Sie sind auf Seite 1von 18

TISSUE-SPECIFIC STEM CELLS

Multipotential Differentiation of Human Urine-Derived Stem Cells:


Potential for Therapeutic Applications in Urology
SHANTARAM BHARADWAJ,a GUIHUA LIU,a YINGAI SHI,a RONGPEI WU,a,b BIN YANG,a,c TONGCHUAN HE,d
YUXIN FAN,e XINYAN LU,f XIAOBO ZHOU,g HONG LIU,h ANTHONY ATALA,a JAN ROHOZINSKI,a,i YUANYUAN ZHANGa
a
Wake Forest Institute of Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North
Carolina, USA; bDepartment of Urology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou,
GuangDong, People’s Republic of China; cDepartment of Urology, Shanghai Tenth People’s Hospital, Tongji
University School of Medicine, Shanghai, People’s Republic of China; dMolecular Oncology Laboratory,
Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois, USA; eJohn Welsh
Cardiovascular Diagnostic Laboratory at Department of Pediatric, Baylor College of Medicine, Houston, Texas,
USA; fClinical Cytogenetics Laboratory, Section of Hematology-Oncology, Department of Pediatrics, Baylor
College of Medicine, Houston, Texas, USA; gRadiology/Translational Biology Department, The Methodist
Hospital Research Institute, Houston, Texas, USA; hCenter for Bioengineering and School of Electrical and
Computer Engineering, University of Oklahoma, Norman, Oklahoma, USA; iDepartment of Obstetrics and
Gynecology, Baylor College of Medicine, Houston, Texas, USA

Key Words. Urine • Bladder • Stem cells • Telomerase • Differentiation • Smooth muscle • Urothelial cells

ABSTRACT
We sought to biologically characterize and identify a When the differentiated USCs were seeded onto a scaf-
subpopulation of urine-derived stem cells (USCs) with fold and subcutaneously implanted into nude mice,
the capacity for multipotent differentiation. We demon- multilayered tissue-like structures formed consisting of
strated that single USCs can expand to a large popula- urothelium and smooth muscle. Additionally, USCs were
tion with 60–70 population doublings. Nine of 15 able to differentiate into endothelial, osteogenic, chon-
individual USC clones expressed detectable levels of telo- drogenic, adipogenic, skeletal myogenic, and neurogenic
merase and have long telomeres. These cells expressed lineages but did not form teratomas during the 1-month
pericyte and mesenchymal stem cell markers. Upon study despite telomerase activity. USCs may be useful in
induction with appropriate media in vitro, USCs differ- cell-based therapies and tissue engineering applications,
entiated into bladder-associated cell types, including including urogenital reconstruction. STEM CELLS
functional urothelial and smooth muscle cell lineages. 2013;31:1840–1856
Disclosure of potential conflicts of interest is found at the end of this article.

differentiation [3]. In the clinical setting, the use of autolo-


INTRODUCTION gous stem cells is advantageous, as they do not induce
immune responses and/or rejection. Because tissue-specific
The ability of stem cells to differentiate into various tissue- stem cells are a very small subpopulation of cells, they are
specific lineages is the basis for exploring their use in cell- difficult to isolate from most differentiated somatic cells pres-
based therapies and tissue regeneration. Tissue-specific stem ent in the tissues and organs [4]. In this article, we demon-
cells and/or other progenitor cells exist in almost every tissue strate that a population of stem cells can be easily isolated
and organ of the body [1]. These cells differ from somatic from human urine; we term these urine-derived stem cells
cells by their ability to differentiate into the many types of (USCs). Single clones of USCs can expand to yield a large
cells required for tissue maintenance, repair, and regeneration population, and they have the capacity for multipotent differ-
[2]. Once isolated, these stem cells can expand in culture, and entiation. These cells express a combination of pericyte and
under appropriate experimental conditions, give rise to a mesenchymal stem cell markers. USCs express detectable lev-
variety of cell types in vitro via induction of lineage-specific els of telomerase and have long telomeres but did not form

Author contributions: S.B. and G.L.: collection and assembly of data, data analysis and interpretation, and manuscript writing; Y.S.,
R.W., B.Y., T.H., and Y.F.: collection and assembly of data and data analysis and interpretation; X.L., X.Z., H.L., and J.R.: data
analysis and interpretation; A.A.: financial support, administrative support, and provision of study material; Y.Z.: conception and design,
data analysis and interpretation, and manuscript writing; S.B., G.L., Y.S., R.W., B.Y., T.H, Y.F., X.L., X.Z., H.L., J.R., A.A., and Y.Z.:
final approval of manuscript. S.B. and G.L. contributed equally to this article.
Correspondence: Yuanyuan Zhang, M.D. Ph.D., Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine,
Medical Center Boulevard, Winston-Salem, NC 27157, USA. Telephone: 336-713-1189; Fax: 336-713-7290; e-mail: yzhang@
wakehealth.edu Received December 11, 2012; accepted for publication April 21, 2013; first published online in STEM CELLS EXPRESS
May 10, 2013. V
C AlphaMed Press 1066-5099/2013/$30.00/0 doi: 10.1002/stem.1424

STEM CELLS 2013;31:1840–1856 www.StemCells.com


Bharadwaj, Liu, Shi et al. 1841

teratomas in vivo during the 1-month study. In this study, we green fluorescent protein (GFP) as a marker for monitoring infec-
have characterized USCs comprehensively and identified multi- tion efficiency. An analogous adenovirus only expressing GFP
lineage differentiation potential of USCs at gene and protein (AdGFP) was used as a control [14, 15, 17-24].
expression levels, cellular function, and tissue formation. We
have extended our previously published data to further confirm Multipotent Differentiation Potential of USCs
the plasticity of USCs in vivo and induced these cells to poten- In Vitro
tially give rise to different tissue types, systematically analyzed In order to characterize the differentiation potential of USC
the function of differentiated cells, particularly urological tis- clones, cells were subjected to the following regimen described
sues such as urothelium, smooth muscle, and microvessel. below, and their changes in morphology and/or histochemical
Obtained via a simple and noninvasive method [5–9], USCs staining for specific components were recorded. The multipoten-
could potentially provide a virtually limitless supply of autolo- tial capability of independent USC clones was evaluated for vary-
gous cells for regenerative medicine and tissue engineering ing levels of response (þ and ) as follows. þþþþ, 80%–100%
applications, in urology [10–12] and other fields as well. differentiated; þþþ, 60%–80% differentiated; þþ, 40%–60%
differentiated; þ, 20%–40% differentiated; 6, 20% or less differ-
entiated; , no differentiation/change in morphology.

MATERIALS AND METHODS Smooth Muscle Cell Induction. USCs were plated at 2,000
cells per centimeter square in smooth muscle differentiation media
Isolation of Human USCs containing equal amounts of Dulbecco’s modified Eagle’s medium
(DMEM) (high glucose) and EFM with 10% FBS and the growth
Approval for this study was obtained from the Wake Forest Uni-
factors transforming growth factor beta 1 (TGF-b1) and platelet
versity Health Sciences Institutional Review Board. To determine
derived growth factor-BB (PDGF-BB) (R&D Systems, Minneapo-
whether USCs can be obtained for cell differentiation from
lis, MN, http://www.rndsystems.com) at concentrations of 2.5 and
healthy individuals at different ages, 59 fresh urine samples were
5 ng/mL, respectively[5]. Cell morphology was recorded before
collected from 17 healthy people ranging from 5 to 75 years old.
and after growth factor additions for up to 14 days.
USCs were isolated and characterized as previously described [6,
8, 9]. Briefly, USCs were grown and expanded in initial/culture
media composed of keratinocyte serum-free medium (KSFM) and Uroepithelial Induction. USCs were plated at 3,000 cells per
embryonic fibroblast medium (EFM) mixed at a ratio of 1:1 with centimeter square in a mixture of medium containing equal
5% fetal bovine serum (FBS) [9]. Only wells in 24 plates that amounts of KSFM and EFM with 2% serum and the growth fac-
contained single cells were scored and cultured. To keep results tor, epidermal growth factor (EGF) (R&D Systems, Minneapolis,
more consistent, cell clones were selected from six donors (3–7 MN) at 30 ng/mL for 14 days [5].
clones per individual) between 28 and 41 years old for further
assessments (Supporting Information Table S1). Endothelial Induction. USCs were plated at a density of
5,000 cells per centimeter square on fibronectin-coated dishes
Cell Morphology and Proliferation (2 lg/cm2) and allowed to grow for 2 days to reach subconfluent
Cell morphology and proliferation capacity were assessed for levels. Endothelial basal medium (EBM2, Lonza) supplemented
each USC clone from 17 donors. For cell population doubling with 50 ng/mL vascular endothelial growth factor (VEGF) was
(PD) studies, 1,720 cells per centimeter square were plated and used to culture USCs for 9 days for induction of endothelial-line-
manual counts were performed 3 or 4 days later (70%–85% con- age differentiation as reported earlier [25].
fluence). The PD and doubling time were calculated. For cell
morphology images, the cells were stained with 0.05% crystal
Neuronal Induction. USCs were seeded in culture dishes
violet dye.
(6,000 cells per centimeter square) and grown in preinduction
Fluorescence Activated Cell Sorting Analysis, medium (PIM) containing DMEM supplemented with 20% FBS
and 10 ng/mL basic fibroblast growth factor (Peprotech, Rocky
Immunofluorescence Analysis, RNA Analysis, Hill, NJ, http://www.peprotech.com). After 24 hours of culture in
Immunoblotting PIM, the medium was substituted with nerve induction medium
Fluorescence activated cell sorting (FACS), immunofluorescence for a further 48 hours.
staining, RNA analysis, immunblotting were used to characterize
the USCs and differentiated USCs according the Supporting In- Osteogenic Induction. USCs were seeded at a density of
formation methods. 4,000 cells per centimeter square and cultured in serum contain-
ing DMEM low-glucose medium with osteogenic supplements.
Determination of Telomerase Activity
USCs isolated from 28 urine samples, collected from 15 healthy
individuals, were used. Whole cell lysates from 2  104 USCs Chondrogenic Induction. For high-density cell pellet cultures,
(p3) were assayed using a Telo TAAGG ELISA kit (Roche 3  105 cells were centrifuged at 600g for 5 minutes in a sterile
Applied Sciences, Upper Bavaria, Germany, http://www.roche- 15 mL conical polypropylene tube and incubated overnight to
applied-science.com), according to the manufacturer’s form an aggregate. The culture medium was replaced with 0.5
instructions. mL of chondrogenic medium for a total of 28 days. USCs were
also cultured at 5,000 cells per centimeter square on regular tis-
Creation of Adenoviruses Expressing BMP2, BMP9, sue culture dishes using chondrogenic medium for 28 days.
and Green Fluorescent Protein
The recombinant adenovirus vectors were generated by using the Adipogenic Induction. USCs were seeded at a density of
AdEasy system [13–17]. Briefly, the coding regions of human 4,000 cells per centimeter square and cultured in serum contain-
BMP2 and BMP9 were polymerase chain reaction (PCR) ing adipogenic medium.
amplified and subcloned into the shuttle vector pAdTrack-TO4.
After sequencing verification, the shuttle vectors were used to Skeletal Myogenic Induction. USCs were induced by two
generate adenoviral recombinants. Adenoviruses were produced methods for 1 month, respectively: (a) using skeletal muscle sup-
and amplified in HEK-293 cells. AdBMP viruses also express plements and DMEM containing 50 lM hydrocortisone, 0.1 lM
www.StemCells.com
1842 Multipotential of Human Urine Derived Stem Cells

Figure 1. Schematic isolation of urine-derived stem cell (USCs) and morphology of USC clone with passage. (A): USCs were derived from
the urine by three major steps: isolation, selection, and propagation. (B): A single USC (inset) is followed through different passages (p0–p12).
The cells were expanded to a colony and were cultured in keratinocyte serum-free medium and embryonic fibroblast medium with 5% serum and
images recorded with passage. Images shown at 100.

dexamethasone, 10% FBS, and 5% horse serum was used to cul- To study the feasibility of adenoviral infection and expression
ture USCs; (b) conditioned medium was collected from skeletal of transgenes in USCs, the GFP expressing transgene-BMP2
muscle cell culture for 12 hours. (Group 1), BMP9 (Group 2), or GFP alone (Group 3, control)
were implanted in 15 female athymic nude (nu/nu) mice (4–6
Cell-Collagen Lattice Contractility Assay weeks old; Harlan Sprague Dawley, five mice per group). Briefly,
the USCs were seeded at subconfluence and infected with adeno-
To measure cell contractile function in vitro, smooth muscle dif-
viruses expressing GFP-BMP2, GFP-BMP9, and GFP at an multi-
ferentiated USCs, noninduced USCs, and human ureter smooth
plicity of infection (MOI) of 10. For experiments involving bone
muscle cells (SMCs) were assessed as reported earlier [26] with
morphogenic protein (BMP) infection, the cells were implanted
minor modifications (Supporting Information Methods).
as described [15, 16, 23, 24, 30-33]. Briefly, subconfluent USCs
were infected with adenoviruses. At 16-hour postinfection, cells
Permeability Studies were harvested and resuspended in phosphate buffered saline
To assess barrier function, urothelial-differentiated USCs, nonin- (PBS) for subcutaneous injection (2  106 per injection) into the
duced USCs, SMCs, and ureter urothelial cells (UCs) were tested. flanks of the mice. At 4 weeks after implantation, animals were
Cells were cultured on 0.4 lm transwell inserts (Becton Dickin- sacrificed, and the implantation sites were retrieved for histologic
son, Franklin Lakes, NJ, http://www.bd.com) placed in six-well evaluation.
dishes as reported earlier [27] with minor modifications (Support-
ing Information Methods). Histochemical Analysis
For Bone. The implanted grafts with induced USCs were har-
In Vitro Tube Formation Assay vested after 28 days and fixed in 95% ethanol prior to histochem-
Endothelial-differentiated USCs (5,000 cells in 0.1 mL medium) ical staining as described in Supporting Information methods.
were seeded on Matrigel and the plates left undisturbed for 18–
20 hours. Bright-field images were then captured for analysis of For Cartilage. The grafts with induced USCs were histochemi-
network formation. Nontreated USCs served as negative or back- cally stained at the end of 21 days after fixing cells with 10% for-
ground controls with human umbilical vein endothelial cells as a malin. Sulfated glycosaminoglycan (GAG) synthesis was visual-
positive control. ized by Alcian blue staining as described in Supporting
Information Methods.
In Vivo Study
Institutional Animal Care and Use Committee approvals for this For Fat. The grafted samples were fixed in 4% formaldehyde
project were obtained for animal surgery. Small intestinal submu- solution for 20 minutes at room temperature and incubated after
cosa (SIS) as a scaffold was harvested from porcine intestine and being stained with 0.5% oil red-O in propylene glycol for
processed as previously described [ 7, 28]. In vitro cell-seeded 50 minutes. Accumulation of lipid globules was visualized after
scaffolds either with urothelially differentiated USCs or smooth extensive water washes under a light microscope.
muscle differentiated USCs were seeded on the mucosal side of For the in vivo cell-seeded SIS grafts and collagen scaffolds,
SIS, as previously described [7, 28, 29]. The cells were main- immunohistochemical triple staining was performed by incubating
tained on scaffolds for 14 days and then cell-scaffold constructs specific antibodies (Supporting Information Table S3) along with
were implanted in 22 immunodeficient female mice for 1 month. human nuclear antibody for detection of implanted human cells.
The constructs were harvested and assessed by histology and For the BMP-infected USCs, serial sections of the paraffin-em-
immunohistochemistry. bedded samples were deparaffinized, then rehydrated in a
Figure 2. Determination of stem cell surface marker expression in USCs by flow cytometry. (A): USCs at passage three were labeled with
mesenchymal stem cell markers (CD29, CD44, CD54, CD73, CD90, CD105, CD166 and STRO-1). (B): USCs (p3) were labeled with hematopoi-
etic stem cell markers (CD11b, CD14, CD19, CD31, CD34, CD45, and MHC-I and -II). No visible positive labeled cells were observed except
for the MHC-class-I protein. (C): Analysis of USCs for pericyte markers; (i) by fluorescence activated cell sorting (FACS) (CD146, CD140b, and
NG2), (ii) semiquantitative reverse transcriptase polymerase chain reaction (RT-PCR) for the three pericyte markers on different passages of cul-
tured USCs (p2-7). (D): Embryonic stem cell marker detection by (i) FACS analysis (Tra-1-60, Tra-1-81, SSEA4); (ii) real-time PCR (sox2,
oct4, c-myc, and klf4), and (iii) immunofluorescence staining (sox2, oct4, c-myc, and klf4). Representative positive cells stained green at their
membranes (SSEA4) or yellow in their nucleus are shown by arrows. Nuclei were counterstained with PI (red). Inset shows positive control
NCCIT cells. Scale ¼ 50 lm. Abbreviations: BMSC, bone marrow mesenchymal stromal cell; ES cell, embryonic stem cell; GAPDH, glyceralde-
hyde 3-phosphate dehydrogenase; MHC, major histocompatibility complex; NG2, NG2 chondroitin sulfate proteoglycan; NCCIT, embryonic car-
cinoma cell line; PDGF-rb, platelet-derived growth factor receptor beta; PI, propidium iodide; SSEA-1, stage specific embryonic antigen-1; TRA,
tumor related antigen; USC, urine-derived stem cell.
1844 Multipotential of Human Urine Derived Stem Cells

Figure 3. Telomerase activity and length in USCs. (A): Nine out of the 15 samples tested showed significant telomerase activity (red bars)
over the background. Positive represents lysates prepared from HEK-293 cells; heat-denatured HEK-293 lysate served as the negative control.
(B): DNA extracted from USCs at different passages analyzed for telomere length. Lanes 1 and 2, USCs at passages 2 and 7, respectively; lanes
3 and 4, low and high telomere length controls; lane 5, DNA marker. Abbreviations: BMSCs, bone marrow mesenchymal stromal cells; SMCs,
human bladder smooth muscle cells; USCs, urine-derived stem cells.

graduated fashion, and stained with hematoxylin and eosin, tion Table S5). Additionally, more than 90% of cultured
Alcian blue, and Masson’s trichrome [15, 16, 23, 24, 30-33]. Oil- USCs and a few cells isolated from fresh urine from a prior
red O staining was carried out on the decalcified and frozen sec- culture expressed pericyte markers (CD146, Fig. 2C).
tion samples, as previously described [15, 16, 23, 24, 30-33]. However, only a few USCs expressed NG2 and PDGF-rb,
Additionally, to determine the origin of the USCs in urine, although we found the cells consistently displayed the NG2
human renal cortex, medullar, renal pelvic and ureter tissues were and PDGF-rb genes in serial passages from p2 to p7 (Fig.2C;
assessed with immunofluorescent staining of the pericyte marker Supporting Information Table S5). USCs expressed the em-
(i.e., CD 146 antibody). bryonic stem cell markers TRA 1-60, TRA 1-81, and SSEA4,
Fluorescence In Situ Hybridization and Amelogenin as well as Sox2, Oct3/4, c-Myc, and klf4. Gene expression
Gene PCR Analysis profiles and nuclear protein levels for these markers are
shown in Figure 2D. Importantly, 9 of 15 clones tested
To determine the origin of the USCs in urine, study of cells with showed telomerase activities more than twice those of the
y-chromosome from sex-mismatched kidneys following renal background levels (0.2 units, p < .005, Student’s t test) (Fig.
transplantation (i.e., male donor-to-female recipient kidney trans-
3A). An assay of terminal restriction fragments [35] showed
plant combination) by fluorescence in situ hybridization (FISH)
and amelogenin gene PCR analysis was performed. FISH using
that USCs had long telomeres; the average telomere length
alpha satellite probe for chromosomes X and Yq12 region from remained constant between passages 2 and 7 (Fig. 3B). How-
Abbott Molecular (Abbott Park, Illinois, U.S.A. http://www. ever, both bone marrow mesenchymal stromal cells (BMSCs)
abbottmolecular.com) was performed according to the manufac- and SMCs displayed telomerase levels below the background
turer’s protocol [34] as described in Supporting Information control levels.
Methods.
Multipotent Differentiation of USCs In Vitro
Statistical Analysis USCs derived from one single clone (p4) were able to differ-
Student’s t test and paired-sample t test were used for statistical entiate into smooth muscle lineage (mesodermal origin) and
analysis. p values of .05 or less were considered significant. One- urothelial lineage (endodermal origin) (Fig. 4A). The differen-
way or two-way ANOVA followed by a Student-Newman-Keuls tiation capacity of USCs was repeatable in each USC clone
post hoc test for multiple comparisons were used when (six clones per donor) from six donors. (Supporting Informa-
appropriate. tion Table S1). Myogenic differentiated USCs became elon-
gated and spindle-shaped (Fig. 4B) and expressed at least a
twofold increase in SMC-specific gene transcripts (desmin,
RESULTS myosin, smoothelin, and a-SMA) upon induction (Fig. 4C,
4D). Up to 80% of induced cells expressed smoothelin, a
smooth muscle-specific protein marker (Fig. 4E–4G).
Characteristics of USCs Although noninduced USCs expressed detectable levels of
Isolation of USCs from urine involves minimal processing a-SMA, the fluorescent intensity of a-SMA increased in a
(Fig. 1A). Single, small, compact ‘‘rice-grain’’ like cells was time-dependent manner upon induction (Fig. 4F, 4G). In cell-
observed 2–3 days after initial seeding. These cells further collagen contractility analysis, myogenic-induced USCs and
formed clones within an additional 4–6 days (Fig. 1B). FACS SMC demonstrated rapid lattice contraction of 24.4% 6 3.5%
analysis showed that USCs consistently expressed mesenchy- and 20.8% 6 1.9%, respectively, which was significantly
mal stem cells (MSCs)-like cell surface markers (CD29, higher than the relative contraction occurred in the
CD44, CD54, CD73, CD90, CD105, CD166 and STRO-1) noninduced USCs (p < .05) (Fig. 4H).
(Fig. 2A), but not hematopoietic stem cell markers (CD11b, Urothelial differentiated cells developed a cobblestone-
CD14, CD19, CD31, CD34, CD45, and MHC-II) except for like morphology (Fig. 4I) and expressed urothelial-specific
MHC-1, endothelial cell markers (CD31), or human leukocyte transcripts and proteins of uroplakin-III, uroplakin-Ia, and
antigen(locus)DR (HLA-DR) (Fig. 2B; Supporting Informa- generic epithelial cell markers CK7 and AE1/AE3 (Fig. 4I–K).
Figure 4. Differentiation of single USC clone into UCs and SMCs. (A): USCs (p3) were used to differentiate into two distinct lineages. Culture
in SMCs-lineage differentiation (2.5 ng/mL TGF-b1 and 5 ng/mL PDGF-BB) and UCs-lineage differentiation (30 ng/mL EGF) medium was used
for 14 days. (B): Morphology of USCs after induction with SMC induction media for 7 and 14 days, respectively. Human ureter SMCs were pos-
itive controls. Scale bar ¼ 100 lm. (C): Semiquantitative reverse transcriptase polymerase chain reaction (RT-PCR) and (D) quantitative real-
time PCR was performed on total RNA using smooth muscle-specific primers (desmin, myosin, aSMA, and smoothelin). Ind-USCs, USCs
induced to myogenic lineage. SMCs RNA was used as a positive control and H2O: negative control. (E): Detection of smooth muscle-specific
proteins by immunoblotting. Specific signals (bands) were seen in lanes with proteins from induced USCs and positive control. (F): Immunofluo-
rescence staining of myogenic induced USCs with time. Ind-USCs-7d and -14d, USCs induced with myogenic medium for 7 and 14 days, respec-
tively. Scale bar ¼ 50 lm. (G): Quantitation of immunofluorescent images for the different markers shown in (F). Differentiation to SMC
lineage significantly increased expression of smooth muscle-specific markers when compared to nontreated USCs as controls. *, p  .05; **, p <
.005. (H): Contractility after myogenic differentiation of USCs. Serum and calcium ionophore promoted significant lattice contraction in SMC
(cross-hatched bars) and induced USCs-collagen lattices (open bars) but not in noninduced USCs-collagen lattice (patterned bars). (I): Morphol-
ogy of USCs after induction with UC induction media following 7 and 14 days. The cell morphology changed from a ‘‘rice-grain-like’’ to a cu-
boidal morphology, which was more pronounced with time. (J): Quantitative real-time PCR (i, ii) and (K) semiquantitative RT-PCR (iii) was
performed on total RNA using urothelial-specific primers (uroplakins-Ia and -III, CK7). Ind-USCs, USCs induced to urothelial lineage. UCs were
used as a positive control and H2O as negative control. (L): Detection of urothelial-specific proteins (uroplakin-Ia, -III, AE1/AE3 and CK7) by
immunoblotting. Specific signals (bands) were seen in lanes with proteins from induced USCs and positive control. (M): Immunofluorescent
staining of urothelial-induced USCs with time. Ind-USC-7d and -14d, USCs induced with urothelial medium for 7 and 14 days, respectively. Spe-
cific marker staining (green) and nuclear staining by propidium iodide (red) are seen. Scale bar ¼ 50 lm. (N): Quantitation of immunofluorescent
images for the different markers shown in (E). Differentiation to an urothelial lineage significantly increased expression of urothelial-specific
markers when compared to nontreated USC controls. *, p  .05; **, p < .005. Functional analysis of urothelial-differentiated USC: (O): Tran-
script analysis by real-time and (P) semiquantitative RT-PCR for functional tight junctions (ZO-1 and E-cadherin). (Q): Immunoblot analysis for
the same tight junction proteins on day 14. (R): Membrane junctions of UC-induced USCs were intensely stained with tight junction antibodies
(ZO-1, occludin, E-cadherin, and cingulin). Inset shows overall staining at a lower magnification. Scale bar ¼ 50 lm. (S): Transmission electron
microscopic analysis for tight junctions in UC differentiated USCs. Arrows indicate position of tight junction; arrow head indicates desmosomes.
(T): Barrier function analysis was performed on USC differentiated to UC-like cells for 3 days. Fluorescein isothiocyanate (FITC)-dextran was
incubated with cells grown on inset; media in the bottom chamber was analyzed after 3 hours. Results were plotted as a percentage of the ‘‘no
cell’’ control (**, p < .05). Abbreviations: ASMA, alpha-smooth muscle actin; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; EGF, epi-
dermal growth factor; Non-USC, noninduced; PI, propidium iodide; SMCs, authentic human ureter smooth muscle cells; UCs, human ureter uro-
thelial cells as control; USCs Con, noninduced USCs control; Ind-USCs, USCs induced to myogenic lineage, urothelial lineage; Ind-USCs-7d and
-14d, USCs induced with myogenic medium, urothelial medium for 7 and 14 days.
1846 Multipotential of Human Urine Derived Stem Cells

Figure 4. (Continued)

In addition, more than 90% of USCs expressed urothelial containing solidified Matrigel (Fig. 5A). In addition, the
markers after 14 days of culture, compared to 55%–70% of differentiated cells expressed endothelial cell-specific gene
USCs that expressed urothelial markers after 7 days of culture transcripts (vWF and CD31), protein markers (CD31, vWF,
(Fig. 4M, 4N). USCs that expressed urothelial protein markers KDR, FLT1, and eNOS), and a tight endothelial junction
increased significantly when the cells were cultured in medium marker (VE-cadherin) (Fig. 5B, 5C).
containing 30 ng/mL EGF (60%–70%), compared to expansion Neurogenic differentiated USCs exhibited several neuro-
medium containing 7.5 ng/mL EGF (30%–40%) [9]. In an genic extensions and processes (arrows) (Fig. 5D) and
assay of cellular barrier function, urothelial differentiated USCs expressed neuronal transcripts (Fig. 5E). Approximately 40%
expressed specific tight junction gene and protein markers (ZO- of induced USCs expressed the neuronal specific protein
1 and E-cadherin) (Fig. 4O–4R). Transmission electron micros- markers nestin, S100, NF200, and GFAP (Fig. 5F).
copy demonstrated the ultrastructure of tight junction formation Skeletal myogenic differentiated USCs displayed an elon-
between neighboring cells (Fig. 4S). In barrier function assays, gated spindle-shaped morphology (Fig. 5G). This change was
urothelial differentiated USCs showed at least a 60% decrease accompanied by expression of skeletal muscle-related tran-
in leakage of fluorescent tracer through the insert in vitro, com- scripts (Fig. 5H). Formation of myotube-like structures was
pared to nondifferentiated USCs (Fig. 4T). seen in 50%–60% of cells stained for MyoD (Fig. 5I, arrows).
Endothelial differentiated USCs exhibited in vitro Nuclear staining with antibodies to myogenin revealed
‘‘vessel’’ formation when they were plated in culture wells differentiated populations (Fig. 5I).
Bharadwaj, Liu, Shi et al. 1847

Figure 4. (Continued)

Finally, USCs were also induced to differentiate into blue, and safranin-O for GAGs and all were positive (Fig. 5J-
osteogenic, adipogenic, and chondrogenic lineages using pub- v,vi,vii,viii). About 60% of the induced USCs that gave rise
lished differentiation protocols [36]. Adipogenic-differentiated to the chondrogenic lineage markers expressed Sox9, collagen
USCs were positive for oil red-O staining (Fig. 5J-i) and II, and aggrecan (Fig. 5K-iii). It appeared that USCs can
30%–40% of adpogenically induced USCs expressed adipo- highly efficiently differentiate into almost all of the above
cyte gene markers including peroxisome proliferator-activated cell lineages except chondrogenic differentiation (Supporting
receptor (PPARc), acetyl-CoA synthase, adiponectin, Information Table S1).
CCAAT/enhancer-binding protein a; fatty acid binding protein This supposition was confirmed with a study performed on
4, and lipoprotein lipase (Fig. 5K-i). Osteogenic-differentiated USC clones from three different donor samples (Fig. 5L; Sup-
USCs appeared to produce mineralized tissue as shown via porting Information Table S1). One of the clones could differ-
von Kossa, alkaline phosphatase, and Alizarin Red staining entiate to multiple lineages (osteogenic, chondrogenic, adipo-
for calcium deposition (Fig. 5J-ii,iii,iv). Induction of the genic myogenic, urothelial, and endothelial) whereas another
osteogenic markers osteocalcin, runt-related transcription was highly restricted to a few lineages (osteogenic, myogenic,
factor-2, and alkaline phosphatase (Fig. 5K-ii) occurred in urothelial, and endothelial). The other clones showed a mixed
70%–80% of osteogenically induced USCs. Chondrogenic-dif- response in vivo. This result confirms the presence of cells at
ferentiated USCs were stained with Alcian blue, toluidine varying stages of differentiation in human urine.
www.StemCells.com
1848 Multipotential of Human Urine Derived Stem Cells

Figure 4. (Continued)

Possible Renal Origin of USCs CD146, pax2, and pax8, were expressed in cultured USCs from
USCs from female recipients of male kidneys showed the different individuals (Fig. 6B, 6C). Importantly, USCs
presence of the Y-chromosome when assessed by FISH and expressed podocytes and parietal cell gene and protein markers
amelogenin gene PCR analysis (Fig. 6A), indicating that (i.e., synaptopodin and podocin) (data not shown). However,
USCs originated from either the upper urinary tract, including these genes were not expressed on cultured ureter UCs. Impor-
kidney, renal pelvic, and/or upper segment of the transplanted tantly, using immunofluorescent staining, we found that parietal
ureters. To further reveal whether USCs were from kidney or cells and podocytes in glomerular tissues and vessels from
ureters, we tested for kidney-specific gene expression. The human kidney cortex expressed CD146 (Fig. 6D), with no
normal renal cell genes and proteins, CD224, CD13, NR3C2, staining in renal tubule epithelial cells or ureter mucosa.
Figure 5. USCs undergo multipotential differentiation in vitro. (A–C): Endothelial differentiation of USCs. USCs (p3) were induced to endothelial
lineage by culture in endothelial basal medium containing vascular endothelial growth factor (VEGF) 50 ng/mL for 14 days. (A): In vitro vessel forma-
tion. Endothelial differentiated USCs were cultured on Matrigel for 18 hours to form branched networks (angiogenesis) and tubular structures. Scale
bar ¼ 100 lm. (B): Expression analysis of endothelial-specific transcripts by reverse transcriptase polymerase chain reaction (RT-PCR). (C): Immuno-
fluorescence staining using endothelial-specific markers revealed enhanced staining of the markers with differentiation (middle row) compared to the
nontreated control (top row). Scale bar ¼ 50 lm. (D–F): Induction of USCs to neuronal lineage. (D): Cell morphology changed significantly, with cell
extensions present after cells were cultured in neuronal induction medium (arrows). Scale bar ¼ 50 lm. (E): Real-time analysis of neuronal transcript
performed on induced USCs for 3 days. A several fold increase in nestin and NF200 transcripts was observed upon induction. (F): Immunofluorescent
staining using lineage-specific neural antibodies (nestin, S-100, NF200, and GFAP) were used for confirming neuronal induction of USCs. Specific
staining is seen in green and nuclei in red (PI). Scale bar ¼ 50 lm. (G–I): Skeletal myogenic induction of USCs. (G): Cells cultured with skeletal-mus-
cle induction medium caused the cells to change to a spindle shape by day 14. Scale bar ¼ 100 lm. (H): Semiquantitative RT-PCR analysis revealed
that cells expressed specific transcripts after induction. (I): Immunofluorescent staining for skeletal muscle markers (myoD and myogenin) showed
increased specific staining (arrows) in induced cells. Scale bar ¼ 50 lm. (J, K): USCs were induced to multiple lineages using specific media cock-
tails. (J): USCs cultured in different induction media were assessed for differentiation to multiple lineages. (i): Adipogenic differentiation-day 28: oil
red O stain for lipid droplets (enlarged image shown in inset 400). (ii–iv): Osteogenic differentiation-day 28: (ii) von Kossa staining for bone miner-
als; (iii) Alizarin Red staining for calcium deposition; (iv) alkaline phosphatase. (v–ix): Chondrogenic differentiation-day 28: (v) toluidine blue stain-
ing; (vi) Alcian blue staining for GAGs in monolayer (vi) and pellet culture (vii). Safranin-O staining of pellet culture (viii) and in monolayer culture
(ix). Scale bar ¼ 100 lm. (K): Semiquantitative RT-PCR analysis for lineage-specific transcripts: (i) adipogenic; (ii) osteogenic, and (iii) chondro-
genic differentiation. (L): Scoring for multipotential capability of independent USC clones. þþþþ, 80%–100% differentiated; þþ, 60%–80% differ-
entiated; þþ, 40%–60% differentiated; þ, 20%–40% differentiated; 6, 20% or less differentiated; , No differentiation/change in morphology.
Abbreviations: BMSC, bone marrow mesenchymal stromal cells; C/EBP, CCAAT/enhancer-binding protein; FABP, fatty acid binding protein; FLT-
1, fms-like tyrosine kinase-1; eNOS, endothelial nitric oxide synthase; GFAP, glial fibrillary acidic protein; GAPDH, glyceraldehyde 3-phosphate de-
hydrogenase (housekeeping gene); HUVEC, human umbilical vein endothelial cell; KDR, kinase insert domain receptor; LpL, lipoprotein lipase; NF,
neurofilament; PPAR, peroxisome proliferator-activated receptor; runx2, runt-related transcription factor-2; sox-9-SRY, sex determining region Y-
box 9; SkM, skeleton muscle; USC, urine derived stem cell; vWF, von willebrand factor; VE, vascular endothelial.
1850 Multipotential of Human Urine Derived Stem Cells

Figure 5. (Continued)

Characterization of Differentiated USCs In Vivo subjected to Masson’s trichrome (Fig. 7C-iv, top), Alcian blue
Myogenic differentiated USCs formed multiple layers of (Fig. 7C-iv, middle), and oil-red O staining (Fig. 7C-iv, bot-
SMCs beneath UC layers in vivo, and the SMCs stained posi- tom). Trichrome staining revealed that BMPs induced USCs
tively for a-SMA, desmin, and myosin. Scaffolds containing to form a highly mature and mineralized osteoid matrix. For-
urothelial differentiated USCs generated stratified layers in mation of cartilage-like matrix and chondrocytes was detected
vivo and stained for the uroplakin-Ia and uroplakin-III urothe- by Alcian blue staining. This staining was more pronounced
lial markers (Fig. 7A, 7B). in BMP expressing USCs compared to the non-BMP-infected
USCs expressing BMP2 and BMP9 formed bony masses, USC controls (data not shown).
which were examined using micro CT imaging 4 weeks post-
implantation (Fig. 7C-i). No masses were found in the non-
BMP-infected USC control animals. The isosurface and den-
sity heat maps for the retrieved masses are shown in Figure DISCUSSION
7C-ii. Masses were excised from the animals, decalcified, em-
bedded in paraffin, and sectioned for hematoxylin and eosin A heterogeneous cell population—including differentiated,
staining (Fig. 7C-iii). Sections of these bony masses were also differentiating, progenitor, and stem cells—exists in human
Bharadwaj, Liu, Shi et al. 1851

Figure 5. (Continued)

voided urine. However, of these, only USCs tend to attach to activity and telomere maintenance are typical features of
culture plastic and continuously expand in culture [9]. These embryonic stem cells, germ cells, some adult stem cells
possess MSC-like features such as clonogenicity, self-renewal, such as hair follicle stem cells and intestinal stem cells.
and multipotent differentiation capacity. In this study, we dem- Telomerase activity is associated with immortality in cancer
onstrated that these cells express a whole panel of pericyte/ cells and correlates with longer lifespan and extended repli-
MSC surface markers, possess telomerase activity, and have cative capacity in cell lines [38]. In this study, telomerase
long telomeres. Importantly, USCs from one clone can simulta- was detected in 9 of 15 independent USC samples. To mea-
neously differentiate into functional SMCs (mesoderm) and sure telomere length of USCs expressing telomerase activity,
UCs (endoderm) in a time-dependent fashion and given the a single USC clone at initial culture generated a large
presence of an optimal amount of growth factors. Additionally, amount of cells with telomerase activity expression; even
USCs can give rise to osteogenic, chondrogenic, adipogenic, these cells reach passages 7–8. The USCs with the highest
myogenic, endothelial (mesoderm), and neurogenic (ectoderm) telomerase activity maintained telomere length. In addition,
lineages in vitro. BMP-2 and -9 transduced USCs can form the USCs retained a complete diploid set of chromosomes
bone, cartilage, and fat tissue in vivo. Together with our previ- with normal karyotype after several passages. Importantly,
ous studies [5–9] describing urine-derived progenitor cells, our USCs obtained from six individuals did not form any terato-
data indicate the existence of a stem cell population in urine. mas in vivo after being subcutaneously implanted in nude
The ability to undergo numerous cycles of cell division mice [5-8, 39]. Compared to BMSCs, which usually stop to
while maintaining an undifferentiated state is associated with grow before passage 10 and reach a PD rate of 30 [40], a
the expression of telomerase in stem cells [37]. Telomerase single USC at initial culture can proliferate for up to 20
www.StemCells.com
1852 Multipotential of Human Urine Derived Stem Cells

Figure 6. Determination of USC source. Several clones of USCs (p3) were cultured and analyzed for expression of kidney-lineage markers.
(A): Fluorescence in situ hybridization (left) and amelogenin gene polymerase chain reaction (PCR) analysis (right) analysis of USCs isolated
from urine obtained from a male donor-to-female recipient kidney transplant for presence of Y-chromosome. (B): Real-time PCR analysis using
specific primers (pax8, NR3C2, and L1-CAM) also showed transcript levels similar to those of the kidney cortex tissue and cultured kidney cells.
(C): Immunofluorescent staining using kidney-specific antibodies (CD224, CD13, NR3C2, and pax8) showed specific staining versus the primary
kidney cortex cells. Scale bar ¼ 50 lm. (D): Histology of human kidney cortex. Immunofluorescent staining of podocytes in glomeruli and ves-
sels showed cells positive for CD146 (green). Nuclei were counter-stained with PI (red). Hematoxylin and eosin staining of a serial section
included as a control. Scale bar ¼ 50 lm. Abbreviations: A4, USCs from male donor-to-female recipient urine sample; F, female control; L,
DNA ladder; M, male control; N, negative control; USC, urine derived stem cell.
Bharadwaj, Liu, Shi et al. 1853

Figure 7. In vivo studies. (A): Myogenic differentiation—nontreated and myogenic-differentiated USCs (p3) were analyzed after 4 weeks in
nude mice for smooth muscle-specific markers (desmin, aSMA, and myosin). Lineage-specific staining is seen in green, human nuclei staining in
red, and nuclear staining in blue (DAPI). (B): Urothelial differentiation—nontreated and urothelial-differentiated USCs (p3) were analyzed after
4 weeks in nude mice for urothelial cell markers (Up-Ia and III). Lineage-specific staining is seen in green, human cells nucleus in red, and all
nucleus (mice and human) stained in blue (DAPI). (C): USCs were infected with bone morphogenetic protein 9 (BMP9) or control green fluores-
cent protein and were injected subcutaneously into nude mice. (i): Bony masses were only observed in mice implanted with BMP-transduced
USCs at week 4. (ii): The harvested bony masses were subjected to microCT imaging revealing the isosurface (left) and density heat maps
(right). (iii): The retrieved masses were also sectioned and stained with hematoxylin and eosin. (iv): Mesodermal lineage differentiation of USCs
in vivo. The retrieved bony masses were stained with Masson’s trichrome (top panel), Alcian blue (middle panel), and oil red O (bottom panel).
Magnification, 400. AC, CM, and OM are indicated by arrows. Abbreviations: ASMA, alpha-smooth muscle actin; AC, adipocytes; CM, chon-
droid matrix; DAPI, 40 ,6-diamidino-2-phenylindole; OM, osteoid matrix; USC, urine derived stem cell.

passages and reach a PD rate of 60–70. This indicates that line of stem cells with biological plasticity. Our study demon-
USCs possess the capacity to generate a sufficient quantity strated that one single USC possesses multipotent differentia-
of cells for potentially use in cell-based therapies. However, tion capability; one USC clone with telomerase activity differ-
further study is needed to determine whether there is a entiated into endodermal, mesodermal, and ectodermal
difference in numbers of USCs expressing telomerase in lineages. One challenge in urological tissue regeneration is
24-hour urine among different age groups. generating urothelial cells from MSC-derived cells. Only 5%
USC clones can be a unique tool to assess whether a sin- of BMSCs can give rise to the cells expressing urothelial
gle adult stem cell is able to generate a genetically identical markers [41]. In contrast, using the same inductive medium
www.StemCells.com
1854 Multipotential of Human Urine Derived Stem Cells

Figure 7. (Continued)

as in the BMSC study [6], 60%–70% of the USCs in this Continuing from the findings of previous studies [9], this
study could not only differentiate into cells expressing uroepi- study showed that USCs can efficiently give rise to functional
thelial cell-specific genes (uroplakin-Ia/III) and protein SMC and urothelial lineage cells. Smooth muscle differenti-
markers but also possessed urothelial barrier function and ated USCs expressed smooth muscle markers at both the gene
tight junction ultrastructures. In addition, urothelial differenti- and protein levels. These markers include: a-SM actin and
ated USCs expressed the genes and proteins ZO-1, E-cad- calponin (early differentiation markers, but not SMC-specific
herin, and cingulin that are associated with tight junctions in markers); desmin and myosin (only displayed in contractile
a dose-dependent and time-dependent manner. Although bar- SMCs); and smoothelin (SMC-specific marker). The mRNA
rier function of the induced USCs did not reach the mature and protein levels of the induced markers increased
function of UCs isolated from bladder tissue, it was signifi- significantly with time in differentiation media. Functional
cantly higher than that observed in noninduced USCs, indicat- studies demonstrated that these SMCs have contractile proper-
ing that USCs possessed stem cell plasticity. ties in vitro.
Bharadwaj, Liu, Shi et al. 1855

USCs can differentiate toward mesodermal cell lineages podocin at a rate similar to that expressed in parietal cells and
using two methods: one is through the use of a commercial podocytes that populate glomeruli, while bladder and ureter
inductive media that includes growth factors, and the second UCs and SMCs did not, indicating that USCs were most
is via gene transfection with BMPs. Several signaling path- likely from translational cells at the parietal cell–podocyte
ways, including those activated by the BMPs, regulate lineage interface in the renal glomerulus. These data suggest that
commitment and terminal differentiation of MSCs. Previously, urine derived from the kidney could be used as a source of
we conducted a comprehensive analysis of 14 types of BMPs healthy cells for tissue-engineered therapeutic strategies for
and compared their abilities to regulate multilineage-specific patients with chronic bladder diseases or from whom adequate
differentiation of MSCs[13, 16, 42]. Most BMPs exhibited cells cannot be obtained by biopsy.
distinct abilities to regulate the expression of Runx2, Sox9,
MyoD, and PPARc2 [15]. Further analysis indicated that
BMP-2, BMP-4, BMP-6, BMP-7, and BMP-9 effectively CONCLUSION
induced both adipogenic and osteogenic differentiation in
vitro and in vivo [15]. In an ongoing study, we are assessing
the relationship between differentiation capacity and expres- In conclusion, USCs can be accessed via a simple, non-inva-
sion of telomerase in USCs. sive, and low-cost approach that avoid surgical procedures. A
Identification of the origin of USCs will lead to a better single cell can expand into many additional cells without
understanding of the biological role of this multipotent MSC feeders. USCs express pericyte/MSC markers, display telo-
population within the urinary tract system. Several types of merase activity with long telomeres, and have the capacity for
MSCs could potentially be shed from the kidney or the multipotent differentiation. A single cell clone isolated from
urothelial mucosa into the urine. These include BMSCs, urine can give rise to the three germinal lineages. Prospec-
urothelial basal cells, renal tubule epithelial cells, parietal tively, USCs may represent one of the most promising cell
cells, and pericytes that originate from capillaries of glomeruli sources for cell therapies in treatment of renal insufficiency,
in kidney. Our serial studies involving morphology and/or urinary incontinence, vesicoureteral reflux, and erectile dys-
growth patterns provided no evidence to suggest that USCs function, and for tissue engineering in bladder and urethral
are actually BMSCs. BSMCs are spindle-shaped and required tissue regeneration.
culture medium containing 10% serum, whereas USCs do not
proliferate well in this media and require 2%–5% serum. The
maximum number of PDs for BMSCs is about 30, while for ACKNOWLEDGMENTS
USCs, it can reach up to 60–70. USCs are also not urothelial
basal cells, because USCs cannot grow in serum-free medium
and initially do not express UC markers [5]. USCs isolated We thank Ms. Karen Klein (Office of Research, Wake Forest
from urine sourced from the upper urinary tract are similar to School of Medicine) for her editorial assistance with this
voided USCs in morphology, cell phenotypes, growth pattern, manuscript.
and bipotent differentiation capacity [5]. Importantly, this
study presents strong evidence that the voided USCs originate
from kidney, because cells obtained from female patients who DISCLOSURE OF POTENTIAL
received a sex-mismatched kidney transplant contained the Y-
chromosome and expressed normal kidney cell gene and pro- CONFLICTS OF INTEREST
tein markers (CD224, CD13, NR3C2, pax2, and pax8 [43]).
They also expressed the marker CD146, synaptopodin, and The authors indicate no potential conflicts of interest.

11 Raya-Rivera A, Esquiliano DR, Yoo JJ et al. Tissue-engineered auto-


REFERENCES logous urethras for patients who need reconstruction: An observational
study. Lancet 2011;377:1175–1182.
12 Chen KL, Eberli D, Yoo JJ et al. Bioengineered corporal tissue for
1 Smith JA, Daniel R. Stem cells and aging: A chicken-or-the-egg structural and functional restoration of the penis. Proc Natl Acad Sci
issue? Aging Dis 2012;3:260–268. USA 2010;107:3346–3350.
2 Baddour JA, Sousounis K, Tsonis PA. Organ repair and regeneration: 13 Cheng H, Jiang W, Phillips FM et al. Osteogenic activity of the four-
An overview. Birth Defects Research C Embryo Today 2012;96:1–29. teen types of human bone morphogenetic proteins (BMPs). J Bone
3 Li W, Jiang K, Ding S. Concise review: A chemical approach to con- Joint Surg Am 2003;85–A:1544–1552.
trol cell fate and function. Stem Cells 2012;30:61–68. 14 He TC, Zhou S, da Costa LT et al. A simplified system for generating
4 Brack AS, Rando TA. Tissue-specific stem cells: Lessons from the recombinant adenoviruses. Proc Natl Acad Sci USA 1998;95:
skeletal muscle satellite cell. Cell Stem Cell 2012;10:504–514. 2509–2514.
5 Bharadwaj S, Liu G, Shi Y et al. Characterization of urine-derived 15 Kang Q, Song WX, Luo Q et al. A comprehensive analysis of the
stem cells obtained from upper urinary tract for use in cell-based uro- dual roles of BMPs in regulating adipogenic and osteogenic differen-
logical tissue engineering. Tissue Eng Part A 2011;17:2123–2132. tiation of mesenchymal progenitor cells. Stem Cells Dev 2009;18:
6 Bodin A, Bharadwaj S, Wu S et al. Tissue-engineered conduit using 545–559.
urine-derived stem cells seeded bacterial cellulose polymer in urinary 16 Kang Q, Sun MH, Cheng H et al. Characterization of the distinct
reconstruction and diversion. Biomaterials 2010;31:8889–8901. orthotopic bone-forming activity of 14 BMPs using recombinant ade-
7 Wu S, Liu Y, Bharadwaj S et al. Human urine-derived stem cells novirus-mediated gene delivery. Gene Ther 2004;11:1312–1320.
seeded in a modified 3D porous small intestinal submucosa scaffold 17 Luo J, Deng ZL, Luo X et al. A protocol for rapid generation of
for urethral tissue engineering. Biomaterials 2011;32:1317–1326. recombinant adenoviruses using the AdEasy system. Nat Protoc 2007;
8 Wu S, Wang Z, Bharadwaj S et al. Implantation of autologous urine 2:1236–1247.
derived stem cells expressing vascular endothelial growth factor for 18 He TC, Chan TA, Vogelstein B et al. PPARdelta is an APC-regu-
potential use in genitourinary reconstruction. J Urol 2011;186:640–647. lated target of nonsteroidal anti-inflammatory drugs. Cell 1999;99:
9 Zhang Y, McNeill E, Tian H et al. Urine derived cells are a potential 335–345.
source for urological tissue reconstruction. J Urol 2008;180: 19 He TC, Sparks AB, Rago C et al. Identification of c-MYC as a target
2226–2233. of the APC pathway. Science 1998;281:1509–1512.
10 Atala A, Bauer SB, Soker S et al. Tissue-engineered autologous blad- 20 Peng Y, Kang Q, Luo Q et al. Inhibitor of DNA binding/differentia-
ders for patients needing cystoplasty. Lancet 2006;367:1241–1246. tion helix-loop-helix proteins mediate bone morphogenetic protein-

www.StemCells.com
1856 Multipotential of Human Urine Derived Stem Cells

induced osteoblast differentiation of mesenchymal stem cells. J Biol 31 Zhang W, Deng ZL, Chen L et al. Retinoic acids potentiate BMP9-
Chem 2004;279:32941–32949. induced osteogenic differentiation of mesenchymal progenitor cells.
21 Luo Q, Kang Q, Si W et al. Connective tissue growth factor (CTGF) Plos One 2010;5:e11917.
is regulated by Wnt and bone morphogenetic proteins signaling in 32 Luo J, Tang M, Huang J et al. TGFbeta/BMP type I receptors ALK1
osteoblast differentiation of mesenchymal stem cells. J Biol Chem and ALK2 are essential for BMP9-induced osteogenic signaling in
2004;279:55958–55968. mesenchymal stem cells. J Biol Chem 2010;285:29588–29598.
22 Si W, Kang Q, LuuHH et al. CCN1/Cyr61 is regulated by the canoni- 33 Chen L, Jiang W, Huang J et al. Insulin-like growth factor 2 (IGF-2)
cal Wnt signal and plays an important role in Wnt3A-induced osteo- potentiates BMP-9-induced osteogenic differentiation and bone forma-
blast differentiation of mesenchymal stem cells. Mol Cell Biol 2006; tion. J Bone Miner Res 2010;25:2447–2459.
26:2955–2964. 34 Trask BJ. Fluorescence in situ hybridization: Applications in cytoge-
23 Sharff KA, Song WX, LuoX et al. Hey1 basic helix-loop-helix protein netics and gene mapping. Trends Genet 1991;7:149–154.
plays an important role in mediating BMP9-induced osteogenic differen- 35 Bryan TM, Englezou A, Dunham MA et al. Telomere length dynam-
tiation of mesenchymal progenitor cells. J Biol Chem 2009;284:649–659. ics in telomerase-positive immortal human cell populations. Exp Cell
24 Tang N, Song WX, Luo J et al. BMP9-induced osteogenic differentiation Res 1998;239:370–378.
of mesenchymal progenitors requires functional canonical Wnt/beta-cat- 36 Pryor JP, Hendry WF. Ejaculatory duct obstruction in subfertile males:
enin signaling. J Cell Mol Med 2009;13:2448–2464. Analysis of 87 patients. Fertil Steril 1991;56:725–730.
25 Oswald J, Boxberger S, Jorgensen B et al. Mesenchymal stem cells 37 Yu J, Vodyanik MA, Smuga-Otto K et al. Induced pluripotent stem cell
can be differentiated into endothelial cells in vitro. Stem Cells 2004; lines derived from human somatic cells. Science 2007;318:1917–1920.
22:377–384. 38 Kim NW, Piatyszek MA, Prowse KR et al. Specific association of
26 Kropp BP, Zhang Y, Tomasek JJ et al. Characterization of cultured human telomerase activity with immortal cells and cancer. Science
bladder smooth muscle cells: Assessment of in vitro contractility. J 1994;266:2011–2015.
Urol 1999;162:1779–1784. 39 Zhang Y. Bladder reconstruction by tissue engineering—With or with-
27 Cross WR, Eardley I, Leese HJ et al. A biomimetic tissue from cul- out cells? J Urol 2008;180:10–11.
tured normal human urothelial cells: Analysis of physiological func- 40 Vidal MA, Walker NJ, Napoli E et al. Evaluation of senescence in
tion. Am J Physiol Renal Physiol 2005;289:F459–F468. mesenchymal stem cells isolated from equine bone marrow, adipose
28 Liu Y, Bharadwaj S, Lee SJ et al. Optimization of a natural collagen tissue, and umbilical cord tissue. Stem Cells Dev 2012;21:273–283.
scaffold to aid cell-matrix penetration for urologic tissue engineering. 41 Tian H, Bharadwaj S, Liu Y et al. Differentiation of human bone mar-
Biomaterials 2009;30:3865–3873. row mesenchymal stem cells into bladder cells: Potential for urologi-
29 Zhang Y, Kropp BP, Moore P et al. Coculture of bladder urothelial cal tissue engineering. Tissue Eng Part A 2010;16:1769–1779.
and smooth muscle cells on small intestinal submucosa: Potential 42 Luu HH, Song WX, Luo X et al. Distinct roles of bone morphogenetic
applications for tissue engineering technology. J Urol 2000;164: proteins in osteogenic differentiation of mesenchymal stem cells. J
928–934; discussion 934–925. Orthop Res 2007;25:665–677.
30 Luo X, Chen J, Song WX et al. Osteogenic BMPs promote tumor 43 Narlis M, Grote D, GaitanY et al. Pax2 and pax8 regulate branching
growth of human osteosarcomas that harbor differentiation defects. morphogenesis and nephron differentiation in the developing kidney. J
Lab Invest 2008;88:1264–1277. Am Soc Nephrol 2007;18:1121–1129.

See www.StemCells.com for supporting information available online.

Das könnte Ihnen auch gefallen