Emerging technologies in regenerative medicine have the potential to restore the β-cell compartment in diabetic patients, thereby overcoming the inadequacies of current treatment strategies and organ supply. Novel approaches include: 1) Encapsulation technology that protects islet transplants from host immune surveillance; 2) stem cell therapies and cellular reprogramming, which seek to regenerate the depleted β-cell compartment; and 3) whole-organ bioengineering, which capitalizes on the innate properties of the pancreas extracellular matrix to drive cellular repopulation. Collaborative efforts across these subfields of regenerative medicine seek to ultimately produce a bioengineered pancreas capable of restoring endocrine function in patients with insulin-dependent diabetes.

Regenerative medicine promises to contribute to the advancement of β-cell replacement strategies through the development and implementation of microencapsulation technology, the production of insulin-producing cells either by regeneration from endogenous cells or reprogramming from nonendocrine adult cell sources, and, more recently, the exploitation of bioengineered microenvironments (1). Considering the shortage of available pancreata, the search for alternative cell sources has recently been categorized within one of the following three “Rs” of pancreatic β-cell replenishment: replacement (from stem cells or xenoislets), regeneration (from endogenous progenitor cells), and reprogramming (from nonendocrine adult cell sources) (2).

The purpose of this article is to comprehensively and critically review the main regenerative medicine−based strategies that are currently being developed to treat type 1 diabetes. The first part of the article will illustrate and discuss islet encapsulation technology, which has been extensively investigated over the past 40 years (3) and represents one of the most promising regenerative medicine−based approaches to achieve immunoisolation of transplantable organs. The second part will focus on cell bioengineering, where different types of progenitor and differentiated cell sources are manipulated to ultimately yield insulin-producing cells. The last part of the article will summarize the most recent advancements in the study of the pancreatic extracellular matrix (ECM) as a platform for endocrine pancreas bioengineering.

Encapsulation technology has been used to protect islets from the host immune system. This strategy holds the promise to allow implantation of islets without immunosuppression not only across genetically different individuals from the same species, but also across species barriers.

Microencapsulation

The first approach to immunoisolate cells consists of the microencapsulation of one to three islets per semipermeable immunoprotective capsule. The spherical configuration of these microcapsules results in a higher surface-to-volume ratio and a higher diffusion rate (4). Furthermore, microcapsules can be injected in large numbers, are durable, and are difficult to disrupt mechanically (5,6). In this area, alginate is and has certainly been the most commonly used polymer to encapsulate islet cells.

Allogeneic Human Islet Microencapsulation

There have been sporadic reports of patients receiving alginate microencapsulated human islets during the last two decades. Soon-Shiong (7) reported on one patient in 1994 who stopped insulin treatment for 9 months concomitant with production of C-peptide (from 0.1 to 0.6 ng/mL) and a decline in HbA1c level from 9.3 to 7.6% following receipt of microencapsulated islets. In 2009, Tuch et al. (8) reported four patients with single or multiple microencapsulated human islets infusions into the peritoneum without significant correction of glycemia or insulin needs, but urinary C-peptide was detectable up to 2.5 years later in one patient. A humoral response was induced and cytotoxic antibodies were found in recipient sera 4−8 weeks following infusion associated with necrosis of the islets at 16 months.

More recently in 2011, Calafiore and colleagues (9) similarly reported four patients who received single or multiple alginate microencapsulated islets infusions with a discrete correction of HbA1c level (−1%) and decrease in exogenous insulin needs. A C-peptide response was evidenced during basal assessment and after stimulation and no alloantibodies were detected during a follow-up of 3 years.

These clinical reports have demonstrated that intraperitoneally infused microencapsulated human islets can be considered safe for up to 3 years (8,9). Although insulin independence was not achieved, glycemic control was slightly improved, with reduction of insulin daily requirements in some cases. The main question remains as to why such a technique has not been broadly used and expanded in well-controlled clinical studies in order to determine whether microencapsulation could be an efficient way to graft human islets without the use of immunosuppression. The lack of human pancreas cells certainly plays a major role and it is not obvious or ethical to use rare human islets in uncertain clinical pilot studies, especially when 1) preclinical microencapsulation studies in large animal models are lacking and 2) solid results have been obtained in some clinical studies with human islet transplantation following the Edmonton protocol. In fact, for diabetic patients who are already under immunosuppression (following kidney transplant), the advantage of encapsulation is not clear. At present, microencapsulation should be considered only for patients who will not be subjected to immunosuppression and are not candidates to receive either an islet or pancreas transplant. A second barrier to the large use of encapsulation could be the long-term stability of microencapsulation, which has not been broadly investigated in preclinical studies. But it has been recently reported that intact alginate microcapsules were present in the peritoneum of one patient for 9.5 years (10,11). A third barrier in the past was inadequate purification of alginate. Currently, however, alginate can be produced with almost no endotoxin contamination, which could make a difference in terms of foreign body reaction, biocompatibility, and stability.

For these reasons, clinicians believe that encapsulation could be considered when the use of an alternative source of insulin-producing cells becomes available. Interestingly, in recent years there have been more patients receiving microencapsulated pig islets rather than allogeneic human islets.

Clinical Xenogeneic Pig Microencapsulation

In 2007, Elliott et al. (10) reported the case of a 41-year-old labile diabetic patient who in 1996 received a single implantation of microencapsulated pig islet cells into the peritoneal cavity to help regulate blood glucose levels and control diabetes. This patient's insulin requirements were reduced 10–30% for over 14 months, and 9.5 years later, a laparoscopic examination revealed intact capsules and some living and functioning pig islet cells in the peritoneal cavity. These findings demonstrated the long-term safety, viability, and functionality of encapsulated porcine islets in one patient, without the use of immunosuppression.

Although larger clinical studies with microencapsulated pig islets grafts are still being conducted (New Zealand, Argentina) or have been completed (Russia) by the same team and Living Cell Technologies (http://www.lctglobal.com), only partial data have been disclosed (12). However, these data are promising as they show that severe hypoglycemia and hypoglycemia unawareness were significantly reduced in patients receiving intraperitoneal pig islets implantation.

Pig-to-Primate Preclinical Data

Until recently, the use of microencapsulated pig islets in humans was not really supported by strong preclinical data, except one historical study in 1996, which demonstrated following transplantation of microencapsulated adult pig islets into spontaneously diabetic cynomolgus monkeys that blood glucose became normalized and the monkeys became insulin-independent for periods ranging from 120 to 804 days (13). Although these results were very encouraging for the clinical application, they may have been dependent on the primate model. In fact, the article does not report a precise status of the “spontaneous” diabetic monkeys used and particularly not the primate C-peptide levels from these animals prior and during the follow-up.

Two more recent studies describing transplantation of microencapsulated neonatal pig islets in an alginate matrix confirmed their biocompatibility in primates as well as their capacity to partially regulate diabetes very transiently (14,15). One study confirmed the biocompatibility for up to 8 weeks after transplantation of encapsulated pig islets in nondiabetic animals; the second demonstrated that these microcapsules could regulate the diabetic state of diabetic recipients. Although the latter showed that daily exogenous insulin requirements were reduced by a mean of 43% compared with control animals transplanted with empty capsules, neither showed significant changes in weekly blood glucose levels (14). The absence of solid consistent data on glucose metabolism renders these experimental studies difficult to consider as evidence for going to clinical trials. More recently, Gianello and colleagues (16) presented data showing that in nondiabetic primates some of the microencapsulated pig islets survived for up to 6 months and were able to respond in vitro to glucose challenge 135 and 180 days after implantation under the kidney capsula. In addition, the microcapsules were intact under the kidney capsule.

Macroencapsulation

Although microencapsulation seems able to protect islet cells from the immune attack, the previous data are insufficient to provide evidence for clinical applications. More interesting data could come from macroencapsulation. In the first preclinical reports of macroencapsulation, a large number of islets were immunoisolated between flat-sheet double membranes (10,11). Although several types of biomaterial have been used to produce macrocapsules, including nitrocellulose, alginate, acrylonitrile, and agarose, these devices activated nonspecific foreign body reactions, resulting in fibrotic overgrowth with subsequent necrosis of the encapsulated tissue (17). Interestingly, a subcutaneously transplanted macrodevice, 4 cm in length, shaped like a teabag, and made of bilayered polytetrafluoroethylene membranes, was found to be biocompatible (11,18).

In humans, Valdés-González et al. (19) reported among 12 nonimmunosuppressed adolescents that half had reduced insulin requirement up to 4 years after transplantation of porcine islets encapsulated in hollow fibers with porcine Sertoli cells, which likely have immunomodulating properties. Two patients became insulin-independent for several months, and porcine insulin was detected following glucose stimulation in three patients. However, this study was done in young patients without preclinical results and was received with skepticism by the scientific community (20).

Recent data using macroencapsulation of pig islets in nonhuman primates are encouraging. In fact, a “monolayer” configuration of macroencapsulated pig islets (monolayer cellular device) implanted subcutaneously has been found to significantly improve diabetes control (HbA1c <7%) in primates for 6 months without any immunosuppression. In this macroencapsulation system, islets were seeded as a monolayer on an acellular collagen matrix, enhancing their interactions with a biologic membrane and increasing islet concentration per unit surface area. In addition, diabetes was controlled for up to 1 year in two diabetic nonhuman primates after retransplantation with new monolayer cellular devices (21). Interestingly, in the same model, diabetic control was obtained for more than 32 weeks after the cotransplantation of adult pig islets and either bone marrow or adipose mesenchymal stem cells (MSCs) (22). The cotransplantation of MSCs increased the vascularization of the monolayer device in terms of increased number of vessels and production of vascular endothelial growth factor (Fig. 1). By using this subcutaneous monolayer cellular device, a phase 1 clinical study is currently ongoing at the Université Catholique de Louvain to assess the safety of this device for allotransplantation of encapsulated islets into humans.

Figure 1

Improvement of monolayer cellular device vascularization after subcutaneous cotransplantation of adipose MSCs and pig islets in diabetic primates. A: Diabetes was induced by streptozotocin in cynomolgus monkeys, and 4 weeks later, they received a subcutaneous patch containing a mixture of adult pig islet cells and either bone marrow−derived MSCs (BM-MSCs) or adipose MSCs (AMSCs). The fasting blood glucose (FBG) was measured during a follow-up over 6 months and showed a correction of the glycemia up to 34 weeks. B: Patches were harvested after and—as seen macroscopically—were still intact. Immunohistochemistry by von Willebrand factor (vWF) clearly showed a significant increase of small capillaries around the patch.

Figure 1

Improvement of monolayer cellular device vascularization after subcutaneous cotransplantation of adipose MSCs and pig islets in diabetic primates. A: Diabetes was induced by streptozotocin in cynomolgus monkeys, and 4 weeks later, they received a subcutaneous patch containing a mixture of adult pig islet cells and either bone marrow−derived MSCs (BM-MSCs) or adipose MSCs (AMSCs). The fasting blood glucose (FBG) was measured during a follow-up over 6 months and showed a correction of the glycemia up to 34 weeks. B: Patches were harvested after and—as seen macroscopically—were still intact. Immunohistochemistry by von Willebrand factor (vWF) clearly showed a significant increase of small capillaries around the patch.

Close modal

Biomaterials for Islet Encapsulation Technology

There are two main categories of polymers that can be used either as water-soluble polymers, such as alginate, or water-insoluble polymers, such as poly(hydroxyethyl methacrylate-methyl methacrylate) (17), but the latter requires organic solvent usually interfering with cellular function (23). Despite their solubility in aqueous solutions, alginate-based capsules have been shown to remain stable for several years in small and large animals and in humans (11,21,2427). In most studies to date, alginate beads were coated with a second layer to reduce the porosity of the capsule membrane (17,28); in some studies, alternating layers of poly-l-lysine and polyornithine were used to surround the alginate core but mechanical instability was described, thereby limiting their application (17,29,30). Several groups have recently reported that encapsulation in simple alginate microbeads can protect pig pancreatic cells against xenorejections in diabetic mice (5,6,31). Although several chemical formulations of alginate (e.g., high-mannuronic/guluronic and high/low viscosity, with or without additional peptide sequences) have been proposed for islet immunoisolation, high-mannuronic alginate was the most suitable to obtain selective impermeability for molecules over 150 kDa and optimal biocompatibility associated with surrounding angiogenesis and sufficient oxygen tension (up to 40 mmHg) (32). This type of alginate was biocompatible not only in rodents but also into a pig-to-primate model of xenotransplantation for up to 8 months (16,33,34).

Possible Sites for Islet Implantation

Islets perform well in a number of ectopic locations and as long as they get appropriately vascularized, islets will be able to function correctly. A site in which encapsulated islets are in close contact with the bloodstream is crucial for clinical applications. Unfortunately, it is difficult to find such a site for encapsulated islets due to the large graft volume needed. Sites reported to allow successful nonencapsulated islet transplantation, such as the liver and spleen, do not meet these requirements because these sites are unable to tolerate the large volumes of capsules (of diameter >600 μm) required for transplantation. Therefore, most transplantations of encapsulated pig islets into preclinical models were intraperitoneal (13,14). Although the technique is easy by laparoscopy, the peritoneal site could be not optimal. In fact, studies in mice found that macrophages and lymphocytes are involved in the rapid degradation of encapsulated pig islets after their transplantation into the peritoneum (31,3538). The peritoneum is a preferential site for inflammation and immunologic reactions. In fact, peritoneal mesothelial cells facilitate the action of powerful innate immune mechanisms by producing (39) significant levels of cytokines, such as tumor necrosis factor-α, interleukin (IL)-1β, IL-10, and intracellular adhesion molecule-I (40). Studies in mice showed that immunosuppression had beneficial effects, improving the biocompatibility and prolonging the survival of encapsulated pig islets transplanted into the peritoneum (35,36,41), but the combined use of encapsulation and immunosuppression probably reduces to nihil the interest of encapsulation.

Encapsulated pig islets transplanted under the kidney capsule and under the skin demonstrated better biocompatibility than capsules transplanted into the peritoneum in both rodents and primates (33). Subcutaneous and kidney capsule implantation resulted in very weak cellular immune reactions (low macrophages recruitment) against encapsulated pig islets, along with improved porcine islet viability, optimal insulin secretion after glucose challenge, and an acceptable oxygen tension (20–40 mmHg) compatible with the function and survival of encapsulated islets (32).

Human Embryonic Stem and Induced Pluripotent Stem Cells

The process of differentiating pluripotent cells toward pancreatic endocrine cells has not been easy or without controversy, but there is a consensus that we have finally reached the “plateau of productivity” often cited when describing the evolution of most technical innovations (42,43). The protocols developed over the second half of the last decade by scientists at ViaCyte (formerly Novocell) (44,45) have paved the way for what is widely expected to be the first human embryonic stem cell−based clinical trials within the next few years (Fig. 2). These methods have circumvented a bottleneck still in place today (our inability to generate fully functional β-cells in vitro [44]) by calling for the transplantation of these cells at the β-cells progenitor stage, long before they are capable of producing insulin (46). By doing so, the recipient’s body provides a microenvironment that is permissive for their efficient functional maturation, even if the process is neither short nor safe (47). Indeed, maturation may typically take anywhere from 2 to 3 months, during which carry-over undifferentiated cells may give rise to teratomas. While further preclinical refinements of the protocol have reportedly taken care of the latter problem (which could also be addressed by selecting only nontumorigenic cells for transplantation [46]), the former is still of concern for those who would rather have a fully characterized cell product prior to transplantation. Regarding the issue of rejection, the first clinical trials are likely to adopt the embodiment proposed by ViaCyte (www.viacyte.com), in which allogeneic progenitor cell populations will be seeded within a subcutaneously implanted immunoisolation device (see Macroencapsulation), which would serve the dual purpose of preventing allorejection and providing a physical containment for any potential tumors that may arise. As this approach does not require immunosuppression of the patient, any conceivable breakage of the device (e.g., by tumorigenic cells) would result in the prompt rejection of the graft.

Figure 2

β-Cell regeneration approaches. Conventional means of pluripotent stem cell procurement entail the extraction and culture of the inner cell mass of preimplantation blastocysts. These are known as embryonic stem (ES) cells (a). An alternative source derived from terminally differentiated somatic cells makes use of reprogramming technology. These reprogrammed cells are called iPS cells (b), and are functionally equivalent to ES cells. Pluripotent cells of both origins can be differentiated along the pancreatic β-cell lineage following multistep protocols that recapitulate the key biological events of pancreatic development. The sequential activation of genes such as Sox17 (definitive endoderm), Pdx1 (pancreatic progenitors), and Ngn3 (pancreatic endocrine progenitors) is commonly observed in these methods. The robust expression of terminal differentiation markers, such as MafA, insulin, glucokinase, or Glut-2, is observed only sporadically in most reported methods, as the efficiency of the last differentiation step is limited thus far. This is why the state of the art calls for the transplantation of pancreatic progenitors rather than terminally differentiated cells. Although some types of adult MSCs could hypothetically be compared with the mesendodermal precursors (which undergo an epithelial-to-mesenchymal transition before becoming definitive endoderm), the ability of such cells to seamlessly join the β-cell differentiation cascade at that juncture is debatable (c). Recently published evidence suggests that endodermal progenitor cells residing in the adult liver and extrahepatic biliary tree may also retain the ability to become either hepatocytes or pancreas, depending on the nature of the ECM onto which they are seeded. If isolated and expanded, these could be potentially used for β-cell differentiation either at the foregut endoderm-equivalent or the pancreatic endoderm-equivalent level (d). Last, reprogramming techniques previously developed for iPS cell generation have also been successfully used to transdifferentiate (or laterally reprogram) either liver or pancreatic acinar tissue (e) directly into insulin-producing β-cells (adapted from Orlando et al. [137], with permission).

Figure 2

β-Cell regeneration approaches. Conventional means of pluripotent stem cell procurement entail the extraction and culture of the inner cell mass of preimplantation blastocysts. These are known as embryonic stem (ES) cells (a). An alternative source derived from terminally differentiated somatic cells makes use of reprogramming technology. These reprogrammed cells are called iPS cells (b), and are functionally equivalent to ES cells. Pluripotent cells of both origins can be differentiated along the pancreatic β-cell lineage following multistep protocols that recapitulate the key biological events of pancreatic development. The sequential activation of genes such as Sox17 (definitive endoderm), Pdx1 (pancreatic progenitors), and Ngn3 (pancreatic endocrine progenitors) is commonly observed in these methods. The robust expression of terminal differentiation markers, such as MafA, insulin, glucokinase, or Glut-2, is observed only sporadically in most reported methods, as the efficiency of the last differentiation step is limited thus far. This is why the state of the art calls for the transplantation of pancreatic progenitors rather than terminally differentiated cells. Although some types of adult MSCs could hypothetically be compared with the mesendodermal precursors (which undergo an epithelial-to-mesenchymal transition before becoming definitive endoderm), the ability of such cells to seamlessly join the β-cell differentiation cascade at that juncture is debatable (c). Recently published evidence suggests that endodermal progenitor cells residing in the adult liver and extrahepatic biliary tree may also retain the ability to become either hepatocytes or pancreas, depending on the nature of the ECM onto which they are seeded. If isolated and expanded, these could be potentially used for β-cell differentiation either at the foregut endoderm-equivalent or the pancreatic endoderm-equivalent level (d). Last, reprogramming techniques previously developed for iPS cell generation have also been successfully used to transdifferentiate (or laterally reprogram) either liver or pancreatic acinar tissue (e) directly into insulin-producing β-cells (adapted from Orlando et al. [137], with permission).

Close modal

As for the star newcomers in the field, namely induced pluripotent stem (iPS) cells (48,49), they have also proved their worth at generating pancreatic endocrine cells in vitro (50). However, as fast as iPS cells rose to relevance, concerns about their stability have somewhat cooled down their prospects as potential replacements for human embryonic stem cells. In particular, it has been reported some of their derivatives senesce prematurely (51), that they maintain some residual epigenetic memory of their parental tissues (5255), and that the formation of potentially oncogenic mutations might be inherent to the process of reprogramming (5659).

Adult Stem Cells

The hypothesis that the ductal system may harbor such progenitor cells has been considered for decades now, and is still being studied very actively. More recently, a unique population of cells within the extrahepatic biliary tree has been characterized for its capacity to turn into both hepatocytes and pancreatic endocrine cells (60,61). Their existence, rather than a mere remnant of the embryonic stage at which liver and pancreas diverted, has been explained in the context of a theoretical process of organogenesis throughout life (62,63). Whether or not these cells can be expanded in sufficient amounts for clinical use is the subject of active investigation.

In parallel to the above efforts, many groups are pursuing the most ubiquitous of all adult stem cells, the MSCs (or stromal cells). MSCs are easily expandable in plastic and can be isolated from most tissues, including the pancreas (64). In a recent review of the subject, Domínguez-Bendala et al. (65) echoed the growing perception that these mesodermal cells are intrinsically incapable of becoming bona fide endodermal β-cells. Their directed differentiation and subsequent transplantation has met only with partial success in preclinical models (6675). However, when directly infused in an undifferentiated state onto diabetic subjects, a variable degree of endogenous regeneration has been observed (7679). It is now broadly believed that MSCs are at their therapeutic best when used in this context, probably due to their well-studied immunomodulatory, anti-inflammatory, proangiogenic, and trophic properties (8087).

Hematopoietic stem cells are another variety of adult stem cells that have been studied for their potential use to treat type 1 diabetes. To an even higher degree than MSCs (because they do it systemically), the main usefulness of hematopoietic stem cells relies on their ability to modulate immunity. Thus, they have been successfully tested for their ability to reset the immunological clock of diabetes (88,89). Cord blood−derived multipotent stem cells (CB-SCs) are also the basis of the novel “stem cell educator” concept, in which lymphocytes of type 1 diabetic patients are circulated through a device preseeded with CB-SCs from healthy donors and reinfused into the patient after a quick process of “re-education” that has been reported to reverse type 1 diabetes (90,91). These findings, however, still require independent confirmation.

Cell Reprogramming

In spite of the concerns previously pointed out, the reprogramming techniques originally developed for iPS cell generation are here to stay, and will probably be used more and more for other applications. From a technical perspective, the use of retroviruses to deliver the reprogramming agents appears now downright primitive in view of the more translational friendly alternatives of synthetic mRNAs, DNA minicircles, episomal vectors, or even transducible proteins (92,93). Early pioneers of the field of pancreatic reprogramming made use of the master gene Pdx1, which, when ectopically expressed in the liver of diabetic mice, led to reversal of hyperglycemia (94). Immunohistochemical analysis of such livers revealed insulin-positive cells in close proximity to blood vessels. It was subsequently found that Pdx1 expression persisted in the liver long after the expected clearance time of the adenoviruses used to carry the gene, suggesting that the initial ectopic expression of the gene may have primed endogenous gene networks that sustain long-term reprogramming (95). Whether these observations were reflective of the reprogramming of terminal hepatocytes or the differentiation of resident progenitor/stem cells was not clear at the time, and in vitro experiments did not prove to be much more informative (96). Additional reports established that combining Pdx1 with other transcription factors, such as BETA2/NeuroD, Ngn3, or MafA, had a synergistic effect (97103). Pancreatic ductal cells, which have been historically thought to contain putative β-cell progenitors, have also been subjected to similar transfection regimens, leading to significant upregulation of insulin expression. An intriguing observation in the field has been that often times reprogramming is seemingly dependent on the activation of immune/innate responses in the host cells. This was evidenced by the fact that the adeno-associated virus (AAV)-mediated delivery of such factors failed to induce liver-to-pancreas transdifferentiation, whereas plasmids plus an irrelevant adenovirus (which are much more immunogenic than AAVs) did (103).

Just around the time when enthusiasm for the directed transdifferentiation to pancreas had started to wane, a new breakthrough rejuvenated the field again. This time, however, the starting material was the exocrine compartment of the pancreas, rather than the liver. Exocrine cells are known to be highly malleable, as shown by their reported conversion to liver (104), ductal (105,106), and β-cell−like cells (107111). Zhou et al. (112) came up with a combination of three key pancreatic transcription factors, Pdx1, Ngn3, and MafA (also used with some success in earlier liver-to-pancreas reprogramming attempts), that, when shuttled aboard adenoviral vector and injected into the pancreatic parenchyma, gave rise to new β-cells in vivo. Upon treatment, diabetic mice exhibited a marked amelioration of their glycemic levels.

More recently, the above results were confirmed in vitro using first the AR42J acinar cell line (113,114), and subsequently primary human pancreatic exocrine cells cultured in conditions that prevented epithelial-to-mesenchymal transition (114). While not as ready as hematopoietic stem cells for clinical prime time, reprogramming of human exocrine tissues offers a most promising alternative to the use of stem cells, especially considering that the exocrine compartment of the pancreas makes up to 95% of the pancreas and is now routinely discarded after every clinical islet preparation.

The pancreatic ECM is a three-dimensional, structural framework of proteins in a state of “dynamic reciprocity” with the cells of the endocrine pancreas (115). Once thought to be a simple scaffold, the ECM has recently being shown to regulate several aspects of islet biology, including development, morphology and differentiation, intracellular signaling, gene expression, adhesion and migration, proliferation, secretion, and survival. The ECM mediates these functions via three-dimensional structure (116,117), signaling molecules, and the secretion and storage of growth factors and cytokines (118). The ECM is therefore essential for effective glucose-stimulated insulin secretion and normoglycemia, the primary function of the endocrine pancreas. Thus, the ECM has become a valuable platform for regenerative medicine investigations, and intact ECM from animal or human whole organs is currently being evaluated for organ bioengineering purposes.

The functional significance of the ECM has been highlighted by the underperformance of isolated islet transplantation (117,119122). The seminal Edmonton protocol (123) attempted to correct hyperglycemia through the transplantation of isolated islets from multiple cadaveric donors. Although the majority of patients reached insulin independence following transplantation, few managed to maintain euglycemia at 2-year follow-up (119). Subsequent studies suggest that the observed transience of therapeutic effect may be due to the loss of native ECM during the islet isolation process. Disruption of the native ECM appears to compromise survival, engraftment, and revascularization of transplanted islets. This implies that preservation of the native ECM may be a necessary condition for successful islet transplantation protocols.

Several features of the ECM appear to be essential for the function of the endocrine pancreas. The three-dimensional structure of native ECM determines the topographical arrangement of pancreatic endocrine cells, which has been shown to influence islet secretory activity (124) and survival (125). Furthermore, the constituent elements of the ECM, including collagens, glycoproteins, and glycosaminoglycans, have been shown to be independently capable of preventing B-cell apoptosis induced by loss of cellular adhesion, termed anoikis (116,124,126130). ECM components have also been shown to enhance insulin secretion, even in the absence of glucose (125). Finally, the ECM is capable of binding, storing, and regulating the activity of growth factors including transforming growth factor-β1, which plays a role in development (131), function, and regeneration (132) of pancreatic islets. The dysregulation of these essential ECM−growth factor interactions have been shown to underlie a variety of pancreatic pathologies, including pancreatitis, fibrosis, and adenocarcinoma.

The permissive effects of ECM structure on islet survival and function have led researchers to investigate the use of biomaterial carriers, which mimic the structure of native ECM. These carriers serve the functions of delivery platform, transitory structural support, and mechanical immune barrier, thus enabling islet transplantation into heterotopic sites (117). The properties of these carriers can also be precisely manipulated, allowing for the study of individual ECM components. Several islet carriers have been investigated, including poly-lactic-coglycolic acid (PLGA), poly vinyl alcohol (PVA), poly(ethylene glycol) (PEG), poly(N-isopropylacrylamide) (NIPA), and biopolymer films. These carriers vary in their structure, strength, stability, biocompatibility, growth factor binding, and amenability to manipulation. However, insights from the fields of bioengineering and regenerative medicine suggest that organ-specific ECM better maintains cell function and phenotype. The use of native ECM has been made possible through the refinement of decellularization protocols capable of removing DNA, cellular material, and cell surface antigens while leaving the attachment sites, structural integrity, and vascular channels intact. Decellularization protocols involve the repeated irrigation of cadaveric tissues with detergents or acids through the innate vasculature, although organs with higher fat content, like the pancreas, often require the addition of lipid solvents, such as alcohol (133).

Pancreas bioengineering lags behind other organs in the field, as only three studies to date report successful recellularization of native pancreatic ECM. De Carlo et al. (134) report the successful subcutaneous implantation of PVA/PEG tubular devices containing slices of rat pancreas and liver recellularized with differentiated murine islets. The pancreatic matrix significantly extended the duration of insulin function, suggesting that pancreas-specific matrix favors physiological response to glucose over the long term. Upon transplantation, the islet devices effectively reduced hyperglycemia, although normalization was not achieved. Conrad et al. (135) report abbreviated findings describing the successful recellularization of murine pancreatic matrix with human islet cells and supportive MSCs. The islets showed preserved glucose-stimulated insulin response, cell viability, subcellular anatomy, and attachments. However, complete findings have yet to be published. In a recent report from the Wake Forest School of Medicine, whole-organ acellular ECM scaffolds were generated through detergent-based decellularization of porcine pancreata (7). The method described therein achieved effective removal of the cellular material and DNA content of the native organ, while preserving ECM proteins and the native vascular tree. The scaffolds were seeded thereafter with human stem cells and porcine pancreatic islets, demonstrating that the decellularized pancreas can support cellular adhesion and maintenance of cell functions (Fig. 3). Similar data have been recently produced in a rodent model (136). However, the successful decellularization and subsequent recellularization of the human pancreatic ECM has not been reported in the bioengineering literature.

Figure 3

Islet survival on decellularized porcine pancreas matrix. A: Islets seeded on porcine acellular pancreatic matrix showed a twofold increase of metabolic activity, from day 3 to day 7, in comparison with islets maintained in culture media, as measured by MTS assay. B: Following 3 days in culture, pig islets maintained in culture media or seeded onto porcine acellular pancreatic matrix were subject to a glucose challenge. Islets seeded onto scaffolds demonstrated a significantly increased insulin secretion ratio (highest peak to mean basal values) compared with unseeded controls. Values expressed as mean ± SD, n = 3, *P < 0.05. C: Porcine islet glucose challenge. Time course data from perifusion of control (unseeded) porcine islets (blue) or islets seeded onto acellular pancreatic matrix (green). Groups were cultured for 3 days then subjected to a glucose challenge, being preperifused for 30 min in low glucose medium prior to collection of samples for insulin secretion analysis. Low glucose levels (3.3 mmol/L) represented physiological euglycemia of 60 mg/dL, and the transition to high glucose (11.1 mmol/L) represented upper limit postprandial levels of 200 mg/dL. Both groups displayed increased insulin secretion during high glucose perifusion, with islets seeded onto scaffolds demonstrating higher peak insulin secretion values, observed specifically at 50, 55, and 75 min. Both groups showed a reduction of insulin secretion after a return to low (basal) glucose concentration (Mirmalek-Sani et al. [1], with permission).

Figure 3

Islet survival on decellularized porcine pancreas matrix. A: Islets seeded on porcine acellular pancreatic matrix showed a twofold increase of metabolic activity, from day 3 to day 7, in comparison with islets maintained in culture media, as measured by MTS assay. B: Following 3 days in culture, pig islets maintained in culture media or seeded onto porcine acellular pancreatic matrix were subject to a glucose challenge. Islets seeded onto scaffolds demonstrated a significantly increased insulin secretion ratio (highest peak to mean basal values) compared with unseeded controls. Values expressed as mean ± SD, n = 3, *P < 0.05. C: Porcine islet glucose challenge. Time course data from perifusion of control (unseeded) porcine islets (blue) or islets seeded onto acellular pancreatic matrix (green). Groups were cultured for 3 days then subjected to a glucose challenge, being preperifused for 30 min in low glucose medium prior to collection of samples for insulin secretion analysis. Low glucose levels (3.3 mmol/L) represented physiological euglycemia of 60 mg/dL, and the transition to high glucose (11.1 mmol/L) represented upper limit postprandial levels of 200 mg/dL. Both groups displayed increased insulin secretion during high glucose perifusion, with islets seeded onto scaffolds demonstrating higher peak insulin secretion values, observed specifically at 50, 55, and 75 min. Both groups showed a reduction of insulin secretion after a return to low (basal) glucose concentration (Mirmalek-Sani et al. [1], with permission).

Close modal

We have reviewed key aspects of regenerative medicine technologies as they relate to the endocrine pancreas and the reconstitution of the β-cell compartment. Advances in islet encapsulation, stem cell therapy, and organ bioengineering are intersecting with the promise to resolve the dire shortage of transplantable organs. Despite steady progress, pancreatic bioengineering lags behind other organs in the field due to the complex architecture and physiology of the native pancreas, and a poor understanding of the interactions between β-cells, vascular supply, growth factors, and the pancreatic ECM. While current research endeavors are promising, the transition to safe and effective clinical implementation faces significant obstacles. Collaborative efforts are required to drive the field toward the production of a bioengineered pancreas capable of restoring endocrine function in patients with end-stage disease.

Duality of Interest.No potential conflicts of interest relevant to this article were reported.

Author Contributions. G.O. conceived and designed the manuscript, wrote the section on ECM technology, edited the manuscript, and accepted its final version. P.G. wrote the second section, edited the manuscript, and accepted its final version. M.S. cowrote the third section, edited the manuscript, and accepted its final version. R.J.S. and S.S. edited the manuscript and accepted its final version. C.R. and J.D.-B. wrote the first section, edited the manuscript, and accepted its final version.

1.
Mirmalek-Sani
SH
,
Orlando
G
,
McQuilling
JP
, et al
.
Porcine pancreas extracellular matrix as a platform for endocrine pancreas bioengineering
.
Biomaterials
2013
;
34
:
5488
5495
[PubMed]
2.
Ricordi
C
,
Inverardi
L
,
Domínguez-Bendala
J
.
From cellular therapies to tissue reprogramming and regenerative strategies in the treatment of diabetes
.
Regen Med
2012
;
7
(
Suppl.
):
41
48
[PubMed]
3.
Gray
DW
.
Encapsulated islet cells: the role of direct and indirect presentation and the relevance to xenotransplantation and autoimmune recurrence
.
Br Med Bull
1997
;
53
:
777
788
[PubMed]
4.
van Schilfgaarde
R
,
de Vos
P
.
Factors influencing the properties and performance of microcapsules for immunoprotection of pancreatic islets
.
J Mol Med (Berl)
1999
;
77
:
199
205
[PubMed]
5.
Duvivier-Kali
VF
,
Omer
A
,
Lopez-Avalos
MD
,
O’Neil
JJ
,
Weir
GC
.
Survival of microencapsulated adult pig islets in mice in spite of an antibody response
.
Am J Transplant
2004
;
4
:
1991
2000
[PubMed]
6.
Omer
A
,
Duvivier-Kali
VF
,
Trivedi
N
,
Wilmot
K
,
Bonner-Weir
S
,
Weir
GC
.
Survival and maturation of microencapsulated porcine neonatal pancreatic cell clusters transplanted into immunocompetent diabetic mice
.
Diabetes
2003
;
52
:
69
75
[PubMed]
7.
Soon-Shiong
P
.
Treatment of type I diabetes using encapsulated islets
.
Adv Drug Deliv Rev
1999
;
35
:
259
270
[PubMed]
8.
Tuch
BE
,
Keogh
GW
,
Williams
LJ
, et al
.
Safety and viability of microencapsulated human islets transplanted into diabetic humans
.
Diabetes Care
2009
;
32
:
1887
1889
[PubMed]
9.
Basta
G
,
Montanucci
P
,
Luca
G
, et al
.
Long-term metabolic and immunological follow-up of nonimmunosuppressed patients with type 1 diabetes treated with microencapsulated islet allografts: four cases
.
Diabetes Care
2011
;
34
:
2406
2409
[PubMed]
10.
Elliott
RB
,
Escobar
L
,
Tan
PL
, et al
.
Live encapsulated porcine islets from a type 1 diabetic patient 9.5 yr after xenotransplantation
.
Xenotransplantation
2007
;
14
:
157
161
11.
Strautz
RL
.
Studies of hereditary-obese mice (obob) after implantation of pancreatic islets in Millipore filter capsules
.
Diabetologia
1970
;
6
:
306
312
[PubMed]
12.
Elliott
RB
Living Cell Technologies
.
Towards xenotransplantation of pig islets in the clinic
.
Curr Opin Organ Transplant
2011
;
16
:
195
200
[PubMed]
13.
Sun
Y
,
Ma
X
,
Zhou
D
,
Vacek
I
,
Sun
AM
.
Normalization of diabetes in spontaneously diabetic cynomologus monkeys by xenografts of microencapsulated porcine islets without immunosuppression
.
J Clin Invest
1996
;
98
:
1417
1422
[PubMed]
14.
Elliott
RB
,
Escobar
L
,
Tan
PL
, et al
.
Intraperitoneal alginate-encapsulated neonatal porcine islets in a placebo-controlled study with 16 diabetic cynomolgus primates
.
Transplant Proc
2005
;
37
:
3505
3508
[PubMed]
15.
Elliott
RB
,
Escobar
L
,
Calafiore
R
, et al
.
Transplantation of micro- and macroencapsulated piglet islets into mice and monkeys
.
Transplant Proc
2005
;
37
:
466
469
[PubMed]
16.
Dufrane
D
,
Goebbels
RM
,
Saliez
A
,
Guiot
Y
,
Gianello
P
.
Six-month survival of microencapsulated pig islets and alginate biocompatibility in primates: proof of concept
.
Transplantation
2006
;
81
:
1345
1353
[PubMed]
17.
de Vos
P
,
Hamel
AF
,
Tatarkiewicz
K
.
Considerations for successful transplantation of encapsulated pancreatic islets
.
Diabetologia
2002
;
45
:
159
173
[PubMed]
18.
Rafael
E
,
Wernerson
A
,
Arner
P
,
Wu
GS
,
Tibell
A
.
In vivo evaluation of glucose permeability of an immunoisolation device intended for islet transplantation: a novel application of the microdialysis technique
.
Cell Transplant
1999
;
8
:
317
326
[PubMed]
19.
Valdés-González
RA
,
Dorantes
LM
,
Garibay
GN
, et al
.
Xenotransplantation of porcine neonatal islets of Langerhans and Sertoli cells: a 4-year study
.
Eur J Endocrinol
2005
;
153
:
419
427
[PubMed]
20.
Birmingham
K
.
Skepticism surrounds diabetes xenograft experiment
.
Nat Med
2002
;
8
:
1047
[PubMed]
21.
Soon-Shiong
P
,
Heintz
RE
,
Merideth
N
, et al
.
Insulin independence in a type 1 diabetic patient after encapsulated islet transplantation
.
Lancet
1994
;
343
:
950
951
[PubMed]
22.
Vériter
S
,
Gianello
P
,
Igarashi
Y
, et al
.
Improvement of subcutaneous bioartificial pancreas vascularization and function by co-encapsulation of pig islets and mesenchymal stem cells in primates
.
Cell Transplant.
4 February
2013 [Epub ahead of print]
[PubMed]
23.
Sefton
MV
.
The good, the bad and the obvious: 1993 Clemson Award for Basic Research—keynote lecture
.
Biomaterials
1993
;
14
:
1127
1134
[PubMed]
24.
De Vos
P
,
De Haan
B
,
Van Schilfgaarde
R
.
Effect of the alginate composition on the biocompatibility of alginate-polylysine microcapsules
.
Biomaterials
1997
;
18
:
273
278
[PubMed]
25.
De Vos
P
,
De Haan
BJ
,
Wolters
GH
,
Strubbe
JH
,
Van Schilfgaarde
R
.
Improved biocompatibility but limited graft survival after purification of alginate for microencapsulation of pancreatic islets
.
Diabetologia
1997
;
40
:
262
270
[PubMed]
26.
De Vos
P
,
Van Straaten
JF
,
Nieuwenhuizen
AG
, et al
.
Why do microencapsulated islet grafts fail in the absence of fibrotic overgrowth?
Diabetes
1999
;
48
:
1381
1388
[PubMed]
27.
Duvivier-Kali
VF
,
Omer
A
,
Parent
RJ
,
O’Neil
JJ
,
Weir
GC
.
Complete protection of islets against allorejection and autoimmunity by a simple barium-alginate membrane
.
Diabetes
2001
;
50
:
1698
1705
[PubMed]
28.
Lim
F
,
Sun
AM
.
Microencapsulated islets as bioartificial endocrine pancreas
.
Science
1980
;
210
:
908
910
[PubMed]
29.
Strand
BL
,
Ryan
TL
,
In’t Veld
P
, et al
.
Poly-l-lysine induces fibrosis on alginate microcapsules via the induction of cytokines
.
Cell Transplant
2001
;
10
:
263
275
[PubMed]
30.
King
A
,
Sandler
S
,
Andersson
A
.
The effect of host factors and capsule composition on the cellular overgrowth on implanted alginate capsules
.
J Biomed Mater Res
2001
;
57
:
374
383
[PubMed]
31.
Korbutt
GS
,
Mallett
AG
,
Ao
Z
,
Flashner
M
,
Rajotte
RV
.
Improved survival of microencapsulated islets during in vitro culture and enhanced metabolic function following transplantation
.
Diabetologia
2004
;
47
:
1810
1818
[PubMed]
32.
Vériter
S
,
Mergen
J
,
Goebbels
RM
, et al
.
In vivo selection of biocompatible alginates for islet encapsulation and subcutaneous transplantation
.
Tissue Eng Part A
2010
;
16
:
1503
1513
[PubMed]
33.
Dufrane
D
,
Steenberghe
Mv
,
Goebbels
RM
,
Saliez
A
,
Guiot
Y
,
Gianello
P
.
The influence of implantation site on the biocompatibility and survival of alginate encapsulated pig islets in rats
.
Biomaterials
2006
;
27
:
3201
3208
[PubMed]
34.
Dufrane
D
,
Goebbels
RM
,
Gianello
P
.
Alginate macroencapsulation of pig islets allows correction of streptozotocin-induced diabetes in primates up to 6 months without immunosuppression
.
Transplantation
2010
;
90
:
1054
1062
[PubMed]
35.
Omer
A
,
Keegan
M
,
Czismadia
E
, et al
.
Macrophage depletion improves survival of porcine neonatal pancreatic cell clusters contained in alginate macrocapsules transplanted into rats
.
Xenotransplantation
2003
;
10
:
240
251
[PubMed]
36.
Safley
SA
,
Kapp
LM
,
Tucker-Burden
C
,
Hering
B
,
Kapp
JA
,
Weber
CJ
.
Inhibition of cellular immune responses to encapsulated porcine islet xenografts by simultaneous blockade of two different costimulatory pathways
.
Transplantation
2005
;
79
:
409
418
[PubMed]
37.
de Vos
P
,
Smedema
I
,
van Goor
H
, et al
.
Association between macrophage activation and function of micro-encapsulated rat islets
.
Diabetologia
2003
;
46
:
666
673
[PubMed]
38.
de Groot
M
,
Schuurs
TA
,
Leuvenink
HG
,
van Schilfgaarde
R
.
Macrophage overgrowth affects neighboring nonovergrown encapsulated islets
.
J Surg Res
2003
;
115
:
235
241
[PubMed]
39.
Hall
JC
,
Heel
KA
,
Papadimitriou
JM
,
Platell
C
.
The pathobiology of peritonitis
.
Gastroenterology
1998
;
114
:
185
196
[PubMed]
40.
Yao
V
,
McCauley
R
,
Cooper
D
,
Platell
C
,
Hall
JC
.
Peritoneal mesothelial cells produce cytokines in a murine model of peritonitis
.
Surg Infect (Larchmt)
2004
;
5
:
229
236
[PubMed]
41.
Hsu
BR
,
Chang
FH
,
Juang
JH
,
Huang
YY
,
Fu
SH
.
The rescue effect of 15-deoxyspergualin on intraperitoneal microencapsulated xenoislets
.
Cell Transplant
1999
;
8
:
307
315
[PubMed]
42.
Fenn
J
,
Raskino
M
.
Mastering the Hype Cycle: How to Choose the Right Innovation at the Right Time. Harvard Business School Press Series
.
Gartner
I
, Ed.
Boston, MA
,
Harvard Business Press
,
2008
43.
Mason
C
,
Manzotti
E
.
Induced pluripotent stem cells: an emerging technology platform and the Gartner hype cycle
.
Regen Med
2009
;
4
:
329
331
[PubMed]
44.
D’Amour
KA
,
Agulnick
AD
,
Eliazer
S
,
Kelly
OG
,
Kroon
E
,
Baetge
EE
.
Efficient differentiation of human embryonic stem cells to definitive endoderm
.
Nat Biotechnol
2005
;
23
:
1534
1541
[PubMed]
45.
McLean
AB
,
D’Amour
KA
,
Jones
KL
, et al
.
Activin a efficiently specifies definitive endoderm from human embryonic stem cells only when phosphatidylinositol 3-kinase signaling is suppressed
.
Stem Cells
2007
;
25
:
29
38
[PubMed]
46.
Kelly
OG
,
Chan
MY
,
Martinson
LA
, et al
.
Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells
.
Nat Biotechnol
2011
;
29
:
750
756
[PubMed]
47.
Kroon
E
,
Martinson
LA
,
Kadoya
K
, et al
.
Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo
.
Nat Biotechnol
2008
;
26
:
443
452
[PubMed]
48.
Takahashi
K
,
Tanabe
K
,
Ohnuki
M
, et al
.
Induction of pluripotent stem cells from adult human fibroblasts by defined factors
.
Cell
2007
;
131
:
861
872
[PubMed]
49.
Yu
J
,
Vodyanik
MA
,
Smuga-Otto
K
, et al
.
Induced pluripotent stem cell lines derived from human somatic cells
.
Science
2007
;
318
:
1917
1920
[PubMed]
50.
Tateishi
K
,
He
J
,
Taranova
O
,
Liang
G
,
D’Alessio
AC
,
Zhang
Y
.
Generation of insulin-secreting islet-like clusters from human skin fibroblasts
.
J Biol Chem
2008
;
283
:
31601
31607
[PubMed]
51.
Feng
Q
,
Lu
SJ
,
Klimanskaya
I
, et al
.
Hemangioblastic derivatives from human induced pluripotent stem cells exhibit limited expansion and early senescence
.
Stem Cells
2010
;
28
:
704
712
[PubMed]
52.
Bar-Nur
O
,
Russ
HA
,
Efrat
S
,
Benvenisty
N
.
Epigenetic memory and preferential lineage-specific differentiation in induced pluripotent stem cells derived from human pancreatic islet beta cells
.
Cell Stem Cell
2011
;
9
:
17
23
[PubMed]
53.
Hu
Q
,
Friedrich
AM
,
Johnson
LV
,
Clegg
DO
.
Memory in induced pluripotent stem cells: reprogrammed human retinal-pigmented epithelial cells show tendency for spontaneous redifferentiation
.
Stem Cells
2010
;
28
:
1981
1991
[PubMed]
54.
Ohi
Y
,
Qin
H
,
Hong
C
, et al
.
Incomplete DNA methylation underlies a transcriptional memory of somatic cells in human iPS cells
.
Nat Cell Biol
2011
;
13
:
541
549
[PubMed]
55.
Rizzi
R
,
Di Pasquale
E
,
Portararo
P
, et al
.
Post-natal cardiomyocytes can generate iPS cells with an enhanced capacity toward cardiomyogenic re-differentation
.
Cell Death Differ
2012
;
19
:
1162
1174
[PubMed]
56.
Hussein
SM
,
Batada
NN
,
Vuoristo
S
, et al
.
Copy number variation and selection during reprogramming to pluripotency
.
Nature
2011
;
471
:
58
62
[PubMed]
57.
Pera
MF
.
Stem cells: the dark side of induced pluripotency
.
Nature
2011
;
471
:
46
47
[PubMed]
58.
Deng
J
,
Shoemaker
R
,
Xie
B
, et al
.
Targeted bisulfite sequencing reveals changes in DNA methylation associated with nuclear reprogramming
.
Nat Biotechnol
2009
;
27
:
353
360
[PubMed]
59.
Gore
A
,
Li
Z
,
Fung
HL
, et al
.
Somatic coding mutations in human induced pluripotent stem cells
.
Nature
2011
;
471
:
63
67
[PubMed]
60.
Deutsch
G
,
Jung
J
,
Zheng
M
,
Lóra
J
,
Zaret
KS
.
A bipotential precursor population for pancreas and liver within the embryonic endoderm
.
Development
2001
;
128
:
871
881
[PubMed]
61.
Wang
Y
,
Lanzoni
G
,
Carpino
G
, et al
.
Biliary tree stem cells, precursors to pancreatic committed progenitors: evidence for possible life-long pancreatic organogenesis
.
Stem Cells
2013
;
31
:
1966
1979
[PubMed]
62.
Cardinale
V
,
Wang
Y
,
Carpino
G
, et al
.
Multipotent stem/progenitor cells in human biliary tree give rise to hepatocytes, cholangiocytes, and pancreatic islets
.
Hepatology
2011
;
54
:
2159
2172
[PubMed]
63.
Cardinale
V
,
Wang
Y
,
Carpino
G
, et al
.
The biliary tree—a reservoir of multipotent stem cells
.
Nat Rev Gastroenterol Hepatol
2012
;
9
:
231
240
[PubMed]
64.
da Silva Meirelles
L
,
Chagastelles
PC
,
Nardi
NB
.
Mesenchymal stem cells reside in virtually all post-natal organs and tissues
.
J Cell Sci
2006
;
119
:
2204
2213
[PubMed]
65.
Domínguez-Bendala
J
,
Lanzoni
G
,
Inverardi
L
,
Ricordi
C
.
Concise review: mesenchymal stem cells for diabetes
.
Stem Cells Transl Med
2012
;
1
:
59
63
[PubMed]
66.
Chen
LB
,
Jiang
XB
,
Yang
L
.
Differentiation of rat marrow mesenchymal stem cells into pancreatic islet beta-cells
.
World J Gastroenterol
2004
;
10
:
3016
3020
[PubMed]
67.
Baertschiger
RM
,
Bosco
D
,
Morel
P
, et al
.
Mesenchymal stem cells derived from human exocrine pancreas express transcription factors implicated in beta-cell development
.
Pancreas
2008
;
37
:
75
84
[PubMed]
68.
Choi
KS
,
Shin
JS
,
Lee
JJ
,
Kim
YS
,
Kim
SB
,
Kim
CW
.
In vitro trans-differentiation of rat mesenchymal cells into insulin-producing cells by rat pancreatic extract
.
Biochem Biophys Res Commun
2005
;
330
:
1299
1305
[PubMed]
69.
Hisanaga
E
,
Park
KY
,
Yamada
S
, et al
.
A simple method to induce differentiation of murine bone marrow mesenchymal cells to insulin-producing cells using conophylline and betacellulin-delta4
.
Endocr J
2008
;
55
:
535
543
[PubMed]
70.
Karnieli
O
,
Izhar-Prato
Y
,
Bulvik
S
,
Efrat
S
.
Generation of insulin-producing cells from human bone marrow mesenchymal stem cells by genetic manipulation
.
Stem Cells
2007
;
25
:
2837
2844
[PubMed]
71.
Li
Y
,
Zhang
R
,
Qiao
H
, et al
.
Generation of insulin-producing cells from PDX-1 gene-modified human mesenchymal stem cells
.
J Cell Physiol
2007
;
211
:
36
44
[PubMed]
72.
Masaka
T
,
Miyazaki
M
,
Du
G
, et al
.
Derivation of hepato-pancreatic intermediate progenitor cells from a clonal mesenchymal stem cell line of rat bone marrow origin
.
Int J Mol Med
2008
;
22
:
447
452
[PubMed]
73.
Moriscot
C
,
de Fraipont
F
,
Richard
MJ
, et al
.
Human bone marrow mesenchymal stem cells can express insulin and key transcription factors of the endocrine pancreas developmental pathway upon genetic and/or microenvironmental manipulation in vitro
.
Stem Cells
2005
;
23
:
594
603
[PubMed]
74.
Wu
XH
,
Liu
CP
,
Xu
KF
, et al
.
Reversal of hyperglycemia in diabetic rats by portal vein transplantation of islet-like cells generated from bone marrow mesenchymal stem cells
.
World J Gastroenterol
2007
;
13
:
3342
3349
[PubMed]
75.
Xu
J
,
Lu
Y
,
Ding
F
,
Zhan
X
,
Zhu
M
,
Wang
Z
.
Reversal of diabetes in mice by intrahepatic injection of bone-derived GFP-murine mesenchymal stem cells infected with the recombinant retrovirus-carrying human insulin gene
.
World J Surg
2007
;
31
:
1872
1882
[PubMed]
76.
Chang
C
,
Wang
X
,
Niu
D
,
Zhang
Z
,
Zhao
H
,
Gong
F
.
Mesenchymal stem cells adopt beta-cell fate upon diabetic pancreatic microenvironment
.
Pancreas
2009
;
38
:
275
281
[PubMed]
77.
Chang
CF
,
Hsu
KH
,
Chiou
SH
,
Ho
LL
,
Fu
YS
,
Hung
SC
.
Fibronectin and pellet suspension culture promote differentiation of human mesenchymal stem cells into insulin producing cells
.
J Biomed Mater Res A
2008
;
86
:
1097
1105
[PubMed]
78.
Dong
QY
,
Chen
L
,
Gao
GQ
, et al
.
Allogeneic diabetic mesenchymal stem cells transplantation in streptozotocin-induced diabetic rat
.
Clin Invest Med
2008
;
31
:
E328
E337
[PubMed]
79.
Ezquer
FE
,
Ezquer
ME
,
Parrau
DB
,
Carpio
D
,
Yañez
AJ
,
Conget
PA
.
Systemic administration of multipotent mesenchymal stromal cells reverts hyperglycemia and prevents nephropathy in type 1 diabetic mice
.
Biol Blood Marrow Transplant
2008
;
14
:
631
640
[PubMed]
80.
Abdi
R
,
Fiorina
P
,
Adra
CN
,
Atkinson
M
,
Sayegh
MH
.
Immunomodulation by mesenchymal stem cells: a potential therapeutic strategy for type 1 diabetes
.
Diabetes
2008
;
57
:
1759
1767
[PubMed]
81.
Ball
SG
,
Shuttleworth
CA
,
Kielty
CM
.
Mesenchymal stem cells and neovascularization: role of platelet-derived growth factor receptors
.
J Cell Mol Med
2007
;
11
:
1012
1030
[PubMed]
82.
Le Blanc
K
,
Ringdén
O
.
Immunomodulation by mesenchymal stem cells and clinical experience
.
J Intern Med
2007
;
262
:
509
525
[PubMed]
83.
Mishra
PK
.
Bone marrow-derived mesenchymal stem cells for treatment of heart failure: is it all paracrine actions and immunomodulation?
J Cardiovasc Med (Hagerstown)
2008
;
9
:
122
128
[PubMed]
84.
Ozaki
K
,
Sato
K
,
Oh
I
, et al
.
Mechanisms of immunomodulation by mesenchymal stem cells
.
Int J Hematol
2007
;
86
:
5
7
[PubMed]
85.
Sadat
S
,
Gehmert
S
,
Song
YH
, et al
.
The cardioprotective effect of mesenchymal stem cells is mediated by IGF-I and VEGF
.
Biochem Biophys Res Commun
2007
;
363
:
674
679
[PubMed]
86.
Wu
Y
,
Chen
L
,
Scott
PG
,
Tredget
EE
.
Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis
.
Stem Cells
2007
;
25
:
2648
2659
[PubMed]
87.
Xu
YX
,
Chen
L
,
Wang
R
, et al
.
Mesenchymal stem cell therapy for diabetes through paracrine mechanisms
.
Med Hypotheses
2008
;
71
:
390
393
[PubMed]
88.
Couri
CE
,
Voltarelli
JC
.
Autologous stem cell transplantation for early type 1 diabetes mellitus
.
Autoimmunity
2008
;
41
:
666
672
[PubMed]
89.
Voltarelli
JC
,
Couri
CE
,
Stracieri
AB
, et al
.
Autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus
.
JAMA
2007
;
297
:
1568
1576
[PubMed]
90.
Zhao
Y
,
Jiang
Z
,
Zhao
T
, et al
.
Reversal of type 1 diabetes via islet β cell regeneration following immune modulation by cord blood-derived multipotent stem cells
.
BMC Med
2012
;
10
:
3
[PubMed]
91.
Zhao
Y
,
Wang
H
,
Mazzone
T
.
Identification of stem cells from human umbilical cord blood with embryonic and hematopoietic characteristics
.
Exp Cell Res
2006
;
312
:
2454
2464
[PubMed]
92.
Kim
D
,
Kim
CH
,
Moon
JI
, et al
.
Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins
.
Cell Stem Cell
2009
;
4
:
472
476
[PubMed]
93.
Lee
J
,
Sayed
N
,
Hunter
A
, et al
.
Activation of innate immunity is required for efficient nuclear reprogramming
.
Cell
2012
;
151
:
547
558
[PubMed]
94.
Ferber
S
,
Halkin
A
,
Cohen
H
, et al
.
Pancreatic and duodenal homeobox gene 1 induces expression of insulin genes in liver and ameliorates streptozotocin-induced hyperglycemia
.
Nat Med
2000
;
6
:
568
572
[PubMed]
95.
Ber
I
,
Shternhall
K
,
Perl
S
, et al
.
Functional, persistent, and extended liver to pancreas transdifferentiation
.
J Biol Chem
2003
;
278
:
31950
31957
[PubMed]
96.
Li
WC
,
Horb
ME
,
Tosh
D
,
Slack
JM
.
In vitro transdifferentiation of hepatoma cells into functional pancreatic cells
.
Mech Dev
2005
;
122
:
835
847
[PubMed]
97.
Kaneto
H
,
Matsuoka
TA
,
Nakatani
Y
, et al
.
A crucial role of MafA as a novel therapeutic target for diabetes
.
J Biol Chem
2005
;
280
:
15047
15052
[PubMed]
98.
Kaneto
H
,
Miyatsuka
T
,
Fujitani
Y
, et al
.
Role of PDX-1 and MafA as a potential therapeutic target for diabetes
.
Diabetes Res Clin Pract
2007
;
77
(
Suppl. 1
):
S127
S137
[PubMed]
99.
Kaneto
H
,
Miyatsuka
T
,
Shiraiwa
T
, et al
.
Crucial role of PDX-1 in pancreas development, beta-cell differentiation, and induction of surrogate beta-cells
.
Curr Med Chem
2007
;
14
:
1745
1752
[PubMed]
100.
Kojima
H
,
Fujimiya
M
,
Matsumura
K
, et al
.
NeuroD-betacellulin gene therapy induces islet neogenesis in the liver and reverses diabetes in mice
.
Nat Med
2003
;
9
:
596
603
[PubMed]
101.
Matsuoka
TA
,
Kaneto
H
,
Stein
R
, et al
.
MafA regulates expression of genes important to islet beta-cell function
.
Mol Endocrinol
2007
;
21
:
2764
2774
[PubMed]
102.
Miyatsuka
T
,
Kaneto
H
,
Kajimoto
Y
, et al
.
Ectopically expressed PDX-1 in liver initiates endocrine and exocrine pancreas differentiation but causes dysmorphogenesis
.
Biochem Biophys Res Commun
2003
;
310
:
1017
1025
[PubMed]
103.
Wang
AY
,
Ehrhardt
A
,
Xu
H
,
Kay
MA
.
Adenovirus transduction is required for the correction of diabetes using Pdx-1 or Neurogenin-3 in the liver
.
Mol Ther
2007
;
15
:
255
263
[PubMed]
104.
Lardon
J
,
De Breuck
S
,
Rooman
I
, et al
.
Plasticity in the adult rat pancreas: transdifferentiation of exocrine to hepatocyte-like cells in primary culture
.
Hepatology
2004
;
39
:
1499
1507
[PubMed]
105.
Rooman
I
,
Heremans
Y
,
Heimberg
H
,
Bouwens
L
.
Modulation of rat pancreatic acinoductal transdifferentiation and expression of PDX-1 in vitro
.
Diabetologia
2000
;
43
:
907
914
[PubMed]
106.
Means
AL
,
Meszoely
IM
,
Suzuki
K
, et al
.
Pancreatic epithelial plasticity mediated by acinar cell transdifferentiation and generation of nestin-positive intermediates
.
Development
2005
;
132
:
3767
3776
[PubMed]
107.
Baeyens
L
,
Bouwens
L
.
Cellular plasticity of the pancreas
.
Biol Chem
2009
;
390
:
995
1001
[PubMed]
108.
Baeyens
L
,
De Breuck
S
,
Lardon
J
,
Mfopou
JK
,
Rooman
I
,
Bouwens
L
.
In vitro generation of insulin-producing beta cells from adult exocrine pancreatic cells
.
Diabetologia
2005
;
48
:
49
57
[PubMed]
109.
Lima
MJ
,
Docherty
HM
,
Chen
Y
,
Docherty
K
.
Efficient differentiation of AR42J cells towards insulin-producing cells using pancreatic transcription factors in combination with growth factors
.
Mol Cell Endocrinol
2012
;
358
:
69
80
[PubMed]
110.
Minami
K
,
Okuno
M
,
Miyawaki
K
, et al
.
Lineage tracing and characterization of insulin-secreting cells generated from adult pancreatic acinar cells
.
Proc Natl Acad Sci USA
2005
;
102
:
15116
15121
[PubMed]
111.
Okuno
M
,
Minami
K
,
Okumachi
A
, et al
.
Generation of insulin-secreting cells from pancreatic acinar cells of animal models of type 1 diabetes
.
Am J Physiol Endocrinol Metab
2007
;
292
:
E158
E165
[PubMed]
112.
Zhou
Q
,
Brown
J
,
Kanarek
A
,
Rajagopal
J
,
Melton
DA
.
In vivo reprogramming of adult pancreatic exocrine cells to beta-cells
.
Nature
2008
;
455
:
627
632
[PubMed]
113.
Zhang
T
,
Saunee
NA
,
Breslin
MB
,
Song
K
,
Lan
MS
.
Functional role of an islet transcription factor, INSM1/IA-1, on pancreatic acinar cell trans-differentiation
.
J Cell Physiol
2012
;
227
:
2470
2479
[PubMed]
114.
Lima
MJ
,
Muir
KR
,
Docherty
HM
, et al
.
Suppression of epithelial-to-mesenchymal transitioning enhances ex vivo reprogramming of human exocrine pancreatic tissue toward functional insulin-producing β-like cells
.
Diabetes
2013
;
62
:
2821
2833
[PubMed]
115.
Song
JJ
,
Ott
HC
.
Organ engineering based on decellularized matrix scaffolds
.
Trends Mol Med
2011
;
17
:
424
432
[PubMed]
116.
Weber
LM
,
Hayda
KN
,
Anseth
KS
.
Cell-matrix interactions improve beta-cell survival and insulin secretion in three-dimensional culture
.
Tissue Eng Part A
2008
;
14
:
1959
1968
[PubMed]
117.
Shimizu
H
,
Ohashi
K
,
Utoh
R
, et al
.
Bioengineering of a functional sheet of islet cells for the treatment of diabetes mellitus
.
Biomaterials
2009
;
30
:
5943
5949
[PubMed]
118.
Beattie
GM
,
Cirulli
V
,
Lopez
AD
,
Hayek
A
.
Ex vivo expansion of human pancreatic endocrine cells
.
J Clin Endocrinol Metab
1997
;
82
:
1852
1856
[PubMed]
119.
Shapiro
AM
,
Ricordi
C
,
Hering
BJ
, et al
.
International trial of the Edmonton protocol for islet transplantation
.
N Engl J Med
2006
;
355
:
1318
1330
[PubMed]
120.
Ruggenenti
P
,
Remuzzi
A
,
Remuzzi
G
.
Decision time for pancreatic islet-cell transplantation
.
Lancet
2008
;
371
:
883
884
[PubMed]
121.
Nagata
NA
,
Inoue
K
,
Tabata
Y
.
Co-culture of extracellular matrix suppresses the cell death of rat pancreatic islets
.
J Biomater Sci Polym Ed
2002
;
13
:
579
590
[PubMed]
122.
Bennet
W
,
Groth
CG
,
Larsson
R
,
Nilsson
B
,
Korsgren
O
.
Isolated human islets trigger an instant blood mediated inflammatory reaction: implications for intraportal islet transplantation as a treatment for patients with type 1 diabetes
.
Ups J Med Sci
2000
;
105
:
125
133
[PubMed]
123.
Shapiro
AM
,
Lakey
JR
,
Ryan
EA
, et al
.
Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen
.
N Engl J Med
2000
;
343
:
230
238
[PubMed]
124.
Montesano
R
,
Mouron
P
,
Amherdt
M
,
Orci
L
.
Collagen matrix promotes reorganization of pancreatic endocrine cell monolayers into islet-like organoids
.
J Cell Biol
1983
;
97
:
935
939
[PubMed]
125.
Lucas-Clerc
C
,
Massart
C
,
Campion
JP
,
Launois
B
,
Nicol
M
.
Long-term culture of human pancreatic islets in an extracellular matrix: morphological and metabolic effects
.
Mol Cell Endocrinol
1993
;
94
:
9
20
[PubMed]
126.
Wang
RN
,
Rosenberg
L
.
Maintenance of beta-cell function and survival following islet isolation requires re-establishment of the islet-matrix relationship
.
J Endocrinol
1999
;
163
:
181
190
[PubMed]
127.
Rosenberg
L
,
Wang
R
,
Paraskevas
S
,
Maysinger
D
.
Structural and functional changes resulting from islet isolation lead to islet cell death
.
Surgery
1999
;
126
:
393
398
[PubMed]
128.
Meda
P
,
Hooghe-Peters
EL
,
Orci
L
.
Monolayer cultures of adult pancreatic islet cells on osmotically disrupted fibroblasts
.
Diabetes
1980
;
29
:
497
500
[PubMed]
129.
Rabinovitch
A
,
Russell
T
,
Mintz
DH
.
Factors from fibroblasts promote pancreatic islet B cell survival in tissue culture
.
Diabetes
1979
;
28
:
1108
1113
[PubMed]
130.
Thivolet
CH
,
Chatelain
P
,
Nicoloso
H
,
Durand
A
,
Bertrand
J
.
Morphological and functional effects of extracellular matrix on pancreatic islet cell cultures
.
Exp Cell Res
1985
;
159
:
313
322
[PubMed]
131.
Crisera
CA
,
Maldonado
TS
,
Kadison
AS
, et al
.
Transforming growth factor-beta 1 in the developing mouse pancreas: a potential regulator of exocrine differentiation
.
Differentiation
2000
;
65
:
255
259
[PubMed]
132.
Han
B
,
Qi
S
,
Hu
B
,
Luo
H
,
Wu
J
.
TGF-beta i promotes islet beta-cell function and regeneration
.
J Immunol
2011
;
186
:
5833
5844
[PubMed]
133.
Crapo
PM
,
Medberry
CJ
,
Reing
JE
, et al
.
Biologic scaffolds composed of central nervous system extracellular matrix
.
Biomaterials
2012
;
33
:
3539
3547
[PubMed]
134.
De Carlo
E
,
Baiguera
S
,
Conconi
MT
, et al
.
Pancreatic acellular matrix supports islet survival and function in a synthetic tubular device: in vitro and in vivo studies
.
Int J Mol Med
2010
;
25
:
195
202
[PubMed]
135.
Conrad
C
, Schuetz C, Clippinger Bet al.
Bio-engineered endocrine pancreas based on decellularized pancreatic matrix and mesenchymal stem cell/islet cell coculture
.
J Am Coll Surg
2010
;
211
:
S62
S62
136.
Goh
SK
,
Bertera
S
,
Olsen
P
, et al
.
Perfusion-decellularized pancreas as a natural 3D scaffold for pancreatic tissue and whole organ engineering
.
Biomaterials
2013
;
34
:
6760
6772
[PubMed]
137.
Orlando
G
,
Bendala
JD
,
Shupe
T
, et al
.
Cell and organ bioengineering technology as applied to gastrointestinal diseases
.
Gut
2013
;
62
:
774
786
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.