Vous êtes sur la page 1sur 14

Archives of Medical Research 48 (2017) 133e146

REVIEW ARTICLE
Mesenchymal Stem Cell-based Therapy as a New Horizon for Kidney Injuries
Amaneh Mohammadi Roushandeh,a Marzie Bahadori,b and Mehryar Habibi Roudkenarc
a
Anatomical Sciences Department, Medicine Faculty, Hamadan University of Medical Sciences, Hamadan, Iran
b
Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
c
Medical Biotechnology Research Center, Paramedicine Faculty, Guilan University of Medical Sciences Rasht, Iran
Received for publication August 29, 2016; accepted February 9, 2017 (ARCMED-D-16-00514).

Today, the prevalence of kidney diseases is increasing around the world, but there has still
been no effective medical treatment. The therapeutic choices are confined to supportive
cares and preventive strategies. Currently, mesenchymal stem cells (MSCs)-based cell
therapy was proposed for the treatment of kidney injuries. However, after the transplan-
tation of MSCs, they are exposed to masses of cytotoxic factors involving an inflamma-
tory cytokine storm, a nutritionally-poor hypoxic environment and oxidative stresses that
finally lead to minimize the efficacy of MSCs based cell therapy. Therefore, several inno-
vative strategies were developed in order to potentiate MSCs to withstand the unfavorable
microenvironments of the injured kidney tissues and improve their therapeutic potentials.
This review aims to introduce MSCs as a new modality in the treatment of renal failure.
Here, we discuss the clinical trials of MSCs-based therapy in kidney diseases as well as
the in vivo studies dealing with MSCs application in kidney injuries mainly from the pro-
liferation, differentiation, migration and survival points of view. The obstacles and chal-
lenges of this new modality in kidney injuries are also discussed. Ó 2017 IMSS.
Published by Elsevier Inc.
Key Words: Renal failure, AKI, MSCs, Cell therapy, Clinical trial.

Introduction and relieve the patients from anemia and other symptoms
of kidney failure such as bone defects and fatigue (10,11).
Acute kidney injury (AKI), an abrupt deficiency in renal
Although the aforementioned therapies increased the
filtration function, has high prevalence and is increasing
survival rate of patients suffering from renal failure, it
annually about 8%. Chronic renal failure has the prevalence
was not satisfactory, and the morbidity and mortality
about 8 to 16% and is a significant clinical problem (1e4).
rates are still high. On the other hand, the expenses of
Currently, three main strategies are applied for the treat-
hemodialysis are high and its side complications such as
ment of renal failure. Hemodialysis is the most common
hypotension, infection, gastrointestinal bleeding and psy-
method used to treat both advanced and permanent kidney
chological problems threaten the patients. Accessibility to
failures (5e7). Kidney transplantation is another strategy
a compatible kidney organ is another challenge. Of note,
in which a healthy kidney from a compatible donor is trans-
the number of patients who need kidney transplantation is
planted to a recipient patient (8,9). The last strategy for the
more than those of organ donors. Hence, some patients
treatment of renal failure is symptom-based treatment. The
have to be in the waiting list, and even may die before
patients neither undergo hemodialysis nor kidney transplan-
receiving the organ. In addition, the effects of conservative
tation, but they receive Pharmacological and dietary regi-
therapy (symptom-based therapy) need more attention and
mens. These treatment strategies alleviate the symptoms
should be carefully examined in older patients (12e14).
Therefore, establishment of a novel treatment is highly
Address reprint requests to: Mehryar Habibi Roudkenar, Medical important in order to improve the life quality of patients
Biotechnology Research Center, Paramedicine Faculty, Guilan University suffering from renal failure, decrease the morbidity and mor-
of Medical Sciences Rasht, Tehran, Islamic Republic of Iran; Phone: tality rates, address the problems of hemodialysis and organ
(þ98) 9126944566; FAX: þ98-31-42565051; E-mail: roudkenar@gums.
ac.ir.
transplantation, and decrease the cost of treatment (14).

0188-4409/$ - see front matter. Copyright Ó 2017 IMSS. Published by Elsevier Inc.
http://dx.doi.org/10.1016/j.arcmed.2017.03.007
134 Roushandeh et al./ Archives of Medical Research 48 (2017) 133e146

Mesenchymal stem cell (MSC)-based cell therapy is an revealed the therapeutic potentials of MSCs, but also inves-
innovative approach in tissue regeneration that has a poten- tigated the effects of different protocols in this field. The
tial capability in the treatment of renal failure. Currently, dose, route and source of MSCs used for cell therapy are
dozens of studies were designed in the fields of stem cells the factors under the focus of investigations. The routes of
with different sources, preconditioning modalities, and MSCs injection include intravenous, intra-arterial or intra-
routes of injection combined with a variety of pharmaco- renal. The main sources of MSCs in this regard are bone
logical agents and doses of in vitro application in preclini- marrow, umbilical and adipose tissues. There is no unique
cal and clinical studies (2,14e21). protocol regarding to the number of injected cells. In some
MSCs are type of multipotent stem cells isolated from clinical trials, the number of injected cells was reported
different tissues such as bone marrow, adipose tissue, cord 150e300 million cells twice per week over the course of
blood, amniotic membrane, human glomeruli, human 2 weeks (38e43). Of note, the number of infused cells was
saphenous vein, human periosteum and human synovial usually normalized to patient’s body weight (BW).
fluid (22e28). They are characterized with the ability of at- Table 1 summarizes the registered studies with regard to
taching to plastic in cell culture, specific surface antigen clinical trials of MSCs application in kidney diseases. As it
expression and osteogenic, chondrogenic, and adipogenic is clear, most of the studies are in the recruitment status and
differentiation (14,15,29). The cells are used in cardio- in phase I/II that shows the first steps in this field. Many other
vascular, autoimmune, neurodegenerative, renal and liver studies are undergoing and their results are not still revealed.
diseases (29,30). However, their renal application is limited The first data of a safe and feasible autologous MSCs
due to the organ complex histopathology, presence of other administration after kidney transplantation was published
diseases such as liver failure, cardiovascular diseases, dia- in 2011 (40). In a multistep study conducted by Perico
betes, inflammation and older age (29). Of note, the effec- et al., the effects of MSCs injection before and after kidney
tiveness of MSCs decreases after injection owing to poor transplantation were investigated in patients at the end stage
micro-environments. When MSCs are isolated from their of kidney disease. In the first step, patient 1 and 2 received
natural niches and cultivated in vitro, they inevitably intravenously 1.7  106 and 2  106 cells/kg autologous
confront challenging situations such as low oxygen, BM-MSCs respectively 7 d after transplantation. Both pa-
ischemia, food deprivation and free radicals. Hence, tients showed a decrease in creatinine (Cr) level after trans-
improving the quality of MSCs before injection is an inno- plantation; however, a slight creatinine increase was
vative strategy in MSCs-based therapy. In vitro cultivation detected 7e14 d after cell therapy. They reported for the first
of MSCs under the conditions of hypoxia or bioactive mol- time that MSCs infusion acted as an immune-modulator that
ecules, augmentation of MSCs with cyto-protective factors could further inhibit memory T cells extension but expand T
via genetic engineering methods and use of modified regulating cells. However, graft dysfunction occurred after
conditioned-medium or micro-vesicles could pave the MSCs transplantation. In the second step of this study, 2
way of obstacles in stem cell therapy in kidney diseases other patients received autologous MSCs at 2  106 cells/
(14,16,19,29,31e37). kg concentration intravenously 1 day before kidney trans-
The present review hopes to comprehensively discuss plantation. Control patients did not receive any MSCs. The
clinical trials of MSCs-based therapy in kidney diseases. patients who received the cell therapy did not show any
Here, we also discuss in vivo studies dealing with MSCs’ negative side effects, and their renal function was restored
survival, proliferation, differentiation and migration. In 3 d after transplantation. Biopsy of the patient number 3 af-
addition, MSCs are considered as a new horizon in the ter 1 year showed interstitial fibrosis and atrophy in tubular
treatment of renal failure. The obstacles and challenges of epithelium. Another patient presented severe lymphocyte
this new strategy will be reviewed in the following sections. infiltration in pre-vascular interestium and tubular epithe-
lium after 1 year. It is suggested that pre-transplant MSCs
rather than those of post-transplantation (after 7 d) could
Clinical Trials of MSCs in Kidney Diseases
prevent the acute graft dysfunction in patient 3. Cell rejec-
Despite the progress of MSCs-based therapy in the treat- tion was observed in patient 4 rather than in patient 3 due
ment of other diseases, its clinical trials in acute and to higher HLA haplotype mismatches (40,41).
chronic kidney injuries remained in primitive steps, and One of the important problems in solid tissue transplanta-
are associated with some problems. tion such as kidney is the application of immunosuppressors
Although, there are several preclinical studies that prom- after surgery as it seems that MSCs could decrease the dose
ise the efficiency of MSCs therapeutic application, most of of immunosuppressors. In a pilot study conducted by Peng
the studies were limited to clinical trial I/II that investigated et al, it was shown that the administration of donors’ bone
the safety and feasibility of employing these cells in human marrow-derived MSCs with tacrolimus (50% of standard
(www.clinicaltrial.gov). dose) was safe enough to protect against acute renal rejec-
Of note, there are only limited clinical data related to tion. They infused the cells in two stages. In the first stage,
MSCs-based therapy in renal disease. These studies not only the experimental group received 5  106 cells/kg directly
Mesenchymal Stem Cells and Kidney Diseases 135

Table 1. Clinical trials of MSCs application in kidney diseases (ClinicalTrails.gov)

ClinicalTrials.gov
Identifier Aim of study Enrollment Phase Status

NCT02166489 Safety of MSCs-based therapy in chronic renal failure 6 Phase I Completed


NCT02409940 To Elucidate the effect of MSCs on the T Cell repertoire of the 30 Phase I Recruiting
kidney transplant patients
NCT02266394 To determine if the MSCs infusion prior to percutaneous 42 (estimated) Phase I Recruiting
transluminal renal angioplasty with stenting (PTRA) further
changes in single kidney blood flow and restoration of kidney
function
NCT02561767 Efficacy and safety of bone marrow-derived MSCs inkidney 120 (estimated) Phase I/II Not yet recruiting
transplantation
NCT01429038 Safety and tolerability of MSCs administration after liver or kidney 40 Phase I/II Recruiting
transplantation
NCT00658073 To evaluate autologous MSCs as an alternative for antibody 165 NM Completed
induction therapy in renal transplantation.
NCT00659620 To find out whether MSCs are effective in preventing organ 20 Phase I/II Unknown
rejection and maintaining kidney function.
NCT01840540 Safety and toxicity of intra-arterial-infused autologous 6 Phase I Active, Not
adipose-derived MSCs in patients with vascular occlusive disease recruitment
of the kidney
NCT01275612 To test the feasibility and safety of systemic infusion of donor 9 Phase I Recruiting
ex-vivo expanded MSCs to repair the kidney and improve the
function in patients with solid organ cancers who develop acute
renal failure after chemotherapy with cisplatin.
NCT02195323 To provide confirmation of the safety of MSCs-based therapy in 7 Phase I Completed
chronic kidney disease (CKD).
NCT02808208 To determine the role of autologous adipose-derived MSCs in the 44 Phase I Not yet recruiting
reduction of hemodialysis arteriovenous fistula failure when
applied during the time of surgical creation
NCT02563366 To investigate whether allogeneic bone marrow-derived MSCs can 120 Phase I/II Recruiting
promote function recovery in patients with poor early graft function
after kidney transplantation in china.
NCT02492490 To determine if autologous Stromal Vascular Fraction 120 Phase I/II Recruiting
(SVF)-derived MSC infusion during and after kidney
transplantation
NCT02490020 To clarify the key role of MSCs applied via renal arterial or 260 Phase I Enrolling by
peripheral vein injection to reduce the rejection and delay graft invitation
function (DGF) after renal transplantation.
NCT02563340 To investigate the efficacy and safety of allogeneic bone 60 Phase I/II Not yet recruiting
marrow-derived MSCs (BM-MSCs) on chronic antibody-mediated
rejection (cAMR) after kidney transplantation
NCT02801890 To evaluate the effect of intravenous injection of autologous 10 Phase I/II Recruiting
adipose-derived MSCs in 10 Peritoneal dialysis patients with ultra
filtration failure (UFF).
NCT00752479 Safety and biological/mechanistic effects of the systemic 4 Phase I/II Terminated
intravenous infusion of syngeneic ex-vivo-expanded MSCs in
living-related kidney transplant recipients (one or two HLA
haplotype mismatches) under basiliximab/low-dose RATG
induction therapy and maintenance immunosuppressive drugs.
NCT01539902 Therapeutic effects of hUC-MSCs in the treatment of proliferative 25 Phase II Unknown
lupus nephritis in remission of lupus nephritis (combined partial
and complete remission) in terms of stabilization and improvement
of renal function.
NCT00659217 To evaluate whether MSCs transplantation will improve the lupus 20 Phase I/II Unknown
disease.
NCT02382874 Therapeutic effects of MSCs in the treatment of Idiopathic 5 Phase I Recruiting
Nephrotic Syndrome (INS) and glomerulus disease.

into the renal allograft artery at the time of kidney transplan- increased in the group that received MSCs. It also decreased
tation, and in the second stage, they received 2  106 cells/kg the immunosuppressive drug, increased the survival rate and
intravenously one month later. The number of T-cells improved kidney functions (42).
136 Roushandeh et al./ Archives of Medical Research 48 (2017) 133e146

Table 2. Preclinical studies using hypoxic preconditioned MSCs in kidney injury

References Animal KI model MSCs source % oxygen Outcomes

Yu et al, 2013 Rat I/R BM 200 mmol/l [ Cell engraftment, survival, proliferation, renal function,
cobalt chloride24 h b-FGF and IGF-1 over-expression
Overath et al, Mouse Cisplatin Adipose 48h, 0.5% O2 Y Serum creatinine, NGAL, IL-b and IL-6
2016 [ Cell survival
Zhang et al, Rat I/R Human 1% O2 24h Y Oxidative stress, Casp3 down-regulation
2014 adipose tissue [ Akt phosphorylation, Angiogenesis, Antioxidants capacity
Liu et al, 2012 Mouse I/R BM %3 O2 24h [ Migration, homing and mitogenic responses, Renal function,
SDF-1-CXCR4/CXCR/7Apoptosis Y

BM, bone marrow; b-FGF, Beta fibroblast growth factor; IGF-1, insulin growth factor; NGAL, Neutrophil gelatinase-associated lipocalin; IL-b, interleukin-b;
IL-6, interleukin 6; Casps3, caspase 3; Akt, Protein kinase B; SDF-1, stromal-derived factor-1; CXCR4, chemokine receptor; I/R, ischemic/reperfusion.

In another clinical trial, 76 patients underwent live-donor in the everolimus/MSC group, and 35 others took part in
renal transplantation and received donor stem cells before the everolimus/standard dose of tacrolimus group. The pa-
transplantation. All patients received donor hematopoietic tients received 2 doses of 1e2  106 cells/kg autologous
stem cells (17.4  5.5  108 nucleated cells/kg BW of recip- BM-MSCs intravenously combined with everolimus, pred-
ient with mean  SD CD34þ count of 2  0.7  104/kg BW) nisolone and tacrolimus with 7 d of time interval 7 weeks
and 55 patients received donor adipose tissue-derived MSCs after transplantation. In the second MSCs injection, tacroli-
(CD45/90þ cells of 5.8  1.5  104/kg BWand CD45/73þ mus was reduced to 50% and completely deleted after
cells of 1.1  0.92  104/kg BW) via omental vein. All 1 week. Control group was treated with routine protocol
patients were treated with cyclophosphamide, rabbit- including everolimus, prednisolone and standard dose of ta-
antithymocyte globulin and rituximab. In addition, depend- crolimus. The aim of the study was to compare some fac-
ing on the patient’s condition, other drugs such as total tors including fibrosis, acute rejection, graft loss or death,
lymphoid irradiation, bortezomib and methylprednisone renal function, proteinuria, infections, and immune moni-
were applied. Immunosuppressors such as calcineurin inhib- toring between the two groups under study (Trial registra-
itor (CNI), cyclosporin, tacrolimus, sirolimus, mycofenolate tion: NCT02057965) (18).
sodium, azathioprine and prednisone were used. All Immu-
nosuppressive drugs except for prednisone were successfully
withdrawn in all patients. On top of that, no organ rejection or Application of Modified MSCs in Preclinical Studies:
negative effects were detected after the drug withdrawal, and In vitro Cultivation and Augmentation of MSCs and
the creatinine level remained normal and constant over the their Preclinical Studies in kidney Disease
withdrawal of the immunosuppressive drugs (43). Renal fail- A plenty of animal studies reported MSCs reno-protective
ure usually accompanies with other diseases such as cardio- effects on both models of acute and chronic injuries. These
vascular disease that make the treatment even more studies varies in terms of MSCs sources, dose of adminis-
complicated. Primary results released by Gooch et al. tration, route of injection, type of kidney injury and pre-
showed that the infusion of low dose of allogenic treatment of cells under different in vitro conditions
BM-MSCs into supra-renal aorta could prevent all symptoms before their administration (3,12,16,34,37). Application of
in 5 patients with acute kidney injury induced after cardio- Modified MSCs is a novel strategy that is attractive to sci-
pulmonary bypass. It also decreased the incidence of AKI entists in preclinical and even clinical studies (21,33).
and shortened the hospitalization period (29,44). These re- The Tables 2e5 summarize some preclinical studies on
sults were consistent with the findings of Togel and Westen- the application of Modified MSCs or MSCs-derived micro-
felder’s study that reported the reno-protective effects of vesicles in the treatment of kidney disease.
intracoronary injection of allogenic BM-MSCs in patients In this section, some studies will be reviewed that are
under cardiac surgery with high risk of AKI (45). dealing with in vitro treatment of MSCs with hypoxia or
In another ongoing clinical trial in the stage of recruiting some bioactive molecules, MSCs genetic modifications
participants, 70 renal allograft recipients, between and the role of micro-vesicles in order to enhance the effi-
18e75 years old, were recruited in a Phase II, open label, ciency of MSCs-based therapy in kidney injuries.
randomized non-blinded, prospective, single center clinical
study. The mentioned study was designed to investigate the
Hypoxia
efficiency and safety of employing concentration-controlled
everolimus and MSCs (everolimus/MSC group) as well as MSCs are usually cultivated in normoxic condition with
everolimus with standard tacrolimus (everolimus/standard 20% O2 concentration in vitro. However, O2 concentrations
dose of tacrolimus group). Hence, 35 patients participated are different among tissues which are the sources of these
Mesenchymal Stem Cells and Kidney Diseases 137

Table 3. Preclinical studies on the pretreatment of MSCs with some bioactive molecules in kidney injury

Preconditioning
References Animal KI model MSCs source modality Outcome

Mias et al, 2008 Rat I/R Adipose Melatonin [ Cell survival, Proliferation, Renal function, SOD-
1 and catalase, [Angiogenesis, YFibrosis
Zhao et al, 2015 Human Cisplatin Human Melatonin [ Cell proliferation, Survival, Migration, Reno-
kidney cells Adipose MSC Protection YInhibition of apoptosis
Chen et al, 2014 Rat Sepsis Adipose Melatonin Y Oxidative stress, Fibrosis, YApoptosis,
[Antioxidants
Xinaris et al, 2013 Mouse Cisplatin BM IGF-1 [ Cell engraftment, SDF-1-CXCR4 pathway
YRenal morphology changes
Liu et al, 2013 Rat Gentamyin BM Vitamin E Y Creatinine, Urea, Tubular epithelium damage,
Apoptosis
Cai et al, 2014 Rat I/R BM Atorvastatin Y Creatinine, Urea, Apoptosis, Oxidative stress,
Inflammatory responses
[ Proliferation in tubular epithelium
Turnsek et al, 2016 Mouse Cisplatin Wharton’s jelly Antithymocyte [ Restored morphology, Antioxidants SOD-1, HO-1
Globulin and GPx, Survival of mice
Y Creatinine, Urea, Apoptosis, Oxidative stress

BM, bone marrow; I/R, ischemic/reperfusion; SOD1, superoxide dismutase 1; SDF-1, stromal derived factor 1; CXCR4, chemokine receptor; HO-1, Heme
oxygenase; GPx, glutathione peroxidase.

cells. O2 concentration in MSCs niches varies from 2e8% the oxidative stress and enhance the antioxidants of MSCs.
(37). Therefore, it seems that MSCs culture under routine Furthermore, it protects the cells against apoptosis, downre-
culture conditions can change the behavior of these cells gulates Casp3 and increase anti-apoptotic factors and Akt
by disturbing both the DNA and protein structures and phosphorylation (16). Administration of hypoxia pre-
consequently alter their functions (37,46,47). conditioned MSCs significantly increased vascular density
Hypoxic preconditioning was confirmed by several in ischemic kidney tissues compared to non-conditioned
research groups as an effective strategy to activate MSCs group (16).
and enhance their therapeutic efficacy to be employed in Hypoxic preconditioning of MSCs resulted in less
ischemic diseases. Injection of human adipose tissue- tubular cell necrosis, loss of brush border and tubular dila-
derived MSCs pretreated with 1% hypoxia could decrease tion following MSCs-based therapy (16). Creatinine and

Table 4. Shows the application of gene-engineering modified-MSCs in preclinical studies in Kidney injury

MSCs Gene- modified


Reference Animal KI model source MSCs Outcome

Mohammadzadeh-vardin Rat Cisplatin BM Nrf2-MSC [ Renal function, Morphological features of kidney,


et al, 2015 Growth factors and antioxidants YCreatinine, Urea
Zhaleh et al, 2016 Rat Glycerol BM Nrf2-MSC YCratinine, Urea, Kim-1 and Cystatin C
[ Renal function, AQP1 and CK-18
Halabian et al, 2015 Rat Cisplatin induced BM Lcn2-MSC [ Reno-protection property, Growth factors,
injury in renal cells antioxidants, Cell proliferation
Y Apoptosis
Hagiwara et al, 2008 Rat I/R BM TK-MSC [ Renal function, Renal tubular epithelium repair
Y Apoptosis, Nitric oxide, Inflammation
Liu et al, 2013 Rat I/R BM CXCR4-MSC [ SDF-1, Cell engraftment, Renal function, Activation
of PI3K/AKT and MAPK pathways, TGF-B1
Liu et al, 2016 Rat Gentamycin hUCMSC IGF-1-MSC [ Cell migration, Renal function, Activation of
antioxidants, anti-inflammatory and anti-apoptotic
signal pathways
Yuan et al, 2011 Mouse Cisplatin BM VEGF-MSC [ Proliferation, Angiogenesis
Y Apoptosis
Wang et al, 2016 Rat Mercuric chloride BM CXCR4-MSC [ CXCR4, homing of injected cell

BM, bone marrow; Nrf2, Nuclear factor erythroid-2 related factor 2; Kim-1, kidneyinjury molecule-1; AQP1, aquaporin 1; CK18, cytokeratin-18; Lcn2,
lipocalin 2; SDF-1, stromal derived factor 1; P13K/AKT, Phosphatidylinositol-4,5-bisphosphate 3-kinase/Protein kinase B; MAPK, Mitogen-activated pro-
tein kinase; TGF-B1, transforming growth facor-B1; CXCR4, chemokine receptor; VEGF, vascular endothelial growth factor; IGF-1, insulin growth fac-
tor; TK, tissue kallikrein; Lcn2, lipocalin 2.
138 Roushandeh et al./ Archives of Medical Research 48 (2017) 133e146

Table 5. Illustrates the evidence of MSCs-derived micro-vesicles application in Kidney regeneration in preclinical studies

References Animals KI model type MSCs SI or MI Pretreatment Outcome

Zou et al, 2014 Rat IRI (Unilateral) Human (WJ) SI No Y Apoptosis, Fibrosis, Inflammation, CX3CL1
[ Proliferation, Renal function
Wang et al, 2015 Mouse UUO mouse (BM) SI erythropoietin Y Changed miRNA profile Fibrosis, Apoptosis
Bruno et al, 2012 Mouse Cisplatin (lethal dose) Mouse SI and M No [ Renal morphology, Tubular cell proliferation,
(6 times) [ Mouse mortality, BUN and Creatinine
Ju et al, 2015 Rat IRI (Unilateral) hUC-MSC SI No [ Restored renal structure and function, Tubular
cell dedifferentiation and growth, Rat HGF
expression
Y Apoptosis
Reis LA et al, 2012 Rat Gentamycin BM SI RNAase and Y Cr, Urea and FENA, necrosis and Apoptosis,
Trypsin RNAase application on reno-protective effects
[ Cell proliferation
Zhou et al, 2013 Rat Cisplatin hUCMSCs SI No [ Renal function, Morphology, Proliferation
Y Oxidative stress, Renal tubular apoptosis
Bruno et al, 2009 Mouse Glycerol BM SI RNAase [ Renal function, Restored renal morphology
Y Creatinine, Urea, RNAase application on
reno-protective effects
Gattie et al, 2011 Rat I/R BM SI No Y Creatinine, Urea, Apoptosis
[ Proliferation, Protection kidney against CKD
Zhang et al, 2016 Rat IRI hWJMSC SI No [ Nrf2/ARE pathway, SOD-1 and HO-1,
restored renal morphology
Y ROS, Creatinine, Urea
Kilpinen et al, 2013 Rat Hypoxia induced hUCBMSC SI IFN-y Diverse Rab proteins, complement and MHCI,
kidney injury no reno-protection

IRI, ischemic/reperfusion injury; SI, single injection; MI, multiple injection; CX3CL, chemokine ligand 1; BUN, Blood urea nitrogen; HGF, hepatocyte
growth factor; Cr, creatinine; FENA, Fractional Excretion of Sodium; CKD, chronic kidney disease; Nrf2/ARE, Nuclear factor erythroid-2 related factor
2/antioxidant respond element; SOD-1, super oxide dismutase; HO-1, heme oxygenase; ROS, reactive oxygen species; MHCI, Major histocompatibility com-
plex; UUO, unilateral ureteric obstruction; WJ, wharton jelley; hUCB, human unbilical cord blood.

BUN decreased in the serum of animals that received migration. Hypoxia mimetic preconditioned MSCs (HMP-
MSCs specially those preconditioned with hypoxia. It is MSC) showed better engraftment in kidney ischemic model
suggested that hypoxia could enhance the efficiency of compared to non-preconditioned MSCs (NP-MSC). Super-
MSCs through anti-apoptotic effects, ROS scavenging paramagnetic iron oxide (SPIO) and fluorescent-labeled
and angiogenesis (16). MSCs were followed by magnetic resonance imaging and
Recently conditioned medium instead of MSCs was immunohistochemistry respectively. Imaging clearly
used in cell therapy. Hypoxia preconditioning of mouse ad- showed that HMP-MSCs exhibited greater migration and
ipose tissue-derived MSCs was performed with 0.5% O2. a longer retention time in the ischemic kidney than
Conditioned medium of hypoxia-pretreated adipose NP-MSCs. Histologically, 72 h after cell infusion, no cell
tissue-derived MSCs from peri-renal fat developed regener- was detected in kidney of NP-MSC but still remained in
ation in mouse models of cisplatin-induced AKI (cisAKI). glumerulli and tubulointerstum in HMP-MSC, 1 week after
The MSCs significantly decreased serum creatinine and cell therapy. Overall, it was confirmed that hypoxia could
neutrophil gelatinase-associated lipocalin values. Moreover, increase survival and proliferation and developed kidney
two inflammatory cytokine, i.e. IL-1 b and IL-6 were functions. Creatinine and urea levels decreased in both
declined in serum indicating its immunomodulatory effects. groups of cell therapy; however, HMP-MSCs had better ef-
Cell survival was more in hypoxia preconditioned group fects on renal recovery. Hypoxia exerts its reno-protective
over 3 d after injection. Kidney injury markers such as renal effects through paracrine secretion such as b-FGF and
KIM-1, Klotho and HMGB-1 were changed after cisplatin IGF-1 (33).
administration. Downregulation of HMGB-1 could sup- It is reported that hypoxia preconditioning may have a
press the immune response in injured tissues (48). role in chemotaxis and homing of MSCs. The mechanism
Cobalt choloride was also used to induce hypoxia. Cul- is mediated by SDF-1-CXCR4/CXCR7 axis. The interac-
ture of rat bone marrow-derived MSCs (BM-MSC) with tion of SDF-1 and its receptor CXCR4 expressed on the
200 mmol/L cobalt significantly increased the expression MSC surface plays an important role in the migration of
of HIF-1 a and CXCR4 and leaded to the increased migra- transplanted cells. MSCs homing decreased when the
tion of MSCs in vitro (49,50). Blocking of these proteins CXCR4 or CXCR7 inhibitors were used. HP-MSCs (3%)
such as HIF-1 with their targeting siRNA declined cell migrated to the ischemic kidney more efficiently than
Mesenchymal Stem Cells and Kidney Diseases 139

NP-MSCs resulting in remarkably improved renal func- injury. In addition, after IGF-1 pretreatment, the cells
tions, accelerated mitogenic responses, and reduced cell mostly settled in peritubular and rarely in renal tubular
death. Histologically, cell therapy groups had less nephro- epithelium. The injected cells were observed for a short time
toxicity, more proliferation and less apoptosis compared in glomerulus of injured kidney after IGF-1 pretreatment.
to the control group (50). Adversely, pretreatment with other factors such as glial cell
line-derived neurotrophic factor (GDNF) and tumor necro-
sis factor 1 (TNF-a) did not enhance the engraftment of
MSCs Preconditioning with Bioactive Molecules
MSCs in animal model of AKI. The mice receiving IGF-
Cytokines, growth factors and other bioactive molecules 1-pretreated MSCs showed more decline in cast formation,
have a critical role in the survival, migration and differen- tubular epithelium necrosis and less changes in ultrastruc-
tiation of MSCs. ture of tubular tissues. It is suggested that the activation of
One of the main problems in cell therapy is the early SDF-1-CXCR4 is the mechanism by which IGF-1 could
death of injected cells because of oxidative stress (51). It induce its potency and enhance renal function (38).
is confirmed that melatonin hormone is able to remove Gentamysin is a toxic antimicrobial drug that its applica-
oxidative damage and stimulates cytokine secretion in tion increased recently because of microbial resistance (60).
injured tissues (52,53). It acts as antioxidant, anti- It causes nephrotoxicity, and results in loss of kidney func-
apoptotic and anti-inflammatory agent (54e56). Pretreat- tion (61). Application of MSCs with vitamin E could pro-
ment of rat MSCs with melatonin enhanced cell survival tect its toxic effects. Vitamin E is an oxygen free radical
and proliferation in AKI mostly through the cellular recep- scavenger and lipid-soluble antioxidant (62). The rats
tors MT1 and MT2. Histological analysis showed no exces- receiving MSCs and vitamin E presented the lowest serum
sive production of extracellular matrix, collagen, calcium creatinine but no change in urea nitrogen decline. Vitamin
deposition or fibrosis in kidney tissue after 2 months. In E protected the kidney against gentamycin toxicity through
addition, Melatonin-treated cells increased the expression decreasing tubular epithelium necrosis and dilation, brush
of two important antioxidants, i.e. superoxide dismutase 1 border loss and suppressing immune responses. Electron
(SOD1) and catalase. It also over-expressed beta fibroblast microscopy showed that gentamycin induced apoptosis in
growth factor (b-FGF) and hepatocyte growth factor renal tubular epithelial cells (RTECs). The combined strat-
(HGF), the two factors in angiogenesis process. Further- egy of cell therapy and vitamin E improved and restored the
more, it improved renal function by decreasing both Cr morphology of injured tissue according to ultrastructure
and BUN levels in rat ischemic/reperfusion (I/R) renal fail- findings through increasing the number of organelles and
ure. It seems that melatonin is an efficient choice in MSCs- decreasing the cytoplasmic vacuoles. Nevertheless, the
based therapy because it is currently used as a complement changes were not significant when MSCs was used alone.
with no side effects in human (57). Conditioned medium Downregulation of some apoptotic genes such as caspase
collected from pretreated human adipose tissue-derived 3 and 9 is the mechanism by which vitamin E could exert
MSCs culture with melatonin for 3 hours showed reno- its potency in combination with MSCs. It is suggested that
protective effects on cisplatin-induced injury in human kid- the application of MSCs along with vitamin E could have
ney cells (58). Pretreated conditioned medium not only better therapeutic effects on AKI rather than MSCs and
enhanced the rate of MSCs proliferation, survival and vitamin E, separately (63).
migration, but also inhibited apoptosis through up- Pretreatment of MSCs with Atorvastatin could enhance
regulation of phosphor-p44/42 MAPK (P-Erk1/2), Bcl-2, renal functions by decreasing apoptosis and ROS. It also in-
SOD-1 and heme oxygenase 1 (HO-1) (58). hibited the immune response (64).
Melatonin also has reno-protective effects in sepsis- Antithymocyte globulin (ATG) is an immunosuppressor
induced AKI and decreases kidney injury through that effectively enhances the renal function when adminis-
increasing antioxidants such as glutathione reductase tered on hWJMSC before transplantation into mouse models
(GRþ), glutathione peroxidase (GPxþ), HO-1 and quinine of cisplatin-induced AKI. ATG increased antioxidants such
acceptor oxidoreductase 1 (NQO-1þ), decreasing oxidative as SOD-1, HO-1 and GPx. More interestingly, it decreased
stress NADPH oxidase -1 (NOX-1) and NOX-2 and the mortality rate of mice injured with cisplatin (65).
declining in fibrosis factors Smad3 and TGF-b. It also in-
hibits apoptosis by changing gene expression of cleaved
Application of Genetically-modified MSCs in Kidney
Caspase 3, Poly ADP (Adenosine Diphosphate)-Ribose
Diseases
Polymerase (PARP) and mitochondrial Bax (59).
Homing of MSCs in injured tissues is very critical in cell MSCs could be engrafted in injured tissues and are the best
therapy. Most of the injected cells are attracted to lung after vehicle for gene transfer in cell therapy. Genetic manipula-
transplantation. Pretreatment of mouse BM-MSCs with in- tion of MSCs could induce the secretion of some proteins
sulin growth factor 1 (IGF-1) significantly increased the involved in cell migration, proliferation and survival.
capability of cell engraftment in cisplatin-induced kidney Different genes could be introduced into MSCs through
140 Roushandeh et al./ Archives of Medical Research 48 (2017) 133e146

viral or non-viral vectors. The later technique is safer, but and urea level and tubular epithelium repair. Six hours after
its efficiency is low. In this section, some genetic modifica- I/R, TK-MSC implantation significantly reduced renal cell
tions will be reviewed that resulted in the development of apoptosis in association with decreased inducible nitric ox-
MSCs therapeutic properties. ide synthase expression and nitric oxide levels. 48h after
The nuclear factor erythroid-2 related factor 2 (Nrf2) is cell therapy, TK-MSCs inhibited the infiltration of the in-
an important transcription factor that protect the cells flammatory cells and decreased myeloperoxidase activity,
against stresses. Transplantation of Nrf2-engineered MSCs superoxide formation and p38 mitogen-activated protein ki-
(Nrf2-MSCs) decreased cisplatin-induced injury in rat nase phosphorylation. Furthermore, it reduced the ex-
models of AKI. It also decreased the cytotoxic effects of pression of tumor necrosis factor-a, monocyte
cisplatin on MSCs in vitro. Nrf2-MSCs could ameliorate chemoattractant protein-1 and intercellular adhesion
the creatinine and urea levels and enhance renal function molecule-1. It is suggested that kallikerin could present
in AKI. Nrf2 overexpression improved the morphological its therapeutic effects through the control of apoptosis
features of renal tubule damaged by cisplatin. Expression and inflammation (81).
of growth factors and antioxidants leaded to better therapeu- MSCs engraftment to target injured tissues is very crit-
tic efficiency of Nrf2-MSCs (66); however, its potency for ical in the outcome of cell therapy. Stromal cell-derived
tumorigenicity should be considered. Transplantation of factor-1 (SDF-1) and its cellular receptor, CXCR4, have a
Nrf2-MSCs into glycerol-induced AKI rats enhanced renal pivotal role in stem cells migration toward injured tissues,
function by decreasing the creatinine and urea levels 14 d af- but the expression of CXCR4 on the surface of BMSCs is
ter cell therapy. More interestingly, the repaired genes such not high. Therefore, overexpression of this receptor on
as Aquapoin 1 (AQP1) and Cytokeratin 18 (CK-18) were the surface of MSCs is a new strategy to increase the effi-
overexpressed in the group that received MSCs or saline ciency of cell therapy. The CXCR4- BMSCs showed a
alone. Moreover, the renal damage markers, i.e. kidney remarkable expression of SDF- 1 in AKI. CXCR4- BMSCs
injury molecule 1 (Kim-1) and Cystatin C decreased signif- transplantation significantly increased the number of en-
icantly after the application of Nrf2-MSCs (3). Actually, the grafted BMSCs in the ischemic renal tissue resulting in
therapeutic effects of Nrf2 could be exerted through the the improvement of renal functions (82). Activation of
expression of genes involved in detoxification, anti- PI3K/AKT and MAPK in BMSCs by SDF-1/CXCR4 axis
oxidation and immunomodulation (3,67,68). could explain the mechanism involved in this process
Overexpression of lipocalin2 in MSCs enhanced their ef- (82,83).
ficiency in in vitro studies (69). Lcn2 belongs to a family of Overexpression of TGF-b1 in genetically-engineered
soluble proteins that is expressed in various normal and path- MSCs increased the homing of the cells in ischemic models
ologic conditions. It increased to 1000 fold after renal tu- of kidney disease and improved the renal function by
bules injury (70). It is reported that lipocaline2 is normally decreasing both the creatinine and urea levels. This study
expressed under some conditions such as stress and inflam- confirmed the role of TGF-b1 in controlling the SDF-1-
mation. It plays a pivotal role in physiological processes such CXCR4 axis in cell engraftment (84).
as cell proliferation, senescence and apoptosis (69,71e77). IGF-1 secreted by MSCs could enhance their therapeu-
It is shown that the co-culture of bone marrow-derived tic potential through cell proliferation and anti-apoptotic
MSCs overexpressing lipocalin2 with HK-2 and actions. After transplantation of modified human umbilical
HEK293 cells had reno-protective potential against cord (UC)-derived MSCs-IGF-1 to gentamycin-induced
cisplatin-induced injury. Lcn2-Overexpressed MSCs AKI, more cell migration was observed in kidney tissue
increased the proliferation rate and prevented cisplatin- compared to MSC-vector or MSCs. It also increased
induced apoptosis when co-cultured with renal cell lines. renal function through recovering the serum and urine
It increased the expression of growth factors such as biochemical indices. Less histological damage to glomer-
HGF, IGF, TGF-b, and FGF as well as antioxidants such ulus, tubular epithelium and collecting tubules were
as SOD1, HO-1 and MT-1 in the kidney cells. Therefore, observed after the application of human UC-MSCs-IGF-
it is suggested that the protective effects of this protein 1. It also up-regulates some signaling pathway genes
could be presented through the secretion of growth factors involved in the properties of anti-oxidation, anti-inflamma-
and production of antioxidants (78). tory, anti-apoptotic and cell migratory capacities of
Tissue kallikrein (TK) was first discovered in human UC-MSCs-IGF-1 (36) (Figure 1).
urine as a hypotensive substance (79). It is remarkably It seems that vascular endothelial growth factor (VEGF)
reduced in mild kidney disease and severe renal failure could have a critical role in reno-protection (85). Human
(80). It is reported that kallikrein has protective effects on MSCs modified with VEGF could enhance the proliferation
tissues damaged by oxidative stresses. Kallikrein-MSCs of nude mouse kidney epithelial cells injured by cisplatin. On
implantation increased protection against AKI through the other hand, it inhibited apoptosis in renal tissue. Interest-
decreasing apoptosis and inflammation. Tissue Kallikrein- ingly, no difference was detected between the migration of
MSCs improved renal function by decreasing creatinine MSCs and MSC-VEGF. Angiogenesis was induced after
Mesenchymal Stem Cells and Kidney Diseases 141

the transplantation of MSC-VEGF in vivo, and the peritubu- therefore, multiple administrations of allogeneic MVs
lar capillary density increased in this group (86). may not activate the immune response (92).
Table 4 shows the application of gene engineering MVs exert their effects directly on cells through transfer-
modified-MSCs in preclinical studies in Kidney injury. ring cell surface receptors or epigenic reprogramming.
Considering these characters and benefits, MVs would be
very attractive in the treatment of renal failure
MSCs and their Micro-vesicle Application in Kidney
(88,90,93,94).
Diseases
Table 5 summarizes some preclinical studies on the
Membranous small vesicles or micro-vesicles (MVs) are application of micro-vesicles in the treatment of kidney
released by MSCs (87e90). They are extracellular vesicles injuries.
including exosomes and micro-vesicles. Exosomes derived Erythropoietin (EPO) is a glycoprotein hormone that in-
from endocytic vesicles have a diameter of 30e100 nm. Shed- volves in the formation and differentiation of erythroid pre-
ding vesicles, also named ectosomes or membrane particles cursor cells in the bone marrow (95). It is expressed in some
tending to be 100 nme1 mm in diameter. They bud directly tissues such as renal tubular cells which also express EpoR
from the plasma membrane. Most extracellular vesicles are and respond to EPO treatment (96). MVs derived from
heterogeneous and consist of exosomes and shedding vesi- MSCs pretreated with EPO showed significantly restored
cles, collectively defined as micro-vesicles (MVs) (90e92). morphology features of renal tissue injured by Unilateral
MVs play a critical role in cell-cell communications. Ureteral Obstruction (UUO) model on 7 and 14 d. It
MVs interact with target cells through surface-expressed li- enhanced kidney function by decreasing urine and creati-
gands, and transfer surface receptors that deliver proteins, nine levels one day after therapy. Extracellular matrix depo-
messenger RNA (mRNA), microRNA (miRNA), and bioac- sition and fibrosis declined in mice that received MVs-EPO.
tive lipids. They contain high amounts of phosphatidylser- It is reported that the expression of a-smooth muscle actin
ine- a protein associated with lipid rafts, and are full of (a-SMA) as a myofibroblast marker decreased while E-cad-
cholesterol, sphingomyelin and ceramide (34,92). herin as an epithelial cell marker increased after the appli-
MVs deliver complex and bioactive molecules derived cation of MVs-EPO (32).
from MSCs to injured cells leading to tissue regeneration. microRNA profiling array results found remarkable
They do not represent histocompatibility antigens; changes in the profile of MVs-MSC and MVs-EPO.

Figure 1. Showes the genes that changed after overexpression of some genes in MSCs. GEM-MSCs, genetic engineering modified-mesenchymal stem cell;
Nrf2, Nuclear factor erythroid-2 related factor 2; Lcn2, lipocalin 2; TK, tissue kallikrein; CXCR4, chemokine receptor; IGF-1, insulin growth factor1; TGF-
B1, transforming growth factor B1. (A color figure can be found in the online version of this article.)
142 Roushandeh et al./ Archives of Medical Research 48 (2017) 133e146

EPO-MVs changed the expression of 212 miRNAs It could improve the proliferation of renal epithelial cells
including miR-299, miR-499, miR-302, and miRNA-200 and inhibit apoptosis. It also recovered the morphology of
from which 70.28 % were overexpressed (32). proximal tubules and decreased cast formation. Human um-
A single administration of MVs immediately after bilical cord MSCs-derived exosomes (hUCMSC-exosome)
ischemic AKI in animal models ameliorated renal injury decreased two oxidants, i.e. 8-hydroxy-20 -deoxyguanosine
at both acute and chronic stages (34). The anti- (8-OHdG) and malondialdehyde (MDA) and increased the
inflammatory effect of MVs seems to be exerted through level of a critical antioxidant, i.e. Glutathione (GSH).
the suppression of CX3CL1. This establishes a substantial hUCMSC-exosome inhibited p38MAPK pathway and inac-
foundation for future research and treatment. Application tivated apoptosis process (97).
of human wharton’s jelley MSCs-MVs (hWJMSC-MVs) Therapeutic effect of MSCs is suggested to be mediated
in AKI rats decreased renal cell apoptosis and induced cell by paracrine interactions with immune cells. It is found that
proliferation. It also decreased immune responses in the human umbilical cord blood MSCs (hUCBMSC)-derived
early stages through decreasing the number of MVs pretreated with interferon-gamma (MVs-IFN-g) had
CD68þ macrophages in the kidney and suppressing no therapeutic effects on I/R-induced kidney injury. It
CX3CL1. Additionally, it enhanced renal tissue restoration was not able to decrease creatinine and urea levels, and
and inhibited fibrosis leading to the improvement of renal no improvement in the recovery of kidney tissue was
function. It is suggested that some mirRNAs such as mir- observed while hUCBMSC MVs had reno-protective ef-
15a, mir15b, mir-16, mir195, mir424 and mir497 are the fects on ischemic injury in kidney. Moreover, protein con-
targets for CX3CL1. These microRNAs might control tent in MVs-IFN-y was significantly higher than that in
CX3CL1, but their functions are not that clear yet (34). non-pretreated MVs (448 proteins versus 220). They pre-
It is suggested that multiple injections of MVs (miMVs) sented diverse Rab proteins and complements and ex-
are more efficient than single that of MVs (siMVs). In a pressed MHCI. It was concluded that the same cell could
study, mouse BM-MSCs-derived miMVs enhanced the sur- produce different types of MVs according to the external
vival of SCID mouse as a model of AKI. It also restored signals (98).
renal tissue and increased renal function for a long time It seems that antioxidant properties of MVs-derived
(21 d). It seems that the expression of anti-apoptotic gene MSCs could enhance renal recovery. It was reported that
after MVs application is the main mechanism of this phe- the application of conditioned medium of human Wharton’s
nomenon (89). Jelly mesenchymal stromal cells could activate Nrf2/anti-
Dedifferentiation of renal tubular cell epithelium is oxidant response element (ARE) in ischemic-induced kid-
important in the repair process after kidney injury. It was ney injury. It increased SOD and HO-1 and decreased
revealed that hepatocyte growth factor has a critical role ROS along with Nrf-2 and ARE. MVs efficiently restored
in this regard. Human umbilical cord (HU) MSCs could ex- renal morphology and function through the decline of creat-
press this growth factor. Furthermore, MVs-HUC MSCs inine, urea and NGAL (99).
could deliver their RNA into the injured tubular cells and
enhance the gene expression of hepatocyte growth factor re-
The Challenges of MSCs Application in Kidney Repair
sulting in cell regeneration. They exerted therapeutic effects
on renal cells through anti-apoptotic and anti-fibrotic activ- Although the application of MSCs is very promising in kid-
ities as well as dedifferentiation. Human HGF mRNA car- ney repair, its therapeutic potentials confront some limita-
ried by micro-vesicles can reside in renal tubular cells and tions that need more attention in the future.
were able to induce higher production of this protein in Unfortunately, there is no updated and adequate good
injured tubular epithelium. Interestingly, MVs treatment manufacturing practices (GMP) guideline in MSCs-based
with RNAase eliminated all its reno-protective effects. Vi- therapy in kidney. In addition, other issues including
mentin and proliferating cell nuclear antigen (PCNA) pro- ethical, legal, technical and safety concerns must be care-
teins increased in tubular epithelium of injured kidney fully considered (91).
after MVs treatment. Vimentin and PCNA up-regulations Although there is no report on the detrimental effects of
leaded to an increase in dedifferentiation and cell growth MSCs-based therapy in clinical trials and it is considered
respectively. MVs activates Erk1/2 pathway to induce safe and feasible, more uncontrolled factors should be
dedifferentiation and growth. An Erk1/2 inhibitor could considered in cell therapy of kidney diseases (27).
block the production of HGF and disturb renal repair (35). Mal-differentiation of stem cells is another challenge
Exosomes are micro-vesicles that are released from that not only impairs the renal tissue recovery, but it also
various cells into the extracellular space when intracellular exacerbates the renal injury by replacing ectopic cells in
MVBs fuse with the plasma membrane of the cells (87). It kidney. Differentiation of injected MSCs to adipocytes
is shown that exosomes derived from human umbilical cord could lead to chronic renal injury (92).
MSCs has reno-protective effects on cisplatin-induced renal Route of MSCs injection is another issue that should be
injury through ameliorating the creatinine and urea levels. considered in renal treatment. Intravascular administration
Mesenchymal Stem Cells and Kidney Diseases 143

of MSCs could lead to vascular occlusion (100). Intrave- MSCs-based Therapy as a New Horizon for Treatment of
nous injection attracts the cells to lung tissues and de- Kidney Diseases
creases its accurate homing in renal tissue (100).
MSCs-based cell therapy is a promising therapeutic
Although the clinical trials of MSCs application are
approach in heart, neurodegenerative and autoimmune dis-
promising, their administration is limited in some dis-
eases. However, kidney diseases have complicated histo-
eases. Their employment to treat various diseases such
pathological configurations that make its routine
as kidney should be with some considerations. More re-
applications even more difficult. Thus, the final outcomes
searches are needed to find out the mechanisms and bio-
are usually not favorable.
logical properties of MSCs to pave the way of their
Application of bone marrow-derived MSCs is routine,
application. MSCs are heterogeneous in population and
and its safety and feasibility are confirmed in plenty of
may result in different outcomes. MSCs ex vivo culture,
their culture protocol and environment, source of the cells, studies. However, using new sources of stem cells with
more therapeutic capability is encouraging. Fortunately,
number of cellular passages, cell number injection are fac-
the presence of a multipotent MSCs population residing
tors of vital importance that should be all standardized and
in human glomeruli has been reported (24). During injury,
optimized (21,101). To obtain better outcomes, more ran-
these cells can differentiate into myofibroblasts which
domized and controlled multicenter clinical trials are
form the cellular basis of tissue repair. It seems that
necessary (101).
tissue-specific stem cells retained superiority in lineage-
Due to the low survival and engraftment of MSCs after
specific differentiation along with their resident tissue
transplantation, a high dose ranging from 150 million to
300 million cells is usually used in clinical trials adminis- origin and natural functions.
tered twice per week over the course of 2 weeks (89). This Recently, MSCs derived from fetal kidney (fKSCs) was
might result in transfusion reaction such as allergic reac- reported (103,104). These cells express mesenchymal and
tion, fever, hypotension and infection. Infection is very crit- renal progenitor markers. They have the ability to differ-
ical; therefore, donor members must be thoroughly entiate into renal epithelial cells that are commonly
examined before bone marrow aspiration (89). injured during kidney failure. Application of fKSCs results
Tumorigenecity is another concern after cell therapy. in rapid restoration of renal function and histology. It up-
Although there has been no malignancy report in clinical regulates the angiogenic proteins such as hypoxia induc-
trials, it should be noticed that most trials have a short ible factor 1 (HIF-1a), VEGF and endothelial nitric oxide
follow up. Hence, long term follow up is necessary to synthase (eNOS) that further induce angiogenesis. There-
assure in this regard (101). fore, studies on fKSCs-mediated renal angiogenesis and
Micro-vesicles and exosomes have valuable therapeutic regeneration may lead to the development of novel phar-
effects on renal tissue recovery after injury; however, their macological therapies in kidney diseases (104).
application should be carefully monitored. It was reported MSCs pretreatment with several factors such as
that miRNA transported by exosomes involve in cancer growth factors and vitamins is a novel strategy to
occurrence (102). They might also increase the risk of neuro- enhance the MSCs efficiency in kidney regeneration. Pre-
degenerative disorders such as prion, tauopathies, Alz- treatment with vitamins, such as vitamin E could be ex-
heimer’s and Parkinson’s diseases. We are not sure whether ploited as a new therapeutic approach in regenerative
all of the MVs cargos are useful in cell therapy or not. There- medicine (63).
fore, finding their complexity, components and interactions Application of natural polymers such as biological scaf-
with other secreted factors should be studied (90e92). folds that carry MSCs for tissue repair is another new strat-
Application of MVs as a non-cellular therapy should be egy to increase their efficiency. These scaffolds include
improved. Therefore, several factors such as the type of alginate, collagen, fibrin, albumin, hyaluronan, platelet-
MVs’ effector molecules, cell source, mechanisms of tar- rich plasma and gelatin. They are constructs with a fully
geting to specific tissues and the genetic manipulation of vascularized implant that can adapt to the hypoxic environ-
the cells they are derived from should be considered to ment in vivo (105,106).
enrich the therapeutic potentials of MVs before clinical Overall MSCs-based cell therapy would be considered
application in human (90). as a new horizon for the treatment of kidney disease. How-
Overall, some challenges such as large-scale production ever, it must be noted that it is still at the preliminary stages
of MVs from cultured stem cells as well as the criteria for of development. Further studies including more clinical tri-
the potency of different MVs preparations should be ad- als are required in order to achieve the routine application
dressed before any clinical use. Although the safety of of MSCs as a new modality for the treatment of kidney in-
MVs was confirmed in preclinical experiments, further juries. We still have to strengthen our knowledge and be
studies are recommended in this regard. patient.
144 Roushandeh et al./ Archives of Medical Research 48 (2017) 133e146

Acknowledgments 20. Morigi M, De Coppi P. Cell therapy for kidney injury: different op-
This study was supported by the Iran National Science Foundation tions and mechanisms-mesenchymal and amniotic fluid stem cells.
Nephron Exp Nephrol 2014;126:59e63.
(INSF) (grand no: 91004674).
21. Liu S, Zhou J, Zhang X, et al. Strategies to Optimize Adult Stem Cell
Conflict of Interest: The authors declare no conflict of interest.
Therapy for Tissue Regeneration. Int J Mol Sci 2016;17:982.
22. De Almeida DC, Donizetti-Oliveira C, Barbosa-Costa P, et al. In
search of mechanisms associated with mesenchymal stem cell-
References based therapies for acute kidney injury. Clin Biochem Rev 2013;
1. Safari S, Yousefifard M, Hashemi B, et al. The Role of Scoring Sys- 34:131e134.
tems and Urine Dipstick in Prediction of Rhabdomyolysis-induced 23. Barton M, Sorokin A. Endothelin and the glomerulus in chronic kid-
Acute Kidney Injury: a Systematic Review. Iran J Kidney Dis ney disease. Semin Nephrol 2015;35:156e167.
2016;10:101e106. 24. Bruno S, Bussolati B, Grange C, et al. Isolation and characterization
2. Eirin A, Lerman LO. Mesenchymal stem cell treatment for chronic of resident mesenchymal stem cells in human glomeruli. Stem Cells
renal failure. Stem Cell Res Ther 2014;5:1e8. Dev 2009;18:867e880.
3. Zhaleh F, Amiri F, Mohammadzadeh-Vardin M, et al. Nuclear factor 25. Bruno S, Camussi G. Isolation and characterization of resident
erythroid-2 related factor 2 overexpressed mesenchymal stem cells mesenchymal stem cells in human glomeruli. Methods Mol Biol
transplantation, improves renal function, decreases injuries markers 2012;879:367e380.
and increases repair markers in glycerol-induced Acute kidney injury 26. Covas DT, Piccinato CE, Orellana MD, et al. Mesenchymal stem
rats. Iran J Basic Med Sci 2016;19:323e329. cells can be obtained from the human saphena vein. Exp Cell Res
4. Wang S, Qu X, Zhao RC. Clinical applications of mesenchymal stem 2005;309:340e344.
cells. J Hematol Oncol 2012;5:19. 27. De Bari C, Dell’Accio F, Vanlauwe J, et al. Mesenchymal multipo-
5. Sherman R, Swartz R, Thomas C. Treatment methods for kidney fail- tency of adult human periosteal cells demonstrated by single-cell
ure: hemodialysis 2009. lineage analysis. Arthritis Rheum 2006;54:1209e1221.
6. Ibrahim A, Ahmed MM, Kedir S, et al. Clinical profile and outcome 28. Jones EA, English A, Henshaw K, et al. Enumeration and phenotypic
of patients with acute kidney injury requiring dialysis—an experi- characterization of synovial fluid multipotential mesenchymal pro-
ence from a haemodialysis unit in a developing country. BMC Neph- genitor cells in inflammatory and degenerative arthritis. Arthritis
rol 2016;17:1. Rheum 2004;50:817e827.
7. Chen JY, Wan EY, Chan KH, et al. Evaluation of the quality of care 29. Squillaro T, Peluso G, Galderisi U. Clinical trials with mesenchymal
of a haemodialysis public-private partnership programme for patients stem cells: an update. Cell Transplant 2016;25:829e848.
with end-stage renal disease. BMC Nephrol 2016;17:1. 30. Amiri F, Molaei S, Bahadori M, et al. Autophagy-Modulated Human
8. Paloyo S, Sageshima J, Gaynor JJ, et al. Negative Impact of Pro- Bone Marrow-Derived Mesenchymal Stem Cells Accelerate Liver
longed Cold Storage Time before Machine Perfusion Preservation Restoration in Mouse Models of Acute Liver Failure. Iran Biomed
in Donation after Circulatory Death Kidney Transplantation. Transpl J 2016;20:135e144.
Int 2016;29:1117e1125. 31. Liu N, Tian J, Cheng J, et al. Effect of erythropoietin on the migra-
9. Spasovski G. Generic DrugseDecreasing Costs and Room for tion of bone marrow-derived mesenchymal stem cells to the acute
Increased Number of Kidney Transplantations. Pril (Makedon Akad kidney injury microenvironment. Exp Cell Res 2013;319:
Nauk Umet Odd Med Nauki) 2015;36:133e138. 2019e2027.
10. Verberne WR, Geers AT, Jellema WT, et al. Comparative survival 32. Wang Y, Lu X, He J, et al. Influence of erythropoietin on micro-
among older adults with advanced kidney disease managed conserva- vesicles derived from mesenchymal stem cells protecting renal
tively versus with dialysis. Clin J Am Soc Nephrol 2016;11: function of chronic kidney disease. Stem Cell Res Ther 2015;
633e640. 6:100.
11. Tam-Tham H, Thomas CM. Does the Evidence Support Conservative 33. Yu X, Lu C, Liu H, et al. Hypoxic preconditioning with cobalt of
Management as an Alternative to Dialysis for Older Patients with bone marrow mesenchymal stem cells improves cell migration and
Advanced Kidney Disease? Clin J Am Soc Nephrol 2016;11: enhances therapy for treatment of ischemic acute kidney injury. PLoS
552e554. One 2013;8:e62703.
12. Madariaga MLL, Ott HC. Bioengineering kidneys for transplanta- 34. Zou X, Zhang G, Cheng Z, et al. Microvesicles derived from human
tion. Semin Nephrol 2014;34:384e393. Wharton’s Jelly mesenchymal stromal cells ameliorate renal
13. Cecka J. Kidney transplantation in the United States. Clin Transplant; ischemia-reperfusion injury in rats by suppressing CX3CL1. Stem
2008:1e18. Cell Res Ther 2014;5:1.
14. Brodie JC, Humes HD. Stem cell approaches for the treatment of 35. Ju GQ, Cheng J, Zhong L, et al. Microvesicles derived from hu-
renal failure. Pharmacol Rev 2005;57:299e313. man umbilical cord mesenchymal stem cells facilitate tubular
15. Kim N, Cho SG. Clinical applications of mesenchymal stem cells. epithelial cell dedifferentiation and growth via hepatocyte growth
Korean J Intern Med 2013;28:387e402. factor induction. PLoS One 2015;10:e0121534.
16. Zhang W, Liu L, Huo Y, et al. Hypoxia-pretreated human MSCs 36. Liu P, Feng Y, Dong D, et al. Enhanced renoprotective effect of IGF-
attenuate acute kidney injury through enhanced angiogenic and anti- 1 modified human umbilical cord-derived mesenchymal stem cells on
oxidative capacities. Biomed Res Int 2014;2014:462472. gentamicin-induced acute kidney injury. Sci Rep 2016;6:1.
17. Humphreys BD, Bonventre JV. Mesenchymal stem cells in acute kid- 37. Amiri F, Jahanian-Najafabadi A, Roudkenar MH. In vitro
ney injury. Annu Rev Med 2008;59:311e325. augmentation of mesenchymal stem cells viability in stressful
18. Reinders ME, Bank JR, Dreyer GJ, et al. Autologous bone marrow microenvironments. Cell stress and chaperones 2015;20:
derived mesenchymal stromal cell therapy in combination with ever- 237e251.
olimus to preserve renal structure and function in renal transplant re- 38. Xinaris C, Morigi M, Benedetti V, et al. A novel strategy to enhance
cipients. J Transl Med 2014;12:331. mesenchymal stem cell migration capacity and promote tissue repair
19. Urt Filho A, Oliveira RJ, Hermeto LC, et al. Mesenchymal stem cell in an injury specific fashion. Cell Transplant 2013;22:423e436.
therapy promotes the improvement and recovery of renal function in 39. Karp JM, Teo GSL. Mesenchymal stem cell homing: the devil is in
a preclinical model. Genet Mol Biol 2016;39:290e299. the details. Cell Stem Cell 2009;4:206e216.
Mesenchymal Stem Cells and Kidney Diseases 145

40. Perico N, Casiraghi F, Introna M, et al. Autologous mesenchymal 60. K€ummerer K. Resistance in the environment. J Antimicrob Chemo-
stromal cells and kidney transplantation: a pilot study of safety and ther 2004;54:311e320.
clinical feasibility. Clin J Am Soc Nephrol 2011;6:412e422. 61. Sweileh WM. A prospective comparative study of gentamicin-and
41. Perico N, Casiraghi F, Gotti E, et al. Mesenchymal stromal cells and amikacin-induced nephrotoxicity in patients with normal baseline
kidney transplantation: pretransplant infusion protects from graft renal function. Fundam Clin Pharmacol 2009;23:515e520.
dysfunction while fostering immunoregulation. Transpl Int 2013; 62. Sies H, Stahl W. Vitamins E and C, beta-carotene, and other caroten-
26:867e878. oids as antioxidants. Am J Clin Nutr 1995;62:1315Se1321S.
42. Peng Y, Ke M, Xu L, et al. Donor-derived mesenchymal stem cells 63. Liu P, Feng Y, Dong C, et al. Study on therapeutic action of bone
combined with low-dose tacrolimus prevent acute rejection after marrow derived mesenchymal stem cell combined with vitamin E
renal transplantation: a clinical pilot study. Transplantation 2013; against acute kidney injury in rats. Life Sci 2013;92:829e837.
95:161e168. 64. Cai J, Yu X, Zhang B, et al. Atorvastatin Improves Survival of Im-
43. Trivedi H, Vanikar A, Kute V, et al. The effect of stem cell transplan- planted Stem Cells in a Rat Model of Renal Ischemia-Reperfusion
tation on immunosuppression in living donor renal transplantation: a Injury. Am J Nephrol 2014;39:466e475.
clinical trial. Int J Organ Transplant Med 2013;4:155e162.  Erman A, Cerar A, et al. Improved protective effect
65. Veceric-Haler Z,
44. Gooch A, Doty J, Flores J, et al. Initial report on a phase I clinical of umbilical cord stem cell transplantation on cisplatin-induced kid-
trial: prevention and treatment of post-operative acute kidney injury ney injury in mice pretreated with antithymocyte globulin. Stem
with allogeneic mesenchymal stem cells in patients who require on- Cells Int; 2016:3585362.
pump cardiac surgery. Cell Ther Transplant 2008;1:31e35. 66. Mohammadzadeh-Vardin M, Roudkenar MH, Jahanian-
45. T€ogel FE, Westenfelder C. Kidney protection and regeneration Najafabadi A. Adenovirus-mediated over-expression of Nrf2 within
following acute injury: progress through stem cell therapy. Am J Kid- mesenchymal stem cells (MSCs) protected rats against acute kidney
ney Dis 2012;60:1012e1022. injury. Adv Pharm Bull 2015;5:201e208.
46. Bizzarri A, Koehler H, Cajlakovic M, et al. Continuous oxygen 67. Kensler TW, Wakabayashi N, Biswal S. Cell survival responses to
monitoring in subcutaneous adipose tissue using microdialysis. Anal environmental stresses via the Keap1-Nrf2-ARE pathway. Annu
Chim Acta 2006;573:48e56. Rev Pharmacol Toxicol 2007;47:89e116.
47. Ma T, Grayson WL, Fr€ohlich M, et al. Hypoxia and stem cell-based 68. Ruiz S, Pergola PE, Zager RA, et al. Targeting the transcription fac-
engineering of mesenchymal tissues. Biotechnol Prog 2009;25: tor Nrf2 to ameliorate oxidative stress and inflammation in chronic
32e42. kidney disease. Kidney Int 2013;83:1029e1041.
48. Overath JM, Gauer S, Oberm€uller N, et al. Short-term precondition- 69. Halabian R, Tehrani HA, Jahanian-Najafabadi A, et al. Lipocalin-2-
ing enhances the therapeutic potential of adipose-derived stromal/ mediated upregulation of various antioxidants and growth factors
stem cell-conditioned medium in cisplatin-induced acute kidney protects bone marrow-derived mesenchymal stem cells against unfa-
injury. Exp Cell Res 2016;342:175e183. vorable microenvironments. Cell stress and chaperones 2013;18:
49. Liu H, Xue W, Ge G, et al. Hypoxic preconditioning advances 785e800.
CXCR4 and CXCR7 expression by activating HIF-1a in MSCs. Bio- 70. Bolignano D, Donato V, Coppolino G, et al. Neutrophil gelatina-
chem Biophys Res Commun 2010;401:509e515. seeassociated lipocalin (NGAL) as a marker of kidney damage.
50. Liu H, Liu S, Li Y, et al. The role of SDF-1-CXCR4/CXCR7 axis in Am J Kidney Dis 2008;52:595e605.
the therapeutic effects of hypoxia-preconditioned mesenchymal stem 71. Roudkenar MH, Kuwahara Y, Baba T, et al. Oxidative stress induced
cells for renal ischemia/reperfusion injury. PLoS One 2012;7:e34608. lipocalin 2 gene expression: addressing its expression under the
51. Kiani AA, Kazemi A, Halabian R, et al. HIF-1a confers resistance to harmful conditions. J Radiat Res 2007;48:39e44.
induced stress in bone marrow-derived mesenchymal stem cells. 72. Roudkenar MH, Halabian R, Ghasemipour Z, et al. Neutrophil
Arch Med Res 2013;44:185e193. Gelatinase-associated Lipocalin acts as a protective factor against
52. Sener G, Toklu H, Kapucu C, et al. Melatonin protects against oxida- H 2 O 2 toxicity. Arch Med Res 2008;39:560e566.
tive organ injury in a rat model of sepsis. Surg Today 2005;35: 73. Roudkenar MH, Halabian R, Roushandeh AM, et al. Lipocalin 2
52e59. regulation by thermal stresses: protective role of Lcn2/NGAL against
53. Sener G, Sehirli AO, € Satıro
glu H, et al. Melatonin improves oxida- cold and heat stresses. Exp Cell Res 2009;315:3140e3151.
tive organ damage in a rat model of thermal injury. Burns 2002;28: 74. Bahmani P, Halabian R, Rouhbakhsh M, et al. Neutrophil gelatinase-
419e425. associated lipocalin induces the expression of heme oxygenase-1 and
54. Cardinali DP, Brusco L, Perez LS, et al. Melatonin in sleep disorders superoxide dismutase 1, 2. Cell stress and chaperones 2010;15:
and jet-lag. Neuro Endocrinol Lett 2002;23:9e13. 395e403.
55. Pevet P. Melatonin and biological rhythms. Therapie 1998;53: 75. Roudkenar MH, Halabian R, Bahmani P, et al. Neutrophil gelatinase-
411e420. associated lipocalin: a new antioxidant that exerts its cytoprotective
56. Skwarlo-Sonta K. Melatonin in immunity: comparative aspects. Neu- effect independent on Heme Oxygenase-1. Free Radic Res 2011;
ro Endocrinol Lett 2002;23:61e66. 45:810e819.
57. Mias C, Trouche E, Seguelas MH, et al. Ex vivo pretreatment with 76. Roudkenar MH, Halabian R, Oodi A, et al. Upregulation of neutro-
melatonin improves survival, proangiogenic/mitogenic activity, and phil gelatinase-associated lipocalin, NGAL/Lcn2, in b-thalassemia
efficiency of mesenchymal stem cells injected into ischemic kidney. patients. Arch Med Res 2008;39:402e407.
Stem Cells 2008;26:1749e1757. 77. Bahmani B, Roudkenar MH, Halabian R, et al. Lipocalin 2 decreases
58. Zhao J, Young YK, Fradette J, et al. Melatonin pretreatment of hu- senescence of bone marrow-derived mesenchymal stem cells under
man adipose tissue-derived mesenchymal stromal cells enhances sub-lethal doses of oxidative stress. Cell stress and chaperones
their prosurvival and protective effects on human kidney cells. Am 2014;19:685e693.
J Physiol Renal Physiol 2015;308:F1474eF1483. 78. Halabian R, Roudkenar MH, Jahanian-Najafabadi A, et al. Co-cul-
59. Chen HH, Lin KC, Wallace CG, et al. Additional benefit of combined ture of bone marrow-derived mesenchymal stem cells overexpress-
therapy with melatonin and apoptotic adipose-derived mesenchymal ing lipocalin 2 with HK-2 and HEK293 cells protects the kidney
stem cell against sepsis-induced kidney injury. J Pineal Res 2014;57: cells against cisplatin-induced injury. Cell Biol Int 2015;39:
16e32. 152e163.
146 Roushandeh et al./ Archives of Medical Research 48 (2017) 133e146

79. Carretero OA, Scicli AG. The renal kallikrein-kinin system in human 93. Gatti S, Bruno S, Deregibus MC, et al. Microvesicles derived from
and in experimental hypertension. Klin Wochenschr 1978;56: human adult mesenchymal stem cells protect against ischaemiaere-
113e1125. perfusion-induced acute and chronic kidney injury. Nephrol Dial
80. Han WK, Bonventre JV. Biologic markers for the early detection Transplant 2011;26:1474e1483.
of acute kidney injury. Curr Opin Crit Care 2004;10:476e482. 94. Reis LA, Borges FT, Simoes MJ, et al. Bone marrow-derived mesen-
81. Hagiwara M, Shen B, Chao L, et al. Kallikrein-modified mesen- chymal stem cells repaired but did not prevent gentamicin-induced
chymal stem cell implantation provides enhanced protection against acute kidney injury through paracrine effects in rats. PLoS One
acute ischemic kidney injury by inhibiting apoptosis and inflamma- 2012;7:e44092.
tion. Hum Gene Ther 2008;19:807e819. 95. Bi B, Guo J, Marlier A, et al. Erythropoietin expands a stromal cell
82. Wang G, Zhang Q, Zhuo Z, et al. Enhanced Homing of population that can mediate renoprotection. Am J Physiol Renal
CXCR-4 Modified Bone MarroweDerived Mesenchymal Stem Physiol 2008;295:F1017eF1022.
Cells to Acute Kidney Injury Tissues by Micro-BubbleeMedi- 96. Sharples EJ, Patel N, Brown P, et al. Erythropoietin protects the kid-
ated Ultrasound Exposure. Ultrasound Med Biol 2016;42: ney against the injury and dysfunction caused by ischemia-reperfu-
539e548. sion. J Am Soc Nephrol 2004;15:2115e2124.
83. Liu N, Tian J, Cheng J, et al. Migration of CXCR4 gene-modified 97. Zhou Y, Xu H, Xu W, et al. Exosomes released by human umbilical
bone marrow-derived mesenchymal stem cells to the acute injured cord mesenchymal stem cells protect against cisplatin-induced renal
kidney. J Cell Biochem 2013;114:2677e2689. oxidative stress and apoptosis in vivo and in vitro. Stem Cell Res
84. Si X, Liu X, Li J, et al. Transforming growth factor-b1 promotes Ther 2013;4:34.
homing of bone marrow mesenchymal stem cells in renal 98. Kilpinen L, Impola U, Sankkila L, et al. Extracellular membrane ves-
ischemia-reperfusion injury. Int J Clin Exp Pathol 2015;8: icles from umbilical cord blood-derived MSC protect against
12368e12378. ischemic acute kidney injury, a feature that is lost after inflammatory
85. Zou X, Gu D, Xing X, et al. Human mesenchymal stromal cell- conditioning. J Extracell Vesicles 2013;2:21927.
derived extracellular vesicles alleviate renal ischemic reperfusion 99. Zhang G, Zou X, Huang Y, et al. Mesenchymal Stromal Cell-Derived
injury and enhance angiogenesis in rats. Am J Transl Res 2016;8: Extracellular Vesicles Protect Against Acute Kidney Injury Through
4289e4299. Anti-Oxidation by Enhancing Nrf2/ARE Activation in Rats. Kidney
86. Yuan L, Wu M-J, Sun H-Y, et al. VEGF-modified human embryonic Blood Press Res 2016;41:119e128.
mesenchymal stem cell implantation enhances protection against 100. Furlani D, Ugurlucan M, Ong L, et al. Is the intravascular adminis-
cisplatin-induced acute kidney injury. Am J Physiol Renal Physiol tration of mesenchymal stem cells safe?: Mesenchymal stem cells
2011;300:F207eF218. and intravital microscopy. Microvasc Res 2009;77:370e376.
87. Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles: arte- 101. Herrera M, Mirotsou M. Stem cells: potential and challenges for kid-
facts no more. Trends cell Biol 2009;19:43e51. ney repair. Am J Physiol Renal Physiol 2014;306:F12eF23.
88. Bruno S, Grange C, Deregibus MC, et al. Mesenchymal stem cell- 102. Iero M, Valenti R, Huber V, et al. Tumour-released exosomes and
derived microvesicles protect against acute tubular injury. J Am their implications in cancer immunity. Cell Death Differ 2008;15:
Soc Nephrol 2009;20:1053e1067. 80e88.
89. Bruno S, Grange C, Collino F, et al. Microvesicles derived from 103. Dekel B, Burakova T, Arditti FD, et al. Human and porcine early kid-
mesenchymal stem cells enhance survival in a lethal model of acute ney precursors as a new source for transplantation. Nat Med 2003;9:
kidney injury. PLoS One 2012;7:e33115. 53e60.
90. Sabin K, Kikyo N. Microvesicles as mediators of tissue regeneration. 104. Gupta AK, Jadhav SH, Tripathy NK, et al. Fetal kidney stem cells
Transl Res 2014;163:286e295. ameliorate cisplatin induced acute renal failure and promote renal
91. Konala VB, Mamidi MK, Bhonde R, et al. The current landscape of angiogenesis. World J Stem Cells 2015;7:776e788.
the mesenchymal stromal cell secretome: A new paradigm for cell- 105. Yu Y, Shao Y, Ding Y, et al. Decellularized kidney scaffold-mediated
free regeneration. Cytotherapy 2016;18:13e24. renal regeneration. Biomaterials 2014;35:6822e6828.
92. Biancone L, Bruno S, Deregibus MC, et al. Therapeutic potential of 106. Xie H, Wang Z, Zhang L, et al. Development of an angiogenesis-
mesenchymal stem cell-derived microvesicles. Nephrol Dial Trans- promoting microvesicle-alginate-polycaprolactone composite graft
plant 2012;27:3037e3042. for bone tissue engineering applications. Peer J 2016;4:e2040.

Vous aimerez peut-être aussi