1
|
Jiang T, Wei F, Xie K. Clinical significance of pancreatic ductal metaplasia. J Pathol 2022; 257:125-139. [PMID: 35170758 DOI: 10.1002/path.5883] [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: 11/21/2021] [Revised: 02/06/2022] [Accepted: 02/14/2022] [Indexed: 11/08/2022]
Abstract
Pancreatic ductal metaplasia (PDM) is the stepwise replacement of differentiated somatic cells with ductal or ductal-like cells in the pancreas. PDM is usually triggered by cellular and environmental insults. PDM development may involve all cell lineages of the pancreas, and acinar cells with the highest plasticity are the major source of PDM. Pancreatic progenitor cells are also involved as cells of origin or transitional intermediates. PDM is heterogeneous at the histological, cellular, and molecular levels and only certain subsets of PDM develop further into pancreatic intraepithelial neoplasia (PanIN) and then pancreatic ductal adenocarcinoma (PDAC). The formation and evolution of PDM is regulated at the cellular and molecular levels through a complex network of signaling pathways. The key molecular mechanisms that drive PDM formation and its progression into PanIN/PDAC remain unclear, but represent key targets for reversing or inhibiting PDM. Alternatively, PDM could be a source of pancreas regeneration, including both exocrine and endocrine components. Cellular aging and apoptosis are obstacles to PDM-to-PanIN progression or pancreas regeneration. Functional identification of the cellular and molecular events driving senescence and apoptosis in PDM and its progression would help not only to restrict the development of PDM into PanIN/PDAC, but may also facilitate pancreatic regeneration. This review systematically assesses recent advances in the understanding of PDM physiology and pathology, with a focus on its implications for enhancing regeneration and prevention of cancer. © 2022 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Tingting Jiang
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, PR China
- Department of Pathology, The South China University of Technology School of Medicine, Guangzhou, PR China
| | - Fang Wei
- Institute of Digestive Diseases Research, The South China University of Technology School of Medicine, Guangzhou, PR China
| | - Keping Xie
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, PR China
- Department of Pathology, The South China University of Technology School of Medicine, Guangzhou, PR China
| |
Collapse
|
2
|
Li X, He J, Xie K. Molecular signaling in pancreatic ductal metaplasia: emerging biomarkers for detection and intervention of early pancreatic cancer. Cell Oncol (Dordr) 2022; 45:201-225. [PMID: 35290607 DOI: 10.1007/s13402-022-00664-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2022] [Indexed: 11/27/2022] Open
Abstract
Pancreatic ductal metaplasia (PDM) is the transformation of potentially various types of cells in the pancreas into ductal or ductal-like cells, which eventually replace the existing differentiated somatic cell type(s). PDM is usually triggered by and manifests its ability to adapt to environmental stimuli and genetic insults. The development of PDM to atypical hyperplasia or dysplasia is an important risk factor for pancreatic intraepithelial neoplasia (PanIN) and pancreatic ductal adenocarcinoma (PDA). Recent studies using genetically engineered mouse models, cell lineage tracing, single-cell sequencing and others have unraveled novel cellular and molecular insights in PDM formation and evolution. Those novel findings help better understand the cellular origins and functional significance of PDM and its regulation at cellular and molecular levels. Given that PDM represents the earliest pathological changes in PDA initiation and development, translational studies are beginning to define PDM-associated cell and molecular biomarkers that can be used to screen and detect early PDA and to enable its effective intervention, thereby truly and significantly reducing the dreadful mortality rate of PDA. This review will describe recent advances in the understanding of PDM biology with a focus on its underlying cellular and molecular mechanisms, and in biomarker discovery with clinical implications for the management of pancreatic regeneration and tumorigenesis.
Collapse
Affiliation(s)
- Xiaojia Li
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, 510006, China
- Department of Pathology, The South China University of Technology School of Medicine, Guangzhou, China
| | - Jie He
- Institute of Digestive Diseases Research, The South China University of Technology School of Medicine, Guangzhou, China
| | - Keping Xie
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, 510006, China.
- Department of Pathology, The South China University of Technology School of Medicine, Guangzhou, China.
| |
Collapse
|
3
|
Li S, Xie K. Ductal metaplasia in pancreas. Biochim Biophys Acta Rev Cancer 2022; 1877:188698. [DOI: 10.1016/j.bbcan.2022.188698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 02/07/2023]
|
4
|
Nano E, Petropavlovskaia M, Rosenberg L. Islet neogenesis associated protein (INGAP) protects pancreatic β cells from IL-1β and IFNγ-induced apoptosis. Cell Death Discov 2021; 7:56. [PMID: 33731692 PMCID: PMC7969959 DOI: 10.1038/s41420-021-00441-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/06/2021] [Accepted: 02/14/2021] [Indexed: 11/11/2022] Open
Abstract
The goal of this study was to determine whether recombinant Islet NeoGenesis Associated Protein (rINGAP) and its active core, a pentadecapeptide INGAP104-118 (Ingap-p), protect β cells against cytokine-induced death. INGAP has been shown to induce islet neogenesis in diabetic animals, to stimulate β-cell proliferation and differentiation, and to improve islet survival and function. Importantly, Ingap-p has shown promising results in clinical trials for diabetes (phase I/II). However, the full potential of INGAP and its mechanisms of action remain poorly understood. Using rat insulinoma cells RINm5F and INS-1 treated with interleukin-1β (IL-1β) and interferon-gamma (IFN-γ), we demonstrate here that both rINGAP and Ingap-p inhibit apoptosis, Caspase-3 activation, inducible nitric oxide synthase (iNOS) expression and nitric oxide (NO) production, and explore the related signaling pathways. As expected, IL-1β induced nuclear factor kappa B (NF-κB), p38, and JNK signaling, whereas interferon-gamma (IFN-γ) activated the JAK2/STAT1 pathway and potentiated the IL-1β effects. Both rINGAP and Ingap-p decreased phosphorylation of IKKα/β, IkBα, and p65, although p65 nuclear translocation was not inhibited. rINGAP, used for further analysis, also inhibited STAT3, p38, and JNK activation. Interestingly, all inhibitory effects of rINGAP were observed for the cytokine cocktail, not IL-1β alone, and were roughly equal to reversing the potentiating effects of INFγ. Furthermore, rINGAP had no effect on IL-1β/NF-κB-induced gene expression (e.g., Ccl2, Sod2) but downregulated several IFNγ-stimulated (Irf1, Socs1, Socs3) or IFNγ-potentiated (Nos2) genes. This, however, was observed again only for the cytokine cocktail, not IFNγ alone, and rINGAP did not inhibit the IFNγ-induced JAK2/STAT1 activation. Together, these intriguing results suggest that INGAP does not target either IL-1β or IFNγ individually but rather inhibits the signaling crosstalk between the two, the exact mechanism of which remains to be investigated. In summary, our study characterizes the anti-inflammatory effects of INGAP, both protein and peptide, and suggests a new therapeutic utility for INGAP in the treatment of diabetes.
Collapse
Affiliation(s)
- Eni Nano
- Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, Department of Surgery, Faculty of Medicine, McGill University, 3755, Cote Ste-Catherine Rd, Montreal, QC, H3T 1E2, Canada
| | - Maria Petropavlovskaia
- Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, Department of Surgery, Faculty of Medicine, McGill University, 3755, Cote Ste-Catherine Rd, Montreal, QC, H3T 1E2, Canada.
| | - Lawrence Rosenberg
- Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, Department of Surgery, Faculty of Medicine, McGill University, 3755, Cote Ste-Catherine Rd, Montreal, QC, H3T 1E2, Canada
| |
Collapse
|
5
|
Gilloteaux J. Primary cilia in the Syrian hamster biliary tract: Bile flow antennae and outlooks about signaling on the hepato-biliary-pancreatic stem cells. TRANSLATIONAL RESEARCH IN ANATOMY 2020. [DOI: 10.1016/j.tria.2020.100063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
6
|
Kowalska M, Rupik W. Development of the duct system during exocrine pancreas differentiation in the grass snakeNatrix natrix(Lepidosauria, Serpentes). J Morphol 2018; 279:724-746. [DOI: 10.1002/jmor.20806] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/25/2018] [Accepted: 02/06/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Magdalena Kowalska
- Department of Animal Histology and Embryology; University of Silesia; Katowice Poland
| | - Weronika Rupik
- Department of Animal Histology and Embryology; University of Silesia; Katowice Poland
| |
Collapse
|
7
|
Heileman KL, Daoud J, Tabrizian M. Elaboration of a finite element model of pancreatic islet dielectric response to gap junction expression and insulin release. Colloids Surf B Biointerfaces 2016; 148:474-480. [PMID: 27665380 DOI: 10.1016/j.colsurfb.2016.09.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 09/07/2016] [Accepted: 09/08/2016] [Indexed: 12/23/2022]
Abstract
Dielectric spectroscopy could potentially be a powerful tool to monitor isolated human pancreatic islets for applications in diabetes therapy and research. Isolated intact human islets provide the most relevant means to understand the cellular and molecular mechanisms associated with diabetes. The advantages of dielectric spectroscopy for continuous islet monitoring are that it is a non-invasive, inexpensive and real-time technique. We have previously assessed the dielectric response of human islet samples during stimulation and differentiation. Because of the complex geometry of islets, analytical solutions are not sufficiently representative to provide a pertinent model of islet dielectric response. Here, we present a finite element dielectric model of a single intact islet that takes into account the tight packing of islet cells and intercellular junctions. The simulation yielded dielectric spectra characteristic of cell aggregates, similar to those produced with islets. In addition, the simulation showed that both exocytosis, such as what occurs during insulin secretion, and differential gap junction expression have significant effects on islet dielectric response. Since the progression of diabetes has some connections with dysfunctional islet gap junctions and insulin secretion, the ability to monitor these islet features with dielectric spectroscopy would benefit diabetes research.
Collapse
Affiliation(s)
| | | | - Maryam Tabrizian
- Biomedical Engineering Department, Canada; Faculty of Dentistry, McGill University, Montreal, Quebec H3A 2B4, Canada.
| |
Collapse
|
8
|
Daoud J, Heileman K, Shapka S, Rosenberg L, Tabrizian M. Dielectric spectroscopy for monitoring human pancreatic islet differentiation within cell-seeded scaffolds in a perfusion bioreactor system. Analyst 2016; 140:6295-305. [PMID: 26280028 DOI: 10.1039/c5an00525f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The long-term in vitro culture and differentiation of human pancreatic islets is still hindered by the inability to emulate a suitable microenvironment mimicking physiological extracellular matrix (ECM) support and nutrient/oxygen perfusion. This is further amplified by the current lack of a non-invasive and rapid monitoring system to readily evaluate cellular processes. In this study, we realized a viable method for non-invasively monitoring isolated human pancreatic islets in vitro. Islets are induced to dedifferentiate into proliferative duct-like structures (DLS) in preparation for potential and subsequent re-differentiation into functional islet-like structures (ILS) in a process reminiscent of islet regeneration strategies. This long-term in vitro process is conducted within a three-dimensional microenvironment involving islets embedded in an optimized ECM gel supported by microfabricated three-dimensional scaffolds. The islet-scaffold is then housed and continuously perfused within chambers of a bioreactor platform. The process in its entirety is monitored through dielectric spectroscopy measurements, yielding an accurate representation of cellular morphology, functionality, and volume fraction. This non-invasive and real-time monitoring tool can be further manipulated to elucidate important information about the optimized cellular microenvironment required for maintaining long-term culture and achieve efficient differentiation for islet regeneration.
Collapse
Affiliation(s)
- J Daoud
- Biomedical Engineering Department, McGill University, 3775 University Street, Montreal, Quebec H3A 2B4, Canada.
| | | | | | | | | |
Collapse
|
9
|
Beamish CA, Strutt BJ, Arany EJ, Hill DJ. Insulin-positive, Glut2-low cells present within mouse pancreas exhibit lineage plasticity and are enriched within extra-islet endocrine cell clusters. Islets 2016; 8:65-82. [PMID: 27010375 PMCID: PMC4987018 DOI: 10.1080/19382014.2016.1162367] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 02/25/2016] [Accepted: 03/01/2016] [Indexed: 01/01/2023] Open
Abstract
Regeneration of insulin-producing β-cells from resident pancreas progenitors requires an understanding of both progenitor identity and lineage plasticity. One model suggested that a rare β-cell sub-population within islets demonstrated multi-lineage plasticity. We hypothesized that β-cells from young mice (postnatal day 7, P7) exhibit such plasticity and used a model of islet dedifferentiation toward a ductal epithelial-cell phenotype to test this theory. RIPCre;Z/AP(+/+) mice were used to lineage trace the fate of β-cells during dedifferentiation culture by a human placental alkaline phosphatase (HPAP) reporter. There was a significant loss of HPAP-expressing β-cells in culture, but remaining HPAP(+) cells lost insulin expression while gaining expression of the epithelial duct cell marker cytokeratin-19 (Ck19). Flow cytometry and recovery of β-cell subpopulations from whole pancreas vs. islets suggest that the HPAP(+)Ck19(+) cells had derived from insulin-positive, glucose-transporter-2-low (Ins(+)Glut2(LO)) cells, representing 3.5% of all insulin-expressing cells. The majority of these cells were found outside of islets within clusters of <5 β-cells. These insulin(+)Glut2(LO) cells demonstrated a greater proliferation rate in vivo and in vitro as compared to insulin(+)Glut2(+) cells at P7, were retained into adulthood, and a subset differentiated into endocrine, ductal, and neural lineages, illustrating substantial plasticity. Results were confirmed using RIPCre;ROSA- eYFP mice. Quantitative PCR data indicated these cells possess an immature β-cell phenotype. These Ins(+)Glut2(LO) cells may represent a resident population of cells capable of forming new, functional β-cells, and which may be potentially exploited for regenerative therapies in the future.
Collapse
Affiliation(s)
- Christine A. Beamish
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
| | - Brenda J. Strutt
- Department of Medicine, Western University, London, ON, Canada
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
| | - Edith J. Arany
- Department of Medicine, Western University, London, ON, Canada
- Department of Pathology, Western University, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
| | - David J. Hill
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Department of Medicine, Western University, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
- Lawson Health Research Institute, St Joseph Health Care, London, ON, Canada
| |
Collapse
|
10
|
Castiello FR, Heileman K, Tabrizian M. Microfluidic perfusion systems for secretion fingerprint analysis of pancreatic islets: applications, challenges and opportunities. LAB ON A CHIP 2016; 16:409-31. [PMID: 26732665 DOI: 10.1039/c5lc01046b] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
A secretome signature is a heterogeneous profile of secretions present in a single cell type. From the secretome signature a smaller panel of proteins, namely a secretion fingerprint, can be chosen to feasibly monitor specific cellular activity. Based on a thorough appraisal of the literature, this review explores the possibility of defining and using a secretion fingerprint to gauge the functionality of pancreatic islets of Langerhans. It covers the state of the art regarding microfluidic perfusion systems used in pancreatic islet research. Candidate analytical tools to be integrated within microfluidic perfusion systems for dynamic secretory fingerprint monitoring were identified. These analytical tools include patch clamp, amperometry/voltametry, impedance spectroscopy, field effect transistors and surface plasmon resonance. Coupled with these tools, microfluidic devices can ultimately find applications in determining islet quality for transplantation, islet regeneration and drug screening of therapeutic agents for the treatment of diabetes.
Collapse
Affiliation(s)
- F Rafael Castiello
- Biomedical Engineering Department, McGill University, Montreal, QC H3A 2B4, Canada.
| | - Khalil Heileman
- Biomedical Engineering Department, McGill University, Montreal, QC H3A 2B4, Canada.
| | - Maryam Tabrizian
- Biomedical Engineering Department, McGill University, Montreal, QC H3A 2B4, Canada.
| |
Collapse
|
11
|
Regnault B, Melanitou E. Genetic architecture of early pre-inflammatory stage transcription signatures of autoimmune diabetes in the pancreatic lymph nodes of the NOD mouse reveals significant gene enrichment on chromosomes 6 and 7. Meta Gene 2015; 6:96-104. [PMID: 26629415 PMCID: PMC4634356 DOI: 10.1016/j.mgene.2015.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/11/2015] [Accepted: 09/18/2015] [Indexed: 12/01/2022] Open
Abstract
Autoimmune diseases are characterized by the stimulation of an excessive immune response to self-tissues by inner and/or outer organism factors. Common characteristics in their etiology include a complex genetic predisposition and environmental triggers as well as the implication of the major histocompatibility (MHC) locus on human chromosome 6p21. A restraint number of non-MHC susceptibility genes, part of the genetic component of type 1 diabetes have been identified in human and in animal models, while the complete spectrum of genes involved remains unknown. We elaborate herein patterns of chromosomal organization of 162 genes differentially expressed in the pancreatic lymph nodes of Non-Obese Diabetic mice, carefully selected by early sub-phenotypic evaluation (presence or absence of insulin autoantibodies). Chromosomal assignment of these genes revealed a non-random distribution on five chromosomes (47%). Significant gene enrichment was observed in particular for two chromosomes, 6 and 7. While a subset of these genes coding for secreted proteins showed significant enrichment on both chromosomes, the overall pool of genes was significantly enriched on chromosome 7. The significance of this unexpected gene distribution on the mouse genome is discussed in the light of novel findings indicating that genes affecting common diseases map to recombination “hotspot” regions of mammalian genomes. The genetic architecture of transcripts differentially expressed in specific stages of autoimmune diabetes offers novel venues towards our understanding of patterns of inheritance potentially affecting the pathological disease mechanisms. Chromosomal organization of differentially expressed genes in early autoimmunity Non-random distribution of type 1 diabetes-related transcripts on the mouse genome High density of genetic polymorphisms on specific loci of chromosomes 6 and 7 Correlation of mapping positions on chromosomes 6 and 7 with known T1D loci Our study offers candidate genes and loci drawn in the genomics of T1D initiation.
Collapse
Affiliation(s)
- Beatrice Regnault
- Department of Genomes and Genetics, Eukaryote Genotyping Platform, Institut Pasteur, 25-28 rue du Dr Roux, 75725 Paris, Cedex 15, France
| | - Evie Melanitou
- Department of Parasites and Insect-Vectors, Institut Pasteur, 25-28 rue du Dr Roux, 75725 Paris, Cedex 15, France
| |
Collapse
|
12
|
Islet Neogenesis Associated Protein (INGAP) induces the differentiation of an adult human pancreatic ductal cell line into insulin-expressing cells through stepwise activation of key transcription factors for embryonic beta cell development. Differentiation 2015; 90:77-90. [DOI: 10.1016/j.diff.2015.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 10/13/2015] [Accepted: 10/22/2015] [Indexed: 01/13/2023]
|
13
|
Role of Islet Glucokinase, Glucose Metabolism, and Insulin Pathway in the Enhancing Effect of Islet Neogenesis-Associated Protein on Glucose-Induced Insulin Secretion. Pancreas 2015; 44:959-66. [PMID: 25906449 DOI: 10.1097/mpa.0000000000000341] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To demonstrate the role of islet glucokinase, glucose metabolism, and intracellular insulin mediators in the enhancing effect of islet neogenesis-associated protein pentadecapeptide (INGAP-PP) on glucose-induced insulin secretion. METHODS Islets from normal rats were cultured for 4 days in the absence or presence of 10 μg/mL INGAP-PP, with/without Wortmannin or LY294002. Islets were incubated with different glucose concentrations to measure insulin secretion and content, hexokinase and glucokinase activity, glucose oxidation and utilization, glucokinase, insulin receptor, insulin receptor substrate (IRS)-1/2, and PI3K concentration and phosphorylation. RESULTS The INGAP-PP significantly increased insulin release at high but not at low glucose concentration, glucokinase activity, glucose metabolism, glucokinase, insulin receptor, IRS-2 and PI3K protein concentration, insulin receptor and IRS-1/2 tyrosine phosphorylation, and the association of p85 with IRS-1. Wortmannin and LY294002 blocked INGAP-PP effect on insulin secretion and glucokinase protein levels in a dose-dependent manner. CONCLUSIONS The enhancing effect of INGAP-PP on glucose-induced insulin release could be partly ascribed to its effect on glucokinase activity and glucose metabolism and is mainly mediated by the PI3K/AKT pathway. These results, together with the low hypoglycemia risk associated with the use of INGAP-PP, offer a new alternative for diabetes prevention and treatment.
Collapse
|
14
|
Hoang DT, Matsunari H, Nagaya M, Nagashima H, Millis JM, Witkowski P, Periwal V, Hara M, Jo J. A conserved rule for pancreatic islet organization. PLoS One 2014; 9:e110384. [PMID: 25350558 PMCID: PMC4211668 DOI: 10.1371/journal.pone.0110384] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/12/2014] [Indexed: 11/18/2022] Open
Abstract
Morphogenesis, spontaneous formation of organism structure, is essential for life. In the pancreas, endocrine , , and cells are clustered to form islets of Langerhans, the critical micro-organ for glucose homeostasis. The spatial organization of endocrine cells in islets looks different between species. Based on the three-dimensional positions of individual cells in islets, we computationally inferred the relative attractions between cell types, and found that the attractions between homotypic cells were slightly, but significantly, stronger than the attractions between heterotypic cells commonly in mouse, pig, and human islets. The difference between cell attraction and cell attraction was minimal in human islets, maximizing the plasticity of islet structures. Our result suggests that although the cellular composition and attractions of pancreatic endocrine cells are quantitatively different between species, the physical mechanism of islet morphogenesis may be evolutionarily conserved.
Collapse
Affiliation(s)
- Danh-Tai Hoang
- Asia Pacific Center for Theoretical Physics, Pohang, Korea
| | - Hitomi Matsunari
- Meiji University International Institute for Bio-Resource Research, Kanagawa, Japan
| | - Masaki Nagaya
- Meiji University International Institute for Bio-Resource Research, Kanagawa, Japan
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Kanagawa, Japan
| | - J. Michael Millis
- Department of Surgery, The University of Chicago, Chicago, IL, United States of America
| | - Piotr Witkowski
- Department of Surgery, The University of Chicago, Chicago, IL, United States of America
| | - Vipul Periwal
- Laboratory of Biological Modeling, NIDDK, NIH, Bethesda, MD, United States of America
| | - Manami Hara
- Department of Medicine, The University of Chicago, Chicago, IL, United States of America
- * E-mail: (MH); (JJ)
| | - Junghyo Jo
- Asia Pacific Center for Theoretical Physics, Pohang, Korea
- Department of Physics, POSTECH, Pohang, Korea
- * E-mail: (MH); (JJ)
| |
Collapse
|
15
|
Abstract
OBJECTIVE The aim of this study was to characterize cells expressing insulin and amylase in adult human pancreas. METHODS We applied serial section and immunohistochemistry to pancreas samples from 5 adult nondiabetic subjects (2 men and 3 women;mean age, 65.8 years; random plasma glucose level, 5.1 mM). Cells expressing insulin and amylase were captured by immunofluorescence and confocal miscroscopy. RESULTS We found a widespread occurrence of insulin-producing cells in exocrine acini and amylase-reactive acinar cells in well-formed islets. The insulin-producing cells in exocrine acini predominantly formed single and double cell units though cell clusters, and islands occurred. Acini containing insulin-producing cells outnumbered the islets with a factor of approximately 5. Confocal microscopy and double immunostaining identified acinar A-cells coexpressing both amylase and insulin. CONCLUSIONS The acinar A-cells represent a distinct category of pancreatic cell populations and might be possible endogenous progenitors of insulin-producing cells in normal and abnormal metabolic homeostasis.
Collapse
|
16
|
Cerf ME. Beta cell dynamics: beta cell replenishment, beta cell compensation and diabetes. Endocrine 2013; 44:303-11. [PMID: 23483434 DOI: 10.1007/s12020-013-9917-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 03/01/2013] [Indexed: 12/19/2022]
Abstract
Type 2 diabetes, characterized by persistent hyperglycemia, arises mostly from beta cell dysfunction and insulin resistance and remains a highly complex metabolic disease due to various stages in its pathogenesis. Glucose homeostasis is primarily regulated by insulin secretion from the beta cells in response to prevailing glycemia. Beta cell populations are dynamic as they respond to fluctuating insulin demand. Beta cell replenishment and death primarily regulate beta cell populations. Beta cells, pancreatic cells, and extra-pancreatic cells represent the three tiers for replenishing beta cells. In rodents, beta cell self-replenishment appears to be the dominant source for new beta cells supported by pancreatic cells (non-beta islet cells, acinar cells, and duct cells) and extra-pancreatic cells (liver, neural, and stem/progenitor cells). In humans, beta cell neogenesis from non-beta cells appears to be the dominant source of beta cell replenishment as limited beta cell self-replenishment occurs particularly in adulthood. Metabolic states of increased insulin demand trigger increased insulin synthesis and secretion from beta cells. Beta cells, therefore, adapt to support their physiology. Maintaining physiological beta cell populations is a strategy for targeting metabolic states of persistently increased insulin demand as in diabetes.
Collapse
Affiliation(s)
- Marlon E Cerf
- Diabetes Discovery Platform, South African Medical Research, PO Box 19070, Tygerberg, 7505, South Africa,
| |
Collapse
|
17
|
Petropavlovskaia M, Daoud J, Zhu J, Moosavi M, Ding J, Makhlin J, Assouline-Thomas B, Rosenberg L. Mechanisms of action of islet neogenesis-associated protein: comparison of the full-length recombinant protein and a bioactive peptide. Am J Physiol Endocrinol Metab 2012; 303:E917-27. [PMID: 22850686 PMCID: PMC3469614 DOI: 10.1152/ajpendo.00670.2011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Islet neogenesis-associated protein (INGAP) was discovered in the partially duct-obstructed hamster pancreas as a factor inducing formation of new duct-associated islets. A bioactive portion of INGAP, INGAP(104-118) peptide (INGAP-P), has been shown to have neogenic and insulin-potentiating activity in numerous studies, including recent phase 2 clinical trials that demonstrated improved glucose homeostasis in both type 1 and type 2 diabetic patients. Aiming to improve INGAP-P efficacy and to understand its mechanism of action, we cloned the full-length protein (rINGAP) and compared the signaling events induced by the protein and the peptide in RIN-m5F cells that respond to INGAP with an increase in proliferation. Here, we show that, although both rINGAP and INGAP-P signal via the Ras/Raf/ERK pathway, rINGAP is at least 100 times more efficient on a molar basis than INGAP-P. For either ligand, ERK1/2 activation appears to be pertussis toxin sensitive, suggesting involvement of a G protein-coupled receptor(s). However, there are clear differences between the peptide and the protein in interactions with the cell surface and in the downstream signaling. We demonstrate that fluorescent-labeled rINGAP is characterized by clustering on the membrane and by slow internalization (≤5 h), whereas INGAP-P does not cluster and is internalized within minutes. Signaling by rINGAP appears to involve Src, in contrast to INGAP-P, which appears to activate Akt in addition to the Ras/Raf/ERK1/2 pathway. Thus our data suggest that interactions of INGAP with the cell surface are important to consider for further development of INGAP as a pharmacotherapy for diabetes.
Collapse
Affiliation(s)
- Maria Petropavlovskaia
- Department of Surgery, the Research Institute of the McGill University Health Center, McGill University, Montreal, Québec, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Daoud J, Asami K, Rosenberg L, Tabrizian M. Dielectric spectroscopy for non-invasive monitoring of epithelial cell differentiation within three-dimensional scaffolds. Phys Med Biol 2012; 57:5097-112. [DOI: 10.1088/0031-9155/57/16/5097] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
19
|
Montanucci P, Pennoni I, Pescara T, Blasi P, Bistoni G, Basta G, Calafiore R. The functional performance of microencapsulated human pancreatic islet-derived precursor cells. Biomaterials 2011; 32:9254-62. [DOI: 10.1016/j.biomaterials.2011.08.052] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 08/16/2011] [Indexed: 11/16/2022]
|
20
|
Hanley SC, Assouline-Thomas B, Makhlin J, Rosenberg L. Epidermal growth factor induces adult human islet cell dedifferentiation. J Endocrinol 2011; 211:231-9. [PMID: 21933872 DOI: 10.1530/joe-11-0213] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Given the inherent therapeutic potential of the morphogenetic plasticity of adult human islets, the identification of factors controlling their cellular differentiation is of interest. The epidermal growth factor (EGF) family has been identified previously in the context of pancreatic organogenesis. We examined the role of EGF in an in vitro model whereby adult human islets are embedded in a collagen gel and dedifferentiated into duct-like epithelial structures (DLS). We demonstrated that DLS formation was EGF dependent, while residual DLS formation in the absence of added EGF was abrogated by EGF receptor inhibitor treatment. With respect to signaling, EGF administration led to an increase in c-Jun NH2-terminal kinase (JNK) phosphorylation early in DLS formation and in AKT and extracellular signal-regulated kinase (ERK) phosphorylation late in the process of DLS formation, concomitant with the increased proliferation of dedifferentiated cells. In the absence of EGF, these phosphorylation changes are not seen and the typical increase in DLS epithelial cell proliferation seen after 10 days in culture is attenuated. Thus, in our model, EGF is necessary for islet cell dedifferentiation, playing an important role in both the onset of DLS formation (through JNK) and in the proliferation of these dedifferentiated cells (through AKT and ERK).
Collapse
Affiliation(s)
- Stephen C Hanley
- Department of Surgery, McGill University, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
21
|
Parikh A, Stephan AF, Tzanakakis ES. Regenerating proteins and their expression, regulation and signaling. Biomol Concepts 2011; 3:57-70. [PMID: 22582090 DOI: 10.1515/bmc.2011.055] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The regenerating (Reg) protein family comprises C-type lectin-like proteins discovered independently during pancreatitis and pancreatic islet regeneration. However, an increasing number of studies provide evidence of participation of Reg proteins in the proliferation and differentiation of diverse cell types. Moreover, Reg family members are associated with various pathologies, including diabetes and forms of gastrointestinal cancer. These findings have led to the emergence of key roles for Reg proteins as anti-inflammatory, antiapoptotic and mitogenic agents in multiple physiologic and disease contexts. Yet, there are significant gaps in our knowledge regarding the regulation of expression of different Reg genes. In addition, the pathways relaying Reg-triggered signals, their targets and potential cross-talk with other cascades are still largely unknown. In this review, the expression patterns of different Reg members in the pancreas and extrapancreatic tissues are described. Moreover, factors known to modulate Reg levels in different cell types are discussed. Several signaling pathways, which have been implicated in conferring the effects of Reg ligands to date, are also delineated. Further efforts are necessary for elucidating the biological processes underlying the action of Reg proteins and their involvement in various maladies. Better understanding of the function of Reg genes and proteins will be beneficial in the design and development of therapies utilizing or targeting this protein group.
Collapse
Affiliation(s)
- Abhirath Parikh
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, Buffalo, NY 14260
| | | | | |
Collapse
|
22
|
β-Cell Generation: Can Rodent Studies Be Translated to Humans? J Transplant 2011; 2011:892453. [PMID: 22007286 PMCID: PMC3189575 DOI: 10.1155/2011/892453] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 07/31/2011] [Accepted: 07/31/2011] [Indexed: 12/26/2022] Open
Abstract
β-cell replacement by allogeneic islet transplantation is a promising approach for patients with type 1 diabetes, but the shortage of organ donors requires new sources of β cells. Islet regeneration in vivo and generation of β-cells ex vivo followed by transplantation represent attractive therapeutic alternatives to restore the β-cell mass. In this paper, we discuss different postnatal cell types that have been envisaged as potential sources for future β-cell replacement therapy. The ultimate goal being translation to the clinic, a particular attention is given to the discrepancies between findings from studies performed in rodents (both ex vivo on primary cells and in vivo on animal models), when compared with clinical data and studies performed on human cells.
Collapse
|
23
|
Daoud J, Rosenberg L, Tabrizian M. Pancreatic Islet Culture and Preservation Strategies: Advances, Challenges, and Future Outlook. Cell Transplant 2010; 19:1523-35. [DOI: 10.3727/096368910x515872] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Postisolation islet survival is a critical step for achieving successful and efficient islet transplantation. This involves the optimization of islet culture in order to prolong survival and functionality in vitro. Many studies have focused on different strategies to culture pancreatic islets in vitro through manipulation of culture media, surface modified substrates, and the use of various techniques such as encapsulation, embedding, scaffold, and bioreactor culture strategies. This review aims to present and discuss the different methodologies employed to optimize pancreatic islet culture in vitro as well as address their respective advantages and drawbacks.
Collapse
Affiliation(s)
- Jamal Daoud
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Lawrence Rosenberg
- Department of Surgery, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
24
|
Liu L, Liu JL, Srikant CB. Reg2 protects mouse insulinoma cells from streptozotocin-induced mitochondrial disruption and apoptosis. Growth Factors 2010; 28:370-8. [PMID: 20919961 DOI: 10.3109/08977194.2010.504721] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We reported previously that pancreas-specific ablation of IGF-I in mice induced an increased expression of regenerating family proteins Reg2 and Reg3β in the pancreas and protected them from streptozotocin (Stz)-induced β-cell damage. We, therefore, assessed the effect of ectopically introduced Reg2 on Stz-induced apoptosis in MIN6 mouse insulinoma cells and report here that Reg2 protects MIN6 cells from Stz-induced apoptosis by attenuating its ability to disrupt mitochondrial membrane integrity, activate caspase-3 and promote poly-ADP ribose polymerase cleavage, and induce apoptosis. These changes correlated with suppression of c-jun N-terminal kinase (JNK) phosphorylation by Stz. Reg2 inhibited Stz-induced proapoptotic events as well as the inactivation of JNK. Inclusion of chemical inhibitor of JNK to Reg2 expressing cells rendered them sensitive to Stz. These data demonstrate that Reg2 protects insulin-producing cells against Stz-induced apoptosis by interfering with its cytotoxic signaling upstream of the intrinsic proapoptotic events by preventing its ability to inactivate JNK.
Collapse
Affiliation(s)
- Lu Liu
- Fraser Laboratories, McGill University Health Centre and Royal Victoria Hospital, Montreal, Quebec, Canada H3A 1A1
| | | | | |
Collapse
|
25
|
Hanley SC, Austin E, Assouline-Thomas B, Kapeluto J, Blaichman J, Moosavi M, Petropavlovskaia M, Rosenberg L. {beta}-Cell mass dynamics and islet cell plasticity in human type 2 diabetes. Endocrinology 2010; 151:1462-72. [PMID: 20176718 DOI: 10.1210/en.2009-1277] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Studies of long-standing type 2 diabetes (T2D) report a deficit in beta-cell mass due to increased apoptosis, whereas neogenesis and replication are unaffected. It is unclear whether these changes are a cause or a consequence of T2D. Moreover, whereas islet morphogenetic plasticity has been demonstrated in vitro, the in situ plasticity of islets, as well as the effect of T2D on endocrine differentiation, is unknown. We compared beta-cell volume, neogenesis, replication, and apoptosis in pancreata from lean and obese (body mass index > or = 27 kg/m(2)) diabetic (5 +/- 2 yr since diagnosis) and nondiabetic cadaveric donors. We also subjected isolated islets from diabetic (3 +/- 1 yr since diagnosis) and nondiabetic donors to an established in vitro model of islet plasticity. Differences in beta-cell volume between diabetic and nondiabetic donors were consistently less pronounced than those reported in long-standing T2D. A compensatory increase in beta-cell neogenesis appeared to mediate this effect. Studies of induced plasticity indicated that islets from diabetic donors were capable of epithelial dedifferentiation but did not demonstrate regenerative potential, as was seen in islets from nondiabetic donors. This deficiency was associated with the overexpression of Notch signaling molecules and a decreased neurogenin-3(+) cell frequency. One interpretation of these results would be that decreased beta-cell volume is a consequence, not a cause, of T2D, mediated by increased apoptosis and attenuated beta-cell (re)generation. However, other explanations are also possible. It remains to be seen whether the morphogenetic plasticity of human islets, deficient in vitro in islets from diabetic donors, is a component of normal beta-cell mass dynamics.
Collapse
Affiliation(s)
- Stephen C Hanley
- M.Eng., Montréal General Hospital C9-128, 1650 Cedar Avenue, Montréal, Québec, Canada H3G 1A4
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Mason MN, Arnold CA, Mahoney MJ. Entrapped collagen type 1 promotes differentiation of embryonic pancreatic precursor cells into glucose-responsive beta-cells when cultured in three-dimensional PEG hydrogels. Tissue Eng Part A 2010; 15:3799-808. [PMID: 19537960 DOI: 10.1089/ten.tea.2009.0148] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Development of an alternative source of functional, transplantable beta-cells to replace or supplement cadaveric tissue is critical to the future success of islet cell transplantation therapy. Embryonic pancreatic precursor cells are desirable as a renewable source of beta-cells as they are both proliferative and inherently capable of pancreatic cell differentiation. We have previously shown that precursor cells undergo selective beta-cell differentiation when dissociated and photoencapsulated in a polyethylene glycol (PEG) hydrogel network; however, these cells remained immature and were not glucose responsive. Collagen type 1 supports mature cell viability and function in many cell types and we hypothesized that incorporating it within our gels may support differentiating beta-cells and facilitate beta-cell maturation. For these studies, collagen-1 was entrapped with dissociated pancreatic precursor cells in a PEG hydrogel matrix (PEGCol) with the following key findings: (1) mature, glucose-responsive, islet-like structures differentiated from spontaneously forming precursor cell clusters in PEGCol, but not unmodified PEG, hydrogels; (2) a balance existed between providing sufficient collagen-1 signaling to support precursor cell development and providing an overabundance of adhesive sites allowing contaminating mesenchymal cells to thrive' and (3) mechanical stability provided by the PEG hydrogel platform is important for successful precursor cell culture, as PEGCol hydrogels encourage glucose responsiveness and high-insulin gene expression, while pure collagen gel cultures, with the same collagen concentration, have negligible insulin gene expression. These results indicate that PEGCol hydrogels are a useful culture platform to promote differentiation of a glucose-responsive beta-cell population from dissociated precursor cells.
Collapse
Affiliation(s)
- Mariah N Mason
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80309, USA
| | | | | |
Collapse
|
27
|
Montanucci P, Basta G, Calafiore R. In Vitro–Cultured Human Islet Cell Monolayers: Stemness Markers and Insulin Recovery upon Streptozotocin Exposure. Tissue Eng Part A 2009; 15:3931-42. [DOI: 10.1089/ten.tea.2009.0124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Pia Montanucci
- Section of Internal Medicine and Endocrine and Metabolic Sciences (Di.M.I.), Department of Internal Medicine, University of Perugia, Perugia, Italy
| | - Giuseppe Basta
- Section of Internal Medicine and Endocrine and Metabolic Sciences (Di.M.I.), Department of Internal Medicine, University of Perugia, Perugia, Italy
| | - Riccardo Calafiore
- Section of Internal Medicine and Endocrine and Metabolic Sciences (Di.M.I.), Department of Internal Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
28
|
Regnault B, Osorio Y Fortea J, Miao D, Eisenbarth G, Melanitou E. Early over expression of messenger RNA for multiple genes, including insulin, in the Pancreatic Lymph Nodes of NOD mice is associated with Islet Autoimmunity. BMC Med Genomics 2009; 2:63. [PMID: 19799787 PMCID: PMC2763872 DOI: 10.1186/1755-8794-2-63] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2009] [Accepted: 10/02/2009] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Autoimmune diabetes (T1D) onset is preceded by a long inflammatory process directed against the insulin-secreting beta cells of the pancreas. Deciphering the early autoimmune mechanisms represents a challenge due to the absence of clinical signs at early disease stages. The aim of this study was to identify genes implicated in the early steps of the autoimmune process, prior to inflammation, in T1D. We have previously established that insulin autoantibodies (E-IAA) predict early diabetes onset delineating an early phenotypic check point (window 1) in disease pathogenesis. We used this sub-phenotype and applied differential gene expression analysis in the pancreatic lymph nodes (PLN) of 5 weeks old Non Obese Diabetic (NOD) mice differing solely upon the presence or absence of E-IAA. Analysis of gene expression profiles has the potential to provide a global understanding of the disease and to generate novel hypothesis concerning the initiation of the autoimmune process. METHODS Animals have been screened weekly for the presence of E-IAA between 3 and 5 weeks of age. E-IAA positive or negative NOD mice at least twice were selected and RNAs isolated from the PLN were used for microarray analysis. Comparison of transcriptional profiles between positive and negative animals and functional annotations of the resulting differentially expressed genes, using software together with manual literature data mining, have been performed. RESULTS The expression of 165 genes was modulated between E-IAA positive and negative PLN. In particular, genes coding for insulin and for proteins known to be implicated in tissue remodelling and Th1 immunity have been found to be highly differentially expressed. Forty one genes showed over 5 fold differences between the two sets of samples and 30 code for extracellular proteins. This class of proteins represents potential diagnostic markers and drug targets for T1D. CONCLUSION Our data strongly suggest that the immune related mechanisms taking place at this early age in the PLN, correlate with homeostatic changes influencing tissue integrity of the adjacent pancreatic tissue. Functional analysis of the identified genes suggested that similar mechanisms might be operating during pre-inflammatory processes deployed in tissues i) hosting parasitic microorganisms and ii) experiencing unrestricted invasion by tumour cells.
Collapse
Affiliation(s)
- Béatrice Regnault
- Immunophysiology and Intracellular Parasitism Unit, Department of Parasitology and Mycology, 75015 Paris, France.
| | | | | | | | | |
Collapse
|
29
|
Assouline-Thomas B, Pilotte A, Petropavlovskaia M, Makhlin J, Ding J, McLeod D, Hanley S, Massie B, Rosenberg L. Production and characterization of the recombinant Islet Neogenesis Associated Protein (rINGAP). Protein Expr Purif 2009; 69:1-8. [PMID: 19635567 DOI: 10.1016/j.pep.2009.07.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 07/20/2009] [Accepted: 07/20/2009] [Indexed: 01/09/2023]
Abstract
Islet Neogenesis Associated Protein (INGAP) is implicated in pancreatic islet neogenesis. INGAP peptide, a pentadecapeptide comprising amino acids 104-118, reverses diabetes in rodents and improves glucose homeostasis in patients with diabetes. The mechanism of INGAP action is unknown, but such studies would benefit from the availability of the full-length recombinant protein (rINGAP). Here we report the production of rINGAP from 293-SF cells following lentiviral transduction, and its characterization by MALDI-TOF and Q-TOF Mass Spectrometry, and HPLC. Importantly, we show that rINGAP exhibits 100x the bioactivity of INGAP peptide on a molar basis in an in vitro assay of human islet regeneration.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/isolation & purification
- Biomarkers, Tumor/biosynthesis
- Biomarkers, Tumor/chemistry
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/isolation & purification
- Cells, Cultured
- Chromatography, High Pressure Liquid
- Cricetinae
- Gene Expression Regulation
- Humans
- Islets of Langerhans/physiology
- Lectins, C-Type/biosynthesis
- Lectins, C-Type/chemistry
- Lectins, C-Type/genetics
- Lectins, C-Type/isolation & purification
- Lentivirus/genetics
- Mass Spectrometry
- Mesocricetus
- Molecular Sequence Data
- Molecular Weight
- Pancreatitis-Associated Proteins
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Recombinant Proteins/biosynthesis
- Recombinant Proteins/chemistry
- Recombinant Proteins/isolation & purification
- Regeneration/physiology
- Subcellular Fractions/metabolism
- Transduction, Genetic
Collapse
|
30
|
Kutlu B, Kayali AG, Jung S, Parnaud G, Baxter D, Glusman G, Goodman N, Behie LA, Hayek A, Hood L. Meta-analysis of gene expression in human pancreatic islets after in vitro expansion. Physiol Genomics 2009; 39:72-81. [PMID: 19622797 DOI: 10.1152/physiolgenomics.00063.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pancreatic islet transplantation as a potential cure for type 1 diabetes (T1D) cannot be scaled up due to a scarcity of human pancreas donors. In vitro expansion of beta-cells from mature human pancreatic islets provides an alternative source of insulin-producing cells. The exact nature of the expanded cells produced by diverse expansion protocols and their potential for differentiation into functional beta-cells remain elusive. We performed a large-scale meta-analysis of gene expression in human pancreatic islet cells, which were processed using three different previously described protocols for expansion and for which redifferentiation was attempted. All three expansion protocols induced dramatic changes in the expression profiles of pancreatic islets; many of these changes are shared among the three protocols. Attempts at redifferentiation of expanded cells induce a limited number of gene expression changes. Nevertheless, these fail to restore a pancreatic islet-like gene expression pattern. Comparison with a collection of public microarray datasets confirmed that expanded cells are highly comparable to mesenchymal stem cells. Genes induced in expanded cells are also enriched for targets of transcription factors important for pluripotency induction. The present data increase our understanding of the active pathways in expanded and redifferentiated islets. Knowledge of the mesenchymal stem cell potential may help development of drug therapeutics to restore beta-cell mass in T1D patients.
Collapse
Affiliation(s)
- B Kutlu
- Institute for Systems Biology, Seattle, Washington, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Cui W, De Jesus K, Zhao H, Takasawa S, Shi B, Srikant CB, Liu JL. Overexpression of Reg3alpha increases cell growth and the levels of cyclin D1 and CDK4 in insulinoma cells. Growth Factors 2009; 27:195-202. [PMID: 19343564 DOI: 10.1080/08977190902863548] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Regenerating gene (Reg) family protein Reg3alpha is normally expressed in pancreatic acinar and endocrine cells. In order to explore its effect on islet beta-cell replication, insulinoma MIN6 cells were stably transfected with murine Reg3alpha cDNA. Determined using real-time PCR and Western blots, the levels of Reg3alpha mRNA and protein in Reg3alpha-transfected clones were increased 10- and 6-fold, respectively. Western blots also revealed that the protein was released into the culture medium, consistent with an endocrine effect. In MTT cell proliferation assay, Reg3alpha-overexpressing cells exhibited a 2-fold increase in the rate of cell growth. In order to investigate the intracellular mechanism, we studied cell cycle regulatory proteins. In Reg3alpha-expressing cells, we detected 2.2- and 2.5-fold increased levels of cyclin D1 and CDK4, respectively, which paralleled a 1.8-fold increase in the rate of Akt phosphorylation. It is established that beta-cell replication is associated with increased cyclin D1 and CDK4 levels; deficiency in CDK4 or cyclin D2 results in reduced beta-cell mass and diabetes. Our results suggest that Reg3alpha stimulates beta-cell replication, by activating Akt kinase and increasing the levels of cyclin D1/CDK4.
Collapse
Affiliation(s)
- Wei Cui
- School of Medicine, The First Affiliated Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, PR China
| | | | | | | | | | | | | |
Collapse
|
32
|
Pittenger GL, Taylor-Fishwick D, Vinik AI. A role for islet neogenesis in curing diabetes. Diabetologia 2009; 52:735-8. [PMID: 19271208 DOI: 10.1007/s00125-009-1322-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Accepted: 02/10/2009] [Indexed: 10/21/2022]
Affiliation(s)
- G L Pittenger
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA.
| | | | | |
Collapse
|
33
|
Abstract
Current therapies do not prevent the complications of diabetes. Furthermore, these therapies do not address the underlying pathology; the lack of functional beta-cell mass that occurs in both types 1 and 2 diabetes. While pancreas and islet transplantation do serve to increase beta-cell mass, a lack of donor organs limits the therapeutic potential of these treatments. As such, expansion of beta-cell mass from endogenous sources, either in vivo or in vitro, represents an area of increasing interest. One potential source of islet progenitors is the islet proper, via the dedifferentiation, proliferation, and redifferentiation of facultative progenitors residing within the islet. We have developed a tissue culture platform whereby isolated adult human pancreatic islets form proliferative duct-like structures expressing ductal and progenitor markers. Short-term treatment with a peptide fragment of islet neogenesis-associated protein (INGAP) induces these structures to reform islet-like structures that resemble freshly isolated islets with respect to the frequency and distribution of the four endocrine cell types, islet gene expression and hormone production, insulin content, and glucose-responsive insulin secretion. As such, the plasticity of adult human islets has significant implications for islet regeneration.
Collapse
Affiliation(s)
- Stephen Hanley
- Department of Surgery, and Centre for Pancreatic Diseases, McGill University Health Centre, Montreal, Quebec, Canada
| | | |
Collapse
|
34
|
Hanley SC, Pilotte A, Massie B, Rosenberg L. Cellular origins of adult human islet in vitro dedifferentiation. J Transl Med 2008; 88:761-72. [PMID: 18490899 DOI: 10.1038/labinvest.2008.41] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cultured human islets can be dedifferentiated to duct-like structures composed mainly of cytokeratin+ and nestin+ cells. Given that these structures possess the potential to redifferentiate into islet-like structures, we sought to elucidate their specific cellular origins. Adenoviral vectors were engineered for beta-, alpha-, delta- or PP-cell-specific GFP expression. A double-stranded system was designed whereby cultures were infected with two vectors: one expressed GFP behind the cumate-inducible promoter sequence, and the other expressed the requisite transactivator behind the human insulin, glucagon, somatostatin or pancreatic polypeptide promoter. This system labels hormone+ cells in the islet in a cell-specific manner, allowing these cells to be tracked during the course of transformation from islet to duct-like structure. Post-infection, islets were cultured to induce dedifferentiation. Fluorescence microscopy demonstrated that alpha-, delta- and PP-cells contributed equally to the cytokeratin+ population, with minimal beta-cell contribution, whereas the converse was true for nestin+ cells. Complementary targeted cell ablation studies, using streptozotocin or similar adenoviral expression of the Bax (Bcl2-associated X protein) toxigene, validated these findings and suggested a redundancy between alpha-, delta- and PP-cells with respect to cytokeratin+ cell derivation. These results call into question the traditional understanding of islet cells as being terminally differentiated and provide support for the concept of adult islet morphogenetic plasticity.
Collapse
|
35
|
|
36
|
|
37
|
Limbert C, Päth G, Jakob F, Seufert J. Beta-cell replacement and regeneration: Strategies of cell-based therapy for type 1 diabetes mellitus. Diabetes Res Clin Pract 2008; 79:389-99. [PMID: 17854943 DOI: 10.1016/j.diabres.2007.06.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2007] [Accepted: 06/20/2007] [Indexed: 01/09/2023]
Abstract
Pancreatic islet transplantation has demonstrated that long-term insulin independence may be achieved in patients suffering from diabetes mellitus type 1. However, because of limited availability of islet tissue, new sources of insulin producing cells that are responsive to glucose are required. Development of pancreatic beta-cell lines from rodent or human origin has progressed slowly in recent years. Current experiments for ex vivo expansion of beta cells and in vitro differentiation of embryonic and adult stem cells into insulin producing beta-cell phenotypes led to promising results. Nevertheless, the cells generated to date lack important characteristics of mature beta cells and generally display reduced insulin secretion and loss of proliferative capacity. Therefore, much better understanding of the mechanisms that regulate expansion and differentiation of stem/progenitor cells is necessary. Here, we review recent advances in the identification of potential cellular sources, and the development of strategies to regenerate or fabricate insulin producing and glucose sensing cells that might enable future cell-based therapies of diabetes mellitus type 1.
Collapse
Affiliation(s)
- C Limbert
- Division of Endocrinology and Diabetology, Department of Internal Medicine II, University Hospital Freiburg, Freiburg, Germany
| | | | | | | |
Collapse
|
38
|
Abstract
OBJECTIVE Adult islet neogenesis is believed to recapitulate elements of pancreatic endocrine development. Identifying factors that regulate islet neogenesis-associated protein (INGAP) gene activity could provide links to pancreas development. METHODS Predicted transcriptional regulators of INGAP were screened in an INGAP-promoter-reporter assay. Based upon their temporal expression, the occurrence of INGAP-positive cells during pancreas embryonic development were studied. RESULTS Pancreatic transcription factors, PDX-1, Ngn3, NeuroD, and Isl-1, activated the INGAP promoter, but PAX4, PAX6, and Nkx2.2 did not. The INGAP-positive cells were present in the developing pancreatic bud of the mouse embryo. Emerging clusters of unorganized endocrine cells were INGAP positive. These cells coexpressed insulin or somatostatin, but glucagon-expressing cells remained distinct. The INGAP-positive cells were also detected in the maturing neonatal endocrine cells organized into islets. In direct contrast to the embryo, glucagon localized with most INGAP-positive cells in the postnatal endocrine cells. The INGAP-positive cells juxtaposed pancreatic duct cells. A subset of 5-bromo-2'-deoxyuridine-positive/INGAP-positive cells was detected in the neonatal pancreas. CONCLUSIONS These data implicate INGAP and/or Reg family proteins in endocrine cell patterning during embryonic development and suggest that INGAP immunoreactivity is a key marker associated with early endocrine cells.
Collapse
|
39
|
Taylor-Fishwick DA, Bowman A, Korngiebel-Rosique M, Vinik AI. Pancreatic islet immunoreactivity to the Reg protein INGAP. J Histochem Cytochem 2007; 56:183-91. [PMID: 17998566 DOI: 10.1369/jhc.7a7365.2007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The Reg-related protein family member INGAP (islet neogenesis-associated protein) is a pleiotropic factor enhancing islet neogenesis, neurite growth, beta-cell protection, and beta-cell function. Using an antibody to the N-termini of INGAP, we have identified that immunoreactivity to INGAP localized to the pancreatic endocrine cells in mouse. INGAP- and insulin-immunoreactive cells are mutually exclusive, with INGAP-immunoreactive cells being preserved after streptozotocin-mediated destruction of beta-cells. Glucagon- and INGAP-immunoreactive cells colocalize, although respective antigen expression occurs in different intracellular locations. These data suggest that INGAP-immunoreactive cells include alpha-cells; however, detection of single INGAP-immunoreactive/glucagon-negative cells indicates that this may not be exclusive. In addition to mouse, detection of islet endocrine cells that were INGAP immunoreactive/glucagon immunoreactive/insulin negative was also observed in islets from human, monkey, and rat. These findings reveal that INGAP and/or related group 3 Reg proteins have a conserved expression in the pancreatic islet.
Collapse
Affiliation(s)
- David A Taylor-Fishwick
- Department of Medicine,, Eastern Virginia Medical School, 855 W. Brambleton Ave., Norfolk, VA, USA.
| | | | | | | |
Collapse
|
40
|
Ballian N, Hu M, Liu SH, Brunicardi FC. Proliferation, hyperplasia, neogenesis, and neoplasia in the islets of Langerhans. Pancreas 2007; 35:199-206. [PMID: 17895838 DOI: 10.1097/mpa.0b013e318074c6ed] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pancreatic disease is responsible for significant morbidity and mortality as a result of pancreatic carcinoma and diabetes mellitus. Regulation of endocrine cell mass is thought to have a central role in the pathogenesis of both these diseases. Islet cell proliferation, hypertrophy, neogenesis, and apoptosis are the main determinants of endocrine cell mass in the pancreas, and their understanding has been improved by new clues of their genetic and molecular basis. Beta cells have attracted most research interest because of potential implications in the treatment of diabetes mellitus and hypoglycemic disorders. The processes that operate during pancreatic adaptation to a changing hormonal milieu are important in pancreatic carcinogenesis. There is evidence that somatostatin and its receptors are fundamental regulators of endocrine cell mass and are involved in islet tumorigenesis.
Collapse
Affiliation(s)
- Nikiforos Ballian
- Department of Surgery, The Johns Hopkins Hospital, Baltimore, MD, USA
| | | | | | | |
Collapse
|
41
|
Moogk D, Hanley S, Ramunas J, Blaylock A, Skorepova J, Rosenberg L, Jervis E. Design and analysis of a long-term live-cell imaging chamber for tracking cellular dynamics within cultured human islets of Langerhans. Biotechnol Bioeng 2007; 97:1138-47. [PMID: 17243145 DOI: 10.1002/bit.21335] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A means of expanding islet cell mass is urgently needed to supplement the limited availability of donor islets of Langerhans for transplant. Live cell imaging of human islets in culture has the potential to identify the specific cells and processes involved in islet expansion. A novel imaging chamber was developed to facilitate long-term three-dimensional imaging of human islets during transformation. Islets have been induced to transform into duct-like epithelial cystic structures and revert back to glucose responsive endocrine cells under appropriate conditions (Jamal et al. Cell Death Differ. 2005 12:702-712). Here we aim to further our understanding by characterizing the process at a single cell level over time-essentially constructing a high resolution recorded history of each cell and its progeny during transformation and reversion. The imaging chamber enables high resolution imaging of three-dimensional islets while maintaining the structure of the islet cells and intercellular matrix components. A mathematical model was developed to validate the imaging chamber design by determining the required chamber dimensions to avoid introduction of oxygen and nutrient transport limitations. Human islets were embedded in collagen in the imaging chamber and differential interference contrast time course images were obtained at 3 min intervals. Immunofluorescent imaging confirmed that islet phenotype was maintained for at least 5 days during imaging. Analysis of the time courses confirms our ability to identify and track individual cells over time and to observe cell death and phenotype transformation in isolated human islets.
Collapse
Affiliation(s)
- Duane Moogk
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
42
|
Feanny MA, Fagan SP, Ballian N, Liu SH, Li Z, Wang X, Fisher W, Brunicardi FC, Belaguli NS. PDX-1 expression is associated with islet proliferation in vitro and in vivo. J Surg Res 2007; 144:8-16. [PMID: 17583748 DOI: 10.1016/j.jss.2007.04.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Revised: 03/21/2007] [Accepted: 04/08/2007] [Indexed: 12/31/2022]
Abstract
BACKGROUND Transcription factor pancreatic duodenal homeobox-1 (PDX-1) is critical for beta-cell differentiation and insulin gene expression. In this study, we investigated the role of PDX-1 in ductal-to-islet cell transdifferentiation, islet cell apoptosis, and proliferation in addition to other regulators associated with these processes in two developing beta-cell models. MATERIALS AND METHODS CAPAN-1 cells were cultured with the GLP-1 analogue Exendin-4 (Ex-4) to induce transdifferentiation to an insulin-producing phenotype. Expression patterns of PDX-1, somatostatin receptors (SSTR) 1, 2, and 5, p27, and p38 were analyzed. To model pancreatic regeneration in vivo, subtotal pancreatectomies were performed in rats and remnant pancreata were compared to sham laparotomy controls to determine islet size, morphology, apoptosis, and PDX-1 expression. RESULTS In Ex-4-treated cells, PDX-1 expression increased 67% above basal levels within 24 h and was followed by a 10-fold decline in expression by the end of the study. Expression of cell-cycle inhibitor p27 was down-regulated by 81% at 24 h, while levels of the pro-apoptotic modulator p38 significantly increased 4-fold. When compared to controls, SSTR1 expression declined, while SSTR2 and SSTR5 expression were significantly up-regulated in treated cells. Immunofluorescence of pancreatic remnants following subtotal pancreatectomy revealed increased PDX-1 staining at 24 h followed by a significant decline at 72 h post-pancreatectomy. CONCLUSION GLP-1 analogue Ex-4 resulted in up-regulation of PDX-1 in CAPAN-1 cells and PDX-1 was up-regulated in proliferating islets following subtotal pancreatectomy in rats. The increase was seen in the first 24 h. These findings suggest a possible relationship between PDX-1 and the state of islet proliferation, islet-to-ductal transdifferentiation, apoptosis, and the expression of SSTRs.
Collapse
Affiliation(s)
- Mark A Feanny
- The Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hanley S, Rosenberg L. Transforming Growth Factor β Is a Critical Regulator of Adult Human Islet Plasticity. Mol Endocrinol 2007; 21:1467-77. [PMID: 17405902 DOI: 10.1210/me.2007-0045] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
AbstractTissue plasticity is well documented in the context of pancreatic regeneration and carcinogenesis, with recent reports implicating dedifferentiated islet cells both as endocrine progenitors and as the cell(s) of origin in pancreatic adenocarcinoma. Accordingly, it is noteworthy that accumulating evidence suggests that TGFβ signaling is essential to pancreatic endocrine development and maintenance, whereas its loss is associated with the progression to pancreatic adenocarcinoma. The aim of this study was to examine the role of TGFβ in an in vitro model of islet morphogenetic plasticity. Human islets were embedded in a collagen gel and cultured under conditions that induced transformation into duct-like epithelial structures (DLS). Addition of TGFβ caused a dose-dependent decrease in DLS formation. Although it was demonstrated that collagen-embedded islets secrete low levels of TGFβ, antibody-mediated neutralization of this endogenously released TGFβ improved DLS formation rates, suggesting local TGFβ concentrations may in fact be higher. Time course studies indicated that TGFβ signaling was associated with an increase in ERK and p38 MAPK phosphorylation, although inhibitor-based studies were consistent with an islet endocrine-stabilizing effect mediated by p38 alone. Localization of TGFβ signaling molecules suggested that the action of TGFβ is directly on the β-cell to inhibit apoptosis and thus stabilize endocrine phenotype.
Collapse
Affiliation(s)
- Stephen Hanley
- Department of Surgery, McGill University, and Centre for Pancreatic Diseases, McGill University Health Centre, Montreal, Quebec, Canada H3G 1A4
| | | |
Collapse
|
44
|
Lipsett MA, Castellarin ML, Rosenberg L. Acinar plasticity: development of a novel in vitro model to study human acinar-to-duct-to-islet differentiation. Pancreas 2007; 34:452-7. [PMID: 17446845 DOI: 10.1097/mpa.0b013e3180335c80] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The plasticity of pancreatic tissue is demonstrated in many pancreatic diseases. It has previously been shown that pancreatic islet-to-duct transformation and acinoductal metaplasia have been associated with both pancreatic regeneration and adenocarcinoma in various in vivo and in vitro settings. Understanding this inherent morphogenetic plasticity of the adult pancreas could lead to new therapeutic approaches to pancreatic disease. METHODS Cadaveric human pancreases (n = 7) were digested, and purified acinar tissue, which was approximately 85% immunoreactive for amylase and approximately 15% immunoreactive for CK-19, was embedded in a type 1 collagen matrix and cultured in a differentiation medium (DM) consisting of Dulbecco modified Eagle/F12 medium supplemented with cholera toxin (100 ng/mL), epidermal growth factor (10 ng/mL), and insulin (24 mU/mL) for 8 days. After this initial period, the resulting tissues were cultured in DM without cholera toxin, supplemented with gastrin (50 nmol/L) and hepatocyte growth factor (HGF; 10 ng/mL), with islet neogenesis-associated protein (INGAP; 167 nmol/L) or with gastrin + HGF + INGAP for 6 days. Tissue samples were then analyzed for amylase, cytokeratin 19, pancreas duodenum homeobox 1, and endocrine hormone immunoreactivity as well as dithizione positivity. RESULTS After 8 days of culture, approximately 90% of acini transformed into ductlike structures. This acinoductal transformation was characterized by a complete absence of amylase staining, with virtually all cells CK-19 immunoreactive. Addition of INGAP led to an approximately 18-fold increase in pancreas duodenum homeobox 1 immunoreactivity, although without an observed increase in insulin production as measured by dithizone positivity. However, when acinar-derived ductlike structures were cultured with gastrin + HGF + INGAP, the total incidence of dithizone-positive structures increased approximately 6-fold (10.9 +/- 2.9% vs 1.7 +/- 0.4%, P = 0.037). Treatment with gastrin + HGF alone led to no significant change in any of the measured parameters. CONCLUSIONS We have developed a novel in vitro model of adult human acinoductal metaplasia that will aid not only in developing new methods of expanding beta-cell mass but also provide insights into pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Mark A Lipsett
- Research Institute of the McGill University Health Centre and the Department of Surgery, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
45
|
Lipsett M, Hanley S, Castellarin M, Austin E, Suarez-Pinzon WL, Rabinovitch A, Rosenberg L. The role of Islet Neogenesis-Associated Protein (INGAP) in islet neogenesis. Cell Biochem Biophys 2007; 48:127-37. [PMID: 17709882 DOI: 10.1007/s12013-007-0028-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/09/2023]
Abstract
Islet Neogenesis-Associated Protein (INGAP) is a member of the Reg family of proteins implicated in various settings of endogenous pancreatic regeneration. The expression of INGAP and other RegIII proteins has also been linked temporally and spatially with the induction of islet neogenesis in animal models of disease and regeneration. Furthermore, administration of a peptide fragment of INGAP (INGAP peptide) has been demonstrated to reverse chemically induced diabetes as well as improve glycemic control and survival in an animal model of type 1 diabetes. Cultured human pancreatic tissue has also been shown to be responsive to INGAP peptide, producing islet-like structures with function, architecture and gene expression matching that of freshly isolated islets. Likewise, studies in normoglycemic animals show evidence of islet neogenesis. Finally, recent clinical studies suggest an effect of INGAP peptide to improve insulin production in type 1 diabetes and glycemic control in type 2 diabetes.
Collapse
Affiliation(s)
- Mark Lipsett
- Department of Surgery, McGill University, Montreal, Quebec, Canada H3G 1A4
| | | | | | | | | | | | | |
Collapse
|
46
|
Strobel O, Dor Y, Stirman A, Trainor A, Fernández-del Castillo C, Warshaw AL, Thayer SP. Beta cell transdifferentiation does not contribute to preneoplastic/metaplastic ductal lesions of the pancreas by genetic lineage tracing in vivo. Proc Natl Acad Sci U S A 2007; 104:4419-24. [PMID: 17360539 PMCID: PMC1815470 DOI: 10.1073/pnas.0605248104] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Inflammatory injury to the pancreas results in regeneration of normal tissue and formation of metaplastic lesions of a ductal phenotype. These metaplastic ductal lesions (MDL) are called tubular complexes (TC), mucinous metaplasia, or pancreatic intraepithelial neoplasia. Because they are regularly found in chronic pancreatitis and pancreatic cancer, their formation is thought to represent a step in inflammation-mediated carcinogenesis. Despite these lesions' ductal character, their origin is controversial. All known pancreatic cell lineages have been suggested as the origin. In vitro studies suggest that differentiated cells in the pancreas remain highly plastic and can transdifferentiate as a mechanism of regeneration and metaplasia. In vivo studies suggest that islets, specifically beta cells, may be the cell of origin. However, in vitro studies are subject to ductal cell contamination, and previous in vivo studies interpret static data rather than direct evidence. Using genetic lineage tracing in vivo, we investigate whether transdifferentiation of beta cells contributes to regeneration or metaplasia in pancreatitis. RIP-CreER;Z/AP mice were used to heritably tag beta cells in the adult pancreas. Injury by cerulein pancreatitis resulted in regeneration of normal tissue and metaplasia with formation of two distinct types of TC and mucinous lesions. Lineage tracing revealed that none of these MDL are of beta cell origin; nor do beta cells contribute to regeneration of normal acinar and ductal tissue, which indicates that the plasticity of differentiated pancreatic islet cells, suggested by earlier static and in vitro studies, plays no role in regeneration, metaplasia, and carcinogenesis in vivo.
Collapse
Affiliation(s)
- Oliver Strobel
- *Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114; and
| | - Yuval Dor
- Department of Cellular Biochemistry and Human Genetics, The Hebrew University–Hadassah Medical School, Jerusalem 91120, Israel
| | - Amy Stirman
- *Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114; and
| | - Amanda Trainor
- *Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114; and
| | | | - Andrew L. Warshaw
- *Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114; and
| | - Sarah P. Thayer
- *Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114; and
- To whom correspondence should be addressed at:
Department of Surgery, Massachusetts General Hospital, 15 Parkman Street, WACC 460, Boston, MA 02114. E-mail:
| |
Collapse
|
47
|
Fleming A, Rosenberg L. Prospects and challenges for islet regeneration as a treatment for diabetes: a review of islet neogenesis associated protein. J Diabetes Sci Technol 2007; 1:231-44. [PMID: 19888412 PMCID: PMC2771469 DOI: 10.1177/193229680700100214] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Diabetes mellitus results from inadequate insulin action, which can be viewed as a consequence of the limited ability to restore beta cells after they are lost as the result of metabolic exhaustion, autoimmune destruction, or surgical insult. Arguably, a uniformly effective therapeutic pathway to address all forms of diabetes would be to reverse the restrictions on beta-cell and islet regeneration. The development from progenitor cells of islets with normal endocrine function does occur in adult humans; it is referred to as islet neogenesis. The induction of islet neogenesis is an important, if not essential, therapeutic approach for curing type 1 diabetes mellitus (T1DM) and could be valuable in the treatment of type 2 diabetes mellitus (T2DM) as well. Islet neogenesis associated protein (INGAP) is the first therapeutic candidate to be identified as the result of a purposeful search for an endogenous molecule with islet neogenic activity. It was found that partial obstruction of the pancreatic duct in hamsters induced islet neogenesis; under this condition, a neogenesis-promoting activity was identified and partially purified from a soluble tissue fraction. A 168-kDa protein product of the cloned gene was found to be responsible for the neogenesis activity. This molecule named INGAP contains an active core sequence of amino acids called INGAP peptide. Results from in vitro, animal, and human studies suggest that INGAP and INGAP peptide are neogenic in at least several vertebrate species, including humans. INGAP has since been found to be a member of the family of Reg proteins, which are found across and in multiple versions within species and are closely associated with embryonic and regenerative processes. Clinical results suggest that INGAP peptide can be a suitable neogenesis therapy, but optimization of the therapy and more data are required to fully access this potential. Understanding of the signaling pathways of INGAP and other related Reg proteins is a promising means of advancing therapeutic development for people with T1DM and T2DM. The quest for the fundamental restorative approach to lost insulin secretion is an enticing target for drug development.
Collapse
Affiliation(s)
- Alexander Fleming
- Kinexum Metabolics, Inc., Harpers Ferry, West Virginia, and formerly Supervisory Medical Officer, Division of Metabolic and Endocrine Drug Products, Food and Drug Administration, Montréal, Québec, Canada
| | - Lawrence Rosenberg
- Department of Surgery, McGill University, and Centre for Pancreatic Diseases, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| |
Collapse
|
48
|
Greer RM, Shah J, Jeske YW, Brown D, Walker RM, Cowley D, Bowling FG, Liaskou D, Harris M, Thomsett MJ, Choong C, Bell JR, Jack MM, Cotterill AM. Genotype-phenotype associations in patients with severe hyperinsulinism of infancy. Pediatr Dev Pathol 2007; 10:25-34. [PMID: 17378627 DOI: 10.2350/06-04-0083.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2006] [Accepted: 07/01/2006] [Indexed: 11/20/2022]
Abstract
In hyperinsulinism of infancy (HI), unregulated insulin secretion causes hypoglycemia. Pancreatectomy may be required in severe cases, most of which result from a defect in the beta-cell KATP channel, encoded by ABCC8 and KCNJ11. Pancreatic histology may be classified as diffuse or focal disease (the latter associated with single paternal ABCC8 mutations), indicated by the presence of islet cell nuclear enlargement in areas of diffuse abnormality. We investigated genotype-phenotype associations in a heterogeneous Australian cohort. ABCC8 and KCNJ11 genes were sequenced and case histology was reviewed in 21 infants who had pancreatectomy. Ninety-eight control DNA samples were tested by single nucleotide polymorphism analysis. Eighteen ABCC8 mutations were identified, 10 novel. Eleven patients (4 compound heterozygote, 4 single mutation, 3 no mutation detected) had diffuse hyperinsulinism. Nine patients had focal hyperinsulinism (6 single paternal mutation, 2 single mutation of undetermined parental origin, 1 none found) with absence of islet cell nuclear enlargement outside the focal area, although centroacinar cell proliferation and/or nesidiodysplasia was present in 7 cases. Regeneration after near-total pancreatectomy was documented in 4 patients, with aggregates of endocrine tissue observed at subsequent operations in 3. Although the absence of enlarged islet cell nuclei is a useful discriminant of focal hyperinsulinism associated with a paternal ABCC8 mutation, further research is needed to understand the pathophysiology of other histological abnormalities in patients with HI, which may have implications for mechanisms of ductal and islet cell proliferation. Previous surgery should be taken into account when interpreting pancreatic histology.
Collapse
Affiliation(s)
- Ristan M Greer
- Department of Paediatrics and Child Health, University of Queensland, Brisbane, and Department of Endocrinology and Diabetes, Princess Margaret Hospital for Children, Western Australia, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lu Y, Ponton A, Okamoto H, Takasawa S, Herrera PL, Liu JL. Activation of the Reg family genes by pancreatic-specific IGF-I gene deficiency and after streptozotocin-induced diabetes in mouse pancreas. Am J Physiol Endocrinol Metab 2006; 291:E50-8. [PMID: 16449294 PMCID: PMC2950860 DOI: 10.1152/ajpendo.00596.2005] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We have recently reported that Pdx1-Cre-mediated whole pancreas inactivation of IGF-I gene [in pancreatic-specific IGF-I gene-deficient (PID) mice] results in increased beta-cell mass and significant protection against both type 1 and type 2 diabetes. Because the phenotype is unlikely a direct consequence of IGF-I deficiency, the present study was designed to explore possible activation of proislet factors in PID mice by using a whole genome DNA microarray. As a result, multiple members of the Reg family genes (Reg2, -3alpha, and -3beta, previously not known to promote islet cell growth) were significantly upregulated in the pancreas. This finding was subsequently confirmed by Northern blot and/or real-time PCR, which exhibited 2- to 8-fold increases in the levels of these mRNAs. Interestingly, these Reg family genes were also activated after streptozotocin-induced beta-cell damage and diabetes (wild-type T1D mice) when islet cells were undergoing regeneration. Immunohistochemistry revealed increased Reg proteins in exocrine as well as endocrine pancreas and suggested their potential role in beta-cell neogenesis in PID or T1D mice. Previously, other Reg proteins (Reg1 and islet neogenesis-associated protein) have been shown to promote islet cell replication and neogenesis. These uncharacterized Reg proteins may play a similar but more potent role, not only in normal islet cell growth in PID mice, but also in islet cell regeneration after T1D.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Crosses, Genetic
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Female
- Gene Expression Regulation
- Immunohistochemistry
- Insulin-Like Growth Factor I/deficiency
- Insulin-Like Growth Factor I/genetics
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Lithostathine/biosynthesis
- Lithostathine/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Transgenic
- Oligonucleotide Array Sequence Analysis
- Pancreatitis-Associated Proteins
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Yarong Lu
- Fraser Laboratories, Rm. M3-15, Royal Victoria Hospital, 687 Pine Ave. West, Montreal, QC H3A 1A1, Canada
| | | | | | | | | | | |
Collapse
|
50
|
Barbosa H, Bordin S, Stoppiglia L, Silva K, Borelli M, Del Zotto H, Gagliardino J, Boschero A. Islet Neogenesis Associated Protein (INGAP) modulates gene expression in cultured neonatal rat islets. ACTA ACUST UNITED AC 2006; 136:78-84. [PMID: 16764950 DOI: 10.1016/j.regpep.2006.04.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2006] [Revised: 04/13/2006] [Accepted: 04/28/2006] [Indexed: 02/07/2023]
Abstract
The Islet Neogenesis Associated Protein (INGAP) increases pancreatic beta-cell mass and potentiates glucose-induced insulin secretion. We currently studied the effects of a pentadecapeptide having the 104-118 amino acid sequence of INGAP (INGAP-PP) on insulin secretion and on transcript profile expression in 4-day-cultured normal pancreatic neonatal rat islets. Islets cultured with INGAP-PP released significantly more insulin in response to 2.8 and 16.7 mM glucose than those cultured without the peptide. The macroarray analysis showed that 210 out of 2352 genes spotted in the nylon membranes were up-regulated while only 4 were down-regulated by INGAP-PP-treatment. The main categories of genes modified by INGAP-PP included several related with islet metabolism, insulin secretion mechanism, beta-cell mass and islet neogenesis. RT-PCR confirmed the macroarray results for ten selected genes involved in growing, maturation, maintenance of pancreatic islet-cells, and exocytosis, i.e., Hepatocyte nuclear factor 3beta (HNF3beta), Upstream stimulatory factor 1 (USF1), K(+)-channel proteins (SUR1 and Kir6.2), PHAS-I protein, Insulin 1 gene, Glucagon gene, Mitogen-activated protein kinase 1 (MAP3K1), Amylin (IAPP), and SNAP-25. INGAP-PP also stimulated PDX-1 expression. The expression of three transcripts (HNF3beta, SUR1, and SNAP-25) was confirmed by Western blotting for the corresponding proteins. In conclusion, our results show that INGAP-PP enhances specifically the secretion of insulin and the transcription of several islet genes, many of them directly or indirectly involved in the control of islet metabolism, beta-cell mass and islet neogenesis. These results, together with other previously reported, strongly indicate an important role of INGAP-PP, and possibly of INGAP, in the regulation of islet function and development.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/physiology
- Biomarkers, Tumor/chemistry
- Biomarkers, Tumor/physiology
- Cells, Cultured
- Cytokines/biosynthesis
- DNA, Complementary/metabolism
- Gene Expression Regulation
- Glucose/metabolism
- Insulin/metabolism
- Insulin Secretion
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Lectins, C-Type/chemistry
- Lectins, C-Type/physiology
- Oligonucleotide Array Sequence Analysis
- Pancreatitis-Associated Proteins
- Peptide Fragments/biosynthesis
- Potassium Channels/chemistry
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Helena Barbosa
- Departamento de Fisiologia e Biofísica, Instituto de Biologia Universidade Estadual de Campinas, 13083-970, Campinas, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|