Vous êtes sur la page 1sur 5

ROAD TO CURES: SCIENCE, TREATMENTS AND ECONOMICS

Stem Cells and Diabetes:


New Trends and Clinical Prospects
By Juan Domínguez-Bendala, Ph.D. and Camillo Ricordi, M.D.

ype I diabetes has been acknowledged, yield increases and the first reports on insulin independence in

T from very early on, as one the top


potential targets of stem cell-based
regenerative therapies. While the treatment of
humans.14 The second was the development of a steroid-free
immunosuppresive regime that helped preserve the survival of
engrafted islets, extending their function for 5 years or longer in a
significant percentage of transplanted patients.15,16 In short, the
other diseases would typically require extensive procedure is based on the enzymatic/mechanical dissociation of islets
bioengineering and/or the recapitulation of a from the pancreatic parenchyma, using a digestion chamber shaken
rather complex cellular niche, the case for type I at a pre-determined speed. Once the digestion is completed, islet cell
diabetes was solidly grounded on the fact that, in clusters are separated by gradient centrifugation and then cultured
and infused in the liver of the patient via the portal vein. Islets are
principle, replacement was needed for only one
known to lodge in the pre-sinusoidal capillaries of the liver, where
cell type – the insulin-producing beta cell. A they get revascularized within weeks of the procedure.9,11,12,17,18 If
tremendous body of work spanning decades of carried out by skilled teams, the procedure has a very low incidence
basic and clinical research had established that of complications, resulting in immediate blood glucose control15 and
beta cells can exert glycemic control in a variety of a significant improvement in the patient’s quality of life.19
ectopic locations, effectively simplifying the Despite the continued refinements of the technique, to this day
islet transplantation remains a “brute force” approach. The digestion
problem. Finally, successful clinical therapies
and isolation procedure may destroy one half of the islets present in
resulting in insulin-independence (islet transplan- any given donor pancreas; and by some estimates, up to 80% of those
tation) had already been in use for two decades, that are infused may also die within hours of the procedure.20 By this
demonstrating the feasibility of cell-based account, the observation that 5 years later only 10-50% of the
approaches for type I diabetes. However, a patients are off-insulin15,21 is not as surprising as the fact that up to
decade after the isolation of the first human 80% are insulin-independent 1 year after the procedure.15 In
summary, our clinical experience is that even very little can go a long
embryonic stem (huES) cells, other a priori less-
way, which bodes well for the applicability of stem cell therapies to
likely targets seem to have taken an early lead in treat this disease.
the race to clinical applications. Informally To avoid redundancy with other articles in this publication, we
organized in “top stories”, this article will review will spare the reader yet another introduction on the basics of stem
the reasons behind this apparent paradox, cells, its many types and prospective uses, proceeding instead to
describing the not-so-simple nature of the describe progress in their use for the treatment of diabetes. In no
particular order of importance, here are some of the most significant
problem, the most promising avenues of research,
advances reported over the last couple of years that are likely to shape
and the challenges that still lie ahead. the overall direction of the field:
Islet Transplantation: The Proof of Concept Functional Beta-like Cells Derived From
Pancreatic transplantation is effective at eliminating the need for huES Cells.
exogenous insulin in type I diabetic patients1, but the limitations
In a triad of groundbreaking communications, the Novocell
common to all solid organ transplantation procedures (including
team led by E. Baetge described in quick succession a method for the
scarcity of donors and complications inherent to major surgery2-7)
efficient generation of definitive endoderm from huES cells22, the first
make it hardly a therapy of choice. Islet transplantation, in contrast,
report on “canonical” huES cell differentiation into beta-like cells23
is a much less invasive approach that allows for the selective
and, finally, a convincing demonstration of in vivo maturation and
heterotopic engraftment of the small percentage of pancreatic cells
function of huES cell-derived pancreatic progenitors.24 The latter
that secrete insulin.8-13 Two major breakthroughs shaped the history of
approach was designed to address the fact that the in vitro protocol,
this practice after the first tentative attempts in the late 70s and early
for all its virtues, was highly inefficient (less than 10% of insulin-
80s. The first one was the invention of a semi-automated method for
producing cells) and did not result in measurable glucose-stimulated
the isolation of islets from a donor pancreas, which led to dramatic
insulin release. The rationale for transplanting immature progenitors

WORLD STEM CELL REPORT 2009 GENETICS POLICY INSTITUTE

73
ROAD TO CURES: SCIENCE, TREATMENTS AND ECONOMICS

was that perhaps the in vivo microenvironment would provide them destroy its own beta cells, immunosuppresion would still be necessary
with the necessary cues to complete their differentiation into no matter the origin of the replacement. It can be argued, however,
functional beta cells. Success was achieved at the expense of a rather that effective iPS-to-beta cell differentiation methods could lead to
long lag period till function (up to three months) and a very high therapies that would tackle two out of the three major problems we
incidence of teratomas, which would obviously hamper the clinical face right now (supply and allorejection), singling out autoimmunity
translation of this method.25 It is unclear at this point the direction as the only remaining hurdle. This would simplify the problem,
the field will take. On the one hand, the full differentiation protocol allowing for a much more effective use of resources to find a cure to
has been reportedly difficult to reproduce with cell lines other than the disease.
those indicated in the original articles, which would argue in favor of
additional research into developing more robust methods that could Infusion of Bone Marrow Stem Cells for
be used with many cell lines. Few would dispute that a fully Type I Diabetes?
functional, ready-to-transplant cell product would be preferable in a It has been hypothesized that some adult stem cells (chiefly of
clinical setting -not only because of the expected reduction in the rate the ubiquitous mesenchymal lineage, obtained from the bone
of tumorigenic events, but also because of the elimination of the lag marrow and other locations) might contribute to the regeneration of
between transplantation and function. In this context, other pancreatic beta cells if delivered either systemically or locally. The
competing methods26 may still prove more viable than Novocell’s. On potential benefits of this approach are clear inasmuch as these cells are
the other hand, progress at sorting out the fully mature cells from the proliferative enough to yield therapeutic loads, can be potentially
carry-over undifferentiated ones, together with the development of obtained from the patient, and pose no ethical concerns. On the
appropriate encapsulation techniques and better controls to identify other hand, premature claims that mesenchymal stem cells (MSCs)
a teratogenic threshold, may still allow for an early therapeutic cells are a safer, non-tumorigenic alternative to huES and iPS cells
implementation of the in vivo maturation approach. have proven wrong. Indeed, their adaptation to in vitro culture can
lead to the development of chromosomal abnormalities resulting in
Directed Differentiation of iPS Cells.
malignant transformation as early as in a few passages.45-50 Also, the
Nothing epitomizes better the frantic pace of stem cell research mesodermal origin of both MSCs and hematopoietic places a big
than the advent and overnight success of induced pluripotent stem question mark on their purported ability to become endodermal
(iPS) cells. Barely two years after the first breakthrough reports on the tissues such as pancreatic beta cells. Early observations that donor
reprogramming of adult fibroblasts by viral transduction of a few bone marrow cells could migrate to the damaged pancreas and
master genes27,28, this technique -which allows for the generation of contribute to the regeneration of islets51 were subsequently disputed
custom-made, patient-matched embryonic stem (ES)-like cells- has by studies that confirmed the migration but not the differentiation.52-54
almost relegated to oblivion the much more cumbersome theoretical The latter of such studies, in fact, provided evidence that the cells
alternative of somatic cell nuclear transfer (SCNT) for “therapeutic recruited to the pancreas were rather of endothelial origin. As is the
cloning”.29,30 The biotechnological feat of reprogramming adult case with MSCs -which have well-documented trophic, angiogenic,
somatic cells in such simple fashion is likely to have enormous immunomodulatory and anti-inflammatory effects55-61, these
medical implications, provided that (a) the newly reprogrammed cells mobilized bone marrow-derived cells could indeed aid in the
are comparable to blastocyst-derived ES cells; and (b) the procedure endogenous regeneration of beta cells, but not by direct
can be done safely. Despite some reports indicating that the differentiation.
molecular signature of iPS cells might be, after all, somewhat Whichever the mechanism may be, autologous intra-pancreatic
different to that of ES cells31, there is very little question now that bone marrow transplantation is currently enjoying the limelight as a
both pluripotent stem cell types are, to a large extent, potential new treatment for type I and II diabetes. Much of this
interchangeable. As for the safety concerns, ongoing efforts at attention is due to the controversial for-profit clinical practice of the
addressing them include the use of non-viral delivery methods32, technique in some countries at a time when there is no evidence of
episomal vectors33, protein transduction34,35, the progressive safety and efficacy in humans. Fortunately, there are already a number
replacement of reprogramming genes by chemical factors36-39 and the of Phase I/II trials looking at the potential benefit of local delivery of
use of adult stem cells as substrates for reprogramming.40 MSCs and bone marrow cells (www.clinicaltrials.gov), including one
As these techniques quickly became mainstream, laboratories in the USA for type II diabetes in which autologous intra-pancreatic
around the world endeavored to replicate with iPS cells results bone marrow cell transplantation is done in conjunction with
previously attained with huES cells.41-43 In this context, the eventual hyperbaric treatment. The role of oxygen in promoting beta cell
use of these cells for direct differentiation into beta-like cells was differentiation and survival has been recently established62-64, and
nothing short of inevitable. The first report was published last year by preliminary results of this combination therapy in human subjects are
Tateishi and colleagues44, who drove skin-derived iPS cells along the highly encouraging.65
beta cell lineage using a protocol similar to that described by Jiang et
al 26 for huES cells. Resetting the Immune System?
The fact that type I diabetes is an autoimmune disorder certainly Most people tend to think about stem cells as “building blocks”
limits enthusiasm for the prospective uses of patient-matched iPS to be used for the repair of damaged tissues. We have described above
cells. After all, since the patient’s immune system is already poised to how bone marrow stem cells may actually help in the regeneration of
WORLD STEM CELL REPORT 2009 GENETICS POLICY INSTITUTE

74
ROAD TO CURES: SCIENCE, TREATMENTS AND ECONOMICS

islets by providing endogenous progenitor cells with the chemical islets72 and then by the unexpected finding that the Ngn3 endocrine
cues required to complete their maturation, or by otherwise differentiation program could be reactivated in the adult pancreas
supporting their action (e.g., by creating anti-inflammatory or following partial duct ligation.73 The physiological significance of this
angiogenic microenvironments). One of the major developments in model is unclear, and it is a safe assumption that self-replication is still
the field over the past few years has to do with another well-known the main engine of adult beta cell regeneration. However, now we
facet of bone marrow stem cells, namely their ability to modulate the know that the pancreas may still have an “ace in the hole”, an
body’s immune response. Thus, in clinical trials conducted on 15 alternative pathway that could be activated under specific
newly diagnosed type I diabetic patients, hemaotopoietic stem cells circumstances and take over regeneration. Further research will be
were mobilized, harvested from peripheral blood and subsequently needed to decipher its molecular determinants, the reasons that make
frozen for later use. Following a harsh immunosuppressive treatment this regeneration model different from all the others (which do not
intended to destroy autoreactive T cell clones, the patients were then induce the Ngn3 program) and the potential ways to apply such
reinfused with their own cryopreserved hematopoietic stem cells, knowledge to our advantage for therapeutic purposes.
which were assumed to be “naïve” (i.e., at a stage prior to the onset of
the disease). In the first communication, the authors reported that 14 Transdifferentiation
out of the 15 patients became insulin-free for extended periods of Also in line with the above strategy, a subtler but equally effective
time.66,67 A recent update of the original study with a longer follow- method to induce beta cell neogenesis in the adult pancreas was
up (21/2 years) confirmed that C-peptide levels increased significantly recently reported by Zhou and colleagues74, who described the
in the treated patients, the majority of whom achieved and “reprogramming” of the acinar tissue of the murine pancreas by
maintained insulin independence.68 Excitement was and remains ectopically expressing a combination of three genes, namely Pdx1,
justified, as such reports are the first ever suggestive of a definitive Ngn3 and MafA. New insulin-producing cells – virtually
“cure”. However, the approach also has substantive limitations. For indistinguishable from the native ones- were detected just days after
one, it has been hypothesized to work only in recently diagnosed the adenoviral delivery of the aforementioned cassettes, and kept
patients, who may still have a significant mass of beta cells susceptible expanding long after the viruses had been cleared from the host.
of regeneration. If such mass is below a still undetermined threshold Clearly inspired in the pioneering work of Ferber and colleagues
(as would be expected in long-standing diabetes), it may still be in the liver75-77, the new approach had the advantage that -perhaps due
necessary to supplement the patients with an exogenous “boost” of to the closeness of the starting material to the desired end product-
beta cells. Secondly, the immunosuppressive regime is highly toxic, the reprogramming process completely abrogated the original
and potentially fatal. Although no deaths have been reported yet in phenotypes; in other words, these were true beta cells, not merely
relation to this therapy, the sample sizes are still very small. A 2005 exocrine cells that secreted insulin. Hyperglycemia was significantly
study evaluating the clinical outcome of autologous hematopoietic ameliorated in the treated animals, even if diabetes was not
stem cell transplantation for a variety of autoimmune conditions completely reversed. This could be due to the fact that, while direct
(including neurological, rheumatologic, hematological and communication between beta cells appears to be critical for the
gastrointestinal disorders) concluded that the treatment was effective, synchronicity and effectiveness of glucose-mediated insulin
but associated with a high rate of morbidity and mortality (7% at 3 secretion78,79, the newly induced cells failed to aggregate in clusters.
years).69 However, better patient selection, refinement of methods, The clinical applicability of this strategy is still unclear. Although
and progressive experience of teams are likely to decrease transplant- adenoviruses are present in the recipient only transiently, they are
related mortality to more acceptable rates. known to elicit serious immune responses. However, these
experiments are proof of principle that exocrine cell reprogramming
Regeneration: From The Duct to Replication is feasible and perhaps doable ex vivo on cultured acinar cells that
(And Back?) would be subsequently transplanted. Non-viral alternatives (such as
Proof of the rapid evolution of an ever-changing field is the the use of chemicals and/or protein transduction) should also be
continuous shift of the paradigm on adult beta cell regeneration. It explored with the goal of improving the safety of this promising
was the conventional wisdom for decades that beta cells arose from approach, thus accelerating its clinical translation.
progenitor cells residing in the pancreatic ducts, although the support
for this hypothesis had always been anecdotal and largely based on New trends in transplantation
two-dimensional pictures of beta cells seemingly budding out from Although the liver has served as the surrogate for the pancreas for
the periductal area.70 A series of elegant lineage-tracing experiments all islet transplantation procedures conducted in humans thus far,
conducted in a transgenic mouse model challenged this notion in there is widespread consensus that this ectopic location is far from
2004, indicating that self-replication was the main mechanism optimal. As mentioned earlier in this article, islets sustain a massive
behind adult beta cell turnover and regeneration.71 While the cell loss upon intra-hepatic infusion. Stem cell-derived beta cells
proponents of the “adult stem cell” theory still contended that human (whichever their origin) are likely to share the same fate, unless
beta cell regeneration might obey to different rules, these clear-cut alternative sites are identified. There has been an active search of such
observations clearly put the burden of proof on their shoulders. Then sites in recent years, including an intriguing approach pioneered by
the new idea was challenged again in 2008, first by new lineage Speier and colleagues in Miami80 by which islets transplanted in the
tracing experiments showing ductal offspring in newly created anterior chamber of the eye can restore normoglycemia in diabetic

WORLD STEM CELL REPORT 2009 GENETICS POLICY INSTITUTE

75
ROAD TO CURES: SCIENCE, TREATMENTS AND ECONOMICS

mice while allowing for a real-time, in vivo follow-up of their currently do when infused through the portal vein. This would enable
engraftment, vascularization and response to the immune attack. the clinician to better monitor the graft while minimizing the
Unfortunately, the generalized view of the eye as an inflammatory responses. Secondly, the use of these devices may
immunoprivileged site81 has not proven true for islet transplantation. eventually allow for the administration of immunosuppresants locally
This, together with the rather limited load of islets that could be at much smaller effective doses than those required if taken
transplanted in a clinical setting, makes the eye an unlikely candidate systemically. This would largely prevent the occurrence of the side
to replace the liver as a preferred location. effects associated with oral immunosuppresion83. Finally, if using
An alternative to finding a suitable site is to create one. Thus, huES/iPS cell-derived tissues as a source of insulin-producing cells,
Pileggi et al82 demonstrated recently that islets infused into a any teratogenic lesion potentially arising from carryover
biocompatible, subcutaneously implanted device – previously undifferentiated cells would be contained within the device.
allowed to vascularize through its stainless-steel mesh walls – were
able to reverse hyperglycemia in diabetic rats. The advantages of such Acknowledgements
an approach are many-fold: first, the islets (or the stem cell-derived CR and JDB are funded by the NIH, JDRF and the Diabetes
beta cells) would not spread throughout an entire organ, as they Research Institute Foundation (DRIF).

Juan Domínguez-Bendala, Ph.D.

Juan Domínguez-Bendala is Director of the cloning of Dolly the sheep. During his training, he acquired
the Pancreatic Development & Stem Cells considerable experience in ES cell research and state-of-the-art
laboratory at the Diabetes Research genetic engineering techniques. His current projects focus on the use
Institute, University of Miami. He of stem cells to obtain pancreatic islets. He co-invented the “oxygen
obtained his Ph.D. at the Roslin Institute sandwich”, a culture device designed to promote differentiation. He
(UK) under the supervision of Dr. is also working on long-term culture/regeneration of pancreatic stem
McWhir, co-author of the 1997 report on cells and reprogramming.

Camillo Ricordi, M.D.

Camillo Ricordi, M.D., is Professor of known for developing the method for large scale isolation of human
Surgery, Medicine, Biomedical pancreatic islets, and for leading the team that performed the first
Engineering, Microbiology and series of successful clinical islet allografts in 1990. Dr. Ricordi’s
Immunology, Director of the Cell research interests include induction of immune tolerance, cellular
Transplant Center and head of the therapies, stem cells and regenerative medicine strategies. Dr. Ricordi
Diabetes Research Institute at the received numerous honors and awards, authored over 600 scientific
University of Miami. Dr. Ricordi is publications and, as inventor, holds nine patents.

WORLD STEM CELL REPORT 2009 GENETICS POLICY INSTITUTE

76
ROAD TO CURES: SCIENCE, TREATMENTS AND ECONOMICS

References
1. Martin-Pagola, A. et al. Diabetologia. 51, 1803-13 (2008). 44. Tateishi, K. et al. J Biol Chem. (2008).
2. Burke, G.W. et al. Transplantation. 77, S62-7 (2004). 45. Aguilar, S. et al. Stem Cells. 25, 1586-94 (2007).
3. Fabrega, A.J. et al. J Intensive Care Med. 9, 281-9 (1994). 46. Miura, M. et al. Stem Cells. 24, 1095-103 (2006).
4. Freise, C.E. et al. West J Med. 170, 11-8 (1999). 47. Rubio, D. et al. Cancer Res. 65, 3035-9 (2005).
5. Landgraf, R. Diabetologia. 39, 1415-24 (1996). 48. Tolar, J. et al. Stem Cells. 25, 371-9 (2007).
6. Melton, L.B. Semin Nephrol. 12, 256-66 (1992). 49. Zhou, Y.F. et al. Cancer Res. 66, 10849-54 (2006).
7. Pirsch, J.D. et al. Am J Kidney Dis. 27, 444-50 (1996). 50. Furlani, D. et al. Cell Transplant. 18, 319-31 (2009).
8. Inverardi, L. et al. Curr Opin Immunol. 15, 507-11 (2003). 51. Ianus, A. et al. J Clin Invest. 111, 843-50 (2003).
9. Merani, S. et al. Clin Sci (Lond). 110, 611-25 (2006). 52. Choi, J.B. et al. Diabetologia. 46, 1366-74 (2003).
10. Murdoch, T.B. et al. Cell Transplant. 13, 605-17 (2004). 53. Lechner, A. et al. Diabetes. 53, 616-23 (2004).
11. Pileggi, A. et al. Minerva Endocrinol. 31, 219-32 (2006). 54. Mathews, V. et al. Diabetes. 53, 91-8 (2004).
12. Ricordi, C. et al. Nat Rev Immunol. 4, 259-68 (2004). 55. Abdi, R. et al. Diabetes. 57, 1759-67 (2008).
13. Seung, E. et al. Curr Diab Rep. 3, 329-35 (2003). 56. Mishra, P.K. J Cardiovasc Med (Hagerstown). 9, 122-8 (2008).
14. Ricordi, C. et al. Diabetes. 37, 413-20 (1988). 57. Le Blanc, K. et al. J Intern Med. 262, 509-25 (2007).
15. Ryan, E.A. et al. Diabetes. 54, 2060-9 (2005). 58. Ozaki, K. et al. Int J Hematol. 86, 5-7 (2007).
16. Shapiro, A.M. et al. N Engl J Med. 343, 230-8 (2000). 59. Wu, Y. et al. Stem Cells. 25, 2648-59 (2007).
17. Ricordi, C. Diabetes. 52, 1595-603 (2003). 60. Ball, S.G. et al. J Cell Mol Med. 11, 1012-30 (2007).
18. Sakuma, Y. et al. Transplant Proc. 40, 343-5 (2008). 61. Xu, Y.X. et al. Med Hypotheses. 71, 390-3 (2008).
19. Poggioli, R. et al. Am J Transplant. 6, 371-8 (2006). 62. Fraker, C., Ricordi, C., Inverardi, L., and Dominguez-Bendala, J.
20. Barshes, N.R. et al. J Leukoc Biol. 77, 587-97 (2005). Culture in perfluorinated platforms results in enhanced human islet
function. in XIV International Society for Cellular Therapy meeting,
21. Bellin, M.D. et al. Am J Transplant. 8, 2463-70 (2008). May 17-20 Vol. 1 81 (Miami, 2008).
22. D’Amour, K.A. et al. Nat Biotechnol. 23, 1534-41 (2005). 63. Fraker, C., Ricordi, C., Inverardi, L., and Dominguez-Bendala, J.
23. D’Amour, K.A. et al. Nat Biotechnol. (2006). Biology of the Cell. 101, 431-440 (2009).
24. Kroon, E. et al. Nat Biotechnol. 26, 443-52 (2008). 64. Fraker, C.A. et al. Stem Cells. 25, 3155-64 (2007).
25. Ricordi, C. et al. Nat Biotechnol. 26, 397-8 (2008). 65. Estrada, E.J. et al. Cell Transplant. 17, 1295-304 (2008).
26. Jiang, J. et al. Stem Cells. 25, 1940-53 (2007). 66. Voltarelli, J.C. et al. Jama. 297, 1568-76 (2007).
27. Takahashi, K. et al. Cell. 131, 861-72 (2007). 67. Couri, C.E. et al. Autoimmunity. 1 (2008).
28. Yu, J. et al. Science. 318, 1917-20 (2007). 68. Couri, C.E. et al. Jama. 301, 1573-9 (2009).
29. Wilmut, I. et al. Nature. 385, 810-3 (1997). 69. Gratwohl, A. et al. Bone Marrow Transplant. 35, 869-79 (2005).
30. Byrne, J.A. et al. Nature. 450, 497-502 (2007). 70. Bonner-Weir, S. et al. Diabetes. 42, 1715-20 (1993).
31. Wilson, K.D. et al. Stem Cells Dev. 18, 749-58 (2009). 71. Dor, Y. et al. Nature. 429, 41-6 (2004).
32. Okita, K. et al. Science. (2008). 72. Bonner-Weir, S. et al. Biochem Soc Trans. 36, 353-6 (2008).
33. Yu, J. et al. Science. (2009). 73. Xu, X. et al. Cell. 132, 197-207 (2008).
34. Bosnali, M. et al. Biol Chem. 389, 851-61 (2008). 74. Zhou, Q. et al. Nature. (2008).
35. Zhou, H., Wu, S., Young Joo, J., Zhu, S., Wook Han, D., Lin, T., 75. Ferber, S. et al. Nat Med. 6, 568-72 (2000).
Trauger, S., Bien, G., Yao, S., Zhu, Y., Siuzdak, G., Scholer, H., Duan, 76. Meivar-Levy, I. et al. Hepatology. 46, 898-905 (2007).
L., Ding, S. Cell Stem Cell. doi:10.1016/j.stem.2009.05.005(2009).
77. Sapir, T. et al. Proc Natl Acad Sci U S A. 102, 7964-9 (2005).
36. Huangfu, D. et al. Nat Biotechnol. 26, 1269-75 (2008).
78. Cabrera, O. et al. Proc Natl Acad Sci U S A. 103, 2334-9 (2006).
37. Huangfu, D. et al. Nat Biotechnol. 26, 795-7 (2008).
79. Konstantinova, I. et al. Cell. 129, 359-70 (2007).
38. Shi, Y. et al. Cell Stem Cell. 3, 568-74 (2008).
80. Speier, S. et al. Nat Med. (2008).
39. Shi, Y. et al. Cell Stem Cell. 2, 525-8 (2008).
81. Taylor, A.W. Eye. (2009).
40. Kim, J.B. et al. Cell. 136, 411-9 (2009).
82. Pileggi, A. et al. Transplantation. 81, 1318-24 (2006).
41. Dimos, J.T. et al. Science. 321, 1218-21 (2008).
83. Fort, A. et al. Cell Transplant. 17, 997-1003 (2008).
42. Tateishi, K. et al. J Cell Mol Med. (2008).
43. Mauritz, C. et al. Circulation. 118, 507-17 (2008).

WORLD STEM CELL REPORT 2009 GENETICS POLICY INSTITUTE

77

Vous aimerez peut-être aussi