To clarify the lineage relationship between cells that express the neural stem cell marker nestin and endocrine cells of the pancreas, we analyzed offspring of a cross between mice carrying a nestin promoter/enhancer-driven cre-recombinase (Nestin-cre) and C57BL/6J-Gtrosa26tm1Sor mice that carry a loxP-disrupted β-galactosidase gene (Rosa26). In nestin-cre+/tg;R26RloxP/+ embryos, cre-recombinase was detected in association with nestin-positive cells in the pancreatic mesenchyme with some of the nestin-positive cells lining vascular channels. In postnatal mice, pancreatic β-galactosidase expression was restricted to vascular endothelial cells of the islet and a subset of cells in the muscularis of arteries in a distribution identical to endogenous nestin expression. Ex vivo explants of mouse pancreatic ducts grew dense cultures that costained for nestin and β-galactosidase, demonstrating recombination in vitro. The cultures could be differentiated into complex stereotypic structures that contain nestin- and insulin-expressing cells. Nestin-cre+/tg;R26RloxP/+-derived duct cultures showed that insulin-positive cells were negative for β-galactosidase. These results indicate that both in vivo and in vitro pancreatic endocrine cells arise independently of nestin-positive precursors. The apparent vascular nature of the nestin-positive cell population and the close association with endocrine cells suggest that nestin-positive cells play an important role in the growth and maintenance of the islet.

Interest in developing a system for the in vitro generation of insulin-producing cells from progenitors has led to a search for a self-renewing stem cell population in the pancreas. Nestin, initially identified as a marker of neural stem or progenitor cells (1), has been suggested to be a marker for multipotent pancreatic stem cells of the pancreas (2). Studies have shown that embryonic stem cell–derived cultures enriched for nestin-expressing cells can be differentiated in vitro to cells that produce insulin and glucagon (3), and adult rat and human islet explants enriched for nestin-positive cells can be differentiated to express insulin and glucagon (4). Human islet–derived cultures enriched for nestin-positive cells express the transporters ABCG2 and MDR-1, showing that these cells have properties consistent with a pluripotent stem cell–like population (5). Islet-derived nestin-positive cells also express glucagon-like peptide 1 receptors and treatment with recombinant glucagon-like peptide 1 enhances differentiation of these cultures to express markers consistent with an endocrine phenotype (6).

Although these studies suggest that nestin is a marker for cells destined for a neuroendocrine fate, others have questioned a direct lineage relationship in vivo (7). Insulin- or glucagon-positive cells often appear within or close to duct-like structures during embryogenesis and during pancreatic regeneration after chemical or surgical damage; thus, it is believed that endocrine cells of the islet arise from pancreatic ducts (811). In the mouse, from embryonic day 10.5 to adulthood, markers of pancreatic progenitors such as Pdx1 and Isl1 are expressed in pancreatic epithelial cells that coexpress duct cell markers; however, these cells do not express nestin (12). Recent studies have suggested that adult pancreatic duct progenitors and endocrine cells diverge early during development (13), raising the possibility that nestin marks an endocrine progenitor that is not expressed in ductal epithelium at later stages of development.

Nestin expression is fairly widespread in the developing embryo (1,1418) including vascular endothelial cells (19) and in the adult (2022), especially under conditions that stimulate neoangiogenesis (23). Recent experiments have suggested that signals arising from endothelial cells are critical to the growth of parenchymal cells, including pancreatic endocrine cells during embryogenesis (24,25) and in the adult (26), raising the possibility that there is a relationship between proliferation of nestin-positive cells and tissue grow and regeneration.

A method that has recently become popular to trace the lineage of cell types uses cre-recombinase, driven by a cell type–specific promoter to activate a “floxed” marker gene, such as β-galactosidase or a fluorescent protein, to genetically mark a cell and all its progeny (13,27). To directly determine the fate of nestin-expressing cells and their relationship to islet endocrine cells in vivo and in vitro, we analyzed β-galactosidase expression after activation by nestin-enhancer–driven cre-recombinase in embryos and adult tissue (nestin-cre+/tg;R26RloxP/+). We found that in the embryo, nestin-positive cells contribute to the developing vasculature in the pancreatic mesenchyme. In the postnatal mouse, nestin-positive cells give rise to cells associated with the microcirculation of the islet but do not contribute to other cells in the pancreas, including endocrine cells. In addition, we show that whereas cultures of primary duct explants from the nestin-cre+/tg;R26RloxP/+ mice give rise to nestin-positive cells intimately associated with insulin-positive cells, the nestin-positive cells do not contribute to the endocrine cell formation.

Transgenic mice.

The Institutional Animal Care and Use Committees of the University of Michigan approved all animal procedures. C57BL/6J-Gtrosa26tm1Sor mice (Jackson Laboratories) were mated with nestin-cre mice (a gift from Rudy Jaenisch, Massachusetts Institute of Technology, and Gail Martin, University of California, San Francisco [28]) that carry a 1.8-kb nestin promoter/enhancer fragment from the nestin second intron that drives the expression of cre-recombinase. F1 mice were killed at 3 weeks or 3 months of age, and pancreata were fixed for immunohistochemistry or β-galactosidase activity (29). F1 mouse embryos were harvested at embryonic day 12.5 from pregnant dams and immediately fixed for sectioning and immunohistochemistry as previously described (29).

Mouse insulin promoter (MIP-GFP) mice were generated by pronuclear injection of a construct containing the mouse insulin I promoter (MIP)–green fluorescent protein (GFP) containing an 8.5-kb fragment of MIP that includes a region from −8.5 kb to 12 bp (relative to the transcriptional start site), the coding region of enhanced GFP (0.76 kb) (Clontech, Palo Alto, CA), and a 2.1-kb fragment of the human growth hormone gene cassette (30).

Duct isolation and culture.

Ducts were isolated by collagenase digestion (P2; Boehringer Mannheim) using a protocol described by Bonner-Weir et al. (31). The ducts were minced, plated onto tissue culture dishes (Primeria; Falcon), and cultured in Dulbecco’s modified Eagle’s medium/F12 medium (1:1) with 10% fetal bovine serum and antibiotics at 37°C. After 7–8 days of growth, the cultures were overlaid with 10% Matrigel, which was removed after 24 h, and the cultures were grown for an additional 14–21 days in the same medium.

Immunohistochemistry.

Fixed tissues were sectioned and stained for nestin (1:100 dilution; Pharmingen, San Diego, CA) and β-galactosidase (1:400; Abcam, Cambridge, U.K.) using a Vectastain ABC-Peroxidase Kit (Vector Laboratories, Burlingame, CA). Biotinylated secondary antibodies and horseradish peroxidase–conjugated anti-biotin were used to detect staining and peroxidase activity visualized by applying diaminobenzidine solution. Sections were then counterstained with hematoxylin, dehydrated, cleared, and mounted. Antisera for the detection of cre-recombinase (1:100; Novagen, Madison, WI) and von Willebrand factor (1:200; Abcam) was incubated with whole mount embryos or pancreatic sections, respectively, treated with Vector Antigen Unmasking Solution for 15 min in a microwave oven. After blocking with 5% donkey serum in PBS with 0.1% Triton X-100, the sections were incubated with the antibodies overnight at 4°C. After rinsing, fluorescein-conjugated donkey anti-rabbit secondary antibody (1:200; Molecular Probes, Eugene, OR) was applied for 90 min at room temperature. After washing with PBS, the second step of immunofluorescent staining with mouse primary antibodies was performed using the Mouse on Mouse (M.O.M.) Immunodetection Fluorescein Kit (Vector).

Explant cultures were fixed in 4% paraformaldehyde at room temperature and blocked in 10% normal goat serum, 0.3% Triton X-100. Monoclonal antibodies to nestin (1:100), rabbit anti–β-galactosidase (1:400), guinea pig anti-insulin (1:2,000; Linco Research, St. Charles, MO), rabbit anti-glucagon (1:1,000; Linco), and rabbit anti-NeuroD/BETA2 (1:200; Abcam) were added for 2 h, and binding was detected with fluorescent secondary antibodies (1:400, Alexa-fluor; Molecular Probes). Nuclei were counterstained with 4′,6-diamidino-2-phenylindole dihydrochloride in VectaShield (Vector Laboratories).

X-gal staining.

Pancreata were fixed for 15 min in 0.5% glutaraldehyde solution with PBS, 2 mmol/l MgCl2, and 1.25 mmol/l EGTA for 4 h at 4°C. Tissues were mounted in Opti-tec and frozen, and 15 μmol/l sections were placed on glass slides and stained for 18 h in X-gal solution (PBS containing 5 mmol/l potassium ferrocyanide, 5 mmol/l potassium ferricyanide, and 1 mg/ml X-Gal). Duct cultures were fixed in 0.5% glutaraldehyde for 1 h at room temperature and stained with X-gal solution for 18 h.

To directly determine the fate of cells within the pancreas that at some time expressed nestin, we analyzed offspring of mice resulting from crossing Rosa26 mice (32) with mice that express a nestin promoter/enhancer-driven cre-recombinase (28) (nestin-cre+/tg;R26RloxP/+) (Fig. 1A). In embryonic day 12.5 embryos, we found that nestin and cre-recombinase were coexpressed in the developing heart and small intestine (not shown) and in the ventricular zone of the brain (Fig. 1B, arrow heads). In the developing pancreas, antisera to cre-recombinase showed primarily nuclear staining with a small amount of cytoplasmic staining, whereas antisera to nestin showed fibrous cytoplasmic staining (Fig. 1C). These findings suggest faithful expression of the cre-recombinase driven by the nestin promoter/enhancer element in these tissues. In the pancreatic mesenchyme, nucleated erythrocytes (which autofluoresce at both 514 and 543 nm) were present in vascular channels lined by nestin-positive cells (Fig. 1C, inset, arrow), suggesting that nestin-positive cells comprise part of the vascular endothelium in the developing pancreas. In the absence of primary antisera to cre-recombinase, no specific peri-epithelial staining was observed; however, the ductal epithelium continued to show fluorescence (Fig. 1D).

To determine the postnatal fate of nestin-positive cells, frozen sections of pancreas from Rosa26 mice were developed with 5-bromo-4-chloro-3-indolyl β-d-galactoside (X-Gal). The tissues showed no specific X-gal staining (Fig. 2A), confirming the absence of endogenous β-galactosidase activity. X-gal staining in pancreatic sections of nestin-cre+/tg;R26RloxP/+ mice was intense in small flattened cells dispersed throughout the islet (Fig. 2B and C, arrows) and in a few cells scattered between acini throughout the exocrine pancreas (Fig. 2B, C, F, and G). We examined >50 islets in animals from 3 weeks to 6 months of age from two independent nestin-cre+/tg;R26RloxP/+ crosses and only observed specific X-gal staining in the flattened cells of the islet and never in the larger rounded endocrine cells. The X-gal+ cells comprised ∼7% of the cells in the islet (63/873). β-Galactosidase expression was also detected in cells in the muscularis layer of arterioles near islets (Fig. 2C, arrows) and in the muscularis layer of larger arteries (Fig. 2D and E). The orientation of X-gal stained cells was in a circumferential pattern in the arteries (Fig. 2D), and in some sections, the X-Gal staining appeared in a spiral pattern around the vascular structure as it entered the islet (Fig. 2B and C, arrow heads). An occasional X-gal+ vascular-like structure was noted between acinar lobes of the exocrine pancreas (Fig. 2F), but the majority of the exocrine pancreas vasculature did not stain with X-gal, indicating that nestin-positive precursors do not contribute significantly to smaller vascular structures outside of the islet. X-gal+ cells could be seen in the subepithelial stroma of ducts (Fig. 2E and G, arrows), the identity of which is uncertain but may be important for generation of nestin-positive cells from the in vitro duct culture (see below). X-gal staining was absent from all other structures within the pancreas, including the acini and large and small pancreatic ducts.

Previous work has demonstrated nestin expression in cells within the islet (2), pericytes (33), and muscle precursor cells (16). Immunohistochemical staining of nestin-cre+/tg;R26RloxP/+ pancreas with nestin antisera showed specific staining of cells in the islet (Fig. 3A and B, arrow) and small arteries (Fig. 3A, arrow) identical to that defined by X-gal staining. To define the characteristics of the nestin-positive cells in the islet, we again exploited the autofluorescence of red blood cells (RBCs) to define the vascular channels in the islets. Confocal images of sections stained with anti-nestin antibodies showed fluorescent nestin-positive cells lining channels containing the RBCs (Fig. 3C–F). Electron microscopy showed that in the majority of sections, only one cell separates the granule containing β-cells and erythrocytes (Fig. 3G) and could represent the nestin-positive cell population. Nestin also stained occasional cells in the walls of small arteries in the pancreas (Fig. 3H, arrow), similar to that seen observed with X-gal staining. Although the endothelial cells of larger arteries of the pancreas could be easily detected with antisera to CD31 (not shown) or von Willebrand factor (Fig. 3I, arrows), these antisera only stained an occasional vessel near the base of the islet (Fig. 3I, asterisk). These data suggest that in the islet, nestin-positive cells may represent endothelial cells with unique antigenic properties.

Although these data suggest that nestin lineage cells contribute to the microvasculature of the islet and not endocrine cells, this does not exclude the possibility that nestin-positive cells cultured in vitro may give rise to alternative cell types, as has been suggested previously (3,4). To characterize the in vitro development of endocrine cells from postnatal mouse pancreata, we prepared primary cultures of islets and pancreatic ducts, per the protocol of Bonner-Weir et al. (31), from 8-week-old transgenic mice that express GFP under the control of an 8.5-kb fragment of MIP (30). Primary islet cultures gave rise to cellular outgrowths that demonstrated fluorescence only in the islets themselves (Fig. 4A and B). After 7 days of growth, examination of living cells from MIP-GFP–derived duct cultures showed no GFP fluorescence in the cells growing from the duct fragment, indicating that contaminating β-cells are not present in the initial duct cultures (Fig. 4C and D). The cells were then treated with Matrigel to induce differentiation, and during the next 7–10 days, the cultures coalesced into bands of densely packed cells (Fig. 4E–H) and eventually formed nodes of even higher cellular density (Fig. 4E–H). The cells in the nodes showed GFP fluorescence (Fig. 4B and D), indicating activation of the insulin promoter. After 14–21 days in culture, GFP+ cell masses in the nodes, termed “islet-like structures,” migrated into the relatively acellular areas or grew on top of the dense cell masses (Fig. 4I–J). Frequent observation of the cultures showed that multiple islet-like structures appear to bud from individual nodes. Examination of Matrigel-differentiated duct-derived cultures from wild-type mice showed no autofluorescence (not shown). These results show that, under the culture conditions used in these studies, differentiation of presumptive endocrine precursors occurs within nodes of higher cell density in a stereotypic manner.

To further define the relationship between nestin-positive cells and endocrine cells developing in vitro, we performed a series of costaining experiments. Duct explant cultures from wild-type mice gave rise to cells, of which ∼30–50% were positive for nestin (not shown). These cultures formed identical dense cell structures after Matrigel treatment and islet-like structures within and extruding from the condensed bands of cells (Fig. 5). The islet-like structures in the nodes immunostained for both insulin and glucagon (Fig. 5A and B) with the insulin-positive cells (Fig. 5B, insert, arrow head) surrounding the lower number of glucagon-positive cells (Fig. 5B, insert, arrow). Nestin expression was always observed in the condensed cell areas and formed a lattice around the developing endocrine cells (Fig. 5C and D, arrow); however, no costaining of nestin and insulin was detected. When stained with antisera to the endocrine cell transcription factor NeuroD/BETA2, differentiated duct-derived cultures showed nuclear immunoreactivity only in nodes (not shown) and in islet-like structures (Fig. 5E–H). Nestin antisera stained elongated cells that were intermeshed between the NeuroD/BETA2+ cells, but, again, there did not appear to be overlap in staining (Fig. 5H).

To further define the role of nestin-positive cells in the generation of insulin-producing cells in vitro, we prepared primary duct cultures of either Rosa26 or nestin-cre+/tg;R26RloxP/+ mice. During the initial growth phase, filamentous nestin staining could be detected in all β-galactosidase–positive cells grown from nestin-cre+/tg;R26RloxP/+ (Fig. 6A and B), indicating that under these culture conditions, there is accurate recombination of the β-galactosidase gene by nestin-driven cre-recombinase in vitro. Nestin-cre+/tg;R26RloxP/+ and Rosa26-derived duct cultures could be differentiated to form condensed cell areas, nodes, and islet-like structures in a manner identical to cultures from wild-type and MIP-GFP mice (Fig. 6CF). When nodes containing islet-like structures were stained for β-galactosidase and insulin, the β-galactosidase–positive cells could be seen coursing through the islet-like structure (Fig. 6C, arrow), but there was no costaining with insulin, showing that the insulin-positive cells did not arise from the nestin-positive cell population. Identical results were observed using sensitive X-gal staining of cultures. β-Galactosidase–positive cells were scattered through the condensed cell areas (Fig. 6D) and in a small number of cells in the islet-like structure (Fig. 6F) in the same pattern as seen in duct-derived cultures stained for nestin (Fig. 5) and β-galactosidase (Fig. 6C). No X-gal staining was observed in cultures from Rosa26 mice (Fig. 6E).

Our data indicate that, in vivo, the intermediate filament protein nestin is expressed only in cells destined to become endothelial cells of the islet vasculature, a subset of cells residing in the wall of arteries and scattered cells in the stroma surrounding the ductal/vascular bundles of the pancreas. This result depends on accurate recombination by the nestin-driven cre-recombinase during embryogenesis and postnatal development. Although we cannot completely rule out alternative nestin promoter/enhancer use in potential endocrine precursor cells, several lines of evidence suggest that the lineage tracing that we observe accurately reflects the in vivo situation. First, we observe transgenic cre-recombinase coexpression in nestin-positive cells in the pancreatic mesenchyme and in the central nervous system (CNS), heart, and intestine, similar to what has been described for nestin expression (1,1418). Second, identical patterns of expression of nestin and β-galactosidase are seen in the pancreas of mature nestin-cre+/tg;R26RloxP/+ mice. Third, in primary pancreatic duct cultures, there is coexpression of nestin and β-galactosidase in the cells grown from these cultures. Thus, we believe that the lack of β-galactosidase expression in pancreatic cells outside of the observed nestin-positive cells, together with earlier studies (12), provide direct evidence that, in vivo, the endocrine cells of the islet arise from non–nestin-positive cells.

Nestin is expressed in the vascular endothelium of a wide variety of tissues during fetal development (23) and is expressed in a subset of cells in the adult animal, including pericytes (33) and astrocytes (34) in the brain, and in stellate cells of the pancreas (5) and liver (35). Nestin expression rises in response to damage in the CNS (23), the liver (35), and the pancreas (36) and is associated with regenerative responses in these tissues. These observations, along with the close association of mesenchymal nestin-positive cells with the pancreatic epithelium during development and our finding of an association during the generation of endocrine cells in vitro, suggest that there is a functional relationship between nestin-positive cells and the growth and differentiation of the endocrine cells in the pancreas. The vascular nature of the nestin-positive cells in the pancreatic mesenchyme and in the mature pancreatic islet is intriguing in context of a recent study demonstrating important inductive signals arising from vascular endothelial cells that are important for endocrine cell development (24). In this study, misexpression of the vascular endothelial growth factor results in localized hypervascularization and ectopic production of insulin-expressing cells. It is evident that the vascular endothelium is important in the growth and differentiation in a number of other tissues including the liver (25), fat (26), and CNS (37). In the latter study, hormonal stimulation of neuronal precursor cells in the avian CNS results in secretion of vascular growth factors. The expanded vasculature in turn secretes factors important for the growth and development of neurons. The finding that nestin is expressed exclusively in the islet microvasculature raises the possibility that these cells participate in sensing the environment of the islet and generate signals to induce the growth of the islet in response to physiological stimuli such as insulin resistance or pregnancy (38). An increase in the islet vasculature has been observed in association with conditions resulting in an expanded islet mass (39,40). In recent studies, we have observed a marked increase in the number and proliferation of nestin-positive cells in the islets of insulin-resistant mice and rats associated with islet hyperplasia but a decline in association with islet failure (M.K.T., C.L.D.-L, C.F.B., unpublished data).

In our studies, CD34 or von Willebrand factor antiserum inconsistently stains the vasculature of the islet, whereas the vasculature of the exocrine pancreas is easily detected by these antisera (data not shown). This result has been reported by others (41,42), providing further evidence for islet microvasculature specialization. Although this study suggests that nestin-positive cells line the vascular channels in the embryonic mesenchyme and in the adult islet, it is possible that nestin also marks other cells, such as pericytes (33,36), within the pancreas. The role of the nestin-positive cells in the walls of the arteries and arterioles is more difficult to understand; however, these cells too may subserve a specific role in islet physiology. The vasculature of the islet apparently responds to environmental influences in a manner that is distinct from that of the exocrine pancreas and regulates blood flow independent of that for the entire pancreas (43). An alternative explanation for these findings is that the vascular nestin-positive population may serve as a reservoir for neovascular growth of the islet during times of increased physiological demand. Further study will determine the exact nature of these cells.

These studies demonstrate a reproducible and inducible developmental process in primary duct cultures that results in the production of both glucagon- and insulin-expressing cells. The generation of these islet-like structures occurs in the context of a network of nestin-positive cells, but nestin-positive cells are not sufficient for islet formation. The finding that nestin-positive cells do not give rise to endocrine cells in vitro is in contrast to findings in earlier studies on nestin-enriched cultures (3,4). Others have suggested that these cells can develop into insulin-secreting cells along a route similar to neurons in the CNS that can express insulin (7,12). The finding of an intimate relationship between cultures enriched for nestin-positive cells and the development of endocrine cells in vitro may be due to the need for signals from the nestin-positive population to allow endocrine cells to develop from precursors present in the cultures. However, we cannot rule out the possibility that under different culture conditions, the nestin-positive population may show more inherent plasticity than under the conditions used in our system.

Finally, a recent publication has generated considerable interest in the possibility of artifacts in identifying “β-cells” grown in in vitro systems (44). We believe that the present study provides strong evidence that there is reliable endocrine differentiation occurring in our system. First, and most importantly, the identification of insulin gene activation in duct-derived cultures from MIP-GFP mice does not rely on antibodies for identification, eliminating the possibility of staining artifact or the possibility of the cells accumulating insulin from the media. This 8.5-kb promoter fragment appears to not be “leaky,” in that only β-cells in vivo show GFP expression. Second, we also observe nuclear staining of NeuroD/BETA2 only in the cellular areas that show insulin staining: in the nodes and in the islet-like structures (and this staining is nuclear and not cytoplasmic). Third, insulin staining is only observed in the same structures that show GFP fluorescence. Finally, we can find islet-like structures that stain for both insulin and glucagon, making nonspecific staining unlikely.

In summary, we show that both in vivo and in vitro, endocrine cell development does not depend on a nestin-positive lineage. However, on the basis of these and earlier studies, we propose that the nestin-positive cells are specialized vascular cells that are important for islet formation in vivo and in vitro by providing signals to true endocrine precursors, similar to what has been proposed to occur in vivo (24) for the pancreas and other tissues (25,26,37,45). Understanding the regulation of growth and development of the nestin-positive population will provide insights into the physiological regulation of islet growth in vivo and provide a tool for use in the development of insulin-producing cells in vitro.

FIG. 1.

Cre-recombinase expression in mouse embryos. A: Schematic of genomic recombination in cre-expressing cells showing the mechanism of activation of β-galactosidase expression. B and C: Sections of embryonic day 12.5 embryos were costained with specific primary antisera and developed with fluorescent secondary antibodies to detect cre-recombinase (red) and nestin (green). Cre-recombinase stained nuclei in the periventricular region (B, arrow heads) in association with multiple nestin-positive cells are shown. C: In the developing pancreas, antisera to nestin stained multiple cells outside of the ductal epithelium (DE) (outlined), whereas cre-recombinase staining was detected primarily in nuclei. Most cells in the mesenchyme showed nestin expression apposed to cre-recombinase–positive nuclei. D: Section of embryonic day 12.5 embryo stained with specific antisera to nestin alone and developed with both anti-mouse (green) an anti-rabbit (red) fluorescent secondary antibodies.

FIG. 1.

Cre-recombinase expression in mouse embryos. A: Schematic of genomic recombination in cre-expressing cells showing the mechanism of activation of β-galactosidase expression. B and C: Sections of embryonic day 12.5 embryos were costained with specific primary antisera and developed with fluorescent secondary antibodies to detect cre-recombinase (red) and nestin (green). Cre-recombinase stained nuclei in the periventricular region (B, arrow heads) in association with multiple nestin-positive cells are shown. C: In the developing pancreas, antisera to nestin stained multiple cells outside of the ductal epithelium (DE) (outlined), whereas cre-recombinase staining was detected primarily in nuclei. Most cells in the mesenchyme showed nestin expression apposed to cre-recombinase–positive nuclei. D: Section of embryonic day 12.5 embryo stained with specific antisera to nestin alone and developed with both anti-mouse (green) an anti-rabbit (red) fluorescent secondary antibodies.

Close modal
FIG. 2.

Vascular nature of nestin-derived cells in the pancreas of mice. A: X-gal staining of a 3-week-old Rosa26 mouse showing no specific X-gal activity. B and F: X-gal staining of pancreatic sections of 3-week-old nestin-cre+/tg;R26RloxP/+. B and C: X-gal staining of pancreatic section staining of a small number of flattened cells within the islet (arrows) and in the walls of arterioles (arrow heads) that show a spiral nature of the stained cell (C). E and F: X-gal staining of a subset of cells in the muscularis layer of a small artery in the cross section (D) and longitudinal section (E). Note the staining of cells beneath the ductal epithelium (E, arrow). F: X-gal staining of a small vascular structure in the exocrine pancreas; note the relative absence of staining in the majority of the exocrine pancreas. G: Immunohistochemistry with anti–β-galactosidase antibodies and diaminobenzidine development (brown staining) demonstrating a number of nestin-positive cells in the stroma of a pancreatic duct (arrows). Note the absence of β-galactosidase staining in the ductal epithelium.

FIG. 2.

Vascular nature of nestin-derived cells in the pancreas of mice. A: X-gal staining of a 3-week-old Rosa26 mouse showing no specific X-gal activity. B and F: X-gal staining of pancreatic sections of 3-week-old nestin-cre+/tg;R26RloxP/+. B and C: X-gal staining of pancreatic section staining of a small number of flattened cells within the islet (arrows) and in the walls of arterioles (arrow heads) that show a spiral nature of the stained cell (C). E and F: X-gal staining of a subset of cells in the muscularis layer of a small artery in the cross section (D) and longitudinal section (E). Note the staining of cells beneath the ductal epithelium (E, arrow). F: X-gal staining of a small vascular structure in the exocrine pancreas; note the relative absence of staining in the majority of the exocrine pancreas. G: Immunohistochemistry with anti–β-galactosidase antibodies and diaminobenzidine development (brown staining) demonstrating a number of nestin-positive cells in the stroma of a pancreatic duct (arrows). Note the absence of β-galactosidase staining in the ductal epithelium.

Close modal
FIG. 3.

Nestin and von Willebrand factor expression in the pancreas. A and B: Expression of nestin in islets using anti-nestin antisera and developed with diaminobenzidine. Original magnification ×10. A: Flattened cells within the islet and small arteriole (arrow) are stained. B: Control sera. C and F: Confocal fluorescence microscopy demonstrating intimate association of erythrocytes (RBCs) with nestin-positive cells in islets. The fixed pancreas was stained with nestin and developed with Alexafluor-labeled secondary antibody. When excited with an argon laser at 488 nm or a helium-neon laser at 543 nm, typical autofluorescence of RBCs (arrows) was observed as green and red, respectively. Red fluorescence of nestin was also seen at 543 nm in B and D. Diffusion interference contrast image demonstrates apparent vascular channels (E). Merged images show the RBCs as orange objects and the red staining nestin-positive cells in intimate association (arrows) (D). G: Transmission electron microscope image of islet. C, endothelial cell; E, endocrine cell; R, RBC. Original magnification ×10,500. H: Nestin staining of small arteries showing only a subset of cells in the muscularis area are stained with diaminobenzidine. I: Staining of vascular endothelial cells with antisera to von Willebrand factor (red, arrows). Only an occasional endothelial cell within islets is positive (*).

FIG. 3.

Nestin and von Willebrand factor expression in the pancreas. A and B: Expression of nestin in islets using anti-nestin antisera and developed with diaminobenzidine. Original magnification ×10. A: Flattened cells within the islet and small arteriole (arrow) are stained. B: Control sera. C and F: Confocal fluorescence microscopy demonstrating intimate association of erythrocytes (RBCs) with nestin-positive cells in islets. The fixed pancreas was stained with nestin and developed with Alexafluor-labeled secondary antibody. When excited with an argon laser at 488 nm or a helium-neon laser at 543 nm, typical autofluorescence of RBCs (arrows) was observed as green and red, respectively. Red fluorescence of nestin was also seen at 543 nm in B and D. Diffusion interference contrast image demonstrates apparent vascular channels (E). Merged images show the RBCs as orange objects and the red staining nestin-positive cells in intimate association (arrows) (D). G: Transmission electron microscope image of islet. C, endothelial cell; E, endocrine cell; R, RBC. Original magnification ×10,500. H: Nestin staining of small arteries showing only a subset of cells in the muscularis area are stained with diaminobenzidine. I: Staining of vascular endothelial cells with antisera to von Willebrand factor (red, arrows). Only an occasional endothelial cell within islets is positive (*).

Close modal
FIG. 4.

Development of insulin-expressing cells in vitro from duct-derived cultures. Pancreata from mice expressing GFP driven by the insulin promoter were digested with collagenase, and pancreatic islets or duct fragments were cultured. After 7 days, some of the cultures were treated with Matrigel for 24 h, after which they were incubated in Dulbecco’s modified Eagle’s medium/F12 with 10% FCS for an additional 21 days. Fluorescence of each culture was photographed before fixation at 489 nm. A and B: Islets cultured for 7 days showing primary cell outgrowths are GFP+. C and J: Primary outgrowths from cultured ducts. After 7 days, no fluorescence was seen in the primary duct cultures (C and D). After 7 days of culture, dense bands of cells could be seen, which showed minimal evidence of insulin expression (E and F). After 10–17 days in cultures, the dense cell areas formed nodes that began expressing insulin (G and H). Finally after 21 days, islet-like structures began budding from the nodes (I and J). d, dense cell areas; ils, islet-like structure; n, nodes.

FIG. 4.

Development of insulin-expressing cells in vitro from duct-derived cultures. Pancreata from mice expressing GFP driven by the insulin promoter were digested with collagenase, and pancreatic islets or duct fragments were cultured. After 7 days, some of the cultures were treated with Matrigel for 24 h, after which they were incubated in Dulbecco’s modified Eagle’s medium/F12 with 10% FCS for an additional 21 days. Fluorescence of each culture was photographed before fixation at 489 nm. A and B: Islets cultured for 7 days showing primary cell outgrowths are GFP+. C and J: Primary outgrowths from cultured ducts. After 7 days, no fluorescence was seen in the primary duct cultures (C and D). After 7 days of culture, dense bands of cells could be seen, which showed minimal evidence of insulin expression (E and F). After 10–17 days in cultures, the dense cell areas formed nodes that began expressing insulin (G and H). Finally after 21 days, islet-like structures began budding from the nodes (I and J). d, dense cell areas; ils, islet-like structure; n, nodes.

Close modal
FIG. 5.

Expression of endocrine cell markers and nestin in differentiating duct cultures. Cultures were grown for 7 days and treated with Matrigel. After 3 weeks, the cultures were fixed and immunostained. Brightfield (A) and immunostaining for insulin (B) (red, arrowhead in insert) and glucagon (green, arrow in insert) in a developing islet-like structure. Brightfield (C) and immunostained image showing insulin-positive cells (D) (red, arrow) in a meshwork of nestin-positive cells (green). A-D: Original magnification ×10, except in insert in B, which is ×20. E and F: NeuroD/BETA2 and nestin staining demonstrate the nestin-positive cells (red) enmeshed islet-like structures in which there is the nuclei stain for NeuroD/BETA2 (green). G: Nuclei counterstained with diaminobenzidine (blue). H: Images in E-G merged showing intercalation of nestin-positive cell in the NeuroD/BETA2+ cell cluster. E-H: Original magnification ×20.

FIG. 5.

Expression of endocrine cell markers and nestin in differentiating duct cultures. Cultures were grown for 7 days and treated with Matrigel. After 3 weeks, the cultures were fixed and immunostained. Brightfield (A) and immunostaining for insulin (B) (red, arrowhead in insert) and glucagon (green, arrow in insert) in a developing islet-like structure. Brightfield (C) and immunostained image showing insulin-positive cells (D) (red, arrow) in a meshwork of nestin-positive cells (green). A-D: Original magnification ×10, except in insert in B, which is ×20. E and F: NeuroD/BETA2 and nestin staining demonstrate the nestin-positive cells (red) enmeshed islet-like structures in which there is the nuclei stain for NeuroD/BETA2 (green). G: Nuclei counterstained with diaminobenzidine (blue). H: Images in E-G merged showing intercalation of nestin-positive cell in the NeuroD/BETA2+ cell cluster. E-H: Original magnification ×20.

Close modal
FIG. 6.

Growth and differentiation of nestin/β-galactosidase-positive duct-derived cultures from Rosa26 or nestin-cre+/tg;R26RloxP/+ mice. A and B: Immunofluorescent costaining of duct-derived cultures after 7 days of growth and before differentiation for β-galactosidase (red) and nestin (green) show a high degree of correlation of expression. Note the filamentous nestin staining in the more diffuse β-galactosidase-stained projection (B, arrow). C: Immunofluorescent costaining of duct-derived cultures after 7 days of growth and 14 days of differentiation for β-galactosidase (red) and insulin (green). Note web of β-galactosidase-positive staining in the insulin-positive cell cluster. D-F: X-gal staining of differentiated cultures from nestin-cre+/tg;R26RloxP/+ mice (D and F) or Rosa26 mouse (E) shows that X-gal staining is positive in only a subset of cells in the nodes (n) of the condensed cell areas (arrow) and is largely excluded from forming islet-like structures (ils) (F). No X-gal staining is seen in the islet-like structure from the duct-derived cultures of the Rosa26 mouse. A and D: Original magnification ×10. B, C, E, and F: Original magnification ×20.

FIG. 6.

Growth and differentiation of nestin/β-galactosidase-positive duct-derived cultures from Rosa26 or nestin-cre+/tg;R26RloxP/+ mice. A and B: Immunofluorescent costaining of duct-derived cultures after 7 days of growth and before differentiation for β-galactosidase (red) and nestin (green) show a high degree of correlation of expression. Note the filamentous nestin staining in the more diffuse β-galactosidase-stained projection (B, arrow). C: Immunofluorescent costaining of duct-derived cultures after 7 days of growth and 14 days of differentiation for β-galactosidase (red) and insulin (green). Note web of β-galactosidase-positive staining in the insulin-positive cell cluster. D-F: X-gal staining of differentiated cultures from nestin-cre+/tg;R26RloxP/+ mice (D and F) or Rosa26 mouse (E) shows that X-gal staining is positive in only a subset of cells in the nodes (n) of the condensed cell areas (arrow) and is largely excluded from forming islet-like structures (ils) (F). No X-gal staining is seen in the islet-like structure from the duct-derived cultures of the Rosa26 mouse. A and D: Original magnification ×10. B, C, E, and F: Original magnification ×20.

Close modal

This work was supported by a grant from the American Diabetes Association (C.F.B.) and U.S. Public Health Service Grants DK-20595 and DK-61245 (M.H.) and KO8HD40288 (D.M.M.).

The authors thank Lanjing Zhang for technical assistance.

1.
Len Dahl U, Zimmerman LB, McKay RD: CNS stem cells express a new class of intermediate filament protein.
Cell
60
:
585
–595,
1990
2.
Hunziker E, Stein M: Nestin-expressing cells in the pancreatic islets of Langerhans.
Biochem Biophys Res Commun
271
:
116
–119,
2000
3.
Lumens N, Blondel O, Laeng P, Velasco I, Ravin R McKay R: Differentiation of embryonic stem cells to insulin-secreting structures similar to pancreatic islets.
Science
292
:
1389
–1394,
2001
4.
Zulewski H, Abraham EJ, Gerlach MJ, Daniel PB, Moritz W, Muller B, Vallejo M, Thomas MK, Habener JF: Multipotential nestin-positive stem cells isolated from adult pancreatic islets differentiate ex vivo into pancreatic endocrine, exocrine, and hepatic phenotypes.
Diabetes
50
:
521
–533,
2001
5.
Lechner A, Leech CA, Abraham EJ, Nolan AL, Habener JF: Nestin-positive progenitor cells derived from adult human pancreatic islets of Langerhans contain side population (SP) cells defined by expression of the ABCG2 (BCRP1) ATP-binding cassette transporter.
Biochem Biophys Res Commun
293
:
670
–674,
2002
6.
Abraham EJ, Leech CA, Lin JC, Zulewski H, Habener JF: Insulinotropic hormone glucagon-like peptide-1 differentiation of human pancreatic islet-derived progenitor cells into insulin-producing cells.
Endocrinology
143
:
3152
–3161,
2002
7.
Edlund H: Organogenesis: pancreatic organogenesis developmental mechanisms and implications for therapy.
Nat Rev Genet
3
:
524
–532,
2002
8.
Edlund H: Pancreas: how to get there from the gut?
Curr Opin Cell Biol
11
:
663
–668,
1999
9.
Gu D, Arnush M, Sarvetnick N: Endocrine/exocrine intermediate cells in streptozotocin-treated Ins-IFN-gamma transgenic mice.
Pancreas
15
:
246
–250,
1997
10.
Bonner-Weir S, Stubbs M, Reitx P, Taneja M, Smith FE: Partial pancreatectomy as a model of pancreatic regeneration. In
Pancreatic Growth and Regeneration.
Sarvetnick N, Ed. Basel, Karger Landes Systems,
1997
, p.
138
–153
11.
Teitelman G, Lee JK, Alpert S: Expression of cell type-specific markers during pancreatic development in the mouse: implications for pancreatic cell lineages.
Cell Tissue Res
250
:
435
–439,
1987
12.
Selander L, Edlund H: Nestin is expressed in mesenchymal and not epithelial cells of the developing mouse pancreas.
Mech Dev
113
:
189
–192,
2002
13.
Gu G, Dubauskaite J, Melton DA: Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors.
Development
129
:
2447
–2457,
2002
14.
Mujtaba T, Mayer-Proschel M, Rao MS: A common neural progenitor for the CNS and PNS.
Dev Biol
200
:
1
–15,
1998
15.
Sejersen T, Lendahl U: Transient expression of the intermediate filament nestin during skeletal muscle development.
J Cell Sci
106
:
1291
–1300,
1993
16.
Kachinsky AM, Dominov JA, Miller JB: Myogenesis and the intermediate filament protein, nestin.
Dev Biol
165
:
216
–228,
1994
17.
Frojdman K, Pelliniemi LJ, Lendahl U, Virtanen I, Eriksson JE: The intermediate filament protein nestin occurs transiently in differentiating testis of rat and mouse.
Differentiation
61
:
243
–249,
1997
18.
Yang J, Bian W, Gao X, Chen L, Jing N: Nestin expression during mouse eye and lens development.
Mech Dev
94
:
287
–291,
2000
19.
Mokry J, Nemecek S: Cerebral angiogenesis shows nestin expression in endothelial cells.
Gen Physiol Biophys
18 (Suppl. 1)
:
25
–29,
1999
20.
Mokry J, Nemecek S: Immunohistochemical detection of intermediate filament nestin.
Acta Medica
41
:
73
–80,
1998
21.
Eaker EY, Sallustio JE: The distribution of novel intermediate filament proteins defines subpopulations of myenteric neurons in rat intestine.
Gastroenterology
107
:
666
–674,
1994
22.
Vaittinen S, Lukka R, Sahlgren C, Rantanen J, Hurme T, Lendahl U, Eriksson JE, Kalimo H: Specific and innervation-regulated expression of the intermediate filament protein nestin at neuromuscular and myotendinous junctions in skeletal muscle.
Am J Pathol
154
:
591
–600,
1999
23.
Mokry J, Nemecek S: Angiogenesis of extra- and intraembryonic blood vessels is associated with expression of nestin in endothelial cells.
Folia Biol (Praha)
44
:
155
–161,
1998
24.
Lammert E, Cleaver O, Melton D: Induction of pancreatic differentiation by signals from blood vessels.
Science
294
:
564
–567,
2001
25.
Matsumoto K, Yoshitomi H, Rossant J, Zaret KS: Liver organogenesis promoted by endothelial cells prior to vascular function.
Science
294
:
559
–563,
2001
26.
Rupnick MA, Panigrahy D, Zhang CY, Dallabrida SM, Lowell BB, Langer R, Folkman MJ: Adipose tissue mass can be regulated through the vasculature.
Proc Natl Acad Sci U S A
99
:
10730
–10735,
2002
27.
Kawaguchi A, Miyata T, Sawamoto K, Takashita N, Murayama A, Akamatsu W, Ogawa M, Okabe M, Tano Y, Goldman SA, Okano H: Nestin-EGFP transgenic mice: visualization of the self-renewal and multipotency of CNS stem cells.
Mol Cell Neurosci
17
:
259
–273,
2001
28.
Trumpp A, Depew MJ, Rubenstein JL, Bishop JM, Martin GR: Cre-mediated gene inactivation demonstrates that FGF8 is required for cell survival and patterning of the first branchial arch.
Genes Dev
13
:
3136
–3148,
1999
29.
Martin DM, Skidmore JM, Fox SE, Gage PJ Camper SA: Pitx2 distinguishes subtypes of terminally differentiated neurons in the developing mouse neuroepithelium.
Dev Biol
252
:
84
–99,
2002
30.
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
:
E177
–E183,
2003
31.
Bonner-Weir S, Taneja M, Weir GC, Tatarkiewicz K, Song KH, Sharma A, O’Neil JJ: In vitro cultivation of human islets from expanded ductal tissue.
Proc Natl Acad Sci U S A
97
:
7999
–8004,
2000
32.
Soriano P: Generalized lacZ expression with the ROSA26 Cre reporter strain.
Nat Genet
21
:
70
–71,
1999
33.
Alliot F, Rutin J, Leenen PJ, Pessac B: Pericytes and periendothelial cells of brain parenchyma vessels co-express aminopeptidase N, aminopeptidase A, and nestin.
J Neurosci Res
58
:
367
–378,
1999
34.
Clarke SR, Shetty AK, Bradley JL, Turner DA: Reactive astrocytes express the embryonic intermediate neurofilament nestin.
Neuroreport
5
:
1885
–1888,
1994
35.
Messing A: Nestin in the liver: lessons from the brain.
Hepatology
29
:
602
–603,
1999
36.
Lardon J, Rooman I, Bouwens L: Nestin expression in pancreatic stellate cells and angiogenic endothelial cells.
Histochem Cell Biol
117
:
535
–540,
2002
37.
Louissaint A Jr, Rao S, Leventhal C, Goldman SA: Coordinated interaction of neurogenesis and angiogenesis in the adult songbird brain.
Neuron
34
:
945
–960,
2002
38.
Bonner-Weir S: Regulation of pancreatic beta-cell mass in vivo.
Recent Prog Horm Res
49
:
91
–104,
1994
39.
Duvillie B, Currie C, Chrones T, Bucchini D, Jami J, Joshi RL, Hill DJ: Increased islet cell proliferation, decreased apoptosis, and greater vascularization leading to beta-cell hyperplasia in mutant mice lacking insulin.
Endocrinology
143
:
1530
–1537,
2002
40.
Mizuno A, Noma Y, Kuwajima M, Murakami T, Zhu M, Shima K: Changes in islet capillary angioarchitecture coincide with impaired B-cell function but not with insulin resistance in male Otsuka-Long-Evans-Tokushima fatty rats: dimorphism of the diabetic phenotype at an advanced age.
Metabolism
48
:
477
–483,
1999
41.
Watanabe H, Sumi S, Urushihata T, Kitamura Y, Iwasaki S, Xu G, Yano S, Nio Y, Tamura K: Immunohistochemical studies on vascular endothelial growth factor and platelet endothelial cell adhesion molecule-1/CD-31 in islet transplantation.
Pancreas
21
:
165
–173,
2000
42.
Mattsson G, Carlsson PO, Olausson K, Jansson L: Histological markers for endothelial cells in endogenous and transplanted rodent pancreatic islets.
Pancreatology
2
:
155
–162,
2002
43.
Jansson L, Carlsson PO, Jansson L, Carlsson P-O: Islet vasculature.
Diabetologia
45
:
749
–763,
2002
44.
Rajagopal J, Anderson WJ, Kume S, Martinez OI, Melton DA: Insulin staining of ES cell progeny from insulin uptake.
Science
299
:
363
,
2003
45.
Song H, Stevens CF, Gage FH: Astroglia induce neurogenesis from adult neural stem cells.
Nature
417
:
39
–44,
2002