Skip to main content
Erschienen in: BMC Immunology 1/2014

Open Access 01.12.2014 | Research article

Mesenchymal stromal cells reset the scatter factor system and cytokine network in experimental kidney transplantation

verfasst von: Marilena Gregorini, Francesca Bosio, Chiara Rocca, Valeria Corradetti, Teresa Valsania, Eleonora Francesca Pattonieri, Pasquale Esposito, Giulia Bedino, Chiara Collesi, Carmelo Libetta, Francesco Frassoni, Antonio Dal Canton, Teresa Rampino

Erschienen in: BMC Immunology | Ausgabe 1/2014

Abstract

Background

In former studies we showed in a rat model of renal transplantation that Mesenchymal Stromal Cells (MSC) prevent acute rejection in an independent way of their endowing in the graft. In this study we investigated whether MSC operate by resetting cytokine network and Scatter Factor systems, i.e. Hepatocyte Growth Factor (HGF), Macrophage Stimulating Protein (MSP) and their receptors Met and RON, respectively.

Methods

MSC were injected into the renal artery soon after reperfusion. Controls were grafted untreated and normal rats. Rats were sacrificed 7 days after grafting. Serum and renal tissue levels of IFN-γ, IL-1, IL-2, IL-4, IL-6, IL-10, MSP/RON, HGF/Met systems, Treg lymphocytes were investigated.

Results

In grafted untreated rats IFN-γ increased in serum and renal tissue and IL-6 rose in serum. MSC prevented both the phenomena, increased IL-10 serum levels and Treg number in the graft. Furthermore MSC increased serum and tissue HGF levels, Met tubular expression and prevented the suppression of tubular MSP/RON expression.

Conclusions

Our results demonstrate that MSC modify cytokine network to a tolerogenic setting, they suppress Th1 cells, inactivate monocytes/macrophage, recruit Tregs. In addition, MSC sustain the expression of the Scatter Factor systems expression, i.e. systems that are committed to defend survival and stimulate regeneration of tubular cells.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12865-014-0044-1) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.
Abkürzungen
Ab
Antibody
ANOVA
Analysis of variance
BCA
Bicinchoninic acid
CD
Cluster differentiation
cDNA
Complementary DNA
EDTA
Ethylenediamine tetraacetic Acid
EGFP
Enhanced GFP
ELISA
Enzyme-linked immunosorbent assay
FAM
6 carboxyfluorescein
FCS
Fetal calf serum
FITC
Fluorescein isothiocyanate
Foxp3
Forkhead box p 3
g
Gram(s)
GFP
Green fluorescent protein
HGF
Hepatocyte growth factor
HPF
High power field
IFN- γ
Interferon gamma
LPS
Lipopolysaccharide
M
Molar
mmol
Millimole(s)
MSC
Mesenchymal stromal cells
MSP
Macrophage stimulating protein
nr
Number
PBMC
Peripheral blood mononuclear cell(s)
PBS
Phosphate buffered saline
PCNA
Proliferating cell nuclear antigen
PE
Phycoerythrin
PIPES
Piperazine N,N’ bis(2 ethanesulfonic acid)
RT-PCR
Reverse transcriptase polymerase chain reaction
SD
Standard deviation
SF
Scatter factor
SLE
Systemic lupus erythematosus
Th
T helper
Treg
Regulatory T cell
Vol
Volume
α
alfa
β
beta
μg
Microgram(s)
μmol
Micromole(s)

Background

Mesenchymal Stromal Cells (MSC) are pluripotent cells that differentiate into various mature cell types [1],[2]. A distinctive property of MSC is that they are not immunogenic [3],[4], and inhibit cell and antibody-mediated immunity in several ways, including the induction of T regulatory cell differentiation [5]-[14]. In a rat model of kidney transplantation we found that MSC injected in the graft improved its function and attenuated renal injury, reducing significantly tubulitis, vasculitis, glomerulitis and immune cell infiltration. Furthermore, we traced MSC in the recipient tissues and found an irrelevant number of them in the kidney. The last finding suggests that mediators account for the protection provided by MSC to the renal graft [15]. A renal protective effect of MSC was shown also in a mouse model of renal transplantation, in which MSC suppressed rejection when they were infused before transplantation, while post-transplant infusion worsened graft outcome [16]. Interestingly, MSC injected before transplantation were found in lymphoid organs and did not localize in the graft, confirming the need for some mediation of MSC effects. Indeed a mediated action of MSC has been proved in diverse experimental disease models [17]-[23], and cytokines have been proposed to play the role of effectors [24]-[32]. However, as yet no evidence has been given that MSC modify the cytokine network in the setting of renal graft, so that we have felt it interesting to test the hypothesis in the present study.
In addition, we have thought of the Scatter Factors as a system that could be reset by MSC and provide protection in the renal graft model. The Scatter Factor (SF) system consists of Hepatocyte Growth Factor (HGF) and Macrophage Stimulating Protein (MSP) and their receptors Met and RON respectively. There are several reasons to think that MSC operate through the SF system. In fact, the SFs are expressed in normal kidney [33]-[35] and participate in the regulation of cell growth and inflammation in various renal diseases including autoimmune forms, e.g. they stimulate renal cell growth and modulate monocyte traffic in the kidney [36]-[42]. Furthermore, we demonstrated that in experimental anti Thy-1 nephritis MSC improve renal injury by modulating SFs [43].
In summary, we investigated whether MSC injected into the renal graft modify the cytokine network and SFs in a way that fits with the concept that these systems are the effectors of MSC-induced graft protection.

Results and discussion

Characterization of rat MSC

As detailed elsewhere the MSC used in the present study were isolated from Sprague Dawley EGFP rats and differentiated into osteogenic and adipogenic cells. Flow fluorocytometry analysis showed that MSC were positive for CD90 (≥95%) and CD73 (≥95%) and were negative for CD45 (<5%), CD11b (<5%), CD34 (<5%), CD79 (<5%).

Tissue and serum levels of cytokines

The expression in renal tissue of IFN-γ, IL-10, IL-6, IL-2, IL-1 was significantly increased in grafted kidneys not injected with MSC compared with native kidneys. MSC injection attenuated significantly the rise of IFN-γ and caused a further increase of IL-10. MSC did not modify tissue levels of any else cytokine. IL-4 tissue levels were similar in all groups (Table 1). In MSC untreated rats grafting was associated with a rise in serum levels of all tested cytokines, except for IL-4. MSC treatment reduced serum IFN-γ and IL-6 and increased IL-10 compared to untreated rats (Table 2).
Table 1
Cytokine levels in renal tissue
pg/mg
A
B
C
B vs C
p
INF-γ
35.2±0.02
206.3±64.4§
116.7±56.1
§ p < 0.005 vs B, C; §p < 0.005 vs C
IL-10
8.6±1.4 §
25.53±0.7§
42.2±10.6
§ p < 0.005 vs B, C; §p < 0.001 vs C
IL-6
117.7±11.36§
245.5±28.8
283.0±32.7
NS
§p < 0.001 vs B, C
IL-2
140.6±0.14§
227±93.1
262±103.4
NS
§p < 0.005 vs B, C
IL-1
235.8±134.4§
505.4±219.7
493±142
NS
§p < 0.005 vs B, C
IL-4
13.05±1.4
26.6±6
32.8±5.2
NS
NS
Groups are defined in Figure 8. A indicates cytokine levels in kidneys of healthy rats, B and C indicate cytokine levels in renal tissue of recipient rats 7 days after transplantation. Data represent means ± SD. The symbols © and ° indicate the different statistical power.
Table 2
Serum cytokine levels
pg/ml
A
B
C
B vs C
p
INF-γ
14±0.08§
151.2±78.6 §
86.7±32.1
§p < 0.0001 vs B, C; § p < 0.05 vs C
IL-10
83.9±9.7*
139.1±20.4
§p < 0.0001 vs B, C; *p < 0.05 vs C
IL-6
183±61.01 #
14.09±0.06
§p < 0.0001 vs B, C; # p < 0.05 vs C
IL-2
61±24.7
94.6±61
NS
§p < 0.05 vs B, C
IL-1
1515±613
1843±670
NS
§p < 0.005 vs B, C;
IL-4
0
0
0
NS
 
Groups are defined in Figure 8. A indicates serum cytokine levels in healthy rats. B and C indicate serum cytokine levels in recipient rats 7 days after transplantation. Data are means±SD. The symbols §, §, *, # indicate the different statistical power.

Tubular necrosis and tubular cell proliferation

Tubules with necrotic cells were significantly less in grafts injected with MSC compared to untreated grafts (Figure 1, panel a). The expression of PCNA was suppressed in grafts not injected with MSC, but it was spared by MSC treatment (Figure 1, panel b).

Foxp3 cells infiltrate

In the grafted kidneys not injected with MSC we found few Foxp3 positive cells (2±1/section) and the number increased in MSC treated rats (10±5/section, p < 0.05). In the spleen we detected 15±10 cells/section (Figure 2).

HGF/Met System

Serum and renal tissue HGF levels, HGF mRNA expression in kidneys

HGF serum levels significantly decreased in grafted rats that were not injected with MSC compared to control normal rats. MSC treatment returned serum HGF to normal levels (Figure 3, upper panel).
Both HGF protein and HGF mRNA were reduced in grafted kidneys compared to native organs. MSC injection prevented the loss of HGF protein (Figure 3, middle panel), although HGF mRNA levels were lower probably for a feedback mechanism (Figure 3, lower panel).

Renal expression of Met

The widespread expression of Met protein in tubules of native kidneys (Figure 4, panel A) was almost completely abrogated in untreated grafted kidneys (Figure 4, panel B). MSC infusion prevented Met loss (Figure 4, panel C). The result was confirmed by Western Blot for Met performed on kidney tissue (Figure 4, lower panel).

MSP/RON system

Renal expression of MSP/RON

Immunohistochemistry showed that MSP was expressed in tubular cells of native kidneys (Figure 5 panel a) and it was undetectable in untreated grafts except for some tubules in which it was distributed with luminal pattern (Figure 5, panel b). MSC restored tubular MSP in transplanted rats, but in a different pattern from normal, i.e. luminal instead of cytoplasmic (Figure 5, panel c). As MSP also its receptor, RON, was diffusely expressed with cytoplasmic pattern in tubules of healthy rats (Figure 5, panel d), while in transplanted rats with acute rejection tubular RON expression was significantly decreased (Figure 5, panel e) and reappeared in tubules of allografts treated with MSC (Figure 5, panel f). In untreated grafts RON was expressed also by some inflammatory cells in the interstitium, but it was absent in infiltrating cells of MSC treated rats (Figure 5, panels e and f).

MSP mRNA expression in kidneys

RT-PCR showed a significant decrease of MSP mRNA in the untreated graft. MSC injection was associated with recovery of MSP mRNA expression (Figure 6).

MSC express MSP mRNA

We performed in vitro experiments aimed to understand whether MSC are possible producers of MSP.
PBMC known to be constitutive MSP producers (44) were used as controls. The expression of MSPmRNA was investigated by quantitative PCR in PBMC and in MSC. The expression of MSP mRNA in PBMC was held as the arbitrary unit. MSC expressed a greater amount of constitutive MSP compared with PBMC (Figure 7).

RON mRNA and RON protein expression in monocytes

RON mRNA expression normalized to the beta-actin significantly increased in monocytes stimulated in vitro for 6 h with LPS and INF-γ than in monocytes cultured in basal condition (C) respectively (LPS: 3,8±0,5 fold increase, p < 0.001 vs C) (IFN-γ: 70,.03±0,3 fold increase, p < 0.001 vs C ) (data not showed).
RON positive monocytes percentage in basal condition (C) was 28,4±2%, but it increased significantly after in vitro monocytes stimulation for 24 h with respectively LPS and INF-γ (LPS: 54,4±4%, p < 0.05 vs C; IFN-γ: 56,7±3.5%, p < 0.05 vs C) (data not showed).

Conclusions

We formerly showed that MSC injected in a rat model of renal transplantation attenuated the severity of rejection and we found also that such effect was independent of MSC endowing in the transplanted kidney. The latter findings indicate that MSC operate through intermediate effectors, as it was elegantly demonstrated by Bi et al. [29] who reproduced MSC effects by substituting MSC for their culture medium.
Aim of the present study was to identify possible effectors of the protection provided by MSC in the renal transplant model. Actually, the study consisted of a straightforward continuation of our former ones in which we demonstrated that MSC prevent the rejection [15],[44].
We have pointed out two systems: (i) the cytokine network that regulates the immune response in acute rejection, and (ii) the Scatter Factors systems that have been shown to promote repair and to modulate immune cell traffic in renal tissue in different models of kidney disease [36]-[42],[45]. In addition to confirming that MSC significantly decrease CD4, CD8 cells and monocytes infiltration [15], here we report that on day 7 of grafting in MSC untreated rats, e.g. rats with acute severe rejection IFN-γ and IL-10 levels rose impressively both in serum and in graft tissue, while MSC injection prevented the rise in IFN-γ and simultaneously it caused a further rise in IL-10 levels. Since IFN-γ is a nominal marker of Th1 lymphocyte activity, while IL-10 is Th2 cells product [46],[47], these results suggest that MSC reset the balance between the two T helper subpopulations, contrasting the prevalence of Th1 over Th2. MSC effects on T cell subsets included also a rise of Foxp3+ cells number, i.e. Treg lymphocytes in the transplanted kidney. This finding confirms that MSC induce Tregs, as shown in patients with SLE [48] and renal graft [49] in which circulating Tregs rose after MSC infusion and in a mouse model of renal transplantation in which pre-transplant MSC infusion was associated with a significant prolongation of graft survival by Treg-dependent mechanism [16]. Altogether the results of our study suggest that MSC reset T cell subpopulations, decreasing the prevalence of Th1 cells that are main effectors of rejection, increasing the activity of the immunosuppressive Th2 subset and recruiting tolerogenic Tregs. In addition to changing the T cell phenotypic distribution, MSC blocked IL-6 overproduction, a major inflammatory product of monocyte/macrophage cell and effector of acute rejection [50],[51]. Therefore, MSC suppressed the cytokines that drive the graft assault by the two major cell effectors of acute rejection, Th1 lymphocytes and monocytes.
A new information given by our study is that MSC have relevant effects on the Scatter Factor systems. In fact, in MSC untreated rats the HGF/Met couple was suppressed in serum (HGF) and in renal tissue (both HGF and Met). MSC prevented such abatement of HGF/Met, thus saving a system that has been proved to protect the kidney in diverse experimental models of renal disease. Actually, HGF has several features that facilitate renal healing: it stimulates proliferation and blocks apoptosis of injured tubular cells, it induces formation of new tubular structures in renal epithelial cells and generates new capillary vessels [52]-[54], it downregulates the inflammatory and immune response, e.g. by inactivating dendritic cells [55],[56], resetting cytokine network and addressing transformation of T cells to the Th2 phenotype [57]. HGF was shown also to interfere with the expression of immune co-stimulatory molecules, and to prolong survival of cardiac allograft by preventing acute rejection in a rodent model [58]. Furthermore, in a model of acute renal rejection, administration of recombinant HGF expanded Treg cell subset [59]. Indeed, there is an impressive overlap between the activities that are attributed to HGF and the changes induced by MSC in grafted rats, e.g. the shift of T cells to a tolerogenic phenotype, the rescue of tubular cells from death and the increase in tubular cell proliferation, that are the most distinctive features of HGF activity on renal tubular cells. Therefore, we believe that HGF/Met is a system that mediates the protective MSC effect on the kidney graft. Interestingly, MSC induced a rise in HGF levels also in another rat model of renal diseases, i.e. anti-Thy 1 nephritis [43] and ischemia-reperfusion injury [60] and in both models HGF overexpression was associated with prevention of renal damage.
In contrast to the abundant literature available on the pleiotropic activities of HGF/Met, the homologous factor MSP and its receptor RON are less known and their role in renal physiology and disease has been scarcely investigated. We have shown that tubular cells produce MSP and that MSP and RON are diffusely expressed in the normal kidney [33]. Studies in vitro have demonstrated that MSP induces in tubular cells proliferation, resistance to apoptosis, migration and branching morphogenesis, i.e. effects that altogether suggest a role of MSP expressed in the kidney as an autocrine/paracrine factor that protects survival and stimulates proliferation of tubular cells. In fact, MSP was shown to attenuate renal injury in the glycerol-induced model of acute renal failure [37]. In the present study we found that MSP and RON were suppressed in renal grafts not injected with MSC i.e. in the setting of unopposed acute rejection. MSC injection had significant effects on MSP/RON system that consisted of (i) recovery of MSP mRNA levels in tubular cells, (ii) recovery of MSP and RON expressed in tubular cells with MSP shifted in a different cell location, i.e. on the cell surface rather than inside the cytoplasm, (iii) suppression of RON in infiltrating monocyte/macrophage cells. We interpret this combination as a series of actions that benefit the kidney by (i) restarting the translation of MSP and the production of MSP protein in an amount that makes it detectable in the tubular cell (ii) shifting the MSP molecule to the cell surface, i.e. in a site where it can meet its receptor and possibly activate it in an autocrine fashion (iii) suppressing RON in monocytes thus restraining their ability to address tubular cells. As for the last effects, in order to understand the underlying mechanism we designed an in vitro experiment that demonstrated for the first time that MSC express constitutively mRNA of MSP in amount greater than that expressed by PBMC, i.e. a reference constitutive MSP producer [61]. RON expression in monocytes is induced by IFN-γ. Since MSC suppress IFN-γ both in serum and in renal tissue, it seems reasonable that this mechanism accounts for the absence of RON in monocytes infiltrating the renal graft injected with MSC.
Recent studies in a mouse model of renal transplantation have shown that MSC effects on graft function depend on the time of their injection, i.e. MSC worsen kidney function when injected after grafting, while they prevent rejection and ameliorate renal outcome when administered 1 or 7 days before the grafting [16]. These observations have a relevant impact on deciding the mode of MSC administration that should be used in clinical transplantation. We have chosen to inject MSC into the renal artery soon after reperfusion of the graft for two reasons: (i) to avoid changes in MSC activity caused by their transit in the lung, and (ii) to administer MSC in a setting that is easily reproducible in the circumstances of transplantation from a cadaver donor, in which the identity of the recipient is known just at the last minute. Our demonstration that this mode is feasible offers a potential advantage, mainly because, once our model is proven safe in humans, it allows to deliver MSC to recipients of kidney from cadaver donors that represent the majority of patients crowding the waiting lists. However, the two models are hardly comparable not only because of the different mode of administration, but also because the studies were performed in different species. Nonetheless it is interesting that both of them give useful information to programme use of MSC in humans, and both identify Tregs as effectors of MSC immunosuppressive action. In summary, we have demonstrated that the prevention of rejection provided by MSC is associated with a shift of T cells to an immune suppressive phenotype and with the recruitment of the Scatter Factor systems. These observations identify candidate mediators of MSC activity.

Methods

Animals and experimental model

The present study was carried out as a straightforward continuation of our former studies in a rat model of kidney transplant in which we demonstrated that MSC prevent severe rejection [15],[44]. In fact, we used specimens of serum and renal tissue that were sampled from the same rats and were stored on purpose. The design of the animal experiments is summarized in Figure 8. In brief, 11-week old Fisher F344 rats were used as kidney donors, 7-week old Lewis RT1 rats were used as recipients and Transgenic Sprague-Dawley rats (n = 5) expressing Enhanced Green Fluorescent Protein (EGFP) (Japan Slc, Hamamatsu, Japan) were used as MSC donors [62].
We studied an allogeneic model (Fisher F344 to Lewis RT1, 20 rats) of renal transplant in which both native kidneys were excised soon before grafting and no immunosuppressive drug therapy was administered. The experimental group consisted of ten rats that were infused into the artery of the grafted kidney with 3-106 MSC in 1 ml of PBS (Sigma Aldrich, St. Louis, MO, USA) (group C), soon after reperfusion. We used two control groups, one of normal not grafted rats (n = 5, group A), one of grafted rats that were infused into the graft artery with 1 ml of saline (n = 10, group B). Blood was drawn from the caudal vein at days 0 and day 7 and serum was stored at –20 00B0C. All the rats were sacrificed at day 7, the kidneys and spleens were removed and cut in two sagittal halves. One half was fixed in 10% formalin, the other one was frozen in liquid nitrogen.
Animal studies have been conducted after the approval by animal ethical committee of the University of Pavia.

Cytokine levels

IFN-γ, IL-1, IL-2, IL-4, IL-6, IL-10 serum levels were measured in rats of control group and 7 days after transplantation in allografted rats by ELISA (R&D Systems, Minneapolis, USA). The same cytokines were measured at day 7 in the graft by protein assay (Search Light Rat Cytokine Array), (Pierce Chemical Company, Rockford, IL, USA). All experiments were quadruplicated.

Renal morphology

Tubular necrosis, tubular cell proliferation, MSP, RON, Met, Foxp3, ED1, GFP antigen expression

3 μm thick sections cut along the sagittal plane of formalin-fixed kidney were stained with periodic acid-Schiff, and evaluated by two investigators in double blind fashion, using an Olympus IX8 microscope connected with a CCD camera and software imaging analysis Cell-R (Olympus America, Center Valley, PA, USA).
We scored for primary necrosis, i.e. necrosis that was not associated with tubulitis, 40 nonoverlapping high-power fields in 5 (HPF) not consecutive renal sections for each animal and quantified tubular necrosis as the ratio between necrotic tubules number and total tubules counted. A tubule was counted as having necrosis when tubular cells had one or more of the following features: isometric vacuolization, cell membrane rupture with loss of cytoplasm, nuclear fragmentation, detachment from basal membrane. Tubular cell proliferation was evaluated counting cells expressing Proliferating Cell Nuclear Antigen (PCNA). Renal expression of PCNA, MSP, RON, Met, ED1 antigen, Foxp3 and Green Fluorescent Protein (GFP) were studied by immunohistochemistry in formalin fixed tissue in 10 non-overlapping high-power fields of each section. The sections of paraffin embedded tissue were collected on poly-L-lysine-coated slides (Dako, Carpinteria, CA, USA), they were dewaxed in xylol, passed in a decreasing series of alcohol, and finally rehydrated with distilled water. Endogenous peroxidase was blocked with H2O2 3.7% vol/vol followed by H2O for 15 min. After 3 washings in 150 mM of PBS the sections underwent microwave antigen retrieval. Subsequently they were exposed overnight at 4° C to the following antibodies: 1) monoclonal mouse anti-PCNA antibody (Ab) (Santa Cruz Biotechnology, Santa Cruz, CA, USA), 1:200; 2) polyclonal goat anti-mouse MSP Ab (Santa Cruz Biotechnology), 1:600; 3) monoclonal mouse anti human RON Ab (Transduction Laboratories, Lexington, KY, USA), 1:750; 4) monoclonal mouse anti-human Met Ab (Novocastra Laboratories Ltd, Newcastle, UK), 1:20, 5) ED1 antigen, 1:80 (Serotec LtD, Oxford, UK), 6) anti Foxp3 Ab, 1:50 (eBioscience Ltd, Hatfield, UK), 7) monoclonal mouse anti-GFP antibody IgG1, diluted 1:1000 (Chemicon International, Temecula, CA, USA). After 3 washings in PBS the immunocomplex was visualized with the biotin-streptavidin-peroxidase complex and 3,3-diaminobenzidine (Dako, Glostrup, Denmark). Sections were faintly counterstained with Harris hematoxylin. Negative controls included both omission of the primary Ab and substitution of IgG for primary antibodies. Positive controls for EGFP positive cells were sections of kidney of EGFP rat. We counted the number of PCNA, Foxp3, GFP positive cells, MSP, RON, Met positive tubules number and RON positive interstitial cells in ten renal sections for each animal.

HGF serum levels

HGF serum levels were measured in control and allografted rats 7 days after transplantation by ELISA (Institute of immunology, Tokyo, Japan). All experiments were quadruplicated.

Western blot for HGF, Met

Tissue samples were frozen at −70° C in liquid nitrogen immediately following nephrectomy. Tissues were pulverized with a Mikro-Dismembrator (B.Braun Biotech International, Melsungen, Germany) in the presence of liquid nitrogen. The powdered whole tissues were washed twice with PBS and homogenized in ice cold buffer containing 10 mM PIPES, pH 6.8, 100 mMol/L NaCl, 5 mMol/L MgCl2, 300 mMol/L sucrose, 5 mMol/L ethylene-glycosbis-(β–aminoethyl ether)-N-N’-tetraacetic acid (DIM buffer), 1% Triton X-100, 100 mol/L sodium ortho-vanadate, and protease inhibitors (aprotinin 10 g/mL, pepstatin 10 g/mL, leupeptin 50 g/mL, soybean trypsin inhibitor 100 g/mL phenylmethanesulfonyl fluoride 1 mMol/L) (Sigma-Aldrich). Equal amounts of protein (800 g), determined using the BCA Protein Assay Reagent Kit (Pierce Chemical Company), were separated on 7,5% sodium dodecylsulfate-polyacrylamide gel electrophoresis and transferred to nitrocellulose Hybond filters (Amersham - GE Healthcare, Little Chalfont, Buckinghamshire, UK). Filters were probed with goat polyclonal anti HGF α Ab (c-20, Santa Cruz) that recognizes the α subunit of active heterodimer HGF, the rabbit polyclonal anti Met Ab that recognizes pro-Met and mature Met β subunit (c-28, Santa Cruz). The specific binding was detected by the enhanced chemiluminescence system ECL-Plus (Pierce Chemical Company).

In vitro experiments

Cell cultures

Peripheral blood mononuclear cells (PBMC) were isolated from rat blood by standard Ficoll hystopaque density gradient separation (Sigma-Aldrich). Monocytes were collected after 45 min of adherence to plastic culture plates and resuspended in RPMI 1640 medium, FCS 10%, and penicillin/streptomycin 1% (Invitrogen, Carlsbad, CA, USA) at 37° C in humidified 5% CO2 atmosphere.
To understand whether IFN-γ modified monocytes RON expression, we studied RON mRNA and RON protein in circulating monocytes soon after sampling and in monocytes cultured in absence and presence of LPS (10 g/ml, Sigma - Aldrich) and INF-γ (10 g/ml, R&D Systems) for 6 h and for 24 h respectively.

Monocytes RON mRNA, mesenchymal stem cells MSP mRNA, Renal MSP mRNA and HGF mRNA expression

Total RNA was extracted from renal tissue and cell cultures using respectively trizol method and guanidine-based RNeasy Mini Kit (QIAGEN GmbH, Hilden, Germany). All RNA was treated with DNase from RNase-Free DNase Set (QIAGEN) and dissolved in nuclease free water. Extracted RNA was tested for quantity and integrity by spectrophotometric analysis (NanoDrop - Thermo Scientific, Waltham, MA, USA). A total of 1 g of RNA per condition was reverse transcribed into complementary DNA (cDNA) through 1st Strand cDNA Synthesis Kit for Real Time (RT) -PCR (AMV) (Roche Applied Science, Penzber, Germany). cDNA was used to perform RT-PCR analysis in 96-well optical reaction plates, using ABI prism 5700 (Applied Biosystems, Foster City, CA, USA) and the 5-exonuclease assay (TaqMan technology) in a volume of 25 reaction containing TaqMan Universal Master Mix, optimized concentrations of FAM-labelled probe, and specific forward and reverse primers for beta-actin, HGF, MSP and RON selected from Assay on Demand (Applied Biosystems). Controls included RT-PCR with water replacing cDNA. The results were analysed using a comparative method, and the values were normalized to the beta-actin expression and converted into fold change, as previously described [63].

Expression of RON receptor in monocytes

A total of 10 ml of EDTA anticoagulated peripheral blood and 3X105 monocytes were incubated for 30 min with 10μ l of mouse anti rat RON mAb (Transduction Laboratories) diluted 1:10. Subsequently the samples were incubated for 30 min with 2μ l of goat anti mouse PE conjugated followed by incubation for 30 min with 2μ l of mouse anti rat CD11b FITC conjugated (BD Biosciences, San Jos, CA) to identify rat monocytes. Before examination samples of whole blood were treated with lysis buffer. All samples were resuspended in 500μ l of PBS and analyzed in FACScan operating with Cell Quest 3.3 software (BD Biosciences). Goat PE- conjugated IgG antibodies were used as negative controls.

Statistical analysis

ANOVA followed by the Newman-Keuls test or Kruskal-Wallis test and Student t test were used for comparison of the medians and means. All data was analysed using Graph Pad Prism software.

Authors’ contributions

MG contributed to the experimental design, renal histopathology studies, analysis of the results and statistical analysis. BF contributed to perform in vivo experiments, biochemical measurements. RC contributed to perform in vivo experiments, molecular and cell biology experiments. CV contributed to perform in vivo experiments, molecular and cell biology experiments. VT contributed to perform in vivo experiments, renal histopathology studies. PEF contributed to perform in vivo experiments, molecular and cell biology experiments. EP contributed to perform molecular and cell biology experiments. BG contributed to perform in vivo experiments, biochemical measurements. CC contributed to perform molecular biology experiments. LC contributed to perform statistical analysis, technical assistance. FF contributed to the experimental design. DCA contributed to the experimental design, final writing and revision of the paper. RT contributed to the experimental design, analysis of the results and writing of the paper. All authors read and approved the final manuscript.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.
Literatur
1.
Zurück zum Zitat Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop D, Horwitz E: Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement. Cytotherapy. 2006, 8: 315-317. 10.1080/14653240600855905.PubMedCrossRef Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop D, Horwitz E: Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement. Cytotherapy. 2006, 8: 315-317. 10.1080/14653240600855905.PubMedCrossRef
2.
Zurück zum Zitat Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR: Multilineage potential of adult human mesenchymal stem cells. Science. 2006, 284: 43-147. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR: Multilineage potential of adult human mesenchymal stem cells. Science. 2006, 284: 43-147.
3.
Zurück zum Zitat Keating A: Mesenchymal stromal cells. Curr Opin in Hematol. 2006, 13: 419-425. 10.1097/01.moh.0000245697.54887.6f.CrossRef Keating A: Mesenchymal stromal cells. Curr Opin in Hematol. 2006, 13: 419-425. 10.1097/01.moh.0000245697.54887.6f.CrossRef
4.
Zurück zum Zitat Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringdén O: HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol. 2003, 31: 890-896. 10.1016/S0301-472X(03)00110-3.PubMedCrossRef Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringdén O: HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol. 2003, 31: 890-896. 10.1016/S0301-472X(03)00110-3.PubMedCrossRef
5.
Zurück zum Zitat Aggarwal S, Pittenger MF: Human mesenchymal stem cells modulate allogenic immune cell response. Blood. 2005, 105: 1815-1822. 10.1182/blood-2004-04-1559.PubMedCrossRef Aggarwal S, Pittenger MF: Human mesenchymal stem cells modulate allogenic immune cell response. Blood. 2005, 105: 1815-1822. 10.1182/blood-2004-04-1559.PubMedCrossRef
6.
Zurück zum Zitat Le Blanc K, Ringdén O: Immunomodulation by mesenchymal stem cells and clinical experience. J Intern Med. 2007, 262: 509-525. 10.1111/j.1365-2796.2007.01844.x.PubMedCrossRef Le Blanc K, Ringdén O: Immunomodulation by mesenchymal stem cells and clinical experience. J Intern Med. 2007, 262: 509-525. 10.1111/j.1365-2796.2007.01844.x.PubMedCrossRef
7.
Zurück zum Zitat Comoli P, Ginevri F, Maccario R, Avanzini MA, Marconi M, Groff A, Cometa A, Cioni M, Porretti L, Barberi W, Frassoni F, Locatelli F: Human mesenchymal stem cells inhibit antibody production induced in vitro by allostimulation. Nephrol Dial Transplant. 2008, 23: 1196-1202. 10.1093/ndt/gfm740.PubMedCrossRef Comoli P, Ginevri F, Maccario R, Avanzini MA, Marconi M, Groff A, Cometa A, Cioni M, Porretti L, Barberi W, Frassoni F, Locatelli F: Human mesenchymal stem cells inhibit antibody production induced in vitro by allostimulation. Nephrol Dial Transplant. 2008, 23: 1196-1202. 10.1093/ndt/gfm740.PubMedCrossRef
8.
Zurück zum Zitat Corcione A, Benvenuto F, Ferretti E, Giunti D, Cappiello V, Cazzanti F, Risso M, Gualandi F, Mancardi GL, Pistoia V, Uccelli A: Human mesenchymal stem cells modulate B-cell functions. Blood. 2006, 107: 367-372. 10.1182/blood-2005-07-2657.PubMedCrossRef Corcione A, Benvenuto F, Ferretti E, Giunti D, Cappiello V, Cazzanti F, Risso M, Gualandi F, Mancardi GL, Pistoia V, Uccelli A: Human mesenchymal stem cells modulate B-cell functions. Blood. 2006, 107: 367-372. 10.1182/blood-2005-07-2657.PubMedCrossRef
9.
Zurück zum Zitat Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L: Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamina 2,3-dioxygenase and prostaglandin E2. Blood. 2008, 111: 1327-1333. 10.1182/blood-2007-02-074997.PubMedCrossRef Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L: Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamina 2,3-dioxygenase and prostaglandin E2. Blood. 2008, 111: 1327-1333. 10.1182/blood-2007-02-074997.PubMedCrossRef
10.
Zurück zum Zitat Le Blanc K, Tammik L, Sundberg B, Haynesworth SE, Ringdén O: Mesenchymal stem cells inhibit and stimulate mixed lymphocyte cultures and mitogenic responses independently of the major histocompatibility complex. Scand J Immunol. 2003, 57: 11-20. 10.1046/j.1365-3083.2003.01176.x.PubMedCrossRef Le Blanc K, Tammik L, Sundberg B, Haynesworth SE, Ringdén O: Mesenchymal stem cells inhibit and stimulate mixed lymphocyte cultures and mitogenic responses independently of the major histocompatibility complex. Scand J Immunol. 2003, 57: 11-20. 10.1046/j.1365-3083.2003.01176.x.PubMedCrossRef
11.
Zurück zum Zitat Yi T, Song SU: Immunomodulatory properties of mesenchymal stem cells and their therapeutic applications. Arch Pharm Res. 2012, 35: 213-221. 10.1007/s12272-012-0202-z.PubMedCrossRef Yi T, Song SU: Immunomodulatory properties of mesenchymal stem cells and their therapeutic applications. Arch Pharm Res. 2012, 35: 213-221. 10.1007/s12272-012-0202-z.PubMedCrossRef
12.
Zurück zum Zitat Prevosto C, Zancolli M, Carnevali P, Zocchi MR, Poggi A: Generation of CD4+ or CD8+ regulatory T cells upon mesenchymal stem cell-lymphocyte interaction. Haematologica. 2007, 92: 881-888. 10.3324/haematol.11240.PubMedCrossRef Prevosto C, Zancolli M, Carnevali P, Zocchi MR, Poggi A: Generation of CD4+ or CD8+ regulatory T cells upon mesenchymal stem cell-lymphocyte interaction. Haematologica. 2007, 92: 881-888. 10.3324/haematol.11240.PubMedCrossRef
13.
Zurück zum Zitat Casiraghi F, Perico N, Remuzzi G: Mesenchymal stromal cells to promote solid organ transplantation tolerance. Curr Opin Organ Transplant. 2013, 18: 51-58. 10.1097/MOT.0b013e32835c5016.PubMedCrossRef Casiraghi F, Perico N, Remuzzi G: Mesenchymal stromal cells to promote solid organ transplantation tolerance. Curr Opin Organ Transplant. 2013, 18: 51-58. 10.1097/MOT.0b013e32835c5016.PubMedCrossRef
14.
Zurück zum Zitat Duffy MM, Ritter T, Ceredig R, Griffin MD: Mesenchymal stem cell effects on T-cell effector pathways. Stem Cell Res Ther. 2011, 2: 34-10.1186/scrt75.PubMedPubMedCentralCrossRef Duffy MM, Ritter T, Ceredig R, Griffin MD: Mesenchymal stem cell effects on T-cell effector pathways. Stem Cell Res Ther. 2011, 2: 34-10.1186/scrt75.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat De Martino M, Zonta S, Rampino T, Gregorini M, Frassoni F, Piotti G, Bedino G, Cobianchi L, Dal Canton A, Dionigi P, Alessiani M: Mesenchymal stem cells infusion prevents acute cellular rejection in rat kidney transplantation. Transplant Proc. 2010, 42: 1331-1335. 10.1016/j.transproceed.2010.03.079.PubMedCrossRef De Martino M, Zonta S, Rampino T, Gregorini M, Frassoni F, Piotti G, Bedino G, Cobianchi L, Dal Canton A, Dionigi P, Alessiani M: Mesenchymal stem cells infusion prevents acute cellular rejection in rat kidney transplantation. Transplant Proc. 2010, 42: 1331-1335. 10.1016/j.transproceed.2010.03.079.PubMedCrossRef
16.
Zurück zum Zitat Casiraghi F, Azzollini N, Todeschini M, Cavinato RA, Cassis P, Solini S, Rota C, Morigi M, Introna M, Maranta R, Perico N, Remuzzi G, Noris M: Localization of mesenchymal stromal cells dictates their immune or proinflammatory effects in kidney transplantation. Am J Transplant. 2012, 12: 2373-2383. 10.1111/j.1600-6143.2012.04115.x.PubMedCrossRef Casiraghi F, Azzollini N, Todeschini M, Cavinato RA, Cassis P, Solini S, Rota C, Morigi M, Introna M, Maranta R, Perico N, Remuzzi G, Noris M: Localization of mesenchymal stromal cells dictates their immune or proinflammatory effects in kidney transplantation. Am J Transplant. 2012, 12: 2373-2383. 10.1111/j.1600-6143.2012.04115.x.PubMedCrossRef
17.
Zurück zum Zitat Morigi M, Imberti B, Zoja C, Corna D, Tomasoni S, Abbate M, Rottoli D, Angioletti S, Benigni A, Perico N, Alison M, Remuzzi G: Mesenchymal stem cells are renotropic, helping to repair the kidney and improve function in acute renal failure. J Am Soc Nephrol. 2004, 15: 1794-1804. 10.1097/01.ASN.0000128974.07460.34.PubMedCrossRef Morigi M, Imberti B, Zoja C, Corna D, Tomasoni S, Abbate M, Rottoli D, Angioletti S, Benigni A, Perico N, Alison M, Remuzzi G: Mesenchymal stem cells are renotropic, helping to repair the kidney and improve function in acute renal failure. J Am Soc Nephrol. 2004, 15: 1794-1804. 10.1097/01.ASN.0000128974.07460.34.PubMedCrossRef
18.
Zurück zum Zitat Herrera MB, Bussolati B, Bruno S, Fonsato V, Romanazzi GM, Camussi G: Mesenchymal stem cells contribute to the renal repair of acute tubular epithelial injury. Int J Mol Med. 2004, 14: 1035-1041.PubMed Herrera MB, Bussolati B, Bruno S, Fonsato V, Romanazzi GM, Camussi G: Mesenchymal stem cells contribute to the renal repair of acute tubular epithelial injury. Int J Mol Med. 2004, 14: 1035-1041.PubMed
19.
Zurück zum Zitat Lange C, Tögel F, Ittrich H, Clayton F, Nolte-Ernsting C, Zander AR, Westenfelder C: Administered mesenchymal stem cells enhance recovery from ischemia/reperfusion-induced acute renal failure. Kidney Int. 2005, 68: 1613-1617. 10.1111/j.1523-1755.2005.00573.x.PubMedCrossRef Lange C, Tögel F, Ittrich H, Clayton F, Nolte-Ernsting C, Zander AR, Westenfelder C: Administered mesenchymal stem cells enhance recovery from ischemia/reperfusion-induced acute renal failure. Kidney Int. 2005, 68: 1613-1617. 10.1111/j.1523-1755.2005.00573.x.PubMedCrossRef
20.
Zurück zum Zitat Bussolati B, Hauser PV, Carvalhosa R, Camussi G: Contribution of stem cells to kidney repair. Curr Stem Cell Res Ther. 2009, 4: 2-8. 10.2174/157488809787169129.PubMedCrossRef Bussolati B, Hauser PV, Carvalhosa R, Camussi G: Contribution of stem cells to kidney repair. Curr Stem Cell Res Ther. 2009, 4: 2-8. 10.2174/157488809787169129.PubMedCrossRef
21.
Zurück zum Zitat Chhabra P, Brayman KL: The use of stem cells in kidney disease. Curr Opin Organ Transplant. 2009, 14: 72-78. 10.1097/MOT.0b013e328320d2f5.PubMedCrossRef Chhabra P, Brayman KL: The use of stem cells in kidney disease. Curr Opin Organ Transplant. 2009, 14: 72-78. 10.1097/MOT.0b013e328320d2f5.PubMedCrossRef
22.
Zurück zum Zitat Tögel F, Cohen A, Zhang P, Yang Y, Hu Z, Westenfelder C: Autologous and allogeneic marrow stromal cells are safe and effective for treatment of acute kidney injury. Stem Cells Dev. 2009, 18: 475-485. 10.1089/scd.2008.0092.PubMedPubMedCentralCrossRef Tögel F, Cohen A, Zhang P, Yang Y, Hu Z, Westenfelder C: Autologous and allogeneic marrow stromal cells are safe and effective for treatment of acute kidney injury. Stem Cells Dev. 2009, 18: 475-485. 10.1089/scd.2008.0092.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Choi S, Park M, Kim J, Hwang S, Park S, Lee Y: The role of mesenchymal stem cells in the functional improvement of chronic renal failure. Stem Cells Dev. 2009, 18: 521-529. 10.1089/scd.2008.0097.PubMedCrossRef Choi S, Park M, Kim J, Hwang S, Park S, Lee Y: The role of mesenchymal stem cells in the functional improvement of chronic renal failure. Stem Cells Dev. 2009, 18: 521-529. 10.1089/scd.2008.0097.PubMedCrossRef
24.
Zurück zum Zitat Doorn J, Moll G, Le Blanc K, van Blitterswijk C, de Boer J: Therapeutic applications of mesenchymal stromal cells: paracrine effects and potential improvements. Tissue Eng Part B Rev. 2012, 18: 101-115. 10.1089/ten.teb.2011.0488.PubMedCrossRef Doorn J, Moll G, Le Blanc K, van Blitterswijk C, de Boer J: Therapeutic applications of mesenchymal stromal cells: paracrine effects and potential improvements. Tissue Eng Part B Rev. 2012, 18: 101-115. 10.1089/ten.teb.2011.0488.PubMedCrossRef
25.
Zurück zum Zitat English K: Mechanisms of mesenchymal stromal cell immunomodulation. Immunol Cell Biol. 2013, 91: 19-26. 10.1038/icb.2012.56.PubMedCrossRef English K: Mechanisms of mesenchymal stromal cell immunomodulation. Immunol Cell Biol. 2013, 91: 19-26. 10.1038/icb.2012.56.PubMedCrossRef
26.
Zurück zum Zitat Casiraghi F, Noris M, Remuzzi G: Immunomodulatory effects of mesenchymal stromal cells in solid organ transplantation. Curr Opin Organ Transplant. 2010, 24: 731-737. 10.1097/MOT.0b013e328340172c.CrossRef Casiraghi F, Noris M, Remuzzi G: Immunomodulatory effects of mesenchymal stromal cells in solid organ transplantation. Curr Opin Organ Transplant. 2010, 24: 731-737. 10.1097/MOT.0b013e328340172c.CrossRef
27.
Zurück zum Zitat Abumaree M, Al Jumah M, Pace RA, Kalionis B: Immunosuppressive properties of mesenchymal stem cells. Stem Cell Rev. 2012, 8: 375-392. 10.1007/s12015-011-9312-0.PubMedCrossRef Abumaree M, Al Jumah M, Pace RA, Kalionis B: Immunosuppressive properties of mesenchymal stem cells. Stem Cell Rev. 2012, 8: 375-392. 10.1007/s12015-011-9312-0.PubMedCrossRef
28.
Zurück zum Zitat Tögel F, Hu Z, Weiss K, Isaac J, Lange C, Westenfelder C: Administered mesenchymal stem cells protect against ischemic acute renal failure through differentiation-independent mechanisms. Am J Physiol Renal Physiol. 2005, 289: F31-F42. 10.1152/ajprenal.00007.2005.PubMedCrossRef Tögel F, Hu Z, Weiss K, Isaac J, Lange C, Westenfelder C: Administered mesenchymal stem cells protect against ischemic acute renal failure through differentiation-independent mechanisms. Am J Physiol Renal Physiol. 2005, 289: F31-F42. 10.1152/ajprenal.00007.2005.PubMedCrossRef
29.
Zurück zum Zitat Bi B, Schmitt R, Israilova M, Nishio H, Cantley LG: Stromal cells protect against acute tubular injury via an endocrine effect. J Am Soc Nephrol. 2007, 18: 2486-2496. 10.1681/ASN.2007020140.PubMedCrossRef Bi B, Schmitt R, Israilova M, Nishio H, Cantley LG: Stromal cells protect against acute tubular injury via an endocrine effect. J Am Soc Nephrol. 2007, 18: 2486-2496. 10.1681/ASN.2007020140.PubMedCrossRef
30.
Zurück zum Zitat Bruno S, Grange C, Deregibus MC, Calogero RA, Saviozzi S, Collino F, Morando L, Busca A, Falda M, Bussolati B, Tetta C, Camussi G: Mesenchymal stem cell-derived microvescicles protect against acute tubular injury. J Am Soc Nephrol. 2009, 20: 1053-1067. 10.1681/ASN.2008070798.PubMedPubMedCentralCrossRef Bruno S, Grange C, Deregibus MC, Calogero RA, Saviozzi S, Collino F, Morando L, Busca A, Falda M, Bussolati B, Tetta C, Camussi G: Mesenchymal stem cell-derived microvescicles protect against acute tubular injury. J Am Soc Nephrol. 2009, 20: 1053-1067. 10.1681/ASN.2008070798.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Camussi G, Deregibus MC, Tetta C: Paracrine/endocrine mechanism of stem cells on kidney repair: role of microvescicle-mediated transfer of genetic information. Curr Opin Nephrol Hypertens. 2010, 19: 7-12. 10.1097/MNH.0b013e328332fb6f.PubMedCrossRef Camussi G, Deregibus MC, Tetta C: Paracrine/endocrine mechanism of stem cells on kidney repair: role of microvescicle-mediated transfer of genetic information. Curr Opin Nephrol Hypertens. 2010, 19: 7-12. 10.1097/MNH.0b013e328332fb6f.PubMedCrossRef
32.
Zurück zum Zitat Soleymaninejadian E, Pramanik K, Samadian E: Immunomodulatory properties of mesenchymal stem cells: cytokines and factors. Am J Reprod Immunol. 2012, 67: 1-8. 10.1111/j.1600-0897.2011.01069.x.PubMedCrossRef Soleymaninejadian E, Pramanik K, Samadian E: Immunomodulatory properties of mesenchymal stem cells: cytokines and factors. Am J Reprod Immunol. 2012, 67: 1-8. 10.1111/j.1600-0897.2011.01069.x.PubMedCrossRef
33.
Zurück zum Zitat Rampino T, Collesi C, Gregorini M, Maggio M, Soccio G, Guallini P, Guallini P, Dal Canton A: Macrophage-stimulating protein is produced by tubular cells and activates mesangial cells. J Am Soc Nephrol. 2002, 13: 649-657.PubMed Rampino T, Collesi C, Gregorini M, Maggio M, Soccio G, Guallini P, Guallini P, Dal Canton A: Macrophage-stimulating protein is produced by tubular cells and activates mesangial cells. J Am Soc Nephrol. 2002, 13: 649-657.PubMed
34.
Zurück zum Zitat Cantley LG, Barros EJ, Gandhi M, Rauchman M, Nigam SK: Regulation of mitogenesis, motogenesis, and tubulogenesis by hepatocyte growth factor in renal collecting duct cells. Am J Physiol. 1994, 267: F271-F280.PubMed Cantley LG, Barros EJ, Gandhi M, Rauchman M, Nigam SK: Regulation of mitogenesis, motogenesis, and tubulogenesis by hepatocyte growth factor in renal collecting duct cells. Am J Physiol. 1994, 267: F271-F280.PubMed
35.
Zurück zum Zitat Liu Y: Hepatocyte growth factor and the kidney. Curr Opin Nephrol Hypertens. 2002, 11: 23-30. 10.1097/00041552-200201000-00004.PubMedCrossRef Liu Y: Hepatocyte growth factor and the kidney. Curr Opin Nephrol Hypertens. 2002, 11: 23-30. 10.1097/00041552-200201000-00004.PubMedCrossRef
36.
Zurück zum Zitat Rampino T, Soccio G, Gregorini M, Guidetti C, Marasà M, Maggio M, Panichi V, Migliori M, Libetta C, Dal Canton A: Neutralization of macrophage-stimulating protein ameliorates renal injury in anti-thy 1 glomerulonephritis. J Am Soc Nephrol. 2007, 18: 1486-1496. 10.1681/ASN.2006060680.PubMedCrossRef Rampino T, Soccio G, Gregorini M, Guidetti C, Marasà M, Maggio M, Panichi V, Migliori M, Libetta C, Dal Canton A: Neutralization of macrophage-stimulating protein ameliorates renal injury in anti-thy 1 glomerulonephritis. J Am Soc Nephrol. 2007, 18: 1486-1496. 10.1681/ASN.2006060680.PubMedCrossRef
37.
Zurück zum Zitat Cantaluppi V, Biancone L, Romanazzi GM, Figliolini F, Beltramo S, Galimi F, Camboni MG, Deriu E, Conaldi P, Bottelli A, Orlandi V, Herrera MB, Pacitti A, Segoloni GP, Camussi G: Macrophage stimulating protein may promote tubular regeneration after acute injury. J Am Soc Nephrol. 2008, 19: 1904-1918. 10.1681/ASN.2007111209.PubMedPubMedCentralCrossRef Cantaluppi V, Biancone L, Romanazzi GM, Figliolini F, Beltramo S, Galimi F, Camboni MG, Deriu E, Conaldi P, Bottelli A, Orlandi V, Herrera MB, Pacitti A, Segoloni GP, Camussi G: Macrophage stimulating protein may promote tubular regeneration after acute injury. J Am Soc Nephrol. 2008, 19: 1904-1918. 10.1681/ASN.2007111209.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Homsi E, Janino P, Amano M, Saraiva Camara NO: Endogenous hepatocyte growth factor attenuates inflammatory response in glycerol-induced acute kidney injury. Am J Nephrol. 2009, 29: 283-291. 10.1159/000159275.PubMedCrossRef Homsi E, Janino P, Amano M, Saraiva Camara NO: Endogenous hepatocyte growth factor attenuates inflammatory response in glycerol-induced acute kidney injury. Am J Nephrol. 2009, 29: 283-291. 10.1159/000159275.PubMedCrossRef
39.
Zurück zum Zitat Mori T, Shimizu A, Masuda Y, Fukuda Y, Yamanaka N: Hepatocyte growth factor-stimulating endothelial cell growth and accelerating glomerular capillary repair in experimental progressive glomerulonephritis. Nephron Exp Nephrol. 2003, 94: e44-e54. 10.1159/000071283.PubMedCrossRef Mori T, Shimizu A, Masuda Y, Fukuda Y, Yamanaka N: Hepatocyte growth factor-stimulating endothelial cell growth and accelerating glomerular capillary repair in experimental progressive glomerulonephritis. Nephron Exp Nephrol. 2003, 94: e44-e54. 10.1159/000071283.PubMedCrossRef
40.
Zurück zum Zitat Takada S, Namiki M, Takahara S, Matsumiya K, Kondoh N, Kokado Y, Matsumoto K, Nakamura T, Okuyama A: Serum HGF levels in acute renal rejection after living related renal transplantation. Transpl Int. 1996, 9: 151-154. 10.1111/j.1432-2277.1996.tb00870.x.PubMedCrossRef Takada S, Namiki M, Takahara S, Matsumiya K, Kondoh N, Kokado Y, Matsumoto K, Nakamura T, Okuyama A: Serum HGF levels in acute renal rejection after living related renal transplantation. Transpl Int. 1996, 9: 151-154. 10.1111/j.1432-2277.1996.tb00870.x.PubMedCrossRef
41.
Zurück zum Zitat Yoshimura R, Watanabe Y, Kasai S, Wada S, Ohyama A, Hase T, Nakatani T, Chargui J, Touraine JL, Nakamura T: Hepatocyte growth factor (HGF) as a rapid diagnostic marker and its potential in the prevention of acute renal rejection. Transpl Int. 2002, 15: 156-162. 10.1111/j.1432-2277.2002.tb00146.x.PubMedCrossRef Yoshimura R, Watanabe Y, Kasai S, Wada S, Ohyama A, Hase T, Nakatani T, Chargui J, Touraine JL, Nakamura T: Hepatocyte growth factor (HGF) as a rapid diagnostic marker and its potential in the prevention of acute renal rejection. Transpl Int. 2002, 15: 156-162. 10.1111/j.1432-2277.2002.tb00146.x.PubMedCrossRef
42.
Zurück zum Zitat Rampino T, Gregorini M, Dal Canton A: Scatter factors in renal disease: Dr. Jeckyll and Mr. Hyde?. Cytokine Growth Factor Rev. 2009, 20: 77-85. 10.1016/j.cytogfr.2009.01.004.PubMedCrossRef Rampino T, Gregorini M, Dal Canton A: Scatter factors in renal disease: Dr. Jeckyll and Mr. Hyde?. Cytokine Growth Factor Rev. 2009, 20: 77-85. 10.1016/j.cytogfr.2009.01.004.PubMedCrossRef
43.
Zurück zum Zitat Rampino T, Gregorini M, Bedino G, Piotti G, Gabanti E, Ibatici A, Sessarego N, Piacenza C, Balenzano CT, Esposito P, Bosio F, Soccio G, Frassoni F, Dal Canton A: Mesenchymal stromal cells improve renal injury in anti-Thy 1 nephritis by modulating inflammatory cytokines and scatter factors. Clin Sci (Lond). 2011, 120: 25-36. 10.1042/CS20100147.CrossRef Rampino T, Gregorini M, Bedino G, Piotti G, Gabanti E, Ibatici A, Sessarego N, Piacenza C, Balenzano CT, Esposito P, Bosio F, Soccio G, Frassoni F, Dal Canton A: Mesenchymal stromal cells improve renal injury in anti-Thy 1 nephritis by modulating inflammatory cytokines and scatter factors. Clin Sci (Lond). 2011, 120: 25-36. 10.1042/CS20100147.CrossRef
44.
Zurück zum Zitat Zonta S, De Martino M, Bedino G, Piotti G, Rampino T, Gregorini M, Frassoni F, Dal Canton A, Dionigi P, Alessiani M: Which is the most suitable and effective route of administration for mesenchymal stem cell-based immunomodulation therapy in experimental kidney transplantation: endovenous or arterial?. Transplant Proc. 2010, 42: 1336-1340. 10.1016/j.transproceed.2010.03.081.PubMedCrossRef Zonta S, De Martino M, Bedino G, Piotti G, Rampino T, Gregorini M, Frassoni F, Dal Canton A, Dionigi P, Alessiani M: Which is the most suitable and effective route of administration for mesenchymal stem cell-based immunomodulation therapy in experimental kidney transplantation: endovenous or arterial?. Transplant Proc. 2010, 42: 1336-1340. 10.1016/j.transproceed.2010.03.081.PubMedCrossRef
45.
Zurück zum Zitat Mizuno S, Nakamura T: Prevention of neutrophil extravasation by hepatocyte growth factor leads to attenuations of tubular apoptosis and renal dysfunction in mouse ischemic kidneys. Am J Pathol. 2005, 166: 1895-1905. 10.1016/S0002-9440(10)62498-4.PubMedPubMedCentralCrossRef Mizuno S, Nakamura T: Prevention of neutrophil extravasation by hepatocyte growth factor leads to attenuations of tubular apoptosis and renal dysfunction in mouse ischemic kidneys. Am J Pathol. 2005, 166: 1895-1905. 10.1016/S0002-9440(10)62498-4.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat O’Garra A, Murphy K: T-cell subsets in autoimmunity. Curr Opin Immunol. 1993, 5: 880-886. 10.1016/0952-7915(93)90100-7.PubMedCrossRef O’Garra A, Murphy K: T-cell subsets in autoimmunity. Curr Opin Immunol. 1993, 5: 880-886. 10.1016/0952-7915(93)90100-7.PubMedCrossRef
47.
Zurück zum Zitat O’Garra A, Vieira P: Regulatory T cells and mechanisms of immune system control. Nat Med. 2004, 10: 801-805. 10.1038/nm0804-801.PubMedCrossRef O’Garra A, Vieira P: Regulatory T cells and mechanisms of immune system control. Nat Med. 2004, 10: 801-805. 10.1038/nm0804-801.PubMedCrossRef
48.
Zurück zum Zitat Carrion F, Nova E, Ruiz C, Diaz F, Inostroza C, Rojo D, Mnckeberg G, Figueroa FE: Autologous mesenchymal stem cell treatment increased T regulatory cells with no effect on disease activity in two systemic lupus erythematosus patients. Lupus. 2010, 19: 317-322. 10.1177/0961203309348983.PubMedCrossRef Carrion F, Nova E, Ruiz C, Diaz F, Inostroza C, Rojo D, Mnckeberg G, Figueroa FE: Autologous mesenchymal stem cell treatment increased T regulatory cells with no effect on disease activity in two systemic lupus erythematosus patients. Lupus. 2010, 19: 317-322. 10.1177/0961203309348983.PubMedCrossRef
49.
Zurück zum Zitat Perico N, Casiraghi F, Introna M, Gotti E, Todeschini M, Cavinato RA, Capelli C, Rambaldi A, Cassis P, Rizzo P, Cortinovis M, Marasà M, Golay J, Noris M, Remuzzi G: Autologous mesenchymal stromal cells and kidney transplantation: a pilot study of safety and clinical feasibility. Clin J Am Soc Nephrol. 2011, 6: 412-422. 10.2215/CJN.04950610.PubMedPubMedCentralCrossRef Perico N, Casiraghi F, Introna M, Gotti E, Todeschini M, Cavinato RA, Capelli C, Rambaldi A, Cassis P, Rizzo P, Cortinovis M, Marasà M, Golay J, Noris M, Remuzzi G: Autologous mesenchymal stromal cells and kidney transplantation: a pilot study of safety and clinical feasibility. Clin J Am Soc Nephrol. 2011, 6: 412-422. 10.2215/CJN.04950610.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Shen H, Goldstein DR: IL-6 and TNF-α synergistically inhibit allograft acceptance. J Am Soc Nephrol. 2009, 20: 1032-1040. 10.1681/ASN.2008070778.PubMedPubMedCentralCrossRef Shen H, Goldstein DR: IL-6 and TNF-α synergistically inhibit allograft acceptance. J Am Soc Nephrol. 2009, 20: 1032-1040. 10.1681/ASN.2008070778.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Nakagiri T, Inoue M, Minami M, Shintani Y, Okumura M: Immunology mini-review: the basics of T(H)17 and interleukin-6 in transplantation. Transplant Proc. 2012, 44: 1035-1040. 10.1016/j.transproceed.2011.12.032.PubMedCrossRef Nakagiri T, Inoue M, Minami M, Shintani Y, Okumura M: Immunology mini-review: the basics of T(H)17 and interleukin-6 in transplantation. Transplant Proc. 2012, 44: 1035-1040. 10.1016/j.transproceed.2011.12.032.PubMedCrossRef
52.
Zurück zum Zitat Nakamura T, Sakai K, Nakamura T, Matsumoto K: Hepatocyte growth factor twenty years on: Much more than a growth factor. J Gastroenterol Hepatol. 2011, 26 (Suppl 1): 188-202. 10.1111/j.1440-1746.2010.06549.x.PubMedCrossRef Nakamura T, Sakai K, Nakamura T, Matsumoto K: Hepatocyte growth factor twenty years on: Much more than a growth factor. J Gastroenterol Hepatol. 2011, 26 (Suppl 1): 188-202. 10.1111/j.1440-1746.2010.06549.x.PubMedCrossRef
53.
Zurück zum Zitat Vargas GA, Hoeflich A, Jehle PM: Hepatocyte growth factor in renal failure: promise and reality. Kidney Int. 2000, 57: 1426-1436. 10.1046/j.1523-1755.2000.00987.x.PubMedCrossRef Vargas GA, Hoeflich A, Jehle PM: Hepatocyte growth factor in renal failure: promise and reality. Kidney Int. 2000, 57: 1426-1436. 10.1046/j.1523-1755.2000.00987.x.PubMedCrossRef
54.
Zurück zum Zitat Matsumoto K, Nakamura T: Hepatocyte growth factor: renotropic role and potential therapeutics for renal diseases. Kidney Int. 2001, 59: 2023-2038. 10.1046/j.1523-1755.2001.00717.x.PubMedCrossRef Matsumoto K, Nakamura T: Hepatocyte growth factor: renotropic role and potential therapeutics for renal diseases. Kidney Int. 2001, 59: 2023-2038. 10.1046/j.1523-1755.2001.00717.x.PubMedCrossRef
55.
Zurück zum Zitat Okunishi K, Dohi M, Nakagome K, Tanaka R, Mizuno S, Matsumoto K, Miyazaki J, Nakamura T, Yamamoto K: A novel role of hepatocyte growth factor as an immune regulator through suppressing dendritic cell function. J Immunol. 2005, 175: 4745-4753. 10.4049/jimmunol.175.7.4745.PubMedCrossRef Okunishi K, Dohi M, Nakagome K, Tanaka R, Mizuno S, Matsumoto K, Miyazaki J, Nakamura T, Yamamoto K: A novel role of hepatocyte growth factor as an immune regulator through suppressing dendritic cell function. J Immunol. 2005, 175: 4745-4753. 10.4049/jimmunol.175.7.4745.PubMedCrossRef
56.
Zurück zum Zitat Rutella S, Bonanno G, Procoli A, Mariotti A, de Ritis DG, Curti A, Danese S, Pessina G, Pandolfi S, Natoni F, Di Febo A, Scambia G, Manfredini R, Salati S, Ferrari S, Pierelli L, Leone G, Lemoli RM: Hepatocyte growth factor favors monocyte differentiation into regulatory interleukin (IL)-10++IL-12low/neg accessory cells with dendritic-cell features. Blood. 2006, 108: 218-227. 10.1182/blood-2005-08-3141.PubMedCrossRef Rutella S, Bonanno G, Procoli A, Mariotti A, de Ritis DG, Curti A, Danese S, Pessina G, Pandolfi S, Natoni F, Di Febo A, Scambia G, Manfredini R, Salati S, Ferrari S, Pierelli L, Leone G, Lemoli RM: Hepatocyte growth factor favors monocyte differentiation into regulatory interleukin (IL)-10++IL-12low/neg accessory cells with dendritic-cell features. Blood. 2006, 108: 218-227. 10.1182/blood-2005-08-3141.PubMedCrossRef
57.
Zurück zum Zitat Futamatsu H, Suzuki J, Mizuno S, Koga N, Adachi S, Kosuge H, Maejima Y, Hirao K, Nakamura T, Isobe M: Hepatocyte growth factor ameliorates the progression of experimental autoimmune myocarditis: a potential role for induction of T helper 2 cytokines. Circ Res. 2005, 96: 823-830. 10.1161/01.RES.0000163016.52653.2e.PubMedCrossRef Futamatsu H, Suzuki J, Mizuno S, Koga N, Adachi S, Kosuge H, Maejima Y, Hirao K, Nakamura T, Isobe M: Hepatocyte growth factor ameliorates the progression of experimental autoimmune myocarditis: a potential role for induction of T helper 2 cytokines. Circ Res. 2005, 96: 823-830. 10.1161/01.RES.0000163016.52653.2e.PubMedCrossRef
58.
Zurück zum Zitat Yamaura K, Ito K, Tsukioka K, Wada Y, Makiuchi A, Sakaguchi M, Akashima T, Fujimori M, Sawa Y, Morishita R, Matsumoto K, Nakamura T, Suzuki J, Amano J, Isobe M: Suppression of acute and chronic rejection by hepatocyte growth factor in a murine model of cardiac transplantation: induction of tolerance and prevention of cardiac allograft vasculopathy. Circulation. 2004, 110: 1650-1657. 10.1161/01.CIR.0000143052.45956.71.PubMedCrossRef Yamaura K, Ito K, Tsukioka K, Wada Y, Makiuchi A, Sakaguchi M, Akashima T, Fujimori M, Sawa Y, Morishita R, Matsumoto K, Nakamura T, Suzuki J, Amano J, Isobe M: Suppression of acute and chronic rejection by hepatocyte growth factor in a murine model of cardiac transplantation: induction of tolerance and prevention of cardiac allograft vasculopathy. Circulation. 2004, 110: 1650-1657. 10.1161/01.CIR.0000143052.45956.71.PubMedCrossRef
59.
Zurück zum Zitat Oku M, Okumi M, Shimizu A, Sahara H, Setoyama K, Nishimura H, Sada M, Scalea J, Ido A, Sachs DH, Tsubouchi H, Yamada K: Hepatocyte growth factor sustains T regulatory cells and prolongs the survival of kidney allografts in major histocompatibility complex-inbred CLAWN-miniature swine. Transplantation. 2012, 93: 148-155. 10.1097/TP.0b013e31823be83f.PubMedCrossRef Oku M, Okumi M, Shimizu A, Sahara H, Setoyama K, Nishimura H, Sada M, Scalea J, Ido A, Sachs DH, Tsubouchi H, Yamada K: Hepatocyte growth factor sustains T regulatory cells and prolongs the survival of kidney allografts in major histocompatibility complex-inbred CLAWN-miniature swine. Transplantation. 2012, 93: 148-155. 10.1097/TP.0b013e31823be83f.PubMedCrossRef
60.
Zurück zum Zitat Du T, Zou X, Cheng J, Wu S, Zhong L, Ju G, Zhu J, Liu G, Zhu Y, Xia S: Human Wharton’s jelly-derived mesenchymal stromal cells reduce renal fibrosis through induction of native and foreign hepatocyte growth factor synthesis in injured tubular epithelial cells. Stem Cell Res Ther. 2013, 4: 59-10.1186/scrt215.PubMedPubMedCentralCrossRef Du T, Zou X, Cheng J, Wu S, Zhong L, Ju G, Zhu J, Liu G, Zhu Y, Xia S: Human Wharton’s jelly-derived mesenchymal stromal cells reduce renal fibrosis through induction of native and foreign hepatocyte growth factor synthesis in injured tubular epithelial cells. Stem Cell Res Ther. 2013, 4: 59-10.1186/scrt215.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Cowin AJ, Kallincos N, Hatzirodos N, Robertson JG, Pickering KJ, Couper J, Belford DA: Hepatocyte growth factor and macrophage-stimulating protein are upregulated during excisional wound repair in rats. Cell Tissue Res. 2001, 306: 239-250. 10.1007/s004410100443.PubMedCrossRef Cowin AJ, Kallincos N, Hatzirodos N, Robertson JG, Pickering KJ, Couper J, Belford DA: Hepatocyte growth factor and macrophage-stimulating protein are upregulated during excisional wound repair in rats. Cell Tissue Res. 2001, 306: 239-250. 10.1007/s004410100443.PubMedCrossRef
62.
Zurück zum Zitat Okabe M, Ikawa M, Kominami K, Nakanishi T, Nishimune Y: `Green mice as a source of ubiquitous green cells. FEBS Lett. 1997, 407: 313-319. 10.1016/S0014-5793(97)00313-X.PubMedCrossRef Okabe M, Ikawa M, Kominami K, Nakanishi T, Nishimune Y: `Green mice as a source of ubiquitous green cells. FEBS Lett. 1997, 407: 313-319. 10.1016/S0014-5793(97)00313-X.PubMedCrossRef
63.
Zurück zum Zitat Overbergh L, Giulietti A, Valckx D, Decallonne R, Bouillon R, Mathieu C: The use of real-time reverse transcriptase PCR for the quantification of cytokine gene expression. J Biomol Tech. 2003, 14: 33-43.PubMedPubMedCentral Overbergh L, Giulietti A, Valckx D, Decallonne R, Bouillon R, Mathieu C: The use of real-time reverse transcriptase PCR for the quantification of cytokine gene expression. J Biomol Tech. 2003, 14: 33-43.PubMedPubMedCentral
Metadaten
Titel
Mesenchymal stromal cells reset the scatter factor system and cytokine network in experimental kidney transplantation
verfasst von
Marilena Gregorini
Francesca Bosio
Chiara Rocca
Valeria Corradetti
Teresa Valsania
Eleonora Francesca Pattonieri
Pasquale Esposito
Giulia Bedino
Chiara Collesi
Carmelo Libetta
Francesco Frassoni
Antonio Dal Canton
Teresa Rampino
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
BMC Immunology / Ausgabe 1/2014
Elektronische ISSN: 1471-2172
DOI
https://doi.org/10.1186/s12865-014-0044-1

Weitere Artikel der Ausgabe 1/2014

BMC Immunology 1/2014 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.