Diabetes Metab J.  2023 Mar;47(2):173-184. 10.4093/dmj.2022.0324.

Heterogeneity of Islet Cells during Embryogenesis and Differentiation

Affiliations
  • 1Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Suita, Japan
  • 2Department of Endocrinology, Diabetes and Metabolism, Kitasato University School of Medicine, Sagamihara, Japan

Abstract

Diabetes is caused by insufficient insulin secretion due to β-cell dysfunction and/or β-cell loss. Therefore, the restoration of functional β-cells by the induction of β-cell differentiation from embryonic stem (ES) and induced-pluripotent stem (iPS) cells, or from somatic non-β-cells, may be a promising curative therapy. To establish an efficient and feasible method for generating functional insulin-producing cells, comprehensive knowledge of pancreas development and β-cell differentiation, including the mechanisms driving cell fate decisions and endocrine cell maturation is crucial. Recent advances in single-cell RNA sequencing (scRNA-seq) technologies have opened a new era in pancreas development and diabetes research, leading to clarification of the detailed transcriptomes of individual insulin-producing cells. Such extensive high-resolution data enables the inference of developmental trajectories during cell transitions and gene regulatory networks. Additionally, advancements in stem cell research have not only enabled their immediate clinical application, but also has made it possible to observe the genetic dynamics of human cell development and maturation in a dish. In this review, we provide an overview of the heterogeneity of islet cells during embryogenesis and differentiation as demonstrated by scRNA-seq studies on the developing and adult pancreata, with implications for the future application of regenerative medicine for diabetes.

Keyword

Cell differentiation; Diabetes mellitus; Embryonic development; Endocrine cells; Regenerative medicine; Single-cell gene expression analysis; Stem cell research

Figure

  • Fig. 1. Heterogeneous properties of pancreatic cells during development. Cell-type transitions during pancreas development in mice that have been clarified by single-cell transcriptome analyses are shown. Cell-specific markers are listed near the cell type. The black arrow indicates the developmental trajectory. CP, common progenitors; EP, endocrine progenitors. Pdx1, pancreatic and duodenal homeobox 1; Sox9, SRY-box transcription factor 9; Foxa2, forkhead box protein A2; Nr2f2, nuclear receptor subfamily 2 group F member 2; Ptf1a, pancreas associated transcription factor 1A; Cpa1, carboxypeptidase A1; Nkx6.1, NK6 homeobox 1; Dcdc2a, doublecortin domain containing 2a; Gcg, glucagon; Mafb, MAF bZIP transcription factor B; Pou3f4, POU class 3 homeobox 4; Rbpjl, recombination signal binding protein for immunoglobulin kappa J region like; Amy1, amylase 1, salivary; Atf3, activating transcription factor 3; Klf5, KLF transcription factor 5; Nr1h4, nuclear receptor subfamily 1 group H member 4; Hnf1b, HNF1 homeobox B; Atoh8, atonal bHLH transcription factor 8; Rfx6, regulatory factor X6; Neurog3, neurogenin 3; Prdx2, peroxiredoxin 2; Neurod2, neurogenic differentiation 2; Vim, vimentin; Isl1, ISL LIM homeobox 1; Chga, chromogranin A; Glis3, GLIS family zinc finger 3; Meis2, Meis homeobox 2; Mnx1, motor neuron and pancreas homeobox 1; Irx2, iroquois homeobox 2; Ins, insulin; Mlxipl, MLX interacting protein like; Ucn3, urocortin 3; Sst, somatostatin; Hhex, hematopoietically expressed homeobox; Ghrl, ghrelin and obestatin prepropeptide; PP, pancreatic polypeptide.

  • Fig. 2. Heterogeneity of newly generated β-cells surrounded by a heterogenous spatial environment. (A) Proposed model of β-cell genesis. Endocrine progenitors differentiate into two types of β-cells; newborn β-cells near the ductal regions (βduct) and newborn β-cells adjacent to blood vessels (βvessel) cells. The βduct cells are observed in the ductal region where neurogenin 3 (Neurog3)-expressing endocrine progenitors emerge. In contrast, a subpopulation of endocrine progenitors migrates away from the ductal area toward the region near the blood vessels, and differentiate into βvessel cells. (B) Cell-type transitions in mouse β-cell development are shown. Cell-specific markers are listed near the cell type. The black arrows indicate the developmental trajectory. MafA, MAF bZIP transcription factor A; Sst, somatostatin; Gcg, glucagon; MafB, MAF bZIP transcription factor B; Nnat, neuronatin; Iapp, islet amyloid polypeptide; G6pc2, glucose-6-phosphatase catalytic subunit 2; Slc2a2, solute carrier family 2 member 2.


Reference

1. International Diabetes Federation. IDF Diabetes Atlas. 10th ed. Brussels: International Diabetes Federation;2021.
2. Fowler MJ. Microvascular and macrovascular complications of diabetes. Clin Diabetes. 2008; 26:77–82.
3. Tabak AG, Jokela M, Akbaraly TN, Brunner EJ, Kivimaki M, Witte DR. Trajectories of glycaemia, insulin sensitivity, and insulin secretion before diagnosis of type 2 diabetes: an analysis from the Whitehall II study. Lancet. 2009; 373:2215–21.
4. Sakuraba H, Mizukami H, Yagihashi N, Wada R, Hanyu C, Yagihashi S. Reduced beta-cell mass and expression of oxidative stress-related DNA damage in the islet of Japanese type II diabetic patients. Diabetologia. 2002; 45:85–96.
5. Sasaki H, Saisho Y, Inaishi J, Watanabe Y, Tsuchiya T, Makio M, et al. Reduced beta cell number rather than size is a major contributor to beta cell loss in type 2 diabetes. Diabetologia. 2021; 64:1816–21.
6. Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC. Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes. 2003; 52:102–10.
7. Mizukami H, Takahashi K, Inaba W, Tsuboi K, Osonoi S, Yoshida T, et al. Involvement of oxidative stress-induced DNA damage, endoplasmic reticulum stress, and autophagy deficits in the decline of β-cell mass in Japanese type 2 diabetic patients. Diabetes Care. 2014; 37:1966–74.
8. Enge M, Arda HE, Mignardi M, Beausang J, Bottino R, Kim SK, et al. Single-cell analysis of human pancreas reveals transcriptional signatures of aging and somatic mutation patterns. Cell. 2017; 171:321–30.
9. Segerstolpe A, Palasantza A, Eliasson P, Andersson EM, Andreasson AC, Sun X, et al. Single-cell transcriptome profiling of human pancreatic islets in health and type 2 diabetes. Cell Metab. 2016; 24:593–607.
10. Ramond C, Beydag-Tasoz BS, Azad A, van de Bunt M, Petersen MB, Beer NL, et al. Understanding human fetal pancreas development using subpopulation sorting, RNA sequencing and single-cell profiling. Development. 2018; 145:dev165480.
11. Petersen MB, Azad A, Ingvorsen C, Hess K, Hansson M, Grapin-Botton A, et al. Single-cell gene expression analysis of a human ESC model of pancreatic endocrine development reveals different paths to β-cell differentiation. Stem Cell Reports. 2017; 9:1246–61.
12. Qiu WL, Zhang YW, Feng Y, Li LC, Yang L, Xu CR. Deciphering pancreatic islet β cell and α cell maturation pathways and characteristic features at the single-cell level. Cell Metab. 2017; 25:1194–205.
13. Zeng C, Mulas F, Sui Y, Guan T, Miller N, Tan Y, et al. Pseudotemporal ordering of single cells reveals metabolic control of postnatal β cell proliferation. Cell Metab. 2017; 25:1160–75.
14. Krentz NA, Lee MY, Xu EE, Sproul SL, Maslova A, Sasaki S, et al. Single-cell transcriptome profiling of mouse and hESC-derived pancreatic progenitors. Stem Cell Reports. 2018; 11:1551–64.
15. Yu XX, Qiu WL, Yang L, Zhang Y, He MY, Li LC, et al. Defining multistep cell fate decision pathways during pancreatic development at single-cell resolution. EMBO J. 2019; 38:e100164.
16. Wells JM, Melton DA. Early mouse endoderm is patterned by soluble factors from adjacent germ layers. Development. 2000; 127:1563–72.
17. Slack JM. Developmental biology of the pancreas. Development. 1995; 121:1569–80.
18. Edlund H. Pancreatic organogenesis: developmental mechanisms and implications for therapy. Nat Rev Genet. 2002; 3:524–32.
19. Zaret KS. Genetic programming of liver and pancreas progenitors: lessons for stem-cell differentiation. Nat Rev Genet. 2008; 9:329–40.
20. Miyatsuka T, Matsuoka TA, Kaneto H. Transcription factors as therapeutic targets for diabetes. Expert Opin Ther Targets. 2008; 12:1431–42.
21. Offield MF, Jetton TL, Labosky PA, Ray M, Stein RW, Magnuson MA, et al. PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development. 1996; 122:983–95.
22. Burlison JS, Long Q, Fujitani Y, Wright CV, Magnuson MA. Pdx-1 and Ptf1a concurrently determine fate specification of pancreatic multipotent progenitor cells. Dev Biol. 2008; 316:74–86.
23. Lynn FC, Smith SB, Wilson ME, Yang KY, Nekrep N, German MS. Sox9 coordinates a transcriptional network in pancreatic progenitor cells. Proc Natl Acad Sci U S A. 2007; 104:10500–5.
24. Kawaguchi Y. Sox9 and programming of liver and pancreatic progenitors. J Clin Invest. 2013; 123:1881–6.
25. Krapp A, Knofler M, Ledermann B, Burki K, Berney C, Zoerkler N, et al. The bHLH protein PTF1-p48 is essential for the formation of the exocrine and the correct spatial organization of the endocrine pancreas. Genes Dev. 1998; 12:3752–63.
26. Kawaguchi Y, Cooper B, Gannon M, Ray M, MacDonald RJ, Wright CV. The role of the transcriptional regulator Ptf1a in converting intestinal to pancreatic progenitors. Nat Genet. 2002; 32:128–34.
27. Jeon J, Correa-Medina M, Ricordi C, Edlund H, Diez JA. Endocrine cell clustering during human pancreas development. J Histochem Cytochem. 2009; 57:811–24.
28. Zhou Q, Law AC, Rajagopal J, Anderson WJ, Gray PA, Melton DA. A multipotent progenitor domain guides pancreatic organogenesis. Dev Cell. 2007; 13:103–14.
29. Jennings RE, Berry AA, Strutt JP, Gerrard DT, Hanley NA. Human pancreas development. Development. 2015; 142:3126–37.
30. Gradwohl G, Dierich A, LeMeur M, Guillemot F. Neurogenin3 is required for the development of the four endocrine cell lineages of the pancreas. Proc Natl Acad Sci U S A. 2000; 97:1607–11.
31. Gu G, Dubauskaite J, Melton DA. Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors. Development. 2002; 129:2447–57.
32. Sheets TP, Park KE, Park CH, Swift SM, Powell A, Donovan DM, et al. Targeted mutation of NGN3 gene disrupts pancreatic endocrine cell development in pigs. Sci Rep. 2018; 8:3582.
33. Miyatsuka T. Chronology of endocrine differentiation and beta-cell neogenesis. Endocr J. 2016; 63:205–11.
34. Larsson LI. On the development of the islets of Langerhans. Microsc Res Tech. 1998; 43:284–91.
35. Villasenor A, Chong DC, Cleaver O. Biphasic Ngn3 expression in the developing pancreas. Dev Dyn. 2008; 237:3270–9.
36. Schwitzgebel VM, Scheel DW, Conners JR, Kalamaras J, Lee JE, Anderson DJ, et al. Expression of neurogenin3 reveals an islet cell precursor population in the pancreas. Development. 2000; 127:3533–42.
37. Miyatsuka T, Li Z, German MS. Chronology of islet differentiation revealed by temporal cell labeling. Diabetes. 2009; 58:1863–8.
38. Jennings RE, Berry AA, Kirkwood-Wilson R, Roberts NA, Hearn T, Salisbury RJ, et al. Development of the human pancreas from foregut to endocrine commitment. Diabetes. 2013; 62:3514–22.
39. Katahira T, Miyatsuka T, Miura M, Suzuki L, Himuro M, Nishida Y, et al. Conversion of pancreatic α cells into insulinproducing cells modulated by β-cell insufficiency and supplemental insulin administration. Biochem Biophys Res Commun. 2020; 521:178–83.
40. Spijker HS, Song H, Ellenbroek JH, Roefs MM, Engelse MA, Bos E, et al. Loss of β-cell identity occurs in type 2 diabetes and is associated with islet amyloid deposits. Diabetes. 2015; 64:2928–38.
41. Shigeno R, Horie I, Miwa M, Ito A, Haraguchi A, Natsuda S, et al. Bihormonal dysregulation of insulin and glucagon contributes to glucose intolerance development at one year postdelivery in women with gestational diabetes: a prospective cohort study using an early postpartum 75-g glucose tolerance test. Endocr J. 2021; 68:919–31.
42. Shih HP, Wang A, Sander M. Pancreas organogenesis: from lineage determination to morphogenesis. Annu Rev Cell Dev Biol. 2013; 29:81–105.
43. Larsen HL, Martin-Coll L, Nielsen AV, Wright CV, Trusina A, Kim YH, et al. Stochastic priming and spatial cues orchestrate heterogeneous clonal contribution to mouse pancreas organogenesis. Nat Commun. 2017; 8:605.
44. Sznurkowska MK, Hannezo E, Azzarelli R, Rulands S, Nestorowa S, Hindley CJ, et al. Defining lineage potential and fate behavior of precursors during pancreas development. Dev Cell. 2018; 46:360–75.
45. Bastidas-Ponce A, Tritschler S, Dony L, Scheibner K, TarquisMedina M, Salinno C, et al. Comprehensive single cell mRNA profiling reveals a detailed roadmap for pancreatic endocrinogenesis. Development. 2019; 146:dev173849.
46. Scavuzzo MA, Hill MC, Chmielowiec J, Yang D, Teaw J, Sheng K, et al. Endocrine lineage biases arise in temporally distinct endocrine progenitors during pancreatic morphogenesis. Nat Commun. 2018; 9:3356.
47. van Gurp L, Muraro MJ, Dielen T, Seneby L, Dharmadhikari G, Gradwohl G, et al. A transcriptomic roadmap to α- and β-cell differentiation in the embryonic pancreas. Development. 2019; 146:dev173716.
48. Wang Y, Li Z, Xu P, Huang L, Tong J, Huang H, et al. Angiomotin-like2 gene (amotl2) is required for migration and proliferation of endothelial cells during angiogenesis. J Biol Chem. 2011; 286:41095–104.
49. Zhao B, Li L, Lu Q, Wang LH, Liu CY, Lei Q, et al. Angiomotin is a novel Hippo pathway component that inhibits YAP oncoprotein. Genes Dev. 2011; 25:51–63.
50. Li Z, Wang Y, Zhang M, Xu P, Huang H, Wu D, et al. The Amotl2 gene inhibits Wnt/β-catenin signaling and regulates embryonic development in zebrafish. J Biol Chem. 2012; 287:13005–15.
51. Mojallal M, Zheng Y, Hultin S, Audebert S, van Harn T, Johnsson P, et al. AmotL2 disrupts apical-basal cell polarity and promotes tumour invasion. Nat Commun. 2014; 5:4557.
52. Jensen J, Pedersen EE, Galante P, Hald J, Heller RS, Ishibashi M, et al. Control of endodermal endocrine development by Hes-1. Nat Genet. 2000; 24:36–44.
53. Cebola I, Rodriguez-Segui SA, Cho CH, Bessa J, Rovira M, Luengo M, et al. TEAD and YAP regulate the enhancer network of human embryonic pancreatic progenitors. Nat Cell Biol. 2015; 17:615–26.
54. Rosado-Olivieri EA, Anderson K, Kenty JH, Melton DA. YAP inhibition enhances the differentiation of functional stem cellderived insulin-producing β cells. Nat Commun. 2019; 10:1464.
55. Sharon N, Vanderhooft J, Straubhaar J, Mueller J, Chawla R, Zhou Q, et al. Wnt signaling separates the progenitor and endocrine compartments during pancreas development. Cell Rep. 2019; 27:2281–91.
56. Szlachcic WJ, Ziojla N, Kizewska DK, Kempa M, Borowiak M. Endocrine pancreas development and dysfunction through the lens of single-cell RNA-sequencing. Front Cell Dev Biol. 2021; 9:629212.
57. Byrnes LE, Wong DM, Subramaniam M, Meyer NP, Gilchrist CL, Knox SM, et al. Lineage dynamics of murine pancreatic development at single-cell resolution. Nat Commun. 2018; 9:3922.
58. Schonhoff SE, Giel-Moloney M, Leiter AB. Neurogenin 3-expressing progenitor cells in the gastrointestinal tract differentiate into both endocrine and non-endocrine cell types. Dev Biol. 2004; 270:443–54.
59. Bechard ME, Bankaitis ED, Hipkens SB, Ustione A, Piston DW, Yang YP, et al. Precommitment low-level Neurog3 expression defines a long-lived mitotic endocrine-biased progenitor pool that drives production of endocrine-committed cells. Genes Dev. 2016; 30:1852–65.
60. Duvall E, Benitez CM, Tellez K, Enge M, Pauerstein PT, Li L, et al. Single-cell transcriptome and accessible chromatin dynamics during endocrine pancreas development. Proc Natl Acad Sci U S A. 2022; 119:e2201267119.
61. Veres A, Faust AL, Bushnell HL, Engquist EN, Kenty JH, Harb G, et al. Charting cellular identity during human in vitro β-cell differentiation. Nature. 2019; 569:368–73.
62. Augsornworawat P, Maxwell KG, Velazco-Cruz L, Millman JR. Single-cell transcriptome profiling reveals β cell maturation in stem cell-derived islets after transplantation. Cell Rep. 2020; 32:108067.
63. Johansson KA, Dursun U, Jordan N, Gu G, Beermann F, Gradwohl G, et al. Temporal control of neurogenin3 activity in pancreas progenitors reveals competence windows for the generation of different endocrine cell types. Dev Cell. 2007; 12:457–65.
64. Schreiber V, Mercier R, Jimenez S, Ye T, Garcia-Sanchez E, Klein A, et al. Extensive NEUROG3 occupancy in the human pancreatic endocrine gene regulatory network. Mol Metab. 2021; 53:101313.
65. Sasaki S, Lee MY, Wakabayashi Y, Suzuki L, Winata H, Himuro M, et al. Spatial and transcriptional heterogeneity of pancreatic beta cell neogenesis revealed by a time-resolved reporter system. Diabetologia. 2022; 65:811–28.
66. Liu J, Banerjee A, Herring CA, Attalla J, Hu R, Xu Y, et al. Neurog3-independent methylation is the earliest detectable mark distinguishing pancreatic progenitor identity. Dev Cell. 2019; 48:49–63.
67. Sharon N, Chawla R, Mueller J, Vanderhooft J, Whitehorn LJ, Rosenthal B, et al. A peninsular structure coordinates asynchronous differentiation with morphogenesis to generate pancreatic islets. Cell. 2019; 176:790–804.
68. Bakhti M, Scheibner K, Tritschler S, Bastidas-Ponce A, Tarquis-Medina M, Theis FJ, et al. Establishment of a high-resolution 3D modeling system for studying pancreatic epithelial cell biology in vitro. Mol Metab. 2019; 30:16–29.
69. Miyatsuka T, Matsuoka TA, Sasaki S, Kubo F, Shimomura I, Watada H, et al. Chronological analysis with fluorescent timer reveals unique features of newly generated β-cells. Diabetes. 2014; 63:3388–93.
70. Miyatsuka T, Kosaka Y, Kim H, German MS. Neurogenin3 inhibits proliferation in endocrine progenitors by inducing Cdkn1a. Proc Natl Acad Sci U S A. 2011; 108:185–90.
71. Miettinen PJ, Huotari M, Koivisto T, Ustinov J, Palgi J, Rasilainen S, et al. Impaired migration and delayed differentiation of pancreatic islet cells in mice lacking EGF-receptors. Development. 2000; 127:2617–27.
72. Greiner TU, Kesavan G, Stahlberg A, Semb H. Rac1 regulates pancreatic islet morphogenesis. BMC Dev Biol. 2009; 9:2.
73. Gribben C, Lambert C, Messal HA, Hubber EL, Rackham C, Evans I, et al. Ductal Ngn3-expressing progenitors contribute to adult β cell neogenesis in the pancreas. Cell Stem Cell. 2021; 28:2000–8.
74. Fukaishi T, Nakagawa Y, Fukunaka A, Sato T, Hara A, Nakao K, et al. Characterisation of Ppy-lineage cells clarifies the functional heterogeneity of pancreatic beta cells in mice. Diabetologia. 2021; 64:2803–16.
75. Bader E, Migliorini A, Gegg M, Moruzzi N, Gerdes J, Roscioni SS, et al. Identification of proliferative and mature β-cells in the islets of Langerhans. Nature. 2016; 535:430–4.
76. Sachs S, Bastidas-Ponce A, Tritschler S, Bakhti M, Bottcher A, Sanchez-Garrido MA, et al. Targeted pharmacological therapy restores β-cell function for diabetes remission. Nat Metab. 2020; 2:192–209.
77. Avrahami D, Wang YJ, Schug J, Feleke E, Gao L, Liu C, et al. Single-cell transcriptomics of human islet ontogeny defines the molecular basis of β-cell dedifferentiation in T2D. Mol Metab. 2020; 42:101057.
78. Mawla AM, Huising MO. Navigating the depths and avoiding the shallows of pancreatic islet cell transcriptomes. Diabetes. 2019; 68:1380–93.
79. Camunas-Soler J, Dai XQ, Hang Y, Bautista A, Lyon J, Suzuki K, et al. Patch-seq links single-cell transcriptomes to human islet dysfunction in diabetes. Cell Metab. 2020; 31:1017–31.
80. Balboa D, Barsby T, Lithovius V, Saarimaki-Vire J, Omar-Hmeadi M, Dyachok O, et al. Functional, metabolic and transcriptional maturation of human pancreatic islets derived from stem cells. Nat Biotechnol. 2022; 40:1042–55.
81. Balboa D, Saarimaki-Vire J, Borshagovski D, Survila M, Lindholm P, Galli E, et al. Insulin mutations impair beta-cell development in a patient-derived iPSC model of neonatal diabetes. Elife. 2018; 7:e38519.
82. Farack L, Golan M, Egozi A, Dezorella N, Bahar Halpern K, Ben-Moshe S, et al. Transcriptional heterogeneity of beta cells in the intact pancreas. Dev Cell. 2019; 48:115–25.
83. Wojtusciszyn A, Armanet M, Morel P, Berney T, Bosco D. Insulin secretion from human beta cells is heterogeneous and dependent on cell-to-cell contacts. Diabetologia. 2008; 51:1843–52.
84. Almaça J, Liang T, Gaisano HY, Nam HG, Berggren PO, Caicedo A. Spatial and temporal coordination of insulin granule exocytosis in intact human pancreatic islets. Diabetologia. 2015; 58:2810–8.
85. Johnston NR, Mitchell RK, Haythorne E, Pessoa MP, Semplici F, Ferrer J, et al. Beta cell hubs dictate pancreatic islet responses to glucose. Cell Metab. 2016; 24:389–401.
86. Dorrell C, Schug J, Canaday PS, Russ HA, Tarlow BD, Grompe MT, et al. Human islets contain four distinct subtypes of β cells. Nat Commun. 2016; 7:11756.
87. Syed SK, Kauffman AL, Beavers LS, Alston JT, Farb TB, Ficorilli J, et al. Ectonucleotidase NTPDase3 is abundant in pancreatic β-cells and regulates glucose-induced insulin secretion. Am J Physiol Endocrinol Metab. 2013; 305:E1319–26.
88. Saunders DC, Brissova M, Phillips N, Shrestha S, Walker JT, Aramandla R, et al. Ectonucleoside triphosphate diphosphohydrolase-3 antibody targets adult human pancreatic β cells for in vitro and in vivo analysis. Cell Metab. 2019; 29:745–54.
89. Docherty FM, Riemondy KA, Castro-Gutierrez R, Dwulet JM, Shilleh AH, Hansen MS, et al. ENTPD3 marks mature stem cell-derived β-cells formed by self-aggregation in vitro. Diabetes. 2021; 70:2554–67.
90. Yoon JS, Sasaki S, Velghe J, Lee MY, Winata H, Nian C, et al. Calcium-dependent transcriptional changes in human pancreatic islet cells reveal functional diversity in islet cell subtypes. Diabetologia. 2022; 65:1519–33.
91. Xiao X, Chen Z, Shiota C, Prasadan K, Guo P, El-Gohary Y, et al. No evidence for β cell neogenesis in murine adult pancreas. J Clin Invest. 2013; 123:2207–17.
92. Qadir MM, Alvarez-Cubela S, Klein D, van Dijk J, Muniz-Anquela R, Moreno-Hernandez YB, et al. Single-cell resolution analysis of the human pancreatic ductal progenitor cell niche. Proc Natl Acad Sci U S A. 2020; 117:10876–87.
93. Wang D, Wang J, Bai L, Pan H, Feng H, Clevers H, et al. Long-term expansion of pancreatic islet organoids from resident Procr+ progenitors. Cell. 2020; 180:1198–211.
94. Oliver-Krasinski JM, Stoffers DA. On the origin of the beta cell. Genes Dev. 2008; 22:1998–2021.
95. Grapin-Botton A, Constam D. Evolution of the mechanisms and molecular control of endoderm formation. Mech Dev. 2007; 124:253–78.
96. Spence JR, Wells JM. Translational embryology: using embryonic principles to generate pancreatic endocrine cells from embryonic stem cells. Dev Dyn. 2007; 236:3218–27.
97. Kim SK, Hebrok M, Melton DA. Notochord to endoderm signaling is required for pancreas development. Development. 1997; 124:4243–52.
98. Hebrok M, Kim SK, Melton DA. Notochord repression of endodermal Sonic hedgehog permits pancreas development. Genes Dev. 1998; 12:1705–13.
99. Lammert E, Cleaver O, Melton D. Induction of pancreatic differentiation by signals from blood vessels. Science. 2001; 294:564–7.
100. Yoshitomi H, Zaret KS. Endothelial cell interactions initiate dorsal pancreas development by selectively inducing the transcription factor Ptf1a. Development. 2004; 131:807–17.
101. Jacquemin P, Yoshitomi H, Kashima Y, Rousseau GG, Lemaigre FP, Zaret KS. An endothelial-mesenchymal relay pathway regulates early phases of pancreas development. Dev Biol. 2006; 290:189–99.
102. Staels W, Heremans Y, Heimberg H, De Leu N. VEGF-A and blood vessels: a beta cell perspective. Diabetologia. 2019; 62:1961–8.
103. Nikolova G, Jabs N, Konstantinova I, Domogatskaya A, Tryggvason K, Sorokin L, et al. The vascular basement membrane: a niche for insulin gene expression and beta cell proliferation. Dev Cell. 2006; 10:397–405.
104. Golosow N, Grobstein C. Epitheliomesenchymal interaction in pancreatic morphogenesis. Dev Biol. 1962; 4:242–55.
105. Landsman L, Nijagal A, Whitchurch TJ, Vanderlaan RL, Zimmer WE, Mackenzie TC, et al. Pancreatic mesenchyme regulates epithelial organogenesis throughout development. PLoS Biol. 2011; 9:e1001143.
106. Cozzitorto C, Mueller L, Ruzittu S, Mah N, Willnow D, Darrigrand JF, et al. A specialized niche in the pancreatic microenvironment promotes endocrine differentiation. Dev Cell. 2020; 55:150–62.
107. Hecksher-Sorensen J, Watson RP, Lettice LA, Serup P, Eley L, De Angelis C, et al. The splanchnic mesodermal plate directs spleen and pancreatic laterality, and is regulated by Bapx1/Nkx3.2. Development. 2004; 131:4665–75.
108. Zhu C, Zhang Y, Li YE, Lucero J, Behrens MM, Ren B. Joint profiling of histone modifications and transcriptome in single cells from mouse brain. Nat Methods. 2021; 18:283–92.
109. Schoof EM, Furtwangler B, Uresin N, Rapin N, Savickas S, Gentil C, et al. Quantitative single-cell proteomics as a tool to characterize cellular hierarchies. Nat Commun. 2021; 12:3341.
110. Nakatani K, Izumi Y, Hata K, Bamba T. An analytical system for single-cell metabolomics of typical mammalian cells based on highly sensitive nano-liquid chromatography tandem mass spectrometry. Mass Spectrom (Tokyo). 2020; 9:A0080.
111. Replogle JM, Saunders RA, Pogson AN, Hussmann JA, Lenail A, Guna A, et al. Mapping information-rich genotype-phenotype landscapes with genome-scale Perturb-seq. Cell. 2022; 185:2559–75.
Full Text Links
  • DMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr