Skip to main content
Islets logoLink to Islets
. 2021 Sep 15;13(5-6):85–105. doi: 10.1080/19382014.2021.1941555

The Potential of Pancreatic Organoids for Diabetes Research and Therapy

Katerina Bittenglova a,b, David Habart a, Frantisek Saudek a, Tomas Koblas c,
PMCID: PMC8528407  PMID: 34523383

ABSTRACT

The success of clinical transplantation of pancreas or isolated pancreatic islets supports the concept of cell-based cure for diabetes. One limitation is the shortage of cadaver human pancreata. The demand–supply gap could potentially be bridged by harnessing the self-renewal capacity of stem cells. Pluripotent stem cells and adult pancreatic stem cells have been explored as possible cell sources. Recently, a system for long-term culture of proposed adult pancreatic stem cells in a form of organoids was developed. Generated organoids partially mimic the architecture and cell-type composition of pancreatic tissue. Here, we review the attempts over the past decade, to utilize the organoid cell culture principles in order to identify, expand, and differentiate the adult pancreatic stem cells from different compartments of mouse and human pancreata. The development of the culture conditions, effects of specific growth factors and small molecules is discussed. The potential utility of the adult pancreatic stem cells is considered in the context of other cell sources.

KEYWORDS: Organoids, adult stem cells, progenitor cells, pancreas, beta cell, stem cell therapy, diabetes, CD133, LGR5, ALDH, PROCR, transplantation, beta cell regeneration

1. Introduction

One great breakthrough in the field of stem cell (SC) research is the recent development of the organoid cell culture system. It can be loosely described as an “organ in a dish,”1 as it simulates organ-like growth in vitro.2 Thus, the organoids gradually make solid organs accessible for stem cell research, which until recently was possible for hematopoietic tissue only.

The cell source in the organoid culture can either be pluripotent stem cells or primary epithelial stem/progenitor cells with the potential to differentiate into organ-specific cell types.1,3,4 According to a generally accepted definition, organoids have the intrinsic capacity to expand cells and spontaneously grow into self-organized three-dimensional (3D) structures, which at least partially mirror tissue architecture, cell-type composition, and functionality of a given organ.2,5–8 As elegantly demonstrated in the intestinal organoids, the stem cells differentiate into organ-specific lineages under specific culture conditions, which activate or inhibit specific signaling pathways.9,10 The extracellular factors directing the fate of stem/progenitor cells were originally conceptualized for hematopoiesis as the “stem cell niche.”11

The term organoid was first used in 1946 to describe the tissue of dermoid cystic “teratoma.”12 Sometimes the term organoid is applied incorrectly to spheroids2 or islet-like cell clusters, which also form 3D cellular structures, but the cells are not attached to the extracellular matrix and the culture media is different from the media used for the organoid culture.13

Organoids provide a useful tool for disease modeling,4,14 drug testing,15,16 or cancer research.17 They also represent an opportunity to study tissue–pathogen interaction in vitro.18,19 Organoids can help in studying stem cell niches20 and organ development.3 In this context, pancreatic organoids have been employed to investigate pancreatic ductal adenocarcinoma,21,22 pancreas development,23–25 or cystic fibrosis,26 and to screen drugs targeting pancreatic diseases.27,28 No information has yet been published on pancreatic organoids derived from diabetic patients in order to investigate the disease pathogenesis, or to screen candidate drugs for diabetes.29 However, pancreatic organoids have been researched as the potential cell source for therapy of diabetes.

Diabetes is a chronic metabolic disease characterized by lost control over blood glucose. Type 1 diabetes is caused by an absolute deficiency of insulin. In type 2 diabetes, relative insulin deficiency results from an increased insulin resistance. At present, the type 1 diabetic patients are mostly managed by the administration of synthetic insulin.30,31 Nevertheless, a small fraction of patients are already cured from diabetes by transplantation of beta cells within cadaver pancreas (more than 48,000 patients)32 or isolated pancreatic islets (more than 1900 allograft and autograft recipients).33 Availability of the cell-based cure of diabetes is circumscribed by the limited amount cadaver pancreata. The demand–supply gap could potentially be bridged by beta cells derived from self-renewing stem cells. The three possible sources of SCs explored over the past two decades include the embryonic stem cells (ESC), induced pluripotent stem cells (iPSC), and adult pancreatic stem cells.

The first insulin-secreting cells derived from genetically modified ESCs were reported to normalize glycemia in streptozotocin-induced diabetic mice in 2000.34 In the following years, the therapeutic potential of ESCs and iPSC has been extensively tested, and high efficacy multi-stage differentiation approaches were developed.35,36 In 2014, Rezania and Pagliuca independently described protocols for in vitro differentiation of human pluripotent stem cells through pancreatic progenitors into beta cells that reversed diabetes when transplanted into mice.37,38 However, the pluripotent stem cells carry the potential for mutagenesis, chromosomal aberrations, and carcinogenesis,39 mainly due to the unphysiological number of cell divisions needed for meaningful therapeutic application.40,41 The safety can be significantly improved by cell encapsulation or by molecules that degrade the remaining undifferentiated cells.42 While the encapsulation also avoids the requirement of immunosuppression, making it safer for patients, fibrosis of the capsule hinders proper vascularization (https://viacyte.com/press-releases/two-year-data-from-viacytes-step-one-clinical-trial-presented-at-ada-2018/ STEP ONE clinical trial). On the other hand, an open system capsule allows a better degree of vascularization at the cost of immunosuppression. Bioengineered pluripotent stem cell therapy and their bottlenecks were reviewed elsewhere.43

Several biotechnology companies have already driven their successful research into pre-clinical and clinical trials.44 Viacyte developed pancreatic progenitors, which differentiate from embryonic stem cells with almost 100% efficiency into islet cells, which are implanted into diabetic patients in closed or open capsule systems (Clinical trials NCT04678557, NCT03163511 https://www.clinicaltrials.gov/ct2/show/NCT04678557?term=viacyte&draw=1&rank=1 and https://www.clinicaltrials.gov/ct2/show/NCT03163511?term=viacyte&draw=1&rank=2).45 Vertex recently registered a clinical trial to test SC-based islet cell therapy VX-880 in patients with hypoglycemia unawareness syndrome (NCT04786262 https://clinicaltrials.gov/ct2/show/NCT04786262?term=VX-880&draw=2&rank=1). Another clinical-stage company Kadimastem, which previously tested clinical grade ESC derived astrocytes (NCT03482050 https://clinicaltrials.gov/ct2/show/NCT03482050), has announced effectiveness of their microencapsulated islet-like clusters (IsletRx https://www.kadimastem.com/post/kadimastem-announces-successful-preclinical-results-of-its-cell-therapy-treatment-for-insulin-depend) in immunocompetent diabetic mice.

While pluripotent SCs appear to be a promising source of cells to cure diabetes, serious complications can still occur in clinical trials. It is therefore reasonable to consider alternative sources of insulin-producing cells, such as the adult pancreatic stem cells, which only recently became amenable to research thanks to the advancement of the organoid cell culture, as reviewed below.

2. Principles of pancreatic organoid culture

2.1. Basic principles of organoid culture

Organoid culture requires three key components: the extracellular matrix substitute, culture media, and source cells. The extracellular matrix substitutes provide specific attachment sites for cell adhesion molecules and three-dimensional support for the constituent cells. The media provide specific soluble factors to modulate signaling pathways or chromatin state. The cell source, from which pancreatic organoids are originated, can either be unselected, such as crude ductal fragments,22,46–50 or highly selected single cells carrying putative stem cell markers.46,47,51–53 The capacity of organoids to expand over numerous passages proves the presence of stem/progenitor cells in the original preparation, as well as the ability of the culture conditions to sustain them.46,47,54,55 By the same token, a failure to expand suggests either the absence of stem/progenitor cells or an inadequate media/matrix composition. When organoid cultures are initiated from unselected raw islet-depleted pancreatic tissue comprising multiple cell types, the organoids gradually prevail, while the other cell types diminish.52 The size of pancreatic organoids ranges between 50 and 2000 µm, dependent on the culture duration, as shown in Figure 1 top (we are grateful to Folia Biologica for the permission to reproduce data recently generated in our lab),52 and media composition. Once an organoid is established, some of the constituent cells can proliferate or differentiate, in response to the appropriate stimuli. Organoid cells are grown within a drop or a ring of Matrigel surrounded with medium, which is changed every 1–3 days. The passaging of organoids (generally every 7–10 days) consists of mechanical or enzymatic digestion of the basement membrane, releasing whole organoids that are further dissociated into small cell clusters or individual cells, that are subsequently replated in new Matrigel drop and culture medium. The three basic components of the organoid culture are discussed in the following chapters.

Figure 1.

Figure 1.

Example of organoid culture established from adult human pancreatic CD133+ cells. Top: Phase contrast microscopy visualizing the expansion of organoids over two weeks. Scale bar: 500 µm. Bottom: Immunohistochemistry visualizing the constituent cells at Day 15, indicating the proliferation activities (Ki67) and cell types (duct, KRT19; epithelial, ECAD; endocrine, CHGA; pancreatic progenitors PDX1, SOX9), including the original CD133+ cells. Scale bar: 200 µm

2.2. Three-dimensional extracellular matrices

Extracellular matrix in 3D organoid culture system permits the growth and expansion of cells in both horizontal and vertical planes, thus distinguishing it from the standard 2D culture system. The mammalian cells grown on a flat surface as opposed to the three-dimensional space are subjected to different mechano-chemical cues, causing differences in cytoskeleton rearrangement, gene expression, cell shape, and function. In two-dimensional culture, no gradients of nutrients and signal molecules are possible. Also, the cells are forced to apical-basal polarity, reducing lateral cell-to-cell adhesions, which are critical for development and function of cells,56 including beta cells.57 Sensitivity to such mechano-chemical cues was demonstrated for stem cells and pancreatic progenitors.58,59 In 3D culture systems, the individual cells utilize a range of surface adhesion molecules to reaggregate among themselves and to interact with fibrillar proteins of the matrix; consequently, chemical gradients are generated.60–62 3D organoid cultures can further be enhanced by co-culturing with additional cell types, e.g. endothelial or mesenchymal cells, to provide signals, which at present cannot be delivered by defined chemical components.1,63

The most commonly used artificial extracellular matrix substitute is Matrigel/BME (Basement membrane extract), which generates 3D scaffold by rapid spontaneous gelification at 37°C. This assortment of gelatinous proteins of the extracellular matrix derived from Engelbreth-Holm-Swarm tumor cell line was shown to support the stem cells' self-renewal potential and preserve their undifferentiated state.64 The major Matrigel components comprise ~60% laminin, ~30% collagen IV, ~8% nidogen/entactin, and Heparan sulfate.65,66 A number of poorly defined growth factors naturally bound to the matrix were reduced in some commercial variants (e.g. BME2) to improve chemical definition and standardization. Matrigel has two major limitations: the xenogeneic cancer cell line origin, hindering its medical use; and the batch to batch variation, hampering research reproducibility.67,68

Synthetic matrices are devoid of growth factors, which are substituted in defined media, thus advancing the organoid field toward Good manufacturing practice (GMP). Defined binding sites for integrins and for other cell adhesion molecules are attached to branched polymers interconnected by a crosslinker, thus forming a hydrogel network. Three such compounds were tested in pancreatic organoid culture. First, the poly(ethylene glycol) (PEG)-based hydrogel was covalently functionalized with laminin-1, and substrates of the FXIIIa enzyme, enabling the crosslinking by Thrombin-activated factor XIIIa. Similar to Matrigel, this hydrogel also supported the morphology, cluster formation, and progenitor maintenance of pancreatic embryonic organoids, but was less potent.54 Second, another PEG-based hydrogel was functionalized with Integrin receptor binding motif containing sequence Arg-Gly-Asp (RGD) while using maleimide for crosslinking. It supported the growth of intestinal organoids derived from human embryonic/induced pluripotent stem cells,69 but the morphology of human pancreatic organoids was altered.70 Third, a dextran polymer was functionalized with RGD and crosslinked with hyaluronic acid. It supported pancreatic organoid morphology and simplified the passaging by digestion with dextranase. However, the expansion was slow and limited to only 5–6 passages in 100 days.48 Additionally, apoly-isocyanopeptide-based hydrogel functionalized with human recombinant laminin-111 supported organoids derived from adult liver,71 making it promising for developmentally related pancreatic organoids. Novel variants of synthetic hydrogels were recently reviewed elsewhere.72

2.3. Basic medium for expanding pancreatic organoids

Culture media for pancreatic organoids were developed from the medium originally established by Sato for intestinal organoids.73 Similarly, Sato’s medium was adopted for derivation of organoids from normal or tumor tissue of other digestive organs, including colon,74 stomach,75 and liver.76 Sato’s medium comprises Advanced DMEM/F12 medium and three key growth factors, EGF, Noggin, and R-Spondin-1, hence ENR medium.73 The rationale for selecting these growth factors is following. EGF was shown to potentiate proliferation, while suppressing differentiation of pancreatic endocrine embryonic progenitors during development and in vitro.77 Noggin, a member of the transforming growth factor superfamily, inhibits the bone morphogenetic protein signaling pathway, which is fundamental for solid organ development,78 including pancreas.79 R-Spondin-1 is an agonist of LGR5 receptor80 of the Wnt-β-catenin signaling pathway (Wnt),81 which, in turn, is essential for development and for self-renewal of several types of adult stem cells.82,83

The Basic Medium for pancreatic organoids is the ENR Medium enriched by additional factors, such as FGF10, Nicotinamide, N-acetylcysteine, and B27 (contains 21 ingredients, mostly antioxidants, and enzymes, such as catalase and superoxide dismutase). FGF10, a natural product of mesenchymal cells, supports the proliferative capacity of PDX1+ (Pancreatic and duodenal homeobox 1) pancreatic progenitors,84 and helps to integrate the growth and the differentiation during pancreatic development.85 Nicotinamide is an inhibitor of sirtuin (a regulator of epigenetic silencing), and poly(ADP-ribose) polymerase (PARP), which regulate protein deacetylation and DNA repair.86,87 Nicotinamide was shown to promote survival and differentiation of human pluripotent stem cells after individualization.87 Within this review, we call it “Basic Medium.”

Basic Medium was variously supplemented with Wnt pathway ligand, WNT3A (WENR medium);74 hormone Gastrin; Rho-associated protein kinase (ROCK) inhibitor, Y-27632; inhibitors of TGFβ pathway, A83-01 or SB431542; Prostaglandin E2; Hepatocyte growth factor; an inhibitor of histone deacetylases, Trichostatin A; an inhibitor of glycogen synthase kinase 3β, CHIR99021; and an activator of adenylyl cyclase, Forskolin, Tables 1, Table 2. Gastrin is produced by G-cells of the developing88 and neonatal pancreas,89 where it has documented proliferative activity. Inhibitor of the ROCK signaling pathway diminished the dissociation-induced apoptosis in embryonic stem cells in vitro.90 The contribution of the individual soluble factors to the expansion capacity of pancreatic organoids was systematically evaluated over 6 months (20 passages) in Basic Medium supplemented with WNT3A, Gastrin, A83-01, and Prostaglandin E2.22 Both human and mouse organoids failed to expand beyond the passages 3–5, when one of the following factors was omitted: EGF, Noggin, R-Spondin-1, WNT3A, Prostaglandin E2, or Nicotinamide. The omission of A83-01 or Gastrin allowed for at least ten passages.22The protein factors can be derived either from specifically designed cell lines, that express these factors and secrete them into the medium,91 or these factors can be added into the medium in the form of pure recombinant proteins, which are amenable for GMP.

Table 1.

Effect on organoid proliferation and differentiation by selected proteins and small molecules

Factor* When the factor is absent from media Reference no.
EGF (133 kDa, 1207 AA)
promotes cell proliferation
• In adult organoids, human: reduced size and expansion (2–5 passages)
• In fetal organoids, mouse/human: reduced proliferation, improved differentiation
46, 22
55
Noggin (58 kDa, 232 AA)
inhibits Bone morphogenic protein pathway
• In adult organoids, human: reduced size and expansion (4 passages)
• In adult organoids, mouse: reduced expansion (2 months)
• In embryonic organoids, mouse: increased cystic morphology
22
46
54
R-Spondin-1 (~28kDa, 263 AA) activates Lgr5 in Wnt/β-catenin pathway • In adult organoids, human: reduced size and expansion of (3 passages)
• In adult organoids, mouse: reduced expansion (2–5 passages)
22
46
WNT3A (39 kDa, 352 AA)
activates Wnt/β-catenin signaling pathway
• In adult organoids, human: reduced expansion (approx. 3 passages) 22
FGF10 (23 kDa, 208 AA), mesenchymal factor • In mouse adult organoids: reduced expansion (2–5 passages)
• In fetal organoids, mouse/human: slower expansion
• In mouse embryonic organoids: reduced acinar diff. and Pdx1 expression
• In mouse embryonic organoids: no effect after 4 days
46
55
54

54
FGF1 (17 kDa, 155 AA), mesenchymal factor • In embryonic organoids, mouse: improved endocrine differentiation
• In embryonic organoids, mouse: diminished Pdx1 expression
54
VEGF (27 kDa, 232 AA), vascular endothelial growth factor • In adult organoids, human: reduced engraftment efficiency (<3 months) 48
Nicotinamide (MW 122.12)
a vitamin B3 form
• In adult organoids, human: reduced expansion (4 passages)
• In adult organoids, mouse: reduced expansion (<2 months)
22
46
Y-27632 (MW 247.34) inhibits ROCK (Rho-associated protein kinase) • In fetal organoids, mouse/human: cell proliferation dramatically decreased
• In embryonic organoids, mouse: reduced org. formation, none Pdx1+
55

54
A83-01 (MW 421.52)
inhibits TGFβ pathway
• In adult organoids, human: reduced expansion (from 20 to 10 passages) 22
Prostaglandin E2 (MW 352.47) • In adult organoids, human: reduced expansion (5 passages) 22
*) UniProtKB/Swiss-Prot: https://genecards.weizmann.ac.il; MW, molecular weight.  

Table 2.

Expansion of pancreatic organoids; cell origin, media composition, and results

Cell selection Basic Medium modifications* Cultivation time, No. of passages Doubling time [hrs] Reference no.
Mouse, hand-picked duct fragments Gastrin 10 months ~60 46
Mouse, Lgr5+ Gastrin, ROCK inhibitor >4 months -
Mouse, Ptf1A+ Gastrin 3–4 passages -
Mouse, EpCAM+ TSQ+ Gastrin, ROCK inhibitor 1 month, no proliferation -
Mouse, hand-picked single ducts Gastrin - - 49
Human, islet-depleted fragments Gastrin, A83-01; w/o Nicotinamide 10 passages 67 47
Human, ALDHhigh (from organoids) Gastrin, A83-01, ROCK inhibitor; w/o Nicotinamide - -
Human, hand-picked duct fragments WNT3A, Gastrin, A83-01, Prostaglandin E2 20 passages, 6 months - 22
Human, CD133+ w/o: B27, N-acetylcysteine >3 months - 53
Human, CD133+ Prostaglandin E2, HGF, Trichostatin A, CHIR99021, SB431542 >5 months 72 52
Human, islet-depleted fragments Gastrin, A83-01, Prostaglandin E2, Forskolin ~5 passages, 70 days 73 50
Mouse, Procr+ cells from islets B27, ITS, EGF, FGF2, heparin, endothelial cells; w/o:Basic Medium 20 passages, 6 months - 161
*Basic Medium: Advanced DMEM/F12, B27, N-acetylcysteine, EGF, Noggin, R-Spondin-1, Nicotinamide, FGF10  

2.4. Media for pancreatic organoid differentiation

Unlike the intestinal organoids, no universal medium has yet been developed for differentiation of pancreatic organoids toward beta cells. Several approaches were tested. The simplest one is the exclusion or reduction of growth factors that stimulate cell proliferation, such as EGF or R-Spondin-1.46,47,52,53,55 Nicotinamide, known to induce differentiation and maturation of fetal pancreatic endocrine cells in non-organoid culture,92 is commonly used in organoid culture.47,52,53,93 The genes required for differentiation of organoid stem cells are presumably inaccessible for the developmental transcription factors. The accessibility of such genes can be improved by chromatin state modulation, using small molecules inhibiting nuclear epigenetic modifiers, such as DNA methyltransferases.52 More direct approach employed in some pancreatic organoid studies is transdifferentiation in vitro by introducing the key transcription factors by viral49,53 or non-viral52 means. Detailed information about differentiation approaches are described in the chapter Organoids derived from Adult Pancreas.

3. Lessons from Embryogenesis and adult organ regeneration

Organoids derived either from embryonic or adult pancreatic tissue could potentially serve as a model system to accelerate the study of some aspects of pancreas and beta cell development. This is particularly important for research of human pancreas, given the limited accessibility. The designing of organoid culture systems draws from the knowledge obtained from studies of the development and regeneration of pancreas and other epithelial organs.

Pancreas develops from dorsal and ventral buds of the foregut endoderm in close contact with endothelium.94–97 The early cellular development as well as the late maintenance of the differentiated state are orchestrated by a hierarchical cascade of stage-specific combinations of transcription factors.98–100 The development of pancreas is divided into three major stages: a primary transition (E9.5–12.5), a secondary transition (E12.5-birth), and the early postnatal period until weaning.101 During the primary transition period, the pancreatic progenitors proliferate within thickening endoderm, which comes into a close proximity of endothelial cells of dorsal aortae.102 The endothelial cells initiate evagination of endoderm,94 which leads to tubulogenesis and branching. During this period, bipotent pancreatic progenitors form the stalk/trunk domains, which give rise to ductal and endocrine cells, while multipotent progenitors present in the tip domains generate endocrine, ductal, and acinar cells.103 Most of the endocrine cell differentiation occurs within the secondary transition period, hand in hand with the extensive exocrine differentiation. Within the secondary transition period, extensive exocrine and most of the endocrine cell differentiation occurs. Multipotent progenitors of the tip domains lose their multipotency and differentiate into acinar cells.99 In the original model of islet formation, the differentiating Ngn3+ endocrine progenitors, derived from bipotent trunk progenitors, undergo epithelial-mesenchymal transition allowing for cell delamination from the ducts, migration toward blood vessels and aggregation within newly formed islets.101 However, recent model omits both the epithelial-mesenchymal transition and cell migration. In the course of islet formation, the cell contacts between the precursors are maintained, bi-layered nascent ”peninsulas” are formed, where alpha cells first develop in the outer layer (E13.5), while beta cells gradually appear beneath them, attached to the epithelial cord producing differentiating cells (E14.5). This spatiotemporal collinearity gradually leads to the core-mantel architecture typical for mature mouse islets.104 Functionally, mouse beta cells mature only after birth, coincident with weaning.105 In the unique human islet architecture, all endocrine cells are attached to blood vessels, heterologous alpha–beta cell contacts are favored, and homologous beta–beta cell contacts are permitted.106 In the formation of human islets, a coalescence of few small ”peninsulas” was proposed.104

Since the 1880s, adult pancreatic cells were considered undividing, but the capacity of pancreas to regenerate remained tenable, due to the early observations of mitotic figures in pancreatic cells, after partial pancreatectomy.107 In humans, beta cells noticeably increase their numbers only in the first few years of life and during pregnancy.108,109 In 1993, Bonner-Weir proposed two pathways of pancreas regeneration after 90% pancreatectomy in mice, when observing a) the replication of preexisting endocrine and exocrine cells, and b) the proliferation and differentiation of ductal cells, forming new lobules, which contained new beta cells, reminiscent of embryonic development.110 Two decades later, the regeneration of adult beta cells remains unsettled. Some genetic cell tracing studies support the original hypothesis110 of beta cell neogenesis from adult ductal progenitors after injury inflicted by duct ligation111,112 or genetic cell targeting.113 Other fate mapping studies failed to find evidence for beta cell neogenesis from adult pancreatic progenitors.111,114–119 Additional genetic cell tracing studies combined with pancreatic duct injury in rodents convincingly demonstrated the replication of beta cells as the dominant regenerative mechanism during adult life120,121 even after partial pancreatectomy120 or targeted beta cell depletion.122 Significance of these complex experiments has been thoroughly reviewed elsewhere.107,123–131 Finally, transdifferentiation of alpha to beta cells was identified as a source of beta cell regeneration after near-total beta cell ablation in adult mice.132 After puberty, this capacity of alpha cells was not altered by age until senescence.133 In juvenile age, dedifferentiation of delta cells was observed, accompanied by subsequent proliferation and differentiation into beta cells after almost complete beta cell ablation.133

An unexpected heterogeneity of regeneration strategies was discovered among epithelial organs, ranging from the presence of multiple types of adult stem cells within adult tissues to the natural plasticity of differentiated cells, which de-differentiate into stem cells and in turn are capable of tissue regeneration.134 The existence of facultative adult progenitors was proposed in some tissues, including liver and pancreas. A picture is emerging, where the Lgr5+ crypt collumnar cells residing at the bottom of the intestinal crypt serve as the intestinal stem cells under normal conditions, while under other conditions, such as radiation injury, the radiation-resistant +4 cells replenish not only the differentiated intestinal cell population, but also lost Lgr5+ crypt collumnar stem cells.135 LGR5 was also used as a positive selection marker to generate adult pancreatic organoids.46

4. Organoids derived from embryonic and fetal pancreatic progenitors

Unlike in adult pancreas, the existence of pancreatic progenitors in the embryonic and fetal tissues is undoubted. The following three studies54,55,93 represent the first attempts to apply the principles of organoid culture in order to replicate the main features of pancreas development in vitro.

Greggio54 optimized culture conditions and achieved expansion and partial differentiation of pancreatic organoids. The organoids originated from single cells or small groups of multipotent pancreatic progenitors isolated from embryonic (E10.5) mice. The mice were genetically modified (e.g. Ngn3-EYFP, Pdx1-nGFP, Sox2-Cre x R26R-lacZ, Sox2-Cre x R26R-YFP, Neurog3 knockout) in order to visualize the lineage tracing with fluorescent or histochemistry labels, in time lapse experiments. The efficiency of organoid structure formation was found to depend on the number of Pdx1+ cells per cluster, giving rise to the organoid (100% efficiency was achieved when at least 12 Pdx1+ cells were present per cluster),54 suggesting the community effect.136 Within seven days, the largest organoids formed lobulated tubes with duct cells marked with mucin. The periphery of terminal buds was crowned with PTF1A+/SOX9+/PDX1+ cells, giving rise to acinar cells (15–20%). In the organoid core bipotent progenitor cells (HNF1B+) differentiated into polarized ductal cells with rare endocrine differentiation. The frequency of mono-hormonal C-peptide+ cells increased up to ~0.7% by omission of FGF1, which however was necessary in the first four days for the proliferation of Pdx1+ progenitor cells. Similarly, ROCK inhibitor was indispensable for the survival of progenitors expressing Pdx1. When the organoids were dissociated and transplanted into E13.5 pancreatic explants (a transplantation assay), the endocrine differentiation increased within 10 days up to 4%, indicating an incompleteness of the artificial niche provided by the in vitro culture conditions.54

Sugiyama93 described an alternative protocol for partial reconstitution of pancreas development, using spherical organoids. Transgenic mice (e.g. Sox9-eGFP/Ngn3-tdTomato, MIP-GFP) were used for cell selection and tracking. FACS-sorted fetal (E11.5) NGN3/SOX9+ multipotent progenitors (expressing also Pdx1, Hnf6, Tcf2 and Hes1, but not endocrine markers) were seeded at clonal density and cultured in Matrigel mixed with mesenchymal cells in PrEBM medium supplemented with FGF10, IGF1, Retinoic acid, insulin, and Transferrin. While the organoids expanded for three passages only, the progenitors here differentiated into multilayered spheres, with the inner layer of mucin+ duct-like cells, and the periphery with SOX9 cells. A subset of SOX9 cells expressed the endocrine markers, e.g. Ngn3, insulin, C-peptide, and glucagon. The endocrine cells on the outer surface were separated from the inner lining of mucin+ cells by another cell layer. The differentiation into glucose-responsive insulin-secreting cells was enhanced by a combination of Nicotinamide, physiological level of oxygen, reduced concentration of FGF10, and in the presence of mesenchymal cells. Uniquely, twofold increase in glucose-stimulated (3 vs. 20 mM) C-peptide secretion was observed in vitro.93

Bonfanti55 took advantage of transgenic mice (Pdx1-eGFP x Ins1-mRFP) to prospectively select and monitor fetal (E12 or E13) pancreatic progenitors in order to study the differentiation dynamics in organoids derived from a single cell.55 Unlike the previous group, the authors demonstrated a high efficiency of organoid formation from individual progenitor cells, however with only partial endocrine differentiation.

In the same paper, Bonfanti investigated the impact of EGF on human fetal pancreatic organoids with the conclusion that EGF potentiates the organoid expansion while suppressing the differentiation toward endocrine fate.55 Human progenitors were isolated after abortion 8–11 weeks post conception; mesenchymal cells were removed and the remaining epithelial tissue was digested, dissociated, mixed with Matrigel and cultured in Basic Medium supplemented with Gastrin and ROCK inhibitor. In the presence of EGF, the organoids expanded for at least 5 months, without changes in their cystic morphology. The organoid architecture contained ductal structures with cell polarization, as detected by MUC1 expression. In the absence of EGF, the organoids grew smaller, some assumed dense rather than cystic morphology, and the possible passage number dropped to ten. Significantly, a spontaneous differentiation toward endocrine fate (e.g. chromogranin A, insulin, glucagon, somatostatin) and acinar fate (e.g. PTF1A) was observed. The authors also observed a decreased proliferation rate after excluding ROCK inhibitor, FGF10, or R-Spondin-1. Their findings demonstrated a similar response between human and mouse fetal progenitors to the same environmental cues.55

5. Organoids derived from adult pancreas

Enzymatic digestion of adult pancreas releases endocrine islets, and non-endocrine fragments comprising the acinar, centro-acinar, and ductal components, which somewhat differ in the tissue density.137 Enrichment of the starting material with the prospective progenitor cells was achieved using techniques, such as tissue fragment separation on density gradient, hand-picking of duct fragments, and single cell sorting using putative markers of the proposed pancreatic stem/progenitor cells, as summarized in Table 2. The mouse and human organoid studies discussed below explored the utility of cell surface markers LGR5, CD133, and PROCR, as well as intracellularly expressed cell markers ALDH and PTF1A. Beta cell differentiation was induced by media composition and/or manipulating key transcription factors, as summarized in Table 3.

Table 3.

Differentiation of expanded organoids from adult murine and human pancreata

Cell source In vitro differentiation In vivo differentiation Maturation by histology C-peptide secretion Reference no.
Mouse, EpCAM+ TSQ CAG+ - Re-aggregated with embryonic cells, transplanted under kidney capsule 2.5% insulin+ (all mono-hormonal) In mice: detectable, response not tested 46
Mouse, hand-picked single ducts Lentiviral delivery: Pdx1, MafA, Ngn3-WT - 7% insulin+ - 49
Lentiviral delivery: Pdx1, MafA, Ngn3-phosphomutant - 28% insulin+ (33% coexpressed somatostatin) -
Plus medium modification: ROCK inhibitor; IWP-2, w/o R-Spondin-1, EGF for the last 2 days - 61% insulin+ (41% coexpressed somatostatin) -
Plus last 2 days the three genes turned off - 61% insulin+ (87% mono-hormonal) -
Human, islet-depleted fragments Nicotinamide containing medium Transplanted under kidney capsule in normoglycemic or hyperglycemic mice 1.5% insulin+ (mono-hormonal) In mice: detectable, not glucose-responsive 47
Human, CD133+ Adenoviral delivery: PDX1, MAFA, NGN3, PAX6; Medium: Retinoic acid; w/o R-spondin Transplanted under kidney capsule 7–11% insulin+ (all mono-hormonal) In vitro: not glucose-responsive; In mice: detectable* 53
Human, CD133+ mRNA delivery: NGN3; Medium: RepSox, PP2, ISX-9, GSK126, 5-aza-2´-deoxycytidin, Forskolin - 5% insulin+ (some coexpressed somatostatin) In vitro: not glucose-responsive 52
Mouse, Procr+ cells from islets B27, ITS, EGF, FGF2, heparin, endothelial cells Organoid transplanted under the kidney capsule were already differentiated in vitro: 30% mono-hormonal insulin+cells, 70% bi-hormonal insulin+ cells In vitro: glucose-responsive, In mice: glucose-responsive 161
*After 3 g/kg of glucose, after o/n starving, there was a trend to incresed C-peptide levels, but statistical significance is not reported.  

Azzarelli49 transdifferentiated mouse ductal organoids in vitro using lentiviral-mediated overexpression of transcription factors Pdx1, MafA, and Neurogenin3. The organoid culture was initiated from hand-picked ducts. Neurogenin3 was administered in two forms: the wild type and a more stable phospho-mutant. The best percentage of transdifferentiation into insulin positive cells (28% versus 7% in the wild type) was observed in organoid cells treated with the stable mutant of Neurogenin3. Significantly, the differentiation further increased up to 61% of insulin positive cells, when Wnt pathway was inhibited (addition of IWP-2, omission of R-Spondin-1) and EGF was removed for the last two days of culture. Moreover, when viral expression of all the transcription factors was turned off, the proportion of mono-hormonal insulin-producing cells increased. In spite of this success, the measurement of glucose responsiveness was inconclusive due to the small number of organoid cells.49

A potential selection marker for adult pancreatic progenitors is LGR5, a plasma membrane receptor for R-Spondin-1, involved in the signaling of canonical Wnt pathway.138 Its deletion is neonatally lethal.139 It was identified in stem cells of the small intestine, colon,83 and in long-lived cycling stem cells in hair follicle.140 Lgr5+ cells in the intestinal crypt were shown to generate all epithelial lineages over more than a year of follow up in vivo study.141 Additionally, liver injury induced the expression of Lgr5 on proposed facultative liver progenitors, as documented by their capacity to expand in vitro in a Wnt-pathway-dependent organoid culture.76 Huch46 stimulated the emergence prospective facultative progenitors in vivo by applying the partial duct ligation model in genetically modified adult mice (e.g. ECad-CFP, CAG-EGFP, Lgr5-LacZ). After pancreatic duct ligation, a population of Lgr5+ stem/progenitor cells appeared in the ducts. Huch46 concluded that adult Lgr5+ cells are bipotential progenitors capable of endocrine and ductal differentiation. In this study, hand-picked ductal fragments were embedded in Matrigel and grown in Basic Medium in Matrigel and grown in Basic Medium supplemented with Gastrin. Ductal fragments formed budding cyst-like organoids composed of duct-like progenitors (expressing Sox9, Pdx1, Muc1, Krt19). The complete medium allowed the organoids expansion for more than ten months, while the omission of EGF, Noggin, R-Spondin-1, FGF10, or Nicotinamide grossly reduced the number of possible passages. The capacity of FACS-sorted single Lgr5+ cells to form organoids was demonstrated with a colony-forming efficiency of 16%, that was in agreement with organoid studies of other digestive organs.75 The potential of the Lgr5+ pancreatic progenitors to differentiate into ductal and endocrine lineages was demonstrated in vivo using organoids derived from single epithelial non-endocrine cells (EpCAM+ TSQ). After 6 weeks of expansion, the organoids were dissociated, re-aggregated, and mixed with late embryonic pancreatic cells (E13.5). Additional stem cell niche signals were provided by transplanting this cell mixture under the kidney capsule of immunodeficient mice. Mainly, differentiated ductal (KRT19+) were observed. Histology also revealed 5% of endocrine cells (synaptophysin+), half of which were mono-hormonal insulin+ and C-peptide+. However, C-peptide in the plasma was not reported.46

Adult Ptf1a+ acinar cells in vivo were shown to regain aspects of embryonic multipotentiality under injury, and subsequent conversion into mature beta cells was revealed.142 PTF1A (Pancreas Transcription Factor 1a) is selectively expressed in pancreas, retina, spinal cord, brain, and enteric nervous system. It is indispensable in controlling the expansion of multipotent progenitor cells as well as the specification and maintenance of the acinar cells.143 Huch46 took advantage of the organoid culture in order to substantiate the hypothesis of the acinar cells as the adult pancreatic progenitors. The authors used PTF1A marker to select single cells for the organoid formation, but their expansion ceased four passages later.

Loomans47 identified in adult human pancreatic organoids an abundant cell population characterized by a high aldehyde dehydrogenase (ALDH) activity, and proposed it as a new marker for adult pancreatic progenitors. ALDH/RALDH (Aldehyde dehydrogenase) participates in All-trans Retinoic acid synthesis, which regulates gene expression by activating specific nuclear receptors during development in various tissues, including pancreas.144,145 In the organoid study by Loomans et al., the transcription profile of ALDH+ cells (CPA1, PDX1, MYC, and PTF1A) corresponded with multipotent embryonic progenitors described by Zhou.103 ALDHhigh cells constituted a quarter of the cells in the primary organoids, which were derived from fragments of islet-depleted pancreatic tissue and cultured in Matrigel and Basic Medium supplemented with Gastrin and TGFβ inhibitor (A83-01), with Nicotinamide omitted. ALDHhigh cells were predominantly localized at the tips of the budding organoids, which expressed mucin-1 at the luminal side and were maintained for at least 10 passages. ALDHhigh cells selected from the primary organoids formed secondary cyst-like colonies, which in turn also expanded, suggesting stemness of the original cells. No endocrine differentiation of the secondary colonies was reported. A small cell fraction (0.5%) in the primary organoids differentiated in vitro into insulin positive cells, when Nicotinamide containing differentiation medium was used.146 No glucagon positivity was reported. One month after the primary organoids were transplanted under the kidney capsule of either normoglycemic or hyperglycemic immunodeficient mice, 1.5% of organoids cells were insulin positive. Human C-peptide was detectable in mouse plasma, but was not glucose-responsive, indicating failed maturation in vivo. Glucagon was detected in mono-hormonal cells only.47 While ALDH+ cells were scarcely detectable in normal adult pancreas,147 they become abundant in regenerating conditions in human (patients with pancreatitis or T1D) as well as in mice (early postnatal, pregnancy),148 corroborating their facultative progenitor status. In this context, Rovira,147 by means of the centro-acinar/terminal ductal cells expressing ALDH1, published the first attempt to utilize the principles of mouse organoid culture in order to identify the putative adult pancreatic progenitor cell. Sorted ALDH positive cells were used to generate clonal pancreatospheres. The spheres contained cells co-expressing ALDH and SOX9, suggesting self-renewal capacity, which however was supported by only miniscule expansion (three passages). Interestingly, spontaneous C-peptide secretion occurred, but glucose responsiveness was tested at 0 vs. 11 mM glucose levels (the standard glucose testing levels are 3 vs. 20 mM), nor was it statistically evaluated.147

Another potentially useful selection marker for adult pancreatic progenitors is CD133 (AC133, Prominin-1), a plasma membrane protein with a large extracellular loop. It was identified in undifferentiated embryonic stem cells,149 and organ-committed stem cells,150 such as hematopoietic151 and neural152 stem cells. Histology of pancreas demonstrated the presence of CD133+ cells within adult ducts.153,154 Several groups utilized it for positive selection of the prospective bipotent or multipotent pancreatic progenitors, employing antibody-based FACS53,155–157 or MACS52,158 cell sorting.

Lee53 transdifferentiated human CD133+ cells derived pancreatic organoids into insulin-secreting cells, using ectopic overexpression of four principal islet transcription factors (PDX1, MAFA, Neurogenin3, and PAX6). The CD133+ cells, selected by FACS from islet-depleted pancreatic tissue, were positive for the ductal marker cytokeratin-19, and negative for acinar and endocrine markers. Single-layer organoids were expanded for up to 3 months in Basic Medium with the omission of B27 supplement. The transdifferentiation protocol involved adenoviral vectors expressing the transcription factors inspired by Zhou.159 After a few days of R-Spondin-1 withdrawal and supplementation with Retinoic acid, the organoids were cultured for subsequent two weeks in a differentiation medium, composition of which was less important than the timing. The differentiated spheres comprised 7–11% insulin positive cells, which secreted C-peptide into the medium, irrespective of the glucose level in the range 2–11 mM. A trend toward glucose-responsiveness (2.4×) was observed only when glucose was increased from starvation level (0.1 mM). However, C-peptide response to non-glucose secretagogues (KCl, sulphonylurea) was observed. After transplantation of transdifferentiated spheres under the kidney capsule of non-diabetic immunodeficient mice, human C-peptide was detectable in plasma for two weeks and a trend toward glucose responsiveness was observed (statistics for these functional observations was not provided).53

Koblas52 achieved reprogramming of human CD133+ organoid cells into insulin-producing cells by the means of non-viral non-integrative introduction of Neurogenin3 in a combination with small molecules altering signaling pathways and epigenetic state. CD133+ cells were immunomagnetically separated from islet-depleted pancreatic tissue and cultivated in Matrigel with Basic Medium replenished with HGF, Prostaglandin E2, Trichostatin A, CHIR99021, and ALK5 inhibitor (SB431542). Single-layer organoids were expanded for at least 5 months. Neurogenin3 was introduced in the form of synthetic mRNA. Differentiation medium contained RepSox, PP2, ISX-9, GSK126, 5-aza-2´-deoxycytidin, and Forskolin. After the differentiation, the organoids comprised ~40% chromogranin A positive cells, including 5% insulin positive cells. C-peptide was detected in the medium at low levels (1/103 of that produced by the same amount of islets), which failed to respond to glucose challenge, possibly due to insufficient expression of MAFA and PAX6. However, the secretion of C-peptide was KCl-responsive. A double-hormonal subpopulation co-expressing insulin and somatostatin was observed, further corroborating the immature character of insulin-producing cells. Glucagon positive cells were not observed.52 The non-integrative approach taken in this study avoids virus-induced inflammatory response and oncogenic transformation.160

The above discussed studies achieved various degrees of expansion of pancreatic organoids derived from different subpopulations of pancreatic duct cells. The maximum length of the growth of human organoids reached six months (20 passages), while ten months for mice. Cyst-like oval shape organoids generally comprised a single layer of predominantly undifferentiated cells oriented around a central lumen (rather than elongated ducts),22,46–53 as exemplified in Figure 1. The culture conditions and the results of the presented studies are summarized in Table 1–3.

Wang et al.161 uniquely searched for the adult endocrine progenitors within the pancreatic islets. The authors identified a novel population of Procr+ progenitors within adult mouse pancreatic islets, using single-cell RNA sequencing (scRNA-seq), and demonstrated their capacity to ameliorate hyperglycemia in diabetic mice. PROCR (EPCR, CD201) is an endothelial receptor for protein C, which was previously identified on hematopoietic stem cells,162 cultured cord blood cells,163 blood vascular endothelial stem cells,164 and breast cancer cells.165 More than 7000 individual cells isolated from islet-enriched preparation were analyzed by scRNA-seq and mapped to clusters representing ten known cell types (alpha, beta, delta, PP, duct, acinar, endothelial, immune, mesenchymal, and stellate cells), and an additional cluster of previously unknown cells was identified. Unique signature genes in this cluster included Wnt pathway agonist gene R-spondin-1 and Wnt target gene Procr, suggesting stem cell character. These Procr+ islet progenitors had the capability to form expanding organoids, to differentiate into four endocrine cell types including insulin-producing beta-like cells, which ameliorated diabetes after transplantation. In Procr-mGFP-2A-LacZ mouse model, Procr+ cells were identified from all pancreas exclusively in the islets, each containing only few such cells. The lineage tracing in adult Procr-CreERT2; RosaConfetti mice several months after tamoxifen pulse revealed clones comprising approximately seven cells. In 70% of the clones, all endocrine cell types were present, while 30% of the clones contained beta cells only. One cell in each clonal population was hormone negative, which was the proposed progenitor cell. When FACS-isolated Procr+ cells were plated at a clonal density in serum-free medium supplemented with B27, ITS, EGF, FGF2, and heparin, one out of 15 cells formed a colony (Procr cells failed to form colonies). However, Procr+ cells could not be maintained for more than 7 days. Co-culture with endothelial cells was revealed as the key factor allowing long-term culture (more than 20 passages) of Procr+ derived organoids. At 15th passage, the organoids contained all hormone+ cell types, and secreted insulin and C-peptide (approximately tenfold and fivefold, respectively) into the medium in the glucose-regulated manner. When 1000 organoids were transplanted under the kidney capsule of diabetic mice, glucose level was ameliorated below 10 mM, which was similar to the control group of diabetic mice transplanted with 300 natural islets. One month after transplantation, intraperitoneal glucose tolerance test (IPGTT) demonstrated the improvement of glucose tolerance to a similar degree as 300 islets transplanted to a control group. Graftectomy after 125 days led to an abrupt blood glucose increase to 25 mM. The clustering of scRNA-seq data from explanted graft revealed alpha, beta, delta, and PP cells matching well with the primary islet cell clusters.161

6. Potential clinical application of adult pancreatic organoids

The first and the only reported attempt to develop a large-scale GMP-level manufacturing procedure for the expansion of organoids for clinical purposes comes from Dossena et al.50 The enzymatic digestion was replaced by a mechanical one, thus avoiding manual duct picking. The organoids were expanded using BME2 matrix and Basic Medium supplemented with Gastrin, Prostaglandin E2, Forskolin, and A83-01. The cystic morphology and the expression profile (positivity for SOX9, PDX1, and MUC) typical for adult ductal organoids were observed. The authors were able to expand the islet-depleted pancreatic tissue, derived from a single cadaver donor, up to 250 × 106 pancreatic organoids with the prospect of future differentiation into insulin-secreting cells.50

The total number of organoid cells reached approximately 250 × 109 cells.50 So far, the best published human differentiation rate achieved in vitro by clinically amenable means reached insulin positivity in 5% of organoid cells.52 If these two approaches were combined, one could hope for 13 × 109 of cells, which however would not yet be functional (steady secretion of 1/103 of the amount of insulin secreted by isolated islets).52 Drawing from the experience with clinical islet transplantation, the minimum number of beta cells to achieve insulin independence is estimated to be 109 of functional beta cells per recipient, given the estimated 1140 beta cells per islet equivalent.166 Some authors demonstrated up to 60% differentiation rate into insulin positive cells, in the case of mouse organoids.49 While the means were unsuitable for clinical practice, these results suggest that a highly efficient though partial transdifferentiation is possible.

The safety of pancreatic organoids derived from adult stem cells for clinical application has not yet been properly evaluated. Only a few pilot studies performed transplantation of expanded or differentiated organoids into the mice without detection of any malignant transformations after maximal observation time for 3 months.48,53

7. Perspective

The existence of adult pancreatic progenitors or facultative progenitors has been contested for decades. Historically, three major assays have been devised to prove the existence of a stem cell in a given tissue: lineage tracing in vivo, clonogenic growth in vitro, and cellular transplantation.124 Here, we reviewed the attempts to resolve the issue by the means of pancreatic organoid culture, which was applied to all four histological compartments of pancreas, comprising the acinar, and centro-acinar/terminal ductal, ductal, and islet cells.

The acinar and centro-acinar/terminal ductal cells failed to expand in long-term organoid culture, suggesting the absence of the stem cells. Although the ductal cells expanded in the long term and differentiated into insulin-producing cells, suggesting the presence of stem/progenitor cells, the differentiated cells remained unresponsive to glucose. Unlike the insulin-secreting cells derived from pluripotent stem cells,167 the insulin positive cells derived from the organoid culture failed to mature in situ after transplantation under the kidney capsule. These findings need to be interpreted with caution, because under the current culture conditions even the embryonic/fetal stem-cells-derived organoids failed to completely differentiate unless co-cultured with mesenchymal cells. One explanation for failed differentiation can be the epigenetic state of the organoid cells preventing sufficient induction of key transcription factors involved in the differentiation process. The beta cell apparatus for metabolically regulated insulin release is quite complex,168 and beta cells naturally mature only after weaning from fat-based to carbohydrate-based food.105 This suggests that current adult non-islet derived organoid cultures might have failed to provide the adequate developmental and maturation cues.

Only Procr+ cells isolated from adult mouse islets constituted organoid culture capable of the long-term expansion as well as the differentiation into glucose-responsive insulin-producing cells, which were uniquely able to ameliorate blood glucose in diabetic mice after transplantation.161 Repeated supplementation of fresh endothelial cells to the organoid co-culture was necessary. The essential role of endothelial cells not only for embryogenesis, but also for proper beta cell function in adulthood was previously described,106,169,170 with the dependence of beta cell on signaling cues from basal membrane, where beta cell cannot synthesize.

Clinical application of adult pancreatic progenitors for stem cell therapy of diabetes is challenged not only by doubtful existence of the adult pancreatic bipotential progenitors, but also by the inaccessibility of the proper pancreatic niche. The unparalleled success of clinical transplantation of hematopoietic stem cells was facilitated by the accessibility of the appropriate niche to the natural adult stem cells. Experimentally, hyperglycemia was indeed ameliorated in mice by transdifferentiation of pancreatic acinar cells in situ, taking advantage of the natural pancreatic niche.159 While current evidence supports other sources of beta cell regeneration,131 the existence of adult bipotential pancreatic progenitors has not yet been abandoned.129,130 The co-expression of Pdx1, Sox9, Nkx6.1, and Hnf6 (Onecut-1) is characteristic for the bipotent endocrine/duct progenitors during embryogenesis, and the same transcription factors are also detected within the pancreatic organoids.46,47,52

Although a large-scale expansion of human adult pancreatic organoids was recently developed, partially at GMP level for putative clinical application, more research would be necessary to develop truly efficient and safe approach. Another drawback of the adult pancreas as the source for cell-based therapy seems to be rather complicated access to this potential cell source, the inherent variability among different donors, and technical difficulties associated with the up-scaling of organoid culture.

Nevertheless, pancreatic organoid culture represents a potentially helpful tool bringing otherwise inaccessible and complex organ in a dish. Hypothetically, a pancreatic biopsy could be used for this purpose, similar to the intestinal organoids from cystic fibrosis patients.16 Diabetes-associated genetic variants could also be introduced into organoids via CRISPR/Cas9 approach. At present, however, the identification of prospective pancreatic progenitor appears to be the most attractive goal for the use of pancreatic organoid system.

At the moment, adult pancreatic organoids are far from becoming a cell source for clinical application, but they remain an invaluable research tool for the investigation of beta cell development and regeneration, which makes more refinement effort worthwhile. It is intriguing whether the recently discovered Procr+ islet progenitor cells in the adult mice exist also in human islets, constituting a novel source of potentially curative beta cell replacement therapy or in situ regeneration. The lessons learned from the differentiation approach so successful in Procr+ islet progenitors might be applicable for the putative extra-islet pancreatic progenitors, thus moving the ten years old field forward.

Abbreviations

ALDH Aldehyde dehydrogenase
ALK5 Activin receptor-like kinase 5
BME Basement membrane extract
CD133 Prominin 1
ECAD E-cadherin
ECM Extracellular matrix
EGF Epidermal growth factor
ENR EGF+Noggin+R-Spondin-1
EpCAM Epithelial cell adhesion molecule
ESCs Embryonic stem cells
EZH2 Enhancer of zeste homolog 2
FACS Flourescent activated cell sorting
FGF1 Fibroblast growth factor 1
FGF10 Fibroblast growth factor 10
GMP Good manufacturing practices
HGF Hepatocyte growth factor
HNF1B Hepatocyte nuclear factor-1 beta
HNF6 Hepatocyte nuclear factor 6, Onecut-1
CHGA Chromogranin A
CHIR99021 Inhibitor of glycogen synthase kinase 3ß
IGF-1 Insulin-like growth factor 1
IPGTT Intraperitoneal glucose tolerance test
iPSC Induced pluripotent stem cells
ISX-9 Izoxazole 9
ITS Insulin-Transferrin-Selenium
IWP-2 Inhibitor of WNT Production-2
Ki67 Proliferation marker
KRT19 Cytokeratine 19
LGR5 Leucine-rich-repeat-containing G-protein-coupled receptor 5
MACS Magnetic-activated cell sorting
MAFA C-maf musculoaponeurotic fibrosarcoma oncogene homolog A
MUC1 Mucin 1
NGN3 Neurogenin3
NKX6.1 Nirenberg and Kim homeobox 6.1
PAX6 Paired box gene 6
PDX1 Pancreatic and duodenal homeobox 1
PEG Poly(ethylene glycol)
PP2 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo[3,4-d]pyrimidine
PrEBM Prostate epithelial cell growth basal medium
Procr Protein C receptor
PTF1A Pancreas associated transcription factor 1A
RepSox Inhibitor of TGFβ type I activin like kinase receptor (ALK5)
RFD Tripeptide Arg-Gly-Asp
R-Spondin-1 Roof plate-specific Spondin-1
SC Stem cell
scRNA-seq Single-cell RNA sequencing
SOX9 SRY-Box Transcription Factor 9
TGFβ Transforming growth factor β superfamily
TSQ Fluorescent chelator for Zn2+ ionts (6-methoxy-8-p-toluenesulfonamidoquilone)
VEGF Vascular endothelial growth factor (A)
Wnt Wingless-INT-#x003B2;-catenin signaling pathway
WNT3A Wnt family member 3A
Y-27632 ROCK inhibitor

Acknowledgments

The authors thank Ivan Leontovyc for reading the manuscript.

This work was supported by the Czech Science Foundation (GACR) under Grant No. 19-07661S.

Funding Statement

This work was supported by the Grantová Agentura České Republiky [19-07661S].

Disclosure statement

No potential conflict of interest was reported by the authors.

References

  • 1.Lancaster MA, Knoblich JA.. Organogenesis in a dish: modeling development and disease using organoid technologies. Science. 2014;345(6194):1247125. doi: 10.1126/science.1247125. [DOI] [PubMed] [Google Scholar]
  • 2.Muthuswamy SK. Bringing together the organoid field: from early beginnings to the road ahead. Development. 2017;144(6):963–967. doi: 10.1242/dev.144444. [DOI] [PubMed] [Google Scholar]
  • 3.Huch M, Koo B-K. Modeling mouse and human development using organoid cultures. Development. 2015;142(18):3113–3125. doi: 10.1242/dev.118570. [DOI] [PubMed] [Google Scholar]
  • 4.Dutta D, Heo I, Clevers H. Disease modeling in stem cell-derived 3D organoid systems. Trends Mol Med. 2017;23(5):393–410. doi: 10.1016/j.molmed.2017.02.007. [DOI] [PubMed] [Google Scholar]
  • 5.Shamir ER, Ewald AJ. Three-dimensional organotypic culture: experimental models of mammalian biology and disease. Nat Rev Mol Cell Bio. 2014;15(10):647–664. doi: 10.1038/nrm3873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.McCauley HA, Wells JM. Pluripotent stem cell-derived organoids: using principles of developmental biology to grow human tissues in a dish. Development. 2017;144(6):958–962. doi: 10.1242/dev.140731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Fatehullah A, Tan SH, Barker N. Organoids as an in vitro model of human development and disease. Nat Cell Biol. 2016;18(3):246–254. doi: 10.1038/ncb3312. [DOI] [PubMed] [Google Scholar]
  • 8.Simian M, Bissell MJ. Organoids: a historical perspective of thinking in three dimensions. J Cell Biol. 2017;216(1):31–40. doi: 10.1083/jcb.201610056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Yin X, Farin HF, Van Es JH, Clevers H, Langer R, Karp JM. Niche-independent high-purity cultures of Lgr5+ intestinal stem cells and their progeny. Nat Methods. 2014;11(1):106–112. doi: 10.1038/nmeth.2737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Basak O, Beumer J, Wiebrands K, Seno H, van Oudenaarden A, Clevers H. Induced quiescence of Lgr5+ stem cells in intestinal organoids enables differentiation of hormone-producing enteroendocrine cells. Cell Stem Cell. 2017;20(2):177–190.e4. doi: 10.1016/j.stem.2016.11.001. [DOI] [PubMed] [Google Scholar]
  • 11.Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells. 1978;4:7–25. [PubMed] [Google Scholar]
  • 12.Smith E, Cochrane WJ. Cystic organoid teratoma; report of a case. Can Med Assoc J. 1946;55:151. [PubMed] [Google Scholar]
  • 13.Kim Y, Kim H, Ko UH, Oh Y, Lim A, Sohn J-W, Shin JH, Kim H, Han Y-M. Islet-like organoids derived from human pluripotent stem cells efficiently function in the glucose responsiveness in vitro and in vivo. Sci Rep-uk. 2016;6(1):35145. doi: 10.1038/srep35145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Lancaster MA, Huch M. Disease modelling in human organoids. Dis Model Mech. 2019;12(7):dmm039347. doi: 10.1242/dmm.039347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kim J, Koo B-K, Knoblich JA. Human organoids: model systems for human biology and medicine. Nat Rev Mol Cell Bio. 2020;21(10):571–584. doi: 10.1038/s41580-020-0259-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Dekkers JF, Berkers G, Kruisselbrink E, Vonk A, de Jonge HR, Janssens HM, Bronsveld I, van de Graaf EA, Nieuwenhuis EES, Houwen RHJ, et al. Characterizing responses to CFTR-modulating drugs using rectal organoids derived from subjects with cystic fibrosis. Sci Transl Med. 2016;8(344):344ra84–344ra84. doi: 10.1126/scitranslmed.aad8278. [DOI] [PubMed] [Google Scholar]
  • 17.Tiriac H, Plenker D, Baker LA, Tuveson DA. Organoid models for translational pancreatic cancer research. Curr Opin Genet Dev. 2019;54:7–11. doi: 10.1016/j.gde.2019.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Schlaermann P, Toelle B, Berger H, Schmidt SC, Glanemann M, Ordemann J, Bartfeld S, Mollenkopf HJ, Meyer TF. A novel human gastric primary cell culture system for modelling Helicobacter pylori infection in vitro. Gut. 2016;65(2):202. doi: 10.1136/gutjnl-2014-307949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.McCracken KW, Catá EM, Crawford CM, Sinagoga KL, Schumacher M, Rockich BE, Tsai Y-H, Mayhew CN, Spence JR, Zavros Y, et al. Modelling human development and disease in pluripotent stem-cell-derived gastric organoids. Nature. 2014;516(7531):400–404. doi: 10.1038/nature13863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Murrow LM, Weber RJ, Gartner ZJ. Dissecting the stem cell niche with organoid models: an engineering-based approach. Development. 2017;144(6):998–1007. doi: 10.1242/dev.140905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Baker LA, Tiriac H, Clevers H, Tuveson DA. Modeling pancreatic cancer with organoids. trends in cancer. 2016;2(4):176–190. doi: 10.1016/j.trecan.2016.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Boj SF, Hwang C-I, Baker LA, Chio IIC, Engle DD, Corbo V, Jager M, Ponz-Sarvise M, Tiriac H, Spector MS, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015;160(1–2):324–338. doi: 10.1016/j.cell.2014.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Grapin-Botton A. Three-dimensional pancreas organogenesis models. Diabetes Obes Metabolism. 2016;18(S1):33–40. doi: 10.1111/dom.12720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Greggio C, Franceschi FD, Grapin-Botton A. Concise reviews: in vitro-produced pancreas organogenesis models in three dimensions: self-organization from few stem cells or progenitors. Stem Cells. 2015;33(1):8–14. doi: 10.1002/stem.1828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Balak JRA, Juksar J, Carlotti F, Nigro AL, de Koning EJP. Organoids from the human fetal and adult pancreas. Curr Diabetes Rep. 2019;19(12):160. doi: 10.1007/s11892-019-1261-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hohwieler M, Illing A, Hermann PC, Mayer T, Stockmann M, Perkhofer L, Eiseler T, Antony JS, Müller M, Renz S, et al. Human pluripotent stem cell-derived acinar/ductal organoids generate human pancreas upon orthotopic transplantation and allow disease modelling. Gut. 2017;66(3):473. doi: 10.1136/gutjnl-2016-312423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hou S, Tiriac H, Sridharan BP, Scampavia L, Madoux F, Seldin J, Souza GR, Watson D, Tuveson D, Spicer TP. Advanced development of primary pancreatic organoid tumor models for high-throughput phenotypic drug screening. Slas Discov. 2018;23:574–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Walsh AJ, Castellanos JA, Nagathihalli NS, Merchant NB, Skala MC. Optical imaging of drug-induced metabolism changes in murine and human pancreatic cancer organoids reveals heterogeneous drug response. Pancreas. 2016;45(6):863–869. doi: 10.1097/MPA.0000000000000543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Tsakmaki A, Pedro PF, Bewick GA. Diabetes through a 3D lens: organoid models. Diabetologia. 2020;63(6):1093–1102. doi: 10.1007/s00125-020-05126-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Tauschmann M, Hovorka R. Technology in the management of type 1 diabetes mellitus — current status and future prospects. Nat Rev Endocrinol. 2018;14(8):464–475. doi: 10.1038/s41574-018-0044-y. [DOI] [PubMed] [Google Scholar]
  • 31.Beck RW, Bergenstal RM, Laffel LM, Pickup JC. Advances in technology for management of type 1 diabetes. Lancet. 2019;394(10205):1265–1273. doi: 10.1016/S0140-6736(19)31142-0. [DOI] [PubMed] [Google Scholar]
  • 32.Gruessner AC, Gruessner RWG. Pancreas transplantation of US and non-US cases from 2005 to 2014 as reported to the United Network for Organ Sharing (UNOS) and the International Pancreas Transplant Registry (IPTR). Rev Diabet Stud. 2016;13:e2016002. doi: 10.1900/RDS.2016.13.e2016002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Collaborative Islet Transplant Registry (CITR) Coordinating Center, CITR 10th Annual Report, Rockville (MD): 2017. Jan 6 [Cited Jun 14, 2021]. Available from:https://citregistry.org/content/citr-10th-annual-report. [Google Scholar]
  • 34.Soria B, Roche E, Berna G, Leon-Quinto T, Reig JA, Martin F. Insulin-secreting cells derived from embryonic stem cells normalize glycemia in streptozotocin-induced diabetic mice. Diabetes. 2000;49(2):157–162. doi: 10.2337/diabetes.49.2.157. [DOI] [PubMed] [Google Scholar]
  • 35.Dayem AA, Lee SB, Kim K, Lim KM, Jeon T, Cho S-G. Recent advances in organoid culture for insulin production and diabetes therapy: methods and challenges. Bmb Rep. 2019;52(5):295–303. doi: 10.5483/BMBRep.2019.52.5.089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Shahjalal H, Dayem AA, Lim KM, Jeon T, Cho S-G. Generation of pancreatic β cells for treatment of diabetes: advances and challenges. Stem Cell Res Ther. 2018;9(1):355. doi: 10.1186/s13287-018-1099-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Rezania A, Bruin JE, Arora P, Rubin A, Batushansky I, Asadi A, O’Dwyer S, Quiskamp N, Mojibian M, Albrecht T, et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol. 2014;32(11):1121–1133. doi: 10.1038/nbt.3033. [DOI] [PubMed] [Google Scholar]
  • 38.Pagliuca FW, Millman JR, Gürtler M, Segel M, Van Dervort A, Ryu JH, Peterson QP, Greiner D, Melton DA. Generation of functional human pancreatic beta cells in vitro. Cell. 2014;159(2):428–439. doi: 10.1016/j.cell.2014.09.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Fujikawa T, Oh S-H, Pi L, Hatch HM, Shupe T, Petersen BE. Teratoma formation leads to failure of treatment for type I diabetes using embryonic stem cell-derived insulin-producing cells. Am J Pathology. 2005;166(6):1781–1791. doi: 10.1016/S0002-9440(10)62488-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Rebuzzini P, Zuccotti M, Redi CA, Garagna S. Achilles’ heel of pluripotent stem cells: genetic, genomic and epigenetic variations during prolonged culture. Cell Mol Life Sci. 2016;73(13):2453–2466. doi: 10.1007/s00018-016-2171-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Nguyen HT, Geens M, Spits C. Genetic and epigenetic instability in human pluripotent stem cells. Hum Reprod Update. 2013;19(2):187–205. doi: 10.1093/humupd/dms048. [DOI] [PubMed] [Google Scholar]
  • 42.Ben-David U, Gan Q-F, Golan-Lev T, Arora P, Yanuka O, Oren YS, Leikin-Frenkel A, Graf M, Garippa R, Boehringer M, et al. Selective elimination of human pluripotent stem cells by an oleate synthesis inhibitor discovered in a high-throughput screen. Cell Stem Cell. 2013;12(2):167–179. doi: 10.1016/j.stem.2012.11.015. [DOI] [PubMed] [Google Scholar]
  • 43.Odorico J, Markmann J, Melton D, Greenstein J, Hwa A, Nostro C, Rezania A, Oberholzer J, Pipeleers D, Yang L, et al. Report of the key opinion leaders meeting on stem cell-derived beta cells. Transplantation. 2018. Apr 30;102(8):1223–1229. doi: 10.1097/TP.0000000000002217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Cade H Top companies developing cell therapy treatments for diabetes. BioInformant. Standford (VA): 2019. Jun 11 [Cited Feb 15, 2021]; Available from: https://bioinformant.com/stem-cells-for-diabetes [Google Scholar]
  • 45.Baharvand H, Totonchi M, Taei A, Seifinejad A, Aghdami N, Salekdeh GH. Human-induced pluripotent stem cells: derivation, propagation, and freezing in serum- and feeder layer-free culture conditions. Methods Mol Biol. 2010;584:425–443. [DOI] [PubMed] [Google Scholar]
  • 46.Huch M, Bonfanti P, Boj SF, Sato T, Loomans CJM, van de Wetering M, Sojoodi M, Li VSW, Schuijers J, Gracanin A, et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. Embo J. 2013;32(20):2708–2721. doi: 10.1038/emboj.2013.204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Loomans CJM, Giuliani NW, Balak J, Ringnalda F, van Gurp L, Huch M, Boj SF, Sato T, Kester L, De S Lopes SMC, et al. Expansion of adult human pancreatic tissue yields organoids harboring progenitor cells with endocrine differentiation potential. Stem Cell Rep. 2018;10(3):712–724. doi: 10.1016/j.stemcr.2018.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Georgakopoulos N, Prior N, Angres B, Mastrogiovanni G, Cagan A, Harrison D, Hindley CJ, Arnes-Benito R, Liau -S-S, Curd A, et al. Long-term expansion, genomic stability and in vivo safety of adult human pancreas organoids. Bmc Dev Biol. 2020;20(1):4. doi: 10.1186/s12861-020-0209-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Azzarelli R, Rulands S, Nestorowa S, Davies J, Campinoti S, Gillotin S, Bonfanti P, Göttgens B, Huch M, Simons B, et al. Neurogenin3 phosphorylation controls reprogramming efficiency of pancreatic ductal organoids into endocrine cells. Sci Rep-uk. 2018;8(1):15374. doi: 10.1038/s41598-018-33838-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Dossena M, Piras R, Cherubini A, Barilani M, Dugnani E, Salanitro F, Moreth T, Pampaloni F, Piemonti L, Lazzari L. Standardized GMP-compliant scalable production of human pancreas organoids. Stem Cell Res Ther. 2020;11(1):94. doi: 10.1186/s13287-020-1585-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Dorrell C, Tarlow B, Wang Y, Canaday PS, Haft A, Schug J, Streeter PR, Finegold MJ, Shenje LT, Kaestner KH, et al. The organoid-initiating cells in mouse pancreas and liver are phenotypically and functionally similar. Stem Cell Res. 2014;13(2):275–283. doi: 10.1016/j.scr.2014.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Koblas T, Leontovyc I, Loukotová S, Saudek F. Reprogramming of human pancreatic organoid cells into insulin-producing β-like cells by small molecules and in vitro transcribed modified mRNA encoding neurogenin 3 transcription factor. Folia Biol-Prague. 2019;65:109–123. [DOI] [PubMed] [Google Scholar]
  • 53.Lee J, Sugiyama T, Liu Y, Wang J, Gu X, Lei J, Markmann JF, Miyazaki S, Miyazaki J, Szot GL, et al. Expansion and conversion of human pancreatic ductal cells into insulin-secreting endocrine cells. Elife. 2013;2:e00940. doi: 10.7554/eLife.00940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Greggio C, Franceschi FD, Figueiredo-Larsen M, Gobaa S, Ranga A, Semb H, Lutolf M, Grapin-Botton A. Artificial three-dimensional niches deconstruct pancreas development in vitro. Development. 2013;140(21):4452–4462. doi: 10.1242/dev.096628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Bonfanti P, Nobecourt E, Oshima M, Albagli-Curiel O, Laurysens V, Stangé G, Sojoodi M, Heremans Y, Heimberg H, Scharfmann R. Ex vivo expansion and differentiation of human and mouse fetal pancreatic progenitors are modulated by epidermal growth factor. Stem Cells Dev. 2015;24(15):1766–1778. doi: 10.1089/scd.2014.0550. [DOI] [PubMed] [Google Scholar]
  • 56.Gray RS, Roszko I, Solnica-Krezel L. Planar cell polarity: coordinating morphogenetic cell behaviors with embryonic polarity. Dev Cell. 2011;21(1):120–133. doi: 10.1016/j.devcel.2011.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Cortijo C, Gouzi M, Tissir F, Grapin-Botton A. Planar cell polarity controls pancreatic beta cell differentiation and glucose homeostasis. Cell Rep. 2012;2(6):1593–1606. doi: 10.1016/j.celrep.2012.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bellas E, Chen CS. Forms, forces, and stem cell fate. Curr Opin Cell Biol. 2014;31:92–97. doi: 10.1016/j.ceb.2014.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Mamidi A, Prawiro C, Seymour PA, de Lichtenberg KH, Jackson A, Serup P, Semb H. Mechanosignalling via integrins directs fate decisions of pancreatic progenitors. Nature. 2018;564(7734):114–118. doi: 10.1038/s41586-018-0762-2. [DOI] [PubMed] [Google Scholar]
  • 60.Armstrong PB. Cell sorting out: the self-assembly of tissues in vitro. Crit Rev Biochem Mol. 1989;24(2):119–149. doi: 10.3109/10409238909086396. [DOI] [PubMed] [Google Scholar]
  • 61.Baker BM, Chen CS. Deconstructing the third dimension – how 3D culture microenvironments alter cellular cues. J Cell Sci. 2012;125:3015–3024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Duval K, Grover H, Han L-H, Mou Y, Pegoraro AF, Fredberg J, Chen Z. Modeling physiological events in 2D vs. 3D cell culture. Physiology. 2017;32(4):266–277. doi: 10.1152/physiol.00036.2016. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
  • 63.Holloway EM, Capeling MM, Spence JR. Biologically inspired approaches to enhance human organoid complexity. Development. 2019;146(8):dev166173. doi: 10.1242/dev.166173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Xu C, Inokuma MS, Denham J, Golds K, Kundu P, Gold JD, Carpenter MK. Feeder-free growth of undifferentiated human embryonic stem cells. Nat Biotechnol. 2001;19(10):971–974. doi: 10.1038/nbt1001-971. [DOI] [PubMed] [Google Scholar]
  • 65.Kleinman HK, McGarvey ML, Hassell JR, Star VL, Cannon FB, Laurie GW, Martin GR. Basement membrane complexes with biological activity. Biochemistry-US. 1986;25(2):312–318. doi: 10.1021/bi00350a005. [DOI] [PubMed] [Google Scholar]
  • 66.Li ML, Aggeler J, Farson DA, Hatier C, Hassell J, Bissell MJ. Influence of a reconstituted basement membrane and its components on casein gene expression and secretion in mouse mammary epithelial cells. Proc National Acad Sci. 1987;84(1):136–140. doi: 10.1073/pnas.84.1.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hughes CS, Postovit LM, Lajoie GA. Matrigel: a complex protein mixture required for optimal growth of cell culture. Proteomics. 2010;10(9):1886–1890. doi: 10.1002/pmic.200900758. [DOI] [PubMed] [Google Scholar]
  • 68.Benton G, Arnaoutova I, George J, Kleinman HK, Koblinski J. Matrigel: from discovery and ECM mimicry to assays and models for cancer research. Adv Drug Deliver Rev. 2014;79:3–18. doi: 10.1016/j.addr.2014.06.005. [DOI] [PubMed] [Google Scholar]
  • 69.Cruz-Acuña R, Quirós M, Farkas AE, Dedhia PH, Huang S, Siuda D, García-Hernández V, Miller AJ, Spence JR, Nusrat A, et al. Synthetic hydrogels for human intestinal organoid generation and colonic wound repair. Nat Cell Biol. 2017;19(11):1326–1335. doi: 10.1038/ncb3632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Broguiere N, Isenmann L, Hirt C, Ringel T, Placzek S, Cavalli E, Ringnalda F, Villiger L, Züllig R, Lehmann R, et al. Growth of epithelial organoids in a defined hydrogel. Adv Mater. 2018;30(43):1801621. doi: 10.1002/adma.201801621. [DOI] [PubMed] [Google Scholar]
  • 71.Ye S, Boeter JWB, Mihajlovic M, Steenbeek FG, Wolferen ME, Oosterhoff LA, Marsee A, Caiazzo M, Laan LJW, Penning LC, et al. A chemically defined hydrogel for human liver organoid culture. Adv Funct Mater. 2020;30(48):2000893. doi: 10.1002/adfm.202000893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Aisenbrey EA, Murphy WL. Synthetic alternatives to Matrigel. Nat Rev Mater. 2020;5(7):539–551. doi: 10.1038/s41578-020-0199-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, Van Es JH, Abo A, Kujala P, Peters PJ, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459(7244):262–265. doi: 10.1038/nature07935. [DOI] [PubMed] [Google Scholar]
  • 74.Sato T, Stange DE, Ferrante M, Vries RGJ, Van Es JH, Van Den Brink S, van Houdt WJ, Pronk A, Van Gorp J, Siersema PD, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and barrett’s epithelium. Gastroenterology. 2011;141(5):1762–1772. doi: 10.1053/j.gastro.2011.07.050. [DOI] [PubMed] [Google Scholar]
  • 75.Barker N, Huch M, Kujala P, van de Wetering M, Snippert HJ, Van Es JH, Sato T, Stange DE, Begthel H, van den Born M, et al. Lgr5+ve stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell. 2010;6(1):25–36. doi: 10.1016/j.stem.2009.11.013. [DOI] [PubMed] [Google Scholar]
  • 76.Huch M, Dorrell C, Boj SF, Van Es JH, Li VSW, van de Wetering M, Sato T, Hamer K, Sasaki N, Finegold MJ, et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature. 2013;494(7436):247–250. doi: 10.1038/nature11826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cras-Meneur C, Elghazi L, Czernichow P, Scharfmann R. Epidermal growth factor increases undifferentiated pancreatic embryonic cells in vitro: a balance between proliferation and differentiation. Diabetes. 2001;50(7):1571–1579. doi: 10.2337/diabetes.50.7.1571. [DOI] [PubMed] [Google Scholar]
  • 78.Wang RN, Green J, Wang Z, Deng Y, Qiao M, Peabody M, Zhang Q, Ye J, Yan Z, Denduluri S, et al. Bone Morphogenetic Protein (BMP) signaling in development and human diseases. Genes Dis. 2014;1:87–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Ahnfelt-Rønne J, Ravassard P, Pardanaud-Glavieux C, Scharfmann R, Serup P. Mesenchymal bone morphogenetic protein signaling is required for normal pancreas development. Diabetes. 2010;59(8):1948–1956. doi: 10.2337/db09-1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Barker N, Clevers H. Leucine-Rich repeat-containing g-protein-coupled receptors as markers of adult stem cells. Gastroenterology. 2010;138(5):1681–1696. doi: 10.1053/j.gastro.2010.03.002. [DOI] [PubMed] [Google Scholar]
  • 81.Carmon KS, Gong X, Lin Q, Thomas A, Liu Q. R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/β-catenin signaling. Proc National Acad Sci. 2011;108(28):11452–11457. doi: 10.1073/pnas.1106083108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Pinto A, Filippi RD, Frigeri F, Corazzelli G, Normanno N. Aging and the hemopoietic system. Crit Rev Oncol Hemat. 2003;48(Suppl):S3–12. doi: 10.1016/j.critrevonc.2003.06.006. [DOI] [PubMed] [Google Scholar]
  • 83.Barker N, Van Es JH, Kuipers J, Kujala P, Van Den Born M, Cozijnsen M, Haegebarth A, Korving J, Begthel H, Peters PJ, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449(7165):1003–1007. doi: 10.1038/nature06196. [DOI] [PubMed] [Google Scholar]
  • 84.Bhushan A, Itoh N, Kato S, Thiery JP, Czernichow P, Bellusci S, Scharfmann R. Fgf10 is essential for maintaining the proliferative capacity of epithelial progenitor cells during early pancreatic organogenesis. Dev Camb Engl. 2001;128:5109–5117. [DOI] [PubMed] [Google Scholar]
  • 85.Norgaard GA, Jensen JN, Jensen J. FGF10 signaling maintains the pancreatic progenitor cell state revealing a novel role of Notch in organ development. Dev Biol. 2003;264(2):323–338. doi: 10.1016/j.ydbio.2003.08.013. [DOI] [PubMed] [Google Scholar]
  • 86.Avalos JL, Bever KM, Wolberger C. Mechanism of sirtuin inhibition by nicotinamide: altering the NAD+ cosubstrate specificity of a Sir2 enzyme. Mol Cell. 2005;17(6):855–868. doi: 10.1016/j.molcel.2005.02.022. [DOI] [PubMed] [Google Scholar]
  • 87.Meng Y, Ren Z, Xu F, Zhou X, Song C, Wang VY-F, Liu W, Lu L, Thomson JA, Chen G. Nicotinamide Promotes Cell Survival and Differentiation as Kinase Inhibitor in Human Pluripotent Stem Cells . Stem Cell Rep. 2018;11(6):1347–56. [DOI] [PMC free article] [PubMed]
  • 88.Furukawa M, Magami Y, Azuma T, Inokuchi H, Nakayama D, Moriyasu F, Kawai K, Hattori T. Proliferation and functional changes of pancreatic gastrin cells in neonatal rat. Pancreas. 2001;23(4):421–426. doi: 10.1097/00006676-200111000-00014. [DOI] [PubMed] [Google Scholar]
  • 89.Suissa Y, Magenheim J, Stolovich-Rain M, Hija A, Collombat P, Mansouri A, Sussel L, Sosa-Pineda B, McCracken K, Wells JM, et al. Gastrin: a distinct fate of neurogenin3 positive progenitor cells in the embryonic pancreas. Plos One. 2013;8(8):e70397. doi: 10.1371/journal.pone.0070397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Watanabe K, Ueno M, Kamiya D, Nishiyama A, Matsumura M, Wataya T, Takahashi JB, Nishikawa S, Nishikawa S, Muguruma K, et al. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat Biotechnol. 2007;25(6):681–686. doi: 10.1038/nbt1310. [DOI] [PubMed] [Google Scholar]
  • 91.VanDussen KL, Sonnek NM, Stappenbeck TS. L-WRN conditioned medium for gastrointestinal epithelial stem cell culture shows replicable batch-to-batch activity levels across multiple research teams. Stem Cell Res. 2019;37:101430. doi: 10.1016/j.scr.2019.101430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Otonkoski T, Beattie GM, Mally MI, Ricordi C, Hayek A. Nicotinamide is a potent inducer of endocrine differentiation in cultured human fetal pancreatic cells. J Clin Invest. 1993;92(3):1459–1466. doi: 10.1172/JCI116723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sugiyama T, Benitez CM, Ghodasara A, Liu L, McLean GW, Lee J, Blauwkamp TA, Nusse R, Wright CVE, Gu G, et al. Reconstituting pancreas development from purified progenitor cells reveals genes essential for islet differentiation. Proc National Acad Sci. 2013;110(31):12691–12696. doi: 10.1073/pnas.1304507110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Lammert E, Cleaver O, Melton DA. Induction of pancreatic differentiation by signals from blood vessels. Science. 2001;294(5542):564–567. doi: 10.1126/science.1064344. [DOI] [PubMed] [Google Scholar]
  • 95.Pictet RL, Clark WR, Williams RH, Rutter WJ. An ultrastructural analysis of the developing embryonic pancreas. Dev Biol. 1972;29(4):436–467. doi: 10.1016/0012-1606(72)90083-8. [DOI] [PubMed] [Google Scholar]
  • 96.Kim SK, Hebrok M, Melton DA. Notochord to endoderm signaling is required for pancreas development. Dev Camb Engl. 1997;124:4243–4252. [DOI] [PubMed] [Google Scholar]
  • 97.Deutsch G, Jung J, Zheng M, Lóra J, Zaret KS. A bipotential precursor population for pancreas and liver within the embryonic endoderm. Dev Camb Engl. 2001;128:871–881. [DOI] [PubMed] [Google Scholar]
  • 98.Habener JF, Kemp DM, Thomas MK. Minireview: transcriptional regulation in pancreatic development. Endocrinology. 2005;146(3):1025–1034. doi: 10.1210/en.2004-1576. [DOI] [PubMed] [Google Scholar]
  • 99.Bonal C, Herrera PL. Genes controlling pancreas ontogeny. Int J Dev Bio. 2008;52(7):823–835. doi: 10.1387/ijdb.072444cb. [DOI] [PubMed] [Google Scholar]
  • 100.Dassaye R, Naidoo S, Cerf ME. Transcription factor regulation of pancreatic organogenesis, differentiation and maturation. Islets. 2015;8(1):13–34. doi: 10.1080/19382014.2015.1075687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Benitez CM, Goodyer WR, Kim SK. Deconstructing pancreas developmental biology. Csh Perspect Biol. 2012;4:a012401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Yoshitomi H, Zaret KS. Endothelial cell interactions initiate dorsal pancreas development by selectively inducing the transcription factor Ptf1a. Development. 2004;131(4):807–817. doi: 10.1242/dev.00960. [DOI] [PubMed] [Google Scholar]
  • 103.Zhou Q, Law AC, Rajagopal J, Anderson WJ, Gray PA, Melton DA. A multipotent progenitor domain guides pancreatic organogenesis. Dev Cell. 2007;13(1):103–114. doi: 10.1016/j.devcel.2007.06.001. [DOI] [PubMed] [Google Scholar]
  • 104.Sharon N, Chawla R, Mueller J, Vanderhooft J, Whitehorn LJ, Rosenthal B, Gürtler M, Estanboulieh RR, Shvartsman D, Gifford DK, et al. A peninsular structure coordinates asynchronous differentiation with morphogenesis to generate pancreatic islets. Cell. 2019;176(4):790–804.e13. doi: 10.1016/j.cell.2018.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Stolovich-Rain M, Enk J, Vikesa J, Nielsen FC, Saada A, Glaser B, Dor Y. Weaning triggers a maturation step of pancreatic β cells. Dev Cell. 2015;32(5):535–545. doi: 10.1016/j.devcel.2015.01.002. [DOI] [PubMed] [Google Scholar]
  • 106.Bosco D, Armanet M, Morel P, Niclauss N, Sgroi A, Muller YD, Giovannoni L, Parnaud G, Berney T. Unique arrangement of alpha- and beta-cells in human islets of Langerhans. Diabetes. 2010;59(5):1202–1210. doi: 10.2337/db09-1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Granger A, Kushner JA. Cellular origins of-cell regeneration: a legacy view of historical controversies. J Intern Med. 2009;266(4):325–338. doi: 10.1111/j.1365-2796.2009.02156.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Butler AE, Cao-Minh L, Galasso R, Rizza RA, Corradin A, Cobelli C, Butler PC. Adaptive changes in pancreatic beta cell fractional area and beta cell turnover in human pregnancy. Diabetologia. 2010;53(10):2167–2176. doi: 10.1007/s00125-010-1809-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Gregg BE, Moore PC, Demozay D, Hall BA, Li M, Husain A, Wright AJ, Atkinson MA, Rhodes CJ. Formation of a human β-cell population within pancreatic islets is set early in life. J Clin Endocrinol Metabolism. 2012;97(9):3197–3206. doi: 10.1210/jc.2012-1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Bonner-Weir S, Baxter LA, Schuppin GT, Smith FE. A second pathway for regeneration of adult exocrine and endocrine pancreas. A possible recapitulation of embryonic development. Diabetes. 1993;42(12):1715–1720. doi: 10.2337/diab.42.12.1715. [DOI] [PubMed] [Google Scholar]
  • 111.Inada A, Nienaber C, Katsuta H, Fujitani Y, Levine J, Morita R, Sharma A, Bonner-Weir S. Carbonic anhydrase II-positive pancreatic cells are progenitors for both endocrine and exocrine pancreas after birth. P Natl Acad Sci USA. 2008;105(50):19915–19919. doi: 10.1073/pnas.0805803105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Xu X, D’Hoker J, Stangé G, Bonné S, Leu ND, Xiao X, Van De Casteele M, Mellitzer G, Ling Z, Pipeleers D, et al. Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell. 2008;132(2):197–207. doi: 10.1016/j.cell.2007.12.015. [DOI] [PubMed] [Google Scholar]
  • 113.Criscimanna A, Speicher JA, Houshmand G, Shiota C, Prasadan K, Ji B, Logsdon CD, Gittes GK, Esni F. Duct cells contribute to regeneration of endocrine and acinar cells following pancreatic damage in adult mice. Gastroenterology. 2011;141(4):1451–1462.e6. doi: 10.1053/j.gastro.2011.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Desai BM, Oliver-Krasinski J, Leon DDD, Farzad C, Hong N, Leach SD, Stoffers DA. Preexisting pancreatic acinar cells contribute to acinar cell, but not islet beta cell, regeneration. J Clin Invest. 2007;117(4):971–977. doi: 10.1172/JCI29988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Teta M, Rankin MM, Long SY, Stein GM, Kushner JA. Growth and regeneration of adult beta cells does not involve specialized progenitors. Dev Cell. 2007;12(5):817–826. doi: 10.1016/j.devcel.2007.04.011. [DOI] [PubMed] [Google Scholar]
  • 116.Solar M, Cardalda C, Houbracken I, Martín M, Maestro MA, Medts ND, Xu X, Grau V, Heimberg H, Bouwens L, et al. Pancreatic exocrine duct cells give rise to insulin-producing cells during embryogenesis but not after birth. Dev Cell. 2009;17(6):849–860. doi: 10.1016/j.devcel.2009.11.003. [DOI] [PubMed] [Google Scholar]
  • 117.Kopp JL, Dubois CL, Schaffer AE, Hao E, Shih HP, Seymour PA, Ma J, Sander M. 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):653–665. doi: 10.1242/dev.056499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Furuyama K, Kawaguchi Y, Akiyama H, Horiguchi M, Kodama S, Kuhara T, Hosokawa S, Elbahrawy A, Soeda T, Koizumi M, 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. doi: 10.1038/ng.722. [DOI] [PubMed] [Google Scholar]
  • 119.Xiao X, Chen Z, Shiota C, Prasadan K, Guo P, El-Gohary Y, Paredes J, Welsh C, Wiersch J, Gittes GK. No evidence for β cell neogenesis in murine adult pancreas. J Clin Invest. 2013;123(5):2207–2217. doi: 10.1172/JCI66323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Dor Y, Brown J, Martinez OI, Melton DA. Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation. Nature. 2004;429(6987):41–46. doi: 10.1038/nature02520. [DOI] [PubMed] [Google Scholar]
  • 121.Stolovich-Rain M, Hija A, Grimsby J, Glaser B, Dor Y. Pancreatic beta cells in very old mice retain capacity for compensatory proliferation. J Biol Chem. 2012;287(33):27407–27414. doi: 10.1074/jbc.M112.350736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Nir T, Melton DA, Dor Y. Recovery from diabetes in mice by β cell regeneration. J Clin Invest. 2007;117(9):2553–2561. doi: 10.1172/JCI32959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Dor Y, Melton DA. Facultative endocrine progenitor cells in the adult pancreas. Cell. 2008;132(2):183–184. doi: 10.1016/j.cell.2008.01.004. [DOI] [PubMed] [Google Scholar]
  • 124.Yanger K, Stanger BZ. Facultative stem cells in liver and pancreas: fact and fancy. Dev Dynam. 2011;240(3):521–529. doi: 10.1002/dvdy.22561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Lysy PA, Weir GC, Bonner-Weir S. Making β cells from adult cells within the pancreas. Curr Diabetes Rep. 2013;13(5):695–703. doi: 10.1007/s11892-013-0400-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Ziv O, Glaser B, Dor Y. The plastic pancreas. Dev Cell. 2013;26(1):3–7. doi: 10.1016/j.devcel.2013.06.013. [DOI] [PubMed] [Google Scholar]
  • 127.Dan YY. Chasing the facultative liver progenitor cell. Hepatology. 2016;64(1):297–300. doi: 10.1002/hep.28642. [DOI] [PubMed] [Google Scholar]
  • 128.Yu K, Fischbach S, Xiao X. Beta cell regeneration in adult mice: controversy over the involvement of stem cells. Curr Stem Cell Res T. 2016;11(7):542–546. doi: 10.2174/1574888X10666141126113110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Afelik S, Rovira M. Pancreatic β-cell regeneration: facultative or dedicated progenitors? Mol Cell Endocrinol. 2017;445:85–94. doi: 10.1016/j.mce.2016.11.008. [DOI] [PubMed] [Google Scholar]
  • 130.Aguayo-Mazzucato C, Bonner-Weir S. Pancreatic β cell regeneration as a possible therapy for diabetes. Cell Metab. 2018;27(1):57–67. doi: 10.1016/j.cmet.2017.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Zhou Q, Melton DA. Pancreas regeneration. Nature. 2018;557(7705):351–358. doi: 10.1038/s41586-018-0088-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Thorel F, Népote V, Avril I, Kohno K, Desgraz R, Chera S, Herrera PL. Conversion of adult pancreatic α-cells to β-cells after extreme β-cell loss. Nature. 2010;464(7292):1149–1154. doi: 10.1038/nature08894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Chera S, Baronnier D, Ghila L, Cigliola V, Jensen JN, Gu G, Furuyama K, Thorel F, Gribble FM, Reimann F, et al. Diabetes recovery by age-dependent conversion of pancreatic δ-cells into insulin producers. Nature. 2014;514(7523):503–507. doi: 10.1038/nature13633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Clevers H, Watt FM. Defining adult stem cells by function, not by phenotype. Annu Rev Biochem. 2018;87(1):1–13. doi: 10.1146/annurev-biochem-062917-012341. [DOI] [PubMed] [Google Scholar]
  • 135.Gehart H, Clevers H. Tales from the crypt: new insights into intestinal stem cells. Nat Rev Gastroentero. 2019;16(1):19–34. doi: 10.1038/s41575-018-0081-y. [DOI] [PubMed] [Google Scholar]
  • 136.Gurdon JB. A community effect in animal development. Nature. 1988;336(6201):772–774. doi: 10.1038/336772a0. [DOI] [PubMed] [Google Scholar]
  • 137.Bonner-Weir S, Taneja M, Weir GC, Tatarkiewicz K, Song KH, Sharma A, O’Neil JJ. In vitro cultivation of human islets from expanded ductal tissue. P Natl Acad Sci USA. 2000;97(14):7999–8004. doi: 10.1073/pnas.97.14.7999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Kumar KK, Burgess AW, Gulbis JM. Structure and function of LGR5: an enigmatic G-protein coupled receptor marking stem cells. Protein Sci. 2014;23(5):551–565. doi: 10.1002/pro.2446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Morita H, Mazerbourg S, Bouley DM, Luo C-W, Kawamura K, Kuwabara Y, Baribault H, Tian H, Hsueh AJW. Neonatal lethality of LGR5 null mice is associated with ankyloglossia and gastrointestinal distension. Mol Cell Biol. 2004;24(22):9736–9743. doi: 10.1128/MCB.24.22.9736-9743.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Jaks V, Barker N, Kasper M, Van Es JH, Snippert HJ, Clevers H, Toftgård R. Lgr5 marks cycling, yet long-lived, hair follicle stem cells. Nat Genet. 2008;40(11):1291–1299. doi: 10.1038/ng.239. [DOI] [PubMed] [Google Scholar]
  • 141.Barker N, Van Es JH, Jaks V, Kasper M, Snippert H, Toftgård R, Clevers H. Very long-term self-renewal of small intestine, colon, and hair follicles from cycling Lgr5+ve STEM CELls. Cold Spring Harb Sym. 2008;73(0):351–356. doi: 10.1101/sqb.2008.72.003. [DOI] [PubMed] [Google Scholar]
  • 142.Pan FC, Bankaitis ED, Boyer D, Xu X, De Casteele MV, Magnuson MA, Heimberg H, Wright CVE. Spatiotemporal patterns of multipotentiality in Ptf1a-expressing cells during pancreas organogenesis and injury-induced facultative restoration. Development. 2013;140(4):751–764. doi: 10.1242/dev.090159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Jin K, Xiang M. Transcription factor Ptf1a in development, diseases and reprogramming. Cell Mol Life Sci. 2019;76(5):921–940. doi: 10.1007/s00018-018-2972-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Öström M, Loffler KA, Edfalk S, Selander L, Dahl U, Ricordi C, Jeon J, Correa-Medina M, Diez J, Edlund H. Retinoic acid promotes the generation of pancreatic endocrine progenitor cells and their further differentiation into β-cells. Plos One. 2008;3(7):e2841. doi: 10.1371/journal.pone.0002841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Ioannou M, Serafimidis I, Arnes L, Sussel L, Singh S, Vasiliou V, Gavalas A. ALDH1B1 is a potential stem/progenitor marker for multiple pancreas progenitor pools. Dev Biol. 2013;374(1):153–163. doi: 10.1016/j.ydbio.2012.10.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Yatoh S, Dodge R, Akashi T, Omer A, Sharma A, Weir GC, Bonner-Weir S. Differentiation of affinity-purified human pancreatic duct cells to beta-cells. Diabetes. 2007;56(7):1802–1809. doi: 10.2337/db06-1670. [DOI] [PubMed] [Google Scholar]
  • 147.Rovira M, Scott S-G, Liss AS, Jensen J, Thayer SP, Leach SD. Isolation and characterization of centroacinar/terminal ductal progenitor cells in adult mouse pancreas. Proc National Acad Sci. 2010;107(1):75–80. doi: 10.1073/pnas.0912589107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Socorro M, Criscimanna A, Riva P, Tandon M, Prasadan K, Guo P, Humar A, Husain SZ, Leach SD, Gittes GK, et al. Identification of newly committed pancreatic cells in the adult mouse pancreas. Sci Rep-uk. 2017;7(1):17539. doi: 10.1038/s41598-017-17884-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Kania G, Corbeil D, Fuchs J, Tarasov KV, Blyszczuk P, Huttner WB, Boheler KR, Wobus AM. Somatic stem cell marker prominin-1/CD133 is expressed in embryonic stem cell–derived progenitors. Stem Cells. 2005;23(6):791–804. doi: 10.1634/stemcells.2004-0232. [DOI] [PubMed] [Google Scholar]
  • 150.Ratajczak MZ, Kucia M, Reca R, Majka M, Janowska-Wieczorek A, Ratajczak J. Stem cell plasticity revisited: CXCR4-positive cells expressing mRNA for early muscle, liver and neural cells “hide out” in the bone marrow. Leukemia. 2004;18(1):29–40. doi: 10.1038/sj.leu.2403184. [DOI] [PubMed] [Google Scholar]
  • 151.Gallacher L, Murdoch B, Wu DM, Karanu FN, Keeney M, Bhatia M. Isolation and characterization of human CD34(-)Lin(-) and CD34(+)Lin(-) hematopoietic stem cells using cell surface markers AC133 and CD7. Blood. 2000;95(9):2813–2820. doi: 10.1182/blood.V95.9.2813.009k20_2813_2820. [DOI] [PubMed] [Google Scholar]
  • 152.Uchida N, Buck DW, He D, Reitsma MJ, Masek M, Phan TV, Tsukamoto AS, Gage FH, Weissman IL. Direct isolation of human central nervous system stem cells. Proc National Acad Sci. 2000;97(26):14720–14725. doi: 10.1073/pnas.97.26.14720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Lardon J, Corbeil D, Huttner WB, Ling Z, Bouwens L. Stem cell marker prominin-1/AC133 is expressed in duct cells of the adult human pancreas. Pancreas. 2008;36(1):e1–6. doi: 10.1097/mpa.0b013e318149f2dc. [DOI] [PubMed] [Google Scholar]
  • 154.Karbanová J, Missol-Kolka E, Fonseca A-V, Lorra C, Janich P, Hollerová H, Jászai J, Ehrmann J, Kolář Z, Liebers C, et al. The stem cell marker CD133 (Prominin-1) is expressed in various human glandular epithelia. J Histochem Cytochem. 2008;56(11):977–993. doi: 10.1369/jhc.2008.951897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Sugiyama T, Rodriguez RT, McLean GW, Kim SK. Conserved markers of fetal pancreatic epithelium permit prospective isolation of islet progenitor cells by FACS. P Natl Acad Sci USA. 2007;104(1):175–180. doi: 10.1073/pnas.0609490104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Oshima Y, Suzuki A, Kawashimo K, Ishikawa M, Ohkohchi N, Taniguchi H. Isolation of mouse pancreatic ductal progenitor cells expressing CD133 and c-Met by flow cytometric cell sorting. Gastroenterology. 2007;132(2):720–732. doi: 10.1053/j.gastro.2006.11.027. [DOI] [PubMed] [Google Scholar]
  • 157.Jin L, Gao D, Feng T, Tremblay JR, Ghazalli N, Luo A, Rawson J, Quijano JC, Chai J, Wedeken L, et al. Cells with surface expression of CD133 high CD71 low are enriched for tripotent colony-forming progenitor cells in the adult murine pancreas. Stem Cell Res. 2016;16(1):40–53. doi: 10.1016/j.scr.2015.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Koblas T, Pektorova L, Zacharovova K, Berkova Z, Girman P, Dovolilova E, Karasova L, Saudek F. Differentiation of CD133-positive pancreatic cells into insulin-producing islet-like cell clusters. Transpl P. 2008;40(2):415–418. doi: 10.1016/j.transproceed.2008.02.017. [DOI] [PubMed] [Google Scholar]
  • 159.Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature. 2008;455(7213):627–632. doi: 10.1038/nature07314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Vorburger SA, Hunt KK. Adenoviral gene therapy. Oncol. 2002;7(1):46–59. doi: 10.1634/theoncologist.7-1-46. [DOI] [PubMed] [Google Scholar]
  • 161.Wang D, Wang J, Bai L, Pan H, Feng H, Clevers H, Zeng YA. Long-Term expansion of pancreatic islet organoids from resident procr+ progenitors. Cell. 2020;180(6):1198–1211.e19. doi: 10.1016/j.cell.2020.02.048. [DOI] [PubMed] [Google Scholar]
  • 162.Balazs AB, Fabian AJ, Esmon CT, Mulligan RC. Endothelial protein C receptor (CD201) explicitly identifies hematopoietic stem cells in murine bone marrow. Blood. 2006;107(6):2317–2321. doi: 10.1182/blood-2005-06-2249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Martin GH, Park CY. EPCR: a novel marker of cultured cord blood HSCs. Blood. 2017;129(25):3279–3280. doi: 10.1182/blood-2017-05-780007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Yu QC, Song W, Wang D, Zeng YA. Identification of blood vascular endothelial stem cells by the expression of protein C receptor. Cell Res. 2016;26(10):1079–1098. doi: 10.1038/cr.2016.85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Wang D, Liu C, Wang J, Jia Y, Hu X, Jiang H, Shao Z, Zeng YA. Protein C receptor stimulates multiple signaling pathways in breast cancer cells. J Biol Chem. 2018;293(4):1413–1424. doi: 10.1074/jbc.M117.814046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Pisania A, Weir GC, O’Neil JJ, Omer A, Tchipashvili V, Lei J, Colton CK, Bonner-Weir S. Quantitative analysis of cell composition and purity of human pancreatic islet preparations. Lab Invest. 2010;90(11):1661–1675. doi: 10.1038/labinvest.2010.124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, Eliazer S, Young H, Richardson M, Smart NG, Cunningham J, et al. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol. 2008;26(4):443–452. doi: 10.1038/nbt1393. [DOI] [PubMed] [Google Scholar]
  • 168.Rorsman P, Braun M. Regulation of insulin secretion in human pancreatic islets. Annu Rev Physiol. 2013;75(1):155–179. doi: 10.1146/annurev-physiol-030212-183754. [DOI] [PubMed] [Google Scholar]
  • 169.Nikolova G, Strilic B, Lammert E. The vascular niche and its basement membrane. Trends Cell Biol. 2007;17(1):19–25. doi: 10.1016/j.tcb.2006.11.005. [DOI] [PubMed] [Google Scholar]
  • 170.Lammert E, Thorn P. The role of the islet niche on beta cell structure and function. J Mol Biol. 2019;432(5):1407–1418. doi: 10.1016/j.jmb.2019.10.032. [DOI] [PubMed] [Google Scholar]

Articles from Islets are provided here courtesy of Taylor & Francis

RESOURCES