Skip to main content
Therapeutic Advances in Endocrinology and Metabolism logoLink to Therapeutic Advances in Endocrinology and Metabolism
. 2016 Jun 6;7(4):182–199. doi: 10.1177/2042018816652059

β-cell replacement sources for type 1 diabetes: a focus on pancreatic ductal cells

Elisa Corritore 1, Yong-Syu Lee 2, Etienne M Sokal 3, Philippe A Lysy 4,
PMCID: PMC4973405  PMID: 27540464

Abstract

Thorough research on the capacity of human islet transplantation to cure type 1 diabetes led to the achievement of 3- to 5-year-long insulin independence in nearly half of transplanted patients. Yet, translation of this technique to clinical routine is limited by organ shortage and the need for long-term immunosuppression, restricting its use to adults with unstable disease. The production of new bona fide β cells in vitro was thus investigated and finally achieved with human pluripotent stem cells (PSCs). Besides ethical concerns about the use of human embryos, studies are now evaluating the possibility of circumventing the spontaneous tumor formation associated with transplantation of PSCs. These issues fueled the search for cell candidates for β-cell engineering with safe profiles for clinical translation. In vivo studies revealed the regeneration capacity of the exocrine pancreas after injury that depends at least partially on facultative progenitors in the ductal compartment. These stimulated subpopulations of pancreatic ductal cells (PDCs) underwent β-cell transdifferentiation through reactivation of embryonic signaling pathways. In vitro models for expansion and differentiation of purified PDCs toward insulin-producing cells were described using cocktails of growth factors, extracellular-matrix proteins and transcription factor overexpression. In this review, we will describe the latest findings in pancreatic β-cell mass regeneration due to adult ductal progenitor cells. We will further describe recent advances in human PDC transdifferentiation to insulin-producing cells with potential for clinical translational studies.

Keywords: β cells, diabetes, differentiation, duct cells, insulin, progenitors

Introduction

Diabetes is a metabolic disorder occurring when the pancreas does not produce enough insulin or when the body does not properly use the released insulin. The common effect is a chronic hyperglycemia that leads to a long-term risk of micro- and macroangiopathy. The worldwide incidence of diabetes is steadily increasing and the World Health Organization estimated that about 552 million people will be affected by the disease by 2030 [Ding et al. 2013].

Type 1 diabetes (T1D) affects 5–10% of all patients with diabetes and usually develops in children and young adults. This disease is characterized by progressive destruction of pancreatic insulin-producing β cells provoked by a B- and T-lymphocyte-dependent autoimmune assault [Atkinson et al. 2011]. Although the origins of the causative autoimmune reactions are still uncertain [Ludvigsson, 2013], several studies have shown correlations between T1D onset and environmental factors such as enteroviral infections [Krogvold et al. 2015], early infant nutrition [Mayer-Davis et al. 2013], or vitamin D deficiency [Mathieu, 2015]. The genetic association between human leucocyte antigen (HLA) and the onset of T1D has also been confirmed by many studies since the 1970s. About 50% of the risk for developing T1D is explained by the polymorphism of more than 50 different HLA loci [Lysy, 2014]. Most of these genes are involved in immune regulation and were associated with the risk of developing other autoimmune disorders such as celiac disease, systemic lupus erythematosus and multiple sclerosis [Noble, 2015]. Current treatments for T1D are primarily focused on insulin supplementation that improves glucose homeostasis but fails to achieve treatment targets for many patients [Lind et al. 2014].

Pancreas and islet transplantation

In this context, the replacement of functional β cells would be the only cure for patients with T1D, as demonstrated by the accumulated experience in whole pancreas [Niederhaus, 2015] and human islet transplantation [Shapiro and Lakey, 2000; Bellin et al. 2012; Barton et al. 2012]. Currently, more than 13,000 patients with diabetes mellitus have benefited from a successful pancreas (i.e. pancreas alone, or pancreas-after-kidney) transplantation [Kerr et al. 2015] and graft survival improved by up to 81.5% 1 year after organ transplant [Kaufman, 2015]. This procedure is often proposed to patients with severe renal failure requiring concomitant kidney transplant [Johannesson et al. 2015]. Despite its curative potential, pancreas transplantation remains a difficult procedure with significant morbidity and mortality (22% mortality rate 10 years after transplant), and with limitations associated to organ shortage [Kandaswamy et al. 2016].

Human islet isolation was developed to provide patients with a minimally invasive cell-replacement protocol, and functionality of transplanted islets was greatly improved in the last decade [Bruni et al. 2014]. A review by Barton and colleagues from the Collaborative Islet Transplant (CIT) Registry showed insulin independence during 3 years after human-islet transplantation in about 44% of patients [Barton et al. 2012]. Recently, Brennan and colleagues showed functional islet engraftment and glucose tolerance 54 months after transplantation under the Edmonton protocol, in patients followed up for 12 years and treated with tacrolimus and sirolimus or mycophenolate mofetil [Brennan et al. 2016]. The development of a new immunosuppressive regimen that combined sirolimus and tacrolimus with classical drugs such as daclizumab and etanercept in addition to granulocyte-colony stimulating (G-CSF) and exenatide showed prolonged graft function in 70% of the patients for about 12 years [Inverardi, 2015]. Previously, Long and colleagues showed the efficiency of rapamycin combined with interleukin-2 (IL-2) as treatment for autoimmune diabetes [Long et al. 2012]. Indeed, nine diabetic patients in a phase I clinical trial were treated with this cocktail (administration of rapamycin for 3 months and IL-2 for 1 month) and showed effective augmentation of Treg cells. However, transient β-cell dysfunction, decreased levels of C-peptide and negative effects on β-cell regeneration represented the greatest limitations of this combinatorial therapy.

Despite significant improvements, islet transplantation still remains limited by the shortage of cadaveric donors, the necessity for long-term immunosuppression, restricting its indication for adults with unstable diabetes (e.g. recurrence of severe hypoglycemias, intractable glucose variability, diabetes-related complications), due to the complexity of cell isolation protocols and islet viability after cryopreservation. Although Manning Fox and colleagues recently showed that human islets cryopreserved for 20 years maintained correct purity, functional intracellular Ca2+ influx and glucose-stimulated insulin secretion (GSIS), these stored cells had decreased levels of insulin content and lower abilities to reverse diabetes in a mouse model as compared with fresh islets [Manning Fox et al. 2015].

Pluripotent stem cells

These difficulties fostered the finding of new cell sources with potential to provide large supplies of insulin-producing cells for clinical settings. Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) represent the most widely investigated candidates for in vitro reprogramming purposes because of their theoretically unlimited proliferation and high differentiation potential [Lysy et al. 2012]. Over the last 10 years, researchers designed protocols for differentiation of pluripotent stem cells (PSCs) that mimicked pancreas embryogenesis by using cocktails of molecules with specific influences on key intracellular pathways [Blyszczuk et al. 2004; Ku et al. 2004; Schroeder et al. 2006; Schiesser and Wells, 2014]. Combinations of activin A, retinoic acid, transforming growth factor (TGF) β, nicotinamide, bone morphogenetic protein (BMP), keratinocyte growth factor (KGF), cyclopamine, basic fibroblastic growth factor (bFGF), Noggin, wingless-type mouse mammary tumor virus (MMTV) integration-site family-member 3a (Wnt3a), sonic hedgehog pathway antagonist (SANT1) and insulin were used to induce the differentiation of ESCs toward pancreatic endocrine precursors through incubations of 3–4 weeks [Ku et al. 2004; Yasunaga et al. 2005; D’Amour et al. 2006; Kroon et al. 2008; Rezania et al. 2012]. During differentiation, ESCs showed acquisition of typical gene profiles of pancreatic stepwise development: expression of OCT4, SOX2, and Brachyury followed by activation of PDX1, NGN3, NKX6.1 until final expression of key β-cell markers such as MAFA, PAX4 and insulin [D’Amour et al. 2006; Jackson et al. 2010; Hrvatin et al. 2014]. In 2006, D’Amour and colleagues made great advances by obtaining ESC-derived β-cell populations containing up to 12% of insulin+ cells after in vitro stepwise protocols [D’Amour et al. 2005, 2006]. These cells completed their reprogramming into functional insulin-producing derivatives 3–4 months after transplantation into rodents [D’Amour et al. 2006; Kroon et al. 2008; Rezania et al. 2012]. Yet PSCs lacked essential properties of bona fide β cells, including GSIS [Nostro et al. 2011; Kelly et al. 2011; Bruin et al. 2014; Hrvatin et al. 2014]. Recently, two groups [Pagliuca et al. 2014; Bruin et al. 2014] demonstrated the possibility of obtaining functional differentiation of human ESCs in vitro with insulin secretion patterns close to human islet cells; yet only the Rezania and Pagliuca teams described the attainment of monohormonal ESC-derived β cells after implantation under the kidney capsule of diabetic mice [Pagliuca et al. 2014; Rezania et al. 2014]. In these studies, human insulin was detected in the mouse bloodstream with subsequent glycemic normalization within 40 days post transplantation. These data are putting PSCs closer to potential exploitation in the clinical setting.

Tumor formation remains a major concern in the use of ESCs, fetal stem cells and iPSCs for transplantation studies. Unwanted ‘off-target’ mesodermal formation was reported with human ESCs in transplantation sites [Kroon et al. 2008; Rezania et al. 2012]. Many groups have tried selection of ESC-derived endocrine cells [Kelly et al. 2015; Jiang and Morahan, 2015] or the use of micro- and macroencapsulation devices [Motte et al. 2014; Agulnick et al. 2015; Song and Roy, 2015] such as the TheracyteTM (Inc. manufactures, Laguna Hills, California) system to circumvent the undesired cell growth, but also to promote angiogenesis of the cells and to protect those from the host immune system [Pepper et al. 2015]. PSC-derived functional insulin-producing cells were recently injected into a Theracyte-based device (called EncaptraTM, San Diego, California) and subsequently transplanted subcutaneously [Agulnick et al. 2015]. Furthermore, this system permitted the survival of mouse neonatal pancreatic tissue maintaining euglycemia up to 95 days post-transplant in diabetic RIP-LCMV.GP mice [Boettler et al. 2015]. Clinical trials in Phase I/II [ClinicalTrials.gov identifiers: NCT02239354 and NCT01996228] are currently assessing long-term tolerability and efficacy of this system, with results eagerly awaited for. Very recently, alginate-based encapsulation was used to intraperitoneally transplant human ESC-derived β-like cells into immunocompetent mice [Vegas et al. 2016]. Grafts retained functionality at 174 days of follow up with significant effects on glucose homeostasis that mirrored C-peptide secretion levels. These important results bring additional evidence of the translational capacity of human ESCs.

Principal modes of β-cell regeneration

Whether progenitors or stem cells reside in the adult human pancreas or not is still unknown, but regeneration of pancreatic compartments were described after birth and after injury [Chintinne et al. 2010; Li et al. 2010; Nakamura et al. 2011]. The principal mechanisms investigated to explain β-cell regeneration within the pancreas include: neogenesis (differentiation of islet cells from facultative pancreatic progenitors), replication of pre-existent β cells and transdifferentiation (being the conversion from one cell type to another) [Bonner-Weir et al. 2010, 2012].

Neogenesis

Neogenesis was largely considered the main process responsible for embryonic development, however, its involvement in the adult pancreas is still under investigation [Bonner-Weir et al. 2012]. Numerous studies describe the existence of a reservoir of progenitor cells within the human exocrine pancreas. The involvement of PDCs as a niche of committed progenitors in the adult pancreas will be discussed below.

Acinar cells represent the major population in the human pancreas; therefore particular attention was paid to their role as progenitors [Houbracken et al. 2010; Mfopou and Bouwens, 2013; Baeyens et al. 2014; Pin et al. 2015]. The conversion of acinar into endocrine fates was previously reported by lineage-tracing studies, showing formation of ectopic insulin+ cells derived from acinar cells after suppression of Ptf1a in zebra fish [Hesselson et al. 2011]. Controversy regarding the in vivo plasticity of adult acinar was generated by a study showing the lack of acinar-to-β transdifferentiation following 70% pancreatectomy (Px), pancreatic duct (PDL), and cerulein-induced pancreatitis in mice [Desai et al. 2007]. In 2008, Zhou and colleagues showed the direct reprogramming of exocrine cells into β cells after in-vivo overexpression of PDX1, NGN3, and MAFA [Zhou et al. 2008]. Adenoviral vectors containing the sequences of the three transcription factors (TFs) (and green fluorescent (GFP) as reporter) were transduced in exocrine cells of Rag1-/- diabetic mice. Only 10 days after infection, new insulin+/GFP+ cells expressing the hallmarks of bona fide β cells were observed. Streptozotocin (STZ)-treated animals showed significant decrease of hyperglycemia without complete disease reversal. These results were confirmed by Cavelti-Weder and colleagues, who showed extensive periods (up to 13 months) of euglycemia in two of the TF-transduced diabetic animals [Cavelti-Weder et al. 2015]. Similarly, a cytokine-based protocol [containing epidermal growth factor (EGF) and ciliary neurotrophic factor (CNTF)] was developed to circumvent the use of adenoviruses for acinar-to-β transdifferentiation [Bonfanti et al. 2015]. In this study, alloxan-induced diabetic mice received EGF and CNTF via intraperitoneal osmotic pumps that allowed 65% of animals to increase their levels of serum insulin and normalize their glycemia within 5 days. Acinar cells were detected to be at the origin of the new β cells via lineage-tracing experiments. Together, these works provide evidence for a major potential of acinar cells for designing in situ transdifferentiation protocols.

Replication

The other mechanism thought to be at play in endogenous replenishment of pancreatic tissues is replication of pre-existing β cells. β-cell proliferation in both human and murine models is normally controlled by activation of proteins that regulate the cell cycle [Vetere et al. 2014]. For example, it has been shown that p16, p27, cyclins D1, D2, and D3 have a central role in pancreas regeneration controlling the replicative potential of the β-cell mass. Many efforts were made to identify compounds that could stimulate β-cell proliferation. An increase of about 50% of the β-cell mass and parallel decrease of hyperglycemia in mice was observed after 6 weeks of treatment with WS6 molecule without affecting their differentiation or viability [Shen et al. 2013; Boerner et al. 2015]. High-throughput screening of more than 850 compounds led to the identification of other molecules such as 5-iodotubercidin and ABT-702, capable of stimulating β-cell expansion in rodent and porcine islets [Annes et al. 2012]. Intraperitoneal injections of these adenosine kinase inhibitors in wild-type mice promoted β-cell turnover within 24 hours, as demonstrated by the increase of PDX1+/insulin+ cells coexpressing Ki-67 while no replication was found in the exocrine compartment.

Although the replication potential of rodent β cells was observed, very little evidence of β-cell proliferation was provided in the human pancreas [Meier et al. 2008]. In this context, Russ and colleagues, in 2011, described an in-vitro system to dedifferentiate, expand and redifferentiate human β cells with the aim of obtaining a large supply of functional insulin-producing cells [Russ et al. 2011]. The dedifferentiating β cells acquired mesenchymal markers, suggesting epithelial-mesenchymal transition (EMT). Furthermore, the maintenance of epigenetic memories allowed fast redifferentiation of β-cell-derived cells into new insulin+ cells, using a combination of soluble factors that included glucose, nicotinamide, exendin-4, and activin A. About 40% of C-peptide+ cells were observed in these EMT populations after 8 days of incubation with these molecules. When EMT-derived β-like cells generated from Notch inhibition protocols were transplanted under the kidney capsule of STZ-treated NOD-SCID mice, Efrat’s group observed a decrease of blood glucose levels and the detection of human C-peptide [Bar et al. 2012]. Collectively, these findings proposed a new system for expansion and generation of a source of functional β cells starting from primary cultures of adult human islets.

Transdifferentiation

The third mechanism evaluated to explain β-cell mass replenishment is the transdifferentiation of pancreatic cells into β cells. Using a transgenic mouse model, Collombat and colleagues, in 2009, described the conversion of glucagon+ cells into functional β cells after overexpression of PAX4 [Collombat et al. 2009] or selective inhibition of aristaless related homeobox (ARX) in α cells [Courtney et al. 2013]. In both studies, in vivo overexpression of PAX4 or misexpression of ARX was sufficient to generate new functional β cells capable to revert STZ-induced diabetes. In parallel, it was observed that the lack of glucagon+ cells triggered an endocrine-specification program occurring after re-expression of NGN3+ in ‘progenitors’ residing in the duct epithelium. This shift of endocrine phenotypes was also observed after diphtheria toxin-induced α-cell death [Thorel et al. 2010]. Other groups reported the in vitro conversion from α to β cell under specific conditions [Fomina-Yadlin et al. 2010; Yang et al. 2011; Zhang et al. 2015], but at rather low yields. Also, the question remains as to whether these phenomena could take place in humans and how this potential could be translated into pharmacological protocols [Courtney et al. 2011; Napolitano et al. 2015; Vieira et al. 2013; Collombat et al. 2010].

Progenitor cells

Theoretically, pancreatic epithelial cells present valuable properties for β-cell reprogramming since they all arise from the same embryonic precursor; the branching morphogenesis of the pancreas, depending on the ductal system, gives rise to all exocrine and endocrine lineages [Pan and Wright, 2011; Lysy et al. 2013] under specific signaling instructions. Pancreas organogenesis is controlled by the embryonic mesoderm that releases molecules such as fibroblastic growth factors (FGFs), retinoic acid, BMPs, Wnt, Sonic Hedgehog, and Notch proteins [Wandzioch and Zaret, 2009; Mfopou et al. 2010; Nostro et al. 2011; Marquez-Aguirre et al. 2015]. Moreover, both exocrine and endocrine compartments are characterized by local expression of specific markers and TFs. For example, Lemaire and colleagues showed that bicaudal C1 (BICC1), a protein normally in duct lining during embryogenesis, is essential for ductal morphogenesis and subsequent progenitor differentiation [Lemaire et al. 2015]. Pancreatic sections of BICC1-/- mice showed a 34% decrease of NGN3-expressing endocrine progenitors within the ductal compartment. BICC1 shutdown affected normal tissue formation: the endocrine mass was decreased by 50% and duct cell overproliferation led to cystic dysplasia of the main and interlobular ducts. These results illustrate the possibility to fate-map molecular signals of pancreas morphogenesis.

In vivo studies on pancreatic progenitors

Postnatal in vivo experiments performed using several murine pancreatic injury models (e.g. partial Px, cellophane wrapping, duct ligation or intraperitoneal cerulein injection [Rosenberg, 1995; De Breuck et al. 2006; Sakaguchi et al. 2006; Bonner-Weir et al. 2008]) revealed partial tissue regeneration from PDCs (Table 1) expressing PDX1, NGN3, HNF1β, with or without SOX9. The Inada group observed β-cell mass replenishment after PDL in carbonic anhydrase II (CAII)-Cre transgenic mice, which were subsequently mated with the strain ROSA26 loxP-Stop-loxP LacZ (R26R) to trace the progeny of ductal cells [Inada et al. 2008]. Two weeks after PDL, mice had 12.1 ± 1.9% of β-galactosidase+ β cells in nonligated portions and 23.6 ± 2.2% in the ligated ones, compared with 5.5 ± 2.0% in control mice. These results suggested, at least partially, the ductal origin of the new β cells. To investigate the role of putative progenitors in pancreas regeneration, Criscimanna and colleagues generated a mouse model of restricted diphtheria-toxin (DT)-receptor expression for induction of global pancreatic damage after toxin injection [Criscimanna et al. 2011]. Extensive and selective ablation of elastase+ acinar and PDX1+ endocrine cells were induced in R6DTR/CRE backcrossed animals. Within 3 to 4 weeks after DT injection, highly proliferative ducts contained cells coexpressing amylase, NGN3, insulin, and glucagon. These findings suggested that the global pancreatic damage provoked by DT injection stimulated the differentiation of cells located in the ductal compartment toward acinar and endocrine cells. Recently, lineage-tracing experiments showed duct-to-β-cell conversion after partial Px in normal young developing mice and in adult mice with TGF-β receptor overexpression [El-Gohary et al. 2016]. Interestingly, this study showed prolongations of the ductal tree inside surrounding islets, at least in young mice and humans. This feature was not observed in adult mice after injury, but well in TGF-β-receptor-mutant animals, showing the implication of this pathway (implicated in EMT) in endocrine reprogramming of the ducts in adult tissues. These results corroborate previous reports about the requirement for duct-lining cells to enter a state of EMT before endocrine reprogramming can occur (see below) [Pfeifer et al. 2013].

Table 1.

Evidence of pancreatic ducts as a source of progenitor cells.

Reference Research model Evidence
Suarez-Pinzon et al. [2005] In vivo Treatment with EGF promoted β-cell neogenesis
Xu et al. [2006] In vivo Treatment with exendin-4 facilitated β-cell neogenesis in pancreatic ducts
Tokui et al. [2006] In vivo Betacellulin promoted ductal differentiation towards insulin-expressing cells
Inada et al. [2008] In vivo β-cell mass replenishment occurred from ductal cells 2 weeks post PDL
Bonner-Weir et al. [2008] In vivo Partial pancreatic tissue regeneration occurred from ductal cells
Xu et al. [2008] In vivo Re-expression of NGN3 in ductal lining cells occurred 1 week after PDL
Criscimanna et al. [2011] In vivo Proliferative ducts expressed acinar and endocrine markers 3/4 weeks post-DT injection
Wang et al. [2012] In vivo Administration of gastrin and EGF increased β-cell neogenesis
Pfeifer et al. [2013] In vivo Requirement of EMT for duct-lining cells differentiation into endocrine cells
Courtney et al. [2013] In vivo Continuous activation of NGN3+ cells along the lining duct occurred after ARX inhibition
Téllez et al. [2014] In vivo Appearance of β-cell clusters from ductal epithelium occurred 3 days post 90% Px
Lemaire et al. [2015] In vivo Bicaudal C1 expressed in duct lining cells was essential for progenitor differentiation
Yamaguchi et al. [2015] In vivo Pancreatic ductal glands contained niches of progenitor cells
Carpino et al. [2015] In vivo Pancreatic ductal glands as container of niches for pancreatic precursors
Napolitano et al. [2015] In vivo Continuous activation of NGN3+ cells along the lining duct occurred after PAX4 overexpression
El-Gohary et al. [2016] In vivo Duct-to-β-cell conversion occurred after partial Px in mice overexpressing TGF-β receptor
Zhang et al. [2016] In vivo Conversion of SOX9+ cells into insulin-producing cells occurred after administration of gastrin and EGF
Seaberg et al. [2004] In vitro Progenitor colonies arose from islet and ducts cultured with EGF and FGF2
Rovira et al. [2010] In vitro Centroacinar/terminal ductal cells with progenitor-like phenotype

ARX, aristaless related homeobox; DT, diphtheria toxin; EGF, epidermal growth factor; EMT, epithelial-mesenchymal transition; PDL, pancreatic duct ligation; Px, pancreatectomy; TGF, transforming growth factor.

The identification of factors with potential to activate β-cell regeneration from progenitors would be a compelling approach to cure diabetes [Piran et al. 2014; Riley et al. 2015; Dominguez-Bendala et al. 2016]. Induction of pancreatic neogenesis in the postnatal pancreas after administration of growth factors in vivo was evaluated in murine models using glucagon-like peptide 1 (GLP-1)/exendin-4, betacellulin, gastrin, INGAP, interferon-γ, soybean trypsin inhibitor and TGF-β. These studies are summarized elsewhere [Bonner-Weir et al. 2012]. Intraperitoneal injection of exendin-4 in STZ-treated wild-type rats, repeated for 10 consecutive days, induced a significant reduction of blood glucose for more than 2 weeks [Xu et al. 2006]. In another study, adenoviral vectors carrying human betacellulin sequences were injected within ducts in imprinting control region (ICR) mice [Tokui et al. 2006]. Only 1 week later, betacellulin and insulin-protein expression were observed in ductal cells, and at 2 and 8 weeks postinfection, numbers of duct-lining cells coexpressing insulin increased [Tokui et al. 2006]. Because of potential implications of gastrin in β-cell mass regeneration in rodents [Suarez-Pinzon et al. 2008; Suissa et al. 2013], a recent study evaluated the role of gastrin in promoting β-cell neogenesis from ductal cells in 90% Px rats [Tellez and Montanya, 2014]. In the gastrin-treated mice, strong proliferation activity was observed in the ductal epithelium 1 day after Px, with cells acquiring expression of β-cell markers such as PDX1, NGN3, NKX6.1, and NeuroD1. Only 3 days after surgery, β-cell clusters and increased β-cell mass appeared next to the ductal epithelium in gastrin-treated animals. However, blood glucose levels were not improved [Tellez and Montanya, 2014].

Since treatment with EGF was reported to induce β-cell neogenesis from adult ductal cells in vitro and in vivo [Suarez-Pinzon et al. 2005; Wang et al. 2012], Zhang and colleagues recently investigated the origin of ductal neogenesis in alloxan-treated diabetic wild-type mice subjected to administration of EGF and gastrin for 56 days [Zhang et al. 2016]. Lineage-tracing experiments showed that conversion of SOX9+ PDCs into insulin-producing cells occurred in hyperglycemic conditions and that differentiation significantly increased after long-term administration of low-dose gastrin and EGF (GE). Interestingly, about 75% of animals reversed medium (300–450 mg/dl) but not high hyperglycemia (>450 mg/dl), suggesting a dependency on glucose levels of duct-to-β reprogramming.

Controversy arose about spontaneous postnatal duct-to-β cell conversion after several studies reported the complete absence of endocrine regeneration from ductal lineages in early postnatal mouse models [Solar et al. 2009; Furuyama et al. 2011; Kopp et al. 2011; Pan et al. 2013; Rankin et al. 2013]. Although β cells appear at day 17.5 in the mouse embryo after activation of NGN3+ cells in the vicinity of the ductal epithelium [Pan and Wright, 2011], Kopinke and Murtaugh could not find evidence of postnatal endocrine specification of PDCs in healthy mice after birth [Kopinke and Murtaugh, 2010]. Using a Cre-lox system within HNF1β locus to track embryonic and adult ductal cells after PDL in R26R reporter mice, Solar and colleagues found that the development of endocrine cells was strictly dependent on HNF1β expression in the embryos but not in the adult tissues [Solar et al. 2009]. Similar findings were obtained in 2011 with Sox9CreERT2 transgenic mouse in which NGN3 expression in PDCs triggered de novo formation of endocrine and acinar cells before birth, but not postnatally [Kopp et al. 2011]. The same year, a study confirmed the absence of endocrine reprogramming of SOX9-expressing cells in adult mice following partial Px, acute pancreatitis, STZ-induced diabetes, or PDL [Furuyama et al. 2011]. In 2013, a study showed the lack of significant changes in β-cell mass and insulin content in adult mice 30 days following PDL [Rankin et al. 2013] despite massive expression of NGN3 in the ligated portion. To investigate β-cell regeneration after extreme β-cell loss, Cavelti-Weder and colleagues performed PDL in STZ-treated adult rats transplanted with syngeneic islets [Cavelti-Weder et al. 2013]. Although acinar cells showed massive regeneration and self-renewal capacities 10 months after surgery, no evidence of significant β-cell replacement was observed in this model. Only few β cells (about 1.5%) could be observed 5 and 10 months after PDL in ligated portions, with low insulin content (0.39 ± 0.1 µg/pancreas).

Existence of stem-cell niches with progenitor potential was recently observed within pancreatic ductal glands (PDGs) [Yamaguchi et al. 2015]. PDGs are a newly identified epithelial compartment residing in the mesenchymal structures surrounding pancreatic ducts. Using a mouse model of pancreatitis and BrdU labeling, Yamaguchi and colleagues observed proliferation in PDCs which migrated from the PDGs to the main pancreatic duct 5 days after acute inflammation. Microarrays showed the enrichment of ESC markers, including SOX2 and NANOG, in the PDGs compared with the ductal epithelium. A more extensive anatomical characterization of PDGs and their role as a niche of pancreatic precursors was performed on healthy human pancreata [Carpino et al. 2015]. PDGs were identified as heterogeneous populations of OCT4-/PDX1+/SOX9+ cells associated with pancreatic ducts and occasionally localized in continuity with islet cluster. A progressive loss of SOX9 expression was detected in PDCs transitioning to the islets; this was interpreted as an indirect demonstration of their commitment to endocrine lineages. Proportion of the duct areas containing PDGs was estimated at 4% in the pancreatic head and in the tail. Few NGN3+ cells were detected in the PDG subpopulation of the main duct but this number increased in interlobular ducts where increased numbers (about 8% of ductal area) of insulin+ or glucagon+ cells were observed. Yet, the function of these PDG-localized insulin-expressing cells remains elusive, in particular, in the context of disease. However, these data suggest the existence of adult-committed progenitors in proximity of intercalated ducts that differ from stem cells and show heterogeneous expression patterns [Yamaguchi et al. 2015].

One of the key elements in driving pancreatic progenitors into endocrine lineage is the activation of NGN3+ expression [Desgraz and Herrera, 2009; Desgraz et al. 2011]. In a study conducted by Xu and colleagues, who observed an increase of about 40% of the β-cell mass 1 week after PDL in adult mice, re-expression of NGN3 occurred throughout the ductal lining [Xu et al. 2008]. Electron microscopy analysis of purified NGN3+ cells showed their structural resemblance to pancreatic progenitor cells in the embryo, whereas their differentiation potential toward functional β cells was reported after microinjection into embryonic NGN3-/- explants. Furthermore, α-to-β cell conversion studies using PAX4 overexpression or ARX inhibition revealed a continuous activation of pancreatic precursors expressing NGN3 that were located along the lining duct [Courtney et al. 2011, 2013; Beucher et al. 2012; Napolitano et al. 2015]. NGN3+ cells differentiated toward α cells to compensate the relative glucagon deficiency. Altogether, these data identified NGN3 as a pro-endocrine gene involved in a continuous cycle of regeneration from duct-lining cells.

In vitro studies on pancreatic progenitors

Two teams discovered multipotent progenitor cells (PMPs) in the adult mouse and human pancreas with capacities for proliferation and β-cell differentiation [Smukler et al. 2011; Razavi et al. 2015]. These PMPs were isolated and identified at low frequency (0.02–0.03%) in both nestin+ and nestin fractions, obtained following islet and ductal dissociation into single cells. Progenitor colonies arose in vitro from islet and ducts cultured at low density with EGF and FGF2 [Seaberg et al. 2004]. About 26.3 ± 1.4 % of insulin+ cells deriving from these PMPs were observed in the normal adult pancreas, whereas few cells coexpressing insulin and NGN3 were identified and accounted as possible progenitor populations. The transplantation of PMP spheres under the kidney capsule of diabetic BalbC and NOD-SCID mice significantly improved hyperglycemia and demonstrated GSIS capacities. Immunostaining analysis of the grafts showed clusters of cells coexpressing insulin and glucagon. These studies support the existence within pancreatic ductal tissue of facultative adult progenitors with extensive proliferation and differentiation capacities.

The endocrine differentiation potential of duct-lining cells was also described with centroacinar/terminal ductal (CA/TD) cell [Rovira et al. 2010]. CA/TD cells contain high levels of aldehyde dehydrogenase type 1 (ALDH1) enzymatic activity that allows their fluorescence-activated cell sorting (FACS) sorting. These cells show a progenitor-like phenotype characterized by PTF1a, SOX9, SCA-1, SDF-1, c-MET, and Nestin expression. In suspension culture, ALDH1+ cells have self-renewal activity and form ‘pancreatospheres’ with endocrine and exocrine differentiation capacities. The spheres responded to glucose challenge (0, 5 and 11 mmol/L of glucose) by secreting up to 0.9 ng/ml of C-peptide in culture media. After injection into cultured microdissected embryonic-dorsal-pancreatic buds isolated from E12.5 mouse embryos, 11.7% and 11.6% of ADLH1+ cells expressed glucagon and insulin/C-peptide, respectively. In vivo, a massive expansion of CA/TD cells was observed after intraperitoneal injections of mice with low-dose cerulein for 3 consecutive weeks [Strobel et al. 2007]. These data suggest the potential application of ADLH1+ cell expansion and transdifferentiation protocols to promote neogenesis in vitro or in vivo.

In vitro expansion of pancreatic ductal cells

PDCs have other interesting features for β-cell engineering: they represent about 35% of the human pancreatic cell mass and are easily purified; they are resistant to organ enzymatic dissociation and cell isolation procedures, and they attach to plastic. These characteristics offer unique opportunities for testing expansion protocols required to obtain a critical mass of transplantable cells.

Human primary PDCs perform one or two rounds of proliferation when plated into classical culture media (i.e. Connaught Medical Research Laboratories (CMRL) or Dulbecco’s Modified Eagle Medium (DMEM)) and rapidly enter cell-cycle arrest if not stimulated [Bonner-Weir et al. 2000]. To foster expansion of PDCs, three-dimensional (3D) culture systems were developed using human exocrine preparations and extracellular matrices such as agarose or rat-tail collagen [Kerr-Conte et al. 1996], or Matrigel [Bonner-Weir et al. 2000]. These studies had in common the development of cystic structures with PDCs increasing their proliferation by a factor of 3–7, and by the budding of endocrine cells coexpressing insulin after more than 1 week in culture. In the mouse, a much higher expansion rate (5 × 105 expansion over 11 weeks) of exocrine tissue was observed after its incorporation in 3D semisolid methylcellulose-based cultures using Matrigel [Jin et al. 2014]. In this model, authors added roof-plate-specific spondin 1 (RSPO1), a wingless-int (Wnt) signaling activator via ligation of LGR4-6 receptors, to the culture media, which greatly enhanced proliferation rates. Huch and colleagues found similar data with Matrigel-embedded mouse pancreatic-duct fragments incubated with RSPO1, EGF, fibroblastic growth factor (FGF) 10 and nicotinamide [Huch et al. 2013]. These authors reported the subpassaging of cyst-like organoids at a 1:4–1:5 ratio weekly for over 10 months, without tumoral formation, chromosomal anomalies or aneuploidy.

Many studies described the use of growth factors such as EGF, hepatocyte growth factor (HGF), KGF or nicotinamide to stimulate human PDC proliferation [Bonner-Weir et al. 2000; Rescan et al. 2005; Hoesli et al. 2012] without successfully avoiding rapid senescence and limited growth. Here again, mouse tissue showed a distinct growth pattern, as shown by Oshima and colleagues, with purified CD133+c-Met+ PDCs cultured in the presence of HGF, EGF, nicotinamide and dexamethasone [Oshima et al. 2007]. These FACS-sorted cells were capable of clonal subculture for 3 months, albeit not to the extent required for massive output of reprogrammable cells. It is noteworthy that most successful protocols for mouse PDC expansion failed to trigger replication of their human counterparts. This may be partly explained by the fact that in human cells, telomere shortening restricts replication and leads to senescence, whereas in mice and rats, telomere length is maintained during subculture [Bonner-Weir et al. 2010].

As with many other epithelial cells [Khuu et al. 2011; Russ et al. 2008], PDCs rapidly lose their phenotype after enzymatic disruption of intracellular junctions and plating in adherent culture [Bonner-Weir et al. 2000; Todorov et al. 2006]. The loss of E-cadherin precedes mesenchymal marker expression, including N-cadherin and Snail1, the master regulator of epithelial-mesenchymal transition (EMT) [Lamouille et al. 2014]. The acquisition of mesenchymal features by PDCs was shown initially on unselected human exocrine cultures [Seeberger et al. 2009; Fanjul et al. 2010], but this was preceded by demonstrations of EMT with human primary islet cultures [Gershengorn et al. 2004; Ouziel-Yahalom et al. 2006] that were able to redifferentiate into insulin-expressing cells after an expansion phase of over 65,000-fold. Russ and colleagues further exploited these properties, confirming the model by lineage tracing [Russ et al. 2008] and produced highly proliferative β-cell derivatives with potential to reverse hyperglycemia in STZ-treated NOD-SCID mice [Russ et al. 2011; Bar et al. 2012]. Similarly, the natural process of EMT can be fostered in human PDCs to overcome early senescence of primary cultures. Using FACS-sorted CA19-9+ PDCs as per the protocol of Yatoh and colleagues [Yatoh et al. 2007], we observed the proliferation of purified cultures using endothelial growth media [Corritore et al. 2014]. The cells identified as human duct-derived cells (HDDCs) shifted from a cobblestone-like to a spindle-shaped morphology and massively expanded, such that 100 × 109 cells could be harvested after 1 month of culture. HDDCs went through an E- to N-cadherin switch, while maintaining low levels of CK19 and SOX9 expression. Incubation of fresh CA19-9+ cells with TGFβ inhibitor A-83-01 blocked the appearance of HDDCs, which were not capable of clonal expansion under serial dilution experiments. Culture conditions were defined to recapitulate the pancreas embryonic development using small molecules and growth factors. After incubation with a 14-day stepwise protocol, HDDCs acquired β-cell features, including insulin secretion capacities, yet not to the extent of glucose sensitivity. Ongoing work is evaluating the possibility of bringing HDDCs to functionality and diabetes-reversal potential in animal models (Corritore et al. 2016).

In vitro reprogramming of pancreatic ductal cells into β cells

Mouse and human studies on growth-factor-based (e.g. EGF, nicotinamide, exendin-4, FGF-7) [Bonner-Weir et al. 2000, 2008; Gao et al. 2003; Kikugawa et al. 2009], spontaneous [Ramiya et al. 2000; Yatoh et al. 2007], or in vivo-induced [Hao et al. 2006] differentiation of PDCs demonstrated the potential of these cells to acquire β-cell-like features with GSIS capacities reported in perifusion models [Gao et al. 2003].

Because of the difficulties in obtaining critical amounts of epithelial PDCs in vitro, many groups investigated the endocrine differentiation potential of ductal-cell lines, such as PANC-1 cells [Zhou et al. 2002; Hardikar et al. 2003; Zhang et al. 2010] derived from a human sample of pancreatic carcinoma. Similarly, human PDCs immortalized by overexpression of E6/E7 genes from human papillomavirus were developed as a surrogate for normal PDCs because of similar genotypic and phenotypic profiles [Yang et al. 1998; Ouyang et al. 2000]. Incubation of these human pancreatic ductal epithelial (HPDE) cell lines with islet neogenesis associated protein (INGAP) favored the upregulation of β-cell markers [Assouline-Thomas et al. 2015]. After short exposure of a week with INGAP, HPDE cells showed the capacity to secrete insulin in response to glucose, albeit the values observed in basal (5.8 mmol of glucose) and stimulated conditions (25 mmol of glucose) were not statistically significant. Interestingly, an increase of glucagon, somatostatin, and pancreatic polypeptide (PPY) gene expression was also detected at day 7 of the incubation period, suggesting the induction of an endocrine program elicited by INGAP. Nevertheless, the outcome of similar differentiation protocols in unmodified primary cultures is still doubtful.

In parallel with studies using cell lines, the β-cell differentiation capacities of the pancreatic exocrine tissue was also investigated, with leaders in the field being Bouwen’s and Baeyens’s groups [Baeyens and Bouwens, 2008; Bouwens et al. 2013] that accumulated in vitro studies both on murine and human tissues. Recent developments in their work showed the potential to induce NGN3 expression in human exocrine cells by lentiviral-induced overexpression of mitogen-activated protein kinase, and signal transducer and activator of transcription 3 [Lemper et al. 2015]. The NGN3-expressing cells entered an endocrine program and specified into functional β cells 42 days after transplantation into normoglycemic immunocompromised mice. Although elastase 2A-expressing acinar cells were shown as the parent of insulin+ cells, a state of acinar-to-ductal dedifferentiation was observed before endocrine reprogramming, as described earlier with human cells [Houbracken et al. 2011; Staels et al. 2015]. This particularity of a ductal intermediate was already suggested in vitro by Gomez and colleagues [Gomez et al. 2015], who showed the possibility to FACS-sort NGN3+ cells from human exocrine tissue cultures based on their selective coexpression of CD133, expressed on the apical surface of CA19–9+ PDCs. Interestingly, these NGN3+CD133+ duct-lining cells, which resembled endocrine progenitors in their expression profile, had features of both acinar and ductal lineages, confirming the continuum between those two exocrine compartments, at least in culture. Similar duct-lining cells coexpressing NGN3 and duct markers (e.g. osteopontin or HNF1β) were purified from adult mice subjected to pancreas injury [Xu et al. 2008] or to genetic modifications to foster α-to-β cell transdifferentiation [Al-Hasani et al. 2013]. How these cells participate in the endocrine reprogramming potential of the exocrine compartment is unclear since in vitro studies involved ipso facto unselected tissues. A recent study by Klein and colleagues using cultures of human islet-depleted exocrine tissue showed that acinar (elastase 3a+) and ductal (CAII+) cells participated poorly to bone morphogenetic 7-induced endocrine reprogramming [Klein et al. 2015]. Instead, cells expressing PDX1 at the time of lentiviral-based tagging participated to almost 50% of the C-peptide+ cells retrieved after differentiation protocols. Although these results might be partly influenced by lineage-dependent transduction efficiency, it clearly highlights issues related with the use of unselected exocrine preparations, that is, contamination with endocrine cells (69% of fresh cells expressed PDX1 protein) and natural EMT processes (79% of the insulin-Cre progeny expressed vimentin after 12 days) leading to overestimation of the initial presence of progenitor cells and/or of the contribution of a cellular subtype in the endocrine reprogramming.

Purification of human PDCs unequivocally addresses the potential of the ductal-cell compartment for in vitro neogenesis. Several membrane proteins were studied for their duct-specific expression, such as lectin (binding peanut agglutinin [Barresi et al. 1993], CA19-9 [Gmyr et al. 2004; Yatoh et al. 2007], CD133 [Lee et al. 2013], or antigens targeted by specific monoclonal antibodies [Dorrell et al. 2008; Hald et al. 2012]. The Bonner-Weir group was one of the first to show spontaneous differentiation of purified human PDCs after aggregation [Yatoh et al. 2007]. In their study, CA19-9+ PDCs had increased capacities to form insulin-producing cells after coculture with pancreatic stromal cells. Likewise, human CD133+ PDCs exclusively expressing duct-specific markers (such as keratin 19) were successfully isolated using flow cytometry [Lee et al. 2013]. These cells were submitted to adenoviral-based concomitant overexpression of PDX1, MAFA, NGN3, and PAX6 that triggered β-cell differentiation to levels close to β cells regarding insulin content (up to 15.2 pmol/µg DNA), GSIS and ultrastructure. This study confirmed previous reports of unselected PDCs that showed reprogramming of PDCs into insulin-producing cells after transduction of NGN3 [Heremans et al. 2002] with or without MYT1 [Swales et al. 2012]. Similar outcomes were obtained in adult mouse PDCs subjected to reprogramming after incubation with a polycistronic construct carrying NGN3, PDX1, and MAFA TFs [Yamada et al. 2015] borrowed from the study by Zhou and colleagues that achieved acinar-to-β transdifferentiation [Cavelti-Weder et al. 2015]. Interestingly, Yamada and colleagues [Yamada et al. 2015] showed the need to add exendin-4 to culture media for PDCs to acquire GSIS, a feature not observed in vivo by other groups [Zhou et al. 2008; Cavelti-Weder et al. 2015].

Conclusion

Over the last 15 years, strategies to generate insulin-producing cells have been thoroughly investigated. Committed progenitor cells with capacity for renewal of the endocrine compartment after tissue damage were identified in adult pancreata within the ductal epithelium. Re-expression of NGN3 in this subpopulation of pancreatic precursors was identified as an essential step anticipating the regeneration process. In vivo experiments performed in adult pancreas showed replenishment of β-cell mass and restoration of glycemic levels in STZ- or alloxan-treated rodents due to conversion of ductal progenitors into insulin-producing cells following treatment with growth factors or small molecules.

In parallel, efforts have been made to exploit PDCs as a safe source of β-like cells for translational studies. Current investigations are focusing on the potential to expand and differentiate PDCs after purification. Recent models for expansion of purified human PDCs were reported, and in vitro differentiation protocols achieved functional β-cell phenotypes within a few weeks. Animal studies will help to determine long-term functionality of these systems and the safety of expanded PDCs after transplantation. Once the proof of concept of diabetes reversal with purified human PDCs is unequivocally demonstrated, PDCs might be considered for developing new strategies for β-cell mass regeneration in the human setting.

Footnotes

Funding: This study was supported by grants from the Belgian Society for Pediatric Endocrinology and Diabetology (BESPEED), the International Society for Pediatric and Adolescent Diabetes (ISPAD), the Institut de Recherche Clinique et Expérimentale (IREC), and the Fonds National de la Recherche Scientifique (FNRS).

Conflict of interest statement: The authors declare that there is no conflict of interest.

Contributor Information

Elisa Corritore, Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium.

Yong-Syu Lee, Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium.

Etienne M. Sokal, Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium

Philippe A. Lysy, Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium.

References

  1. Agulnick A., Ambruzs D., Moorman M., Bhoumik A., Cesario R., Payne J., et al. (2015) Insulin-producing endocrine cells differentiated in vitro from human embryonic stem cells function in macroencapsulation devices in vivo. Stem Cells Transl Med 4: 1214–1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Al-Hasani K., Pfeifer A., Courtney M., Ben-Othman N., Gjernes E., Vieira A., et al. (2013) Adult duct-lining cells can reprogram into beta-like cells able to counter repeated cycles of toxin-induced diabetes. Dev Cell 26: 86–100. [DOI] [PubMed] [Google Scholar]
  3. Annes J., Ryu J., Lam K., Carolan P., Utz K., Hollister-Lock J., et al. (2012) Adenosine kinase inhibition selectively promotes rodent and porcine islet beta-cell replication. Proc Natl Acad Sci USA 109: 3915–3920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Assouline-Thomas B., Ellis D., Petropavlovskaia M., Makhlin J., Ding J., Rosenberg L. (2015) Islet neogenesis associated protein (INGAP) induces the differentiation of an adult human pancreatic ductal cell line into insulin-expressing cells through stepwise activation of key transcription factors for embryonic beta cell development. Differentiation 90: 77–90. [DOI] [PubMed] [Google Scholar]
  5. Atkinson M., Bluestone J., Eisenbarth G., Hebrok M., Herold K., Accili D., et al. (2011) How does type 1 diabetes develop?: the notion of homicide or beta-cell suicide revisited. Diabetes 60: 1370–1379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Baeyens L., Bouwens L. (2008) Can beta-cells be derived from exocrine pancreas? Diabetes Obes Metab 10: 170–178. [DOI] [PubMed] [Google Scholar]
  7. Baeyens L., Lemper M., Leuckx G., De Groef S., Bonfanti P., Stange G., et al. (2014) Transient cytokine treatment induces acinar cell reprogramming and regenerates functional beta cell mass in diabetic mice. Nat Biotechnol 32: 76–83. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  8. Bar Y., Russ H., Sintov E., Anker-Kitai L., Knoller S., Efrat S. (2012) Redifferentiation of expanded human pancreatic beta-cell-derived cells by inhibition of the notch pathway. J Biol Chem 287: 17269–17280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Barresi G., Vitarelli E., Grosso M., Tuccari G. (1993) Peanut lectin binding sites in human foetal and neonatal pancreas. Eur J Histochem 37: 329–334. [PubMed] [Google Scholar]
  10. Barton F., Rickels M., Alejandro R., Hering B., Wease S., Naziruddin B., et al. (2012) Improvement in outcomes of clinical islet transplantation: 1999–2010. Diabetes Care 35: 1436–1445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bellin M., Barton F., Heitman A., Harmon J., Kandaswamy R., Balamurugan A., et al. (2012) Potent induction immunotherapy promotes long-term insulin independence after islet transplantation in type 1 diabetes. Am J Transplant 12: 1576–1583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Beucher A., Martin M., Spenle C., Poulet M., Collin C., Gradwohl G. (2012) Competence of failed endocrine progenitors to give rise to acinar but not ductal cells is restricted to early pancreas development. Dev Biol 361: 277–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Blyszczuk P., Asbrand C., Rozzo A., Kania G., St-Onge L., Rupnik M., et al. (2004) Embryonic stem cells differentiate into insulin-producing cells without selection of nestin-expressing cells. Int J Dev Biol 48: 1095–1104. [DOI] [PubMed] [Google Scholar]
  14. Boerner B., George N., Mir S., Sarvetnick N. (2015) Ws6 induces both alpha and beta cell proliferation without affecting differentiation or viability. Endocr J 62: 379–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Boettler T., Schneider D., Cheng Y., Kadoya K., Brandon E., Martinson L., et al. (2015) Pancreatic tissue transplanted in theracyte encapsulation devices are protected and prevent hyperglycemia in a mouse model of immune-mediated diabetes. Cell Transplant 25: 559–574. [DOI] [PubMed] [Google Scholar]
  16. Bonfanti P., Nobecourt E., Oshima M., Albagli-Curiel O., Laurysens V., Stange G., et al. (2015) Ex vivo expansion and differentiation of human and mouse fetal pancreatic progenitors are modulated by epidermal growth factor. Stem Cells Dev 24: 1766–1778. [DOI] [PubMed] [Google Scholar]
  17. Bonner-Weir S., Guo L., Li W., Ouziel-Yahalom L., Lysy P., Weir G., et al. (2012) Islet neogenesis: a possible pathway for beta-cell replenishment. Rev Diabet Stud 9: 407–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Bonner-Weir S., Inada A., Yatoh S., Li W.C., Aye T., Toschi E., et al. (2008) Transdifferentiation of pancreatic ductal cells to endocrine beta-cells. Biochem Soc Trans 36: 353–356. [DOI] [PubMed] [Google Scholar]
  19. Bonner-Weir S., Li W., Ouziel-Yahalom L., Guo L., Weir G., Sharma A. (2010) Beta-cell growth and regeneration: replication is only part of the story. Diabetes 59: 2340–2348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Bonner-Weir S., Taneja M., Weir G., Tatarkiewicz K., Song K., Sharma A., et al. (2000) In vitro cultivation of human islets from expanded ductal tissue. Proc Natl Acad Sci USA 97: 7999–8004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Bouwens L., Houbracken I., Mfopou J. (2013) The use of stem cells for pancreatic regeneration in diabetes mellitus. Nat Rev Endocrinol 9: 598–606. [DOI] [PubMed] [Google Scholar]
  22. Brennan D., Kopetskie H., Sayre P., Alejandro R., Cagliero E., Shapiro A., et al. (2016)Long-term follow-up of the edmonton protocol of islet transplantation in the United States. Am J Transplant 16: 509–517. [DOI] [PubMed] [Google Scholar]
  23. Bruin J., Erener S., Vela J., Hu X., Johnson J., Kurata H., et al. (2014) Characterization of polyhormonal insulin-producing cells derived in vitro from human embryonic stem cells. Stem Cell Res 12: 194–208. [DOI] [PubMed] [Google Scholar]
  24. Bruni A., Gala-Lopez B., Pepper A., Abualhassan N., Shapiro A. (2014) Islet cell transplantation for the treatment of type 1 diabetes: recent advances and future challenges. Diabetes Metab Syndr Obes 7: 211–223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Carpino G., Renzi A., Cardinale V., Franchitto A., Onori P., Overi D., et al. (2015) Progenitor cell niches in the human pancreatic duct system and associated pancreatic duct glands: an anatomical and immunophenotyping study. J Anat 228: 474–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Cavelti-Weder C., Li W., Zumsteg A., Stemann-Andersen M., Zhang Y., Yamada T., et al. (2015) Hyperglycaemia attenuates in vivo reprogramming of pancreatic exocrine cells to beta cells in mice. Diabetologia 59: 522–532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Cavelti-Weder C., Shtessel M., Reuss J., Jermendy A., Yamada T., Caballero F., et al. (2013) Pancreatic duct ligation after almost complete beta-cell loss: exocrine regeneration but no evidence of beta-cell regeneration. Endocrinology 154: 4493–4502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Chintinne M., Stange G., Denys B. In ‘T Veld P., Hellemans K., Pipeleers-Marichal M., et al. (2010) Contribution of postnatally formed small beta cell aggregates to functional beta cell mass in adult rat pancreas. Diabetologia 53: 2380–2388. [DOI] [PubMed] [Google Scholar]
  29. Collombat P., Xu X., Heimberg H., Mansouri A. (2010) Pancreatic beta-cells: from generation to regeneration. Semin Cell Dev Biol 21: 838–844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Collombat P., Xu X., Ravassard P., Sosa-Pineda B., Dussaud S., Billestrup N., et al. (2009) The ectopic expression of PAX4 in the mouse pancreas converts progenitor cells into alpha and subsequently beta cells. Cell 138: 449–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Corritore E., Dugnani E., Pasquale V., Misawa R., Witkowski P., Lei J., et al. (2014) Beta-cell differentiation of human pancreatic duct-derived cells after in vitro expansion. Cell Reprogram 16: 456–466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Corritore E., Lee Y.S., Pasquale V., Liberati D., Hsu M.J., Lombard C., Van Der Smissen P., Vetere A., Bonner-Weir S., Piemonti L., Sokal E., Lysy P.A. (2016). MAFA smRNA drives reprogramming of human pancreatic duct-derived cells into insulin-secreting cells. Stem Cells Transl Med. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Courtney M., Gjernes E., Druelle N., Ravaud C., Vieira A., Ben-Othman N., et al. (2013) The inactivation of ARX in pancreatic alpha-cells triggers their neogenesis and conversion into functional beta-like cells. PLoS Genet 9: e1003934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Courtney M., Pfeifer A., Al-Hasani K., Gjernes E., Vieira A., Ben-Othman N., et al. (2011) In vivo conversion of adult alpha-cells into beta-like cells: a new research avenue in the context of type 1 diabetes. Diabetes Obes Metab 13: 47–52. [DOI] [PubMed] [Google Scholar]
  35. Criscimanna A., Speicher J., Houshmand G., Shiota C., Prasadan K., Ji B., et al. (2011) Duct cells contribute to regeneration of endocrine and acinar cells following pancreatic damage in adult mice. Gastroenterology 141: 1451–1462; e1451–1456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. D’Amour K., Agulnick A., Eliazer S., Kelly O., Kroon E., Baetge E. (2005) Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat Biotechnol 23: 1534–1541. [DOI] [PubMed] [Google Scholar]
  37. D’Amour K., Bang A., Eliazer S., Kelly O., Agulnick A., Smart N., et al. (2006) Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol 24: 1392–1401. [DOI] [PubMed] [Google Scholar]
  38. De Breuck S., Baeyens L., Bouwens L. (2006) Expression and function of leukaemia inhibitory factor and its receptor in normal and regenerating rat pancreas. Diabetologia 49: 108–116. [DOI] [PubMed] [Google Scholar]
  39. Desai B., Oliver-Krasinski J., De Leon D., Farzad C., Hong N., Leach S., et al. (2007) Preexisting pancreatic acinar cells contribute to acinar cell, but not islet beta cell, regeneration. J Clin Invest 117: 971–977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Desgraz R., Bonal C., Herrera P. (2011) Beta-cell regeneration: the pancreatic intrinsic faculty. Trends Endocrinol Metab 22: 34–43. [DOI] [PubMed] [Google Scholar]
  41. Desgraz R., Herrera P. (2009) Pancreatic neurogenin 3-expressing cells are unipotent islet precursors. Development 136: 3567–3574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Ding L., Gysemans C., Mathieu C. (2013) Beta-cell differentiation and regeneration in type 1 diabetes. Diabetes Obes Metab 15: 98–104. [DOI] [PubMed] [Google Scholar]
  43. Dominguez-Bendala J., Lanzoni G., Klein D., Alvarez-Cubela S., Pastori R. (2016) The human endocrine pancreas: new insights on replacement and regeneration. Trends Endocrinol Metab 27: 153–162. [DOI] [PubMed] [Google Scholar]
  44. Dorrell C., Abraham S., Lanxon-Cookson K., Canaday P., Streeter P., Grompe M. (2008) Isolation of major pancreatic cell types and long-term culture-initiating cells using novel human surface markers. Stem Cell Res 1: 183–194. [DOI] [PubMed] [Google Scholar]
  45. El-Gohary Y., Wiersch J., Tulachan S., Xiao X., Guo P., Rymer C., et al. (2016) Intraislet pancreatic ducts can give rise to insulin-positive cells. Endocrinology 157: 166–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Fanjul M., Gmyr V., Sengenes C., Ratovo G., Dufresne M., Lefebvre B., et al. (2010) Evidence for epithelial-mesenchymal transition in adult human pancreatic exocrine cells. J Histochem Cytochem 58: 807–823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Fomina-Yadlin D., Kubicek S., Walpita D., Dancik V., Hecksher-Sorensen J., Bittker J., et al. (2010) Small-molecule inducers of insulin expression in pancreatic alpha-cells. Proc Natl Acad Sci USA 107: 15099–15104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Furuyama K., Kawaguchi Y., Akiyama H., Horiguchi M., Kodama S., Kuhara T., et al. (2011) Continuous cell supply from a SOX9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nat Genet 43: 34–41. [DOI] [PubMed] [Google Scholar]
  49. Gao R., Ustinov J., Pulkkinen M., Lundin K., Korsgren O., Otonkoski T. (2003) Characterization of endocrine progenitor cells and critical factors for their differentiation in human adult pancreatic cell culture. Diabetes 52: 2007–2015. [DOI] [PubMed] [Google Scholar]
  50. Gershengorn M., Hardikar A., Wei C., Geras-Raaka E., Marcus-Samuels B., Raaka B. (2004) Epithelial-to-mesenchymal transition generates proliferative human islet precursor cells. Science 306: 2261–2264. [DOI] [PubMed] [Google Scholar]
  51. Gmyr V., Belaich S., Muharram G., Lukowiak B., Vandewalle B., Pattou F., et al. (2004) Rapid purification of human ductal cells from human pancreatic fractions with surface antibody Ca19-9. Biochem Biophys Res Commun 320: 27–33. [DOI] [PubMed] [Google Scholar]
  52. Gomez D., O’Driscoll M., Sheets T., Hruban R., Oberholzer J., McGarrigle J., et al. (2015) Neurogenin 3 expressing cells in the human exocrine pancreas have the capacity for endocrine cell fate. PLoS One 10: e0133862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Hald J., Galbo T., Rescan C., Radzikowski L., Sprinkel A., Heimberg H., et al. (2012) Pancreatic islet and progenitor cell surface markers with cell sorting potential. Diabetologia 55: 154–165. [DOI] [PubMed] [Google Scholar]
  54. Hao E., Tyrberg B., Itkin-Ansari P., Lakey J., Geron I., Monosov E., et al. (2006) Beta-cell differentiation from nonendocrine epithelial cells of the adult human pancreas. Nat Med 12: 310–316. [DOI] [PubMed] [Google Scholar]
  55. Hardikar A., Marcus-Samuels B., Geras-Raaka E., Raaka B., Gershengorn M. (2003) Human pancreatic precursor cells secrete FGF2 to stimulate clustering into hormone-expressing islet-like cell aggregates. Proc Natl Acad Sci USA 100: 7117–7122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Heremans Y., Van De Casteele M., In’t Veld P., Gradwohl G., Serup P., Madsen O., et al. (2002) Recapitulation of embryonic neuroendocrine differentiation in adult human pancreatic duct cells expressing neurogenin 3. J Cell Biol 159: 303–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Hesselson D., Anderson R., Stainier D. (2011) Suppression of PTF1a activity induces acinar-to-endocrine conversion. Curr Biol 21: 712–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Hoesli C., Johnson J., Piret J. (2012) Purified human pancreatic duct cell culture conditions defined by serum-free high-content growth factor screening. PLoS One 7: e33999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Houbracken I., Bouwens L. (2010) The quest for tissue stem cells in the pancreas and other organs, and their application in beta-cell replacement. Rev Diabet Stud 7: 112–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Houbracken I., De Waele E., Lardon J., Ling Z., Heimberg H., Rooman I., et al. (2011) Lineage tracing evidence for transdifferentiation of acinar to duct cells and plasticity of human pancreas. Gastroenterology 141: 731–741, 741 e731–734. [DOI] [PubMed] [Google Scholar]
  61. Hrvatin S., O’Donnell C., Deng F., Millman J., Pagliuca F., Diiorio P., et al. (2014) Differentiated human stem cells resemble fetal, not adult, beta cells. Proc Natl Acad Sci USA 111: 3038–3043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Huch M., Bonfanti P., Boj S., Sato T., Loomans C., Van De Wetering M., et al. (2013) Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the LGR5/r-spondin axis. EMBO J 32: 2708–2721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Inada A., Nienaber C., Katsuta H., Fujitani Y., Levine J., Morita R., et al. (2008) Carbonic anhydrase II-positive pancreatic cells are progenitors for both endocrine and exocrine pancreas after birth. Proc Natl Acad Sci USA 105: 19915–19919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Inverardi L. (2015) Improved graft survival in islet transplant recipients treated with filgrastim and exenatide. 75th scientific sessions of American Diabetes Association, June, Boston, MA. [Google Scholar]
  65. Jackson S., Schiesser J., Stanley E., Elefanty A. (2010) Differentiating embryonic stem cells pass through ‘temporal windows’ that mark responsiveness to exogenous and paracrine mesendoderm inducing signals. PLoS One 5: e10706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Jiang F., Morahan G. (2015) Multipotent pancreas progenitors: inconclusive but pivotal topic. World J Stem Cells 7: 1251–1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Jin L., Feng T., Chai J., Ghazalli N., Gao D., Zerda R., et al. (2014) Colony-forming progenitor cells in the postnatal mouse liver and pancreas give rise to morphologically distinct insulin-expressing colonies in 3D cultures. Rev Diabet Stud 11: 35–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Johannesson B., Sui L., Freytes D., Creusot R., Egli D. (2015) Toward beta cell replacement for diabetes. EMBO J 34: 841–855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Kandaswamy R., Skeans M., Gustafson S., Carrico R., Prentice M., Israni A., et al. (2016) Pancreas. Am J Transplant 16: 47–68. [DOI] [PubMed] [Google Scholar]
  70. Kaufman D. (2015) State of the art of solid organ pancreas transplantation. 75th scientific sessions of American Diabetes Association, June, Boston, MA. [Google Scholar]
  71. Kelly K., Hollingsworth M., Brand R., Liu C., Singh V., Srivastava S., et al. (2015) Advances in biomedical imaging, bioengineering, and related technologies for the development of biomarkers of pancreatic disease: summary of a National Institute of Diabetes and Digestive and Kidney Diseases and National Institute of Biomedical Imaging and Bioengineering workshop. Pancreas 44: 1185–1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Kelly O., Chan M., Martinson L., Kadoya K., Ostertag T., Ross K., et al. (2011) Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells. Nat Biotechnol 29: 750–756. [DOI] [PubMed] [Google Scholar]
  73. Kerr H., Hatipoglu B., Krishnamurthi V. (2015) Pancreas transplant for diabetes mellitus. Cleve Clin J Med 82: 738–744. [DOI] [PubMed] [Google Scholar]
  74. Kerr-Conte J., Pattou F., Lecomte-Houcke M., Xia Y., Boilly B., Proye C., et al. (1996) Ductal cyst formation in collagen-embedded adult human islet preparations. A means to the reproduction of nesidioblastosis in vitro. Diabetes 45: 1108–1114. [DOI] [PubMed] [Google Scholar]
  75. Khuu D., Scheers I., Ehnert S., Jazouli N., Nyabi O., Buc-Calderon P., et al. (2011) In vitro differentiated adult human liver progenitor cells display mature hepatic metabolic functions: a potential tool for in vitro pharmacotoxicological testing. Cell Transplant 20: 287–302. [DOI] [PubMed] [Google Scholar]
  76. Kikugawa R., Katsuta H., Akashi T., Yatoh S., Weir G., Sharma A., et al. (2009) Differentiation of copas-sorted non-endocrine pancreatic cells into insulin-positive cells in the mouse. Diabetologia 52: 645–652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Klein D., Alvarez-Cubela S., Lanzoni G., Vargas N., Prabakar K., Boulina M., et al. (2015) BMP-7 induces adult human pancreatic exocrine-to-endocrine conversion. Diabetes 64: 4123–4134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Kopinke D., Murtaugh L. (2010) Exocrine-to-endocrine differentiation is detectable only prior to birth in the uninjured mouse pancreas. BMC Dev Biol 10: 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Kopp J., Dubois C., Schaffer A., Hao E., Shih H., Seymour P., et al. (2011) SOX9+ ductal cells are multipotent progenitors throughout development but do not produce new endocrine cells in the normal or injured adult pancreas. Development 138: 653–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Krogvold L., Edwin B., Buanes T., Frisk G., Skog O., Anagandula M., et al. (2015) Detection of a low-grade enteroviral infection in the islets of langerhans of living patients newly diagnosed with type 1 diabetes. Diabetes 64: 1682–1687. [DOI] [PubMed] [Google Scholar]
  81. Kroon E., Martinson L., Kadoya K., Bang A., Kelly O., Eliazer S., et al. (2008) Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol 26: 443–452. [DOI] [PubMed] [Google Scholar]
  82. Ku H., Zhang N., Kubo A., O’Connor R., Mao M., Keller G., et al. (2004) Committing embryonic stem cells to early endocrine pancreas in vitro. Stem Cells 22: 1205–1217. [DOI] [PubMed] [Google Scholar]
  83. Lamouille S., Xu J., Derynck R. (2014) Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 15: 178–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Lee J., Sugiyama T., Liu Y., Wang J., Gu X., Lei J., et al. (2013) Expansion and conversion of human pancreatic ductal cells into insulin-secreting endocrine cells. Elife 2: e00940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Lemaire L., Goulley J., Kim Y., Carat S., Jacquemin P., Rougemont J., et al. (2015) Bicaudal C1 promotes pancreatic neurog3+ endocrine progenitor differentiation and ductal morphogenesis. Development 142: 858–870. [DOI] [PubMed] [Google Scholar]
  86. Lemper M., Leuckx G., Heremans Y., German M., Heimberg H., Bouwens L., et al. (2015) Reprogramming of human pancreatic exocrine cells to beta-like cells. Cell Death Differ 22: 1117–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Li W., Rukstalis J., Nishimura W., Tchipashvili V., Habener J., Sharma A., et al. (2010) Activation of pancreatic-duct-derived progenitor cells during pancreas regeneration in adult rats. J Cell Sci 123: 2792–2802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Lind M., Svensson A., Kosiborod M., Gudbjornsdottir S., Pivodic A., Wedel H., et al. (2014) Glycemic control and excess mortality in type 1 diabetes. N Engl J Med 371: 1972–1982. [DOI] [PubMed] [Google Scholar]
  89. Long S., Rieck M., Sanda S., Bollyky J., Samuels P., Goland R., et al. (2012) Rapamycin/Il-2 combination therapy in patients with type 1 diabetes augments tregs yet transiently impairs beta-cell function. Diabetes 61: 2340–2348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Ludvigsson J. (2013) C-peptide in diabetes diagnosis and therapy. Front Biosci (Elite Ed) 5: 214–223. [DOI] [PubMed] [Google Scholar]
  91. Lysy P. (2014) Update on genetic determinants of type 1 diabetes. J Mol Genet Med 8: 126. [Google Scholar]
  92. Lysy P., Weir G., Bonner-Weir S. (2012) Concise review: pancreas regeneration: recent advances and perspectives. Stem Cells Transl Med 1: 150–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Lysy P., Weir G., Bonner-Weir S. (2013) Making beta cells from adult cells within the pancreas. Curr Diab Rep 13: 695–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Manning Fox J., Lyon J., Dai X., Wright R., Hayward J., Van De Bunt M., et al. (2015) Human islet function following 20 years of cryogenic biobanking. Diabetologia 58: 1503–1512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Marquez-Aguirre A., Canales-Aguirre A., Padilla-Camberos E., Esquivel-Solis H., Diaz-Martinez N. (2015) Development of the endocrine pancreas and novel strategies for beta-cell mass restoration and diabetes therapy. Braz J Med Biol Res 48: 765–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Mathieu C. (2015) Vitamin D and diabetes: where do we stand? Diabetes Res Clin Pract 108: 201–209. [DOI] [PubMed] [Google Scholar]
  97. Mayer-Davis E., Dabelea D., Crandell J., Crume T., D’Agostino R., Jr., Dolan L., et al. (2013) Nutritional factors and preservation of c-peptide in youth with recently diagnosed type 1 diabetes: Search Nutrition Ancillary Study. Diabetes Care 36: 1842–1850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Meier J., Butler A., Saisho Y., Monchamp T., Galasso R., Bhushan A., et al. (2008) Beta-cell replication is the primary mechanism subserving the postnatal expansion of beta-cell mass in humans. Diabetes 57: 1584–1594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Mfopou J., Bouwens L. (2013) [Differentiation of pluripotent stem cells into pancreatic lineages]. Med Sci (Paris) 29: 736–743. [DOI] [PubMed] [Google Scholar]
  100. Mfopou J., Chen B., Mateizel I., Sermon K., Bouwens L. (2010) Noggin, retinoids, and fibroblast growth factor regulate hepatic or pancreatic fate of human embryonic stem cells. Gastroenterology 138: 2233–2245; e2231–2214. [DOI] [PubMed] [Google Scholar]
  101. Motte E., Szepessy E., Suenens K., Stange G., Bomans M., Jacobs-Tulleneers-Thevissen D., et al. (2014) Composition and function of macroencapsulated human embryonic stem cell-derived implants: comparison with clinical human islet cell grafts. Am J Physiol Endocrinol Metab 307: e838–846. [DOI] [PubMed] [Google Scholar]
  102. Nakamura K., Minami K., Tamura K., Iemoto K., Miki T., Seino S. (2011) Pancreatic beta-cells are generated by neogenesis from non-beta-cells after birth. Biomed Res 32: 167–174. [DOI] [PubMed] [Google Scholar]
  103. Napolitano T., Avolio F., Courtney M., Vieira A., Druelle N., Ben-Othman N., et al. (2015) PAX4 acts as a key player in pancreas development and plasticity. Semin Cell Dev Biol 44: 107–114. [DOI] [PubMed] [Google Scholar]
  104. Niederhaus S. (2015) Pancreas transplant alone. Curr Opin Organ Transplant 20: 115–120. [DOI] [PubMed] [Google Scholar]
  105. Noble J. (2015) Immunogenetics of type 1 diabetes: a comprehensive review. J Autoimmun 64: 101–112. [DOI] [PubMed] [Google Scholar]
  106. Nostro M., Sarangi F., Ogawa S., Holtzinger A., Corneo B., Li X., et al. (2011) Stage-specific signaling through TGF beta family members and wnt regulates patterning and pancreatic specification of human pluripotent stem cells. Development 138: 861–871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Oshima Y., Suzuki A., Kawashimo K., Ishikawa M., Ohkohchi N., Taniguchi H. (2007) Isolation of mouse pancreatic ductal progenitor cells expressing CD133 and c-met by flow cytometric cell sorting. Gastroenterology 132: 720–732. [DOI] [PubMed] [Google Scholar]
  108. Ouyang H., Mou L., Luk C., Liu N., Karaskova J., Squire J., et al. (2000) Immortal human pancreatic duct epithelial cell lines with near normal genotype and phenotype. Am J Pathol 157: 1623–1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Ouziel-Yahalom L., Zalzman M., Anker-Kitai L., Knoller S., Bar Y., Glandt M., et al. (2006) Expansion and redifferentiation of adult human pancreatic islet cells. Biochem Biophys Res Commun 341: 291–298. [DOI] [PubMed] [Google Scholar]
  110. Pagliuca F., Millman J., Gurtler M., Segel M., Van Dervort A., Ryu J., et al. (2014) Generation of functional human pancreatic beta cells in vitro. Cell 159: 428–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Pan F., Bankaitis E., Boyer D., Xu X., Van De Casteele M., Magnuson M., et al. (2013) Spatiotemporal patterns of multipotentiality in PTF1a-expressing cells during pancreas organogenesis and injury-induced facultative restoration. Development 140: 751–764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Pan F., Wright C. (2011) Pancreas Organogenesis: From Bud to Plexus to Gland. Dev Dyn 240: 530–565. [DOI] [PubMed] [Google Scholar]
  113. Pepper A., Pawlick R., Gala-Lopez B., Macgillivary A., Mazzuca D., White D., et al. (2015) Diabetes is reversed in a murine model by marginal mass syngeneic islet transplantation using a subcutaneous cell pouch device. Transplantation 99: 2294–2300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Pfeifer A., Courtney M., Ben-Othman N., Al-Hasani K., Gjernes E., Vieira A., et al. (2013) Induction of multiple cycles of pancreatic beta-cell replacement. Cell Cycle 12: 3243–3244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Pin C., Ryan J., Mehmood R. (2015) Acinar cell reprogramming: a clinically important target in pancreatic disease. Epigenomics 7: 267–281. [DOI] [PubMed] [Google Scholar]
  116. Piran R., Lee S., Li C., Charbono A., Bradley L., Levine F. (2014) Pharmacological induction of pancreatic islet cell transdifferentiation: relevance to type I diabetes. Cell Death Dis 5: e1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Ramiya V., Maraist M., Arfors K., Schatz D., Peck A., Cornelius J. (2000) Reversal of insulin-dependent diabetes using islets generated in vitro from pancreatic stem cells. Nat Med 6: 278–282. [DOI] [PubMed] [Google Scholar]
  118. Rankin M., Wilbur C., Rak K., Shields E., Granger A., Kushner J. (2013) Beta-cells are not generated in pancreatic duct ligation-induced injury in adult mice. Diabetes 62: 1634–1645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Razavi R., Najafabadi H., Abdullah S., Smukler S., Arntfield M., Van Der Kooy D. (2015) Diabetes enhances the proliferation of adult pancreatic multipotent progenitor cells and biases their differentiation to more beta-cell production. Diabetes 64: 1311–1323. [DOI] [PubMed] [Google Scholar]
  120. Rescan C., Le Bras S., Lefebvre V., Frandsen U., Klein T., Foschi M., et al. (2005) EGF-induced proliferation of adult human pancreatic duct cells is mediated by the MEK/ERK cascade. Lab Invest 85: 65–74. [DOI] [PubMed] [Google Scholar]
  121. Rezania A., Bruin J., Arora P., Rubin A., Batushansky I., Asadi A., et al. (2014) Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol 32: 1121–1133. [DOI] [PubMed] [Google Scholar]
  122. Rezania A., Bruin J., Riedel M., Mojibian M., Asadi A., Xu J., et al. (2012) Maturation of human embryonic stem cell-derived pancreatic progenitors into functional islets capable of treating pre-existing diabetes in mice. Diabetes 61: 2016–2029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Riley K., Pasek R., Maulis M., Peek J., Thorel F., Brigstock D., et al. (2015) Connective tissue growth factor modulates adult beta-cell maturity and proliferation to promote beta-cell regeneration in mice. Diabetes 64: 1284–1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Rosenberg L. (1995) In vivo cell transformation: neogenesis of beta cells from pancreatic ductal cells. Cell Transplant 4: 371–383. [DOI] [PubMed] [Google Scholar]
  125. Rovira M., Scott S., Liss A., Jensen J., Thayer S., Leach S. (2010) Isolation and characterization of centroacinar/terminal ductal progenitor cells in adult mouse pancreas. Proc Natl Acad Sci USA 107: 75–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Russ H., Bar Y., Ravassard P., Efrat S. (2008) In vitro proliferation of cells derived from adult human beta-cells revealed by cell-lineage tracing. Diabetes 57: 1575–1583. [DOI] [PubMed] [Google Scholar]
  127. Russ H., Sintov E., Anker-Kitai L., Friedman O., Lenz A., Toren G., et al. (2011) Insulin-producing cells generated from dedifferentiated human pancreatic beta cells expanded in vitro. PLoS One 6: e25566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Sakaguchi Y., Inaba M., Kusafuka K., Okazaki K., Ikehara S. (2006) Establishment of animal models for three types of pancreatitis and analyses of regeneration mechanisms. Pancreas 33: 371–381. [DOI] [PubMed] [Google Scholar]
  129. Schiesser J., Wells J. (2014) Generation of beta cells from human pluripotent stem cells: are we there yet? Ann NY Acad Sci 1311: 124–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Schroeder I., Rolletschek A., Blyszczuk P., Kania G., Wobus A. (2006) Differentiation of mouse embryonic stem cells to insulin-producing cells. Nat Protoc 1: 495–507. [DOI] [PubMed] [Google Scholar]
  131. Seaberg R., Smukler S., Kieffer T., Enikolopov G., Asghar Z., Wheeler M., et al. (2004) Clonal identification of multipotent precursors from adult mouse pancreas that generate neural and pancreatic lineages. Nat Biotechnol 22: 1115–1124. [DOI] [PubMed] [Google Scholar]
  132. Seeberger K., Eshpeter A., Rajotte R., Korbutt G. (2009) Epithelial cells within the human pancreas do not coexpress mesenchymal antigens: epithelial-mesenchymal transition is an artifact of cell culture. Lab Invest 89: 110–121. [DOI] [PubMed] [Google Scholar]
  133. Shapiro A., Lakey J. (2000) Future trends in islet cell transplantation. Diabetes Technol Ther 2: 449–452. [DOI] [PubMed] [Google Scholar]
  134. Shen W., Tremblay M., Deshmukh V., Wang W., Filippi C., Harb G., et al. (2013) Small-molecule inducer of beta cell proliferation identified by high-throughput screening. J Am Chem Soc 135: 1669–1672. [DOI] [PubMed] [Google Scholar]
  135. Smukler S., Arntfield M., Razavi R., Bikopoulos G., Karpowicz P., Seaberg R., et al. (2011) The adult mouse and human pancreas contain rare multipotent stem cells that express insulin. Cell Stem Cell 8: 281–293. [DOI] [PubMed] [Google Scholar]
  136. Solar M., Cardalda C., Houbracken I., Martin M., Maestro M., De Medts N., et al. (2009) Pancreatic exocrine duct cells give rise to insulin-producing beta cells during embryogenesis but not after birth. Dev Cell 17: 849–860. [DOI] [PubMed] [Google Scholar]
  137. Song S., Roy S. (2015) Progress and challenges in macroencapsulation approaches for type 1 diabetes (T1D) treatment: cells, biomaterials, and devices. Biotechnol Bioeng. 28 November 2015. [epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Staels W., Heremans Y., Heimberg H. (2015) Reprogramming of human exocrine pancreas cells to beta cells. Best Pract Res Clin Endocrinol Metab 29: 849–857. [DOI] [PubMed] [Google Scholar]
  139. Strobel O., Dor Y., Stirman A., Trainor A., Fernandez-Del Castillo C., Warshaw A., et al. (2007) Beta cell transdifferentiation does not contribute to preneoplastic/metaplastic ductal lesions of the pancreas by genetic lineage tracing in vivo. Proc Natl Acad Sci USA 104: 4419–4424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Suarez-Pinzon W., Lakey J., Brand S., Rabinovitch A. (2005) Combination therapy with epidermal growth factor and gastrin induces neogenesis of human islet {beta}-cells from pancreatic duct cells and an increase in functional {beta}-cell mass. J Clin Endocrinol Metab 90: 3401–3409. [DOI] [PubMed] [Google Scholar]
  141. Suarez-Pinzon W., Lakey J., Rabinovitch A. (2008) Combination therapy with glucagon-like peptide-1 and gastrin induces beta-cell neogenesis from pancreatic duct cells in human islets transplanted in immunodeficient diabetic mice. Cell Transplant 17: 631–640. [DOI] [PubMed] [Google Scholar]
  142. Suissa Y., Magenheim J., Stolovich-Rain M., Hija A., Collombat P., Mansouri A., et al. (2013) Gastrin: a distinct fate of neurogenin3 positive progenitor cells in the embryonic pancreas. PLoS One 8: e70397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Swales N., Martens G., Bonne S., Heremans Y., Borup R., Van De Casteele M., et al. (2012) Plasticity of adult human pancreatic duct cells by neurogenin3-mediated reprogramming. PLoS One 7: e37055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Tellez N., Montanya E. (2014) Gastrin induces ductal cell dedifferentiation and beta-cell neogenesis after 90% pancreatectomy. J Endocrinol 223: 67–78. [DOI] [PubMed] [Google Scholar]
  145. Thorel F., Nepote V., Avril I., Kohno K., Desgraz R., Chera S., et al. (2010) Conversion of adult pancreatic alpha-cells to beta-cells after extreme beta-cell loss. Nature 464: 1149–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Todorov I., Omori K., Pascual M., Rawson J., Nair I., Valiente L., et al. (2006) Generation of human islets through expansion and differentiation of non-islet pancreatic cells discarded (pancreatic discard) after islet isolation. Pancreas 32: 130–138. [DOI] [PubMed] [Google Scholar]
  147. Tokui Y., Kozawa J., Yamagata K., Zhang J., Ohmoto H., Tochino Y., et al. (2006) Neogenesis and proliferation of beta-cells induced by human betacellulin gene transduction via retrograde pancreatic duct injection of an adenovirus vector. Biochem Biophys Res Commun 350: 987–993. [DOI] [PubMed] [Google Scholar]
  148. Vegas A., Veiseh O., Gurtler M., Millman J., Pagliuca F., Bader A., et al. (2016) Long-term glycemic control using polymer-encapsulated human stem cell-derived beta cells in immune-competent mice. Nat Med 22: 306–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Vetere A., Choudhary A., Burns S., Wagner B. (2014) Targeting the pancreatic beta-cell to treat diabetes. Nat Rev Drug Discov 13: 278–289. [DOI] [PubMed] [Google Scholar]
  150. Vieira A., Druelle N., Courtney M., Avolio F., Ben-Othman N., Pfeifer A., et al. (2013) [Reprogramming pancreatic cells to beta cells]. Med Sci (Paris) 29: 749–755. [DOI] [PubMed] [Google Scholar]
  151. Wandzioch E., Zaret K. (2009) Dynamic signaling network for the specification of embryonic pancreas and liver progenitors. Science 324: 1707–1710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Wang M., Racine J., Song X., Li X., Nair I., Liu H., et al. (2012) Mixed chimerism and growth factors augment beta cell regeneration and reverse late-stage type 1 diabetes. Sci Transl Med 4: 133ra159. [DOI] [PubMed] [Google Scholar]
  153. Xu G., Kaneto H., Lopez-Avalos M., Weir G., Bonner-Weir S. (2006) GLP-1/exendin-4 facilitates beta-cell neogenesis in rat and human pancreatic ducts. Diabetes Res Clin Pract 73: 107–110. [DOI] [PubMed] [Google Scholar]
  154. Xu X., D’Hoker J., Stange G., Bonne S., De Leu N., Xiao X., et al. (2008) Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell 132: 197–207. [DOI] [PubMed] [Google Scholar]
  155. Yamada T., Cavelti-Weder C., Caballero F., Lysy P., Guo L., Sharma A., et al. (2015) Reprogramming mouse cells with a pancreatic duct phenotype to insulin-producing beta-like cells. Endocrinology 156: 2029–2038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Yamaguchi J., Liss A., Sontheimer A., Mino-Kenudson M., Castillo C., Warshaw A., et al. (2015) Pancreatic duct glands (PDGS) are a progenitor compartment responsible for pancreatic ductal epithelial repair. Stem Cell Res 15: 190–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Yang J., Liu C., Domer P., Iannaccone P. (1998) Expression of lymphomagenic oncogenes in T-cell lymphomas of HPV 16 transgenic mice. Cancer Detect Prev 22: 405–415. [DOI] [PubMed] [Google Scholar]
  158. Yang Y., Thorel F., Boyer D., Herrera P., Wright C. (2011) Context-specific alpha-to-beta-cell reprogramming by forced PDX1 expression. Genes Dev 25: 1680–1685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Yasunaga M., Tada S., Torikai-Nishikawa S., Nakano Y., Okada M., Jakt L., et al. (2005) Induction and monitoring of definitive and visceral endoderm differentiation of mouse ES cells. Nat Biotechnol 23: 1542–1550. [DOI] [PubMed] [Google Scholar]
  160. Yatoh S., Dodge R., Akashi T., Omer A., Sharma A., Weir G., et al. (2007) Differentiation of affinity-purified human pancreatic duct cells to beta-cells. Diabetes 56: 1802–1809. [DOI] [PubMed] [Google Scholar]
  161. Zhang M., Lin Q., Qi T., Wang T., Chen C., Riggs A., et al. (2016) Growth factors and medium hyperglycemia induce SOX9+ ductal cell differentiation into beta cells in mice with reversal of diabetes. Proc Natl Acad Sci USA 113: 650–655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Zhang T., Wang H., Saunee N., Breslin M., Lan M. (2010) Insulinoma-associated antigen-1 zinc-finger transcription factor promotes pancreatic duct cell trans-differentiation. Endocrinology 151: 2030–2039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Zhang Y., Fava G., Wang H., Mauvais-Jarvis F., Fonseca V., Wu H. (2015) PAX4 gene transfer induces alpha-to-beta cell phenotypic conversion and confers therapeutic benefits for diabetes treatment. Mol Ther 24: 251–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Zhou J., Pineyro M., Wang X., Doyle M., Egan J. (2002) Exendin-4 differentiation of a human pancreatic duct cell line into endocrine cells: involvement of PDX-1 and HNF3beta transcription factors. J Cell Physiol 192: 304–314. [DOI] [PubMed] [Google Scholar]
  165. Zhou Q., Brown J., Kanarek A., Rajagopal J., Melton D. (2008) In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature 455: 627–632. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Endocrinology and Metabolism are provided here courtesy of SAGE Publications

RESOURCES