Skip to main content
Top
Published in: Current Diabetes Reports 5/2016

01-05-2016 | Immunology and Transplantation (L Piemonti and V Sordi, Section Editors)

New Insights into Diabetes Cell Therapy

Authors: Philippe A. Lysy, Elisa Corritore, Etienne M. Sokal

Published in: Current Diabetes Reports | Issue 5/2016

Login to get access

Abstract

Since insulin discovery, islet transplantation was the first protocol to show the possibility to cure patients with type 1 diabetes using low-risk procedures. The scarcity of pancreas donors triggered a burst of studies focused on the production of new β cells in vitro. These were rapidly dominated by pluripotent stem cells (PSCs) demonstrating diabetes-reversal potential in diabetic mice. Subsequent enthusiasm fostered a clinical trial with immunoisolated embryonic-derived pancreatic progenitors. Yet safety is the Achilles’ heel of PSCs, and a whole branch of β cell engineering medicine focuses on transdifferentiation of adult pancreatic cells. New data showed the possibility to chemically stimulate acinar or α cells to undergo β cell neogenesis and provide opportunities to intervene in situ without the need for a transplant, at least after weighing benefits against systemic adverse effects. The current studies suggested the pancreas as a reservoir of facultative progenitors (e.g., in the duct lining) could be exploited ex vivo for expansion and β cell differentiation in timely fashion and without the hurdles of PSC use. Diabetes cell therapy is thus a growing field not only with great potential but also with many pitfalls to overcome for becoming fully envisioned as a competitor to the current treatment standards.
Literature
1.
go back to reference van Belle TL, Coppieters KT, von Herrath MG. Type 1 diabetes: etiology, immunology, and therapeutic strategies. Physiol Rev. 2011;91(1):79–118.CrossRefPubMed van Belle TL, Coppieters KT, von Herrath MG. Type 1 diabetes: etiology, immunology, and therapeutic strategies. Physiol Rev. 2011;91(1):79–118.CrossRefPubMed
2.
go back to reference Michels A et al. Prediction and prevention of type 1 diabetes: update on success of prediction and struggles at prevention. Pediatr Diabetes. 2015;16(7):465–84.CrossRefPubMed Michels A et al. Prediction and prevention of type 1 diabetes: update on success of prediction and struggles at prevention. Pediatr Diabetes. 2015;16(7):465–84.CrossRefPubMed
3.
go back to reference Cameron FJ, Wherrett DK. Care of diabetes in children and adolescents: controversies, changes, and consensus. Lancet. 2015;385(9982):2096–106.CrossRefPubMed Cameron FJ, Wherrett DK. Care of diabetes in children and adolescents: controversies, changes, and consensus. Lancet. 2015;385(9982):2096–106.CrossRefPubMed
4.
go back to reference Lind M et al. Glycemic control and excess mortality in type 1 diabetes. N Engl J Med. 2014;371(21):1972–82.CrossRefPubMed Lind M et al. Glycemic control and excess mortality in type 1 diabetes. N Engl J Med. 2014;371(21):1972–82.CrossRefPubMed
5.
go back to reference Hampp C et al. Use of antidiabetic drugs in the U.S., 2003–2012. Diabetes Care. 2014;37(5):1367–74.CrossRefPubMed Hampp C et al. Use of antidiabetic drugs in the U.S., 2003–2012. Diabetes Care. 2014;37(5):1367–74.CrossRefPubMed
6.
go back to reference Kelly WD et al. Allotransplantation of the pancreas and duodenum along with the kidney in diabetic nephropathy. Surgery. 1967;61(6):827–37.PubMed Kelly WD et al. Allotransplantation of the pancreas and duodenum along with the kidney in diabetic nephropathy. Surgery. 1967;61(6):827–37.PubMed
7.
go back to reference Kaufman, D. State of the art of solid organ pancreas transplantation. in 75th Scientific Sessions of the American Diabetes Association. 2015. Boston, USA. Kaufman, D. State of the art of solid organ pancreas transplantation. in 75th Scientific Sessions of the American Diabetes Association. 2015. Boston, USA.
9.
10.
go back to reference Redfield RR, Scalea JR, Odorico JS. Simultaneous pancreas and kidney transplantation: current trends and future directions. Curr Opin Organ Transplant. 2015;20(1):94–102.CrossRefPubMedPubMedCentral Redfield RR, Scalea JR, Odorico JS. Simultaneous pancreas and kidney transplantation: current trends and future directions. Curr Opin Organ Transplant. 2015;20(1):94–102.CrossRefPubMedPubMedCentral
11.
go back to reference Ricordi C et al. Automated method for isolation of human pancreatic islets. Diabetes. 1988;37(4):413–20.CrossRefPubMed Ricordi C et al. Automated method for isolation of human pancreatic islets. Diabetes. 1988;37(4):413–20.CrossRefPubMed
12.
go back to reference Shapiro AM et al. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 2000;343(4):230–8.CrossRefPubMed Shapiro AM et al. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 2000;343(4):230–8.CrossRefPubMed
14.••
go back to reference Balamurugan AN et al. Islet product characteristics and factors related to successful human islet transplantation from the Collaborative Islet Transplant Registry (CITR) 1999–2010. Am J Transplant. 2014;14(11):2595–606. This work highlights the efforts of the CITR to develop successful state-of-the-art human islet transplantation protocols.CrossRefPubMedPubMedCentral Balamurugan AN et al. Islet product characteristics and factors related to successful human islet transplantation from the Collaborative Islet Transplant Registry (CITR) 1999–2010. Am J Transplant. 2014;14(11):2595–606. This work highlights the efforts of the CITR to develop successful state-of-the-art human islet transplantation protocols.CrossRefPubMedPubMedCentral
15.
go back to reference Brennan, D.C., et al., Long-term follow-up of the Edmonton Protocol of islet transplantation in the United States. Am J Transplant, 2015. Brennan, D.C., et al., Long-term follow-up of the Edmonton Protocol of islet transplantation in the United States. Am J Transplant, 2015.
16.
go back to reference Ricordi, C. Clinical islet transplantation update. in 75th Scientific Sessions of the American Diabetes Association. 2015. Boston, USA. Ricordi, C. Clinical islet transplantation update. in 75th Scientific Sessions of the American Diabetes Association. 2015. Boston, USA.
17.
go back to reference Inverardi, L. Improved graft survival in islet transplant recipients treated with G-CSF (filgrastim) and exenatide. in 75th Scientific Sessions of the American Diabetes Association. 2015. Boston, USA. Inverardi, L. Improved graft survival in islet transplant recipients treated with G-CSF (filgrastim) and exenatide. in 75th Scientific Sessions of the American Diabetes Association. 2015. Boston, USA.
18.
go back to reference Zoso A et al. Human fibrocytic myeloid-derived suppressor cells express IDO and promote tolerance via Treg-cell expansion. Eur J Immunol. 2014;44(11):3307–19.CrossRefPubMed Zoso A et al. Human fibrocytic myeloid-derived suppressor cells express IDO and promote tolerance via Treg-cell expansion. Eur J Immunol. 2014;44(11):3307–19.CrossRefPubMed
19.
go back to reference Haller MJ et al. Anti-thymocyte globulin/G-CSF treatment preserves beta cell function in patients with established type 1 diabetes. J Clin Invest. 2015;125(1):448–55.CrossRefPubMedPubMedCentral Haller MJ et al. Anti-thymocyte globulin/G-CSF treatment preserves beta cell function in patients with established type 1 diabetes. J Clin Invest. 2015;125(1):448–55.CrossRefPubMedPubMedCentral
20.
go back to reference Keymeulen B et al. Correlation between beta cell mass and glycemic control in type 1 diabetic recipients of islet cell graft. Proc Natl Acad Sci U S A. 2006;103(46):17444–9.CrossRefPubMedPubMedCentral Keymeulen B et al. Correlation between beta cell mass and glycemic control in type 1 diabetic recipients of islet cell graft. Proc Natl Acad Sci U S A. 2006;103(46):17444–9.CrossRefPubMedPubMedCentral
21.
go back to reference Piemonti L et al. Effects of cryopreservation on in vitro and in vivo long-term function of human islets. Transplantation. 1999;68(5):655–62.CrossRefPubMed Piemonti L et al. Effects of cryopreservation on in vitro and in vivo long-term function of human islets. Transplantation. 1999;68(5):655–62.CrossRefPubMed
22.
go back to reference Lakey JR, Anderson TJ, Rajotte RV. Novel approaches to cryopreservation of human pancreatic islets. Transplantation. 2001;72(6):1005–11.CrossRefPubMed Lakey JR, Anderson TJ, Rajotte RV. Novel approaches to cryopreservation of human pancreatic islets. Transplantation. 2001;72(6):1005–11.CrossRefPubMed
24.
go back to reference Pepper AR et al. A prevascularized subcutaneous device-less site for islet and cellular transplantation. Nat Biotechnol. 2015;33(5):518–23.CrossRefPubMed Pepper AR et al. A prevascularized subcutaneous device-less site for islet and cellular transplantation. Nat Biotechnol. 2015;33(5):518–23.CrossRefPubMed
25.
go back to reference Phelps EA et al. Vasculogenic bio-synthetic hydrogel for enhancement of pancreatic islet engraftment and function in type 1 diabetes. Biomaterials. 2013;34(19):4602–11.CrossRefPubMedPubMedCentral Phelps EA et al. Vasculogenic bio-synthetic hydrogel for enhancement of pancreatic islet engraftment and function in type 1 diabetes. Biomaterials. 2013;34(19):4602–11.CrossRefPubMedPubMedCentral
26.
go back to reference Samikannu B et al. Dipeptidyl peptidase IV inhibition activates CREB and improves islet vascularization through VEGF-A/VEGFR-2 signaling pathway. PLoS One. 2013;8(12):e82639.CrossRefPubMedPubMedCentral Samikannu B et al. Dipeptidyl peptidase IV inhibition activates CREB and improves islet vascularization through VEGF-A/VEGFR-2 signaling pathway. PLoS One. 2013;8(12):e82639.CrossRefPubMedPubMedCentral
27.
go back to reference Hajizadeh-Saffar E et al. Inducible VEGF expression by human embryonic stem cell-derived mesenchymal stromal cells reduces the minimal islet mass required to reverse diabetes. Sci Rep. 2015;5:9322.CrossRefPubMedPubMedCentral Hajizadeh-Saffar E et al. Inducible VEGF expression by human embryonic stem cell-derived mesenchymal stromal cells reduces the minimal islet mass required to reverse diabetes. Sci Rep. 2015;5:9322.CrossRefPubMedPubMedCentral
28.
go back to reference Calafiore R, Basta G. Clinical application of microencapsulated islets: actual prospectives on progress and challenges. Adv Drug Deliv Rev. 2014;67–68:84–92.CrossRefPubMed Calafiore R, Basta G. Clinical application of microencapsulated islets: actual prospectives on progress and challenges. Adv Drug Deliv Rev. 2014;67–68:84–92.CrossRefPubMed
29.
go back to reference Boettler, T., et al., Pancreatic tissue transplanted in TheraCyte encapsulation devices are protected and prevent hyperglycemia in a mouse model of immune-mediated diabetes. Cell Transplant, 2015. Boettler, T., et al., Pancreatic tissue transplanted in TheraCyte encapsulation devices are protected and prevent hyperglycemia in a mouse model of immune-mediated diabetes. Cell Transplant, 2015.
31.
go back to reference Hering BJ et al. Prolonged diabetes reversal after intraportal xenotransplantation of wild-type porcine islets in immunosuppressed nonhuman primates. Nat Med. 2006;12(3):301–3.CrossRefPubMed Hering BJ et al. Prolonged diabetes reversal after intraportal xenotransplantation of wild-type porcine islets in immunosuppressed nonhuman primates. Nat Med. 2006;12(3):301–3.CrossRefPubMed
32.
go back to reference 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(10):1054–62.CrossRefPubMed 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(10):1054–62.CrossRefPubMed
33.
go back to reference Elliott RB et al. Live encapsulated porcine islets from a type 1 diabetic patient 9.5 yr after xenotransplantation. Xenotransplantation. 2007;14(2):157–61.CrossRefPubMed Elliott RB et al. Live encapsulated porcine islets from a type 1 diabetic patient 9.5 yr after xenotransplantation. Xenotransplantation. 2007;14(2):157–61.CrossRefPubMed
34.
go back to reference Klymiuk N et al. Genetic modification of pigs as organ donors for xenotransplantation. Mol Reprod Dev. 2010;77(3):209–21.PubMed Klymiuk N et al. Genetic modification of pigs as organ donors for xenotransplantation. Mol Reprod Dev. 2010;77(3):209–21.PubMed
35.•
go back to reference Shin JS et al. Long-term control of diabetes in immunosuppressed nonhuman primates (NHP) by the transplantation of adult porcine islets. Am J Transplant. 2015;15(11):2837–50. This study shows the efficacy of combination immunotherapy in achieving diabetes reversal in primates after pig islet transplantation.CrossRefPubMed Shin JS et al. Long-term control of diabetes in immunosuppressed nonhuman primates (NHP) by the transplantation of adult porcine islets. Am J Transplant. 2015;15(11):2837–50. This study shows the efficacy of combination immunotherapy in achieving diabetes reversal in primates after pig islet transplantation.CrossRefPubMed
36.
go back to reference Soria B et al. Insulin-secreting cells derived from embryonic stem cells normalize glycemia in streptozotocin-induced diabetic mice. Diabetes. 2000;49(2):157–62.CrossRefPubMed Soria B et al. Insulin-secreting cells derived from embryonic stem cells normalize glycemia in streptozotocin-induced diabetic mice. Diabetes. 2000;49(2):157–62.CrossRefPubMed
37.
go back to reference Lumelsky N et al. Differentiation of embryonic stem cells to insulin-secreting structures similar to pancreatic islets. Science. 2001;292(5520):1389–94.CrossRefPubMed Lumelsky N et al. Differentiation of embryonic stem cells to insulin-secreting structures similar to pancreatic islets. Science. 2001;292(5520):1389–94.CrossRefPubMed
38.
go back to reference Hori Y et al. Growth inhibitors promote differentiation of insulin-producing tissue from embryonic stem cells. Proc Natl Acad Sci U S A. 2002;99(25):16105–10.CrossRefPubMedPubMedCentral Hori Y et al. Growth inhibitors promote differentiation of insulin-producing tissue from embryonic stem cells. Proc Natl Acad Sci U S A. 2002;99(25):16105–10.CrossRefPubMedPubMedCentral
39.
go back to reference Kroon E et al. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol. 2008;26(4):443–52.CrossRefPubMed Kroon E et al. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol. 2008;26(4):443–52.CrossRefPubMed
40.
go back to reference D’Amour KA et al. Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol. 2006;24(11):1392–401.CrossRefPubMed D’Amour KA et al. Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol. 2006;24(11):1392–401.CrossRefPubMed
41.
go back to reference Fujikawa T et al. Teratoma formation leads to failure of treatment for type I diabetes using embryonic stem cell-derived insulin-producing cells. Am J Pathol. 2005;166(6):1781–91.CrossRefPubMedPubMedCentral Fujikawa T et al. Teratoma formation leads to failure of treatment for type I diabetes using embryonic stem cell-derived insulin-producing cells. Am J Pathol. 2005;166(6):1781–91.CrossRefPubMedPubMedCentral
42.
go back to reference Blum B, Benvenisty N. The tumorigenicity of diploid and aneuploid human pluripotent stem cells. Cell Cycle. 2009;8(23):3822–30.CrossRefPubMed Blum B, Benvenisty N. The tumorigenicity of diploid and aneuploid human pluripotent stem cells. Cell Cycle. 2009;8(23):3822–30.CrossRefPubMed
43.
go back to reference Kahan B et al. Elimination of tumorigenic stem cells from differentiated progeny and selection of definitive endoderm reveals a Pdx1+ foregut endoderm stem cell lineage. Stem Cell Res. 2011;6(2):143–57.CrossRefPubMedPubMedCentral Kahan B et al. Elimination of tumorigenic stem cells from differentiated progeny and selection of definitive endoderm reveals a Pdx1+ foregut endoderm stem cell lineage. Stem Cell Res. 2011;6(2):143–57.CrossRefPubMedPubMedCentral
44.
go back to reference Jiang W et al. CD24: a novel surface marker for PDX1-positive pancreatic progenitors derived from human embryonic stem cells. Stem Cells. 2011;29(4):609–17.CrossRefPubMed Jiang W et al. CD24: a novel surface marker for PDX1-positive pancreatic progenitors derived from human embryonic stem cells. Stem Cells. 2011;29(4):609–17.CrossRefPubMed
45.
go back to reference Kelly OG et al. Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells. Nat Biotechnol. 2011;29(8):750–6.CrossRefPubMed Kelly OG et al. Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells. Nat Biotechnol. 2011;29(8):750–6.CrossRefPubMed
46.
go back to reference Rezania A et al. Maturation of human embryonic stem cell-derived pancreatic progenitors into functional islets capable of treating pre-existing diabetes in mice. Diabetes. 2012;61(8):2016–29.CrossRefPubMedPubMedCentral Rezania A et al. Maturation of human embryonic stem cell-derived pancreatic progenitors into functional islets capable of treating pre-existing diabetes in mice. Diabetes. 2012;61(8):2016–29.CrossRefPubMedPubMedCentral
47.
go back to reference Bruin JE et al. Maturation and function of human embryonic stem cell-derived pancreatic progenitors in macroencapsulation devices following transplant into mice. Diabetologia. 2013;56(9):1987–98.CrossRefPubMed Bruin JE et al. Maturation and function of human embryonic stem cell-derived pancreatic progenitors in macroencapsulation devices following transplant into mice. Diabetologia. 2013;56(9):1987–98.CrossRefPubMed
48.••
go back to reference Rezania A et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol. 2014;32(11):1121–33. This study demonstrate the potential of hESCs to acquire functional β cell-like functionality both in vitro and in vivo.CrossRefPubMed Rezania A et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol. 2014;32(11):1121–33. This study demonstrate the potential of hESCs to acquire functional β cell-like functionality both in vitro and in vivo.CrossRefPubMed
49.••
go back to reference Pagliuca FW et al. Generation of functional human pancreatic beta cells in vitro. Cell. 2014;159(2):428–39. This team reported near-to-normal β-cell features of human pluripotent stem cells (ESCs and iPSCs) after cocktail-induced reprogramming.CrossRefPubMedPubMedCentral Pagliuca FW et al. Generation of functional human pancreatic beta cells in vitro. Cell. 2014;159(2):428–39. This team reported near-to-normal β-cell features of human pluripotent stem cells (ESCs and iPSCs) after cocktail-induced reprogramming.CrossRefPubMedPubMedCentral
50.
go back to reference Nostro MC et al. Stage-specific signaling through TGFbeta family members and WNT regulates patterning and pancreatic specification of human pluripotent stem cells. Development. 2011;138(5):861–71.CrossRefPubMedPubMedCentral Nostro MC et al. Stage-specific signaling through TGFbeta family members and WNT regulates patterning and pancreatic specification of human pluripotent stem cells. Development. 2011;138(5):861–71.CrossRefPubMedPubMedCentral
51.
go back to reference Osafune K et al. Marked differences in differentiation propensity among human embryonic stem cell lines. Nat Biotechnol. 2008;26(3):313–5.CrossRefPubMed Osafune K et al. Marked differences in differentiation propensity among human embryonic stem cell lines. Nat Biotechnol. 2008;26(3):313–5.CrossRefPubMed
52.
go back to reference Mfopou JK et al. Noggin, retinoids, and fibroblast growth factor regulate hepatic or pancreatic fate of human embryonic stem cells. Gastroenterology. 2010;138(7):2233–45. 2245 e1-14.CrossRefPubMed Mfopou JK et al. Noggin, retinoids, and fibroblast growth factor regulate hepatic or pancreatic fate of human embryonic stem cells. Gastroenterology. 2010;138(7):2233–45. 2245 e1-14.CrossRefPubMed
54.
go back to reference Matveyenko AV et al. Inconsistent formation and nonfunction of insulin-positive cells from pancreatic endoderm derived from human embryonic stem cells in athymic nude rats. Am J Physiol Endocrinol Metab. 2010;299(5):E713–20.CrossRefPubMedPubMedCentral Matveyenko AV et al. Inconsistent formation and nonfunction of insulin-positive cells from pancreatic endoderm derived from human embryonic stem cells in athymic nude rats. Am J Physiol Endocrinol Metab. 2010;299(5):E713–20.CrossRefPubMedPubMedCentral
55.
go back to reference Agulnick AD et al. Insulin-producing endocrine cells differentiated in vitro from human embryonic stem cells function in macroencapsulation devices in vivo. Stem Cells Transl Med. 2015;4(10):1214–22.CrossRefPubMedPubMedCentral Agulnick AD et al. Insulin-producing endocrine cells differentiated in vitro from human embryonic stem cells function in macroencapsulation devices in vivo. Stem Cells Transl Med. 2015;4(10):1214–22.CrossRefPubMedPubMedCentral
56.
go back to reference Laurent LC et al. Dynamic changes in the copy number of pluripotency and cell proliferation genes in human ESCs and iPSCs during reprogramming and time in culture. Cell Stem Cell. 2011;8(1):106–18.CrossRefPubMedPubMedCentral Laurent LC et al. Dynamic changes in the copy number of pluripotency and cell proliferation genes in human ESCs and iPSCs during reprogramming and time in culture. Cell Stem Cell. 2011;8(1):106–18.CrossRefPubMedPubMedCentral
61.
go back to reference Gershengorn MC et al. Epithelial-to-mesenchymal transition generates proliferative human islet precursor cells. Science. 2004;306(5705):2261–4.CrossRefPubMed Gershengorn MC et al. Epithelial-to-mesenchymal transition generates proliferative human islet precursor cells. Science. 2004;306(5705):2261–4.CrossRefPubMed
62.
go back to reference Russ HA et al. In vitro proliferation of cells derived from adult human beta-cells revealed by cell-lineage tracing. Diabetes. 2008;57(6):1575–83.CrossRefPubMed Russ HA et al. In vitro proliferation of cells derived from adult human beta-cells revealed by cell-lineage tracing. Diabetes. 2008;57(6):1575–83.CrossRefPubMed
63.
64.
go back to reference Toren-Haritan G, Efrat S. TGFbeta pathway inhibition redifferentiates human pancreatic islet beta cells expanded in vitro. PLoS One. 2015;10(9):e0139168.CrossRefPubMedPubMedCentral Toren-Haritan G, Efrat S. TGFbeta pathway inhibition redifferentiates human pancreatic islet beta cells expanded in vitro. PLoS One. 2015;10(9):e0139168.CrossRefPubMedPubMedCentral
65.
go back to reference Lenz A, Toren-Haritan G, Efrat S. Redifferentiation of adult human beta cells expanded in vitro by inhibition of the WNT pathway. PLoS One. 2014;9(11):e112914.CrossRefPubMedPubMedCentral Lenz A, Toren-Haritan G, Efrat S. Redifferentiation of adult human beta cells expanded in vitro by inhibition of the WNT pathway. PLoS One. 2014;9(11):e112914.CrossRefPubMedPubMedCentral
66.
go back to reference Bar Y et al. Redifferentiation of expanded human pancreatic beta-cell-derived cells by inhibition of the NOTCH pathway. J Biol Chem. 2012;287(21):17269–80.CrossRefPubMedPubMedCentral Bar Y et al. Redifferentiation of expanded human pancreatic beta-cell-derived cells by inhibition of the NOTCH pathway. J Biol Chem. 2012;287(21):17269–80.CrossRefPubMedPubMedCentral
67.
go back to reference Seaberg RM et al. Clonal identification of multipotent precursors from adult mouse pancreas that generate neural and pancreatic lineages. Nat Biotechnol. 2004;22(9):1115–24.CrossRefPubMed Seaberg RM et al. Clonal identification of multipotent precursors from adult mouse pancreas that generate neural and pancreatic lineages. Nat Biotechnol. 2004;22(9):1115–24.CrossRefPubMed
68.
go back to reference Smukler SR et al. The adult mouse and human pancreas contain rare multipotent stem cells that express insulin. Cell Stem Cell. 2011;8(3):281–93.CrossRefPubMed Smukler SR et al. The adult mouse and human pancreas contain rare multipotent stem cells that express insulin. Cell Stem Cell. 2011;8(3):281–93.CrossRefPubMed
69.
go back to reference Razavi R et al. Diabetes enhances the proliferation of adult pancreatic multipotent progenitor cells and biases their differentiation to more beta-cell production. Diabetes. 2015;64(4):1311–23.CrossRefPubMed Razavi R et al. Diabetes enhances the proliferation of adult pancreatic multipotent progenitor cells and biases their differentiation to more beta-cell production. Diabetes. 2015;64(4):1311–23.CrossRefPubMed
70.
71.
go back to reference Collombat P et al. The ectopic expression of Pax4 in the mouse pancreas converts progenitor cells into alpha and subsequently beta cells. Cell. 2009;138(3):449–62.CrossRefPubMedPubMedCentral Collombat P et al. The ectopic expression of Pax4 in the mouse pancreas converts progenitor cells into alpha and subsequently beta cells. Cell. 2009;138(3):449–62.CrossRefPubMedPubMedCentral
73.
go back to reference Cavelti-Weder C et al. Pancreatic duct ligation after almost complete beta-cell loss: exocrine regeneration but no evidence of beta-cell regeneration. Endocrinology. 2013;154(12):4493–502.CrossRefPubMedPubMedCentral Cavelti-Weder C et al. Pancreatic duct ligation after almost complete beta-cell loss: exocrine regeneration but no evidence of beta-cell regeneration. Endocrinology. 2013;154(12):4493–502.CrossRefPubMedPubMedCentral
74.
75.••
go back to reference Courtney M et al. The inactivation of Arx in pancreatic alpha-cells triggers their neogenesis and conversion into functional beta-like cells. PLoS Genet. 2013;9(10):e1003934. By showing the possibility to drive α-to-β reprogramming through inhibitory signals, this team confirmed the opportunity for pharmacological induction of β-cell replenishment.CrossRefPubMedPubMedCentral Courtney M et al. The inactivation of Arx in pancreatic alpha-cells triggers their neogenesis and conversion into functional beta-like cells. PLoS Genet. 2013;9(10):e1003934. By showing the possibility to drive α-to-β reprogramming through inhibitory signals, this team confirmed the opportunity for pharmacological induction of β-cell replenishment.CrossRefPubMedPubMedCentral
76.
77.
go back to reference Fomina-Yadlin D et al. GW8510 increases insulin expression in pancreatic alpha cells through activation of p53 transcriptional activity. PLoS One. 2012;7(1):e28808.CrossRefPubMedPubMedCentral Fomina-Yadlin D et al. GW8510 increases insulin expression in pancreatic alpha cells through activation of p53 transcriptional activity. PLoS One. 2012;7(1):e28808.CrossRefPubMedPubMedCentral
78.
go back to reference Zhang, Y., et al., PAX4 Gene transfer induces alpha-to-beta cell phenotypic conversion and confers therapeutic benefits for diabetes treatment. Mol Ther, 2015. Zhang, Y., et al., PAX4 Gene transfer induces alpha-to-beta cell phenotypic conversion and confers therapeutic benefits for diabetes treatment. Mol Ther, 2015.
79.
go back to reference Sangan CB et al. In vitro reprogramming of pancreatic alpha cells towards a beta cell phenotype following ectopic HNF4alpha expression. Mol Cell Endocrinol. 2015;399:50–9.CrossRefPubMed Sangan CB et al. In vitro reprogramming of pancreatic alpha cells towards a beta cell phenotype following ectopic HNF4alpha expression. Mol Cell Endocrinol. 2015;399:50–9.CrossRefPubMed
80.
go back to reference Zhou Q et al. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature. 2008;455(7213):627–32.CrossRefPubMed Zhou Q et al. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature. 2008;455(7213):627–32.CrossRefPubMed
81.••
go back to reference Li W et al. Long-term persistence and development of induced pancreatic beta cells generated by lineage conversion of acinar cells. Nat Biotechnol. 2014;32(12):1223–30. This group confirmed its own data about acinar-to-β reprogramming and obtained functional β cells after extended follow-up periods.CrossRefPubMed Li W et al. Long-term persistence and development of induced pancreatic beta cells generated by lineage conversion of acinar cells. Nat Biotechnol. 2014;32(12):1223–30. This group confirmed its own data about acinar-to-β reprogramming and obtained functional β cells after extended follow-up periods.CrossRefPubMed
82.
go back to reference Akinci E et al. Reprogramming of pancreatic exocrine cells towards a beta (beta) cell character using Pdx1, Ngn3 and MafA. Biochem J. 2012;442(3):539–50.CrossRefPubMedPubMedCentral Akinci E et al. Reprogramming of pancreatic exocrine cells towards a beta (beta) cell character using Pdx1, Ngn3 and MafA. Biochem J. 2012;442(3):539–50.CrossRefPubMedPubMedCentral
83.
go back to reference Yamada T et al. Reprogramming mouse cells with a pancreatic duct phenotype to insulin-producing beta-like cells. Endocrinology. 2015;156(6):2029–38.CrossRefPubMed Yamada T et al. Reprogramming mouse cells with a pancreatic duct phenotype to insulin-producing beta-like cells. Endocrinology. 2015;156(6):2029–38.CrossRefPubMed
84.••
go back to reference Baeyens L et al. Transient cytokine treatment induces acinar cell reprogramming and regenerates functional beta cell mass in diabetic mice. Nat Biotechnol. 2014;32(1):76–83. A groundbreaking work showing the possibility for pharmacological induction of acinar-to-β differentiation.CrossRefPubMedPubMedCentral Baeyens L et al. Transient cytokine treatment induces acinar cell reprogramming and regenerates functional beta cell mass in diabetic mice. Nat Biotechnol. 2014;32(1):76–83. A groundbreaking work showing the possibility for pharmacological induction of acinar-to-β differentiation.CrossRefPubMedPubMedCentral
85.
86.
go back to reference Hurtado M et al. Activation of the epidermal growth factor signalling pathway by tissue plasminogen activator in pancreas cancer cells. Gut. 2007;56(9):1266–74.CrossRefPubMedPubMedCentral Hurtado M et al. Activation of the epidermal growth factor signalling pathway by tissue plasminogen activator in pancreas cancer cells. Gut. 2007;56(9):1266–74.CrossRefPubMedPubMedCentral
87.
go back to reference Ray KC et al. Heparin-binding epidermal growth factor-like growth factor eliminates constraints on activated Kras to promote rapid onset of pancreatic neoplasia. Oncogene. 2014;33(7):823–31.CrossRefPubMedPubMedCentral Ray KC et al. Heparin-binding epidermal growth factor-like growth factor eliminates constraints on activated Kras to promote rapid onset of pancreatic neoplasia. Oncogene. 2014;33(7):823–31.CrossRefPubMedPubMedCentral
88.
go back to reference Voudouri K et al. Insulin-like growth factor and epidermal growth factor signaling in breast cancer cell growth: focus on endocrine resistant disease. Anal Cell Pathol (Amst). 2015;2015:975495. Voudouri K et al. Insulin-like growth factor and epidermal growth factor signaling in breast cancer cell growth: focus on endocrine resistant disease. Anal Cell Pathol (Amst). 2015;2015:975495.
89.
go back to reference Sasaki S et al. Activation of GLP-1 and gastrin signalling induces in vivo reprogramming of pancreatic exocrine cells into beta cells in mice. Diabetologia. 2015;58(11):2582–91.CrossRefPubMed Sasaki S et al. Activation of GLP-1 and gastrin signalling induces in vivo reprogramming of pancreatic exocrine cells into beta cells in mice. Diabetologia. 2015;58(11):2582–91.CrossRefPubMed
90.
go back to reference Houbracken I et al. Lineage tracing evidence for transdifferentiation of acinar to duct cells and plasticity of human pancreas. Gastroenterology. 2011;141(2):731–41. 741 e1-4.CrossRefPubMed Houbracken I et al. Lineage tracing evidence for transdifferentiation of acinar to duct cells and plasticity of human pancreas. Gastroenterology. 2011;141(2):731–41. 741 e1-4.CrossRefPubMed
91.
go back to reference Pan FC, Wright C. Pancreas organogenesis: from bud to plexus to gland. Dev Dyn. 2011;240(3):530–65.CrossRefPubMed Pan FC, Wright C. Pancreas organogenesis: from bud to plexus to gland. Dev Dyn. 2011;240(3):530–65.CrossRefPubMed
93.
go back to reference Kopp JL et al. Sox9+ ductal cells are multipotent progenitors throughout development but do not produce new endocrine cells in the normal or injured adult pancreas. Development. 2011;138(4):653–65.CrossRefPubMedPubMedCentral Kopp JL et al. Sox9+ ductal cells are multipotent progenitors throughout development but do not produce new endocrine cells in the normal or injured adult pancreas. Development. 2011;138(4):653–65.CrossRefPubMedPubMedCentral
94.
go back to reference Furuyama K et al. Continuous cell supply from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nat Genet. 2011;43(1):34–41.CrossRefPubMed Furuyama K et al. Continuous cell supply from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nat Genet. 2011;43(1):34–41.CrossRefPubMed
95.
go back to reference Solar M et al. Pancreatic exocrine duct cells give rise to insulin-producing beta cells during embryogenesis but not after birth. Dev Cell. 2009;17(6):849–60.CrossRefPubMed Solar M et al. Pancreatic exocrine duct cells give rise to insulin-producing beta cells during embryogenesis but not after birth. Dev Cell. 2009;17(6):849–60.CrossRefPubMed
96.
go back to reference Al-Hasani K et al. Adult duct-lining cells can reprogram into beta-like cells able to counter repeated cycles of toxin-induced diabetes. Dev Cell. 2013;26(1):86–100.CrossRefPubMed Al-Hasani K et al. Adult duct-lining cells can reprogram into beta-like cells able to counter repeated cycles of toxin-induced diabetes. Dev Cell. 2013;26(1):86–100.CrossRefPubMed
97.
go back to reference Yatoh S et al. Differentiation of affinity-purified human pancreatic duct cells to beta-cells. Diabetes. 2007;56(7):1802–9.CrossRefPubMed Yatoh S et al. Differentiation of affinity-purified human pancreatic duct cells to beta-cells. Diabetes. 2007;56(7):1802–9.CrossRefPubMed
98.
go back to reference Lee J et al. Expansion and conversion of human pancreatic ductal cells into insulin-secreting endocrine cells. Elife. 2013;2:e00940.PubMedPubMedCentral Lee J et al. Expansion and conversion of human pancreatic ductal cells into insulin-secreting endocrine cells. Elife. 2013;2:e00940.PubMedPubMedCentral
99.
go back to reference Baertschiger RM et al. Mesenchymal stem cells derived from human exocrine pancreas express transcription factors implicated in beta-cell development. Pancreas. 2008;37(1):75–84.CrossRefPubMed Baertschiger RM et al. Mesenchymal stem cells derived from human exocrine pancreas express transcription factors implicated in beta-cell development. Pancreas. 2008;37(1):75–84.CrossRefPubMed
100.
go back to reference Seeberger KL et al. Expansion of mesenchymal stem cells from human pancreatic ductal epithelium. Laboratory investigation; a journal of technical methods and pathology. 2006;86(2):141–53.CrossRefPubMed Seeberger KL et al. Expansion of mesenchymal stem cells from human pancreatic ductal epithelium. Laboratory investigation; a journal of technical methods and pathology. 2006;86(2):141–53.CrossRefPubMed
101.
Metadata
Title
New Insights into Diabetes Cell Therapy
Authors
Philippe A. Lysy
Elisa Corritore
Etienne M. Sokal
Publication date
01-05-2016
Publisher
Springer US
Published in
Current Diabetes Reports / Issue 5/2016
Print ISSN: 1534-4827
Electronic ISSN: 1539-0829
DOI
https://doi.org/10.1007/s11892-016-0729-3

Other articles of this Issue 5/2016

Current Diabetes Reports 5/2016 Go to the issue

Microvascular Complications—Nephropathy (AP Maxwell, Section Editor)

The Promise of Mesenchymal Stem Cell Therapy for Diabetic Kidney Disease

Immunology and Transplantation (L Piemonti and V Sordi, Section Editors)

Targeting Homeostatic T Cell Proliferation to Control Beta-Cell Autoimmunity

Immunology and Transplantation (L Piemonti and V Sordi, Section Editors)

Bone Marrow-Derived Stem Cells: a Mixed Blessing in the Multifaceted World of Diabetic Complications

Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.