当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2006年 > 第2期 > 正文
编号:11256967
Failure of Transplanted Bone Marrow Cells to Adopt a Pancreatic -Cell Fate
     1 Hematopoietic Stem Cell Laboratory, Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, Lund University, Lund, Sweden

    2 Department of Physiological Sciences, Lund University, Lund Sweden

    3 Department of Developmental Biology, Hagedorn Research Institute, Copenhagen, Denmark

    Key Words: GFP, green fluorescence protein Nkx6.1, homeodomain protein Pdx-1, pancreatic duodenal homeobox factor-1

    ABSTRACT

    Recent studies in normal mice have suggested that transplanted bone marrow cells can transdifferentiate into pancreatic -cells at relatively high efficiency. Herein, adopting the same and alternative approaches to deliver and fate map-transplanted bone marrow cells in the pancreas of normal as well as diabetic mice, we further investigated the potential of bone marrow transplantation as an alternative approach for -cell replacement. In contrast to previous studies, transplanted bone marrow cells expressing green fluorescence protein (GFP) under the control of the mouse insulin promoter failed to express GFP in the pancreas of normal as well as diabetic mice. Although bone marrow cells expressing GFP under the ubiquitously expressed -actin promoter efficiently engrafted the pancreas of normal and hyperglycemic mice, virtually all expressed CD45 and Mac-1/Gr-1, demonstrating that they adopt a hematopoietic rather than -cell fate, a finding further substantiated by the complete absence of GFP+ cells expressing insulin and the -cell transcription factors pancreatic duodenal homeobox factor-1 and homeodomain protein. Thus, transplanted bone marrow cells demonstrated little, if any, capacity to adopt a -cell fate.

    Anumber of recent studies (1eC8) have suggested that bone marrow transplantation may give rise to nonhematopoietic cell lineages in multiple organs by a process termed transdifferentiation. However, these data are controversial, as other studies (9eC14) have failed to document such transdifferentiation, although in some cases bone marroweCderived hematopoietic cells have been demonstrated to contribute at very low frequencies to other cell lineages through cell fusion (12,15eC19). In parallel with this work, other studies (20eC24) have extended the concept of developmental plasticity of transplanted bone marrow cells. One of the most surprising and compelling claims of cell plasticity following bone marrow transplantation suggests that transplanted bone marrow cells can give rise to fully competent insulin-producing pancreatic -cells (25). Whereas most other reports (13,26,27) of bone marrow plasticity have indicated that transdifferentiation occurs at a very low rate, the studies of Ianus et al. (25) suggested that as much as 1.7eC3% of the insulin-expressing cells in the islets of transplanted mice were derived from reconstituting bone marrow cells. If these findings can be confirmed, bone marrow cells might clearly play a role in the adaptive increase in -cell mass that occurs physiologically during, for example, obesity and pregnancy. Moreover, bone marrow cells may also represent an attractive source for cell replacement therapy of type 1 and type 2 diabetes, not least in view of the limited supply of human islets and ethical issues surrounding embryonic stem cell research (28eC31). Although some subsequent studies (32,33) have obtained results compatible with the generation of insulin-expressing pancreatic cells from transplanted bone marrow cells, the reported efficiency has been several orders of magnitude lower than initially reported, and other studies have failed to detect any transdifferentiation into -cells at all (34). Even when insulin-expressing cells were obtained following bone marrow transplantation, these did not express any of the transcription factors characterizing the mature -cell (35). Thus, the capacity of transplanted bone marrow cells to contribute to -cell regeneration remains controversial. However, until this controversy has been resolved, bone marrow cells continue to be promoted as a candidate source for cell replacement therapy for diabetes (31). A problem in this context is the different transplantation, conditioning, and transgenic fate-mapping approaches used in the previous studies (25,32,34,35). Here, we have adopted different experimental strategies to follow the fate of transplanted bone marrow cells in the pancreas of normal as well as hyperglycemic mice. Although we found evidence for extensive bone marrow cell engraftment in control and damaged islets, the engraftment proved to be almost exclusively of the hematopoietic lineage, and no evidence was obtained for bone marrow cells giving rise to cells expressing insulin or the essential -cell transcription factors pancreatic duodenal homeobox factor-1 (Pdx-1) and homeodomain protein (Nkx6.1).

    RESEARCH DESIGN AND METHODS

    Two mouse strains on a C57BL/6 background, transgenic for green fluorescence protein (GFP) and driven by mouse insulin I gene promoter (MIP/GFP; kindly provided by Dr. Manami Hara, University of Chicago, Chicago, IL) (36) or the -actin promoter (/GFP; kindly provided by Dr. M. Okabe, Osaka University, Suita, Japan) (37), were used as donors of bone marrow cells. Both of these strains express the CD45.2 isoform of the panhematopoietic cell surface antigen CD45. As recipient mice, we used wild-type C57BL/6 mice expressing CD45.1. All mice were given sterile food and autoclaved acidified water and housed under pathogen-free conditions. All procedures were performed with consent from the local ethical committee at Lund University.

    Generation of bone marrow chimeric mice.

    Whole bone marrow cells were collected from femurs and tibias of 10- to 14-week-old CD45.2 MIP/GFP or /GFP donor mice. Femurs and tibias were gently crushed in a mortar, and the cell suspension was filtered through a 70-e蘭 mesh filter (Becton Dickinson Falcon, Erembodegem, Belgium). The fraction of dead cells in the suspension was determined with Trypan blue (Sigma-Aldrich, St. Louis, MO), and the volume used for transplantation was adjusted accordingly. MIP/GFP or /GFP (both CD45.2) bone marrow cells were transplanted into lethally irradiated (975 cGy of irradiator from a Cs137 source) wild-type C57BL/6 (CD45.1) recipient mice. Hematopoietic chimerism and blood lineage distribution was evaluated in peripheral blood of recipient mice 4eC6 weeks after transplantation by flow cytometry (FACSCalibur; Becton Dickinson, San Jose, CA) as previously described (12). Data were analyzed using FlowJo software (TreeStar, San Carlos, CA).

    Alloxan-induced diabetes and cytokine mobilization of bone marrow chimeric mice.

    Wild-type mice with high levels of GFP+ multilineage hematopoietic reconstitution underwent cytokine mobilization and alloxan treatment (Sigma) 8 weeks after bone marrow transplantation (Fig. 4A). Starting 2 days before alloxan injection, bone marrow cells were, as previously described (12), mobilized to peripheral blood by five daily injections (5 e蘥 · mouseeC1 · dayeC1) with recombinant human fms-like tyrosine kinase-3 ligand and recombinant murine granulocyte macrophage colonyeCstimulating factor (kind gifts of Amgen, Thousand Oaks, CA). Diabetes was induced by a single intravenous injection of alloxan monohydrate at 80 mg/kg body wt as previously described (38,39). Blood insulin and glucose levels were evaluated 5 days later, and 4eC6 weeks after alloxan administration, mice were killed and pancreata harvested and fixed for immunohistochemistry.

    Exendin-4 treatment.

    Six weeks after MIP/GFP bone marrow transplantation, mice were treated with 10 daily intraperitoneal injections of exendin-4, an analog of glucagon-like peptide-1 (24 nmol/kg) (Sigma-Aldrich). Mice were killed for immunohistochemical analysis 4 weeks after the last injection of exendin-4 treatment.

    Antibodies.

    The following primary antibodies were used: rabbit antieCGST-STF1 (Pdx-1), rabbit antieCGST-Nkx6.1 (both from Hagedorn Research Institute, Copenhagen, Denmark), guinea-pig anti-insulin/proinsulin (B65-1) (Euro-Diagnostica, Malm, Sweden), chicken anti-GFP (Chemicon, Temecula, CA), rat anti-mouse CD45 (NeoMarkers, Fermont, CA), and rat anti-mouse CD4 (145eC2C11), CD8 (53-6.7), B220 (RA3-6B2), Mac-1 (M1/70), Gr-1 (RB6-8C5), CD45.2 (104), and CD45.1 (A20) antibodies (all from PharMingen, San Diego, CA). Secondary antibodies included fluorescein isothiocyanateeCand Texas redeCconjugated donkey anti-chicken IgG, Cy5-conjugated donkey anti-rat IgG, Texas redeCconjugated donkey antieCguinea pig IgG (all from Jackson Immuno Research Laboratories, West Grove, PA), and Alexa Fluor 555 conjugated with goat anti-rat IgG or goat anti-rabbit IgG (Molecular Probes, Eugene, OR).

    Phenotypic and morphological evaluations of pancreatic sections.

    Dissected pancreata as well as livers were fixed with Stefanini fixative solution overnight at 4°C followed by equilibration overnight in 20% sucrose at 4°C, embedded in Tissue-Tek O.C.T. (Sakura Finetek Europe B.V., Zoeterwoude, the Netherlands), cryosectioned (8eC12 e蘭), and stored at eC80°C until examination. For immunohistochemistry, the sections were permeabilized with 0.25% Triton X100 (Sigma) for 10 min, washed three times with PBS, and blocked with 5% donkey or goat serum (Sigma) for 1 h at room temperature. Primary antibodies were diluted with 5% donkey or goat serum and incubated overnight at 4°C. After washing with PBS three times, appropriate secondary antibodies were incubated for 45 min at room temperature. Finally, to visualize the cell nuclei, sections were washed and incubated with Hoechst 33342 for nuclear staining (Sigma). Staining with control antibodies, as well as staining of negative and positive control tissues, was always performed to control the specificity of the antibodies. -CelleCspecific antibodies were titrated on sections of viable pancreatic islets from wild-type and /GFP transgenic mice. Hematopoietic marker-specific antibodies were titrated on samples from bone marrow and spleen. Immunoreactivity was visualized and images taken using conventional fluorescence (Olympus, London, U.K.) or confocal (LSM 510 Meta; Zeiss, Jena, Germany) microscopy. In nondiabetic mice, an average of 60 pancreatic sections as well as 30 liver sections were analyzed per mouse, and in alloxan-treated mice an average of 30eC40 pancreatic sections were examined per mouse, equally distributed between the pancreatic head and the tail. For all experimental groups, a minimum of 14 mice (except for the exendin-treated cohort, in which only 5 mice were used) were examined, for a total of 420eC840 pancreatic sections representing 3 x 104 to 7.5 x 105 -cells. All datasets were independently confirmed by at least two investigators in two different laboratories involved in the studies.

    RESULTS

    Bone marrow cells engrafted into the pancreas adopt hematopoietic, but not -cell, fates in nondiabetic mice.

    We first used the same experimental strategy as Ianus et al. (25), in which bone marrow cells were reported to acquire a -cell fate in vivo upon transplantation into nondiabetic mice. Lethally irradiated wild-type mice were first transplanted intravenously with a high number (1 x 107) of unfractionated bone marrow cells from mice in which GFP is expressed under control of the mouse insulin promoter (MIP/GFP) (36). As expected, GFP expression in MIP/GFP mice was restricted to pancreatic -cells (Fig. 1A), and no peripheral blood cells derived from MIP/GFP bone marrow cells expressed GFP (Fig. 1B). Transplanted mice were killed, as in the studies of Ianus et al. (25), 4eC6 weeks posttransplantation, at which time transplanted bone marrow cells contributed to >80% of peripheral blood cells (Fig. 1B, Table 1). Examination by direct fluorescence microscopy of as much as 800 pancreatic islet sections (representing 7.5 x 105 -cells) from 14 MIP/GFP bone marroweCtransplanted mice failed to reveal pancreatic engraftment of any GFP+ cells even when using an anti-GFP antibody to enhance sensitivity (Fig. 2A, Table 1). We also failed to detect any GFP expression in the liver of transplanted mice (420 liver sections from 14 transplanted mice analyzed). The same negative picture was obtained when the pancreata were analyzed 3 months posttransplantation (J.T., S.E.W.J., unpublished observation). Although the use of MIP/GFP bone marrow cells allows specific and high-resolution tracking of bone marroweCderived cells expressing insulin, such cells cannot be used to determine their level of pancreatic engraftment and fate determination. Thus, we next performed the same experimental bone marrow transplantation protocol in nondiabetic mice, using bone marrow cells in which GFP was instead expressed under the ubiquitously expressed -actin promoter (/GFP) (37) (Fig. 1A and B). In contrast to MIP/GFP bone marroweCtransplanted mice, high levels of engrafted GFP+ cells were observed in the pancreas of all mice transplanted with /GFP bone marrow cells (Fig. 2, Table 1). However, in 420 sections (containing 4.0 x 105 -cells), we found no cells coexpressing GFP and insulin (Fig. 2A, Table 1). Furthermore, no bone marroweCderived GFP+ cells coexpressed the -cell transcription factors Pdx-1 or Nkx6.1 (Fig. 2B and C, Table 1). By contrast, virtually every GFP+ cell engrafted into the pancreas expressed the panhematopoietic marker CD45 (>99.9%; Fig. 3A, Table 1) as well as myeloid antigens Mac-1/Gr-1, whereas they were negative for T-cell (CD4, CD8) and B-cell (B220) markers (Fig. 3B, Table 1). Thus, transplanted bone marrow cells have the capacity to engraft into the pancreas of nondiabetic recipients but then exclusively adopt a hematopoietic rather than a -cell fate.

    Lack of evidence for bone marroweCderived -cells in diabetic mice.

    A number of studies (12,15,17,18) have suggested that the ability of bone marroweCderived cells to adapt nonhematopoietic fates in vivo, primarily through cell fusion, is dependent on or enhanced by organ-specific insults. In addition, it has been suggested that cytokine-induced mobilization of bone marrow cells to peripheral blood can promote their migration to nonhematopoietic tissues and nonhematopoietic fate conversion (12,40). Thus, we next explored the possibility that mobilized bone marrow cells might adapt a -cell fate in mice rendered diabetic following treatment with the -celleCspecific toxin alloxan (38,39). Wild-type mice reconstituted with MIP/GFP or /GFP bone marrow cells were cytokine mobilized before and after alloxan treatment (Fig. 4A). To verify the extent of alloxan-induced -cell injury, blood insulin and glucose levels were measured 5 days later. Alloxan-treated mice had reduced blood insulin levels (4 ± 2 e蘒/ml) compared with untreated controls (25 ± 7 e蘒/ml), and blood glucose was elevated 2.5-fold from 6 ± 2 mmol/l in untreated mice to 15 ± 4 mmol/l. Furthermore, immunohistochemical analysis of pancreatic islets from alloxan-treated mice revealed extensive pancreatic islets damage (Fig. 4B). However, in accordance with the data from nondiabetic mice, no GFP+ cells were observed in the pancreata of alloxan-treated mice reconstituted with MIP/GFP bone marrow cells (Fig. 4C, Table 2). In total, we investigated sections representing 50,000 pancreatic -cells in 21 mice. Alloxan-treated mice reconstituted with /GFP cells consistently exhibited pancreatic engraftment with GFP+ bone marroweCderived cells (Fig. 4C, Table 2). However, investigation of sections representing >30,000 pancreatic -cells demonstrated that >99% of the bone marroweCderived GFP+ cells coexpressed the hematopoietic markers CD45 and Mac-1/Gr-1, whereas not a single GFP+ cell was found to express insulin, Pdx-1, or Nkx6.1 (Fig. 4C, Table 2).

    Finally, mice reconstituted with MIP/GFP bone marrow cells were treated with exendin-4, a glucagon-like peptide-1 analog that, in addition to its insulin-releasing action, stimulates pancreatic -cell growth with resultant increase in pancreatic -cell mass (41,42). However, exendin-4 had no detectable effect when this cohort (alloxan-treated mice) was analyzed 4 weeks after exendin-4 treatment and no GFP+ cells were observed in investigated pancreatic or liver sections (not shown).

    DISCUSSION

    The recent report (25) of bone marrow cells giving rise to up to 3% insulin-expressing cells when transplanted in vivo has raised the exciting possibility that bone marrow cells could be used for cellular replacement therapy in diabetes (28,31). Because they are so readily accessible and can be delivered by autologous transplantation, bone marrow cells would represent a particularly attractive source of -cells for cell replacement therapy. However, before embarking on a route for clinical development, it is essential to first unequivocally establish that bone marroweCderived -cell transdifferentiation does indeed occur reproducibly. Many claims of bone marrow transdifferentiation have proven to be difficult to reproduce (10,12,14,32,43). Therefore, it has been proposed that a key criterion for demonstration of transdifferentation of bone marrow cells is that the finding should be independently reproduced in more than one laboratory (26,44). Unfortunately, several recent studies have partially (32) or fully (34) failed to reproduce the initial findings of bone marroweCderived -cell generation (25). However, because alternative procedures were used for tracing transplanted bone marrow cells and/or conditioning the recipients, it remains possible that bone marrow transdifferentiation to insulin-expressing cells only occurs under certain conditions. Furthermore, the cellular fate(s) of a large fraction of bone marroweCderived cells engrafted in the pancreas was not established in these studies, leaving open the possibility that they might represent bone marrow cells that have partially embarked on a differentiation path toward -cells (32,34). Here, we systematically compared multiple approaches to promote and investigate the -cell potential of the transplanted bone marrow cells. One of these was the same as that used by Ianus et al. (25) and involved analysis of pancreatic sections for presence of GFP+ cells 4eC6 weeks after bone marrow transplantation into lethally irradiated mice of the same background as in the original study. To enhance resolution, we used bone marrow cells in which GFP was under the control of an insulin-specific promoter. This was essential to allow potentially very low levels of transdifferentiation to be detected. Although the levels of hematopoietic reconstitution obtained with bone marrow transplantation were high, we failed to detect any GFP+ cells among the high number of transplanted mice and -cells investigated. Importantly, if the levels of transdifferentiation had been similar to Ianus et al. (25) (1.7eC3%), we would have expected to find at least 1 x 104 GFP+ cells in the investigated sections. Performing similar experiments with bone marrow cells in which GFP was instead under the control of the ubiquitous -actin promoter, we were able to demonstrate considerable GFP+ engraftment in the pancreas of bone marroweCtransplanted mice. However, none of the engrafted cells expressed insulin or the transcription factors Pdx-1 and Nkx6.1 (45,46). Most importantly, in contrast to Choi et al. (34) and Lechner et al. (32), who used a different anti-CD45 antibody than us, we were able to demonstrate that >99.9% of the bone marroweCderived GFP+ cells expressed the panhematopoietic marker CD45 as well as the myeloid antigen Mac-1 (12). Thus, our data clearly demonstrate that /GFP bone marroweCderived cells engraft efficiently in the pancreas but adopt almost exclusively a hematopoietic cell fate. As we failed to detect any bone marroweCderived -cells in steady state, we also investigated the ability of MIP/GFP as well as /GFP bone marrow cells to generate -cells in mice rendered diabetic by alloxan treatment. However, not a single bone marroweCderived cell was found to express insulin, Pdx-1, or Nkx6.1, whereas close to 100% of them expressed the blood cell antigens CD45 and Mac-1/Gr-1. It is unclear why we were unable to confirm the observations of Ianus et al. If this is attributable to subtle differences in the experimental protocols, the implication is that the phenomenon of in vivo bone marroweCderived -cell differentiation is not robust. Consequently, the requisite conditions must be much better defined before even considering any clinical applications. However, as already emphasized by others (44), it is also necessary to reinvestigate the experimental approaches, as cells can show considerable autofluorescence and unspecific binding of antibodies. Although our studies provided no support of the hypothesis that fresh bone marrow cells can transdifferentiate into -cells upon in vivo transplantation, they do not exclude that cells with such potential might be derived from cultured bone marrow cells as recently described (47,48). Furthermore, recent studies have suggested that bone marrow transplantation might promote endogenous pancreatic -cell regeneration (35,49).

    ACKNOWLEDGMENTS

    This work was supported by the Juvenile Diabetes Research Foundation, the Swedish Diabetes Foundation, the Swedish Research Council and ALF (Government Public Health Grant). The Lund Stem Cell Center is supported by a center of excellence grant from the Swedish Foundation for Strategic Research.

    We thank L. Wittman for technical assistance.

    FOOTNOTES

    P.S. is employed by, is a stock shareholder of, and receives grant support from Novo Nordisk.

    REFERENCES

    Gussoni E, Soneoka Y, Strickland CD, Buzney EA, Khan MK, Flint AF, Kunkel LM, Mulligan RC: Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature 401:390eC394, 1999

    Petersen BE, Bowen WC, Patrene KD, Mars WM, Sullivan AK, Murase N, Boggs SS, Greenberger JS, Goff JP: Bone marrow as a potential source of hepatic oval cells. Science 284:1168eC1170, 1999

    Mezey E, Chandross KJ, Harta G, Maki RA, McKercher SR: Turning blood into brain: cells bearing neuronal antigens generated in vivo from bone marrow. Science 290:1779eC1782, 2000

    Brazelton TR, Rossi FM, Keshet GI, Blau HM: From marrow to brain: expression of neuronal phenotypes in adult mice. Science 290:1775eC1779, 2000

    Lagasse E, Connors H, Al-Dhalimy M, Reitsma M, Dohse M, Osborne L, Wang X, Finegold M, Weissman IL, Grompe M: Purified hematopoietic stem cells can differentiate into hepatocytes in vivo. Nat Med 6:1229eC1234, 2000

    Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, McKay R, Nadal-Ginard B, Bodine DM, Leri A, Anversa P: Bone marrow cells regenerate infarcted myocardium. Nature 410:701eC705, 2001

    Jackson KA, Majka SM, Wang H, Pocius J, Hartley CJ, Majesky MW, Entman ML, Michael LH, Hirschi KK, Goodell MA: Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells. J Clin Invest 107:1395eC1402, 2001

    Bjornson CR, Rietze RL, Reynolds BA, Magli MC, Vescovi AL: Turning brain into blood: a hematopoietic fate adopted by adult neural stem cells in vivo. Science 283:534eC537, 1999

    Wagers AJ, Sherwood RI, Christensen JL, Weissman IL: Little evidence for developmental plasticity of adult hematopoietic stem cells. Science 297:2256eC2259, 2002

    Castro RF, Jackson KA, Goodell MA, Robertson CS, Liu H, Shine HD: Failure of bone marrow cells to transdifferentiate into neural cells in vivo. Science 297:1299, 2002

    McKinney-Freeman SL, Jackson KA, Camargo FD, Ferrari G, Mavilio F, Goodell MA: Muscle-derived hematopoietic stem cells are hematopoietic in origin. Proc Natl Acad Sci U S A 99:1341eC1346, 2002

    Nygren JM, Jovinge S, Breitbach M, Sawen P, Roll W, Hescheler J, Taneera J, Fleischmann BK, Jacobsen SE: Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nat Med 10:494eC501, 2004

    Anderson DJ, Gage FH, Weissman IL: Can stem cells cross lineage boundaries Nat Med 7:393eC395, 2001

    Morshead CM, Benveniste P, Iscove NN, van der Kooy D: Hematopoietic competence is a rare property of neural stem cells that may depend on genetic and epigenetic alterations. Nat Med 8:268eC273, 2002

    Wang X, Willenbring H, Akkari Y, Torimaru Y, Foster M, Al-Dhalimy M, Lagasse E, Finegold M, Olson S, Grompe M: Cell fusion is the principal source of bone-marrow-derived hepatocytes. Nature 422:897eC901, 2003

    Terada N, Hamazaki T, Oka M, Hoki M, Mastalerz DM, Nakano Y, Meyer EM, Morel L, Petersen BE, Scott EW: Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion. Nature 416:542eC545, 2002

    Alvarez-Dolado M, Pardal R, Garcia-Verdugo JM, Fike JR, Lee HO, Pfeffer K, Lois C, Morrison SJ, Alvarez-Buylla A: Fusion of bone-marrow-derived cells with Purkinje neurons, cardiomyocytes and hepatocytes. Nature 425:968eC973, 2003

    Vassilopoulos G, Wang PR, Russell DW: Transplanted bone marrow regenerates liver by cell fusion. Nature 422:901eC904, 2003

    Ying QL, Nichols J, Evans EP, Smith AG: Changing potency by spontaneous fusion. Nature 416:545eC548, 2002

    Priller J, Persons DA, Klett FF, Kempermann G, Kreutzberg GW, Dirnagl U: Neogenesis of cerebellar Purkinje neurons from gene-marked bone marrow cells in vivo. J Cell Biol 155:733eC738, 2001

    LaBarge MA, Blau HM: Biological progression from adult bone marrow to mononucleate muscle stem cell to multinucleate muscle fiber in response to injury. Cell 111:589eC601, 2002

    Laflamme MA, Myerson D, Saffitz JE, Murry CE: Evidence for cardiomyocyte repopulation by extracardiac progenitors in transplanted human hearts. Circ Res 90:634eC640, 2002

    Krause DS, Theise ND, Collector MI, Henegariu O, Hwang S, Gardner R, Neutzel S, Sharkis SJ: Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell 105:369eC377, 2001

    Quaini F, Urbanek K, Beltrami AP, Finato N, Beltrami CA, Nadal-Ginard B, Kajstura J, Leri A, Anversa P: Chimerism of the transplanted heart. N Engl J Med 346:5eC15, 2002

    Ianus A, Holz GG, Theise ND, Hussain MA: In vivo derivation of glucose-competent pancreatic endocrine cells from bone marrow without evidence of cell fusion. J Clin Invest 111:843eC850, 2003

    Wagers AJ, Weissman IL: Plasticity of adult stem cells. Cell 116:639eC648, 2004

    Camargo FD, Green R, Capetanaki Y, Jackson KA, Goodell MA: Single hematopoietic stem cells generate skeletal muscle through myeloid intermediates. Nat Med 9:1520eC1527, 2003

    Lee VM, Stoffel M: Bone marrow: an extra-pancreatic hideout for the elusive pancreatic stem cell J Clin Invest 111:799eC801, 2003

    Blyszczuk P, Wobus AM: Stem cells and pancreatic differentiation in vitro. J Biotechnol 113:3eC13, 2004

    Weir GC: Can we make surrogate beta-cells better than the original Semin Cell Dev Biol 15:347eC357, 2004

    Hussain MA, Theise ND: Stem-cell therapy for diabetes mellitus. Lancet 364:203eC205, 2004

    Lechner A, Yang YG, Blacken RA, Wang L, Nolan AL, Habener JF: No evidence for significant transdifferentiation of bone marrow into pancreatic -cells in vivo. Diabetes 53:616eC623, 2004

    Kojima H, Fujimiya M, Matsumura K, Nakahara T, Hara M, Chan L: Extrapancreatic insulin-producing cells in multiple organs in diabetes. Proc Natl Acad Sci U S A 101:2458eC2463, 2004

    Choi JB, Uchino H, Azuma K, Iwashita N, Tanaka Y, Mochizuki H, Migita M, Shimada T, Kawamori R, Watada H: Little evidence of transdifferentiation of bone marrow-derived cells into pancreatic beta cells. Diabetologia 46:1366eC1374, 2003

    Hess D, Li L, Martin M, Sakano S, Hill D, Strutt B, Thyssen S, Gray DA, Bhatia M: Bone marrow-derived stem cells initiate pancreatic regeneration. Nat Biotechnol 21:763eC770, 2003

    Hara M, Wang X, Kawamura T, Bindokas VP, Dizon RF, Alcoser SY, Magnuson MA, Bell GI: Transgenic mice with green fluorescent protein-labeled pancreatic beta-cells. Am J Physiol Endocrinol Metab 284:E177eCE183, 2003

    Okabe M, Ikawa M, Kominami K, Nakanishi T, Nishimune Y: ‘Green mice’ as a source of ubiquitous green cells. FEBS Lett 407:313eC319, 1997

    Rerup C, Lundquist I: Blood glucose level in mice. 2. A quantitative study of alloxan diabetes. Acta Endocrinol (Copenh) 54:514eC526, 1967

    Szkudelski T: The mechanism of alloxan and streptozotocin action in B cells of the rat pancreas. Physiol Res 50:537eC546, 2001

    Orlic D, Kajstura J, Chimenti S, Limana F, Jakoniuk I, Quaini F, Nadal-Ginard B, Bodine DM, Leri A, Anversa P: Mobilized bone marrow cells repair the infarcted heart, improving function and survival. Proc Natl Acad Sci U S A 98:10344eC10349, 2001

    Tourrel C, Bailbe D, Meile MJ, Kergoat M, Portha B: Glucagon-like peptide-1 and exendin-4 stimulate -cell neogenesis in streptozotocin-treated newborn rats resulting in persistently improved glucose homeostasis at adult age. Diabetes 50:1562eC1570, 2001

    Li Y, Hansotia T, Yusta B, Ris F, Halban PA, Drucker DJ: Glucagon-like peptide-1 receptor signaling modulates beta cell apoptosis. J Biol Chem 278:471eC478, 2003

    Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO, Rubart M, Pasumarthi KB, Virag JI, Bartelmez SH, Poppa V, Bradford G, Dowell JD, Williams DA, Field LJ: Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature 428:664eC668, 2004

    Chien KR: Stem cells: lost in translation. Nature 428:607eC608, 2004

    Jonsson J, Carlsson L, Edlund T, Edlund H: Insulin-promoter-factor 1 is required for pancreas development in mice. Nature 371:606eC609, 1994

    Sander M, Sussel L, Conners J, Scheel D, Kalamaras J, Dela Cruz F, Schwitzgebel V, Hayes-Jordan A, German M: Homeobox gene Nkx6.1 lies downstream of Nkx2.2 in the major pathway of beta-cell formation in the pancreas. Development 127:5533eC5540, 2000

    Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad M, Du J, Aldrich S, Lisberg A, Low WC, Largaespada DA, Verfaillie CM: Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 418:41eC49, 2002

    Tang DQ, Cao LZ, Burkhardt BR, Xia CQ, Litherland SA, Atkinson MA, Yang LJ: In vivo and in vitro characterization of insulin-producing cells obtained from murine bone marrow. Diabetes 53:1721eC1732, 2004

    Banerjee M, Kumar A, Bhonde RR: Reversal of experimental diabetes by multiple bone marrow transplantation. Biochem Biophys Res Commun 328:318eC325, 2005(Jalal Taneera, Anders Ros)