOR signaling pathway

Accepted Manuscript Human umbilical cord-derived mesenchymal stem cells enhanced HK-2 cell autophagy through MicroRNA-145 by inhibiting the PI3K/AKT/m...

895KB Sizes 0 Downloads 14 Views

Accepted Manuscript Human umbilical cord-derived mesenchymal stem cells enhanced HK-2 cell autophagy through MicroRNA-145 by inhibiting the PI3K/AKT/mTOR signaling pathway Jin Xiang, Tingting Jiang, Wenying Zhang, Wei Xie, Xun Tang, Jun Zhang PII:

S0014-4827(18)31047-4

DOI:

https://doi.org/10.1016/j.yexcr.2019.03.019

Reference:

YEXCR 11355

To appear in:

Experimental Cell Research

Received Date: 2 October 2018 Revised Date:

10 March 2019

Accepted Date: 11 March 2019

Please cite this article as: J. Xiang, T. Jiang, W. Zhang, W. Xie, X. Tang, J. Zhang, Human umbilical cord-derived mesenchymal stem cells enhanced HK-2 cell autophagy through MicroRNA-145 by inhibiting the PI3K/AKT/mTOR signaling pathway, Experimental Cell Research (2019), doi: https:// doi.org/10.1016/j.yexcr.2019.03.019. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

Human Umbilical Cord-Derived Mesenchymal Stem Cells Enhanced HK-2 Cell Autophagy Through MicroRNA-145 By Inhibiting The

RI PT

PI3K/AKT/mTOR Signaling Pathway

SC

Jin Xiang1, 2#, Tingting Jiang3#, Wenying Zhang1, Wei Xie1, Xun Tang1*, Jun Zhang1*

Guangdong Province, 510282, China

M AN U

1 Department of Nephrology, Zhujiang Hospital of Southern Medical University, Guangzhou,

2 Department of Nephrology, People's Hospital of Yuxi City, Yuxi, Yunnan Province, 653100, China

TE D

3 Department of Nephrology, The People’s Hospital of Guangxi Zhuang Autonomous Region,

*

These authors contributed equally to this work.

AC C

#

EP

Nanning, Guangxi Province, 530021, China

corresponding author: Jun Zhang, Department of Nephrology, Zhujiang Hospital of Southern Medical University, 253 Gongye Road, Guangzhou, Guangdong, 510282 (PR China). E-mail: [email protected]

Xun Tang, Department of Nephrology, Zhujiang Hospital of

ACCEPTED MANUSCRIPT Southern Medical University, 253 Gongye Road,

Guangzhou, Guangdong, 510282 (PR China).

SC

RI PT

E-mail: [email protected]

Abstract: Recent studies have shown that autophagy exhibits a protective role in

M AN U

acute kidney injury (AKI), and the accumulation of advanced oxidation protein products (AOPP) participates in the progression of kidney diseases. Our previous study indicated that AOPP induced injury in renal tubular epithelial cells (RTECs) through autophagy inhibition. Besides, we found that human umbilical cord-derived

TE D

mesenchymal stem cells (hUC-MSCs) enhanced autophagy in AOPP-treated RTECs, but the underlying mechanism remains unclear. We regulated microRNA-145

EP

(miR-145) expression in HK-2 cells (a cell line of RTECs), or co-cultured hUC-MSCs with HK-2 cells and studied the autophagic activity in HK-2 cells to explore the

AC C

underlying mechanism. Our data demonstrated that upregulated miR-145 increased LC3 II and Beclin 1 levels, decreased p62 level, three autophagy related proteins, inhibited the phosphorylation of PI3K/AKT/mTOR, and increased LC3B-positive staining and the autophagosome number. Furthermore, hUC-MSCs enhanced autophagy and inhibited phosphorylation of PI3K/AKT/mTOR in AOPP-treated HK-2 cells, which was then partially rescued using miR-145 knockdown in the hUC-MSCs co-culture system. In conclusion, our study showed that hUC-MSCs

ACCEPTED MANUSCRIPT enhanced autophagy in AOPP-treated HK-2 cells mediated by miR-145 via inhibition of the PI3K/AKT/mTOR pathway, which indicated that hUC-MSCs might serve as a

RI PT

prospective therapy for AKI.

SC

Keywords: hUC-MSCs; miR-145; autophagy; HK-2 cells; PI3K/AKT/mTOR

M AN U

Introduction

Acute kidney injury (AKI) is a global public health concern and is associated with high morbidity, mortality, and healthcare costs; AKI impacts approximately 13.3 million patients per year [1]. Long-term outcomes may include the development of

TE D

chronic kidney disease (CKD) [2] and end-stage renal disease (ESRD) [3], which require renal replacement therapy and cause a heavy economic burden. Therefore, it is

EP

important to understand the mechanisms of AKI and develop an effective treatment strategy to reduce the occurrence of AKI or slow the development of CKD and

AC C

ESRD.

A growing number of studies have revealed the role of autophagy in kidney

diseases [4] [5], both in the tubulointerstitial compartment and in glomeruli. Pathologically, AKI is characterized by tubular cell injury and death. In addition, the role of autophagy in tubules, which is the key determinant of kidney prognosis, has been demonstrated in renal tubular epithelial cells (RTECs) [6] and is generally

ACCEPTED MANUSCRIPT accepted as a protective cellular response in AKI under various causes [7] [8]. Advanced oxidation protein products (AOPP) is a harmful protein product that is highly expressed in patients with kidney diseases. Furthermore, phosphatidylinositol

RI PT

3-kinase (PI3K)/AKT/mTOR signaling pathway is a crucial negative modulator of autophagy[9]. Our work team have made some researches about the harmful effects of AOPP on kidney diseases. In the latest investigation, our data showed that AOPP

SC

inhibited autophagy of RTECs trough activation of the PI3K/AKT/mTOR signaling

M AN U

pathway. Moreover, we also reported that autophagy inhibition mediated AOPP-induced RTEC injury in this study. [10]. Therefore, enhancing RTEC autophagy may be critical for suppressing the progression of kidney diseases including AKI.

TE D

Currently, stem cell therapy has therapeutic potential to repair the impaired kidney. The mesenchymal stem cell (MSC) is a mesodermal stem cell that has the ability to

EP

self-renew and differentiate into other mesodermal cell lineages. Compared with MSCs from other sources, human umbilical cord-derived MSCs (hUC-MSCs) have

AC C

received much attention because of their abundance, ease of separation, strong growth capacity, low immunogenicity, and lack of harm to mothers and newborns [11]. It is interesting to note that a systematic review and meta-analysis entering 71 articles showed that MSC therapy could delay the progression of CKD according to subgroup analysis, blood biochemical indexes, different related models and cell related factors [12]. Furthermore, the study of MSCs in the treatment of kidney diseases has become one of the hot topics in international nephrology research, including diabetic

ACCEPTED MANUSCRIPT nephropathy [13], renal transplantation [14], and lupus nephritis [15]. More excitingly, we have found that hUC-MSCs enhanced autophagy in AOPP-injured RTEC, which may perform a protective role in renal tissue and cells (the article is under review.)

RI PT

An increasing number of studies have demonstrated that MSCs regulate the microenvironment of renal cells to play a protective role by secreting a variety of

SC

cytokines, proteins and miRNAs [16] [17]. Notably, the potential therapeutic role of microRNA-145 (miR-145) secreted from MSCs has attracted increasing attention. A

M AN U

previous study found that hUC-MSCs inhibited the TGF-β/SMAD2 signaling pathway to protect the body from burn injury by secreting miR-145 [18]. Another study showed that MSCs inhibited the migration and self-renewal of glioma cells and glioma stem cells by secreting miR-145 [19]. More excitingly, it was reported that

TE D

miR-145 repaired myocardial infarction by promoting myocardial cell autophagy [20]. In addition, there was another study showing that upregulation of miR-145 induced

EP

inhibition of the PI3K/AKT/mTOR pathway in laryngeal squamous cell carcinoma[21]. However, whether miR-145 secreted from hUC-MSCs plays

AC C

protective roles in AKI by enhancing RTEC autophagy and the underlying mechanism need to be explored.

To further investigate the relationship between MSCs and renal autophagy, we

performed the current study to investigate the role of hUC-MSCs on autophagy inhibited by AOPP in HK-2 cells (a cell line of human RTECs) and the underlying mechanism.

ACCEPTED MANUSCRIPT

Material and Methods

RI PT

1. Materials and Reagents Bovine serum albumin (BSA) was purchased from Sigma (St. Louis, USA). Antibodies against LC3B, Beclin 1, p62, p-mTOR, mTOR, p-AKT, AKT, and p-PI3K

SC

were obtained from Cell Signaling Technology (Beverly, USA). The GAPDH antibody was purchased from Bioworld (Minnesota, USA). Antibodies against CD90,

M AN U

CD105, CD73, CD34, CD45, CD11b, CD14, and HLA-DR for flow cytometry were purchased from Becton Dickinson and Company (New Jersey, USA). The U6 and miR-145 primers were obtained from Ribobio Company (Guangzhou, China). Sequencing of miR-145 siRNA, miR-145 mimics, and negative siRNA was

TE D

performed by Shanghai GenePharma Co. Ltd (Shanghai, China).

EP

2. AOPP Preparation

AOPP was prepared as previously described [22]. Briefly, HOCl (200 mmol/L) was

AC C

mixed with BSA solution for 30 min at room temperature, and then dialyzed against PBS at 4°C to remove free HOCl for 24 h. Native BSA was dissolved in PBS alone as the control. The endotoxin levels were measured and were required to be <0.025 EU/mL. The AOPP content was measured at 340 nm to obtain the absorbance under acidic conditions and calibrated using chloramine-T in the presence of potassium iodide.

3. HK-2 Cell Culture and Treatment

ACCEPTED MANUSCRIPT HK-2 cells were purchased from the American Type Culture Collection (Rockville, USA) and cultured in DMEM/F12 (Gibco, USA) supplemented with 10% heat-inactivated FBS and maintained in a humidified incubator containing a 5% CO2

until they reached approximately 70-80% confluence.

SC

4. hUC-MSCs Isolation and Co-culture with HK-2 cells

RI PT

atmosphere at 37°C. Cells were incubated in BSA (200 µg/mL) or AOPP (200 µg/mL)

We isolated hUC-MSCs using an adherent tissue method[23]. Briefly, a 10 cm

M AN U

umbilical cord from a full-term healthy newborn was washed with PBS (containing 1% penicillin-streptomycin double-resistant solution) 3 times. Then, we cut the cord into small pieces, dislodged the umbilical vein and umbilical artery, and left Wharton's

TE D

jelly at last. Wharton's jelly was then cut into 1 mm × 1 mm × 1 mm and cultured in DMEM/F12 medium containing 5% FBS; 3-6 passages (P3-6) were selected for the following experiments. We used a co-culture chamber to block the immediate contact

EP

between the hUC-MSCs and HK-2 cells to explore the paracrine action of hUC-MSCs

AC C

in the co-culture system.

5. Flow Cytometry and hUC-MSCs Identification hUC-MSCs were trypsinized, and the cell concentration was adjusted to 1×106/mL

in PBS. Then, 200 µl of the suspension was incubated with 5 µl of antibodies against CD90, CD105, CD73, CD34, CD45, CD11b, CD14, and HLA-DR without light for 30 min. Primary antibodies were directly conjugated with FITC and phycoerythrin. For isotype control, non-specific FITC-conjugated IgG was substituted for the

ACCEPTED MANUSCRIPT primary antibodies. Lastly, the samples were analyzed using flow cytometry.

6. RNA Extraction and Quantitative Real-time PCR (RT-qPCR) Analysis

RI PT

Total RNA was extracted from treated HK-2 cells using TRIzol reagent (TaKaRa, Japan). 1 µg RNA was reverse transcribed using the MMLV reverse transcriptase kit according to the manufacturer’s instructions (TaKaRa, Japan). The U6 and miR-145

SC

primers were provided directly by RiboBio Company and the sequences were kept

U6.

7. Western Blotting Analysis

M AN U

secreted by RiboBio Company. All data were normalized using the internal control

We extracted total protein using RIPA lysis buffer. Then, 20-50 µg of protein was

TE D

separated using SDS-PAGE and transferred to PVDF membranes. Subsequently, the membranes were blocked in 5% nonfat milk or BSA at room temperature for 2 h,

EP

followed by incubation of the primary antibodies at 4°C overnight. The primary antibodies were as follows: anti-LC3B, anti-Beclin 1, anti-p62, anti-p-mTOR,

AC C

anti-mTOR, anti-p-AKT, anti-AKT, anti-p-PI3K (dilution, 1:1000) and anti-GAPDH (dilution,

1:5000).

The membranes

were

incubated

with

the

appropriate

HRP-conjugated secondary antibodies at room temperature for 2 h and were finally detected using an enhanced chemiluminescence (ECL) system. GAPDH was used as the internal control and semiquantitative analysis was performed using the ImageJ system.

8. Immunofluorescence Staining

ACCEPTED MANUSCRIPT Cells plated in 96-well plates were fixed with 4% paraformaldehyde, permeabilized with 0.5% Triton X-100 for 10 minutes and incubated in a blocking buffer containing 5% BSA for 30 min at room temperature. Then, cells were incubated with LC3B Finally, we incubated cells with

RI PT

antibody (1:50 dilution) overnight at 4°C.

fluorescently labeled secondary antibodies (Alexa Fluor 488, 1:400, Gongsi) for 1 h at room temperature in the dark, followed by incubation with 0.1% DAPI for 10 min.

SC

We used an inverted fluorescence microscope to observe and record the

M AN U

LC3B-positive staining.

9. Transmission Electron Microscopy (TEM)

HK-2 cells were gently harvested using trypsin with EDTA and were centrifuged at a speed

TE D

of 1000 r/m for 5 min. Then, the sediment was washed twice with cold PBS and fixed in 2.5% glutaraldehyde for 2 h at 4°C. Cells were conventionally dehydrated, embedded, sliced into 60 nm sections and stained with uranyl acetate for 15 min at room temperature, followed by

EP

lead citrate for 15 min at room temperature. Autophagosome (AP) and autolysosome (AL)

AC C

formation were observed using TEM at a magnification of low-power field (x6000, x8000, x10000) or high-power field (x40000) operating at 60 kV.

10. HK-2 Cell Transfection And hUC-MSC Transfection

HK-2 cells were transfected with miR-145 siRNA, miR-145 mimics, or negative siRNA, and hUC-MSCs was transfected with miR-145 siRNA, using Lipofectamine 2000 (Invitrogen, CA, USA) according to the manufacturer’s instructions. Targeting sequences were as follows: miR-145 siRNA: sense (5’-

ACCEPTED MANUSCRIPT AGGGAUUCCUGGGAAAACUGGAC-3’); miR-145 mimics: sense (5’GUCCAGUUUUCCCAGGAAUCCCU-3’), antisense (5’GGAUUCCUGGGAAAACUGGACUU-3’); negative siRNA: sense

(5’-ACGUGACACGUUCGGAGAATT-3’).

SC

11. Statistical Analysis

RI PT

(5’-UUCUCCGAACGUGUCACGUTT-3’), antisense

Statistical analyses were performed using SPSS 20.0 software. Continuous

M AN U

variables are presented as the mean ± SD. One-way ANOVA was carried out to determine differences among groups. For the comparison of 2 groups, the LSD method was used when the assumption of variance was equal. Otherwise, the Dunnett

EP

Results

TE D

T3 method was used. P values < 0.05 indicated a statistically significant difference.

1. Identification results of hUC-MSCs

AC C

We chose P3 hUC-MSCs to identify the cell phenotype by flow cytometry.

Inverted phase contrast microscopy showed that P4 hUC-MSCs presented 50% fusion and 100% vortex fusion, as shown in in Figure 1A and 1B. Flow cytometry analysis of the cell phenotype showed that the hUC-MSCs were positive for CD90, CD105, and CD73, but were negative for CD34, CD45, CD11b, CD14, and HLA-DR (Figure 1C-J).

ACCEPTED MANUSCRIPT 2. miR-145 was upregulated in HK-2 cells under hUC-MSCs co-cultured system We co-cultured hUC-MSCs with HK-2 cells and used RT-qPCR to measure the expression of miR-145 in HK-2 cells. The result showed that when HK-2 cells were

RI PT

co-cultured with hUC-MSCs, the mRNA level of miR-145 was significantly upregulated. Next, we knocked down miR-145 in the hUC-MSCs and then

SC

co-cultured it with HK-2 cells. We found that in the later co-culture system, miR-145 did not increased in HK-2 cells (Figure 2). The data indicated that hUC-MSCs

M AN U

secreted miR-145 and transferred it to HK-2 cells through paracrine action.

3. miR-145 affected HK-2 cell autophagy

We first downregulated or upregulated the expression of miR-145 in HK-2 cells

TE D

and examined the mRNA level of miR-145. The RT-qPCR results showed that miR-145 siRNA downregulated miR-145; however, miR-145 mimics upregulated miR-145 in HK-2 cells. At the same time, the expression of miR-145 showed no

EP

difference between negative siRNA and the normal control group (Figure 3A).

AC C

We then assayed the autophagy-related proteins LC3B, Beclin 1, and p62 by

western blotting. The results in figure 3(B-C) showed that when we inhibited the expression of miR-145 by siRNA, the protein levels of LC3 II and Beclin 1 decreased and the protein level of p62 increased. However, when we upregulated the expression of miR-145, the protein levels of those markers mentioned above had the opposite effect. In addition, there was no difference between negative siRNA and the normal control group.

ACCEPTED MANUSCRIPT Furthermore, the autophagic activity of HK-2 cells was assessed using immunofluorescence technology and TEM. LC3B-positive staining was significantly decreased in cells transfected with miR-145 siRNA but increased in cells transfected

RI PT

with miR-145 mimics compared with that of the normal control group (Figure 3D). Similarly, the TEM results demonstrated a decrease in the number of typical Aps (double membrane vacuoles engulfing cytoplasmic structures) and ALs (single

SC

membrane vacuole structures containing high density materials) in miR-145 siRNA

M AN U

transfected cells but revealed an increase in the number of Aps and ALs in miR-145 transfected cells compared with those in the normal control group (Figure 3E). Collectively, these results suggested that overexpression of miR-145 activated HK-2 cell autophagy.

cells

TE D

4. miR-145 mediated hUC-MSCs enhanced-autophagy in AOPP-treated HK-2

EP

To further confirm that hUC-MSCs-enhanced autophagy was mediated by the

AC C

secretion of miR-145, we transfected HK-2 cells with miR-145 siRNA after AOPP-treated HK-2 cells were co-cultured with hUC-MSCs to investigate the effect of hUC-MSCs on HK-2 cells. We first measured the miR-145 expression in transfected and co-culture system. RT-qPCR analysis showed that the level of miR-145 was increased in the hUC-MSCs and AOPP co-culture group compared with that in the normal control group, but the miR-145 level was decreased in the transfected and co-culture system compared with the hUC-MSCs and AOPP co-culture group (Figure 4A).

ACCEPTED MANUSCRIPT Next, we detected the autophagous activity after transfection with miR-145 siRNA in the hUC-MSCs and AOPP co-culture system. The western blotting results showed that when we decreased the expression of miR-145 by siRNA, the protein levels of

those in

the hUC-MSCs

co-culture system

RI PT

LC3 II and Beclin 1 decreased and the p62 protein level increased compared with (Figure 4B-C).

Furthermore,

LC3B-positive staining was significantly decreased in cells transfected with miR-145

SC

siRNA compared with that in the hUC-MSCs co-culture system (Figure 4D).

M AN U

Similarly, the TEM results demonstrated a decrease in the numbers of typical Aps and ALs in HK-2 cells in siRNA transfected with the hUC-MSCs co-culture system compared with those in the hUC-MSCs co-culture system alone (Figure 4E). Taken together, these results indicated that miR-145 secreted from hUC-MSCs enhanced

5.

TE D

HK-2 cell autophagy.

miR-145 mediated hUC-MSCs enhanced-autophagy in HK-2 cells by

EP

inhibiting the PI3K/AKT/mTOR signaling pathway

AC C

To verify the underlying pathway, we detected PI3K/AKT/mTOR signaling pathway related proteins after regulating miR-145 expression in HK-2 cells or transfection with miR-145 siRNA in the hUC-MSCs co-culture system. The western blotting results showed that when we decreased the expression of miR-145 by siRNA, the phosphorylation of PI3K, AKT, and mTOR was induced. However, when we upregulated the expression of miR-145 by mimics, the phosphorylation of PI3K, AKT, and mTOR was inhibited. In addition, there was no difference between negative siRNA and the normal control group (Figure 5A-B). Finally, transfection with

ACCEPTED MANUSCRIPT miR-145 siRNA in the hUC-MSCs co-culture system promoted the phosphorylation of PI3K, AKT, and mTOR, which was inhibited by hUC-MSCs (Figure 5C-D). We concluded that miR-145 mediated hUC-MSCs enhanced-autophagy in HK-2 cells via

RI PT

inhibiting the PI3K/AKT/mTOR signaling pathway.

SC

Discussion

M AN U

The present study demonstrated the significant role of hUC-MSCs in HK-2 cell autophagy in AKI. We provided evidence that hUC-MSCs increased HK-2 cell autophagy via secretion of miR-145 through inhibition of the PI3K/AKT/mTOR signaling pathway. The current results implicate hUC-MSCs as a promising

TE D

therapeutic strategy in protecting AKI through its paracrine action.

A considerable number of studies have focused on the role of autophagy in renal

EP

diseases. An increasing number of findings have highlighted that autophagy is rapidly induced during AKI to protect tubular cells from injury and death, and

AC C

autophagy-related gene deficiency leads to impaired renal function and tubular cell apoptosis and necrosis in conditional knockout mouse models [24] [25]. However, our study found that AOPP inhibited HK-2 cell autophagy and induced HK-2 cell injury instead of inducing autophagy to protect cells [10]. Whether the noxious stimuli induce or inhibit cell autophagy may depend on the tissue, magnitude, time or other factors that cells suffer. Livingston and colleagues [26] summarized a similar opinion, proposing that it was very important to monitor autophagy in the correct tissue at the

ACCEPTED MANUSCRIPT correct time to determine the optimal conditions and therapeutic window, thus ensuring that autophagy induction would yield protective effects in renal diseases. Moreover, given the evidence that autophagy plays an important role in the

RI PT

development and severity of AKI, targeting autophagy could be a novel and potential strategy for the treatment of AKI and even other renal diseases. In this study, we discovered a new mechanism under autophagy regulation by hUC-MSCs in HK-2

SC

cells.

M AN U

The first key finding of this study was that miR-145 was upregulated in hUC-MSCs and HK-2 cells in a co-culture system. It is general knowledge that MSCs demonstrate paracrine mechanisms, including the secretion of various nutrition factors, proteins, mRNA, or miRNA. In this way, MSCs directly or indirectly induce regeneration,

TE D

rescuing injured cells and accelerating organ repair [27]. We co-cultured hUC-MSCs with HK-2 cells and found that miR-145 expression was upregulated in HK-2 cells in

EP

this system. We then knocked down miR-145 in the hUC-MSCs before it co-cultured with HK-2 cells and found that miR-145 did not increased in HK-2 cells, which

AC C

indicated that hUC-MSCs secreted miR-145 and transported it to HK-2 cells through paracrine action. Previous studies showed that MSCs delivered exogenous miRNAs to neural cells [28] or myofibroblasts [18]. In addition, it has been proposed that MSC-derived extracellular vesicles protect against renal injury [29] [30]. However, the underlying mechanism of MSC treatment on renal diseases and whether the upregulated miR-145 in HK-2 cells derived from hUC-MSCs contribute to the renal protection are unclear.

ACCEPTED MANUSCRIPT The second crucial finding of this study was that regulation of miR-145 affected HK-2 cell autophagy. Currently, may studies have indicated that abnormal expression of miRNA participates in the regulation and progression of various renal diseases,

RI PT

such as AKI[31], diabetic nephropathy[32], and IgA nephropathy [33]. Furthermore, multiple studies have previously demonstrated that miRNA affects renal function or the pathology and physiological of renal cells through various mechanisms. Zou et al.

SC

[34] demonstrated that miRNA mediated epithelial mesenchymal transition in fibrotic

M AN U

diseases, including renal fibrosis. Dewanjee et al. [35] concluded in their review that miRNA regulated various molecular pathways in diabetic nephropathy, including the TGF-β signaling pathway, PI3K/AKT signaling pathway, and collagen gene expression. To investigate whether miR-145 influences HK-2 cell autophagy, we first

TE D

downregulated or upregulated the expression of miR-145 in HK-2 cells. The results showed that when we inhibited the expression of miR-145 by siRNA, the protein levels of LC3 II and Beclin 1 decreased and the protein level of p62 increased and

EP

phosphorylation of PI3K, AKT, and mTOR increased, but the expression of these

AC C

markers showed the opposite change when we upregulated the overexpressed miR-145 with mimics. In addition, LC3B-positive staining and the numbers of typical Aps and ALs were significantly decreased in cells transfected with miR-145 siRNA but increased in cells transfected with miR-145 mimics compared with those in the normal control group. Collectively, these results suggested that overexpression of miR-145 activated HK-2 cell autophagy and that miR-145 knockdown inhibited HK-2 cell autophagy. It was reported that miR-145 repaired myocardium by accelerating

ACCEPTED MANUSCRIPT cardiomyocyte autophagy [20]. There was also another investigation showing that miRNA-145 promoted osteosarcoma cell autophagy by targeting HDAC4 [36]. In addition, there was a study showing that upregulation of miR-145 induced inhibition

RI PT

of the PI3K/AKT/mTOR pathway in laryngeal squamous cell carcinoma[21]. Excitingly, our study was the first time to show that miR-145 affects RTEC

SC

autophagy by inhibiting PI3K/AKT/mTOR signaling pathway.

Finally, we confirmed that hUC-MSCs enhanced HK-2 cell autophagy mediated by

M AN U

the secretion of miR-145 through inhibiting the PI3K/AKT/mTOR signaling pathway. In our previous investigation, we proposed that hUC-MSCs enhanced autophagy in AOPP-treated HK-2 cells, but the underlying mechanism requires further investigation. Accumulating evidence suggests that MSCs enhance autophagy through

TE D

various classical pathways or molecules. Li et al. [37] showed that bone marrow-derived MSCs enhanced autophagy via PI3K/AKT signaling to reduce the

EP

severity of ischaemia/reperfusion-induced lung injury. Liu et al. [38] suggested that MSC induced cardiomyocyte autophagy via the AMPK and AKT signaling pathways

AC C

to rescue myocardial ischaemia/reperfusion injury. To our knowledge, MSCs primarily possess paracrine or endocrine functions, which allow them to play a protective role. Wang et al. [39] indicated that hUC-MSCs-derived exosomes increased the expression of the autophagic marker protein LC3B and the autophagy-related genes ATG5 and ATG7 in cisplatin-treated NRK-52E cells to relieve the nephrotoxicity of cisplatin. In addition, Jia et al. [40] presented a similar conclusion in another study, showing that hUC-MSCs-derived exosomes enhanced

ACCEPTED MANUSCRIPT autophagy via transmission of 14-3-3zeta and modulation of ATG16L in preventing cisplatin-induced AKI. Since paracrine secretion of miR-145 from hUC-MSCs is delivered to HK-2 cells, to further uncover whether hUC-MSCs enhance autophagy

RI PT

through secretion of miR-145, we transfected HK-2 cells with miR-145 siRNA after AOPP-treated HK-2 cells were co-cultured with hUC-MSCs. We found that when we decreased the expression of miR-145 by siRNA, the protein levels of LC3 II and

SC

Beclin 1 decreased, the p62 protein level increased, the LC3B-positive staining and

M AN U

numbers of typical Aps and ALs decreased, and the phosphorylation of PI3K, AKT, and mTOR increased compared with those in hUC-MSCs co-culture system. Taken together, in this study, we presented for the first time a new mechanism of hUC-MSCs-enhanced cell autophagy by secreting miR-145 through inhibition of the

TE D

PI3K/AKT/mTOR signaling pathway.

We did not explore how hUC-MSCs transported secreted miR-145 to HK-2 to

EP

enhance cell autophagy. Previous studies found that MSCs transported RNA or protein to target cells through MSCs-derived exosomes to repair damaged cells and

AC C

tissues[41]. Future in-depth studies will explore whether exosomes act as the mechanism of hUC-MSCs on HK-2 cells. Additionally, the amount of miR-145 expressed in transfection is not the same with that secreted by hUC-MSCs, and we will further regulate and investigate the effects of different dosage of miR-145 secreted by hUC-MSCs on autophagy in HK-2 cells. Finally, further in vitro studies are warranted to validate the significance of hUC-MSCs and secreted miR-145 in the enhancement of autophagy in HK-2 cells and primary cells and even in AKI models.

ACCEPTED MANUSCRIPT

Conclusion

RI PT

In conclusion, these results indicated that hUC-MSCs played an autophagy enhancement role via overexpression of miR-145, which provided a novel mechanism for the investigation of hUC-MSCs in AKI treatment. MSCs represent an amazing

SC

therapeutic tool in regenerative medicine; therefore, developing a better understanding

kidney diseases.

Conflict of interest statement

EP

TE D

None.

M AN U

of the mechanism of MSCs in renal protection is a promising therapeutic strategy for

Acknowledgements

AC C

The present study was supported by the Natural Science Foundation of Guangdong Province (grant no. 2018A030313557).

References [1]

R. Mehta, J. Cerdá, E. Burdmann, M. Tonelli, G. García-García, V. Jha, P. Susantitaphong, M. Rocco, R. Vanholder, M. Sever, D. Cruz, B. Jaber, N. Lameire, R. Lombardi, A. Lewington, J. Feehally, F. Finkelstein, N. Levin, N. Pannu, B. Thomas, E. Aronoff-Spencer, G. Remuzzi, International Society of Nephrology's 0by25 initiative for acute kidney injury (zero

ACCEPTED MANUSCRIPT preventable deaths by 2025): a human rights case for nephrology, Lancet 385 (2015) 2616-2643. [2]

L. Chawla, P. Eggers, R. Star, P. Kimmel, Acute kidney injury and chronic kidney disease as interconnected syndromes, N. Engl. J. Med. 371 (2014) 58-66.

[3]

A. Ishani, J. Xue, J. Himmelfarb, P. Eggers, P. Kimmel, B. Molitoris, A. Collins, Acute kidney

[4]

RI PT

injury increases risk of ESRD among elderly, J. Am. Soc. Nephrol. 20 (2009) 223-228.

O. Lenoir, M. Jasiek, C. Hénique, L. Guyonnet, B. Hartleben, T. Bork, A. Chipont, K. Flosseau, I. Bensaada, A. Schmitt, J. Massé, M. Souyri, T. Huber, P. Tharaux, Endothelial cell and podocyte

(2015) 1130-1145. [5]

O. Lenoir, P. Tharaux, T. Huber, Autophagy in kidney disease and aging: lessons from rodent

M AN U

models, Kidney Int. 90 (2016) 950-964. [6]

SC

autophagy synergistically protect from diabetes-induced glomerulosclerosis, Autophagy 11

S. Mackensen-Haen, R. Bader, K. Grund, A. Bohle, Correlations between renal cortical interstitial fibrosis, atrophy of the proximal tubules and impairment of the glomerular filtration rate, Clin. Nephrol. 15 (1981) 167-171.

[7]

X. Guan, Y. Qian, Y. Shen, L. Zhang, Y. Du, H. Dai, J. Qian, Y. Yan, Autophagy protects renal tubular cells against ischemia / reperfusion injury in a time-dependent manner, Cell. Physiol.

[8]

TE D

Biochem. 36 (2015) 285-298.

T. Kimura, A. Takahashi, Y. Takabatake, T. Namba, T. Yamamoto, J. Kaimori, I. Matsui, H. Kitamura, F. Niimura, T. Matsusaka, T. Soga, H. Rakugi, Y. Isaka, Autophagy protects kidney

EP

proximal tubule epithelial cells from mitochondrial metabolic stress, Autophagy 9 (2013) 1876-1886. [9]

D. Heras-Sandoval, J.M. Perez-Rojas, J. Hernandez-Damian, J. Pedraza-Chaverri, The role of

AC C

PI3K/AKT/mTOR pathway in the modulation of autophagy and the clearance of protein aggregates in neurodegeneration, Cellular signalling 26 (2014) 2694-2701.

[10]

J. Zhang, X. Xiang, S. Shu, C. Zhang, Y. Liang, T. Jiang, W. Zhang, T. Guo, X. Liang, X. Tang,

Advanced oxidation protein products inhibit the autophagy of renal tubular epithelial cells, Exp Ther Med 15 (2018) 3908-3916.

[11]

M. Detamore, Human umbilical cord mesenchymal stromal cells in regenerative medicine, Stem Cell Res Ther 4 (2013) 142.

[12]

D. Papazova, N. Oosterhuis, H. Gremmels, A. van Koppen, J. Joles, M. Verhaar, Cell-based therapies for experimental chronic kidney disease: a systematic review and meta-analysis, Dis

ACCEPTED MANUSCRIPT Model Mech 8 (2015) 281-293. [13]

T. Griffin, W. Martin, N. Islam, T. O'Brien, M. Griffin, The Promise of Mesenchymal Stem Cell Therapy for Diabetic Kidney Disease, Curr. Diab. Rep. 16 (2016) 42.

[14]

F. Casiraghi, N. Perico, M. Cortinovis, G. Remuzzi, Mesenchymal stromal cells in renal

[15]

RI PT

transplantation: opportunities and challenges, Nat Rev Nephrol 12 (2016) 241-253. S. Tian, B. Feldman, J. Beyene, P. Brown, E. Uleryk, E. Silverman, Immunosuppressive

Therapies for the Maintenance Treatment of Proliferative Lupus Nephritis: A Systematic Review and Network Metaanalysis, J. Rheumatol. 42 (2015) 1392-1400. [16]

M. Khubutiya, A. Vagabov, A. Temnov, A. Sklifas, Paracrine mechanisms of proliferative,

acute organ injury, Cytotherapy 16 (2014) 579-585.

A. Caplan, J. Dennis, Mesenchymal stem cells as trophic mediators, J. Cell. Biochem. 98 (2006)

M AN U

[17]

1076-1084. [18]

SC

anti-apoptotic and anti-inflammatory effects of mesenchymal stromal cells in models of

S. Fang, C. Xu, Y. Zhang, C. Xue, C. Yang, H. Bi, X. Qian, M. Wu, K. Ji, Y. Zhao, Y. Wang, H. Liu, X. Xing, Umbilical Cord-Derived Mesenchymal Stem Cell-Derived Exosomal MicroRNAs Suppress Myofibroblast Differentiation by Inhibiting the Transforming Growth Factor-β/SMAD2

[19]

TE D

Pathway During Wound Healing, Stem Cells Transl Med 5 (2016) 1425-1439. H. Lee, S. Finniss, S. Cazacu, E. Bucris, A. Ziv-Av, C. Xiang, K. Bobbitt, S. Rempel, L. Hasselbach, T. Mikkelsen, S. Slavin, C. Brodie, Mesenchymal stem cells deliver synthetic microRNA mimics to glioma cells and glioma stem cells and inhibit their cell migration and self-renewal,

[20]

EP

Oncotarget 4 (2013) 346-361.

K. Higashi, Y. Yamada, S. Minatoguchi, S. Baba, M. Iwasa, H. Kanamori, M. Kawasaki, K. Nishigaki, G. Takemura, M. Kumazaki, Y. Akao, S. Minatoguchi, MicroRNA-145 repairs

AC C

infarcted myocardium by accelerating cardiomyocyte autophagy, Am. J. Physiol. Heart Circ. Physiol. 309 (2015) H1813-1826.

[21]

X. Zhu, R. Zhu, Curcumin suppresses the progression of laryngeal squamous cell carcinoma

through the upregulation of miR-145 and inhibition of the PI3K/Akt/mTOR pathway, OncoTargets and therapy 11 (2018) 3521-3531.

[22]

X. Tang, G. Rong, Y. Bu, S. Zhang, M. Zhang, J. Zhang, X. Liang, Advanced oxidation protein products induce hypertrophy and epithelial-to-mesenchymal transition in human proximal tubular cells through induction of endoplasmic reticulum stress, Cell. Physiol. Biochem. 35 (2015) 816-828.

ACCEPTED MANUSCRIPT [23]

李艳琪, 王洪一, 姚尧, 刘晶晶, 徐潇, 张宇, 刘洋, 吴祖泽, 靳继德, 人脐带源间充质干 细胞分离培养方法的改进, 中国组织工程研究 18 (2014) 1609-1614.

[24]

S. Liu, B. Hartleben, O. Kretz, T. Wiech, P. Igarashi, N. Mizushima, G. Walz, T. Huber, Autophagy plays a critical role in kidney tubule maintenance, aging and ischemia-reperfusion injury, Autophagy 8 (2012) 826-837. T. Kimura, Y. Takabatake, A. Takahashi, J. Kaimori, I. Matsui, T. Namba, H. Kitamura, F. Niimura,

RI PT

[25]

T. Matsusaka, T. Soga, H. Rakugi, Y. Isaka, Autophagy protects the proximal tubule from degeneration and acute ischemic injury, J. Am. Soc. Nephrol. 22 (2011) 902-913.

M. Livingston, Z. Dong, Autophagy in acute kidney injury, Semin. Nephrol. 34 (2014) 17-26.

[27]

S. Keshtkar, N. Azarpira, M. Ghahremani, Mesenchymal stem cell-derived extracellular

SC

[26]

vesicles: novel frontiers in regenerative medicine, Stem Cell Res Ther 9 (2018) 63. H. Lee, S. Finniss, S. Cazacu, C. Xiang, C. Brodie, Mesenchymal stem cells deliver exogenous

M AN U

[28]

miRNAs to neural cells and induce their differentiation and glutamate transporter expression, Stem Cells Dev. 23 (2014) 2851-2861. [29]

X. Zou, D. Gu, X. Xing, Z. Cheng, D. Gong, G. Zhang, Y. Zhu, Human mesenchymal stromal cell-derived extracellular vesicles alleviate renal ischemic reperfusion injury and enhance

[30]

TE D

angiogenesis in rats, Am J Transl Res 8 (2016) 4289-4299.

G. Zhang, X. Zou, Y. Huang, F. Wang, S. Miao, G. Liu, M. Chen, Y. Zhu, 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 (2016) 119-128. K. Ramachandran, J. Saikumar, V. Bijol, J. Koyner, J. Qian, R. Betensky, S. Waikar, V. Vaidya,

EP

[31]

Human miRNome profiling identifies microRNAs differentially present in the urine after

AC C

kidney injury, Clin. Chem. 59 (2013) 1742-1752. [32]

F. He, F. Peng, X. Xia, C. Zhao, Q. Luo, W. Guan, Z. Li, X. Yu, F. Huang, MiR-135a promotes renal

fibrosis in diabetic nephropathy by regulating TRPC1, Diabetologia 57 (2014) 1726-1736.

[33]

H. Bao, H. Chen, X. Zhu, M. Zhang, G. Yao, Y. Yu, W. Qin, C. Zeng, K. Zen, Z. Liu, MiR-223

downregulation promotes glomerular endothelial cell activation by upregulating importin α4 and α5 in IgA nephropathy, Kidney Int. 85 (2014) 624-635.

[34]

X. Zou, T. Liu, Z. Gong, C. Hu, Z. Zhang, MicroRNAs-mediated epithelial-mesenchymal transition in fibrotic diseases, Eur. J. Pharmacol. 796 (2017) 190-206.

[35]

S. Dewanjee, N. Bhattacharjee, MicroRNA: A new generation therapeutic target in diabetic nephropathy, Biochem. Pharmacol. 155 (2018) 32-47.

ACCEPTED MANUSCRIPT [36]

G. Wu, W. Yu, M. Zhang, R. Yin, Y. Wu, Q. Liu, MicroRNA-145-3p suppresses proliferation and promotes apotosis and autophagy of osteosarcoma cell by targeting HDAC4, Artif Cells Nanomed Biotechnol (2018) 1-8.

[37]

J. Li, J. Zhou, D. Zhang, Y. Song, J. She, C. Bai, Bone marrow-derived mesenchymal stem cells enhance autophagy via PI3K/AKT signalling to reduce the severity of

[38]

RI PT

ischaemia/reperfusion-induced lung injury, J. Cell. Mol. Med. 19 (2015) 2341-2351. L. Liu, X. Jin, C. Hu, R. Li, Z. Zhou, C. Shen, Exosomes Derived from Mesenchymal Stem Cells

Rescue Myocardial Ischaemia/Reperfusion Injury by Inducing Cardiomyocyte Autophagy Via AMPK and Akt Pathways, Cell. Physiol. Biochem. 43 (2017) 52-68.

B. Wang, H. Jia, B. Zhang, J. Wang, C. Ji, X. Zhu, Y. Yan, L. Yin, J. Yu, H. Qian, W. Xu,

SC

[39]

Pre-incubation with hucMSC-exosomes prevents cisplatin-induced nephrotoxicity by

[40]

M AN U

activating autophagy, Stem Cell Res Ther 8 (2017) 75.

H. Jia, W. Liu, B. Zhang, J. Wang, P. Wu, N. Tandra, Z. Liang, C. Ji, L. Yin, X. Hu, Y. Yan, F. Mao, X. Zhang, J. Yu, W. Xu, H. Qian, HucMSC exosomes-delivered 14-3-3ζ enhanced autophagy via modulation of ATG16L in preventing cisplatin-induced acute kidney injury, Am J Transl Res 10 (2018) 101-113.

[41]

C. Merino-Gonzalez, F.A. Zuniga, C. Escudero, V. Ormazabal, C. Reyes, E. Nova-Lamperti, C.

TE D

Salomon, C. Aguayo, Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote

AC C

EP

Angiogenesis: Potencial Clinical Application, Frontiers in physiology 7 (2016) 24.

Figure legends

SC

Fig. 1 hUC-MSCs identification

RI PT

ACCEPTED MANUSCRIPT

A: Inverted phase contrast microscope images (× 50) showing that P4 hUC-MSCs presented 50%

M AN U

fusion; B: Inverted phase contrast microscope images (× 50) showing that P4 hUC-MSCs presented 100% vortex fusion; C-J: Flow cytometry showed that hUC-MSCs were positive for CD90, CD105,

AC C

EP

TE D

and CD73, but were negative for CD34, CD45, CD11b, CD14, and HLA-DR.

SC

RI PT

ACCEPTED MANUSCRIPT

M AN U

Fig. 2 Detection of miR-145 in HK-2 cells in the hUC-MSCs co-culture system

The RT-qPCR results showed that miR-145 expression was upregulated in HK-2 cells in the hUC-MSCs co-culture system (hUC-MSCs group). Next, we knocked down miR-145 in the hUC-MSCs and then co-cultured it with HK-2 cells. We found that in the later co-culture system

TE D

(hUC-MSCs siRNA group), miR-145 did not increased in HK-2 cells. Data are expressed as the means

AC C

EP

± SD from 3 independent experiments; *P < 0.05 vs. Ctrl group. Ctrl: normal control.

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

Fig. 3 miR-145 affected HK-2 cell autophagy.

We inhibited or promoted the expression of miR-145 in HK-2 cells and detected autophagy related

TE D

markers. A: The RT-qPCR results showed that miR-145 siRNA downregulated miR-145 but miR-145 mimics upregulated miR-145 in HK-2 cells. B-C: Western blotting results showed that LC3 II and

EP

Beclin 1 were decreased and p62 was increased in the miR-145 siRNA group, but the opposite trend was found in the miR-145 mimics group. D: Immunofluorescence result showed that LC3B-positive

AC C

staining decreased in cells transfected with miR-145 siRNA but increased in cells transfected with miR-145 mimics. E: TEM experiments revealed that typical Aps and ALs were significantly decreased in cells transfected with miR-145 siRNA but increased in cells transfected with miR-145 mimics (the above is the low-power field, x8000, or x10000, and the below is the high-power field, x40000). Data are expressed as the means ± SD from 3 independent experiments; *P < 0.05 vs. Ctrl group. Ctrl: normal control.

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

Fig. 4 hUC-MSCs enhanced autophagy in AOPP-treated HK-2 cells by secretion of miR-145.

We transfected HK-2 cells with miR-145 siRNA in the hUC-MSCs and AOPP co-culture system. A: RT-qPCR analysis showed that the increased miR-145 secreted from hUC-MSCs was partially decreased in the miR-145 siRNA transfected and co-culture system compared with that in the

ACCEPTED MANUSCRIPT hUC-MSCs and AOPP co-culture group. B-C: Western blotting showed that LC3 II and Beclin 1 was decreased and p62 was increased when HK-2 cells were transfected with miR-145 siRNA compared with those in the hUC-MSCs co-culture system. D: Immunofluorescence showed that LC3B-positive

RI PT

staining was decreased in cells transfected with miR-145 siRNA. E: TEM experiments presented that typical Aps and ALs were significantly decreased in cells transfected with miR-145 siRNA (the above is the low-power field, x6000, or x8000, and the below is the high-power field, x40000). Data are

SC

expressed as the means ± SD from 3 independent experiments; *P < 0.05 vs. AOPP group; # P < 0.05

AC C

EP

TE D

M AN U

vs. AOPP+hUC-MSCs group.

RI PT

ACCEPTED MANUSCRIPT

Fig. 5 miR-145 mediated hUC-MSCs enhanced-autophagy in HK-2 cells by inhibiting the

SC

PI3K/AKT/mTOR signaling pathway.

We regulated miR-145 expression in HK-2 cells and measured PI3K/AKT/mTOR signaling pathway

M AN U

related proteins. A-B: Western blotting results showed that when we decreased the expression of miR-145 by siRNA, phosphorylation of PI3K, AKT, and mTOR was induced, but was inhibited when we upregulated the expression of miR-145 by mimics. C-D; Western blotting results indicated that transfection with miR-145 siRNA in the hUC-MSCs co-culture system partially rescued the

TE D

phosphorylation of PI3K, AKT, and mTOR, which was inhibited by hUC-MSCs. Data are expressed as the means ± SD from 3 independent experiments; *P < 0.05 vs. Ctrl group (A, B) or AOPP group (C,

AC C

EP

D); # P < 0.05 vs. AOPP+hUC-MSCs group.

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT