1
|
Peng G, Mosleh E, Yuhas A, Katada K, Kasinathan D, Cherry C, Golson ML. FOXM1 cooperates with ERα to regulate functional β-cell mass. Am J Physiol Endocrinol Metab 2025; 328:E804-E821. [PMID: 40261794 DOI: 10.1152/ajpendo.00438.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/02/2024] [Accepted: 04/11/2025] [Indexed: 04/24/2025]
Abstract
The transcription factor forkhead box (FOX)M1 regulates β-cell proliferation and insulin secretion. Our previous work demonstrates that expressing a constitutively active form of FOXM1 (FOXM1*) in β-cells increases β-cell function, proliferation, and mass in male mice. However, in contrast to what is observed in males, we demonstrate here that in female mice expression of FOXM1* in β-cells does not affect β-cell proliferation or glucose tolerance. Similarly, FOXM1* transduction of male but not female human islets enhances insulin secretion in response to elevated glucose. We therefore examined the mechanism behind this sexual dimorphism. Estrogen contributes to diabetes susceptibility differences between males and females, and estrogen receptor (ER)α is the primary mediator of β-cell estrogen signaling. Moreover, in breast cancer cells, ERα and FOXM1 work together to drive gene expression. We therefore examined whether FOXM1 and ERα functionally interact in β-cells. FOXM1* rescued elevated fasting glucose, glucose intolerance, and homeostatic model assessment of β-cell function (HOMA-B) in female mice with a β-cell-specific ERα deletion. Furthermore, in the presence of estrogen, the FOXM1 and ERα cistromes exhibit significant overlap in βTC6 β-cells. In addition, FOXM1 and ERα binding sites frequently occur in complex enhancers co-occupied by other islet transcription factors. These data indicate that FOXM1 and nuclear ERα cooperate to regulate β-cell function and suggest a general mechanism contributing to the lower incidence of diabetes observed in women.NEW & NOTEWORTHY Here we investigate why the effects of increasing FOXM1 activity in β-cells observed in male mice are not seen in female mice. ERα likely collaborates with FOXM1 and other transcription factors to enhance gene expression related to β-cell function. Higher estrogen levels in females may contribute to their increased insulin secretion and the more severe consequences of losing transcription factors like FOXM1 in males. Overall, these findings shed light on sex differences in diabetes susceptibility.
Collapse
Affiliation(s)
- Guihong Peng
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Elham Mosleh
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Andrew Yuhas
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Kay Katada
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Devi Kasinathan
- Department of Physiology, Johns Hopkins University, Baltimore, Maryland, United States
| | | | - Maria L Golson
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
2
|
Villalba A, Gitton Y, Aiello V, Toupin M, Mazaud-Guittot S, Chédotal A, Scharfmann R. Imaging Human Pancreatic Endocrinogenesis During Early Prenatal Life. Diabetes 2025; 74:368-375. [PMID: 39602451 PMCID: PMC11842602 DOI: 10.2337/db24-0641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Murine pancreatic endocrinogenesis has been extensively studied, but human data remain scarce due to limited sample availability. Here, we first built a large collection of human embryonic and fetal pancreases covering the first trimester of pregnancy to explore human endocrinogenesis. Using an experimental pipeline combining in toto staining, tissue clearing, and light-sheet fluorescence microscopy, we show that insulin-, glucagon-, and somatostatin-positive cells appear simultaneously at Carnegie stage (CS) 16. This contrasts with rodents, in which glucagon-positive cells appear first, followed by insulin-positive and, finally, somatostatin-positive cells and highlights interspecies differences. We also detected bihormonal endocrine cells in 7 of 9 human pancreases between CS16 and CS18, which were no longer detected at later stages. We observed that cell distribution within human fetal islets resembles adult mouse islets, with a core of β-cells surrounded by α- and δ-cells, differing from a more complex arrangement in adult human islets. This, in connection with the small size of human fetal islets when compared with adult islets, suggests that adult human islets may form by fusion of preexisting islets, in contrast to the mouse fission model. Together, our study provides a detailed and comprehensive description of the spatiotemporal dynamics of human pancreatic endocrinogenesis. ARTICLE HIGHLIGHTS Data on human pancreas development are limited and derived from two-dimensional staining. We overcome this using in toto staining, tissue clearing, and light-sheet imaging. We sought to understand when and where endocrine cells first emerge and how they cluster. First, endocrine cell types appear simultaneously, and early pancreases contain bihormonal cells. There are morphometric differences between fetal and adult islets. We propose a mechanism of adult islet formation by fusion: a new base to reconstitute in vitro islet neogenesis.
Collapse
Affiliation(s)
- Adrian Villalba
- Institut Cochin, CNRS, INSERM, Université Paris Cité, Paris, France
| | - Yorick Gitton
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Virginie Aiello
- Institut Cochin, CNRS, INSERM, Université Paris Cité, Paris, France
| | - Maryne Toupin
- Inserm, EHESP, Institut de Recherche en Santé, Environnement et Travail, UMR_S 1085, Université Rennes, Rennes, France
| | - Séverine Mazaud-Guittot
- Inserm, EHESP, Institut de Recherche en Santé, Environnement et Travail, UMR_S 1085, Université Rennes, Rennes, France
| | - Alain Chédotal
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
- Institut de Pathologie, Groupe Hospitalier Est, Hospices Civils de Lyon, Lyon, France
- MeLiS (Mechanisms in Integrated Life Sciences), CNRS UMR5284, INSERM U1314, University Claude Bernard Lyon 1, Lyon, France
| | | |
Collapse
|
3
|
Andersson-Rolf A, Groot K, Korving J, Begthel H, Hanegraaf MAJ, VanInsberghe M, Salmén F, van den Brink S, Lopez-Iglesias C, Peters PJ, Krueger D, Beumer J, Geurts MH, Alemany A, Gehart H, Carlotti F, de Koning EJP, Chuva de Sousa Lopes SM, van Oudenaarden A, van Es JH, Clevers H. Long-term in vitro expansion of a human fetal pancreas stem cell that generates all three pancreatic cell lineages. Cell 2024; 187:7394-7413.e22. [PMID: 39626658 DOI: 10.1016/j.cell.2024.10.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/18/2024] [Accepted: 10/18/2024] [Indexed: 12/29/2024]
Abstract
The mammalian pancreas consists of three epithelial compartments: the acini and ducts of the exocrine pancreas and the endocrine islets of Langerhans. Murine studies indicate that these three compartments derive from a transient, common pancreatic progenitor. Here, we report derivation of 18 human fetal pancreas organoid (hfPO) lines from gestational weeks 8-17 (8-17 GWs) fetal pancreas samples. Four of these lines, derived from 15 to 16 GWs samples, generate acinar-, ductal-, and endocrine-lineage cells while expanding exponentially for >2 years under optimized culture conditions. Single-cell RNA sequencing identifies rare LGR5+ cells in fetal pancreas and in hfPOs as the root of the developmental hierarchy. These LGR5+ cells share multiple markers with adult gastrointestinal tract stem cells. Organoids derived from single LGR5+ organoid-derived cells recapitulate this tripotency in vitro. We describe a human fetal tripotent stem/progenitor cell capable of long-term expansion in vitro and of generating all three pancreatic cell lineages.
Collapse
Affiliation(s)
- Amanda Andersson-Rolf
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands.
| | - Kelvin Groot
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Jeroen Korving
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Harry Begthel
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Maaike A J Hanegraaf
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Michael VanInsberghe
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Fredrik Salmén
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Stieneke van den Brink
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Carmen Lopez-Iglesias
- The Maastricht Multimodal Molecular Imaging Institute, 6229 ER Maastricht, the Netherlands
| | - Peter J Peters
- The Maastricht Multimodal Molecular Imaging Institute, 6229 ER Maastricht, the Netherlands
| | - Daniel Krueger
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Joep Beumer
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche innovation Centre, 4070 Basel, Switzerland
| | - Maarten H Geurts
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Princess Maxima Centre for Pediatric Oncology, 3584 CS Utrecht, the Netherlands
| | - Anna Alemany
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Helmuth Gehart
- ETH Zurich, Institute of Molecular Health Sciences, 8093 Zürich, Schweiz
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Eelco J P de Koning
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | | | - Alexander van Oudenaarden
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Johan H van Es
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Hans Clevers
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Princess Maxima Centre for Pediatric Oncology, 3584 CS Utrecht, the Netherlands; Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche innovation Centre, 4070 Basel, Switzerland.
| |
Collapse
|
4
|
Villalba A, Gitton Y, Inoue M, Aiello V, Blain R, Toupin M, Mazaud-Guittot S, Rachdi L, Semb H, Chédotal A, Scharfmann R. A 3D atlas of the human developing pancreas to explore progenitor proliferation and differentiation. Diabetologia 2024; 67:1066-1078. [PMID: 38630142 PMCID: PMC11058870 DOI: 10.1007/s00125-024-06143-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/07/2024] [Indexed: 04/30/2024]
Abstract
AIMS/HYPOTHESIS Rodent pancreas development has been described in great detail. On the other hand, there are still gaps in our understanding of the developmental trajectories of pancreatic cells during human ontogenesis. Here, our aim was to map the spatial and chronological dynamics of human pancreatic cell differentiation and proliferation by using 3D imaging of cleared human embryonic and fetal pancreases. METHODS We combined tissue clearing with light-sheet fluorescence imaging in human embryonic and fetal pancreases during the first trimester of pregnancy. In addition, we validated an explant culture system enabling in vitro proliferation of pancreatic progenitors to determine the mitogenic effect of candidate molecules. RESULTS We detected the first insulin-positive cells as early as five post-conceptional weeks, two weeks earlier than previously observed. We observed few insulin-positive clusters at five post-conceptional weeks (mean ± SD 9.25±5.65) with a sharp increase to 11 post-conceptional weeks (4307±152.34). We identified a central niche as the location of onset of the earliest insulin cell production and detected extra-pancreatic loci within the adjacent developing gut. Conversely, proliferating pancreatic progenitors were located in the periphery of the epithelium, suggesting the existence of two separated pancreatic niches for differentiation and proliferation. Additionally, we observed that the proliferation ratio of progenitors ranged between 20% and 30%, while for insulin-positive cells it was 1%. We next unveiled a mitogenic effect of the platelet-derived growth factor AA isoform (PDGFAA) in progenitors acting through the pancreatic mesenchyme by increasing threefold the number of proliferating progenitors. CONCLUSIONS/INTERPRETATION This work presents a first 3D atlas of the human developing pancreas, charting both endocrine and proliferating cells across early development.
Collapse
Affiliation(s)
- Adrian Villalba
- Institut Cochin, CNRS, Inserm, Université Paris Cité, Paris, France
| | - Yorick Gitton
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Megumi Inoue
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Virginie Aiello
- Institut Cochin, CNRS, Inserm, Université Paris Cité, Paris, France
| | - Raphaël Blain
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Maryne Toupin
- Inserm, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, Université Rennes, Rennes, France
| | - Séverine Mazaud-Guittot
- Inserm, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, Université Rennes, Rennes, France
| | - Latif Rachdi
- Institut Cochin, CNRS, Inserm, Université Paris Cité, Paris, France
| | - Henrik Semb
- Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, München, Germany
| | - Alain Chédotal
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France.
- Institut de pathologie, groupe hospitalier Est, hospices civils de Lyon, Lyon, France.
- MeLiS, CNRS UMR5284, Inserm U1314, University Claude Bernard Lyon 1, Lyon, France.
| | | |
Collapse
|
5
|
Rout S, Amirtham SM, Prasad M, Cherian AG, B SR, Sudhakar Y, Prince N. In Vitro Human Fetal Pancreatic Islets to Redefine Pancreatic Research. Cureus 2023; 15:e43244. [PMID: 37692623 PMCID: PMC10491859 DOI: 10.7759/cureus.43244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
BACKGROUND In vitro studies with human fetal islets of different gestational ages (GA) would be a great tool to generate information on the developmental process of the islets as this would help to recontextualize diabetes research and clinical practice. Pancreatic islets from human cadavers and other animal species are extensively researched to explore their suitability for islet transplantation procedure, one of the upcoming treatment strategies for insulin-dependent diabetes mellitus. Although human fetal islets are also considered for islet transplantation, ethical issues and limited knowledge constraints their use. The fetal islets could be explored to address the information lacunae on the maturity process of pancreatic islets and the endocrine-exocrine signaling mechanisms. AIM This study aimed to assess the feasibility of isolating viable islets and study the cytoarchitecture of the fetal pancreas of GA 22-29 weeks, not reported otherwise. METHODOLOGY Pancreas obtained from the aborted fetuses of GA 22-29 weeks were subjected to collagenase digestion and were further cultured to determine the viability in vitro. Parameters assessed were expression of markers for endocrine cell lineages and insulin release to glucose challenge. RESULTS Islets were viable in vitro and islets were shown to maintain cues for post-digestion re-aggregation and expansion in culture. The immunofluorescent staining showed islets of varying sizes, homogenous cell clusters aggregating to form heterogenous cell clusters, otherwise not reported for this GA. On stimulation with different concentrations of glucose (2.8 and 28 mM), the fetal islets in the culture exhibited insulin release, and this response confirmed their viability in vitro. CONCLUSION Our findings showed that viable islets could be isolated and cultured in vitro for further in-depth studies to explore their proliferative potential as well as for the identification of pancreatic progenitors, a good strategy to take forward.
Collapse
Affiliation(s)
- Sipra Rout
- Anatomy, All India Institute of Medical Sciences, Bhubaneswar, IND
| | | | - Mythraeyee Prasad
- Anatomy, Velammal Medical College Hospital and Research Institute, Madurai, IND
| | | | - Sandya Rani B
- Research, Christian Medical College and Hospital, Vellore, IND
| | - Yesudas Sudhakar
- Biochemistry, Christian Medical College and Hospital, Vellore, IND
| | - Neetu Prince
- Physiology, Christian Medical College and Hospital, Vellore, IND
| |
Collapse
|
6
|
Gondek S, Ogledzinski M, Lin W, Milejczyk K, Juengel B, Potter L, Bachul PJ, Basto L, Perea L, Wang LJ, Tibudan M, Witkowska Z, Barth R, Fung J, Witkowski P. Persistence of long-term insulin independence after islet transplantation and two subsequent pregnancies. EUROPEAN JOURNAL OF TRANSLATIONAL AND CLINICAL MEDICINE 2023; 6:9-13. [PMID: 37693941 PMCID: PMC10485871 DOI: 10.31373/ejtcm/166178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Pregnancy increases metabolic demand for insulin and may lead to the exhaustion of intraportally transplanted islets and post-gestational hyperglycemia. To prevent these complications, we implemented preemptive insulin supplementation during two subsequent pregnancies in an insulin-independent islet transplant recipient. This strategy resulted in optimal blood glucose control during the pregnancies, the preservation of the optimal islet graft function and the postpartum maintenance of long-term insulin independence.
Collapse
Affiliation(s)
- Sarah Gondek
- The Transplantation Institute, University of Chicago, USA
| | | | - William Lin
- The Transplantation Institute, University of Chicago, USA
| | | | - Braden Juengel
- The Transplantation Institute, University of Chicago, USA
| | - Lisa Potter
- The Transplantation Institute, University of Chicago, USA
| | - Piotr J Bachul
- The Transplantation Institute, University of Chicago, USA
| | - Lindsay Basto
- The Transplantation Institute, University of Chicago, USA
| | | | - Ling-Jia Wang
- The Transplantation Institute, University of Chicago, USA
| | - Martin Tibudan
- The Transplantation Institute, University of Chicago, USA
| | | | - Rolf Barth
- The Transplantation Institute, University of Chicago, USA
| | - John Fung
- The Transplantation Institute, University of Chicago, USA
| | | |
Collapse
|
7
|
Jiang Y, Wiersch J, Wu W, Qian J, Adama MPR, Wu N, Yang W, Chen C, Zhu L, Prasadan K, Gittes GK, Xiao X. Bone-marrow derived cells do not contribute to new beta-cells in the inflamed pancreas. Front Immunol 2023; 14:1084056. [PMID: 36733483 PMCID: PMC9887320 DOI: 10.3389/fimmu.2023.1084056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/03/2023] [Indexed: 01/18/2023] Open
Abstract
The contribution of bone-marrow derived cells (BMCs) to a newly formed beta-cell population in adults is controversial. Previous studies have only used models of bone marrow transplantation from sex-mismatched donors (or other models of genetic labeling) into recipient animals that had undergone irradiation. This approach suffers from the significant shortcoming of the off-target effects of irradiation. Partial pancreatic duct ligation (PDL) is a mouse model of acute pancreatitis with a modest increase in beta-cell number. However, the possibility that recruited BMCs in the inflamed pancreas may convert into beta-cells has not been examined. Here, we used an irradiation-free model to track the fate of the BMCs from the donor mice. A ROSA-mTmG red fluorescent mouse was surgically joined to an INS1Cre knock-in mouse by parabiosis to establish a mixed circulation. PDL was then performed in the INS1Cre mice 2 weeks after parabiosis, which was one week after establishment of the stable blood chimera. The contribution of red cells from ROSA-mTmG mice to beta-cells in INS1Cre mouse was evaluated based on red fluorescence, while cell fusion was evaluated by the presence of green fluorescence in beta-cells. We did not detect any red or green insulin+ cells in the INS1Cre mice, suggesting that there was no contribution of BMCs to the newly formed beta-cells, either by direct differentiation, or by cell fusion. Thus, the contribution of BMCs to beta-cells in the inflamed pancreas should be minimal, if any.
Collapse
Affiliation(s)
- Yinan Jiang
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - John Wiersch
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Wei Wu
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of General Surgery, Children’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Jieqi Qian
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Ultrasound in Medicine, the Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Maharana Prathap R. Adama
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Nannan Wu
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Weixia Yang
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Congde Chen
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Pediatric Surgery, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lingyan Zhu
- Department of Endocrinology, the First Affiliated Hospital of NanChang University, Nanchang, China
| | - Krishna Prasadan
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - George K. Gittes
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Xiangwei Xiao
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
8
|
Abstract
This review focuses on the human pancreatic islet-including its structure, cell composition, development, function, and dysfunction. After providing a historical timeline of key discoveries about human islets over the past century, we describe new research approaches and technologies that are being used to study human islets and how these are providing insight into human islet physiology and pathophysiology. We also describe changes or adaptations in human islets in response to physiologic challenges such as pregnancy, aging, and insulin resistance and discuss islet changes in human diabetes of many forms. We outline current and future interventions being developed to protect, restore, or replace human islets. The review also highlights unresolved questions about human islets and proposes areas where additional research on human islets is needed.
Collapse
Affiliation(s)
- John T Walker
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Diane C Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marcela Brissova
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
9
|
Lee SY, Yuk HG, Ko SG, Cho SG, Moon GS. Gut Microbiome Prolongs an Inhibitory Effect of Korean Red Ginseng on High-Fat-Diet-Induced Mouse Obesity. Nutrients 2021; 13:nu13030926. [PMID: 33809267 PMCID: PMC7999605 DOI: 10.3390/nu13030926] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 01/14/2023] Open
Abstract
Although the anti-obesity effect of Korean red ginseng (Panax ginseng Meyer) has been revealed, its underlying mechanisms are not clearly understood. Here, we demonstrate an involvement of gut microbiome in the inhibitory effect of Korean red ginseng on high-fat-diet (HFD)-induced mouse obesity, and further provides information on the effects of saponin-containing red ginseng extract (SGE) and saponin-depleted red ginseng extract (GE). Mice were fed with either SGE or GE every third day for one month, and their food intakes, fat weights, plasma glucose, and insulin and leptin levels were measured. Immunofluorescence assays were conducted to measure pancreatic islet size. Stools from the mice were subjected to metagenomic analysis. Both SGE and GE attenuated HFD-induced gain of body weight, reducing HFD-induced increase of food intakes and fat weights. They also reduced HFD-increased plasma glucose, insulin, and leptin levels, decreased both fasting and postprandial glucose concentrations, and improved both insulin resistance and glucose intolerance. Immunofluorescence assays revealed that they blocked HFD-induced increase of pancreatic islet size. Our pyrosequencing of the 16S rRNA gene V3 region from stools revealed that both SGE and GE modulated HFD-altered composition of gut microbiota. Therefore, we conclude that Korean red ginseng inhibits HFD-induced obesity and diabetes by altering gut microbiome.
Collapse
Affiliation(s)
- Seo Yeon Lee
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea; (S.Y.L.); (S.G.K.)
| | - Hyun Gyun Yuk
- Department of Food Science and Technology, Korea National University of Transportation, 61 Daehak-ro, Jeungpyeong, Chungbuk 27909, Korea;
| | - Seong Gyu Ko
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea; (S.Y.L.); (S.G.K.)
| | - Sung-Gook Cho
- Department of Biotechnology, Korea National University of Transportation, 61 Daehak-ro, Jeungpyeong, Chungbuk 27909, Korea
- Correspondence: (S.-G.C.); (G.-S.M.); Tel.: +82-43-820-5254 (S.-G.C.); +82-43-820-5272 (G.-S.M.)
| | - Gi-Seong Moon
- Department of Biotechnology, Korea National University of Transportation, 61 Daehak-ro, Jeungpyeong, Chungbuk 27909, Korea
- Correspondence: (S.-G.C.); (G.-S.M.); Tel.: +82-43-820-5254 (S.-G.C.); +82-43-820-5272 (G.-S.M.)
| |
Collapse
|
10
|
Scharfmann R, Staels W, Albagli O. The supply chain of human pancreatic β cell lines. J Clin Invest 2019; 129:3511-3520. [PMID: 31478912 PMCID: PMC6715382 DOI: 10.1172/jci129484] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Patients with type 1 or type 2 diabetes have an insufficiency in their functional β cell mass. To advance diabetes treatment and to work toward a cure, a better understanding of how to protect the pancreatic β cells against autoimmune or metabolic assaults (e.g., obesity, gestation) will be required. Over the past decades, β cell protection has been extensively investigated in rodents both in vivo and in vitro using isolated islets or rodent β cell lines. Transferring these rodent data to humans has long been challenging, at least partly for technical reasons: primary human islet preparations were scarce and functional human β cell lines were lacking. In 2011, we described a robust protocol of targeted oncogenesis in human fetal pancreas and produced the first functional human β cell line, and in subsequent years additional lines with specific traits. These cell lines are currently used by more than 150 academic and industrial laboratories worldwide. In this Review, we first explain how we developed the human β cell lines and why we think we succeeded where others, despite major efforts, did not. Next, we discuss the use of such functional human β cell lines and share some perspectives on their use to advance diabetes research.
Collapse
Affiliation(s)
- Raphael Scharfmann
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| | - Willem Staels
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels, Belgium
| | - Olivier Albagli
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| |
Collapse
|
11
|
Mesodermal induction of pancreatic fate commitment. Semin Cell Dev Biol 2018; 92:77-88. [PMID: 30142440 DOI: 10.1016/j.semcdb.2018.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/29/2018] [Accepted: 08/20/2018] [Indexed: 12/27/2022]
Abstract
The pancreas is a compound gland comprised of both exocrine acinar and duct cells as well as endocrine islet cells. Most notable amongst the latter are the insulin-synthesizing β-cells, loss or dysfunction of which manifests in diabetes mellitus. All exocrine and endocrine cells derive from multipotent pancreatic progenitor cells arising from the primitive gut epithelium via inductive interactions with adjacent mesodermal tissues. Research in the last two decades has revealed the identity of many of these extrinsic cues and they include signaling molecules used in many other developmental contexts such as retinoic acid, fibroblast growth factors, and members of the TGF-β superfamily. As important as these inductive cues is the absence of other signaling molecules such as hedgehog family members. Much has been learned about the interactions of extrinsic factors with fate regulators intrinsic to the pancreatic endoderm. This new knowledge has had tremendous impact on the development of directed differentiation protocols for converting pluripotent stem cells to β-cells in vitro.
Collapse
|
12
|
Petersen MB, Gonçalves CA, Kim YH, Grapin-Botton A. Recapitulating and Deciphering Human Pancreas Development From Human Pluripotent Stem Cells in a Dish. Curr Top Dev Biol 2018; 129:143-190. [DOI: 10.1016/bs.ctdb.2018.02.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
13
|
Donadel G, Pastore D, Della-Morte D, Capuani B, Lombardo MF, Pacifici F, Bugliani M, Grieco FA, Marchetti P, Lauro D. FGF-2b and h-PL Transform Duct and Non-Endocrine Human Pancreatic Cells into Endocrine Insulin Secreting Cells by Modulating Differentiating Genes. Int J Mol Sci 2017; 18:2234. [PMID: 29068419 PMCID: PMC5713204 DOI: 10.3390/ijms18112234] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 12/12/2022] Open
Abstract
Background: Diabetes mellitus (DM) is a multifactorial disease orphan of a cure. Regenerative medicine has been proposed as novel strategy for DM therapy. Human fibroblast growth factor (FGF)-2b controls β-cell clusters via autocrine action, and human placental lactogen (hPL)-A increases functional β-cells. We hypothesized whether FGF-2b/hPL-A treatment induces β-cell differentiation from ductal/non-endocrine precursor(s) by modulating specific genes expression. Methods: Human pancreatic ductal-cells (PANC-1) and non-endocrine pancreatic cells were treated with FGF-2b plus hPL-A at 500 ng/mL. Cytofluorimetry and Immunofluorescence have been performed to detect expression of endocrine, ductal and acinar markers. Bromodeoxyuridine incorporation and annexin-V quantified cells proliferation and apoptosis. Insulin secretion was assessed by RIA kit, and electron microscopy analyzed islet-like clusters. Results: Increase in PANC-1 duct cells de-differentiation into islet-like aggregates was observed after FGF-2b/hPL-A treatment showing ultrastructure typical of islets-aggregates. These clusters, after stimulation with FGF-2b/hPL-A, had significant (p < 0.05) increase in insulin, C-peptide, pancreatic and duodenal homeobox 1 (PDX-1), Nkx2.2, Nkx6.1, somatostatin, glucagon, and glucose transporter 2 (Glut-2), compared with control cells. Markers of PANC-1 (Cytokeratin-19, MUC-1, CA19-9) were decreased (p < 0.05). These aggregates after treatment with FGF-2b/hPL-A significantly reduced levels of apoptosis. Conclusions: FGF-2b and hPL-A are promising candidates for regenerative therapy in DM by inducing de-differentiation of stem cells modulating pivotal endocrine genes.
Collapse
Affiliation(s)
- Giulia Donadel
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Donatella Pastore
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - David Della-Morte
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy.
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy.
| | - Barbara Capuani
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Marco F Lombardo
- Agenzia regionale per la protezione ambientale (ARPA) Lazio, Sezione di Roma, 00173 Rome, Italy.
| | - Francesca Pacifici
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Marco Bugliani
- Endocrinology and Metabolism of Transplantation, Azienda Ospedaliero-Universitaria (A.O.U.) Pisana, 56126 Pisa, Italy.
| | - Fabio A Grieco
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy.
| | - Piero Marchetti
- Endocrinology and Metabolism of Transplantation, Azienda Ospedaliero-Universitaria (A.O.U.) Pisana, 56126 Pisa, Italy.
| | - Davide Lauro
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy.
| |
Collapse
|
14
|
Iuamoto LR, Franco AS, Suguita FY, Essu FF, Oliveira LT, Kato JM, Torsani MB, Meyer A, Andraus W, Chaib E, D'Albuquerque LAC. Human islet xenotransplantation in rodents: A literature review of experimental model trends. Clinics (Sao Paulo) 2017; 72:238-243. [PMID: 28492724 PMCID: PMC5401612 DOI: 10.6061/clinics/2017(04)08] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/16/2016] [Indexed: 01/19/2023] Open
Abstract
Among the innovations for the treatment of type 1 diabetes, islet transplantation is a less invasive method of treatment, although it is still in development. One of the greatest barriers to this technique is the low number of pancreas donors and the low number of pancreases that are available for transplantation. Rodent models have been chosen in most studies of islet rejection and type 1 diabetes prevention to evaluate the quality and function of isolated human islets and to identify alternative solutions to the problem of islet scarcity. The purpose of this study is to conduct a review of islet xenotransplantation experiments from humans to rodents, to organize and analyze the parameters of these experiments, to describe trends in experimental modeling and to assess the viability of this procedure. In this study, we reviewed recently published research regarding islet xenotransplantation from humans to rodents, and we summarized the findings and organized the relevant data. The included studies were recent reports that involved xenotransplantation using human islets in a rodent model. We excluded the studies that related to isotransplantation, autotransplantation and allotransplantation. A total of 34 studies that related to xenotransplantation were selected for review based on their relevance and current data. Advances in the use of different graft sites may overcome autoimmunity and rejection after transplantation, which may solve the problem of the scarcity of islet donors in patients with type 1 diabetes.
Collapse
Affiliation(s)
- Leandro Ryuchi Iuamoto
- Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| | | | | | | | | | | | | | - Alberto Meyer
- Departamento de Gastroenterologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Wellington Andraus
- Departamento de Gastroenterologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Eleazar Chaib
- Departamento de Gastroenterologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | |
Collapse
|
15
|
Larsen HL, Grapin-Botton A. The molecular and morphogenetic basis of pancreas organogenesis. Semin Cell Dev Biol 2017; 66:51-68. [PMID: 28089869 DOI: 10.1016/j.semcdb.2017.01.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 01/08/2023]
Abstract
The pancreas is an essential endoderm-derived organ that ensures nutrient metabolism via its endocrine and exocrine functions. Here we review the essential processes governing the embryonic and early postnatal development of the pancreas discussing both the mechanisms and molecules controlling progenitor specification, expansion and differentiation. We elaborate on how these processes are orchestrated in space and coordinated with morphogenesis. We draw mainly from experiments conducted in the mouse model but also from investigations in other model organisms, complementing a recent comprehensive review of human pancreas development (Jennings et al., 2015) [1]. The understanding of pancreas development in model organisms provides a framework to interpret how human mutations lead to neonatal diabetes and may contribute to other forms of diabetes and to guide the production of desired pancreatic cell types from pluripotent stem cells for therapeutic purposes.
Collapse
Affiliation(s)
- Hjalte List Larsen
- DanStem, University of Copenhagen, 3 B Blegdamsvej, DK-2200 Copenhagen N, Denmark
| | - Anne Grapin-Botton
- DanStem, University of Copenhagen, 3 B Blegdamsvej, DK-2200 Copenhagen N, Denmark.
| |
Collapse
|
16
|
Scharfmann R, Didiesheim M, Richards P, Chandra V, Oshima M, Albagli O. Mass production of functional human pancreatic β-cells: why and how? Diabetes Obes Metab 2016; 18 Suppl 1:128-36. [PMID: 27615142 DOI: 10.1111/dom.12728] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022]
Abstract
Diabetes (either type 1 or type 2) is due to insufficient functional β-cell mass. Research has, therefore, aimed to discover new ways to maintain or increase either β-cell mass or function. For this purpose, rodents have mainly been used as model systems and a large number of discoveries have been made. Meanwhile, although we have learned that rodent models represent powerful systems to model β-cell development, function and destruction, we realize that there are limitations when attempting to transfer the data to what is occurring in humans. Indeed, while human β-cells share many similarities with rodent β-cells, they also differ on a number of important parameters. In this context, developing ways to study human β-cell development, function and death represents an important challenge. This review will describe recent data on the development and use of convenient sources of human β-cells that should be useful tools to discover new ways to modulate functional β-cell mass in humans.
Collapse
Affiliation(s)
- R Scharfmann
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France.
| | - M Didiesheim
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - P Richards
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - V Chandra
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - M Oshima
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - O Albagli
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| |
Collapse
|
17
|
Pauerstein PT, Park KM, Peiris HS, Wang J, Kim SK. Research Resource: Genetic Labeling of Human Islet Alpha Cells. Mol Endocrinol 2016; 30:248-53. [PMID: 26745668 DOI: 10.1210/me.2015-1220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The 2 most abundant human pancreatic islet cell types are insulin-producing β-cells and glucagon-producing α-cells. Defined cis-regulatory elements from rodent Insulin genes have permitted genetic labeling of human islet β-cells, enabling lineage tracing and generation of human β-cell lines, but analogous elements for genetically labeling human α-cells with high specificity do not yet exist. To identify genetic elements that specifically direct reporter expression to human α-cells, we investigated noncoding sequences adjacent to the human GLUCAGON and ARX genes, which are expressed in islet α-cells. Elements with high evolutionary conservation were cloned into lentiviral vectors to direct fluorescent reporter expression in primary human islets. Based on the specificity of reporter expression for α- and β-cells, we found that rat glucagon promoter was not specific for human α-cells but that addition of human GLUCAGON untranslated region sequences substantially enhanced specificity of labeling in both cultured and transplanted islets to a degree not previously reported, to our knowledge. Specific transgene expression from these cis-regulatory sequences in human α-cells should enable targeted genetic modification and lineage tracing.
Collapse
Affiliation(s)
- Philip T Pauerstein
- Department of Developmental Biology (P.T.P., K.M.P., H.S.P., J.W., S.K.K.) and Howard Hughes Medical Institute (S.K.K.), Stanford University School of Medicine, Stanford, California 94305
| | - Keon Min Park
- Department of Developmental Biology (P.T.P., K.M.P., H.S.P., J.W., S.K.K.) and Howard Hughes Medical Institute (S.K.K.), Stanford University School of Medicine, Stanford, California 94305
| | - Heshan S Peiris
- Department of Developmental Biology (P.T.P., K.M.P., H.S.P., J.W., S.K.K.) and Howard Hughes Medical Institute (S.K.K.), Stanford University School of Medicine, Stanford, California 94305
| | - Jing Wang
- Department of Developmental Biology (P.T.P., K.M.P., H.S.P., J.W., S.K.K.) and Howard Hughes Medical Institute (S.K.K.), Stanford University School of Medicine, Stanford, California 94305
| | - Seung K Kim
- Department of Developmental Biology (P.T.P., K.M.P., H.S.P., J.W., S.K.K.) and Howard Hughes Medical Institute (S.K.K.), Stanford University School of Medicine, Stanford, California 94305
| |
Collapse
|
18
|
Prel A, Caval V, Gayon R, Ravassard P, Duthoit C, Payen E, Maouche-Chretien L, Creneguy A, Nguyen TH, Martin N, Piver E, Sevrain R, Lamouroux L, Leboulch P, Deschaseaux F, Bouillé P, Sensébé L, Pagès JC. Highly efficient in vitro and in vivo delivery of functional RNAs using new versatile MS2-chimeric retrovirus-like particles. Mol Ther Methods Clin Dev 2015; 2:15039. [PMID: 26528487 PMCID: PMC4613645 DOI: 10.1038/mtm.2015.39] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 09/02/2015] [Accepted: 09/03/2015] [Indexed: 12/25/2022]
Abstract
RNA delivery is an attractive strategy to achieve transient gene expression in research projects and in cell- or gene-based therapies. Despite significant efforts investigating vector-directed RNA transfer, there is still a requirement for better efficiency of delivery to primary cells and in vivo. Retroviral platforms drive RNA delivery, yet retrovirus RNA-packaging constraints limit gene transfer to two genome-molecules per viral particle. To improve retroviral transfer, we designed a dimerization-independent MS2-driven RNA packaging system using MS2-Coat-retrovirus chimeras. The engineered chimeric particles promoted effective packaging of several types of RNAs and enabled efficient transfer of biologically active RNAs in various cell types, including human CD34(+) and iPS cells. Systemic injection of high-titer particles led to gene expression in mouse liver and transferring Cre-recombinase mRNA in muscle permitted widespread editing at the ROSA26 locus. We could further show that the VLPs were able to activate an osteoblast differentiation pathway by delivering RUNX2- or DLX5-mRNA into primary human bone-marrow mesenchymal-stem cells. Thus, the novel chimeric MS2-lentiviral particles are a versatile tool for a wide range of applications including cellular-programming or genome-editing.
Collapse
Affiliation(s)
- Anne Prel
- Université François Rabelais de Tours, INSERM UMR 966, Tours, France
- UMR UPS/CNRS 5273, EFS-PM, INSERM U1031, Toulouse, France
| | - Vincent Caval
- Université François Rabelais de Tours, INSERM UMR 966, Tours, France
| | - Régis Gayon
- Vectalys, Bâtiment Canal Biotech 2, Parc Technologique du Canal 3, Toulouse, France
| | - Philippe Ravassard
- Institut du Cerveau et de la Moelle (ICM), Université Pierre et Marie Curie, CNRS UMR7225; INSERM U1127, Biotechnologies and Biothérapies Team, Paris, France
| | - Christine Duthoit
- Vectalys, Bâtiment Canal Biotech 2, Parc Technologique du Canal 3, Toulouse, France
| | - Emmanuel Payen
- CEA/Université Paris Sud (UMR-E 007), Institut of Emerging Diseases and Innovative Therapies (iMETI), CEA de Fontenay aux Roses, Fontenay aux Roses, France
| | - Leila Maouche-Chretien
- CEA/Université Paris Sud (UMR-E 007), Institut of Emerging Diseases and Innovative Therapies (iMETI), CEA de Fontenay aux Roses, Fontenay aux Roses, France
| | - Alison Creneguy
- INSERM UMRS 1064, Centre Hospitalier Universitaire (CHU) Hôtel Dieu, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), Université de Nantes, Nantes, France
| | - Tuan Huy Nguyen
- INSERM UMRS 1064, Centre Hospitalier Universitaire (CHU) Hôtel Dieu, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), Université de Nantes, Nantes, France
| | - Nicolas Martin
- Vectalys, Bâtiment Canal Biotech 2, Parc Technologique du Canal 3, Toulouse, France
| | - Eric Piver
- Université François Rabelais de Tours, INSERM UMR 966, Tours, France
- CHRU de Tours, Laboratoire de biochimie et biologie moléculaire, Tours, France
| | - Raphaël Sevrain
- Vectalys, Bâtiment Canal Biotech 2, Parc Technologique du Canal 3, Toulouse, France
| | - Lucille Lamouroux
- Vectalys, Bâtiment Canal Biotech 2, Parc Technologique du Canal 3, Toulouse, France
| | - Philippe Leboulch
- CEA/Université Paris Sud (UMR-E 007), Institut of Emerging Diseases and Innovative Therapies (iMETI), CEA de Fontenay aux Roses, Fontenay aux Roses, France
| | | | - Pascale Bouillé
- Vectalys, Bâtiment Canal Biotech 2, Parc Technologique du Canal 3, Toulouse, France
| | - Luc Sensébé
- UMR UPS/CNRS 5273, EFS-PM, INSERM U1031, Toulouse, France
| | - Jean-Christophe Pagès
- Université François Rabelais de Tours, INSERM UMR 966, Tours, France
- CHRU de Tours, Laboratoire de biochimie et biologie moléculaire, Tours, France
| |
Collapse
|
19
|
Pauerstein PT, Sugiyama T, Stanley SE, McLean GW, Wang J, Martín MG, Kim SK. Dissecting Human Gene Functions Regulating Islet Development With Targeted Gene Transduction. Diabetes 2015; 64:3037-49. [PMID: 25901096 PMCID: PMC4512220 DOI: 10.2337/db15-0042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 04/09/2015] [Indexed: 01/19/2023]
Abstract
During pancreas development, endocrine precursors and their progeny differentiate, migrate, and cluster to form nascent islets. The transcription factor Neurogenin 3 (Neurog3) is required for islet development in mice, but its role in these dynamic morphogenetic steps has been inferred from fixed tissues. Moreover, little is known about the molecular genetic functions of NEUROG3 in human islet development. We developed methods for gene transduction by viral microinjection in the epithelium of cultured Neurog3-null mutant fetal pancreas, permitting genetic complementation in a developmentally relevant context. In addition, we developed methods for quantitative assessment of live-cell phenotypes in single developing islet cells. Delivery of wild-type NEUROG3 rescued islet differentiation, morphogenesis, and live cell deformation, whereas the patient-derived NEUROG3(R107S) allele partially restored indicators of islet development. NEUROG3(P39X), a previously unreported patient allele, failed to restore islet differentiation or morphogenesis and was indistinguishable from negative controls, suggesting that it is a null mutation. Our systems also permitted genetic suppression analysis and revealed that targets of NEUROG3, including NEUROD1 and RFX6, can partially restore islet development in Neurog3-null mutant mouse pancreata. Thus, advances described here permitted unprecedented assessment of gene functions in regulating crucial dynamic aspects of islet development in the fetal pancreas.
Collapse
Affiliation(s)
- Philip T Pauerstein
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA
| | - Takuya Sugiyama
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA
| | - Susan E Stanley
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA
| | - Graeme W McLean
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA
| | - Jing Wang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA
| | - Martín G Martín
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
20
|
Markovič R, Stožer A, Gosak M, Dolenšek J, Marhl M, Rupnik MS. Progressive glucose stimulation of islet beta cells reveals a transition from segregated to integrated modular functional connectivity patterns. Sci Rep 2015; 5:7845. [PMID: 25598507 PMCID: PMC4297961 DOI: 10.1038/srep07845] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 12/16/2014] [Indexed: 01/11/2023] Open
Abstract
Collective beta cell activity in islets of Langerhans is critical for the supply of insulin within an organism. Even though individual beta cells are intrinsically heterogeneous, the presence of intercellular coupling mechanisms ensures coordinated activity and a well-regulated exocytosis of insulin. In order to get a detailed insight into the functional organization of the syncytium, we applied advanced analytical tools from the realm of complex network theory to uncover the functional connectivity pattern among cells composing the intact islet. The procedure is based on the determination of correlations between long temporal traces obtained from confocal functional multicellular calcium imaging of beta cells stimulated in a stepwise manner with a range of physiological glucose concentrations. Our results revealed that the extracted connectivity networks are sparse for low glucose concentrations, whereas for higher stimulatory levels they become more densely connected. Most importantly, for all ranges of glucose concentration beta cells within the islets form locally clustered functional sub-compartments, thereby indicating that their collective activity profiles exhibit a modular nature. Moreover, we show that the observed non-linear functional relationship between different network metrics and glucose concentration represents a well-balanced setup that parallels physiological insulin release.
Collapse
Affiliation(s)
- Rene Markovič
- Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000 Maribor, Slovenia
| | - Andraž Stožer
- 1] Institute of Physiology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia [2] Centre for Open Innovations and Research, University of Maribor, Slomškov trg 15, 2000 Maribor, Slovenia
| | - Marko Gosak
- 1] Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000 Maribor, Slovenia [2] Centre for Open Innovations and Research, University of Maribor, Slomškov trg 15, 2000 Maribor, Slovenia [3] Faculty of Education, University of Maribor, Koroška cesta 160, 2000 Maribor, Slovenia
| | - Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Marko Marhl
- 1] Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000 Maribor, Slovenia [2] Centre for Open Innovations and Research, University of Maribor, Slomškov trg 15, 2000 Maribor, Slovenia [3] Faculty of Education, University of Maribor, Koroška cesta 160, 2000 Maribor, Slovenia
| | - Marjan Slak Rupnik
- 1] Institute of Physiology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia [2] Centre for Open Innovations and Research, University of Maribor, Slomškov trg 15, 2000 Maribor, Slovenia [3] Institute of Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraβe 17, A-1090 Vienna, Austria
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW We highlight some of the major recent advances in characterizing human pancreas development and endocrine cell differentiation. RECENT FINDINGS Extensive research efforts have helped to define crucial events in the mouse pancreas organogenesis. Information gained from these studies was used to develop human embryonic stem cell (hESC) differentiation protocols with the goal of generating functional glucose-responsive, insulin-producing human β-cells. In spite of remarkable progress in hESC differentiation, current protocols based on mouse developmental biology can produce human β-cells only in vivo. New differentiation markers and recently generated reagents may provide an unprecedented opportunity to develop a high-density expression map of human fetal pancreas and pancreatic islets that could serve as a reference point for in vitro hESC differentiation. SUMMARY Integrating an increased knowledge of human pancreas development into hESC differentiation protocols has the potential to greatly advance our ability to generate functional insulin-producing cells for β-cell replacement therapy.
Collapse
Affiliation(s)
- Fong Cheng Pan
- Department of Cell and Developmental Biology and Vanderbilt University Program in Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marcela Brissova
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
22
|
Proshchina AE, Krivova YS, Barabanov VM, Saveliev SV. Ontogeny of neuro-insular complexes and islets innervation in the human pancreas. Front Endocrinol (Lausanne) 2014; 5:57. [PMID: 24795697 PMCID: PMC4001005 DOI: 10.3389/fendo.2014.00057] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 04/08/2014] [Indexed: 11/13/2022] Open
Abstract
The ontogeny of the neuro-insular complexes (NIC) and the islets innervation in human pancreas has not been studied in detail. Our aim was to describe the developmental dynamics and distribution of the nervous system structures in the endocrine part of human pancreas. We used double-staining with antibodies specific to pan-neural markers [neuron-specific enolase (NSE) and S100 protein] and to hormones of pancreatic endocrine cells. NSE and S100-positive nerves and ganglia were identified in the human fetal pancreas from gestation week (gw) 10 onward. Later the density of S100 and NSE-positive fibers increased. In adults, this network was sparse. The islets innervation started to form from gw 14. NSE-containing endocrine cells were identified from gw 12 onward. Additionally, S100-positive cells were detected both in the periphery and within some of the islets starting at gw 14. The analysis of islets innervation has shown that the fetal pancreas contained NIC and the number of these complexes was reduced in adults. The highest density of NIC is detected during middle and late fetal periods, when the mosaic islets, typical for adults, form. The close integration between the developing pancreatic islets and the nervous system structures may play an important role not only in the hormone secretion, but also in the islets morphogenesis.
Collapse
Affiliation(s)
- Alexandra E. Proshchina
- Laboratory of Nervous System Development, Scientific Research Institute of Human Morphology, Department of Medical Biological Sciences, Russian Academy of Medical Science, Moscow, Russia
- *Correspondence: Alexandra E. Proshchina, Laboratory of Nervous System Development, Scientific Research Institute of Human Morphology, Department of Medical Biological Sciences, Russian Academy of Medical Science, Tsurupi Street 3, Moscow 117418, Russia e-mail:
| | - Yulia S. Krivova
- Laboratory of Nervous System Development, Scientific Research Institute of Human Morphology, Department of Medical Biological Sciences, Russian Academy of Medical Science, Moscow, Russia
| | - Valeriy M. Barabanov
- Laboratory of Nervous System Development, Scientific Research Institute of Human Morphology, Department of Medical Biological Sciences, Russian Academy of Medical Science, Moscow, Russia
| | - Sergey V. Saveliev
- Laboratory of Nervous System Development, Scientific Research Institute of Human Morphology, Department of Medical Biological Sciences, Russian Academy of Medical Science, Moscow, Russia
| |
Collapse
|
23
|
Revascularization of transplanted pancreatic islets and role of the transplantation site. Clin Dev Immunol 2013; 2013:352315. [PMID: 24106517 PMCID: PMC3782812 DOI: 10.1155/2013/352315] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 08/09/2013] [Indexed: 12/16/2022]
Abstract
Since the initial reporting of the successful reversal of hyperglycemia through the transplantation of pancreatic islets, significant research efforts have been conducted in elucidating the process of revascularization and the influence of engraftment site on graft function and survival. During the isolation process the intrinsic islet vascular networks are destroyed, leading to impaired revascularization after transplant. As a result, in some cases a significant quantity of the beta cell mass transplanted dies acutely following the infusion into the portal vein, the most clinically used site of engraftment. Subsequently, despite the majority of patients achieving insulin independence after transplant, a proportion of them recommence small, supplemental exogenous insulin over time. Herein, this review considers the process of islet revascularization after transplant, its limiting factors, and potential strategies to improve this critical step. Furthermore, we provide a characterization of alternative transplant sites, analyzing the historical evolution and their role towards advancing transplant outcomes in both the experimental and clinical settings.
Collapse
|
24
|
Xiao X, Guo P, Shiota C, Prasadan K, El-Gohary Y, Wiersch J, Gaffar I, Gittes GK. Neurogenin3 activation is not sufficient to direct duct-to-beta cell transdifferentiation in the adult pancreas. J Biol Chem 2013; 288:25297-25308. [PMID: 23867457 PMCID: PMC3757194 DOI: 10.1074/jbc.m113.484022] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 07/12/2013] [Indexed: 12/25/2022] Open
Abstract
It remains controversial whether adult pancreatic ducts harbor facultative beta cell progenitors. Because neurogenin3 (Ngn3) is a key determinant of pancreatic endocrine cell neogenesis during embryogenesis, many studies have also relied upon Ngn3 expression as evidence of beta cell neogenesis in adults. Recently, however, Ngn3 as a marker of adult beta cell neogenesis has been called into question by reports of Ngn3 expression in fully-developed beta cells. Nevertheless, direct evidence as to whether Ngn3 activation in adult pancreatic duct cells may lead to duct-to-beta cell transdifferentiation is lacking. Here we studied two models of Ngn3 activation in adult pancreatic duct cells (low-dose alloxan treatment and pancreatic duct ligation) and lineage-traced Ngn3-activated duct cells by labeling them through intraductal infusion with a cell-tagging dye, CFDA-SE No dye-labeled beta cells were found during the follow-up in either model, suggesting that activation of Ngn3 in duct cells is not sufficient to direct their transdifferentiation into beta cells. Therefore, Ngn3 activation in duct cells is not a signature for adult beta cell neogenesis.
Collapse
Affiliation(s)
- Xiangwei Xiao
- From the Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224.
| | - Ping Guo
- From the Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Chiyo Shiota
- From the Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Krishna Prasadan
- From the Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Yousef El-Gohary
- From the Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - John Wiersch
- From the Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Iljana Gaffar
- From the Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - George K Gittes
- From the Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224.
| |
Collapse
|
25
|
Wei R, Yang J, Hou W, Liu G, Gao M, Zhang L, Wang H, Mao G, Gao H, Chen G, Hong T. Insulin-producing cells derived from human embryonic stem cells: comparison of definitive endoderm- and nestin-positive progenitor-based differentiation strategies. PLoS One 2013; 8:e72513. [PMID: 23951327 PMCID: PMC3741181 DOI: 10.1371/journal.pone.0072513] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 07/16/2013] [Indexed: 12/26/2022] Open
Abstract
Human embryonic stem cells (hESCs) are pluripotent and capable of undergoing multilineage differentiation into highly specialized cells including pancreatic islet cells. Thus, they represent a novel alternative source for targeted therapies and regenerative medicine for diabetes. Significant progress has been made in differentiating hESCs toward pancreatic lineages. One approach is based on the similarities of pancreatic β cell and neuroepithelial development. Nestin-positive cells are selected as pancreatic β cell precursors and further differentiated to secrete insulin. The other approach is based on our knowledge of developmental biology in which the differentiation protocol sequentially reproduces the individual steps that are known in normal β cell ontogenesis during fetal pancreatic development. In the present study, the hESC cell line PKU1.1 was induced to differentiate into insulin-producing cells (IPCs) using both protocols. The differentiation process was dynamically investigated and the similarities and differences between both strategies were explored. Our results show that IPCs can be successfully induced with both differentiation strategies. The resulting IPCs from both protocols shared many similar features with pancreatic islet cells, but not mature, functional β cells. However, these differently-derived IPC cell types displayed specific morphologies and different expression levels of pancreatic islet development-related markers. These data not only broaden our outlook on hESC differentiation into IPCs, but also extend the full potential of these processes for regenerative medicine in diabetes.
Collapse
Affiliation(s)
- Rui Wei
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Haidian District, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Ninov N, Hesselson D, Gut P, Zhou A, Fidelin K, Stainier DYR. Metabolic regulation of cellular plasticity in the pancreas. Curr Biol 2013; 23:1242-50. [PMID: 23791726 DOI: 10.1016/j.cub.2013.05.037] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 03/21/2013] [Accepted: 05/21/2013] [Indexed: 12/27/2022]
Abstract
Obese individuals exhibit an increase in pancreatic β cell mass; conversely, scarce nutrition during pregnancy has been linked to β cell insufficiency in the offspring [reviewed in 1, 2]. These phenomena are thought to be mediated mainly through effects on β cell proliferation, given that a nutrient-sensitive β cell progenitor population in the pancreas has not been identified. Here, we employed the fluorescent ubiquitination-based cell-cycle indicator system to investigate β cell replication in real time and found that high nutrient concentrations induce rapid β cell proliferation. Importantly, we found that high nutrient concentrations also stimulate β cell differentiation from progenitors in the intrapancreatic duct (IPD). Furthermore, using a new zebrafish line where β cells are constitutively ablated, we show that β cell loss and high nutrient intake synergistically activate these progenitors. At the cellular level, this activation process causes ductal cell reorganization as it stimulates their proliferation and differentiation. Notably, we link the nutrient-dependent activation of these progenitors to a downregulation of Notch signaling specifically within the IPD. Furthermore, we show that the nutrient sensor mechanistic target of rapamycin (mTOR) is required for endocrine differentiation from the IPD under physiological conditions as well as in the diabetic state. Thus, this study reveals critical insights into how cells modulate their plasticity in response to metabolic cues and identifies nutrient-sensitive progenitors in the mature pancreas.
Collapse
Affiliation(s)
- Nikolay Ninov
- Department of Biochemistry and Biophysics, Programs in Developmental and Stem Cell Biology, Genetics and Human Genetics, the Diabetes Center, Institute for Regeneration Medicine and Liver Center, University of California, San Francisco, 1550 4(th) Street, San Francisco, CA 94158, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Scharfmann R, Rachdi L, Ravassard P. Concise review: in search of unlimited sources of functional human pancreatic beta cells. Stem Cells Transl Med 2012; 2:61-7. [PMID: 23283495 DOI: 10.5966/sctm.2012-0120] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
It is well-established that insulin-producing pancreatic beta cells are central in diabetes. In type 1 diabetes, beta cells are destroyed by an autoimmune mechanism, whereas in type 2 diabetes, there is a decrease in functional beta-cell mass. In this context, studying beta cells is of major importance. Beta cells represent only 1% of total pancreatic cells and are found dispersed in the pancreatic gland. During the past decades, many tools and approaches have been developed to study rodent beta cells that efficiently pushed the field forward. However, rodent and human beta cells are not identical, and our knowledge of human beta cells has not progressed as quickly as our understanding of rodent beta cells. We believe that one of the reasons for this inefficient progress is the difficulty of accessing unlimited sources of functional human pancreatic beta cells. The main focus of this review concerns recent strategies to generate new sources of human pancreatic beta cells.
Collapse
|
28
|
Morán I, Akerman İ, van de Bunt M, Xie R, Benazra M, Nammo T, Arnes L, Nakić N, García-Hurtado J, Rodríguez-Seguí S, Pasquali L, Sauty-Colace C, Beucher A, Scharfmann R, van Arensbergen J, Johnson PR, Berry A, Lee C, Harkins T, Gmyr V, Pattou F, Kerr-Conte J, Piemonti L, Berney T, Hanley NA, Gloyn AL, Sussel L, Langman L, Brayman KL, Sander M, McCarthy MI, Ravassard P, Ferrer J. Human β cell transcriptome analysis uncovers lncRNAs that are tissue-specific, dynamically regulated, and abnormally expressed in type 2 diabetes. Cell Metab 2012; 16:435-448. [PMID: 23040067 PMCID: PMC3475176 DOI: 10.1016/j.cmet.2012.08.010] [Citation(s) in RCA: 351] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 07/30/2012] [Accepted: 08/31/2012] [Indexed: 02/08/2023]
Abstract
A significant portion of the genome is transcribed as long noncoding RNAs (lncRNAs), several of which are known to control gene expression. The repertoire and regulation of lncRNAs in disease-relevant tissues, however, has not been systematically explored. We report a comprehensive strand-specific transcriptome map of human pancreatic islets and β cells, and uncover >1100 intergenic and antisense islet-cell lncRNA genes. We find islet lncRNAs that are dynamically regulated and show that they are an integral component of the β cell differentiation and maturation program. We sequenced the mouse islet transcriptome and identify lncRNA orthologs that are regulated like their human counterparts. Depletion of HI-LNC25, a β cell-specific lncRNA, downregulated GLIS3 mRNA, thus exemplifying a gene regulatory function of islet lncRNAs. Finally, selected islet lncRNAs were dysregulated in type 2 diabetes or mapped to genetic loci underlying diabetes susceptibility. These findings reveal a new class of islet-cell genes relevant to β cell programming and diabetes pathophysiology.
Collapse
Affiliation(s)
- Ignasi Morán
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - İldem Akerman
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Martijn van de Bunt
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
| | - Ruiyu Xie
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California, USA
| | - Marion Benazra
- Centre de recherche de l’institut du cerveau et de la moelle, Biotechnology & Biotherapy team, CNRS UMR7225; INSERM U975; University Pierre et Marie Curie, Paris, France
| | - Takao Nammo
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Department of Metabolic Disorders, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
| | - Luis Arnes
- Department of Genetics and Development, Russ Berrie Medical Pavilion, Columbia University, New York, USA
| | - Nikolina Nakić
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Javier García-Hurtado
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Santiago Rodríguez-Seguí
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Lorenzo Pasquali
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Claire Sauty-Colace
- Centre de recherche de l’institut du cerveau et de la moelle, Biotechnology & Biotherapy team, CNRS UMR7225; INSERM U975; University Pierre et Marie Curie, Paris, France
| | - Anthony Beucher
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Raphael Scharfmann
- Institut National de la Santé et de la Recherche Médicale (INSERM) U845, Research Center Growth and Signalling, Paris Descartes University, Sorbonne Paris Cité, Necker Hospital, Paris, France
| | - Joris van Arensbergen
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Paul R Johnson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
- Oxford Islet Transplant Programme, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Andrew Berry
- Developmental Biomedicine Research Group, School of Biomedicine, Manchester Academic Health Sciences Centre, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Clarence Lee
- Genome Sequencing Collaborations Group, Life Technologies, Beverly, Massachusetts USA
| | - Timothy Harkins
- Genome Sequencing Collaborations Group, Life Technologies, Beverly, Massachusetts USA
| | - Valery Gmyr
- University of Lille Nord de France, INSERM U859 Biotherapies of Diabete, Lille, France
| | - François Pattou
- University of Lille Nord de France, INSERM U859 Biotherapies of Diabete, Lille, France
| | - Julie Kerr-Conte
- University of Lille Nord de France, INSERM U859 Biotherapies of Diabete, Lille, France
| | - Lorenzo Piemonti
- Diabetes research institute (HSR-DRI), San Raffaele Scientific Institute, Milano, Italy
| | - Thierry Berney
- Cell Isolation and Transplantation Center, Geneva, Switzerland
| | - Neil A Hanley
- Developmental Biomedicine Research Group, School of Biomedicine, Manchester Academic Health Sciences Centre, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Anna L Gloyn
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Lori Sussel
- Department of Genetics and Development, Russ Berrie Medical Pavilion, Columbia University, New York, USA
| | - Linda Langman
- Division of Transplantation, Department of Surgery, Center for Cellular Therapy and Biotherapeutics, University of Virginia, USA
| | - Kenneth L Brayman
- Division of Transplantation, Department of Surgery, Center for Cellular Therapy and Biotherapeutics, University of Virginia, USA
| | - Maike Sander
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California, USA
| | - Mark I. McCarthy
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
- Diabetes research institute (HSR-DRI), San Raffaele Scientific Institute, Milano, Italy
| | - Philippe Ravassard
- Centre de recherche de l’institut du cerveau et de la moelle, Biotechnology & Biotherapy team, CNRS UMR7225; INSERM U975; University Pierre et Marie Curie, Paris, France
| | - Jorge Ferrer
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Department of Endocrinology and Nutrition, Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
29
|
Ravassard P, Czernichow P, Scharfmann R. [First immortalised cell lines phenotypically and functionally equivalent to human insulin-secreting pancreatic beta cells]. Med Sci (Paris) 2012; 28:149-51. [PMID: 22377301 DOI: 10.1051/medsci/2012282011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
30
|
|
31
|
Riedel MJ, Asadi A, Wang R, Ao Z, Warnock GL, Kieffer TJ. Immunohistochemical characterisation of cells co-producing insulin and glucagon in the developing human pancreas. Diabetologia 2012; 55:372-81. [PMID: 22038519 DOI: 10.1007/s00125-011-2344-9] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 08/30/2011] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS In adult human islets, insulin and glucagon production is largely restricted to individual cell populations. The production of these hormones is less segregated during development and during the differentiation of human pluripotent stem cells towards pancreatic lineages. We therefore sought to characterise the transcription factor profile of these cells that co-produce insulin and glucagon in the developing human pancreas, and thus to gain insight into their potential fate during normal pancreas development. METHODS An immunohistochemical analysis was performed on human pancreas sections from fetal donors aged 9 to 21 weeks and from adult donors between the ages of 17 and 55 years. RESULTS Endocrine cells were observed within the pancreas at all ages examined, with cells co-producing insulin and glucagon observed as early as 9 weeks of fetal age. The population of cells that co-produce insulin and glucagon generally decreased in prevalence with age, with negligible numbers in adult pancreas. From 9 to 16 weeks, the population of glucagon-only cells increased, while the insulin-only cells decreased in abundance. Cells that co-produced insulin and glucagon also produced the alpha cell transcription factor, aristaless related homeobox (ARX), and lacked the beta cell transcription factors pancreatic and duodenal homeobox 1 (PDX1), NK6 homeobox 1 (NKX6.1) and v-maf musculoaponeurotic fibrosarcoma oncogene homologue A (MAFA). CONCLUSIONS/INTERPRETATION Our results indicate that cells co-producing insulin and glucagon in the developing human pancreas share a transcription factor profile that is similar to that of mature alpha cells and suggest that some maturing alpha cells briefly exhibit ectopic insulin expression. Thus cells that co-produce insulin and glucagon may represent a transient cell population, which gives rise to mature alpha cells.
Collapse
Affiliation(s)
- M J Riedel
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada V6T 1Z3
| | | | | | | | | | | |
Collapse
|
32
|
Ravassard P, Hazhouz Y, Pechberty S, Bricout-Neveu E, Armanet M, Czernichow P, Scharfmann R. A genetically engineered human pancreatic β cell line exhibiting glucose-inducible insulin secretion. J Clin Invest 2011; 121:3589-97. [PMID: 21865645 DOI: 10.1172/jci58447] [Citation(s) in RCA: 451] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 06/15/2011] [Indexed: 12/12/2022] Open
Abstract
Despite intense efforts over the past 30 years, human pancreatic β cell lines have not been available. Here, we describe a robust technology for producing a functional human β cell line using targeted oncogenesis in human fetal tissue. Human fetal pancreatic buds were transduced with a lentiviral vector that expressed SV40LT under the control of the insulin promoter. The transduced buds were then grafted into SCID mice so that they could develop into mature pancreatic tissue. Upon differentiation, the newly formed SV40LT-expressing β cells proliferated and formed insulinomas. The resulting β cells were then transduced with human telomerase reverse transcriptase (hTERT), grafted into other SCID mice, and finally expanded in vitro to generate cell lines. One of these cell lines, EndoC-βH1, expressed many β cell-specific markers without any substantial expression of markers of other pancreatic cell types. The cells secreted insulin when stimulated by glucose or other insulin secretagogues, and cell transplantation reversed chemically induced diabetes in mice. These cells represent a unique tool for large-scale drug discovery and provide a preclinical model for cell replacement therapy in diabetes. This technology could be generalized to generate other human cell lines when the cell type-specific promoter is available.
Collapse
Affiliation(s)
- Philippe Ravassard
- Université Pierre et Marie Curie-Paris 6, Biotechnology and Biotherapy Team, Centre de Recherche de l’Institut du Cerveau et de la Moelle épinière (CRICM), UMRS 975, Paris, France.
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Pancreas oganogenesis comprises a coordinated and highly complex interplay of signaling events and transcriptional networks that guide a step-wise process of organ development from early bud specification all the way to the final mature organ state. Extensive research on pancreas development over the last few years, largely driven by a translational potential for pancreatic diseases (diabetes, pancreatic cancer, and so on), is markedly advancing our knowledge of these processes. It is a tenable goal that we will one day have a clear, complete picture of the transcriptional and signaling codes that control the entire organogenetic process, allowing us to apply this knowledge in a therapeutic context, by generating replacement cells in vitro, or perhaps one day to the whole organ in vivo. This review summarizes findings in the past 5 years that we feel are amongst the most significant in contributing to the deeper understanding of pancreas development. Rather than try to cover all aspects comprehensively, we have chosen to highlight interesting new concepts, and to discuss provocatively some of the more controversial findings or proposals. At the end of the review, we include a perspective section on how the whole pancreas differentiation process might be able to be unwound in a regulated fashion, or redirected, and suggest linkages to the possible reprogramming of other pancreatic cell-types in vivo, and to the optimization of the forward-directed-differentiation of human embryonic stem cells (hESC), or induced pluripotential cells (iPSC), towards mature β-cells.
Collapse
|
34
|
Liew CG. Generation of insulin-producing cells from pluripotent stem cells: from the selection of cell sources to the optimization of protocols. Rev Diabet Stud 2010; 7:82-92. [PMID: 21060967 DOI: 10.1900/rds.2010.7.82] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The pancreas arises from Pdx1-expressing progenitors in developing foregut endoderm in early embryo. Expression of Ngn3 and NeuroD1 commits the cells to form endocrine pancreas, and to differentiate into subsets of cells that constitute islets of Langerhans. β-cells in the islets transcribe gene-encoding insulin, and subsequently process and secrete insulin, in response to circulating glucose. Dysfunction of β-cells has profound metabolic consequences leading to hyperglycemia and diabetes mellitus. β-cells are destroyed via autoimmune reaction in type 1 diabetes (T1D). Type 2 diabetes (T2D), characterized by impaired β-cell functions and reduced insulin sensitivity, accounts for 90% of all diabetic patients. Islet transplantation is a promising treatment for T1D. Pluripotent stem cells provide an unlimited cell source to generate new β-cells for patients with T1D. Furthermore, derivation of induced pluripotent stem cells (iPSCs) from patients captures "disease-in-a-dish" for autologous cell replacement therapy, disease modeling, and drug screening for both types of diabetes. This review highlights essential steps in pancreas development, and potential stem cell applications in cell regeneration therapy for diabetes mellitus.
Collapse
Affiliation(s)
- Chee-Gee Liew
- UCR Stem Cell Center, University of California, Riverside, CA 92521, USA.
| |
Collapse
|
35
|
Abstract
Over the last years, there has been great success in driving stem cells toward insulin-expressing cells. However, the protocols developed to date have some limitations, such as low reliability and low insulin production. The most successful protocols used for generation of insulin-producing cells from stem cells mimic in vitro pancreatic organogenesis by directing the stem cells through stages that resemble several pancreatic developmental stages. Islet cell fate is coordinated by a complex network of inductive signals and regulatory transcription factors that, in a combinatorial way, determine pancreatic organ specification, differentiation, growth, and lineage. Together, these signals and factors direct the progression from multipotent progenitor cells to mature pancreatic cells. Later in development and adult life, several of these factors also contribute to maintain the differentiated phenotype of islet cells. A detailed understanding of the processes that operate in the pancreas during embryogenesis will help us to develop a suitable source of cells for diabetes therapy. In this chapter, we will discuss the main transcription factors involved in pancreas specification and beta-cell formation.
Collapse
|
36
|
Desgraz R, Herrera PL. Pancreatic neurogenin 3-expressing cells are unipotent islet precursors. Development 2009; 136:3567-74. [PMID: 19793886 DOI: 10.1242/dev.039214] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pancreatic islet endocrine cells arise during development from precursors expressing neurogenin 3 (Ngn3). As a population, Ngn3(+) cells produce all islet cell types, but the potential of individual Ngn3(+) cells, an issue central to organogenesis in general and to in vitro differentiation towards cell-based therapies, has not been addressed. We performed in vivo clonal analyses in mice to study the proliferation and differentiation of very large numbers of single Ngn3(+) cells using MADM, a genetic system in which a Cre-dependent chromosomal translocation labels, at extremely low mosaic efficiency, a small number of Ngn3(+) cells. We scored large numbers of progeny arising from single Ngn3(+) cells. In newborns, labeled islets frequently contained just a single tagged endocrine cell, indicating for the first time that each Ngn3(+) cell is the precursor of a single endocrine cell. In adults, small clusters of two to three Ngn3(+) progeny were detected, but all expressed the same hormone, indicating a low rate of replication from birth to adult stages. We propose a model whereby Ngn3(+) cells are monotypic (i.e. unipotent) precursors, and use this paradigm to refocus ideas on how cell number and type must be regulated in building complete islets of Langerhans.
Collapse
Affiliation(s)
- Renaud Desgraz
- Department of Genetic Medicine and Development, University of Geneva Medical School, 1 rue Michel-Servet, 1211 Geneva-4, Switzerland
| | | |
Collapse
|
37
|
A new strategy to generate functional insulin-producing cell lines by somatic gene transfer into pancreatic progenitors. PLoS One 2009; 4:e4731. [PMID: 19266046 PMCID: PMC2649535 DOI: 10.1371/journal.pone.0004731] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Accepted: 02/04/2009] [Indexed: 01/25/2023] Open
Abstract
Background There is increasing interest in developing human cell lines to be used to better understand cell biology, but also for drug screening, toxicology analysis and future cell therapy. In the endocrine pancreatic field, functional human beta cell lines are extremely scarce. On the other hand, rodent insulin producing beta cells have been generated during the past years with great success. Many of such cell lines were produced by using transgenic mice expressing SV40T antigen under the control of the insulin promoter, an approach clearly inadequate in human. Our objective was to develop and validate in rodent an alternative transgenic-like approach, applicable to human tissue, by performing somatic gene transfer into pancreatic progenitors that will develop into beta cells. Methods and Findings In this study, rat embryonic pancreases were transduced with recombinant lentiviral vector expressing the SV40T antigen under the control of the insulin promoter. Transduced tissues were next transplanted under the kidney capsule of immuno-incompetent mice allowing insulinoma development from which beta cell lines were established. Gene expression profile, insulin content and glucose dependent secretion, normalization of glycemia upon transplantation into diabetic mice validated the approach to generate beta cell lines. Conclusions Somatic gene transfer into pancreatic progenitors represents an alternative strategy to generate functional beta cell lines in rodent. Moreover, this approach can be generalized to derive cells lines from various tissues and most importantly from tissues of human origin.
Collapse
|