• No results found

MicroRNA-9 modified bone marrow-derived mesenchymal stem cells (BMSCs) repair severe acute pancreatitis (SAP) via inducing angiogenesis in rats

N/A
N/A
Protected

Academic year: 2020

Share "MicroRNA-9 modified bone marrow-derived mesenchymal stem cells (BMSCs) repair severe acute pancreatitis (SAP) via inducing angiogenesis in rats"

Copied!
15
0
0

Loading.... (view fulltext now)

Full text

(1)

R E S E A R C H

Open Access

MicroRNA-9 modified bone marrow-derived

mesenchymal stem cells (BMSCs) repair

severe acute pancreatitis (SAP) via inducing

angiogenesis in rats

Daohai Qian

1,2,3

, Guodong Song

2

, Zhilong Ma

2

, Guannan Wang

1

, Lei Jin

4

, Minghua Hu

1

, Zhenshun Song

2*

and Xiaoming Wang

1*

Abstract

Background:Severe acute pancreatitis (SAP) is an acute abdominal disease characterized by pancreatic necrosis and systemic disease. In a previous study, we showed that bone marrow-derived mesenchymal stem cells (BMSCs) can reduce SAP by secreting microRNA (miR)-9; however, the underlying mechanism remains unclear. The present study investigated the mechanism underlying BMSC-induced pancreatic regeneration.

Methods:BMSCs were isolated, and miR-9 modified/antagonized BMSCs (pri-miR-9-BMSCs/TuD-BMSCs) were generated and injected into SAP rats. The levels of inflammatory cytokines and histopathologic changes were examined using ELISA and H&E staining. Angiogenesis was analyzed by qRT-PCR, western blotting, and immunohistochemistry. Cell function tests, dual luciferase reporter assays, cell co-culture, western blotting, and cell tracing were used to explore the mechanisms underlying miR-9 induced angiogenesis.

Results: Pri-miR-9-BMSCs induced angiogenesis in SAP rats (Ang-1↑, TIE-2↑, and CD31↑) and repaired damaged vascular endothelial cells (VECs) in vitro, promoting angiogenesis (Ang-1↑, TIE-2↑, PI3K↑, AKT↑, p-AKT↑, CD31↑, and CD34↑). Pri-miR-9-BMSCs released miR-9 into VECs or injured pancreatic tissue, targeting the VE-cadherin gene and promoting PI3K/AKT signaling to treat SAP (VE-cadherin↓,β-catenin↓, PI3K↑, p-AKT↑), whereas antagonizing miR-9 in BMSCs did not alleviate or aggravated SAP.

Conclusions:Pri-miR-9-BMSCs can repair injured pancreatic tissue by secreting miR-9 and promoting angiogenesis.

Background

Acute pancreatitis (AP) is an acute abdominal disease [1]. Gallstone disease and excess alcohol ingestion are the most common causes of AP and are involved in > 90% of patients [2]. Approximately 10–20% of AP cases can evolve into severe acute pancreatitis (SAP), which is associated with a high rate of morbidity and mortality [2]. Although the pathogenesis of AP remains unclear,

abnormal activation of trypsinogen, which causes the self-digestion of pancreatic acinar cells, is associated with AP [2]. In addition, excessive activation of white cells and systemic inflammatory responses contribute to the occur-rence and progression of AP [2]. The release of pro-inflammatory cytokines also plays an important role in AP, and it can aggravate the local inflammatory re-sponse and give rise to systemic inflammatory rere-sponse syndrome (SIRS) and multiple organ dysfunction syn-drome (MODS) [1,2]. Understanding the pathogenesis of AP may help develop therapeutic strategies, and the in-flammatory response may become a key target for the treatment of AP [1]. However, the inflammatory response in AP is considered as a vascular reaction [3]. Vascular endothelial cells (VECs) are initially damaged by

* Correspondence:zs_song@hotmail.com;david00091@163.com

Daohai Qian, Guodong Song and Zhilong Ma are co-first authors. Daohai Qian, Guodong Song and Zhilong Ma contributed equally to this work.

2

Department of General Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai 200072, China

1Department of General Surgery, Yijishan Hospital, Wannan Medical College,

Wuhu 241001, Anhui, China

Full list of author information is available at the end of the article

(2)

pro-inflammatory cytokines, which alters cellular integrity and increases permeability, leading to microcirculatory disturbances, tissue edema, infiltration of inflammatory cells, and the release of pro-inflammatory cytokines [3,4]. Therefore, the repair of injured blood vessels contributes to decreasing the local/systemic inflammatory response and improves the local/systemic microcirculation [5]. Injury to blood vessels occurs before the development of AP [3]. Therefore, the development of methods to repair injured blood vessels has become a research hot-spot and could be a new target for the treatment of AP. An adequate blood supply provides essential nutrients to pancreatic cells, which is important for supporting metabolism and growth [4, 6]. In addition, the injured pancreas can be repaired in the presence of an adequate blood supply to support the self-renewal of pancreatic cells [4]. Tissue regeneration associated with anti- and pro-angiogenic signaling pathways mainly depends on the formation of new blood vessels, which is mediated by a complex process [7]. The PI3K/AKT signaling path-way, which can promote the proliferation and migration of VECs to trigger angiogenesis, has been investigated extensively [8–10]. Conversely, the VE-cadherin-catenin complex can strongly stabilize endothelial junctions against the migration of VECs, which can inhibit angio-genesis [11–13]. Cellular growth factors also play an important role in inducing angiogenesis by acting on their receptors to start downstream signal transduc-tion and promote the proliferatransduc-tion and migratransduc-tion of VECs [4]. For example, vascular endothelial growth factor (VEGF) and its receptor, VEGFR, can activate the PI3K/AKT pathway to trigger revascularization [12]. Angiopoietin-1 (ang-1), which is also related to endothelial cell survival, proliferation, and migration, can reduce endothelial permeability and promote the maturation and stability of newly formed blood vessels by interacting with the tyrosine kinase TIE-2 receptor [6, 14].

Mesenchymal stem cells (MSCs) are adult stem cells of low immunogenicity that possess specific properties such as self-renewal, multilineage differentiation, im-munosuppression, directed migration, and paracrine functions [15]. Therefore, MSCs are considered as the ideal seed cells for treating human diseases including autoimmune and liver diseases [16]. Bone marrow-de-rived mesenchymal stem cells (BMSCs) have been inves-tigated extensively. Recent studies including ours demonstrated that infused BMSCs can decrease the local systemic inflammatory response and repair injured pan-creatic tissue. Transplanted BMSCs promote the expres-sion of VEGF-A, Ang-1, HGF, and TGF-β in the damaged pancreas and induce angiogenesis [4]. How-ever, the underlying mechanism remains unknown. Des-pite extensive efforts, necrotic pancreatic tissues cannot

be completely repaired in the early stage of SAP. There-fore, exploring the potential of infused MSCs for the treatment of SAP and how to improve their curative ef-fect are important issues that need to be addressed.

MicroRNAs (miRNAs), which are endogenous noncod-ing RNAs of 18–24 nucleotides, are involved in virus defense, hematopoiesis, organ formation, cell proliferation, inflammatory responses, apoptosis, and lipid metabolism. miRNAs function by binding to the 3′untranslated region (3′-UTR) of target genes, promoting mRNA degradation or repressing protein translation [17–19]. Recent studies show that miRNAs are useful as biomarkers/targets for the diagnosis/treatment of AP [18, 19]. In our previous study, we showed that miR-9 is positively correlated with the severity of AP, and injection of miR-9 mimics acts on SAP by targeting the NF-κB1 gene and inhibiting NF-κB signaling. Studies show that miR-9 has the ability to pro-mote the migration of vascular endothelial cells and in-duce angiogenesis [20, 21]. In our previous study, we investigated that BMSCs can repair SAP through miR-9, which could inhibit the inflammatory response [17]. How-ever, we also observed that BMSCs promoted the regener-ation of the pancreas. In the present study, we investigated the effect of BMSCs on promoting the repair of necrotized pancreatic tissues and the mechanism by which miR-9-modified BMSCs induce the angiogenesis.

Methods Materials

Na-taurocholate (NaT), poly-l-lysine, nitrocellulose mem-brane, trypan blue, 4′, 6-diamidino-2-phenylindole (DAPI), polybrene, dimethyl sulfoxide (DMSO), 3-(4, 5-dimethylthiazol-2-yl)-2, S-diphenyltetrazolium bromide (MTT), lipopolysaccharide (LPS), and secondary anti-bodies were purchased from Sigma-Aldrich (Brooklyn, NY, USA). The apoptosis detection kit was from BD com-pany (Becton, Dickinson and Comcom-pany, NY, USA). The amylase and lipase activity assay kits were from Biovision (Palo Alto, California, USA). SPION (Fe2O3, 30 nm) were

from Dk Nanotechnology Company (Beijing, China). Penicillin, streptomycin, streptomycin, Cell Tracker CM-Dil, TRIzol, TRIzol LS Reagent, Lipofectamine 2000 (Lipo2000), and the Histostain-Plus Kit (DAB, Broad Spectrum) were from Invitrogen (Carlsbad, California, USA). RIPA lysis buffer, BCA protein concentration assay kit, phenylmethanesulfonyl fluoride (PMSF, 100 mM), Dulbecco’s modified Eagle’s medium–high/low glucose (DMEM-H/LG), 0.25% Trypsin-EDTA and fetal bo-vine serum (FBS) were from Gibico (Middleton, WI, USA). The Prussian Blue staining kit were from Beyo-time Biotechnology (Nantong, Jiangsu Province, China). Agarose was from Biowest (Spain). Antibodies directed against Glyceraldehyde-phosphate dehydro-genase (GAPDH), VEGFA, angiopoietin-1(Ang-1) and

(3)

TIE-2 are from ProteinTech (Wuhan, Hubei Province, China), β-catenin, VE-cadherin, PI3K, AKT, and p-AKT from CST (Danvers, MA, USA), CXCR4 from Abcam (Cambridge, MA, USA), PECAM-1 (CD31) from Santa Cruz Biotechnology (Santa Cruz, CA, USA). IL-1β, IL-4, IL-6, IL-10, TNF-α, and TGF-βenzyme-linked immuno-sorbent assays (ELISAs) kits were purchased from R&D Systems (Minneapolis, MN, USA). Restriction endonucle-ase, competentEscherichia coli (DH5α), Taq enzyme, Pri-meScript Reverse Transcriptase Reagent Kit and Primer STAR Max DNA Polymerase, MutanBEST Kit, and T4 polynucleotide kinase are from Takara Biotechnology (Da-lian, Liaoning Province, China). DNA purification kit, Dual Luciferase Reporter Assay System, was from Pro-mega corporation (Beijing, China), TIANprep Mini Plas-mid Kit and TIANgel Midi Purification Kit are from Tiangen Biotechnology company (Beijing, China).

Cell culture

Bone marrow-derived mesenchymal stem cells (BMSCs) were isolated from the bone of 3–4 weeks of Sprague-Dawley (SD) rats and cultured in DMEM-LG complete medium as previously described [4, 17]. HEK-293T cells (human embryonic kidney-293 cells ex-pressing the large T-antigen of simian virus 40) were purchased from the cell bank of Chinese Academy of Sciences and cultured in DMEM-HG supplemented with 10% FBS, 100 U/ml penicillin, and 100μg/ml strepto-mycin. Vascular endothelial cells (VECs) were isolated from the abdominal aortic of SD rats as previously de-scribed [22] and cultured in DMEM-HG complete medium. Finally, these cells were digested and passaged when reaching 80% of confluence.

Construction of cell lines of pri-miR-9-BMSCs and TuD-BMSCs

The establishment of cell lines of pri-miR-9-BMSCs and TuD-BMSCs was performed as previously described [17]. In brief, rat genomic DNA was extracted using a DNA purification kit, and a 368-bp DNA fragment con-taining themiR-9-1 sequence (NC_005101.4) was ampli-fied and inserted into the PCDH plasmid to form the

recombinant vector of pri-miR-9-PCDH. The lentivirus en-coding miR-9 was produced by a lentivirus packaging sys-tem (Syssys-tem Biosciences, CA, and USA) as previously described [17]. The anti-miR-9 plasmid was constructed by applying the RNA tough decoy (TuD) technique as previ-ously described [23]. The lentivirus encoding TuD was also produced by a lentivirus packaging system. BMSCs were in-fected with pri-miR-9-, empty-, and TuD-lentivirus to es-tablish the cell lines of pri-miR-9-BMSCs, empty virus BMSCs (EV-BMSCs), and TuD-BMSCs.

Transfection of BMSCs with Cy3-miR-9a-5p mimics and co-culture with VECs

BMSCs were transfected with Cy3-miR-9a-5p mimics or miR-9a-5p control at a final concentration of 100 nM using Lipo2000 as previously described [17]. At 24 h after transfection, they were digested, added into the upper chambers of six cluster plates (Costar Transwell™ Permeable Supports, 0.4-μm), and co-cultured with VECs. The mRNAs and proteins were then extracted using the TRIzol reagent and RIPA lysis buffer at 48 and 72 h, respectively. The expression of genes of interested was verified by general PCR (gPCR) and western blot-ting. The experiments were repeated three times.

Cell apoptosis and proliferation

The cell apoptosis was performed by the apoptosis detec-tion kit as previously described [16]. In brief, BMSCs transfected with miR-9 mimic or miR-9 control was treated with or without LPS (1μg/ml) for 24 h. Then, cells were preincubated with annexin V at room temperature in the dark for 15 min, followed by the addition of propidium iodide (PI). Finally, the percentage of apoptotic cells was analyzed by flow cytometry (BD Biosciences). To evaluate the effect of miR-9 on VECs, the cell proliferation test (MTT) was also conducted as previously described [4]. First, VECs were transfected with miR-9 mimic or miR-9 control. Then, MTT solution (20 μl of 5 mg/ml) was added for 6 days (12 h, 36 h, 60 h, 84 h, 108 h, and 132 h). Third, the medium was removed and 150 μl of DMSO was added to each well, which was shaken slowly for 10 min. Finally, the absorbance was measured by an

Table 1The sequence of primers

Gene Forward (5′-3′) Reverse (5′-3′)

TIE-2 CTGCGGCATGACATGTGCAG GCAAATGATGGTCTCTCATAAGG

Ang-1 CAAGGCTTGGTTACTCGTCAG CCATGAGCTCCAGTTGTTGC

GAPDH CCGTTGTGGATCTGACATGC CTCTTGCTCTCAGTATCCTTGC

Primers miR-9 (5′-3′) U6 (5′-3′)

Reverse transcription GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACTCATAC ACGCTTCACGAATTTGCGTGTC

Forward GGCTCTTTGGTTATCTAGCT CTGCTTCGGCAGCACATATACT

(4)

Fig. 1(See legend on next page.)

(5)

ELISA plate reader at 490 nm. This experiment was re-peated three times.

Cell migration and angiogenesis

VECs transfected with miR-9 mimic or miR-9 control were added to the upper chamber of the Transwell appar-atus. After incubation for 12 h, the upper chamber was fixed with 4% paraformaldehyde for 30 min and stained with 0.1% crystal violet (sigma) for 10 min in the dark and photographed by the phase-contrast microscopy. Finally, the crystal violet was dissolved in 300μl of 33% acetic acid (sigma) and the absorbance of the solution was measured by an ELISA plate reader (Gene Company Limited, HK, China). To investigate the effect of the miR-9 on the an-giogenic activity of VECs in vitro, we performed a tube formation assay. Ninety-six-well culture dishes were coated with 50 μl of matrigel matrix (BD company) and incubated for 30 min at 37 °C. VECs transfected with miR-9 mimic or miR-9 control were seeded onto the so-lidified gels at a density of 2 × 105cells/well in 50μl of cul-ture medium. After incubation for 12 h, the total tube-like structures were photographed by phase-contrast micros-copy (× 100) and counted. The above experiments were repeated three times.

Animal models

Male SD rats weighing 200–250 g (n = 100) were pur-chased from Shanghai Laboratory Animal Co. Ltd. (Shanghai, China) and fed in a suitable environment with 25 °C and 12-h dark/light cycle, given free access to water and food. The AP models were induced by the peritoneal injection of caerulein (100 μg/kg) for three times or the retrograde injection of 3%NaT (1 ml/kg) as previously described [15, 16]. All the procedures con-form to the Ethics of Yijishan Hospital, affiliated to Wannan Medical School (Wuhu City, Anhui Province, China) and the Ethics of Shanghai Tenth People’s Hos-pital, affiliated to Tongji University (Shanghai, China).

Cell transplantation, animal grouping, and sample preparation

Rats were randomly injected with pri-miR-9-BMSCs, EV-BMSCs (EV-BMSCs), TuD-BMSCs, or BMSCs

(1 × 107cells/kg) by the tail vein at postoperative day 1 as previously described [15,16] and divided into NC (n =6), Sham (n = 6), SAP (n = 6), SAP+PBS (PBS treatment) (n = 6), BMSCs (n = 6), pri-miR-9-BMSCs (n = 6), EV-BMSCs (n = 6), and TuD-BMSCs (n = 6) groups. To investigate the relationship between miR-9 and AP, several AP models were established as follows: NC (n = 3), Sham (n = 3), caerulein (n = 3), 1% NaT (n= 3), and 3% NaT (n = 3). Rats were humanely killed at day 3 after BMSC treatment. The serum was collected by centrifugation at 8000×g at 4 °C for 20 min and stored at −80 °C. The tissues were ob-tained by surgical vehicles and stored in liquid nitro-gen or at −80 °C or fixed in 4% paraformaldehyde.

Hematoxylin–eosin (H&E) staining, ELISAs, and the levels of amylase and lipase

The H&E staining of paraffin-embedded pancreatic tis-sues was performed for assessing the severity of AP as previously described [4, 17]. The levels of serum IL-1β,

IL-4, IL-6, IL-10, TNF-α, and TGF-β were detected by ELISA kit as previously described. The activities of serum amylase and lipase were assayed by the amylase and lipase assay kit as previously described [16].

General PCR (gPCR) and quantitative real-time PCR (qRT-PCR)

Total RNA was extracted using TRIzol or TRIzolLS agent. cDNA was synthesized using a PrimeScript Re-verse Transcriptase Reagent Kit. The expression of genes of interest was detected by gPCR or qRT-PCR as previ-ously described [17]. GAPDH and U6 were used as en-dogenous controls. The sequences of the primers are listed in Table 1. Each measurement was performed in three repeats.

Immunoblotting and immunohistochemistry

The immunoblotting (western blot) procedure used in the study was described previously [17]. In brief, total proteins were extracted using RIPA lysis buffer with PMSF (1:100) and protease inhibitor cocktail tablets (Roche Applied Science, Shanghai, China), and the BCA method was used for the protein quantification. Proteins

(See figure on previous page.)

Fig. 1miR-9 modified BMSCs (pri-miR-9-BMSCs) alleviate SAP.aBMSCs infected by pri-miR-9 and empty virus expressing GFP.bThe recombinant pri-miR-9-1-PCDH plasmid was identified by dual-enzyme digestion.cpri-miR-9-1 sequence was amplified from pri-miR-9-1 plasmid using special primers.d,eThe expression of mature miR-9 in pri-miR-9-BMSCs was higher than in empty virus BMSCs (EV-BMSCs) by gPCR and qRT-PCR. Data are shown as the mean ± SD for at least 3 separate experiments.###p< 0.001, compared with EV-BMSCs by paired t test, gPCR, General PCR, qRT-PCR, quantitative real-time PCR.f,g,h,ipri-miR-9-BMSCs significantly reduced pancreatic edema, infiltration, hemorrhage and necrosis, the release of serum amylase, lipase and pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) and increased anti-inflammatory cytokines (IL-4, IL-10, and TGF-β) compared with SAP, SAP+PBS, BMSCs, EV-BMSCs and TuD-BMSCs groups. Data are shown as the mean ± SD for at least three separate experiments.

%

p< 0.05,%%p< 0.01,%%%p< 0.001, compared with NC, **p< 0.01 and ***p< 0.001, compared with NC,@@p < 0.01 and@@@p< 0.001, compared with SAP,&&p< 0.01 and&&&p< 0.001, compared with PBS treatment,#p< 0.05,##p< 0.01, and###p< 0.001, compared with BMSCs,$p< 0.05,

$$

(6)

Fig. 2(See legend on next page.)

(7)

were transferred to a nitrocellulose membrane, which was detected by the Odyssey 3.0 analysis software (LI-COR Biotechnology, Nebraska, USA) after incuba-tion with primary and secondary antibodies. Immuno-histochemistry was performed as previously described [17]. The experiments were repeated three times.

MiRNAs target prediction

The prediction of miRNA target genes was performed by the algorithms of TargetScan [24], PicTar [24], micro-RNAorg [25], and miRWalk Targets [26]. The results of predicted targets were intersected by using MatchMiner [27], showing that the VE-cadherin gene was the target gene of miR-9.

Dual luciferase reporter assays

A fragment of VE-cadherin 3′-UTR (222 bp) containing the putative miR-9 binding site was amplified by Primer STAR Max DNA Polymerase using the following primers:

5′-GAAGCCAGAAACCGGACCCCTGGG-3′ and 5′-GC

CACGGGGATGGAAGTGTCTTTG-3′ and cloned into psiCHECK-2 vector (Promega, Beijing, USA). Finally, the recombinant psiCHECK-2-VE-cadherin 3′-UTR (wtUTR) plasmid was identified by the sequence analysis. Besides, a mutant VE-cadherin 3’UTR fragment (222 bp) with five base mutation (CAAAG→TCGCT) was generated by TaKaRa MutanBEST Kit using the following primers:

5′-GAAGCCAGAAACCGGACCCCTGGG-3′ and 5′-G

CCACGGGGATGGAAGTGTAGCGAGTACCCACAGGC AAG-3′and then cloned into the psiCHECK-2 vector at the XhoI and Not I sites to produce the recombinant psiCHECK-2-mutant VE-cadherin 3′-UTR (mutUTR) plasmid. Finally, the mutUTR plasmid was identified by the sequence analysis. wtUTR or mutUTR plasmid (1μg) was co-transfected with miR-9a-5p mimics (50 nM) into 293T cells by using Lipo2000. The firefly luciferase activity was measured by Dual Luciferase Reporter Assay System at 48 h after transfection. The experiment was repeated above three times.

CM-Dil- and SPION-labeled BMSCs and tracing

To track the migration of injected BMSCs in vivo, CM-Dil was selected for labeling as previously described [4, 15–17]. The pancreas, liver, lung, spleen, heart, and

duodenum were collected and fixed with 4% paraformal-dehyde for 24 h, then dehydrated in a 30% sucrose solu-tion for 2 h. The tissues were then embedded using the Tissue-Tek O.C.T. Compound (SAKURA, USA) and so-lidified into a tissue block at −80 °C. The tissue block was cut into 5μm frozen sections and observed under a fluorescence microscope. The red particles were counted in five randomly selected fields at × 200 magnification. SPION was also used for labeling BMSCs to trace their distribution in vivo as previously described [4].

Tracking of cy3-miR-9a-5p in vivo after the transplantation of EV-BMSCs transfected with Cy3-miR-9a-5p

The liver, heart, spleen, lung, pancreas, kidney, and duo-denum were collected on day 3 after the transplantation of empty virus BMSCs transfected with Cy3-miR-9a-5p and fixed in 4% paraformaldehyde for 24 h. Then, the organs were dehydrated in a 30% sucrose solution and embedded using the Tissue-Tek O.C.T. Compound. Fro-zen sections were observed and photographed using a fluorescence microscope.

Image processing and statistical analysis

Adobe Photoshop 6.0 (Adobe Systems Inc., San Jose, CA), Image-Pro Plus version 6.0 (Media Cybernetics, USA), and ImageJ (National Institutes of Health, USA) were applied for image typesetting, analysis, and pro-cessing. GraphPad Prism 5.1 (GraphPad Co., USA) was used for mapping. The statistical analyses were con-ducted by SPSS 18.0 statistical software (Chicago, IL). Experimental data are shown as means ± standard devia-tions (SD) and compared with Student’s or a paired t test or one-way ANOVA. A value ofP< 0.05 was deemed to indicate significant differences.

Results

Infused BMSCs reduce SAP and inhibit systemic inflammatory responses in a miR-9-dependent manner

A rat model of SAP was established, and BMSCs were iso-lated and cultured as previously described [4]. Then, BMSCs were infused into SAP rats and their effect was in-vestigated. Detection of the expression of miR-9 in

(See figure on previous page.)

Fig. 2pri-miR-9-BMSCs promote angiogenesis.aAngiogenesis-related genes (CXCR4, Ang-1, TIE-2, and CD31) were significantly upregulated after pri-miR-9-BMSC transplantation, compared with SAP, SAP+PBS, BMSCs, or TuD-BMSCs groups. Anti-angiogenic genes (VE-cadherin,β-catenin) were significantly decreased compared with SAP, SAP+PBS, BMSCs, EV-BMSCs, or TuD-BMSCs groups. The PI3K/AKT signaling pathway (PI3K, p-AKT, AKT) was activated by pri-miR-9-BMSCs. Data are shown as the mean ± SD for at least 3 separate experiments. *p< 0.05, **p< 0.01 and ***p< 0.001, compared with NC,++p< 0.01 and+++p< 0.001, compared with SAP,$p< 0.05 and$$p< 0.01, compared with SAP+PBS,#p< 0.01,

##

(8)

pancreatic tissues showed that miR-9 levels were lower in the SAP group and higher in the BMSC group. miR-9 modified/antagonized BMSCs (pri-miR-9-BMSCs/TuD-BMSCs) and empty virus BMSCs (EV-(pri-miR-9-BMSCs/TuD-BMSCs) were injected into SAP rats as previously described [4, 15, 17].

The results showed that pri-miR-9-BMSCs markedly pro-moted the repair of damaged pancreatic tissues (pancre-atic edema↓, infiltration↓, hemorrhage↓, and necrosis↓), decreased the levels of amylase and lipase, and inhibited systemic inflammatory responses (TNF-α↓, IL-1β↓, IL-6↓, Fig. 3(a,b) The distributions in vivo of CM-Dil- and SPION-labeled BMSCs were observed by fluorescence microscopy and Prussian Blue staining. The number of cells migrating to the damaged pancreas was smaller than that of cells migrating to the liver, spleen and lung. Data are shown as the mean ± SD. ***p< 0.001, compared with liver,###p< 0.001, compared with spleen,@@@p< 0.001, compared with lung by using two-tailedttest

(9)
(10)

and IL-4↑, IL-10↑, TGF-β↑). TuD-BMSCs had a minor or no effect (Fig.1).

Pri-miR-9-BMSCs repair injured pancreatic tissues by inducing angiogenesis

Pri-miR-9-BMSCs were previously shown to promote pancreatic regeneration [17]. To examine the underlying mechanism, we first detected the expression of angiogenesis-related genes, and the results showed that pri-miR-9-BMSCs upregulated Ang-1, CXCR4, and TIE-2. Treatment with pri-miR-9-BMSCs activated the downstream PI3K-AKT pathway (p-AKT↑, PI3K↑), and significantly upregulated CD31 and CD34 compared with the SAP, PBS + SAP, BMSCs, EV-BMSCs, and TuD-BMSCs groups. However, TuD-BMSCs did not promote the expression of Ang-1, CXCR4, and TIE-2 or activate the PI3K/AKT pathway (Fig.2).

Few BMSCs could move to the damaged pancreatic tissue

To observe the migration of the infused cells in SAP rats, CM-Dil and superparamagnetic iron oxide nano-particles (SPION) were used for labeling BMSCs, and the number of red and blue particles in pancreatic tis-sues was counted by fluorescence microscopy or iron staining as previously described [4, 17]. The results showed that transplanted BMSCs could migrate to the injured pancreas; however, the number of cells migrating to the pancreas was small and lower than that of cells migrating to the lung, spleen, and liver at 3 days after transplantation. Therefore, the direct effect of BMSCs on repairing injured pancreatic tissues on day 3 after cell transplantation was difficult to explain (Fig.3).

BMSCs deliver exogenous miR-9 into the pancreas and other organs, which can promote the proliferation, migration, and angiogenesis of vascular endothelial cells

To trace the distribution of transplanted BMSCs in vivo, synthetic Cy3-miR-9a-5p (red) was transplanted into GFP-BMSCs (green) and these cells were infused into

SAP rats by the tail vein as previously described. The re-sults showed that Cy3-miR-9a-5p was released from GFP-BMSCs into the damaged pancreas and other or-gans; however, a higher number of infused BMSCs migrated to the lung, liver, and spleen than to the pancreas (Fig. 4a, b). miR-9 inhibited apoptosis and promoted the proliferation, migration, and angiogen-esis of VECs (Fig. 5).

MiR-9 is a protective factor and positively correlated with angiogenesis in AP

The expression of miR-9 was lower in the AP group than in the NC/Sham group, suggesting a negative rela-tionship with the severity of AP. The levels of CXCR4, p-AKT, VEGFA, Ang-1, and TIE-2 were significantly lower in AP than in the NC and Sham groups. Pearson’s correlation showed that the expression of CXCR4, p-AKT, VEGFA, Ang-1, and TIE-2 was negatively corre-lated with the severity of AP. The expression of miR-9 was positively correlated with that of CXCR4, p-AKT, VEGFA, Ang-1, and TIE-2 (Fig.6).

VE-cadherin is a target of miR-9

The VE-cadherin transcript and the miR-9a-5p seed se-quence have eight pairs of bases at both putative target sites. VE-cadherin and ß-catenin were markedly down-regulated in VECs after miR-9a-5p transient overexpres-sion. To determine whether miR-9a-5p can target the VE-cadherin gene, we constructed luciferase reporter vectors for VE-cadherin wild-type (wtUTR) or mutant 3′-UTR (mutUTR) harboring the predicted binding sites for miR-9a-5p. The results showed that the relative ac-tivity of firefly luciferase in 293T cells was markedly de-creased after transfection of miR-9a-5p mimics. The luciferase reporter repression was rescued by mutating the VE-cadherin 3′-UTR. Taken together, these results indicated that VE-cadherin was the target gene of miR-9a-5p (Fig.4c–g).

(See figure on previous page.)

Fig. 4miR-9, released from BMSCs into VECs, can target the VE-cadherin gene and induce the expression of angiogenic genes in VECs.amiR-9 in pancreatic tissues was downregulated in the PBS + SAP group compared with the NC group, whereas it was significantly upregulated by pri-miR-9-BMSCs, compared with SAP, SAP+PBS, BMSCs, EV-BMSCs, and TuD-BMSCs. Data are shown as the mean ± SD for at least three separate experiments.

%

p< 0.05, compared with NC,**p< 0.01, compared with NC,@@p< 0.01, compared with SAP,&&p< 0.01, compared with PBS treatment,#p< 0.05, compared with BMSCs,$p< 0.05, compared with EV-BMSCs,+p< 0.05, compared with TuD-BMSCs by using two-tailedttest.bGFP-BMSCs could deliver exogenous Cy3-miR-9a-5p to the liver, spleen, lung, and pancreas, and the number was higher in the liver and spleen.cEight paired base between miR-9 and VE-cadherin and the structure of the wild-type VE-cadherin 3′-UTR (wtUTR) or mutant VE-cadherin 3′-UTR (CAAAG→TCGCT) (mutUTR) were cloned into the psiCHECK-2 plasmid to produce the recombinant vectors, wtUTR psiCHECK-2 and mutUTR psiCHECK-2, harboring the predicted binding sites of miR-9.dThe activity of firefly luciferase was significantly decreased by miR-9 and rescued by mutant of VE-cadherin 3′-UTR. e–gCo-culture of VECs and BMSCs transfected with cy3-miR-9 showed that BMSCs secreted cy3-miR-9 into VECs and inhibited the expression of VE-cadherin andβ-catenin and upregulated Ang-1, CXCR4, TIE-2 and p-AKT.*p< 0.05,**p< 0.01, and***p< 0.001, compared with NC group,@p< 0.05,

@@

p< 0.01, and@@@p< 0.001, compared with miR-9 con by using paired t test. VECs, vascular endothelial cells; BMSCs, bone marrow-derived mesenchymal stem cells; GFP, green fluorescent protein; gPCR, general PCR; qRT-PCR, quantitative real-time PCR; SAP, severe acute pancreatitis; NC, normal control; miR-9 con, miR-9 mimic control

(11)
(12)

Discussion

The incidence of AP is increasing gradually, and the underlying pathogenetic mechanism remains undefined [28]. Most AP cases can be cured by conventional med-ical treatment. However, 10 to 20% of AP cases can evolve into SAP, which is a systemic disease character-ized by pancreatic necrosis, systemic inflammatory re-sponses, and multiple organ dysfunction [1, 2, 28]. Preventing the occurrence of SAP is currently not pos-sible, and the disease process is not completely under-stood. Repairing injured pancreatic tissues thoroughly cannot be achieved, and the mortality of SAP remains at approximately 40% [2,4, 28]. The morbidity of SAP also affects the quality of life of patients, who often require pancreatic enzymes or insulin injections to support their lives. The high mortality and morbidity not only makes patients suffer, but is also associated with negative effects on the national economy. Thus, it is very urgent to find effective therapeutic strategies for treating those patients [28].

The current treatment methods are mostly combinations of internal medicine and surgery [2]. Biotherapy, which is gradually being accepted by patients, is based on stem cells as a novel therapeutic method that is expected to cure SAP completely [4, 6,15–17,29–31]. MSCs, which are import-ant members of the family of stem cells, are characterized by specific properties such as self-renewal, multilineage dif-ferentiation, immunosuppressive effects, and paracrine ef-fects and are also used as vectors for exogenous genes [31]. Animal experiments show that BMSCs can ameliorate SAP by secreting cellular growth factors, regulating immune re-sponses, and protecting gastrointestinal functions [15–17]. In our previous study, we showed that BMSCs could pro-mote the repair of injured pancreatic tissues, although the underlying mechanism remains unclear [17]. miR-9 plays a key role in the inhibition of inflammatory responses in SAP. BMSCs release miR-9 into inflammatory cells and re-duce the secretion of pro-inflammatory cytokines by sup-pressing the NF-κB signaling pathway [17]. In addition, we showed that genes related to pancreatic regeneration, such as pancreas transcription factor 1 complex (PTF1), pancre-atic and duodenal homeobox 1(PDX1), and regenerating islet-derived protein 4 (Reg4), were significantly upregu-lated and pancreatic necrosis was markedly decreased after the injection of BMSCs. Based on these results, we con-cluded that BMSCs could treat SAP. In the present study, we investigated the mechanism underlying the effect of BMSCs on promoting the recovery of SAP, and the

results showed that BMSCs induced angiogenesis by re-leasing miR-9. miRNAs are involved in the pathologic process of SAP, and several miRNAs serve as bio-markers for diagnosis and therapeutic targets in SAP [17–19, 32]. However, few studies have revealed that miRNAs can promote pancreatic regeneration. In the present study, we demonstrated that miR-9 plays a role as a regeneration factor of inducing angiogenesis by suppressing VE-cadherin/-catenin signaling pathway.β

Pancreatic diseases are often irreversible, leading to patient suffering and decreased quality of life [33, 34]. Therefore, many researchers have started to focus on pancreatic regeneration, which can eradicate pancreatic diseases and alleviate pain. The process of regeneration must be inseparable from angiogenesis, which is essen-tial for tissue regeneration [35,36].

BMSCs have the potential for secreting cellular growth factors and promoting angiogenesis, although the underlying mechanism remains unknown [4]. In the present study, we explored the possible mechanism using genetic engineering methods and by constructing miR-9 modified/antagonizing BMSCs (pri-miR-9-BMSCs and TuD-(pri-miR-9-BMSCs). The results showed that pri-miR-9-BMSCs promoted angiogenesis more effect-ively than BMSCs and TuD-BMSCs. The expression levels of Ang-1, TIE-2, CXCR4, and p-AKT in pri-miR-9-BMSCs were also the highest and the lowest in TuD-BMSCs. These results showed that angiogenesis was regulated by miR-9 in BMSC therapy for SAP.

Further, we demonstrated that the angiogenesis was inhibited in injured pancreatic tissues, showing the correl-ation with the severity of pancreatitis and miR-9 was posi-tively correlated with the angiogenesis. Besides, transfection of miR-9 mimics into VECs showed that miR-9 significantly inhibited the expression of angiogenesis-related genes (VEGF, Ang-1, TIE-2, CXCR4, and p-AKT). These data demonstrated that miR-9 was involved in the process of angiogenesis and could promote its occurrence.

To further reveal the mechanism underlying the effect of miR-9 on promoting angiogenesis, we predicted the target gene of miR-9 using online software and found that VE-cadherin is a target gene of miR-9. To confirm that miR-9 can repress the expression of VE-cadherin by directly binding to the VE-cadherin 3′-UTR, we per-formed a dual luciferase reporter assay and detected the expression of the VE-cadherin gene in VECs after co-culture with BMSCs transfected with miR-9 mimics. The results showed that miR-9 could reduce the activity

(See figure on previous page.)

Fig. 5miR-9 promoted proliferation, migration and angiogenesis, and inhibited apoptosis in VECs.a–bmiR-9 significantly inhibited apoptosis in VECs induced by LPS compared with LPS + miR-9 control. *p< 0.05, compared with LPS + miR-9 control.cmiR-9 promoted the proliferation of VECs.**p< 0.001 and ***p< 0.001, compared with miR-9 control group.d–gmiR-9 increased migration and induced angiogenesis. *p< 0.05, compared with miR-9 control

(13)
(14)

of firefly luciferase in a dose-dependent manner, and this could be rescued by anti-miR-9 (TuD) or the 3′-UTR mutant (mutUTR). Meanwhile, transfer of exogenous miR-9 mimics from BMSCs to VECs downregulated the expression of VE-cadherin in VECS. Therefore, the VE-cadherin gene was confirmed as the target gene of miR-9. The expression of VE-cadherin and-catenin in in-β jured pancreatic tissues was detected after the injection

of BMSCs, and the results showed that

pri-miR-9-BMSCs upregulated VE-cadherin and -catenin.β These data demonstrated that miR-9 targets VE-cad-herin and affects the activity of the-catenin signalingβ pathway to regulate angiogenesis.

The repair of damaged blood vessels affects the sys-temic inflammatory response. In the present study, the levels of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) were decreased and the levels of anti-inflammatory cytokines (IL-4, IL-10, and TGF-β) were significantly in-creased in the pri-miR-9-BMSC group, consistent with the results of angiogenesis.

Further, we used two kinds of materials for labeling BMSCs, CM-Dil and SPION, to assay the contributions of BMSCs in vivo. The results showed that the CM-Dil-or SPION-labeled BMSCs could migrate to the pancreas, although at lower number than those migrating to the lung. Hence, infused BMSCs may repair injured pancre-atic tissues mainly by paracrine effects rather than the direct interaction at the early phase of BMSCs therapy. Moreover, transplanted BMSCs could deliver miR-9 to the liver, spleen, lung, and pancreas, suggesting that it is possible to repair SAP through the secretions of BMSCs. Co-culture of BMSCs transfected with exogenous miR-9 and VECs showed that miR-9 could migrate to VECs from BMSCs to regulate the functions of VECs and the expression of target genes.

In addition, in another study [4], we traced the distribu-tions of infused BMSCs at postoperative day 1, 3, 5, 7, 10 and the result showed that SPION-labeled BMSCs grad-ually migrated to injured pancreas and peaked on postop-erative days 5–7. We speculate on that and come to the conclusion that miR-9-induced angiogenesis might help BMSCs migrate to injured pancreas (Additional file 1). Furthermore, we investigated that SPION-labeled BMSCs migrating to injured pancreas obviously decreased at

postoperative day 10. The possible reason for the sharp decline is that those BMSCs might have differentiated into pancreatic cells and regenerate necrotized pancreatic tissue.

In a word, the process of BMSCs promoting pancreatic regeneration is very complex, and the mechanism is dif-ficult to understand. At present, most of the studies on pancreatic regeneration mainly focus on the regener-ation of insulin-producing β-cells rather than the regen-erative process of digestive enzyme-producing acinar cells [33]. Thus, little is known about the regeneration of acinar cells. In this study, we revealed that infused BMSCs could induce the angiogenesis at day 3 after cell transplantation by secreting miR-9. Combining with the previous study, we found that miR-9 released by BMSCs can induce angiogenesis, which could help more BMSCs migrate to the injured pancreas and promote the regen-eration of necrotized pancreatic tissue.

Conclusions

miR-9 is an important protective factor in SAP, and BMSCs can repair SAP through miR-9, which targets the VE-cadherin gene, modulates-catenin signaling, and in-β duces angiogenesis. These findings suggest the potential of BMSCs for the treatment of SAP and identify a new therapeutic target.

Additional file

Additional file 1:The possible mechanisms of speculation on BMSC repairing necrotized pancreatic tissue. (JPG 17 kb)

Acknowledgements

We sincerely thank Hui Wang for editing our language and dialog expression. We also appreciate Ge Wei, Chenglei Xu, and Zhigang He for offering us the vector. Finally, thanks to Deng Yuan, Jie Hua and Jian Gong for teaching us the method of isolating bone marrow-derived mesenchymal stem cells (BMSCs).

Funding

The present study was supported by research grants from the National Natural Science Foundation of China (No. 81670582) and College Natural Science Foundation of Anhui Province (KJ2017A271) and talent introduction fund of Yijishan Hospital of Wannan Medical School (YR201601).

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study. If you want, please contact author for data requests.

(See figure on previous page.)

Fig. 6a,bAP was induced by caerulein and 3% NaT, and H&E staining showed that pancreatic damage was more severe in the 3% NaT group than in the caerulein group.c,dmiR-9 expression was lower in damaged pancreatic tissues and the serum of AP rats than in the NC or Sham group. Data are shown as the mean ± SD.&&&p< 0.001 and###p< 0.01, compared with NC, *p< 0.05, compared with NC,%%%p< 0.001, compared with Sham,$$p< 0.01, compared with Caerulein group by using paired t test.eWestern blot analysis showed that angiogenic genes (VEGFA, Ang-1, TIE-2, p-AKT, CXCR4) were downregulated in damaged pancreatic tissues.fThe level of miR-9 and the expression of angiogenic genes were negatively correlated with the severity of AP, and miR-9 was positively correlated with angiogenesis-related as determined by Pearson’s correlation analysis (p< 0.05). AP, acute pancreatitis; H&E, hematoxylin eosin; NC, normal control; NaT, sodium taurocholate; SD, standard deviation; miR-9, microRNA-9

(15)

Authors’contributions

DQ, GS and ZM performed this experiment. XW and ZS designed the study and wrote the main manuscript. GW and MH collected the data and analyzed the results. LJ checked the data and prepared the pictures. All authors read and approved the final manuscript.

Ethics approval

All animal experiments are approved by the Ethics committee of Yijishan Hospital, affiliated to Wannan Medical School (Wuhu City, Anhui Province, China) and the Ethics of Shanghai Tenth Peoples Hospital, affiliated to Tongji University (Shanghai, China).

Consent for publication

All authors have reviewed the manuscript and approved the publication.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author details 1

Department of General Surgery, Yijishan Hospital, Wannan Medical College, Wuhu 241001, Anhui, China.2Department of General Surgery, Shanghai

Tenth Peoples Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai 200072, China.3Department of Pharmacology and

Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, USA.4Department of Gastroenterology, The Second Affiliated Hospital

of Wannan Medical College, Wuhu 241001, Anhui, China.

Received: 28 June 2018 Revised: 11 September 2018 Accepted: 28 September 2018

References

1. Maheshwari R, Subramanian RM. Severe acute pancreatitis and necrotizing pancreatitis. Crit Care Clin. 2016;32:279–90.

2. Portelli M, Jones CD. Severe acute pancreatitis: pathogenesis, diagnosis and surgical management. Hepatobiliary Pancreat Dis Int. 2017;16:155–9. 3. Zheng YJ, Zhou B, Ding G, et al. Effect of serum from patients with severe

acute pancreatitis on vascular endothelial permeability. Pancreas. 2013;42: 6339.

4. Qian D, Gong J, He Z, et al. Bone marrow-derived mesenchymal stem cells repair necrotic pancreatic tissue and promote angiogenesis by secreting cellular growth factors involved in the SDF-1 alpha /CXCR4 axis in rats. Stem Cells Int. 2015;2015:306836.

5. Ge N, Xia Q, Yang ZH, et al. Vascular endothelial injury and apoptosis in rats with severe acute pancreatitis. Gastroenterol Res Pract. 2015;2015:235017. 6. Hua J, He ZG, Qian DH, et al. Angiopoietin-1 gene-modified human

mesenchymal stem cells promote angiogenesis and reduce acute pancreatitis in rats. Int J Clin Exp Pathol. 2014;7(7):3580–95. 7. Kargozar S, Baino F, Hamzehlou S, et al. Bioactive glasses: sprouting

angiogenesis in tissue engineering. Trends Biotechnol. 2018;36:430–44. 8. Wang Y, Yin D, Xu C, et al. Roxarsone induces angiogenesis via PI3K/Akt

signaling. Cell Biosci. 2016;6:54.

9. Yuan X, Han L, Fu P, et al. Cinnamaldehyde accelerates wound healing by promoting angiogenesis via up-regulation of PI3K and MAPK signaling pathways. Lab Invest. 2018;98(6):78398.

10. Wagner NM, Bierhansl L, Noldge-Schomburg G, et al. Toll-like receptor 2-blocking antibodies promote angiogenesis and induce ERK1/2 and AKT signaling via CXCR4 in endothelial cells. Arterioscler Thromb Vasc Biol. 2013; 33:194351.

11. Gaengel K, Niaudet C, Hagikura K, et al. The sphingosine-1-phosphate receptor S1PR1 restricts sprouting angiogenesis by regulating the interplay between VE-cadherin and VEGFR2. Dev Cell. 2012;23:587–99.

12. Hrgovic I, Doll M, Pinter A, et al. Histone deacetylase inhibitors interfere with angiogenesis by decreasing endothelial VEGFR-2 protein half-life in part via a VE-cadherin-dependent mechanism. Exp Dermatol. 2017;26:194–201. 13. Warren NA, Voloudakis G, Yoon Y, et al. The product of the

gamma-secretase processing of ephrinB2 regulates VE-cadherin complexes and angiogenesis. Cell Mol Life Sci. 2018;75(15):2813–26.

14. Isidori AM, Venneri MA, Fiore D. Angiopoietin-1 and Angiopoietin-2 in metabolic disorders: therapeutic strategies to restore the highs and lows of angiogenesis in diabetes. J Endocrinol Investig. 2016;39:1235–46. 15. Jung KH, Song SU, Yi T, et al. Human bone marrow-derived clonal

mesenchymal stem cells inhibit inflammation and reduce acute pancreatitis in rats. Gastroenterology. 2011;140:9981008.

16. He Z, Hua J, Qian D, et al. Intravenous hMSCs ameliorate acute pancreatitis in mice via secretion of tumor necrosis factor-alpha stimulated gene/ protein 6. Sci Rep. 2016;6:38438.

17. Qian D, Wei G, Xu C, et al. Bone marrow-derived mesenchymal stem cells (BMSCs) repair acute necrotized pancreatitis by secreting microRNA-9 to target the NF-kappaB1/p50 gene in rats. Sci Rep. 2017;7:581.

18. Xiang H, Tao X, Xia S, et al. Targeting microRNA function in acute pancreatitis. Front Physiol. 2017;8:726.

19. Zhang XX, Deng LH, Chen WW, et al. Circulating microRNA 216 as a marker for the early identification of severe acute pancreatitis. Am J Med Sci. 2017; 353:178–86.

20. Cui Y, Han Z, Hu Y, et al. MicroRNA-181b and microRNA-9 mediate arsenic-induced angiogenesis via NRP1. J Cell Physiol. 2012;227:772–83.

21. Qu J, Lu D, Guo H, et al. MicroRNA-9 regulates osteoblast differentiation and angiogenesis via the AMPK signaling pathway. Mol Cell Biochem. 2016;411: 2333.

22. Li C, Yang L, Wu H, et al. Paeonol inhibits oxidized low-density lipoprotein-induced vascular endothelial cells autophagy by upregulating the expression of miRNA-30a. Front Pharmacol. 2018;9:95.

23. Haraguchi T, Ozaki Y, Iba H. Vectors expressing efficient RNA decoys achieve the long-term suppression of specific microRNA activity in mammalian cells. Nucleic Acids Res. 2009;37:e43.

24. Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009;19:92105.

25. Betel D, Wilson M, Gabow A, et al. The microRNA.org resource: targets and expression. Nucleic Acids Res. 2008;36:D149–53.

26. Dweep H, Sticht C, Pandey P, et al. miRWalk--database: prediction of possible miRNA binding sites bywalkingthe genes of three genomes. J Biomed Inform. 2011;44:839–47.

27. Bussey KJ, Kane D, Sunshine M, Narasimhan S, Nishizuka S, Reinhold WC, Zeeberg B, Ajay W, Weinstein JN. MatchMiner: a tool for batch navigation among gene and gene product identifiers. Genome Biol. 2003;4:R27. 28. Garber A, Frakes C, Arora Z, et al. Mechanisms and management of acute

pancreatitis. Gastroenterol Res Pract. 2018;2018:6218798.

29. Tu XH, Huang SX, Li WS, et al. Mesenchymal stem cells improve intestinal integrity during severe acute pancreatitis. Mol Med Rep. 2014;10:181320. 30. Yin G, Hu G, Wan R, et al. Role of microvesicles from bone marrow

mesenchymal stem cells in acute pancreatitis. Pancreas. 2016;45:1282–93. 31. Kawakubo K, Ohnishi S, Kuwatani M, et al. Mesenchymal stem cell therapy

for acute and chronic pancreatitis. J Gastroenterol. 2018;53:15. 32. Lu P, Wang F, Wu J, et al. Elevated serum 7, 9, 122, and

miR-141 are noninvasive biomarkers of acute pancreatitis. Dis Markers. 2017; 2017:7293459.

33. Kim HS, Lee MK. Beta-cell regeneration through the transdifferentiation of pancreatic cells: pancreatic progenitor cells in the pancreas. J Diabetes Investig. 2016;7:286–96.

34. Minami K, Seino S. Current status of regeneration of pancreatic beta-cells. J Diabetes Investig. 2013;4:13141.

35. Bulgin D. Therapeutic angiogenesis in ischemic tissues by growth factors and bone marrow mononuclear cells administration: biological foundation and clinical prospects. Curr Stem Cell Res Ther. 2015;10:509–22.

Figure

Table 1 The sequence of primers
Fig. 1 (See legend on next page.)
Fig. 2 (See legend on next page.)
Fig. 3 (a, b) The distributions in vivo of CM-Dil- and SPION-labeled BMSCs were observed by fluorescence microscopy and Prussian Blue staining.The number of cells migrating to the damaged pancreas was smaller than that of cells migrating to the liver, sple
+4

References

Related documents

Figure (3-a): Combined effect of molar ratio and catalyst loading on the biodiesel yield during reactive extraction via ultrasonication of castor seed at room temperature (3D

The future directions for Playground include including symbolic execution for systematic exploration of the ap- plications’ state space and to make Playground even more stealthy

This Research Study has been undertaken as a single exploratory case study with one of the reputed Sports Goods Manufacturing Industry from India for Empirical Examination of

Multiple sequence alignment of CnPMK amino acid sequences with PMKs from other species showed high sequence similarity (Fig. The multiple alignments analysis revealed

In general, semiautonomous teleoperated systems are compose with two parts, the first is the human controlled situations (non-autonomous component) and the second

We conclude that an extra-articular resection of the hip using the Bernese periacetabular technique, followed by reconstruction with an extracorporeally irradiated acetabular

For the statement that there is no positive Global Effect of Product Manufacturing on the Nigeria Business Competitive Position on Some Selected Institutions in

trauma, posttraumatic stress disorder, comparative effectiveness research, intervention studies, child,