1
|
Wright JJ, Eskaros A, Windon A, Bottino R, Jenkins R, Bradley AM, Aramandla R, Philips S, Kang H, Saunders DC, Brissova M, Powers AC. Exocrine Pancreas in Type 1 and Type 2 Diabetes: Different Patterns of Fibrosis, Metaplasia, Angiopathy, and Adiposity. Diabetes 2024; 73:1140-1152. [PMID: 37881846 PMCID: PMC11189834 DOI: 10.2337/db23-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/18/2023] [Indexed: 10/27/2023]
Abstract
The endocrine and exocrine compartments of the pancreas are spatially related but functionally distinct. Multiple diseases affect both compartments, including type 1 diabetes (T1D), pancreatitis, cystic fibrosis, and pancreatic cancer. To better understand how the exocrine pancreas changes with age, obesity, and diabetes, we performed a systematic analysis of well-preserved tissue sections from the pancreatic head, body, and tail of organ donors with T1D (n = 20) or type 2 diabetes (T2D) (n = 25) and donors with no diabetes (ND; n = 74). Among ND donors, we found that the incidence of acinar-to-ductal metaplasia (ADM), angiopathy, and pancreatic adiposity increased with age, and ADM and adiposity incidence also increased with BMI. Compared with age- and sex-matched ND organs, T1D pancreata had greater rates of acinar atrophy and angiopathy, with fewer intralobular adipocytes. T2D pancreata had greater rates of ADM and angiopathy and a higher total number of T lymphocytes, but no difference in adipocyte number, compared with ND organs. Although total pancreatic fibrosis was increased in both T1D and T2D, the patterns were different, with periductal and perivascular fibrosis occurring more frequently in T1D pancreata and lobular and parenchymal fibrosis occurring more frequently in T2D. Thus, the exocrine pancreas undergoes distinct changes as individuals age or develop T1D or T2D. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Jordan J. Wright
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN
| | - Adel Eskaros
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Annika Windon
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Rita Bottino
- Imagine Islet Center, Imagine Pharma, Pittsburgh, PA
- Institute of Cellular Therapeutics, Allegheny-Singer Research Institute, Allegheny Health Network, Pittsburgh, PA
| | - Regina Jenkins
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Amber M. Bradley
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Radhika Aramandla
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Sharon Philips
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Hakmook Kang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Diane C. Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Human Pancreas Analysis Program, Nashville, TN; Philadelphia, PA; and Gainesville, FL
| | - Marcela Brissova
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Human Pancreas Analysis Program, Nashville, TN; Philadelphia, PA; and Gainesville, FL
| | - Alvin C. Powers
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN
- Human Pancreas Analysis Program, Nashville, TN; Philadelphia, PA; and Gainesville, FL
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
2
|
Li R, Yang T, Zhang M, Ren K, Li J, Sato I, Yi SQ. A new histopathological phenomenon: Pancreatic islet cell loss in the elderly population. Dig Liver Dis 2024; 56:1039-1045. [PMID: 38065700 DOI: 10.1016/j.dld.2023.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 05/28/2024]
Abstract
BACKGROUND We observed the phenomenon of pancreatic islet cell loss (ICL) in our previous histopathological study. Multiple studies have reported that a decrease in β-cells is correlated with diabetes or chronic pancreatitis. Few studies have reported ICL in a healthy population. METHODS Thirty-three pancreatic tissue samples were obtained from cadavers (age: 65-104 years) who had never been diagnosed with any pancreatic diseases before death. The pancreatic body sections were used for an immunohistochemical study of pancreatic islet cells, and area calculations were performed using ImageJ to determine the degree of ICL and islet cell proportions. RESULTS The proportion of β-cells showed a downward trend as the degree of ICL increased (r=-0.414, P = 0.011), and the proportion of women with severe ICL was significantly higher than that of men with severe ICL (P = 0.016). The probability of severe ICL decreased with age in the population over 70 years of age (P = 0.069, linear correlation). Severe ICL may be associated with higher pancreatic intraepithelial neoplasia lesions (P = 0.059). CONCLUSION The phenomenon of ICL in the elderly population was mainly due to pancreatic β-cell reduction. It may be one of the direct causes of age-related diabetes.
Collapse
Affiliation(s)
- Rujia Li
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 116-8551 Tokyo, Japan
| | - Ting Yang
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 116-8551 Tokyo, Japan
| | - Mingshou Zhang
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 116-8551 Tokyo, Japan
| | - Ke Ren
- Faculty of Physical Education, Qu Jing Normal University, Yun Nan, China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Iwao Sato
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Shuang-Qin Yi
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 116-8551 Tokyo, Japan.
| |
Collapse
|
3
|
Zook HN, Quijano JC, Ortiz JA, Donohue C, Lopez K, Li W, Erdem N, Jou K, Crook CJ, Garcia I, Kandeel F, Montero E, Ku HT. Activation of ductal progenitor-like cells from adult human pancreas requires extracellular matrix protein signaling. iScience 2024; 27:109237. [PMID: 38433896 PMCID: PMC10904999 DOI: 10.1016/j.isci.2024.109237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 12/22/2023] [Accepted: 02/09/2024] [Indexed: 03/05/2024] Open
Abstract
Ductal progenitor-like cells are a sub-population of ductal cells in the adult human pancreas that have the potential to contribute to regenerative medicine. However, the microenvironmental cues that regulate their activation are poorly understood. Here, we establish a 3-dimensional suspension culture system containing six defined soluble factors in which primary human ductal progenitor-like and ductal non-progenitor cells survive but do not proliferate. Expansion and polarization occur when suspension cells are provided with a low concentration (5% v/v) of Matrigel, a sarcoma cell product enriched in many extracellular matrix (ECM) proteins. Screening of ECM proteins identified that collagen IV can partially recapitulate the effects of Matrigel. Inhibition of integrin α1β1, a major collagen IV receptor, negates collagen IV- and Matrigel-stimulated effects. These results demonstrate that collagen IV is a key ECM protein that stimulates the expansion and polarization of human ductal progenitor-like and ductal non-progenitor cells via integrin α1β1 receptor signaling.
Collapse
Affiliation(s)
- Heather N. Zook
- Irell & Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Janine C. Quijano
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jose A. Ortiz
- Irell & Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Cecile Donohue
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Kassandra Lopez
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Wendong Li
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Neslihan Erdem
- Irell & Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Diabetes Immunology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Kevin Jou
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Christiana J. Crook
- Irell & Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Isaac Garcia
- Department of Diabetes Immunology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Enrique Montero
- Department of Diabetes Immunology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Hsun Teresa Ku
- Irell & Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
4
|
Turniak-Kusy M, Studzian M, Szpakowski P, Kuchta P, Smietanka K, Mattern C, Pulaski L, Bielecki B. Testosterone Inhibits Secretion of the Pro-Inflammatory Chemokine CXCL1 from Astrocytes. Curr Issues Mol Biol 2024; 46:2105-2118. [PMID: 38534751 DOI: 10.3390/cimb46030135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 02/25/2024] [Accepted: 03/01/2024] [Indexed: 03/28/2024] Open
Abstract
Astrocytes play an important role in the regulation of the inflammatory response in the CNS, e.g., in demyelinating diseases. Since the chemokine CXCL1 is known to be secreted by astrocytes and to have a pro-inflammatory effect on immune cells in the CNS, we verified the effect of testosterone on its secretion in vitro (in the astrocytic cell line DI TNC1). Testosterone reduced the increase in CXCL1 production caused by the pro-inflammatory agent lysophosphatidylcholine and restored the basal production level of CXCL1. The androgen receptor (present and functional in the studied cell line) was strongly suggested to mediate this effect-its non-steroid ligand flutamide exerted an agonist-like effect, mimicking the activity of testosterone itself on CXCL1 secretion. This novel mechanism has important implications for the known immunomodulatory effect of testosterone and potentially other androgenic hormones. It provides a potential explanation on the molecular level and shows that astrocytes are important players in inflammatory homeostasis in the CNS and its hormonal regulation. Therefore, it suggests new directions for the development of the therapeutic intervention.
Collapse
Affiliation(s)
| | - Maciej Studzian
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, Polish Academy of Sciences, 90-364 Lodz, Poland
| | - Piotr Szpakowski
- Department of Neurology and Stroke, Medical University of Lodz, 90-549 Lodz, Poland
| | - Piotr Kuchta
- Faculty of Medicine, Medical University of Lodz, 90-419 Lodz, Poland
| | - Kaja Smietanka
- Department of Neurology and Stroke, Medical University of Lodz, 90-549 Lodz, Poland
| | - Claudia Mattern
- Oceanographic Center, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
- M&P Pharma AG, 6376 Emmetten, Switzerland
| | - Lukasz Pulaski
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, Polish Academy of Sciences, 90-364 Lodz, Poland
| | - Bartosz Bielecki
- Department of Neurology, Laboratory of Neuroimmunology, Medical University of Lodz, 90-153 Lodz, Poland
| |
Collapse
|
5
|
Diao H, Li X, Xu Y, Xing X, Pang S. Asprosin, a novel glucogenic adipokine implicated in type 2 diabetes mellitus. J Diabetes Complications 2023; 37:108614. [PMID: 37769508 DOI: 10.1016/j.jdiacomp.2023.108614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/12/2023] [Accepted: 09/17/2023] [Indexed: 10/03/2023]
Abstract
Asprosin, encoded by penultimate two exons (exon 65 and exon 66) of the gene Fibrillin 1 (FBN1), has been recently discovered to be a novel hormone secreted by white adipose tissues during fasting. The glucose metabolism disorders are often accompanied by increased asprosin level. Previous research suggests that asprosin may contribute to the development of diabetes by regulating glucose homeostasis, appetite, insulin secretion, and insulin sensitivity. In this review, we summarize the recent findings from studies on asprosin and its association with Type 2 diabetes mellitus, and discusses its mechanisms from various aspects, so as to provide clinical diagnosis and treatment ideas for T2DM.
Collapse
Affiliation(s)
- Hongcui Diao
- Department of Endocrinology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Xue Li
- Department of Infectious Diseases, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yeqiu Xu
- Department of Eye, Oral & Plastic Surgery, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiuli Xing
- Department of Endocrinology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shuguang Pang
- Department of Endocrinology, Jinan Central Hospital, Shandong University, Jinan, China; Department of Endocrinology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
6
|
Kulkarni S, Posgai AL, Kusmartseva I, Wasserfall CH, Atkinson MA, Butler AE. Exocrine and Endocrine Inflammation Increases Cellular Replication in the Pancreatic Duct Compartment in Type 1 Diabetes. J Endocr Soc 2022; 6:bvac136. [PMID: 36249412 PMCID: PMC9557836 DOI: 10.1210/jendso/bvac136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Indexed: 01/21/2023] Open
Abstract
Context We recently demonstrated increased cellular proliferation in the pancreatic ductal gland (PDG) compartment of organ donors with type 1 diabetes, suggesting that PDGs may harbor progenitor cells capable of pancreatic regeneration. Objective We evaluated the impact of diabetes and pancreatic inflammation on PDG and interlobular duct (ILD) cellular proliferation and profiles. Methods Endocrine hormone expression (insulin, glucagon, somatostatin, pancreatic polypeptide) and proliferating Ki67+ cells were localized within the PDG and ILD compartments by multicolor immunohistochemistry in cross-sections from the head, body, and tail regions of pancreata from those with (n = 31) or without type 1 diabetes (n = 43). Whole-slide scanned images were analyzed using digital pathology. Results Type 1 diabetes donors with insulitis or histologically identified pancreatitis had increased cellular replication in the ILD and PDG compartments. Interestingly, while cellular proliferation within the pancreatic ductal tree was significantly increased in type 1 diabetes (PDG mean = 3.36%, SEM = 1.06; ILD mean = 2.78%, SEM = 0.97) vs nondiabetes(ND) subjects without pancreatic inflammation (PDG mean = 1.18%, SEM = 0.42; ILD mean = 0.74%, SEM = 0.15, P < 0.05), robust replication was also observed in ND donors with pancreatitis (PDG mean = 3.52%, SEM = 1.33; ILD mean = 2.18%, SEM = 0.54, P < 0.05). Few polyhormonal cells were present in the ILD (type 1 diabetes = 0.04 ± 0.02%; ND = 0.08 ± 0.03%, P = 0.40) or PDG compartment (type 1 diabetes = 0.02 ± 0.01%; ND = 0.08 ± 0.13%, P = 0.63). Conclusion These data suggest that increased pancreatic ductal cell replication is associated with sustained pancreatic inflammation; however, as replicating cells were hormone-negative, PDGs do not appear to represent a compelling endogenous source of hormone-positive endocrine cells.
Collapse
Affiliation(s)
- Shweta Kulkarni
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Irina Kusmartseva
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Clive H Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32611, USA
- Department of Pediatrics, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Alexandra E Butler
- Department of Research, Royal College of Surgeons in Ireland-Bahrain, 15503 Adliya, Bahrain
| |
Collapse
|
7
|
Abstract
In this review, Lee and Olefsky discuss the characteristics of chronic inflammation in the major metabolic tissues and how obesity triggers these events, including a focus on the role of adipose tissue hypoxia and macrophage-derived exosomes. Obesity is the most common cause of insulin resistance, and the current obesity epidemic is driving a parallel rise in the incidence of T2DM. It is now widely recognized that chronic, subacute tissue inflammation is a major etiologic component of the pathogenesis of insulin resistance and metabolic dysfunction in obesity. Here, we summarize recent advances in our understanding of immunometabolism. We discuss the characteristics of chronic inflammation in the major metabolic tissues and how obesity triggers these events, including a focus on the role of adipose tissue hypoxia and macrophage-derived exosomes. Last, we also review current and potential new therapeutic strategies based on immunomodulation.
Collapse
Affiliation(s)
- Yun Sok Lee
- Department of Medicine, Division of Endocrinology and Metabolism, University of California at San Diego, La Jolla, California 92093, USA
| | - Jerrold Olefsky
- Department of Medicine, Division of Endocrinology and Metabolism, University of California at San Diego, La Jolla, California 92093, USA
| |
Collapse
|
8
|
Kandikattu HK, Venkateshaiah SU, Mishra A. Chronic Pancreatitis and the Development of Pancreatic Cancer. Endocr Metab Immune Disord Drug Targets 2021; 20:1182-1210. [PMID: 32324526 DOI: 10.2174/1871530320666200423095700] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/31/2019] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
Abstract
Pancreatitis is a fibro-inflammatory disorder of the pancreas that can occur acutely or chronically as a result of the activation of digestive enzymes that damage pancreatic cells, which promotes inflammation. Chronic pancreatitis with persistent fibro-inflammation of the pancreas progresses to pancreatic cancer, which is the fourth leading cause of cancer deaths across the globe. Pancreatic cancer involves cross-talk of inflammatory, proliferative, migratory, and fibrotic mechanisms. In this review, we discuss the role of cytokines in the inflammatory cell storm in pancreatitis and pancreatic cancer and their role in the activation of SDF1α/CXCR4, SOCS3, inflammasome, and NF-κB signaling. The aberrant immune reactions contribute to pathological damage of acinar and ductal cells, and the activation of pancreatic stellate cells to a myofibroblast-like phenotype. We summarize several aspects involved in the promotion of pancreatic cancer by inflammation and include a number of regulatory molecules that inhibit that process.
Collapse
Affiliation(s)
- Hemanth K Kandikattu
- Department of Medicine, Tulane Eosinophilic Disorders Centre (TEDC), Section of Pulmonary Diseases, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Sathisha U Venkateshaiah
- Department of Medicine, Tulane Eosinophilic Disorders Centre (TEDC), Section of Pulmonary Diseases, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Anil Mishra
- Department of Medicine, Tulane Eosinophilic Disorders Centre (TEDC), Section of Pulmonary Diseases, Tulane University School of Medicine, New Orleans, LA 70112, United States
| |
Collapse
|
9
|
Yagihashi S. What can we learn from the islet of Joslin Medalists? J Diabetes Investig 2020; 11:1117-1119. [PMID: 32343033 PMCID: PMC7477497 DOI: 10.1111/jdi.13283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 11/28/2022] Open
Abstract
This commentary refers to the recent study of Joslin Medalists that showed evidence of residual endocrine cells still functioning in response to glucose stimuli. Islet pathology showed the ongoing remodeling influenced by autoimmune attack and glucose toxicity. The results showed the presence of functional endocrine cells that contribute to the longevity of patients with type 1 diabetes.
Collapse
Affiliation(s)
- Soroku Yagihashi
- Department of Exploratory Medicine for Nature, Life and HumanToho University School of MedicineChibaJapan
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| |
Collapse
|
10
|
Ueberberg S, Nauck MA, Uhl W, Montemurro C, Tannapfel A, Clark A, Meier JJ. Islet Amyloid in Patients With Diabetes Due to Exocrine Pancreatic Disorders, Type 2 Diabetes, and Nondiabetic Patients. J Clin Endocrinol Metab 2020; 105:5818378. [PMID: 32271378 DOI: 10.1210/clinem/dgaa176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/08/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Amyloid deposits are a typical finding in pancreatic islets from patients with type 2 diabetes. Whether this is linked to the pathogenesis of type 2 diabetes is currently unknown. Therefore, we compared the occurrence of islet amyloid in patients with type 2 diabetes, diabetes secondary to pancreatic disorders, and nondiabetic individuals. PATIENTS AND METHODS Pancreatic tissue from 15 nondiabetic patients, 22 patients with type 2 diabetes, and 11 patients with diabetes due to exocrine pancreatic disorders (chronic pancreatitis, pancreatic carcinoma) were stained for insulin, amyloid, and apoptosis. β-cell area, amyloid deposits, and β-cell apoptosis were quantified by morphometric analysis. RESULTS The proportion of islets containing amyloid deposits was significantly higher in both type 2 diabetes and diabetes due to exocrine pancreatic disorders than in healthy subjects. Islets with both amyloid and apoptosis were observed more frequently in type 2 diabetes and significantly more so in diabetes due to exocrine pancreatic disorders. In both diabetic groups, apoptotic ß-cells were found significantly more frequently in islets with more prominent amyloid deposits. CONCLUSIONS The occurrence of amyloid deposits in both type 2 diabetes and diabetes secondary to exocrine pancreatic disorders suggests that islet amyloid formation is a common feature of diabetes mellitus of different etiologies and may be associated with a loss of pancreatic ß-cells.
Collapse
Affiliation(s)
- Sandra Ueberberg
- Diabetes Division St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Michael A Nauck
- Diabetes Division St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Waldemar Uhl
- Department of General and Visceral Surgery, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Chiara Montemurro
- Diabetes Division St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
- David Geffen School of Medicine, University of California, Los Angeles, CA, US
| | | | - Anne Clark
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Juris J Meier
- Diabetes Division St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
11
|
Wang L, Liu T, Liang R, Wang G, Liu Y, Zou J, Liu N, Zhang B, Liu Y, Ding X, Cai X, Wang Z, Xu X, Ricordi C, Wang S, Shen Z. Mesenchymal stem cells ameliorate β cell dysfunction of human type 2 diabetic islets by reversing β cell dedifferentiation. EBioMedicine 2020; 51:102615. [PMID: 31918404 PMCID: PMC7000334 DOI: 10.1016/j.ebiom.2019.102615] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/05/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023] Open
Abstract
Background A physiological hallmark of patients with type 2 diabetes mellitus (T2DM) is β cell dysfunction. Despite adequate treatment, it is an irreversible process that follows disease progression. Therefore, the development of novel therapies that restore β cell function is of utmost importance. Methods This study aims to unveil the mechanistic action of mesenchymal stem cells (MSCs) by investigating its impact on isolated human T2DM islets ex vivo and in vivo. Findings We propose that MSCs can attenuate β cell dysfunction by reversing β cell dedifferentiation in an IL-1Ra-mediated manner. In response to the elevated expression of proinflammatory cytokines in human T2DM islet cells, we observed that MSCs was activated to secret IL-1R antagonist (IL-1Ra) which acted on the inflammed islets and reversed β cell dedifferentiation, suggesting a crosstalk between MSCs and human T2DM islets. The co-transplantation of MSCs with human T2DM islets in diabetic SCID mice and intravenous infusion of MSCs in db/db mice revealed the reversal of β cell dedifferentiation and improved glycaemic control in the latter. Interpretation This evidence highlights the potential of MSCs in future cell-based therapies regarding the amelioration of β cell dysfunction.
Collapse
Affiliation(s)
- Le Wang
- Organ Transplant Centre, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China; NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China
| | - Tengli Liu
- NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China; Diabetes Research Institute Federation, Hollywood, FL 33021, USA
| | - Rui Liang
- NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China; Diabetes Research Institute Federation, Hollywood, FL 33021, USA
| | - Guanqiao Wang
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Yaojuan Liu
- NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China
| | - Jiaqi Zou
- NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China
| | - Na Liu
- NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China
| | - Boya Zhang
- Organ Transplant Centre, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Yan Liu
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Xuejie Ding
- Organ Transplant Centre, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Xiangheng Cai
- The First Central Clinical College, Tianjin Medical University, Tianjin, 300192, China
| | - Zhiping Wang
- Organ Transplant Centre, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Xiumin Xu
- Diabetes Research Institute, Cell Transplant Centre; Department of Surgery; Department Medicine; Miller School of Medicine, University of Miami, Miami, FL 33136, USA; The Cure Alliance, Miami, FL 33137, USA; Diabetes Research Institute Federation, Hollywood, FL 33021, USA
| | - Camillo Ricordi
- Diabetes Research Institute, Cell Transplant Centre; Department of Surgery; Department Medicine; Miller School of Medicine, University of Miami, Miami, FL 33136, USA; The Cure Alliance, Miami, FL 33137, USA; Diabetes Research Institute Federation, Hollywood, FL 33021, USA
| | - Shusen Wang
- Organ Transplant Centre, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China; NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China; Diabetes Research Institute Federation, Hollywood, FL 33021, USA.
| | - Zhongyang Shen
- Organ Transplant Centre, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China; Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China.
| |
Collapse
|
12
|
IAPP toxicity activates HIF1α/PFKFB3 signaling delaying β-cell loss at the expense of β-cell function. Nat Commun 2019; 10:2679. [PMID: 31213603 PMCID: PMC6581914 DOI: 10.1038/s41467-019-10444-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 05/13/2019] [Indexed: 02/07/2023] Open
Abstract
The islet in type 2 diabetes (T2D) is characterized by amyloid deposits derived from islet amyloid polypeptide (IAPP), a protein co-expressed with insulin by β-cells. In common with amyloidogenic proteins implicated in neurodegeneration, human IAPP (hIAPP) forms membrane permeant toxic oligomers implicated in misfolded protein stress. Here, we establish that hIAPP misfolded protein stress activates HIF1α/PFKFB3 signaling, this increases glycolysis disengaged from oxidative phosphorylation with mitochondrial fragmentation and perinuclear clustering, considered a protective posture against increased cytosolic Ca2+ characteristic of toxic oligomer stress. In contrast to tissues with the capacity to regenerate, β-cells in adult humans are minimally replicative, and therefore fail to execute the second pro-regenerative phase of the HIF1α/PFKFB3 injury pathway. Instead, β-cells in T2D remain trapped in the pro-survival first phase of the HIF1α injury repair response with metabolism and the mitochondrial network adapted to slow the rate of cell attrition at the expense of β-cell function. Type 2 diabetes is associated with islet amyloid deposits derived from islet amyloid polypeptide (IAPP) expressed by β-cells. Here the authors show that IAPP misfolded protein stress induces the hypoxia inducible factor 1 alpha injury repair pathway and activates survival metabolic changes mediated by PFKFB3.
Collapse
|
13
|
Okano S, Yasui A, Kanno SI, Satoh K, Igarashi M, Nakajima O. Karyopherin Alpha 2-Expressing Pancreatic Duct Glands and Intra-Islet Ducts in Aged Diabetic C414A-Mutant-CRY1 Transgenic Mice. J Diabetes Res 2019; 2019:7234549. [PMID: 31179341 PMCID: PMC6507265 DOI: 10.1155/2019/7234549] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/04/2019] [Accepted: 03/17/2019] [Indexed: 11/22/2022] Open
Abstract
Our earlier studies demonstrated that cysteine414- (zinc-binding site of mCRY1-) alanine mutant mCRY1 transgenic mice (Tg mice) exhibit diabetes characterized by the reduction of β-cell proliferation and by β-cell dysfunction, presumably caused by senescence-associated secretory phenotype- (SASP-) like characters of islets. Earlier studies also showed that atypical duct-like structures in the pancreas developed age-dependently in Tg mice. Numerous reports have described that karyopherin alpha 2 (KPNA2) is highly expressed in cancers of different kinds. However, details of the expression of KPNA2 in pancreatic ductal atypia and in normal pancreatic tissues remain unclear. To assess the feature of the expression of KPNA2 in the development of the ductal atypia and islet architectures, we scrutinized the pancreas of Tg mice histopathologically. Results showed that considerable expression of KPNA2 was observed in pancreatic β-cells, suggesting its importance in maintaining the functions of β-cells. In mature stages, the level of KPNA2 expression was lower in islets of Tg mice than in wild-type controls. At 4 weeks, the expression levels of KPNA2 in islets of Tg mice were the same as those in wild-type controls. These results suggest that the reduction of KPNA2 might contribute to β-cell dysfunction in mature Tg mice. Additionally, the formation of mucin-producing intra-islet ducts, islet fibrosis, and massive T cell recruitment to the islet occurred in aged Tg mice. In exocrine areas, primary pancreatic intraepithelial neoplasias (PanINs) with mucinous pancreatic duct glands (PDGs) emerged in aged Tg mice. High expression of KPNA2 was observed in the ductal atypia. By contrast, KPNA2 expression in normal ducts was quite low. Thus, upregulation of KPNA2 seemed to be correlated with progression of the degree of atypia in pancreatic ductal cells. The SASP-like microenvironment inside islets might play stimulatory roles in the formation of ductal metaplasia inside islets and in islet fibrosis in Tg mice.
Collapse
Affiliation(s)
- Satoshi Okano
- Research Center for Molecular Genetics, Institute for Promotion of Medical Science Research, Faculty of Medicine, Yamagata University, Yamagata 990-9585, Japan
- Department of Functional Genomics, Innovative Medical Science Research, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| | - Akira Yasui
- Division of Dynamic Proteome in Cancer and Aging, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Shin-ichiro Kanno
- Division of Dynamic Proteome in Cancer and Aging, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Kennichi Satoh
- Division of Gastroenterology, Tohoku Medical and Pharmaceutical University, Sendai 983-8512, Japan
| | - Masahiko Igarashi
- Division of Diabetes and Endocrinology, Yamagata City Hospital Saiseikan, Yamagata 990-8533, Japan
| | - Osamu Nakajima
- Research Center for Molecular Genetics, Institute for Promotion of Medical Science Research, Faculty of Medicine, Yamagata University, Yamagata 990-9585, Japan
- Department of Functional Genomics, Innovative Medical Science Research, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| |
Collapse
|
14
|
Li X, Jia L, Chen X, Dong Y, Ren X, Dong Y, Chen Y, Xie L, Liu M, Shiota C, Gittes GK, Rui L, Chen Z. Islet α-cell Inflammation Induced By NF-κB inducing kinase (NIK) Leads to Hypoglycemia, Pancreatitis, Growth Retardation, and Postnatal Death in Mice. Theranostics 2018; 8:5960-5971. [PMID: 30613274 PMCID: PMC6299425 DOI: 10.7150/thno.28960] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/04/2018] [Indexed: 11/29/2022] Open
Abstract
Islet α-cell dysfunction has been shown to contribute to type 2 diabetes; however, whether islet α-cell inflammation is involved in the occurrence of pancreatitis is largely unknown. The aims of this study were to investigate how NF-κB inducing kinase (NIK) regulates pancreatic α-cell function, both in vitro and in vivo, and to assess how islet α-cell inflammation induced by NIK affects the development of pancreatitis. Methods: We utilized adenovirus-mediated NIK overexpression, ELISA, qPCR, RNA-seq, and Western blot analyses to study the role of NIK in islet α cells in vitro. Islet α-cell-specific NIK overexpressing (α-NIK-OE) mice were generated, and pancreatic α/β-cell function and the occurrence of pancreatitis in these mice were assessed via ELISA, qPCR, and immunohistochemical analyses. Results: The LTβR/noncanonical NF-κB signaling pathway is present in islet α cells. Overexpression of NIK in αTC1-6 cells induces inflammation and cell death, contributing to a decrease in the expression and secretion of glucagon. Additionally, α-cell specific overexpression of NIK (α-NIK-OE) results in α-cell death, lower serum glucagon levels, and hypoglycemia in mice. Strikingly, α-NIK-OE mice also display a reduced β-cell mass, growth retardation, pancreatitis, and postnatal death. Conclusions: Islet α-cell specific overexpression of NIK results in islet α-cell dysfunction and causes islet β-cell death and pancreatitis, which are most likely due to paracrine secretion of cytokines and chemokines from islet α cells, thus leading to hypoglycemia, growth retardation, and postnatal death in mice.
Collapse
Affiliation(s)
- Xinzhi Li
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, 130024, China
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Linna Jia
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, 130024, China
| | - Xiaoyue Chen
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, 130024, China
| | - Ying Dong
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, 130024, China
| | - Xiaomeng Ren
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, 130024, China
| | - Yuefan Dong
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, 130024, China
| | - Ying Chen
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, 130024, China
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou 510070, China
| | - Ming Liu
- Department of endocrinology and metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Chiyo Shiota
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - George K. Gittes
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Liangyou Rui
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| |
Collapse
|
15
|
Butler AE, Kirakossian D, Gurlo T, Gao F, Coppola G, Butler PC. In the setting of β-cell stress, the pancreatic duct gland transcriptome shows characteristics of an activated regenerative response. Am J Physiol Gastrointest Liver Physiol 2018; 315:G848-G854. [PMID: 30095296 PMCID: PMC6293255 DOI: 10.1152/ajpgi.00177.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The pancreatic duct gland (PDG) compartment has been proposed as a potential stem cell niche based on its coiled tubular structure embedded in mesenchyme, its proliferation and expansion in response to pancreatic injury, and the fact that it contains endocrine and exocrine epithelial cells. Little is known of the molecular signature of the PDG compartment in either a quiescent state or the potentially activated state during β-cell stress characteristic of diabetes. To address this, we performed RNA sequencing on RNA obtained from PDGs of wild-type vs. prediabetic HIP rats, a model of type 2 diabetes. The transcriptome of the PDG compartment, compared with a library of 84 tissue types, placed PDGs midpoint between the exocrine and endocrine pancreas and closely related to seminiferous tubules, consistent with a role as a stem cell niche for the exocrine and endocrine pancreas. Standard differential expression analysis (permissive threshold P < 0.005) identified 245 genes differentially expressed in PDGs from HIP rats vs. WT rats, with overrepresentation of transcripts involved in acute inflammatory responses, regulation of cell proliferation, and tissue development, while pathway analysis pointed to enrichment of cell movement-related pathways. In conclusion, the transcriptome of the PDG compartment is consistent with a pancreatic stem cell niche that is activated by ongoing β-cell stress signals. The documented PDG transcriptome provides potential candidates to be exploited for lineage tracing studies of this as yet little investigated compartment. NEW & NOTEWORTHY The pancreatic duct gland (PDG) compartment has been proposed as a potential stem cell niche. Transcriptome analysis of the PDG gland placed it midpoint between exocrine and endocrine tissues with adaptation toward response to inflammation and increased cell movement in a model of type 2 diabetes with ongoing β-cell apoptosis. These findings support the proposal that PDGs may act as a pancreatic stem cell niche.
Collapse
Affiliation(s)
- Alexandra E. Butler
- 1Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - David Kirakossian
- 1Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Tatyana Gurlo
- 1Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Fuying Gao
- 2Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Giovanni Coppola
- 2Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Peter C. Butler
- 1Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
16
|
Moin ASM, Cory M, Choi J, Ong A, Dhawan S, Dry SM, Butler PC, Rizza RA, Butler AE. Increased Chromogranin A-Positive Hormone-Negative Cells in Chronic Pancreatitis. J Clin Endocrinol Metab 2018; 103:2126-2135. [PMID: 29659906 PMCID: PMC6456995 DOI: 10.1210/jc.2017-01562] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 03/30/2018] [Indexed: 01/09/2023]
Abstract
CONTEXT Chronic pancreatitis (CP) is characterized by inflammation, fibrosis, and a loss of pancreatic acinar cells, which can result in exocrine and eventually endocrine deficiency. Pancreatitis has been reported to induce formation of new endocrine cells (neogenesis) in mice. Our recent data have implicated chromogranin A-positive hormone-negative (CPHN) cells as potential evidence of neogenesis in humans. OBJECTIVE We sought to establish if CPHN cells were more abundant in CP in humans. DESIGN, SETTING, AND PARTICIPANTS We investigated the frequency and distribution of CPHN cells and the expression of the chemokine C-X-C motif ligand 10 (CXCL10) and its receptor chemokine C-X-C motif receptor 3 in pancreas of nondiabetic subjects with CP. RESULTS CPHN cell frequency in islets was increased sevenfold in CP [2.1% ± 0.67% vs 0.35% ± 0.09% CPHN cells in islets, CP vs nonpancreatitis (NP), P < 0.01], as were the CPHN cells found as scattered cells in the exocrine areas (17.4 ± 2.9 vs 4.2 ± 0.6, CP vs NP, P < 0.001). Polyhormonal endocrine cells were also increased in CP (2.7 ± 1.2 vs 0.1 ± 0.04, CP vs NP, % of polyhormonal cells of total endocrine cells, P < 0.01), as was expression of CXCL10 in α and β cells. CONCLUSION There is increased islet endogenous expression of the inflammation marker CXCL10 in islets in the setting of nondiabetic CP and an increase in polyhormonal (insulin-glucagon expressing) cells. The increase in CPHN cells in CP, often in a lobular distribution, may indicate foci of attempted endocrine cell regeneration.
Collapse
Affiliation(s)
- Abu Saleh Md Moin
- Diabetes and Metabolism Research Institute, City of Hope, Duarte, California
| | - Megan Cory
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Jennifer Choi
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Allison Ong
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Sangeeta Dhawan
- Diabetes and Metabolism Research Institute, City of Hope, Duarte, California
| | - Sarah M Dry
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California
| | - Peter C Butler
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Robert A Rizza
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Alexandra E Butler
- Anti Doping Laboratory Qatar, Doha, Qatar
- Correspondence and Reprint Requests: Alexandra E. Butler, MBBS, Anti-Doping Laboratory Qatar, PO Box 27775, Doha, Qatar. E-mail:
| |
Collapse
|