Download as pdf or txt
Download as pdf or txt
You are on page 1of 5

190 Current Stem Cell Research & Therapy, 2010, 5, 190-194

Adipose Tissue Derived Stem Cells for Regeneration and Differentiation into Insulin-
Producing Cells

Song Cheol Kim*,1,2, Duck Jong Han1 and Ji Yeon Lee2

1
Department of Surgery, University of Ulsan College of Medicine & Asan Medical Center, Korea; 2Laboratory of Cell Therapy, Asan
Institute for Life Science, Seoul, Korea

Abstract: Stem cells are considered an ideal tool for the supply of insulin-producing cells or repairing damaged pancreatic tissues to treat
diabetes mellitus, with the possibility of unlimited sources. This cell population includes embryonic, adult bone marrow, pancreatic stem
cells, extra pancreatic (such as hepatic cells) and adipose-derived stem cells. Multipotent adipose tissue-derived stem cells (ADSCs) are
abundant in the human body, and thus are an ideal donor source for autologous transplantation to generate insulin-producing cells.
Moreover these cells are better sources than bone marrow stem cells (BMSCs) for clinical applications, owing to minimal invasive pro-
cedures, high proliferation and multi-differentiation potential. Human adipose tissue-derived stem cells (hADSCs) may thus provide an
alternative stem cell source, replacing BM-MSCs or embryonic stem cells (ESCs) for future clinical use in diabetes mellitus treatment.

Keywords: Stem cell, adipose tissue derived stem cells, insulin-producing cells, diabetes, regeneration, cell therapy.

INTRODUCTION and transdifferentiate into pancreatic beta-cells in vivo [10-14].


Curative treatment of insulin-deficient diabetes mellitus with However, the in vivo transdifferentiation of BM-MSCs remains a
minute-to-minute control can be achieved by: i) restoring the en- controversial issue [15-17]. The highly invasive procedure, limited
dogenous insulin source and ii) transplanting an insulin-producing quantity of bone marrow obtainable from a single donor and low
pancreatic or non pancreatic source of cells. To accomplish this cell numbers are significant obstacles in the production of sufficient
goal, several clinical trials investigating stem cell potential in hu- BM-MSCs for clinical applications. Stem cells in adult tissues, such
man or non-human subjects, such as regeneration of residual islet as liver [18], pancreas duct [19], umbilical cord blood [20] and
cells in the diseased pancreas or reprogramming of the regenerative placenta [21], differentiate into pancreatic beta-cells. A recent re-
power by precursor or stem cells [1], and islet-cell transplantation port indicates that human adipose tissue contains multipotent stem
[2] are underway. Among these strategies, transplantation of islets cells that have the potential to differentiate into adipogenic, chon-
from cadaveric donors is a promising cell-based therapy for diabe- drogenic, myogenic, and osteogenic lineages [22]. Pancreatic dif-
tes mellitus. However, limited availability of donor cells and im- ferentiation from human adipose tissue-derived stem cells (hASCs)
mune suppression are major obstacles in islet transplantation. has additionally been reported by Timper et al. [23] and Lee et al.
Therefore, considerable effort has been devoted to identifying alter- [24]. Here, we discuss the various stem cell types that are able to
native stem cell sources for generating transplantable pancreatic differentiate into insulin-producing cells.
beta-cells. Here, we review the use of stem cells for pancreatic re-
1. Embryonic Stem Cells
generation and differentiation into insulin-producing cells, and
adipose-derived stem cell strategies for clinical use in the genera- Embryonic stem cells can differentiate into almost all cell types.
tion of insulin-producing cells. The application of these pluripotent cells in the field of regenerative
medicine is currently under investigation [25]. Murine and human
STEM CELL STRATEGY FOR PANCREATIC REGEN- ESCs generate embryoid bodies that contain cells with a beta-cell-
ERATION AND DIFFERENTIATION INTO INSULIN- like phenotype in vitro, and differentiated ESCs lower blood glu-
PRODUCING CELLS cose levels in rodents [26-27]. Segev and colleagues [4] reported
Stem cells are an ideal tool for supplying cells producing spe- enhanced expression of beta-cell-specific genes in ESCs after ma-
cific hormones or proteins or repairing damaged tissues. A major nipulation of culture medium, while Soria et al. [28] observed that
advantage of the stem cell approach is the supply of an unlimited insulin expression also occurs spontaneously during culture of un-
number of cells that have the potential to become a functional or- differentiated ES cells.
gan. Adult stem cells have the advantage of differentiating into Lumelsky and co-workers [29] initially reported that a nestin-
different tissue-specific cell types, along with no significant ethical positive pancreatic islet cell-like structure can be selected, ex-
concerns. Different sources of stem cells are proposed for the pro- panded, and differentiated from ES cells. These cells displayed 50-
duction of beta cells essential to control hyperglycemia in the hu- fold lower intracellular insulin content than normal pancreatic islet
man body. Embryonic stem cells are pluripotent, and differentiate cells, but were not able to reverse the diabetic status of streptozoto-
into almost all cell types. A number of studies have demonstrated cin-induced diabetic mice. As an alternative trial to enhance the
insulin-producing pancreatic differentiation of ESCs [3-9]. How- phenotype of insulin-producing cells, treatment with enhancers
ever, the ethical issues and risk of teratoma formation remain to be (such as inhibitors of phosphoinositide 3 kinase) and overexpres-
resolved. MSCs from bone marrow are the most extensively inves- sion of transcription factors, such as PAX4, PDX1, and NKX6.1,
tigated adult stem cells to date, and several research groups report was investigated, but with minimal success. While the potential
that BM-MSCs differentiate into insulin-producing cells in vitro application of ESCs for producing beta cells has brought idealism
into the field of diabetic research, a significant drawback requires
some considerations. It is suggested that beta cells derived from
ESCs represent aberrant products from uncontrolled differentiation
*Address correspondence to this author at the Department of Surgery, Uni- programs. An earlier study shows that animals develop tumors
versity of Ulsan College of Medicine and Asan Medical Center, 388-1 when transplanted with ESC-derived insulin-producing cells [30].
Pungnap-dong, Songpa-gu, Seoul 138-736, Korea; Tel: 82-2-3010-3936; Thus, it is crucial to assess the efficacy of ESCs in detail before
Fax: 82-2-474-9027; E-mail: drksc@amc.seoul.kr

1574-888X/10 $55.00+.00 © 2010 Bentham Science Publishers Ltd.


Adipose Tissue-Derived Stem Cells Generate Insulin-Producing Cells Current Stem Cell Research & Therapy, 2010, Vol. 5, No. 2 191

recommending these cells as a suitable source of insulin-producing Nestin-positive cells represent a well-known endocrine-derived
cells in diabetes treatment. stem cell type. Nestin is detected in various adult CNS lineages.
These cells have been identified in murine and human pancreas
2. BM-Derived Stem Cells [39]. Nestin-positive cells represent a population of islet-derived
Bone marrow-derived stem cells (BMSCs) are multipotent, ca- stem cells, and these progenitor cells are able to differentiate in
pable of self-renewal, and a well known source of stem cells for vitro into cells with liver, pancreatic exocrine and/or ductal and
blood cells. These cells have the ability to generate other cells or endocrine phenotypes. After differentiation, cells produce insulin,
tissue types, due to their ability to differentiate into all embryonic glucagon, glucagon-like peptide-1 (GLP-1), and the transcription
lineages [31], migrate towards the site of damage, and differentiate factor PDX1 [40]. However, nestin is regarded as a functional pro-
under the influence of factors from the microenvironment [32]. tein expressed in beta cell precursors due to heterogeneous expres-
Ianus and colleagues showed that adult bone marrow stem cells sion in pancreas and other tissues. The issue of whether nestin-
differentiate into insulin-producing cells after engrafting to recipi- positive cells function as b-cell precursors remains a subject of
ent pancreas parenchyma, express beta cell markers, and produce controversy. Other possible islet-related cells include “small cells”
insulin after the glucose challenge test. They showed that this phe- within the pancreatic islet expressing four types of endocrine hor-
nomenon was the result of donor cell differentiation with the CRE- mones, specifically, PDX-1, synaptophysin, Bcl-2, and insulin [41].
LoxP cell tracking system [33]. BMSCs cultured in vitro can pro- Guz and colleagues showed that the islet contains these cells, and
duce de novo insulin and express insulin upon challenge with high that islets in streptozotocin-damaged pancreas contain cells express-
glucose [10]. Overall, current research suggests that BMSCs do not ing insulin and somatostatin, thereby validating a role of this cell
differentiate into beta cells in vivo. However, BMSCs support pan- population in beta cell differentiation [42].
creatic growth in vivo, and can be manipulated in vitro to differenti-
ate into beta cells. 5. Other Sources of Non-Pancreatic Organ-Derived Stem Cells
The main advantage of extrapancreatic sources is that these tis-
3. Human Umbilical Cord Blood Cells sues (such as adipose tissue) can be obtained easily. Extrapancreatic
Human umbilical cord blood (UCB) cells can differentiate into organ-derived stem cells, including those from hepatocytes [43],
insulin-producing cells with the advantages of generating sufficient human adipose-tissue derived mesenchymal stem cells [23,24], and
autologous stem cells to overcome the immune reaction, as shown splenocytes [44], can differentiate into islet cells. Kojima et al. [45]
by a number of researchers. Yoshida et al. [34] intravenously trans- reported islet neogenesis in the portal triad area by delivery of a
planted human CB–derived cells into non-obese diabetic/severe combination of NeuroD and betacellulin. Delivery of the transcrip-
combined immunodeficient/2-microglobulin null mice. After tion factor, PDX1, or a combination of BETA2 and betacellulin to
transplantation of T cell-depleted mononuclear cells, human CB- the medium [45, 46] can be successfully applied to promote differ-
derived cells generated insulin-producing cells at a frequency of entiation into a beta cell phenotype.
0.65 ± 0.64% in xenogeneic hosts. Kang et al. [20] isolated a popu-
lation of stem cells from human cord blood (UCB) expressing the 6. Adipose Tissue-Derived Stem Cells Generate Insulin-
embryonic stage-specific maker, SSEA-4, and the multipotential Producing Cells
stem cell marker, Oct4. These cells were induced to differentiate Human adipose tissue is available in large quantities as a waste
into insulin-producing islet-like structures co-expressing insulin and product of cosmetic liposuction, which involves a less invasive
C-peptide. Chao and colleagues [35] transplanted human umbilical procedure than obtaining human bone marrow. Human adipose
cord mesenchymal cells into the livers of streptozotocin-induced tissue contains multipotent stem cells that have the potential to
diabetic rats. These islet-like cell clusters contain the human C- differentiate into adipogenic, chondrogenic, osteogenic, myogenic,
peptide and release human insulin in response to glucose. Real-time neurogenic [22], endothelial [47], hepatic [43], and pancreatic line-
RT-PCR was employed to detect the expression of insulin and other ages [23]. Lack of HLA-DR expression and immunosuppressive
pancreatic beta cell-related genes (Pdx1, Hlxb9, Nkx2.2, Nkx6.1, properties of hASCs have been reported by Puissant et al. [48].
and Glut-2). Hyperglycemia in streptozotocin-induced diabetic rats Fang and colleagues published preliminary data showing that se-
was significantly alleviated after xenotransplantation of islet-like vere refractory acute graft-versus-host disease (GVHD) could be
cell clusters without immunosuppressants. treated with human adipose tissue-derived mesenchymal cells from
HLA-mismatched donors [49]. Zhu and co-workers further pro-
4. Pancreatic Stem Cells posed that ADSCs are a better cell source than BMSCs [50]. They
Specific populations of cells located in the adult pancreas are investigated the proliferation and multi-differentiation potential of
capable of differentiating into islet cells after partial pancreatec- ADSCs via cell phenotype analysis. ADSCs can be continuously
tomy., Islet cell production from these stem cells may be a useful cultured in vitro for up to 1 month without passage, and undergo
tool to correct islet cell deficiency in the clinical setting [36]. Pan- several logarithmic growth phases during the culture period.
creatic stem cells constitute different populations, including those Moreover, ADSCs express high levels of stem cell-related antigens,
of endocrine origin (islet-derived population) as well as non endo- but not hematopoiesis-related antigens or the human leukocyte
crine-derived stem cells (ductal epithelial and acinar cells). Non- antigen, HLA-DR. The stem cell-related transcription factors,
endocrine stem cells are mainly pancreatic ductal epithelial cells Nanog, Oct-4, Sox-2, and Rex-1, are positively expressed in
expressing the ductal markers, CK-19 and PDX1, which possess the ADSCs.
ability to expand and differentiate into endocrine cells. Early pan- Only four reports on the possibility of ADSC conversion to in-
creatic development from the endoderm bud progresses to cy- sulin-producing cells are currently documented in the English lit-
tokeratin-expressing ductal structures, followed by eventual loss of erature, including our study. An earlier investigation by Timper et
cytokeratin and expression of the endocrine or exocrine phenotype al. [23] showed that human adipose tissue-derived MSCs from four
[37]. Recently Hao and colleagues reported beta cell differentiation healthy donors differentiated into insulin, somatostatin, and gluca-
from non endocrine cells procured after human islet isolation to gon-expressing cells. During the proliferation period, cells ex-
adult pancreatic stem cells. They cultured the mixture fractions with pressed the stem cell markers nestin, ABCG2, SCF and Thy-1, as
G 418 to eliminate mesenchymal cells, leaving highly purified well as the pancreatic endocrine transcription factor, Isl-1. Cells
nonendocrine pancreatic epithelial cells (NEPECs). NEPECs were were induced to differentiate into a pancreatic endocrine phenotype
capable of endocrine differentiation upon cotransplantation with within 3 days by culturing in defined conditions. Quantitative PCR
fetal pancreatic cells [38]. revealed downregulation of ABCG2 and upregulation of the pan-
192 Current Stem Cell Research & Therapy, 2010, Vol. 5, No. 2 Kim et al.

creatic developmental transcription factors, Isl-1, Ipf-1, and Ngn3, epididymal fat pads of Swiss albino mice. They used a 10-day dif-
together with induction of the islet hormones, insulin, glucagon, ferentiation protocol involving progressive changes in the differen-
and somatostatin. tiation cocktail on days 1, 3 and 5. These cell populations contained
We subcultured ADSCs every 14 days instead of the normal 5 pancreatic endoderm-expressing and hormone-expressing cells.
days [24]. ADSCs maintained strong proliferation ability, pheno- Calcium-alginate encapsulated day10 ICAs, when transplanted
type, and displayed stronger multi-differentiation potential after 25 intraperitoneally into STZ-induced diabetic mice, restored normo-
passages. Pancreatic regeneration is reported in partially pancre- glycemia within 2 weeks.
atectomized rats, and treatment of the extract from the remnant During the preparation of this review, an interesting article was
pancreas after partial pancreatectomy initiates islet neogenesis and published online by Lin et al. [55]. They reported the derivation of
normoglycemia in streptozotocin (STZ)-induced diabetic mice [51]. insulin-producing cells from human or rat ADSCs by transduction
To determine whether hADSCs differentiate into pancreatic linea- with the pancreatic duodenal homeobox 1 (Pdx1) gene. RT-PCR
ges by regenerating the pancreatic extract (RPE), hADSCs were analyses revealed that native ADSCs expressed insulin, glucagon,
treated with RPE. After differentiation, increased expression levels and NeuroD genes, which were upregulated following Pdx1-
of transcription factors (FoxA2, Hlxb9, NeuroD, and Nkx2.2) re- transduction. ELISA analyses showed that transduced cells secreted
lated to beta cell development were observed in cultures. The levels higher amounts of insulin in response to increasing concentrations
of Sox17, a definitive endoderm marker, and IPF-1, which is not of glucose. Transplantation of these cells under the renal capsule of
only expressed in pancreatic beta-cells but also involved in early streptozotocin-induced diabetic rats resulted in lower blood glu-
endocrine development, were examined in differentiated cultures. cose, higher glucose tolerance, smoother fur, and less cataracts.
Somatostatin was expressed in both control and RPE-treated cul- Moreover, histological examination confirmed that the transplanted
tures. Undifferentiated cells expressed Pax6 and Isl-1, and levels of cells formed tissue-like structures and expressed insulin. These
these genes were slightly increased in RPE-treated cultures. These results support the suitability of ADSCs for the treatment of diabe-
genes are not only expressed in early pancreatic cells, but are also tes mellitus.
active in the developing and mature central nervous system. This
finding supports the proposal that hADSCs have the potential to CONCLUSIONS
differentiate into neuronal lineages. To confirm that insulin mRNA Stem cell approaches capable of supplying an unlimited number
in differentiated cells was newly transcribed, we performed immu- of cells that have the potential to become a functional organ are
nocytochemistry analysis for the C-peptide. Notably, C-peptide- emerging as an effective tool for the curative treatment of insulin-
positive cells were observed in differentiated cultures. However, we deficient diabetes mellitus. Among these stem cell populations,
were unable to confirm detectable levels of C-peptide in differenti- hASCs are a better cell source than BMSCs for clinical applications
ated cultures, possibly due to the insufficient number of differenti- owing to minimal invasive procedures, high proliferation rates, and
ated cells induced. multi-differentiation potential. Human adipose tissue-derived stem
Recently Trivedi and colleagues showed that hADSCs com- cells may thus be used as an alternative source, successfully replac-
bined with human bone marrow hematopoietic cells synthesize ing BM-MSCs or ESCs for future clinical use in diabetes mellitus
insulin [52]. Five insulinopenic DM patients were intraportally therapy
administered xenogeneic-free h-ADMSC (mean dose, 1.5 mL; cell
counts, 2.1 x 103/μL). The CD45-/90+/73+ cells (29.8/16.8%) con- ACKNOWLEDGEMENT
tained 3.08 ng/mL c-peptide and 1578μIU/mL insulin. Aliquots This study was supported by a Grant (07-211) from the Asan
were supplemented with bone marrow hematopoietic cells contain- Institute for Life Sciences, Seoul, Korea.
ing 0.93% CD45-/34+. All patients were successfully infused with
h-ADMSC containing bone marrow hematopoietic cells with no Table 1. Benefits of Using ADSCs for the Generation of Insulin-
side-effects, and showed 30% to 50% decrease in insulin require- Producing Cells
ment and 4- to 26-fold increased serum c-peptide levels, at a mean
follow-up of 2.9 months. 1. Less invasive procedure
Kang and colleagues [53] demonstrated that differentiated 2.Unlimited numbers of cell sources
HEACs (human eyelid adipose cells) directly regulate blood glu- 3.Possibility of high proliferation and multi-differentiation potential
cose levels in diabetic mice by releasing human insulin. Stem cells
from HEACs displayed characteristics of neural crest cells, and 4.Lower possibility of immunogenicity
HEACs were differentiated into insulin-secreting cells using two- 5.Better availability
step culture conditions combined with nicotinamide, activin, and/or
GLP-1. They verified the in vivo effects of differentiated cells via REFERENCES
transplantation experiments. They observed normalization of hy-
[1] Tang DQ, Cao LZ, Burkhardt BR, et al. In vivo and in vitro charac-
perglycemia in 10 of 20 recipients until sacrifice after two months terization of insulin producing cells obtained from murine bone
in streptozotocin-treated immunocompetent diabetic mice without marrow. Diabetes 2004; 53: 1721-32.
immunosuppression. Diminished or null expression of some im- [2] Shapiro AM, Lakey JT, Ryane E, et al. Islet transplantation in
mune-related genes was additionally observed. Interestingly, Kang seven patients with type 1 diabetes mellitus using a glucocorticoid-
and colleagues maintained that HEACs are markedly different from free immunosuppressive regimen. N Engl J Med 2000; 343: 230-8.
MSCs isolated from other adipose tissues. They showed that undif- [3] Assady S, Maor G, Amit M. Itskovitz-Eldor J, Skorecki KL,
ferentiated HEACs spontaneously express several neural cell- Tzukerman M. Insulin production by human embryonic stem cells.
related mRNAs and a variety of neural proteins, including GABA, Diabetes 2001; 50: 1691-7.
GalC, serotonin, tubulin III, and GFAP, most of which are also [4] Segev H, Fishman B, Ziskind A, Shulman M, Itskovitz-Eldor J.
identified in human neural crest and neuroblastoma cells, while Differentiation of human embryonic stem cells into insulin-
MSCs from the trunk adipose tissues do not display most of these producing clusters. Stem Cells 2004; 22: 265-74.
[5] Leon-Quinto T, Jones J, Skoudy A, Burcin M, Soria B. In vitro
neural cell-like characteristics unless cultured under neurogenic
directed differentiation of mouse embryonic stem cells into insulin-
conditions comprising particular chemicals, growth factors, and producing cells. Diabetologia 2004; 47: 1442-51.
neural tissues. [6] Brolen GKC, Heins N, Edsbagge J, Semb H. Signals from the
Chandra et al. [54] successfully generated pancreatic hormone- embryonic mouse pancreas induce differentiation of human embry-
expressing islet-like cell aggregates from ASCs cultured from
Adipose Tissue-Derived Stem Cells Generate Insulin-Producing Cells Current Stem Cell Research & Therapy, 2010, Vol. 5, No. 2 193

onic stem cells into insulin-producing b-cell-like cells, Diabetes embryonic stem cells normalize glycemia in streptozotocin-induced
2005; 54: 2867-74. diabetic mice. Diabetes 2000: 49: 157-62.
[7] Bai L, Meredith G, Tuch BE. Glucagon-like peptide-1 enhances [29] Lumelsky N, Blonde O, Laeng P, Velasc I, Ravin R, McKay R.
production of insulin in insulin-producing cells derived from mouse Differentiation of embryonic stem cells to insulin-secreting struc-
embryonic stem cells. J Endocrinol 2005; 186; 343-52. tures similar to pancreatic islets. Science 2001; 292: 1389-94.
[8] Vaca P, Martin F, Vegara-Meseguer JM, Rovira JM, Berna G, [30] Smith AG. Embryo-derived stem cells: of mice and men. Annu Rev
Soria B. Induction of differentiation of embryonic stem cells into Cell Dev Biol 2001; 17: 435-62.
insulin-secreting cells by fetal soluble factors. Stem Cells 2006; 24: [31] Poulsom R, Alison MR, Forbes SJ, Wright NA. Adult stem cell
258-65. plasticity. J Pathol 2002; 197: 441-56.
[9] D’Amour KA, Bang AG, Eliazer S, et al. Production of pancreatic [32] Lombaert IM, Wierenga PK, Kok T, Kampinga HH, deHaan G,
hormone-expressing endocrine cells from human embryonic stem Coppes RP. Mobilization of bone marrow stem cells by granulo-
cells. Nat Biotechnol 2006; 24: 1392-401. cyte colony-stimulating factor ameliorates radiation-induced dam-
[10] Oh SH, Muzzonigro TM, Bae SH, et al. Adult bone marrow- age to salivary glands. Clin Cancer Res 2006; 12: 1804-12.
derived cells trans-differentiating into insulin-producing cells for [33] Ianus A, Holz GG, Theise ND, Hussain MA. In vivo derivation of
the treatment of type I diabetes. Lab Invest 2004; 84: 607-17. glucose-competent pancreatic endocrine cells from bone marrow
[11] Tang DQ, Cao LZ, Burkhardt BR, et al. In vivo and in vitro charac- without evidence of cell fusion. J Clin Investig 2003; 111: 843-50.
terization of insulin-producing cells obtained from murine bone [34] Yoshida S, Ishikawa F, Kawano N, et al. Human cord blood–
marrow. Diabetes 2004; 53: 1721-32. derived cells generate insulin-producing cells in vivo. Stem Cells
[12] Moriscot C, Fraipont F, Richard MJ, et al. Human bone marrow 2005; 23: 1409-16.
mesenchymal stem cells can express insulin and key transcription [35] Chao KC, Chao KF, Fu YS, Liu SH. Islet-like clusters derived
factors of the endocrine pancreas developmental pathway upon ge- from mesenchymal stem cells in Wharton’s Jelly of the human um-
netic and/or microenvironmental manipulation in vitro. Stem Cells bilical cord for transplantation to control type 1 diabetes. PLoS
2005; 23: 594-603. ONE 2008; 3: e1451. doi:10.1371/journal.pone.0001451.
[13] Choi K, Shin JS, Lee JJ, Kim YS, Kim SB, Kim CW. In vitro [36] Guz Y, Nasir I, Teitelman G. Regeneration of pancreatic b cells
transdifferentiation of rat mesenchymal cells into insulin-producing from intra-islet precursor cells in an experimental model of diabe-
cells by rat pancreatic extract. Biochem Biophys Res Commun tes. Endocrinology 2001; 142: 4956-68.
2005; 330: 1299-305. [37] Bouwens L, DeBlay E. Islet morphogenesis and stem cell markers
[14] Li Y, Zhang R, Qiao H, et al. Generation of insulin-producing cells in rat pancreas. J Histochem Cytochem 1996; 44: 947-51.
from PDX-1 gene modified human mesenchymal stem cells. J Cell [38] Hao E, Tyrber B, Pamela IA, et al. Beta cell differentailation from
Physiol 2007; 211: 36-44. non endocrine epithelial cells of the adult human pancreas. Nat
[15] Choi JB, Uchino H, Azuma K, et al. Little evidence of transdiffer- Med 2006; 12: 310-6.
entiation of bone marrow-derived cells into pancreatic beta cells. [39] Hunziker E, Stein M. Nestin-expressing cells in the pancreatic
Diabetologia 2003; 46: 1366-74. islets of Langerhans. Biochem Biophys Res Commun 2000; 271:
[16] Lechner A, Yang YG, Blacken RA, Wang L, Nolan AL, Habener 116-9.
JF. No evidence of significant transdifferentiation of bone marrow [40] Zulewski H, Abraham EJ, Gerlach MJ, et al. Multipotential nestin-
into pancreatic bcells in vivo. Diabetes 2004; 53: 616-23. positive stem cells isolated from adult pancreatic islets differentiate
[17] Taneera J, Rosengren A, Renstrom E, et al. Failure of transplanted ex vivo into pancreatic endocrine, exocrine, and hepatic pheno-
bone marrow cells to adopt a pancreatic beta cell fate. Diabetes types. Diabetes 2001; 50: 521-33.
2006; 55: 290-6. [41] Petropavlovskaia M, Rosenberg L. Identification and characteriza-
[18] Yang L, Li S, Hatch H, et al. In vitro trans-differentiation of adult tion of small cells in the adult pancreas: Potential progenitor cells?
hepatic stem cells into pancreatic endocrine hormone-producing Cell Tissue Res 2002; 310: 51-8.
cells. Proc Natl Acad Sci USA 2002; 99: 8078-83. [42] Guz Y, Nasir J, Teitelman G. Regeneration of pancreatic beta-cells
[19] Ramiya VK, Maraist M, Arfors KE, Schatz DA, Peck AB, from intra-islet precursor cells in an experimental model of diabe-
Cornelius JG. Reversal of insulin-dependent diabetes using islets tes. Endocrinology 2001; 142: 4956-68.
generated in vitro from pancreatic stem cells. Nat Med 2000; 6: [43] Li WC, Horb ME, Tosh D, Slack JMW. In vitro transdifferentiation
278-82. of hepatoma cells into functional pancreatic cells. Mech Dev 2005;
[20] Sun B, Roh KH, Lee SR, Lee YS, Kang KS. Induction of human 122: 835-47.
umbilical cord blood-derived stem cells with embryonic stem cell [44] Kodama S, Kuhtreiber W, Fujimura S, Dale EA, Faustman DL.
phenotypes into insulin producing islet-like structure. Biochem Islet regeneration during the reversal of autoimmune diabetes in
Biophys Res Commun 2007; 354: 919-23. NOD mice. Science 2003; 302: 1223-7.
[21] Chang CM, Kao CL, Chang YL, et al. Placenta-derived multipotent [45] Kojima H, Fujimiya M, Matsumura K, et al. NeuroD-betacellulin
stem cells induced to differentiate into insulin-positive cells. Bio- gene therapy induces islet neogenesis in the liver and reverses dia-
chem Biophys Res Commun 2007; 357: 414-20 betes in mice. Nat Med 2003; 9: 596-603.
[22] Zuk PA, Zhu N, Mizuno H, et al. Multilineage cells from human [46] Zalzman M, Anker-Kitai L, Efrat S. Differentiation of human liver-
adipose tissue: implications for cell-based therapies. Tissue Eng derived,insulin-producing cells toward the b-cell phenotype. Diabe-
2001; 7: 211-28. tes 2005; 54: 2568-75.
[23] Timper K, Seboek D, Eberhardt M, et al. Human adipose tissue- [47] Planat-Benard V, Silvestre JS, Cousin B, et al. Plasticity of human
derived mesenchymal stem cells differentiate into insulin, soma- adipose lineage cells toward endothelial cells: physiological and
tostatin, and glucagons expressing cells. Biochem Biophys Res therapeutic perspectives. Circulation 2004; 109: 656-63.
Commun 2006; 341: 1135-40. [48] Puissant B, Barreau C, Bourin P, et al. Immunomodulatory effect
[24] Lee J, Han DJ, Kim SC. In vitro differentiation of human adipose of human adipose tissue-derived adult stem cells: comparison with
tissuederived stem cells into cells with pancreatic phenotype by re- bone marrow mesenchymal stem cells. Br J Haematol 2005; 129:
generating pancreas extract. Biochem Biophys Res Commun 2008; 118-29.
375: 547-51. [49] Fang B, Song Y, Lin Q, et al. Human adipose tissue-derived mes-
[25] Czyz J, Wiese J, Rolletscheck C, et al. Potential of embryonic and enchymal stromal cells as salvage therapy for treatment of severe
adult stem cells in vitro. Biol Chem 2003; 384: 1391-409. refractory acute graft-vs.-host disease in two children. Pediatr
[26] Assady S, Maor G, Amit M, Itskovitz-Eldor J, Skorecki KL, Transplant 2007; 11: 814-7.
Tzukerman M. Insulin production by human embryonic stem cells. [50] Zhu Y, Liu T, Song K, Fan X, Ma X, Cui X. Adipose-derived stem
Diabetes 2001; 50: 1691-7. cell: a better stem cell than BMSC. Cell Biochem Funct 2008; 26:
[27] Hussain MA, Theise ND. Stem-cell therapy for diabetes mellitus. 664-75.
Lancet 2004; 364: 203-5. [51] Hardikar AA, Bhonde RR. Modulating experimental diabetes by
[28] Soria B, Roche E, Berna G, Leon-Quinto T, Reig JA, Martin F. treatment with cytosolic extract from the regenerating pancreas.
Insulin-secreting cells derived from embryonic stem cells normal- Diabetes Res Clin Pract 1999; 46: 203-11.
ize glycemia in streptozotocin-induced diabetic mice. derived from
194 Current Stem Cell Research & Therapy, 2010, Vol. 5, No. 2 Kim et al.

[52] Trivedi H, Vanikar A, Thakker U, et al. Human adipose tissue- [54] Chandra V, Swetha G, Phadnis S, Nair PD, Bhonde RR. Genera-
derived mesenchymal stem cells combined with hematopoietic tion of pancreatic hormone expressing islet like cell aggregates
stem cell transplantation synthesize Insulin. Transpl Proc 2008; 40: from murine adipose tissue-derived stem cells. Stem Cells 2009;
1135-9. 27: 1941-53.
[53] Kang HM, Kim JY, Park S, et al. Insulin-Secreting Cells from [55] Lin G, Wang G, Liu G, et al. Treatment of type 1 diabetes with
Human Eyelid-Derived Stem Cells Alleviate Type I Diabetes in adipose tissue-derived stem cells expressing pancreatic duodenal
Immunocompetent Mice. Stem Cells 2009; 27: 1999-2008. homeobox 1. Stem Cells Dev 2009; 18: 1399-406.

Received: June 07, 2009 Revised: July 13, 2009 Accepted: August 04, 2009

You might also like