Review

Review

Organogenesis 10:2, 225–230; April/May/June 2014; © 2014 Landes Bioscience

In vitro reconstitution of pancreatic islets Nobuhiko Kojima Graduate School of Nanobioscience; Yokohama City University; Yokohama, Japan

The lack of transplantable pancreatic islets is a serious problem that affects the treatment of patients with type 1 diabetes mellitus. Beta cells can be induced from various sources of stem or progenitor cells, including induced pluripotent stem cells in the near future; however, the reconstitution of islets from β cells in culture dishes is challenging. The generation of highly functional islets may require three-dimensional spherical cultures that resemble intact islets. This review discusses recent advances in the reconstitution of islets. Several factors affect the reconstitution of pseudoislets with higher functions, such as architectural similarity, cell-to-cell contact, and the production method. The actual transplantation of naked or encapsulated pseudoislets and islet-like cell clusters from various stem cell sources is also discussed. Advancing our understanding of the methods used to reconstitute pseudoislets should expand the range of potential strategies available for developing de novo islets for therapeutic applications.

Introduction Transplantation of the pancreas or pancreatic islets is the most efficient means of treating type 1 diabetes mellitus.1,2 Whole pancreas transplantation allows the patients to remain insulinindependent for more than 10 y.3 Islet transplantation does not need major surgery and the function of the islet grafts maintained over five years.4,5 However, a shortage of donors prevents this therapy from being implemented, although the methodology is well established. The generation of artificial pancreas is an alternative means of providing transplantable islets; islets can be isolated from pigs or other animals with the hydrogel capsule and separation membrane attached to prevent the access of immunoglobulins, which would lead to immunoreactions.6-8 Another approach is to reconstitute pancreatic islets in vitro. At present, it is not possible to induce fully differentiated β cells from induced pluripotent stem (iPS) cells. However, after these methods have been established, it will be possible to obtain a cell source to form islets, which would be adapted to the immune systems of patients. Various tissue engineering methods are also needed to handle the cells so that they form islets with an appropriate architecture. Thus, it would be useful to understand the process of reconstituting islet structures using freshly isolated Correspondence to: Nobuhiko Kojima; Email: [email protected] Submitted: 12/18/2013; Revised: 02/19/2014; Accepted: 02/24/2014; Published Online: 03/03/2014; http://dx.doi.org/10.4161/org.28351

primary islet endocrine cells, established cell lines, and β cells, such as cells induced from iPS or embryonic stem (ES) cells. In this review, we focus on in vitro organogenesis to form islet-like tissues using primary endocrine cells, cell lines, and various stem cells.

Pseudoislets from Single-Cell Preparations The first reports of pseudoislet formation appeared around 1980. Scharp et al. reported the digestion of dog pancreas using trypsin to obtain single endocrine cells, where subsequent rotational culture yielded selective aggregation in 4–8 d. Pseudoislets contained all islet cell types and were stable for 4 wk, releasing hormones in response to appropriate stimuli.9 They also succeeded in forming pseudoislets from neonatal pig islets.10 Pseudoislet formation was also confirmed using adult rats,11-14 neonatal rats,11,15-17 cadaveric pancreata from children,18 and neonatal humans.19 These studies clearly demonstrate that pancreatic endocrine cells, such as α, β, and delta, and pancreatic polypeptide-producing cells have the potential to reconstitute their original architecture and form islets. Halban et al. also demonstrated that their pseudoislets were remarkably similar to adult rat islets in terms of the cellular composition and organization, although the pseudoislets were approximately half the size of the native islets.14 This encouraged the generation of islets in vitro using islet cells derived from various sources as well as primary cells.

Mechanisms of Pseudoislet Formation with an Appropriate Architecture Aggregates can form with similar cell positioning to intact islets. Thus, determining the mechanisms that underlie this process would improve our understanding of islet regenerative medicine. Halban et al. compared pseudoislets formed from native pancreatic β cells and transformed (RINm5F) cells.20 On mixing native β cells, non-β cells, and RINm5F cells, they observed that the native β cells were centrally located and were surrounded by zones of non-β cells, as reported previously, whereas the RINm5F cells were found only in the peripheral region. They concluded that a cell surface antigen recognized by a monoclonal antibody was important for the behavior of native β cells in pseudoislets. In 1991, a related study observed that E-cadherin was important for the aggregation of islet cells and that calcium-independent cell adhesion molecules distinguished the islet cell types.21 The cell surface calcium-independent adhesion molecule expressed

www.landesbioscience.com Organogenesis 225

©2014 Landes Bioscience. Do not distribute.

Keywords: pancreatic islets, pseudoislets, islet-like cell clusters, diabetes mellitus, tissue engineering

Using single-cell suspensions of pancreatic cells Non-adherent dish under static condition

Refs. 12, 13, 17, 20, 22, 23, 29, 38, 39, 45, 46, 47, 48, 50, 54, 55, 56

Non-adherent dish with shaking

Refs. 9, 10, 21, 39, 40, 45, 49

Adherent dish

Refs. 18, 36

On a gelatin-coated dish

Refs. 30, 31, 32, 33, 34, 35, 37, 41

Round-bottom 96-well plate

Ref. 57

Droplets on an untreated culture dish

Ref. 14

Centrifugation in tubes

Ref. 11

In collagen gel or synthetic polymer gel

Refs. 15, 59, 62, 63, 64

In methylcellulose sol

Ref. 42

Microencapsulation

Ref. 53

Specific surfaces

Ref. 51

Dielectrophoresis

Ref. 52

Using small pieces of pancreatic tissue Non-adherent dish

Ref. 19

by β cells that regulates β cell aggregation was shown to be controlled by tumor necrosis factor-α signaling.22 They found that neural cell adhesion molecule was expressed by non-β cells and showed that it had a role in islet cell type segregation.23 These studies demonstrate the role of cell surface molecules in selective aggregation and the formation of the islet-like architecture of pseudoislets. However, the relationships between cell positioning and the functions of pseudoislets remain to be elucidated and may help in designing a better method for the production of more functional pseudoislets for use in regenerative medicine.

Relationship between Cell-to-Cell Contact and Function It has been reported that the architecture of islets (the arrangement of endocrine cells) is important for their functions such as insulin secretion, thereby indicating that simple aggregates may exhibit relatively weak functions.24-28 Pseudoislets are suitable for studying the importance of structure and the relationships between the configurations of cells in islets, rather than β cells alone. It has been shown that cell-to-cell contact is indispensable for the initiation of insulin secretion when stimulating intact islets with glucose. Pseudoislet formation is sufficient for reversing the loss of structure–function relationships in single-cell preparations of islets cells from adult rat islets.29 Pseudoislets were also formed using MIN6, an established pancreatic β cell line, to show that cell-to-cell interactions are essential for integrated responses to nutrient stimuli.30 The β cell line-based pseudoislet system is different from pseudoislets formed using whole endocrine cells, although it is a powerful

tool for studying various aspects of islets in vitro, as shown by Jones et al. in several studies.31-38 The reconstitution of islets using two types of cells can help to elucidate the relationships between them. For example, Josefsen et al. separated islet cells obtained from rat islets into β cells and non-β cells using fluorescence-activated cell sorting. They reconstituted the separated cells and found that α cells are necessary for insulin secretion but not for glucose sensing.39 On the other hand, Hamaguchi et al. reported pseudoislets formed from α and β cell lines and showed that cell-to-cell contact reduced the level of insulin secretion by β cells.40 Brereton et al. concluded that α cells did not affect insulin secretion by β cells via cell-to-cell contact,41 although these results are controversial. Importantly, insufficient studies have used pseudoislets to test this hypothesis. Thus, the roles of interactions between endocrine cells must be intensively analyzed to facilitate the effective reconstitution of islets for use in regenerative medicine. A possible explanation for previous contrasting results is the ratio of α and β cells. Thus, we tested broader ranges of cell type ratios compared with previous studies, i.e., Hamaguchi et al. used α:β ratios of 1:2, 1:1, or 2:1, and Brereton et al. used an α:β ratio of 1:3. We found that the level of insulin secretion was higher when the α:β ratio was 1:8 than when only β cells were used. When the percentage of α cells was 20% or more, insulin secretion was not induced, remained the same or reduced.42 These results may be reflecting the importance of endocrine cell ratio in islet. For example Brissova et al. reported that mouse islets were composed of 14–19% α cells, 75–80% β cells, and 6% delta cells.43 They also reported the population of human islets, 34% α cells, 54% β cells, and 10% delta cell.43 Kim et al. compared islet architecture including β cell ratio between different species, human (64%), monkey (79%), pig (89%), rabbit (79%), bird (46%), and mouse (90%).44 It is an issue to be addressed whether the optimal cell ratio and arrangement in pseudoislets is different when we use endocrine cells from other species.

Effective Methods for Pseudoislet Production and Maintenance of Their Functions The formation of pseudoislets is not difficult, which is an advantage for pancreas studies using pseudoislets. Early studies of pseudoislets reported culture in an untreated culture dish with or without shaking (Table 1). However, analyses of the effective factors that facilitate the formation of pseudoislets with higher functions are indispensable. Matta et al. studied the conditions of islet dissociation and reaggregation of islated endcrine cells for effective pseudoislet formation, and found that dissociation with dispase and reaggregation in rotation culture with a high glucose medium (11 mM) was better than the condition using trypsin, static culture, and low glucose medium (4 mM).45 Supplementation with all-trans retinoic acid or a conditioned medium obtained from exocrine cell culture also helped in effective aggregation of dissociated islet cells, which exhibited normal insulin secretion after glucose stimulation.46

226 Organogenesis Volume 10 Issue 2

©2014 Landes Bioscience. Do not distribute.

Table 1. Methods used to form pseudoislets

Transplantation of Pseudoislets Better results are obtained in transplantation using pseudoislets that form successfully. Tze and Tai reported the allotransplantation and xenotransplantation of rat-derived pseudoislets intraportally into diabetic rats and mice.12,54 Rejection is one of the major problems of transplantation. Wolf-Jochim et al. compared the survival time after transplantation of fresh or cultured pancreatic islets, pseudoislets, and single cells beneath the kidney capsule in non-immunosuppressed diabetic rats. They concluded that transplantation of pseudoislets resulted in the prolonged acceptance of allografts, without immunosuppression of the host.55 Vascularization is another problem that affects pseudoislet transplantation. Beger et al. reported the vascularization of pseudoislets in the dorsal skin folds using Syrian golden hamsters.56 The vascularization of pseudoislet after transplantation was slower than that of native islets containing the original endothelial cells. However, they argued that pseudoislets did not disable the formation of a microvasculature. To improve the vascularization in pseudoislets, Penko et al. formed mosaic pseudoislets, which comprised β cells with interspersed vasculogenic endothelial progenitor cells.57 They did not report the results of transplantation, although these mosaic pseudoislets exhibited higher insulin secretion after glucose stimulation than normal pseudoislets that comprised only β cells.

Production of Bioartificial Islets Using Reconstituted Islets The use of immunoisolation membranes is effective for protecting islet tissues from the immune system, which is generally achieved by producing bioartificial islets using intact islets.58 This method is also adaptable to pseudoislets. Ohgawara et al. formed pseudoislets using a mouse β cell line and transferred them into diffusion chambers to create a bioartificial endocrine pancreas (Bio-AEP).59 The implantation of Bio-AEP into stereptozotocin-induced diabetic rats resulted in normoglycemia for ≥12 wk without any immunosuppressants. As mentioned previously, the capsulation of pseudoislets is another technique that facilitates immunoprotection and helps control the size of islets.53 Sakai et al. reported advanced methods for forming cell aggregates with outer cell layers as a model of islets.60 Onoe et al. established islet tissues by reconstituting them in hydrogel fibers.61 One of the advantages of producing islets in fibers is their ease of handling during transplantation or the removal of fibers. In contrast to particles beneath the kidney capsule, the fibers remain fixed; thus, it is easy to locate them after transplantation.

Islet-Like Cell Clusters (ICCs) from Various Stem Cells The term ICCs refers to islet-like cellular aggregates derived from stem cell culture. They differ from pseudoislets because ICCs are “grown” by culturing cells on tissue culture plates, which includes no step to dissociate cells into single-cell suspensions. ICCs have been produced from pancreatic tissue-derived stem cells such as rat embryonic pancreatic precursor cells,62-64 chick islet-derived stellate-like cells,65 human non-islet pancreatic cells,66 human mesenchymal stem cells (MSCs) derived from pancreatic islets,67 and human adult pancreatic endocrine progenitors.68 MSCs derived from the bone marrow of mice,69,70 rats,71,72 and humans73 have also been used to produce ICCs. Adipose tissue-derived stem cells from mice74 or humans75,76 are sources of pseudoislets. Additional stem cell sources include human peripheral blood monocytes,77 human umbilical cord blood-derived stem cells,78 human skin fibroblasts,79 human umbilical cord matrix-derived MSCs,80 human placenta-derived MSCs,81 human amnion-derived MSCs,82 human multipotent dermal fibroblasts,83 human placenta-derived multipotent stem cells,84 and human dental pulp stem cells.85 Previous studies have induced ICCs from mouse ES cells,86-89 monkey ES cells,90 and human ES cells.91-94 Mouse and human iPS cells can also be adapted to induce ICCs.95,96 In particular, the islet-like tissues induced by Saito et al. exhibited distinct three-dimensional structures, which were similar to adult pancreatic islets, and they secreted insulin in response to glucose concentrations. If the induction efficiency of islets can be improved, these systems for inducing ICCs from stem cells will be very important methods to compensate for the lack of transplantable islets. In addition, although the structure is not similar to that of native islets, it is

www.landesbioscience.com Organogenesis 227

©2014 Landes Bioscience. Do not distribute.

The addition of nicotinamide to the culture medium also maintained or enhanced the functions of pseudoislets obtained from pig neonatal or human fetal islets.47-49 Genetically engineered islets may have various potential applications. Compared with cells in native islets, dissociated endocrine cells on plates are easier to transfect with exogenous genes using viral or chemical methods. Caton et al. reported the lentivirus-mediated transduction of endocrine cells on plates and the formation of pseudoislets with gene-transferred cells,50 which were used to elucidate the functions of connexin during insulin secretion. The expression or knockdown of specific genes would be indispensable for enhancing or regulating pseudoislets in regenerative medicine. Recent trials have investigated the formation of pseudoislets using various biomaterials, including different surfaces. Yang et al. reported that poly(3-hydroxybutyrate-co-3-hydroxyhexanoate) is a good surface for the aggregation of pseudoislets that exhibit higher insulin production.51 Pethig et al. demonstrated the dielectrophoretic assembly of a β cell line to form pseudoislets at approximately 1000 cells/aggregate in a 10 × 10 array.52 The β cell aggregates included nanosensors to detect the pH and cellular oxygen levels. Tsang et al. aggregated adult human pancreatic islet cells in the intracapsular spaces of alginate-polyl-lysine microcapsules.53 These capsules had potent effects on reducing hyperglycemia and produced human C-peptide after transplantation into mice.

Conclusions Pancreatic endocrine cells have the potential to reorganize islet-like structures even when they are dissociated into single cells. This characteristic is highly beneficial for the in vitro formation of islets (pseudoislets or ICCs), which could compensate References 1.

2.

3.

4.

5.

6.

7.

8.

9.

10.

Vardanyan M, Parkin E, Gruessner C, Rodriguez Rilo HL. Pancreas vs. islet transplantation: a call on the future. Curr Opin Organ Transplant 2010; 15:12430; PMID:20009930; http://dx.doi.org/10.1097/ MOT.0b013e32833553f8 Ludwig B, Ludwig S, Steffen A, Saeger HD, Bornstein SR. Islet versus pancreas transplantation in type 1 diabetes: competitive or complementary? Curr Diab Rep 2010; 10:506-11; PMID:20830612; http:// dx.doi.org/10.1007/s11892-010-0146-y Ricordi C, Strom TB. Clinical islet transplantation: advances and immunological challenges. Nat Rev Immunol 2004; 4:259-68; PMID:15057784; http:// dx.doi.org/10.1038/nri1332 Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL, Kneteman NM, Rajotte RV. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med 2000; 343:230-8; PMID:10911004; http://dx.doi.org/10.1056/ NEJM200007273430401 Ryan EA, Lakey JR, Rajotte RV, Korbutt GS, Kin T, Imes S, Rabinovitch A, Elliott JF, Bigam D, Kneteman NM, et al. Clinical outcomes and insulin secretion after islet transplantation with the Edmonton protocol. Diabetes 2001; 50:710-9; PMID:11289033; http://dx.doi.org/10.2337/diabetes.50.4.710 Kizilel S, Garfinkel M, Opara E. The bioartificial pancreas: progress and challenges. Diabetes Technol Ther 2005; 7:968-85; PMID:16386103; http:// dx.doi.org/10.1089/dia.2005.7.968 Narang AS, Mahato RI. Biological and biomaterial approaches for improved islet transplantation. Pharmacol Rev 2006; 58:194-243; PMID:16714486; http://dx.doi.org/10.1124/pr.58.2.6 Wilson JT, Chaikof EL. Challenges and emerging technologies in the immunoisolation of cells and tissues. Adv Drug Deliv Rev 2008; 60:12445; PMID:18022728; http://dx.doi.org/10.1016/j. addr.2007.08.034 Scharp DW, Downing R, Merrell RC, Greider M. Isolating the elusive islet. Diabetes 1980; 29(Suppl 1):19-30; PMID:6243591; http://dx.doi. org/10.2337/diab.29.1.S19 Britt LD, Stojeba PC, Scharp CR, Greider MH, Scharp DW. Neonatal pig pseudo-islets. A product of selective aggregation. Diabetes 1981; 30:5803; PMID:7018963; http://dx.doi.org/10.2337/ diab.30.7.580

for the lack of transplantable pancreas because the structure of the islets is important for their function. In addition, the best structures for specific functions may differ from those of native islets. Thus, the in vitro reconstitution of islets should be tested depending on factors such as the ratio of α:β cells. Expansion of the concept of islets is also important for enhancing the effectiveness of islets for transplantation. “Fiber-formed islets” represent a breakthrough in this area. Many studies have induced ICCs using various types of stem cells, some of which were able to induce ICCs that resembled native islets. However, although the structures of ICCs may be very different from intact islets, it may be possible to form pseudoislets using single cells obtained from dissociated ICCs. Thus, it is essential to integrate the concepts of pseudoislets, ICCs, and artificial islets to facilitate their practical use. Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

11. Halban PA, Wollheim CB, Blondel B, Meda P, Niesor EN, Mintz DH. The possible importance of contact between pancreatic islet cells for the control of insulin release. Endocrinology 1982; 111:8694; PMID:6123433; http://dx.doi.org/10.1210/ endo-111-1-86 12. Tze WJ, Tai J. Preparation of pseudoislets for morphological and functional studies. Transplantation 1982; 34:228-31; PMID:6815845; http://dx.doi. org/10.1097/00007890-198210000-00019 13. Hopcroft DW, Mason DR, Scott RS. Insulin secretion from perifused rat pancreatic pseudoislets. In Vitro Cell Dev Biol 1985; 21:421-7; PMID:3897182; http://dx.doi.org/10.1007/BF02620828 14. Halban PA, Powers SL, George KL, Bonner-Weir S. Spontaneous reassociation of dispersed adult rat pancreatic islet cells into aggregates with three-dimensional architecture typical of native islets. Diabetes 1987; 36:783-90; PMID:3556277; http://dx.doi. org/10.2337/diab.36.7.783 15. 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; 97:935-9; PMID:6350323; http://dx.doi.org/10.1083/jcb.97.3.935 16. Schröder D, Wegner U, Hehmke B, Besch W, Zühlke H. Pancreatic islet cell suspensions of newborn rats and the formation of pseudo-islets in culture. Acta Biol Med Ger 1982; 41:1145-50; PMID:6201029 17. Schröder D, Wegner U, Besch W, Zühlke H. Characterization of pseudo-islets formed from pancreatic islet cell suspensions of neonatal rats. Mol Cell Endocrinol 1983; 32:179-93; PMID:6357894; http://dx.doi.org/10.1016/0303-7207(83)90081-3 18. Kuo CY, Herrod HG, Burghen GA. Formation of pseudoislets from human pancreatic cultures. Pancreas 1992; 7:320-5; PMID:1375749; http:// dx.doi.org/10.1097/00006676-199205000-00008 19. Farkas G, Joó F. Simple and reliable conditions for routine, long-term culturing of fetal human pancreatic tissue fragments. Diabetes 1984; 33:11658; PMID:6389232; http://dx.doi.org/10.2337/ diab.33.12.1165 20. Halban PA, Powers SL, George KL, Bonner-Weir S. Altered differentiated cell surface properties of transformed (RINm5F) compared with native adult rat pancreatic B cells. Endocrinology 1988; 123:1139; PMID:3289893; http://dx.doi.org/10.1210/ endo-123-1-113

21. Rouiller DG, Cirulli V, Halban PA. Uvomorulin mediates calcium-dependent aggregation of islet cells, whereas calcium-independent cell adhesion molecules distinguish between islet cell types. Dev Biol 1991; 148:233-42; PMID:1936561; http://dx.doi. org/10.1016/0012-1606(91)90332-W 22. Cirulli V, Halban PA, Rouiller DG. Tumor necrosis factor-alpha modifies adhesion properties of rat islet B cells. J Clin Invest 1993; 91:1868-76; PMID:8098044; http://dx.doi.org/10.1172/ JCI116403 23. Cirulli V, Baetens D, Rutishauser U, Halban PA, Orci L, Rouiller DG. Expression of neural cell adhesion molecule (N-CAM) in rat islets and its role in islet cell type segregation. J Cell Sci 1994; 107:1429-36; PMID:7962186 24. Orci L, Malaisse-Lagae F, Ravazzola M, Rouiller D, Renold AE, Perrelet A, Unger R. A morphological basis for intercellular communication between alphaand beta-cells in the endocrine pancreas. J Clin Invest 1975; 56:1066-70; PMID:1099118; http://dx.doi. org/10.1172/JCI108154 25. Orci L, Unger RH. Functional subdivision of islets of Langerhans and possible role of D cells. Lancet 1975; 2:1243-4; PMID:53729; http://dx.doi.org/10.1016/ S0140-6736(75)92078-4 26. Unger RH, Dobbs RE, Orci L. Insulin, glucagon, and somatostatin secretion in the regulation of metabolism. Annu Rev Physiol 1978; 40:307-43; PMID:205166; http://dx.doi.org/10.1146/annurev. ph.40.030178.001515 27. Orci L. Macro- and micro-domains in the endocrine pancreas. Diabetes 1982; 31:538-65; PMID:6759269; http://dx.doi.org/10.2337/diab.31.6.538 28. Weir GC, Bonner-Weir S. Islets of Langerhans: the puzzle of intraislet interactions and their relevance to diabetes. J Clin Invest 1990; 85:9837; PMID:1969425; http://dx.doi.org/10.1172/ JCI114574 29. Hopcroft DW, Mason DR, Scott RS. Structurefunction relationships in pancreatic islets: support for intraislet modulation of insulin secretion. Endocrinology 1985; 117:2073-80; PMID:2864239; http://dx.doi.org/10.1210/endo-117-5-2073 30. Hauge-Evans AC, Squires PE, Persaud SJ, Jones PM. Pancreatic beta-cell-to-beta-cell interactions are required for integrated responses to nutrient stimuli: enhanced Ca2+ and insulin secretory responses of MIN6 pseudoislets. Diabetes 1999; 48:14028; PMID:10389845; http://dx.doi.org/10.2337/ diabetes.48.7.1402

228 Organogenesis Volume 10 Issue 2

©2014 Landes Bioscience. Do not distribute.

possible that single cells from trypsinized ICCs can be used to reconstitute pseudoislets. Blastocyst complementation allows pancreatic replacement by donor cells in pigs,97 thereby indicating that human ES or iPS cells may contribute to pancreatic organogenesis. This approach may be used to generate pancreatic islets with an intact architecture via capsulation with immunoisolation materials, because blood vessels would not be able to replace the donor cells. Human endocrine cells isolated from humanized pancreas may also be used to form pseudoislets.

45. Matta SG, Wobken JD, Williams FG, Bauer GE. Pancreatic islet cell reaggregation systems: efficiency of cell reassociation and endocrine cell topography of rat islet-like aggregates. Pancreas 1994; 9:439-49; PMID:7937692; http://dx.doi. org/10.1097/00006676-199407000-00005 46. Lee DY, Park SJ, Nam JH, Byun Y. Optimal aggregation of dissociated islet cells for functional islet-like cluster. J Biomater Sci Polym Ed 2008; 19:441-52; PMID:18318957; http://dx.doi. org/10.1163/156856208783719527 47. Ohgawara H, Mochizuki N, Karibe S, Omori Y. Survival and B-cell function of neonatal pig pancreatic islet-like cell clusters in an extracellular matrix. Pancreas 1991; 6:625-30; PMID:1723523; http:// dx.doi.org/10.1097/00006676-199111000-00002 48. Otonkoski T, Beattie GM, Mally MI, Ricordi C, Hayek A. Nicotinamide is a potent inducer of endocrine differentiation in cultured human fetal pancreatic cells. J Clin Invest 1993; 92:145966; PMID:8104197; http://dx.doi.org/10.1172/ JCI116723 49. Beattie GM, Rubin JS, Mally MI, Otonkoski T, Hayek A. Regulation of proliferation and differentiation of human fetal pancreatic islet cells by extracellular matrix, hepatocyte growth factor, and cell-cell contact. Diabetes 1996; 45:1223-8; PMID:8772726; http://dx.doi.org/10.2337/diab.45.9.1223 50. Caton D, Calabrese A, Mas C, Serre-Beinier V, Charollais A, Caille D, Zufferey R, Trono D, Meda P. Lentivirus-mediated transduction of connexin cDNAs shows level- and isoform-specific alterations in insulin secretion of primary pancreatic beta-cells. J Cell Sci 2003; 116:2285-94; PMID:12697840; http://dx.doi.org/10.1242/jcs.00442 51. Yang XD, Li HM, Chen M, Zou XH, Zhu LY, Wei CJ, Chen GQ. Enhanced insulin production from murine islet beta cells incubated on poly(3-hydroxybutyrate-co-3-hydroxyhexanoate). J Biomed Mater Res A 2010; 92:548-55; PMID:19235213 52. Pethig R, Menachery A, Heart E, Sanger RH, Smith PJ. Dielectrophoretic assembly of insulinoma cells and fluorescent nanosensors into three-dimensional pseudo-islet constructs. IET Nanobiotechnol 2008; 2:31; PMID:18500910; http://dx.doi.org/10.1049/ iet-nbt:20070027 53. Tsang WG, Zheng T, Wang Y, Tang J, Rind HB, Francki A, Bufius N. Generation of functional islet-like clusters after monolayer culture and intracapsular aggregation of adult human pancreatic islet tissue. Transplantation 2007; 83:685-93; PMID:17414699; http://dx.doi.org/10.1097/01. tp.0000256178.57359.4f Tze WJ, Tai J. Allotransplantation and 54. xenotransplantation of pseudoislets in diabetic rats and mice. Transplantation 1984; 38:438-40; PMID:6437026; http://dx.doi. org/10.1097/00007890-198410000-00026 55. Wolf-Jochim M, Wöhrle M, Federlin K, Bretzel RG. Comparison of the survival of fresh or cultured pancreatic islets, pseudoislets and single cells following allotransplantation beneath the kidney capsule in non-immunosuppressed diabetic rats. Exp Clin Endocrinol Diabetes 1995; 103(Suppl 2):118-22; PMID:8839267; http://dx.doi. org/10.1055/s-0029-1211407 56. Beger C, Cirulli V, Vajkoczy P, Halban PA, Menger MD. Vascularization of purified pancreatic islet-like cell aggregates (pseudoislets) after syngeneic transplantation. Diabetes 1998; 47:55965; PMID:9568687; http://dx.doi.org/10.2337/ diabetes.47.4.559 57. Penko D, Mohanasundaram D, Sen S, Drogemuller C, Mee C, Bonder CS, Coates PT, Jessup CF. Incorporation of endothelial progenitor cells into mosaic pseudoislets. Islets 2011; 3:73-9; PMID:21478677; http://dx.doi.org/10.4161/ isl.3.3.15392

58. Teramura Y, Iwata H. Bioartificial pancreas microencapsulation and conformal coating of islet of Langerhans. Adv Drug Deliv Rev 2010; 62:82740; PMID:20138097; http://dx.doi.org/10.1016/j. addr.2010.01.005 59. Ohgawara H, Miyazaki J, Karibe S, Tashiro F, Akaike T, Hashimoto Y. Embedded-culture of pancreatic beta-cells derived from transgenic mouse insulinoma as a potential source for xenotransplantation using a diffusion chamber. Cell Transplant 1995; 4:307-13; PMID:7640870; http://dx.doi. org/10.1016/0963-6897(95)00001-E 60. Sakai S, Inagaki H, Inamoto K, Taya M. Wrapping tissues with a pre-established cage-like layer composed of living cells. Biomaterials 2012; 33:67217; PMID:22770525; http://dx.doi.org/10.1016/j. biomaterials.2012.06.027 61. Onoe H, Okitsu T, Itou A, Kato-Negishi M, Gojo R, Kiriya D, Sato K, Miura S, Iwanaga S, KuribayashiShigetomi K, et al. Metre-long cell-laden microfibres exhibit tissue morphologies and functions. Nat Mater 2013; 12:584-90; PMID:23542870; http://dx.doi. org/10.1038/nmat3606 62. Mason MN, Mahoney MJ. Selective beta-cell differentiation of dissociated embryonic pancreatic precursor cells cultured in synthetic polyethylene glycol hydrogels. Tissue Eng Part A 2009; 15:134352; PMID:19072086; http://dx.doi.org/10.1089/ten. tea.2008.0290 63. Mason MN, Arnold CA, Mahoney MJ. Entrapped collagen type 1 promotes differentiation of embryonic pancreatic precursor cells into glucose-responsive beta-cells when cultured in three-dimensional PEG hydrogels. Tissue Eng Part A 2009; 15:3799-808; PMID:19537960; http://dx.doi.org/10.1089/ten. tea.2009.0148 64. Mason MN, Mahoney MJ. Inhibition of gammasecretase activity promotes differentiation of embryonic pancreatic precursor cells into functional islet-like clusters in poly(ethylene glycol) hydrogel culture. Tissue Eng Part A 2010; 16:2593-603; PMID:20236034; http://dx.doi.org/10.1089/ten. tea.2010.0015 65. Datar SP, Bhonde RR. Islet-derived stellate-like cells as a novel source for islet neogenesis in chicks. Poult Sci 2009; 88:654-60; PMID:19211538; http:// dx.doi.org/10.3382/ps.2007-00440 66. Todorov I, Omori K, Pascual M, Rawson J, Nair I, Valiente L, Vuong T, Matsuda T, Orr C, Ferreri K, et al. Generation of human islets through expansion and differentiation of non-islet pancreatic cells discarded (pancreatic discard) after islet isolation. Pancreas 2006; 32:130-8; PMID:16552331; http:// dx.doi.org/10.1097/01.mpa.0000202945.78331.93 67. Zanini C, Bruno S, Mandili G, Baci D, Cerutti F, Cenacchi G, Izzi L, Camussi G, Forni M. Differentiation of mesenchymal stem cells derived from pancreatic islets and bone marrow into isletlike cell phenotype. PLoS One 2011; 6:e28175; PMID:22194812; http://dx.doi.org/10.1371/journal. pone.0028175 68. Koblas T, Pektorova L, Zacharovova K, Berkova Z, Girman P, Dovolilova E, Karasova L, Saudek F. Differentiation of CD133-positive pancreatic cells into insulin-producing islet-like cell clusters. Transplant Proc 2008; 40:415-8; PMID:18374086; http:// dx.doi.org/10.1016/j.transproceed.2008.02.017 69. Tang DQ, Cao LZ, Burkhardt BR, Xia CQ, Litherland SA, Atkinson MA, Yang LJ. In vivo and in vitro characterization of insulin-producing cells obtained from murine bone marrow. Diabetes 2004; 53:1721-32; PMID:15220196; http://dx.doi. org/10.2337/diabetes.53.7.1721 70. Thatava T, Ma B, Rohde M, Mayer H. Chromatinremodeling factors allow differentiation of bone marrow cells into insulin-producing cells. Stem Cells 2006; 24:2858-67; PMID:16990588; http://dx.doi. org/10.1634/stemcells.2006-0109

www.landesbioscience.com Organogenesis 229

©2014 Landes Bioscience. Do not distribute.

31. Squires PE, Hauge-Evans AC, Persaud SJ, Jones PM. Synchronization of Ca(2+)-signals within insulin-secreting pseudoislets: effects of gap-junctional uncouplers. Cell Calcium 2000; 27:28796; PMID:10859595; http://dx.doi.org/10.1054/ ceca.2000.0117 32. Hauge-Evans AC, Squires PE, Belin VD, RoderigoMilne H, Ramracheya RD, Persaud SJ, Jones PM. Role of adenine nucleotides in insulin secretion from MIN6 pseudoislets. Mol Cell Endocrinol 2002; 191:167-76; PMID:12062900; http://dx.doi. org/10.1016/S0303-7207(02)00051-5 33. Luther MJ, Davies E, Muller D, Harrison M, Bone AJ, Persaud SJ, Jones PM. Cell-to-cell contact influences proliferative marker expression and apoptosis in MIN6 cells grown in islet-like structures. Am J Physiol Endocrinol Metab 2005; 288:E5029; PMID:15479950; http://dx.doi.org/10.1152/ ajpendo.00424.2004 34. Luther MJ, Hauge-Evans A, Souza KL, Jörns A, Lenzen S, Persaud SJ, Jones PM. MIN6 beta-cell-betacell interactions influence insulin secretory responses to nutrients and non-nutrients. Biochem Biophys Res Commun 2006; 343:99-104; PMID:16529716; http://dx.doi.org/10.1016/j.bbrc.2006.02.003 35. Brereton HC, Carvell MJ, Asare-Anane H, Roberts G, Christie MR, Persaud SJ, Jones PM. Homotypic cell contact enhances insulin but not glucagon secretion. Biochem Biophys Res Commun 2006; 344:9951000; PMID:16643853; http://dx.doi.org/10.1016/j. bbrc.2006.03.214 36. Carvell MJ, Marsh PJ, Persaud SJ, Jones PM. E-cadherin interactions regulate beta-cell proliferation in islet-like structures. Cell Physiol Biochem 2007; 20:617-26; PMID:17762188; http://dx.doi. org/10.1159/000107545 37. Kitsou-Mylona I, Burns CJ, Squires PE, Persaud SJ, Jones PM. A role for the extracellular calciumsensing receptor in cell-cell communication in pancreatic islets of langerhans. Cell Physiol Biochem 2008; 22:557-66; PMID:19088438; http://dx.doi. org/10.1159/000185540 38. Reers C, Hauge-Evans AC, Morgan NG, Wilcox A, Persaud SJ, Jones PM. Down-regulation of proliferation does not affect the secretory function of transformed β-cell lines regardless of their anatomical configuration. Islets 2011; 3:80-8; PMID:21505275; http://dx.doi.org/10.4161/isl.3.3.15428 39. Josefsen K, Stenvang JP, Kindmark H, Berggren PO, Horn T, Kjaer T, Buschard K. Fluorescence-activated cell sorted rat islet cells and studies of the insulin secretory process. J Endocrinol 1996; 149:14554; PMID:8676047; http://dx.doi.org/10.1677/ joe.0.1490145 40. Hamaguchi K, Utsunomiya N, Takaki R, Yoshimatsu H, Sakata T. Cellular interaction between mouse pancreatic alpha-cell and beta-cell lines: possible contact-dependent inhibition of insulin secretion. Exp Biol Med (Maywood) 2003; 228:1227-33; PMID:14610265 41. Brereton H, Carvell MJ, Persaud SJ, Jones PM. Islet al.ha-cells do not influence insulin secretion from beta-cells through cell-cell contact. Endocrine 2007; 31:61-5; PMID:17709899; http://dx.doi. org/10.1007/s12020-007-0004-0 42. Kojima N, Takeuchi S, Sakai Y. Engineering of Pseudoislets: Effect on Insulin Secretion Activity by Cell Number, Cell Population, and Microchannel Networks. Transplant Proc 2014; (Forthcoming) 43. Brissova M, Fowler MJ, Nicholson WE, Chu A, Hirshberg B, Harlan DM, Powers AC. Assessment of human pancreatic islet architecture and composition by laser scanning confocal microscopy. J Histochem Cytochem 2005; 53:1087-97; PMID:15923354; http://dx.doi.org/10.1369/jhc.5C6684.2005 44. Kim A, Miller K, Jo J, Kilimnik G, Wojcik P, Hara M. Islet architecture: A comparative study. Islets 2009; 1:129-36; PMID:20606719; http://dx.doi. org/10.4161/isl.1.2.9480

80. Kadam SS, Bhonde RR. Islet neogenesis from the constitutively nestin expressing human umbilical cord matrix derived mesenchymal stem cells. Islets 2010; 2:112-20; PMID:21099303; http://dx.doi. org/10.4161/isl.2.2.11280 81. Kadam S, Muthyala S, Nair P, Bhonde R. Human placenta-derived mesenchymal stem cells and isletlike cell clusters generated from these cells as a novel source for stem cell therapy in diabetes. Rev Diabet Stud 2010; 7:168-82; PMID:21060975; http:// dx.doi.org/10.1900/RDS.2010.7.168 82. Kadam SS, Sudhakar M, Nair PD, Bhonde RR. Reversal of experimental diabetes in mice by transplantation of neo-islets generated from human amnion-derived mesenchymal stromal cells using immuno-isolatory macrocapsules. Cytotherapy 2010; 12:982-91; PMID:20807019; http://dx.doi.org/10.3 109/14653249.2010.509546 83. Bi D, Chen FG, Zhang WJ, Zhou GD, Cui L, Liu W, Cao Y. Differentiation of human multipotent dermal fibroblasts into islet-like cell clusters. BMC Cell Biol 2010; 11:46; PMID:20579360; http://dx.doi. org/10.1186/1471-2121-11-46 84. Chiou SH, Chen SJ, Chang YL, Chen YC, Li HY, Chen DT, Wang HH, Chang CM, Chen YJ, Ku HH. MafA promotes the reprogramming of placenta-derived multipotent stem cells into pancreatic islets-like and insulin+ cells. J Cell Mol Med 2011; 15:612-24; PMID:20158571; http://dx.doi. org/10.1111/j.1582-4934.2010.01034.x 85. Govindasamy V, Ronald VS, Abdullah AN, Nathan KR, Ab Aziz ZA, Abdullah M, Musa S, Kasim NH, Bhonde RR. Differentiation of dental pulp stem cells into islet-like aggregates. J Dent Res 2011; 90:646-52; PMID:21335539; http://dx.doi. org/10.1177/0022034510396879 86. Hori Y, Rulifson IC, Tsai BC, Heit JJ, Cahoy JD, Kim SK. Growth inhibitors promote differentiation of insulin-producing tissue from embryonic stem cells. Proc Natl Acad Sci U S A 2002; 99:1610510; PMID:12441403; http://dx.doi.org/10.1073/ pnas.252618999 87. Schroeder IS, Rolletschek A, Blyszczuk P, Kania G, Wobus AM. Differentiation of mouse embryonic stem cells to insulin-producing cells. Nat Protoc 2006; 1:495-507; PMID:17406275; http://dx.doi. org/10.1038/nprot.2006.71 88. Wang X, Ye K. Three-dimensional differentiation of embryonic stem cells into islet-like insulin-producing clusters. Tissue Eng Part A 2009; 15:1941-52; PMID:19196138; http://dx.doi.org/10.1089/ten. tea.2008.0181

89. Kubo A, Stull R, Takeuchi M, Bonham K, GouonEvans V, Sho M, Iwano M, Saito Y, Keller G, Snodgrass R. Pdx1 and Ngn3 overexpression enhances pancreatic differentiation of mouse ES cell-derived endoderm population. PLoS One 2011; 6:e24058; PMID:21931641; http://dx.doi.org/10.1371/journal. pone.0024058 90. Yue F, Cui L, Johkura K, Ogiwara N, Sasaki K. Glucagon-like peptide-1 differentiation of primate embryonic stem cells into insulin-producing cells. Tissue Eng 2006; 12:2105-16; PMID:16968152; http://dx.doi.org/10.1089/ten.2006.12.2105 91. Segev H, Fishman B, Ziskind A, Shulman M, Itskovitz-Eldor J. Differentiation of human embryonic stem cells into insulin-producing clusters. Stem Cells 2004; 22:265-74; PMID:15153604; http:// dx.doi.org/10.1634/stemcells.22-3-265 92. Jiang J, Au M, Lu K, Eshpeter A, Korbutt G, Fisk G, Majumdar AS. Generation of insulin-producing isletlike clusters from human embryonic stem cells. Stem Cells 2007; 25:1940-53; PMID:17510217; http:// dx.doi.org/10.1634/stemcells.2006-0761 93. Mao GH, Chen GA, Bai HY, Song TR, Wang YX. The reversal of hyperglycaemia in diabetic mice using PLGA scaffolds seeded with islet-like cells derived from human embryonic stem cells. Biomaterials 2009; 30:1706-14; PMID:19135250; http://dx.doi. org/10.1016/j.biomaterials.2008.12.030 94. Schulz TC, Young HY, Agulnick AD, Babin MJ, Baetge EE, Bang AG, Bhoumik A, Cepa I, Cesario RM, Haakmeester C, et al. A scalable system for production of functional pancreatic progenitors from human embryonic stem cells. PLoS One 2012; 7:e37004; PMID:22623968; http://dx.doi. org/10.1371/journal.pone.0037004 95. Saito H, Takeuchi M, Chida K, Miyajima A. Generation of glucose-responsive functional islets with a three-dimensional structure from mouse fetal pancreatic cells and iPS cells in vitro. PLoS One 2011; 6:e28209; PMID:22145030; http://dx.doi. org/10.1371/journal.pone.0028209 96. Thatava T, Nelson TJ, Edukulla R, Sakuma T, Ohmine S, Tonne JM, Yamada S, Kudva Y, Terzic A, Ikeda Y. Indolactam V/GLP-1-mediated differentiation of human iPS cells into glucose-responsive insulin-secreting progeny. Gene Ther 2011; 18:28393; PMID:21048796; http://dx.doi.org/10.1038/ gt.2010.145 97. Matsunari H, Nagashima H, Watanabe M, Umeyama K, Nakano K, Nagaya M, Kobayashi T, Yamaguchi T, Sumazaki R, Herzenberg LA, et al. Blastocyst complementation generates exogenic pancreas in vivo in apancreatic cloned pigs. Proc Natl Acad Sci U S A 2013; 110:4557-62; PMID:23431169; http://dx.doi. org/10.1073/pnas.1222902110

230 Organogenesis Volume 10 Issue 2

©2014 Landes Bioscience. Do not distribute.

71. Oh SH, Muzzonigro TM, Bae SH, LaPlante JM, Hatch HM, Petersen BE. Adult bone marrow-derived cells trans-differentiating into insulin-producing cells for the treatment of type I diabetes. Lab Invest 2004; 84:607-17; PMID:15034596; http://dx.doi. org/10.1038/labinvest.3700074 72. Li L, Li F, Qi H, Feng G, Yuan K, Deng H, Zhou H. Coexpression of Pdx1 and betacellulin in mesenchymal stem cells could promote the differentiation of nestin-positive epithelium-like progenitors and pancreatic islet-like spheroids. Stem Cells Dev 2008; 17:815-23; PMID:18439098; http://dx.doi. org/10.1089/scd.2008.0060 73. Xie QP, Huang H, Xu B, Dong X, Gao SL, Zhang B, Wu YL. Human bone marrow mesenchymal stem cells differentiate into insulin-producing cells upon microenvironmental manipulation in vitro. Differentiation 2009; 77:483-91; PMID:19505629; http://dx.doi.org/10.1016/j.diff.2009.01.001 74. Chandra V, G S, Phadnis S, Nair PD, Bhonde RR. Generation of pancreatic hormone-expressing isletlike cell aggregates from murine adipose tissuederived stem cells. Stem Cells 2009; 27:1941-53; PMID:19544426; http://dx.doi.org/10.1002/ stem.117 75. Okura H, Komoda H, Fumimoto Y, Lee CM, Nishida T, Sawa Y, Matsuyama A. Transdifferentiation of human adipose tissue-derived stromal cells into insulin-producing clusters. J Artif Organs 2009; 12:12330; PMID:19536630; http://dx.doi.org/10.1007/ s10047-009-0455-6 76. Chandra V, Swetha G, Muthyala S, Jaiswal AK, Bellare JR, Nair PD, Bhonde RR. Islet-like cell aggregates generated from human adipose tissue derived stem cells ameliorate experimental diabetes in mice. PLoS One 2011; 6:e20615; PMID:21687731; http:// dx.doi.org/10.1371/journal.pone.0020615 77. Ruhnke M, Ungefroren H, Nussler A, Martin F, Brulport M, Schormann W, Hengstler JG, Klapper W, Ulrichs K, Hutchinson JA, et al. Differentiation of in vitro-modified human peripheral blood monocytes into hepatocyte-like and pancreatic islet-like cells. Gastroenterology 2005; 128:177486; PMID:15940611; http://dx.doi.org/10.1053/j. gastro.2005.03.029 78. Sun B, Roh KH, Lee SR, Lee YS, Kang KS. Induction of human umbilical cord blood-derived stem cells with embryonic stem cell phenotypes into insulin producing islet-like structure. Biochem Biophys Res Commun 2007; 354:919-23; PMID:17274951; http://dx.doi.org/10.1016/j.bbrc.2007.01.069 79. 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; 283:31601-7; PMID:18782754; http://dx.doi. org/10.1074/jbc.M806597200

In vitro reconstitution of pancreatic islets.

The lack of transplantable pancreatic islets is a serious problem that affects the treatment of patients with type 1 diabetes mellitus. Beta cells can...
632KB Sizes 1 Downloads 3 Views