1
|
Paradiž Leitgeb E, Pohorec V, Križančić Bombek L, Skelin Klemen M, Duh M, Gosak M, Dolenšek J, Stožer A. Calcium Imaging and Analysis in Beta Cells in Acute Mouse Pancreas Tissue Slices. Methods Mol Biol 2025; 2861:223-246. [PMID: 39395109 DOI: 10.1007/978-1-0716-4164-4_17] [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] [Indexed: 10/14/2024]
Abstract
Ca2+ ions play a central role in the stimulus-secretion coupling cascade of pancreatic beta cells. The use of confocal microscopy in conjunction with the acute pancreas tissue slice technique offers valuable insights into changes in the intracellular calcium concentration following stimulation by secretagogues. This allows the study of beta cells on a single cell level, as well as their behavior on a multicellular scale within an intact environment. With the use of advanced analytical tools, this approach offers insight into how single cells contribute to the functional unit of islets of Langerhans and processes underlying insulin secretion. Here we describe a comprehensive protocol for the preparation and utilization of acute pancreas tissue slices in mice, the use of high-resolution confocal microscopy for observation of glucose-stimulated calcium dynamics in beta cells, and the computational analysis for objective evaluation of calcium signals.
Collapse
Affiliation(s)
| | - Viljem Pohorec
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | | | | | - Maja Duh
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Marko Gosak
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Alma Mater Europaea University, Maribor, Slovenia
| | - Jurij Dolenšek
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia.
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.
| |
Collapse
|
2
|
Alver CG, Dominguez-Bendala J, Agarwal A. Engineered tools to study endocrine dysfunction of pancreas. BIOPHYSICS REVIEWS 2024; 5:041303. [PMID: 39449867 PMCID: PMC11498943 DOI: 10.1063/5.0220396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Pancreas, a vital organ with intricate endocrine and exocrine functions, is central to the regulation of the body's glucose levels and digestive processes. Disruptions in its endocrine functions, primarily regulated by islets of Langerhans, can lead to debilitating diseases such as diabetes mellitus. Murine models of pancreatic dysfunction have contributed significantly to the understanding of insulitis, islet-relevant immunological responses, and the optimization of cell therapies. However, genetic differences between mice and humans have severely limited their clinical translational relevance. Recent advancements in tissue engineering and microfabrication have ushered in a new era of in vitro models that offer a promising solution. This paper reviews the state-of-the-art engineered tools designed to study endocrine dysfunction of the pancreas. Islet on a chip devices that allow precise control of various culture conditions and noninvasive readouts of functional outcomes have led to the generation of physiomimetic niches for primary and stem cell derived islets. Live pancreatic slices are a new experimental tool that could more comprehensively recapitulate the complex cellular interplay between the endocrine and exocrine parts of the pancreas. Although a powerful tool, live pancreatic slices require more complex control over their culture parameters such as local oxygenation and continuous removal of digestive enzymes and cellular waste products for maintaining experimental functionality over long term. The combination of islet-immune and slice on chip strategies can guide the path toward the next generation of pancreatic tissue modeling for better understanding and treatment of endocrine pancreatic dysfunctions.
Collapse
Affiliation(s)
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Ashutosh Agarwal
- Author to whom correspondence should be addressed:. Tel.: +1 305 243-8925
| |
Collapse
|
3
|
Gukovskaya AS, Lerch MM, Mayerle J, Sendler M, Ji B, Saluja AK, Gorelick FS, Gukovsky I. Trypsin in pancreatitis: The culprit, a mediator, or epiphenomenon? World J Gastroenterol 2024; 30:4417-4438. [DOI: 10.3748/wjg.v30.i41.4417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/19/2024] [Accepted: 07/16/2024] [Indexed: 10/23/2024] Open
Abstract
Pancreatitis is a common, life-threatening inflammatory disease of the exocrine pancreas. Its pathogenesis remains obscure, and no specific or effective treatment is available. Gallstones and alcohol excess are major etiologies of pancreatitis; in a small portion of patients the disease is hereditary. Pancreatitis is believed to be initiated by injured acinar cells (the main exocrine pancreas cell type), leading to parenchymal necrosis and local and systemic inflammation. The primary function of these cells is to produce, store, and secrete a variety of enzymes that break down all categories of nutrients. Most digestive enzymes, including all proteases, are secreted by acinar cells as inactive proforms (zymogens) and in physiological conditions are only activated when reaching the intestine. The generation of trypsin from inactive trypsinogen in the intestine plays a critical role in physiological activation of other zymogens. It was proposed that pancreatitis results from proteolytic autodigestion of the gland, mediated by premature/inappropriate trypsinogen activation within acinar cells. The intra-acinar trypsinogen activation is observed in experimental models of acute and chronic pancreatitis, and in human disease. On the basis of these observations, it has been considered the central pathogenic mechanism of pancreatitis - a concept with a century-old history. This review summarizes the data on trypsinogen activation in experimental and genetic rodent models of pancreatitis, particularly the more recent genetically engineered mouse models that mimic mutations associated with hereditary pancreatitis; analyzes the mechanisms mediating trypsinogen activation and protecting the pancreas against its’ damaging effects; discusses the gaps in our knowledge, potential therapeutic approaches, and directions for future research. We conclude that trypsin is not the culprit in the disease pathogenesis but, at most, a mediator of some pancreatitis responses. Therefore, the search for effective therapies should focus on approaches to prevent or normalize other intra-acinar pathologic processes, such as defective autophagy leading to parenchymal cell death and unrelenting inflammation.
Collapse
Affiliation(s)
- Anna S Gukovskaya
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90073, United States
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States
| | - Markus M Lerch
- Department of Medicine, Ludwig Maximilian University Hospital, Munich 81377, Germany
| | - Julia Mayerle
- Department of Medicine II, Ludwig Maximilian University of Munich, Munich 81377, Germany
| | - Matthias Sendler
- Department of Medicine A, University of Greifswald, Greifswald 17475, Germany
| | - Baoan Ji
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, United States
| | - Ashok K Saluja
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Fred S Gorelick
- Departments of Cell Biology and Internal Medicine, Yale University School of Medicine and VA West Haven, New Haven, CT 06519, United States
| | - Ilya Gukovsky
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90073, United States
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States
| |
Collapse
|
4
|
Sgarminato V, Madrid-Wolff J, Boniface A, Ciardelli G, Tonda-Turo C, Moser C. 3D in vitromodeling of the exocrine pancreatic unit using tomographic volumetric bioprinting. Biofabrication 2024; 16:045034. [PMID: 39121863 DOI: 10.1088/1758-5090/ad6d8d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer, a leading cause of cancer-related deaths globally. Initial lesions of PDAC develop within the exocrine pancreas' functional units, with tumor progression driven by interactions between PDAC and stromal cells. Effective therapies require anatomically and functionally relevantin vitrohuman models of the pancreatic cancer microenvironment. We employed tomographic volumetric bioprinting, a novel biofabrication method, to create human fibroblast-laden constructs mimicking the tubuloacinar structures of the exocrine pancreas. Human pancreatic ductal epithelial (HPDE) cells overexpressing the KRAS oncogene (HPDE-KRAS) were seeded in the multiacinar cavity to replicate pathological tissue. HPDE cell growth and organization within the structure were assessed, demonstrating the formation of a thin epithelium covering the acini inner surfaces. Immunofluorescence assays showed significantly higher alpha smooth muscle actin (α-SMA) vs. F-actin expression in fibroblasts co-cultured with cancerous versus wild-type HPDE cells. Additionally,α-SMA expression increased over time and was higher in fibroblasts closer to HPDE cells. Elevated interleukin (IL)-6 levels were quantified in supernatants from co-cultures of stromal and HPDE-KRAS cells. These findings align with inflamed tumor-associated myofibroblast behavior, serving as relevant biomarkers to monitor early disease progression and target drug efficacy. To our knowledge, this is the first demonstration of a 3D bioprinted model of exocrine pancreas that recapitulates its true 3-dimensional microanatomy and shows tumor triggered inflammation.
Collapse
Affiliation(s)
- Viola Sgarminato
- Laboratory of Applied Photonics Devices, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Jorge Madrid-Wolff
- Laboratory of Applied Photonics Devices, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Antoine Boniface
- Laboratory of Applied Photonics Devices, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Chiara Tonda-Turo
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Christophe Moser
- Laboratory of Applied Photonics Devices, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
5
|
Drotar DM, Mojica-Avila AK, Bloss DT, Cohrs CM, Manson CT, Posgai AL, Williams MD, Brusko MA, Phelps EA, Wasserfall CH, Speier S, Atkinson MA. Impaired islet function and normal exocrine enzyme secretion occur with low inter-regional variation in type 1 diabetes. Cell Rep 2024; 43:114346. [PMID: 38850534 PMCID: PMC11251461 DOI: 10.1016/j.celrep.2024.114346] [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] [Received: 02/15/2024] [Revised: 05/03/2024] [Accepted: 05/24/2024] [Indexed: 06/10/2024] Open
Abstract
Histopathological heterogeneity in the human pancreas is well documented; however, functional evidence at the tissue level is scarce. Herein, we investigate in situ glucose-stimulated islet and carbachol-stimulated acinar cell secretion across the pancreas head (PH), body (PB), and tail (PT) regions in donors without diabetes (ND; n = 15), positive for one islet autoantibody (1AAb+; n = 7), and with type 1 diabetes (T1D; <14 months duration, n = 5). Insulin, glucagon, pancreatic amylase, lipase, and trypsinogen secretion along with 3D tissue morphometrical features are comparable across regions in ND. In T1D, insulin secretion and beta-cell volume are significantly reduced within all regions, while glucagon and enzymes are unaltered. Beta-cell volume is lower despite normal insulin secretion in 1AAb+, resulting in increased volume-adjusted insulin secretion versus ND. Islet and acinar cell secretion in 1AAb+ are consistent across the PH, PB, and PT. This study supports low inter-regional variation in pancreas slice function and, potentially, increased metabolic demand in 1AAb+.
Collapse
Affiliation(s)
- Denise M Drotar
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | - Ana Karen Mojica-Avila
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), München, Neuherberg, Germany
| | - Drew T Bloss
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | - Christian M Cohrs
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), München, Neuherberg, Germany
| | - Cameron T Manson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | - MacKenzie D Williams
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | - Maigan A Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | - Edward A Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Clive H Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA; Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Stephan Speier
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), München, Neuherberg, Germany
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA; Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA.
| |
Collapse
|
6
|
Alver CG, Álvarez-Cubela S, Altilio I, Hutchison E, Warrner E, Viso ME, Vitale G, Oliver D, Pastori RL, Dominguez-Bendala J, Agarwal A. SliceChip: a benchtop fluidic platform for organotypic culture and serial assessment of human and rodent pancreatic slices. LAB ON A CHIP 2024; 24:1557-1572. [PMID: 38205530 PMCID: PMC10939771 DOI: 10.1039/d3lc00850a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Enzymatically isolated pancreatic islets are the most commonly used ex vivo testbeds for diabetes research. Recently, precision-cut living slices of human pancreas are emerging as an exciting alternative because they maintain the complex architecture of the endocrine and exocrine tissues, and do not suffer from the mechanical and chemical stress of enzymatic isolation. We report a fluidic pancreatic SliceChip platform with dynamic environmental controls that generates a warm, oxygenated, and bubble-free fluidic pathway across singular immobilized slices with continuous deliver of fresh media and the ability to perform repeat serial perfusion assessments. A degasser ensures the system remains bubble-free while systemic pressurization with compressed oxygen ensures slice medium remains adequately oxygenated. Computational modeling of perfusion and oxygen dynamics within SliceChip guide the system's physiomimetic culture conditions. Maintenance of the physiological glucose dependent insulin secretion profile across repeat perfusion assessments of individual pancreatic slices kept under physiological oxygen levels demonstrated the culture capacity of our platform. Fluorescent images acquired every 4 hours of transgenic murine pancreatic slices were reliably stable and recoverable over a 5 day period due to the inclusion of a 3D-printed bioinert metallic anchor that maintained slice position within the SliceChip. Our slice on a chip platform has the potential to expand the useability of human pancreatic slices for diabetes pathogenesis and the development of new therapeutic approaches, while also enabling organotypic culture and assessment of other tissue slices such as brain and patient tumors.
Collapse
Affiliation(s)
- Charles G Alver
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Isabella Altilio
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Emily Hutchison
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Emma Warrner
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Mariana E Viso
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Giana Vitale
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - David Oliver
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Ricardo L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
7
|
Drotar DM, Mojica-Avila AK, Bloss DT, Cohrs CM, Manson CT, Posgai AL, Williams MD, Brusko MA, Phelps EA, Wasserfall CH, Speier S, Atkinson MA. Impaired islet function with normal exocrine enzyme secretion is consistent across the head, body, and tail pancreas regions in type 1 diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.08.579175. [PMID: 38405840 PMCID: PMC10888906 DOI: 10.1101/2024.02.08.579175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Histopathological heterogeneity in human pancreas has been well documented; however, functional evidence at the tissue level is scarce. Herein we investigated in situ glucose-stimulated islet and carbachol-stimulated acinar cell secretion across the pancreas head (PH), body (PB), and tail (PT) regions in no diabetes (ND, n=15), single islet autoantibody-positive (1AAb+, n=7), and type 1 diabetes donors (T1D, <14 months duration, n=5). Insulin, glucagon, pancreatic amylase, lipase, and trypsinogen secretion along with 3D tissue morphometrical features were comparable across the regions in ND. In T1D, insulin secretion and beta-cell volume were significantly reduced within all regions, while glucagon and enzymes were unaltered. Beta-cell volume was lower despite normal insulin secretion in 1AAb+, resulting in increased volume-adjusted insulin secretion versus ND. Islet and acinar cell secretion in 1AAb+ were consistent across PH, PB and PT. This study supports low inter-regional variation in pancreas slice function and potentially, increased metabolic demand in 1AAb+.
Collapse
Affiliation(s)
- Denise M. Drotar
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
| | - Ana Karen Mojica-Avila
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Drew T. Bloss
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
| | - Christian M. Cohrs
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Cameron T. Manson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL USA
| | - Amanda L. Posgai
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
| | - MacKenzie D. Williams
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
| | - Maigan A. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
| | - Edward A. Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL USA
| | - Clive H. Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL USA
| | - Stephan Speier
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Mark A. Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL USA
| |
Collapse
|
8
|
Cohrs CM, Chen C, Atkinson MA, Drotar DM, Speier S. Bridging the Gap: Pancreas Tissue Slices From Organ and Tissue Donors for the Study of Diabetes Pathogenesis. Diabetes 2024; 73:11-22. [PMID: 38117999 PMCID: PMC10784654 DOI: 10.2337/dbi20-0018] [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: 05/04/2021] [Accepted: 10/14/2023] [Indexed: 12/22/2023]
Abstract
Over the last two decades, increased availability of human pancreatic tissues has allowed for major expansions in our understanding of islet biology in health and disease. Indeed, studies of fixed and frozen pancreatic tissues, as well as efforts using viable isolated islets obtained from organ donors, have provided significant insights toward our understanding of diabetes. However, the procedures associated with islet isolation result in distressed cells that have been removed from any surrounding influence. The pancreas tissue slice technology was developed as an in situ approach to overcome certain limitations associated with studies on isolated islets or fixed tissue. In this Perspective, we discuss the value of this novel platform and review how pancreas tissue slices, within a short time, have been integrated in numerous studies of rodent and human islet research. We show that pancreas tissue slices allow for investigations in a less perturbed organ tissue environment, ranging from cellular processes, over peri-islet modulations, to tissue interactions. Finally, we discuss the considerations and limitations of this technology in its future applications. We believe the pancreas tissue slices will help bridge the gap between studies on isolated islets and cells to the systemic conditions by providing new insight into physiological and pathophysiological processes at the organ level. ARTICLE HIGHLIGHTS Human pancreas tissue slices represent a novel platform to study human islet biology in close to physiological conditions. Complementary to established technologies, such as isolated islets, single cells, and histological sections, pancreas tissue slices help bridge our understanding of islet physiology and pathophysiology from single cell to intact organ. Diverse sources of viable human pancreas tissue, each with distinct characteristics to be considered, are available to use in tissue slices for the study of diabetes pathogenesis.
Collapse
Affiliation(s)
- Christian M. Cohrs
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Chunguang Chen
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Mark A. Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL
| | - Denise M. Drotar
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL
| | - Stephan Speier
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
9
|
Mastracci TL, Apte M, Amundadottir LT, Alvarsson A, Artandi S, Bellin MD, Bernal-Mizrachi E, Caicedo A, Campbell-Thompson M, Cruz-Monserrate Z, El Ouaamari A, Gaulton KJ, Geisz A, Goodarzi MO, Hara M, Hull-Meichle RL, Kleger A, Klein AP, Kopp JL, Kulkarni RN, Muzumdar MD, Naren AP, Oakes SA, Olesen SS, Phelps EA, Powers AC, Stabler CL, Tirkes T, Whitcomb DC, Yadav D, Yong J, Zaghloul NA, Pandol SJ, Sander M. Integrated Physiology of the Exocrine and Endocrine Compartments in Pancreatic Diseases: Workshop Proceedings. Diabetes 2023; 72:433-448. [PMID: 36940317 PMCID: PMC10033248 DOI: 10.2337/db22-0942] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/29/2022] [Indexed: 03/22/2023]
Abstract
The Integrated Physiology of the Exocrine and Endocrine Compartments in Pancreatic Diseases workshop was a 1.5-day scientific conference at the National Institutes of Health (Bethesda, MD) that engaged clinical and basic science investigators interested in diseases of the pancreas. This report provides a summary of the proceedings from the workshop. The goals of the workshop were to forge connections and identify gaps in knowledge that could guide future research directions. Presentations were segregated into six major theme areas, including 1) pancreas anatomy and physiology, 2) diabetes in the setting of exocrine disease, 3) metabolic influences on the exocrine pancreas, 4) genetic drivers of pancreatic diseases, 5) tools for integrated pancreatic analysis, and 6) implications of exocrine-endocrine cross talk. For each theme, multiple presentations were followed by panel discussions on specific topics relevant to each area of research; these are summarized here. Significantly, the discussions resulted in the identification of research gaps and opportunities for the field to address. In general, it was concluded that as a pancreas research community, we must more thoughtfully integrate our current knowledge of normal physiology as well as the disease mechanisms that underlie endocrine and exocrine disorders so that there is a better understanding of the interplay between these compartments.
Collapse
Affiliation(s)
- Teresa L. Mastracci
- Department of Biology, Indiana University–Purdue University Indianapolis, Indianapolis, IN
| | - Minoti Apte
- Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | | | - Alexandra Alvarsson
- Diabetes, Obesity, and Metabolism Institute, Mount Sinai Hospital, New York, NY
| | - Steven Artandi
- Department of Internal Medicine, Stanford University, Stanford, CA
| | - Melena D. Bellin
- Departments of Pediatrics and Surgery, University of Minnesota Medical School, Minneapolis, MN
| | - Ernesto Bernal-Mizrachi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Alejandro Caicedo
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Martha Campbell-Thompson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Zobeida Cruz-Monserrate
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - Kyle J. Gaulton
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Andrea Geisz
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Manami Hara
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Rebecca L. Hull-Meichle
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA
| | - Alexander Kleger
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University, Ulm, Germany
| | - Alison P. Klein
- Department of Pathology and Medicine, Johns Hopkins School of Medicine, Baltimore MD
| | - Janel L. Kopp
- Department of Cellular & Physiological Sciences, The University of British Columbia, Vancouver, Canada
| | | | - Mandar D. Muzumdar
- Departments of Genetics and Internal Medicine (Oncology), Yale University School of Medicine, New Haven, CT
| | | | - Scott A. Oakes
- Department of Pathology, The University of Chicago, Chicago, IL
| | - Søren S. Olesen
- Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Edward A. Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Alvin C. Powers
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN
| | - Cherie L. Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Temel Tirkes
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN
| | | | - Dhiraj Yadav
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Jing Yong
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Norann A. Zaghloul
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Stephen J. Pandol
- Department of Gastroenterology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Maike Sander
- Department of Pediatrics and Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA
| |
Collapse
|
10
|
Takano T, Yule DI. Ca 2+ signals in pancreatic acinar cells in response to physiological stimulation in vivo. J Physiol 2023. [PMID: 36965132 DOI: 10.1113/jp284469] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/08/2023] [Indexed: 03/27/2023] Open
Abstract
The exocrine pancreas secretes fluid and digestive enzymes in response to parasympathetic release of acetylcholine (ACh) via the vagus nerve and the gut hormone cholecystokinin (CCK). Both secretion of fluid and exocytosis of secretory granules containing enzymes and zymogens are dependent on an increase in the cytosolic [Ca2+ ] in acinar cells. It is thought that the specific spatiotemporal characteristics of the Ca2+ signals are fundamental for appropriate secretion and that these properties are disrupted in disease states in the pancreas. While extensive research has been performed to characterize Ca2+ signalling in acinar cells, this has exclusively been achieved in ex vivo preparations of exocrine cells, where it is difficult to mimic physiological conditions. Here we have developed a method to optically observe pancreatic acinar Ca2+ signals in vivo using a genetically expressed Ca2+ indicator and imaged with multi-photon microscopy in live animals. In vivo, acinar cells exhibited baseline activity in fasted animals, which was dependent on CCK1 receptors (CCK1Rs). Both stimulation of intrinsic nervous input and administration of systemic CCK induced oscillatory activity in a proportion of the cells, but the maximum frequencies were vastly different. Upon feeding, oscillatory activity was also observed, which was dependent on CCK1Rs. No evidence of a vago-vagal reflex mediating the effects of CCK was observed. Our in vivo method revealed the spatial and temporal profile of physiologically evoked Ca2+ signals, which will provide new insights into future studies of the mechanisms underlying exocrine physiology and that are disrupted in pathological conditions. KEY POINTS: In the exocrine pancreas, the spatiotemporal properties of Ca2+ signals are fundamentally important for the appropriate stimulation of secretion by the neurotransmitter acetylcholine and gut hormone cholecystokinin. These characteristics were previously defined in ex vivo studies. Here we report the spatiotemporal characteristics of Ca2+ signals in vivo in response to physiological stimulation in a mouse engineered to express a Ca2+ indicator in acinar cells. Specific Ca2+ 'signatures' probably important for stimulating secretion are evoked in vivo in fasted animals, by feeding, neural stimulation and cholecystokinin administration. The Ca2+ signals are probably the result of the direct action of ACh and CCK on acinar cells and not indirectly through a vago-vagal reflex.
Collapse
Affiliation(s)
- Takahiro Takano
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| |
Collapse
|
11
|
Petersen OH. The 2022 George E Palade Medal Lecture: Toxic Ca 2+ signals in acinar, stellate and endogenous immune cells are important drivers of acute pancreatitis. Pancreatology 2023; 23:1-8. [PMID: 36539315 PMCID: PMC10809214 DOI: 10.1016/j.pan.2022.12.010] [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: 12/10/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 01/11/2023]
Abstract
In this account of the 2022 Palade Medal Lecture, an attempt is made to explain, as simply as possible, the most essential features of normal physiological control of pancreatic enzyme secretion, as they have emerged from more than 50 years of experimental work. On that basis, further studies on the mechanism by which acute pancreatitis is initiated are then described. Calcium ion signaling is crucially important for both the normal physiology of secretion control as well as for the development of acute pancreatitis. Although acinar cell processes have, rightly, been central to our understanding of pancreatic physiology and pathophysiology, attention is here drawn to the additional critical influence of calcium signaling events in stellate and immune cells in the acinar environment. These signals contribute significantly to the crucially important inflammatory response in acute pancreatitis.
Collapse
Affiliation(s)
- Ole H Petersen
- School of Biosciences, Sir Martin Evans Building, Cardiff University, Wales, CF10 3AX, UK.
| |
Collapse
|
12
|
Kang F, Xie L, Qin T, Miao Y, Kang Y, Takahashi T, Liang T, Xie H, Gaisano HY. Plasma membrane flipping of Syntaxin-2 regulates its inhibitory action on insulin granule exocytosis. Nat Commun 2022; 13:6512. [PMID: 36316316 PMCID: PMC9622911 DOI: 10.1038/s41467-022-33986-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 10/05/2022] [Indexed: 11/07/2022] Open
Abstract
Enhancing pancreatic β-cell secretion is a primary therapeutic target for type-2 diabetes (T2D). Syntaxin-2 (Stx2) has just been identified to be an inhibitory SNARE for insulin granule exocytosis, holding potential as a treatment for T2D, yet its molecular underpinnings remain unclear. We show that excessive Stx2 recruitment to raft-like granule docking sites at higher binding affinity than pro-fusion syntaxin-1A effectively competes for and inhibits fusogenic SNARE machineries. Depletion of Stx2 in human β-cells improves insulin secretion by enhancing trans-SNARE complex assembly and cis-SNARE disassembly. Using a genetically-encoded reporter, glucose stimulation is shown to induce Stx2 flipping across the plasma membrane, which relieves its suppression of cytoplasmic fusogenic SNARE complexes to promote insulin secretion. Targeting the flipping efficiency of Stx2 profoundly modulates secretion, which could restore the impaired insulin secretion in diabetes. Here, we show that Stx2 acts to assist this precise tuning of insulin secretion in β-cells, including in diabetes.
Collapse
Affiliation(s)
- Fei Kang
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada ,grid.231844.80000 0004 0474 0428Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4 Canada
| | - Li Xie
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Tairan Qin
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Yifan Miao
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Youhou Kang
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Toshimasa Takahashi
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Tao Liang
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada ,grid.231844.80000 0004 0474 0428Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4 Canada
| | - Huanli Xie
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Herbert Y. Gaisano
- grid.17063.330000 0001 2157 2938Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada ,grid.231844.80000 0004 0474 0428Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4 Canada
| |
Collapse
|
13
|
Li Y, Cui ZJ. Transmembrane Domain 3 Is a Transplantable Pharmacophore in the Photodynamic Activation of Cholecystokinin 1 Receptor. ACS Pharmacol Transl Sci 2022; 5:539-547. [PMID: 35983279 PMCID: PMC9379944 DOI: 10.1021/acsptsci.2c00031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cholecystokinin 1 receptor (CCK1R) is activated in photodynamic action by singlet oxygen, but detailed molecular mechanisms are not elucidated. To identify the pharmacophore(s) in photodynamic CCK1R activation, we examined photodynamic activation of point mutants CCK1RM121/3.32A, CCK1RM121/3.32Q, and a chimeric receptor with CCK1R transmembrane domain 3 (TM3) transplanted to muscarinic ACh receptor 3 (M3R) which is unaffected by photodynamic action. These engineered receptors were tagged at the N-terminus with genetically encoded protein photosensitizer miniSOG, and their light-driven photodynamic activation was compared to wild type CCK1R and M3R, as monitored by Fura-2 fluorescent calcium imaging. Photodynamic activations of miniSOG-CCK1RM121/3.32A and miniSOG-CCK1RM121/3.32Q were found to be 55% and 73%, respectively, when compared to miniSOG-CCK1R (100%), whereas miniSOG-M3R was not affected (0% activation). Notably, the chimeric receptor miniSOG-M3R-TM3CCK1R was effectively activated photodynamically (65%). These data suggest that TM3 is an important pharmacophore in photodynamic CCK1R activation, readily transplantable to nonsusceptible M3R for photodynamic activation.
Collapse
Affiliation(s)
- Yuan Li
- Institute of Cell Biology, Beijing Normal University, Beijing 100875, China
| | - Zong Jie Cui
- Institute of Cell Biology, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
14
|
Voronina S, Chvanov M, De Faveri F, Mayer U, Wileman T, Criddle D, Tepikin A. Autophagy, Acute Pancreatitis and the Metamorphoses of a Trypsinogen-Activating Organelle. Cells 2022; 11:cells11162514. [PMID: 36010591 PMCID: PMC9406838 DOI: 10.3390/cells11162514] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 01/18/2023] Open
Abstract
Recent studies have highlighted the importance of autophagy and particularly non-canonical autophagy in the development and progression of acute pancreatitis (a frequent disease with considerable morbidity and significant mortality). An important early event in the development of acute pancreatitis is the intrapancreatic activation of trypsinogen, (i.e., formation of trypsin) leading to the autodigestion of the organ. Another prominent phenomenon associated with the initiation of this disease is vacuolisation and specifically the formation of giant endocytic vacuoles in pancreatic acinar cells. These organelles develop in acinar cells exposed to several inducers of acute pancreatitis (including taurolithocholic acid and high concentrations of secretagogues cholecystokinin and acetylcholine). Notably, early trypsinogen activation occurs in the endocytic vacuoles. These trypsinogen-activating organelles undergo activation, long-distance trafficking, and non-canonical autophagy. In this review, we will discuss the role of autophagy in acute pancreatitis and particularly focus on the recently discovered LAP-like non-canonical autophagy (LNCA) of endocytic vacuoles.
Collapse
Affiliation(s)
- Svetlana Voronina
- Department of Molecular Physiology and Cell Signalling, University of Liverpool, Liverpool L69 3BX, UK
| | - Michael Chvanov
- Department of Molecular Physiology and Cell Signalling, University of Liverpool, Liverpool L69 3BX, UK
| | - Francesca De Faveri
- Department of Molecular Physiology and Cell Signalling, University of Liverpool, Liverpool L69 3BX, UK
| | - Ulrike Mayer
- Biomedical Research Centre, School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Tom Wileman
- Quadram Institute Bioscience and Norwich Medical School, University of East Anglia, Norwich NR4 7UQ, UK
| | - David Criddle
- Department of Molecular Physiology and Cell Signalling, University of Liverpool, Liverpool L69 3BX, UK
| | - Alexei Tepikin
- Department of Molecular Physiology and Cell Signalling, University of Liverpool, Liverpool L69 3BX, UK
- Correspondence:
| |
Collapse
|
15
|
Rafiei N, Moghadam MG, Au A, Regeenes R, Chidambaram S, Liang T, Wang Y, Yip CM, Gaisano H, Rocheleau JV. Design of a versatile microfluidic device for imaging precision-cut-tissue slices. Biofabrication 2022; 14. [PMID: 35793653 DOI: 10.1088/1758-5090/ac7eea] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/06/2022] [Indexed: 11/12/2022]
Abstract
Precision-cut-tissues (PCTs), which preserve many aspects of a tissue's microenvironment, are typically imaged using conventional sample dishes and chambers. These can require large amounts of reagent and, when used for flow-through experiments, the shear forces applied on the tissues are often ill-defined. Their physical design also makes it difficult to image large volumes and repetitively image smaller regions of interest in the living slice. We report here on the design of a versatile microfluidic device capable of holding mouse or human pancreas PCTs for 3D fluorescence imaging using confocal and selective plane illumination microscopy (SPIM). Our design positions PCTs within a 5 × 5 mm × 140µm deep chamber fitted with 150µm tall channels to facilitate media exchange. Shear stress in the device is localized to small regions on the surface of the tissue and can be easily controlled. This design allows for media exchange at flowrates ∼10-fold lower than those required for conventional chambers. Finally, this design allows for imaging the same immunofluorescently labeled PCT with high resolution on a confocal and with large field of view on a SPIM, without adversely affecting image quality.
Collapse
Affiliation(s)
- Nafiseh Rafiei
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON, Canada
| | - Mohammadamir G Moghadam
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Aaron Au
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Romario Regeenes
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | | | - Tao Liang
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Yufeng Wang
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Christopher M Yip
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Herbert Gaisano
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Jonathan V Rocheleau
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
16
|
Marolt U, Paradiž Leitgeb E, Pohorec V, Lipovšek S, Venglovecz V, Gál E, Ébert A, Menyhárt I, Potrč S, Gosak M, Dolenšek J, Stožer A. Calcium imaging in intact mouse acinar cells in acute pancreas tissue slices. PLoS One 2022; 17:e0268644. [PMID: 35657915 PMCID: PMC9165796 DOI: 10.1371/journal.pone.0268644] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/04/2022] [Indexed: 12/22/2022] Open
Abstract
The physiology and pathophysiology of the exocrine pancreas are in close connection to changes in intra-cellular Ca2+ concentration. Most of our knowledge is based on in vitro experiments on acinar cells or acini enzymatically isolated from their surroundings, which can alter their structure, physiology, and limit our understanding. Due to these limitations, the acute pancreas tissue slice technique was introduced almost two decades ago as a complementary approach to assess the morphology and physiology of both the endocrine and exocrine pancreas in a more conserved in situ setting. In this study, we extend previous work to functional multicellular calcium imaging on acinar cells in tissue slices. The viability and morphological characteristics of acinar cells within the tissue slice were assessed using the LIVE/DEAD assay, transmission electron microscopy, and immunofluorescence imaging. The main aim of our study was to characterize the responses of acinar cells to stimulation with acetylcholine and compare them with responses to cerulein in pancreatic tissue slices, with special emphasis on inter-cellular and inter-acinar heterogeneity and coupling. To this end, calcium imaging was performed employing confocal microscopy during stimulation with a wide range of acetylcholine concentrations and selected concentrations of cerulein. We show that various calcium oscillation parameters depend monotonically on the stimulus concentration and that the activity is rather well synchronized within acini, but not between acini. The acute pancreas tissue slice represents a viable and reliable experimental approach for the evaluation of both intra- and inter-cellular signaling characteristics of acinar cell calcium dynamics. It can be utilized to assess many cells simultaneously with a high spatiotemporal resolution, thus providing an efficient and high-yield platform for future studies of normal acinar cell biology, pathophysiology, and screening pharmacological substances.
Collapse
Affiliation(s)
- Urška Marolt
- Clinical department for abdominal and general surgery, University Medical Centre Maribor, Maribor, Slovenia
- * E-mail: (UM); (JD); (AS)
| | - Eva Paradiž Leitgeb
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Viljem Pohorec
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Saška Lipovšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia
| | - Viktória Venglovecz
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Eleonóra Gál
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Attila Ébert
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - István Menyhárt
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Stojan Potrč
- Clinical department for abdominal and general surgery, University Medical Centre Maribor, Maribor, Slovenia
| | - Marko Gosak
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- * E-mail: (UM); (JD); (AS)
| | - Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- * E-mail: (UM); (JD); (AS)
| |
Collapse
|
17
|
Abstract
The enteroendocrine system coordinates the physiological response to food intake by regulating rates of digestion, nutrient absorption, insulin secretion, satiation and satiety. Gut hormones with important anorexigenic and/or insulinotropic roles include glucagon-like peptide 1 (GLP-1), peptide YY (PYY3-36), cholecystokinin (CCK) and glucose-dependent insulinotropic peptide (GIP). High BMI or obesogenic diets do not markedly disrupt this enteroendocrine system, which represents a critical target for inducing weight loss and treating co-morbidities in individuals with obesity.
Collapse
|
18
|
Mareninova OA, Dillon DL, Wightman CJM, Yakubov I, Takahashi T, Gaisano HY, Munson K, Ohmuraya M, Dawson D, Gukovsky I, Gukovskaya AS. Rab9 Mediates Pancreatic Autophagy Switch From Canonical to Noncanonical, Aggravating Experimental Pancreatitis. Cell Mol Gastroenterol Hepatol 2021; 13:599-622. [PMID: 34610499 PMCID: PMC8715155 DOI: 10.1016/j.jcmgh.2021.09.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Autophagosome, the central organelle in autophagy process, can assemble via canonical pathway mediated by LC3-II, the lipidated form of autophagy-related protein LC3/ATG8, or noncanonical pathway mediated by the small GTPase Rab9. Canonical autophagy is essential for exocrine pancreas homeostasis, and its disordering initiates and drives pancreatitis. The involvement of noncanonical autophagy has not been explored. We examine the role of Rab9 in pancreatic autophagy and pancreatitis severity. METHODS We measured the effect of Rab9 on parameters of autophagy and pancreatitis responses using transgenic mice overexpressing Rab9 (Rab9TG) and adenoviral transduction of acinar cells. Effect of canonical autophagy on Rab9 was assessed in ATG5-deficient acinar cells. RESULTS Pancreatic levels of Rab9 and its membrane-bound (active) form decreased in rodent pancreatitis models and in human disease. Rab9 overexpression stimulated noncanonical and inhibited canonical/LC3-mediated autophagosome formation in acinar cells through up-regulation of ATG4B, the cysteine protease that delipidates LC3-II. Conversely, ATG5 deficiency caused Rab9 increase in acinar cells. Inhibition of canonical autophagy in Rab9TG pancreas was associated with accumulation of Rab9-positive vacuoles containing markers of mitochondria, protein aggregates, and trans-Golgi. The shift to the noncanonical pathway caused pancreatitis-like damage in acinar cells and aggravated experimental pancreatitis. CONCLUSIONS The results show that Rab9 regulates pancreatic autophagy and indicate a mutually antagonistic relationship between the canonical/LC3-mediated and noncanonical/Rab9-mediated autophagy pathways in pancreatitis. Noncanonical autophagy fails to substitute for its canonical counterpart in protecting against pancreatitis. Thus, Rab9 decrease in experimental and human pancreatitis is a protective response to sustain canonical autophagy and alleviate disease severity.
Collapse
Affiliation(s)
- Olga A Mareninova
- Department of Medicine, Los Angeles, California; VA Greater Los Angeles Healthcare System, Los Angeles, California
| | - Dustin L Dillon
- Department of Medicine, Los Angeles, California; VA Greater Los Angeles Healthcare System, Los Angeles, California
| | - Carli J M Wightman
- Department of Medicine, Los Angeles, California; VA Greater Los Angeles Healthcare System, Los Angeles, California
| | | | | | - Herbert Y Gaisano
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Keith Munson
- Department of Physiology, Los Angeles, California; VA Greater Los Angeles Healthcare System, Los Angeles, California
| | - Masaki Ohmuraya
- Department of Genetics, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - David Dawson
- Department of Pathology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Ilya Gukovsky
- Department of Medicine, Los Angeles, California; VA Greater Los Angeles Healthcare System, Los Angeles, California
| | - Anna S Gukovskaya
- Department of Medicine, Los Angeles, California; VA Greater Los Angeles Healthcare System, Los Angeles, California.
| |
Collapse
|
19
|
Sangnes DA, Sandvik Bergmann E, Moss RM, Engjom T, Søfteland E. Pancreatic exocrine insufficiency in diabetes is associated with autonomic dysfunction. Scand J Gastroenterol 2021; 56:1222-1228. [PMID: 34491874 DOI: 10.1080/00365521.2021.1957496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Pancreatic exocrine insufficiency (PEI) is prevalent in diabetes. Pathophysiological theories imply autoimmune destruction, lack of trophic effects of insulin or impaired neuronal stimulation, but the relationship between PEI and autonomic dysfunction is largely unknown. In a pilot study, we aimed to investigate if patients with diabetes and PEI had impaired autonomic function. METHODS We measured faecal elastase in 59 patients with type 1 or 2 diabetes, using a cut-off-value <200 μg/g to define PEI. Based on faecal elastase results, patients were stratified into matched case (n = 8) and control groups (n = 13). We used heart rate variability, baroreflex sensitivity and orthostatic hypotension tests to assess autonomic dysfunction. RESULTS All baroreflex sensitivity parameters were reduced in cases with PEI compared with controls (all p < .05). The heart rate variability parameters root mean square of successive RR interval differences (p = .05) and high frequency (p = .04) were also reduced. We found no difference in orthostatic hypotension between the groups. CONCLUSIONS In this first-of-its-kind study, we found that diabetes patients with PEI had reduced autonomic function compared with matched controls. Although numbers are small, results support the hypothesis that autonomic dysfunction could be a contributor to PEI in diabetes.
Collapse
Affiliation(s)
- Dag André Sangnes
- Department of Medicine, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Norway
| | | | - Rose Marie Moss
- Department of Clinical Medicine, University of Bergen, Norway
| | - Trond Engjom
- Department of Medicine, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Norway
| | - Eirik Søfteland
- Department of Medicine, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Norway.,Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
20
|
Mareninova OA, Vegh ET, Shalbueva N, Wightman CJ, Dillon DL, Malla S, Xie Y, Takahashi T, Rakonczay Z, French SW, Gaisano HY, Gorelick FS, Pandol SJ, Bensinger SJ, Davidson NO, Dawson DW, Gukovsky I, Gukovskaya AS. Dysregulation of mannose-6-phosphate-dependent cholesterol homeostasis in acinar cells mediates pancreatitis. J Clin Invest 2021; 131:146870. [PMID: 34128834 PMCID: PMC8321573 DOI: 10.1172/jci146870] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/11/2021] [Indexed: 01/18/2023] Open
Abstract
Disordered lysosomal/autophagy pathways initiate and drive pancreatitis, but the underlying mechanisms and links to disease pathology are poorly understood. Here, we show that the mannose-6-phosphate (M6P) pathway of hydrolase delivery to lysosomes critically regulates pancreatic acinar cell cholesterol metabolism. Ablation of the Gnptab gene encoding a key enzyme in the M6P pathway disrupted acinar cell cholesterol turnover, causing accumulation of nonesterified cholesterol in lysosomes/autolysosomes, its depletion in the plasma membrane, and upregulation of cholesterol synthesis and uptake. We found similar dysregulation of acinar cell cholesterol, and a decrease in GNPTAB levels, in both WT experimental pancreatitis and human disease. The mechanisms mediating pancreatic cholesterol dyshomeostasis in Gnptab-/- and experimental models involve a disordered endolysosomal system, resulting in impaired cholesterol transport through lysosomes and blockage of autophagic flux. By contrast, in Gnptab-/- liver the endolysosomal system and cholesterol homeostasis were largely unaffected. Gnptab-/- mice developed spontaneous pancreatitis. Normalization of cholesterol metabolism by pharmacologic means alleviated responses of experimental pancreatitis, particularly trypsinogen activation, the disease hallmark. The results reveal the essential role of the M6P pathway in maintaining exocrine pancreas homeostasis and function, and implicate cholesterol disordering in the pathogenesis of pancreatitis.
Collapse
Affiliation(s)
- Olga A. Mareninova
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Eszter T. Vegh
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Pathophysiology, University of Szeged, Szeged, Hungary
| | - Natalia Shalbueva
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Carli J.M. Wightman
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Dustin L. Dillon
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Sudarshan Malla
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Yan Xie
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Zoltan Rakonczay
- Department of Pathophysiology, University of Szeged, Szeged, Hungary
| | - Samuel W. French
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, California, USA
| | | | - Fred S. Gorelick
- Departments of Cell Biology and Internal Medicine, Yale University School of Medicine and VA West Haven, West Haven, Connecticut, USA
| | - Stephen J. Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | | - Nicholas O. Davidson
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David W. Dawson
- Department of Pathology and Laboratory Medicine and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Ilya Gukovsky
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Anna S. Gukovskaya
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| |
Collapse
|
21
|
Domínguez-Bendala J, Qadir MMF, Pastori RL. Temporal single-cell regeneration studies: the greatest thing since sliced pancreas? Trends Endocrinol Metab 2021; 32:433-443. [PMID: 34006411 PMCID: PMC8239162 DOI: 10.1016/j.tem.2021.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/12/2021] [Accepted: 04/16/2021] [Indexed: 01/01/2023]
Abstract
The application of single-cell analytic techniques to the study of stem/progenitor cell niches supports the emerging view that pancreatic cell lineages are in a state of flux between differentiation stages. For all their value, however, such analyses merely offer a snapshot of the cellular palette of the tissue at any given time point. Conclusions about potential developmental/regeneration paths are solely based on bioinformatics inferences. In this context, the advent of new techniques for the long-term culture and lineage tracing of human pancreatic slices offers a virtual window into the native organ and presents the field with a unique opportunity to serially resolve pancreatic regeneration dynamics at the single-cell level.
Collapse
Affiliation(s)
- Juan Domínguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Mirza Muhammad Fahd Qadir
- Section of Endocrinology and Metabolism, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - Ricardo Luis Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
22
|
Petersen OH, Gerasimenko JV, Gerasimenko OV, Gryshchenko O, Peng S. The roles of calcium and ATP in the physiology and pathology of the exocrine pancreas. Physiol Rev 2021; 101:1691-1744. [PMID: 33949875 DOI: 10.1152/physrev.00003.2021] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
This review deals with the roles of calcium ions and ATP in the control of the normal functions of the different cell types in the exocrine pancreas as well as the roles of these molecules in the pathophysiology of acute pancreatitis. Repetitive rises in the local cytosolic calcium ion concentration in the apical part of the acinar cells not only activate exocytosis but also, via an increase in the intramitochondrial calcium ion concentration, stimulate the ATP formation that is needed to fuel the energy-requiring secretion process. However, intracellular calcium overload, resulting in a global sustained elevation of the cytosolic calcium ion concentration, has the opposite effect of decreasing mitochondrial ATP production, and this initiates processes that lead to necrosis. In the last few years it has become possible to image calcium signaling events simultaneously in acinar, stellate, and immune cells in intact lobules of the exocrine pancreas. This has disclosed processes by which these cells interact with each other, particularly in relation to the initiation and development of acute pancreatitis. By unraveling the molecular mechanisms underlying this disease, several promising therapeutic intervention sites have been identified. This provides hope that we may soon be able to effectively treat this often fatal disease.
Collapse
Affiliation(s)
- Ole H Petersen
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | | | | | | | - Shuang Peng
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
23
|
Huber MK, Drotar DM, Hiller H, Beery ML, Joseph P, Kusmartseva I, Speier S, Atkinson MA, Mathews CE, Phelps EA. Observing Islet Function and Islet-Immune Cell Interactions in Live Pancreatic Tissue Slices. J Vis Exp 2021:10.3791/62207. [PMID: 33900291 PMCID: PMC8314551 DOI: 10.3791/62207] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Live pancreatic tissue slices allow for the study of islet physiology and function in the context of an intact islet microenvironment. Slices are prepared from live human and mouse pancreatic tissue embedded in agarose and cut using a vibratome. This method allows for the tissue to maintain viability and function in addition to preserving underlying pathologies such as type 1 (T1D) and type 2 diabetes (T2D). The slice method enables new directions in the study of the pancreas through the maintenance of the complex structures and various intercellular interactions that comprise the endocrine and exocrine tissues of the pancreas. This protocol demonstrates how to perform staining and time-lapse microscopy of live endogenous immune cells within pancreatic slices along with assessments of islet physiology. Further, this approach can be refined to discern immune cell populations specific for islet cell antigens using major histocompatibility complex-multimer reagents.
Collapse
Affiliation(s)
- Mollie K Huber
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida
| | - Denise M Drotar
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden; Institute of Physiology, Faculty of Medicine, Technische Universität Dresden; German Center for Diabetes Research (DZD)
| | - Helmut Hiller
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida
| | - Maria L Beery
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida
| | - Paul Joseph
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida
| | - Irina Kusmartseva
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida
| | - Stephan Speier
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden; Institute of Physiology, Faculty of Medicine, Technische Universität Dresden; German Center for Diabetes Research (DZD)
| | - Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida
| | - Clayton E Mathews
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida
| | - Edward A Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida;
| |
Collapse
|
24
|
Dolai S, Takahashi T, Qin T, Liang T, Xie L, Kang F, Miao YF, Xie H, Kang Y, Manuel J, Winter E, Roche PA, Cattral MS, Gaisano HY. Pancreas-specific SNAP23 depletion prevents pancreatitis by attenuating pathological basolateral exocytosis and formation of trypsin-activating autolysosomes. Autophagy 2020; 17:3068-3081. [PMID: 33213278 DOI: 10.1080/15548627.2020.1852725] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Intrapancreatic trypsin activation by dysregulated macroautophagy/autophagy and pathological exocytosis of zymogen granules (ZGs), along with activation of inhibitor of NFKB/NF-κB kinase (IKK) are necessary early cellular events in pancreatitis. How these three pancreatitis events are linked is unclear. We investigated how SNAP23 orchestrates these events leading to pancreatic acinar injury. SNAP23 depletion was by knockdown (SNAP23-KD) effected by adenovirus-shRNA (Ad-SNAP23-shRNA/mCherry) treatment of rodent and human pancreatic slices and in vivo by infusion into rat pancreatic duct. In vitro pancreatitis induction by supraphysiological cholecystokinin (CCK) or ethanol plus low-dose CCK were used to assess SNAP23-KD effects on exocytosis and autophagy. Pancreatitis stimuli resulted in SNAP23 translocation from its native location at the plasma membrane to autophagosomes, where SNAP23 would bind and regulate STX17 (syntaxin17) SNARE complex-mediated autophagosome-lysosome fusion. This SNAP23 relocation was attributed to IKBKB/IKKβ-mediated SNAP23 phosphorylation at Ser95 Ser120 in rat and Ser120 in human, which was blocked by IKBKB/IKKβ inhibitors, and confirmed by the inability of IKBKB/IKKβ phosphorylation-disabled SNAP23 mutant (Ser95A Ser120A) to bind STX17 SNARE complex. SNAP23-KD impaired the assembly of STX4-driven basolateral exocytotic SNARE complex and STX17-driven SNARE complex, causing respective reduction of basolateral exocytosis of ZGs and autolysosome formation, with consequent reduction in trypsinogen activation in both compartments. Consequently, pancreatic SNAP23-KD rats were protected from caerulein and alcoholic pancreatitis. This study revealed the roles of SNAP23 in mediating pathological basolateral exocytosis and IKBKB/IKKβ's involvement in autolysosome formation, both where trypsinogen activation would occur to cause pancreatitis. SNAP23 is a strong candidate to target for pancreatitis therapy.
Collapse
Affiliation(s)
- Subhankar Dolai
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | | | - Tairan Qin
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tao Liang
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Li Xie
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Fei Kang
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yi-Fan Miao
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Huanli Xie
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Youhou Kang
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Justin Manuel
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Erin Winter
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Paul A Roche
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark S Cattral
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Herbert Y Gaisano
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Bernard H, Teijeiro A, Chaves-Pérez A, Perna C, Satish B, Novials A, Wang JP, Djouder N. Coxsackievirus B Type 4 Infection in β Cells Downregulates the Chaperone Prefoldin URI to Induce a MODY4-like Diabetes via Pdx1 Silencing. CELL REPORTS MEDICINE 2020; 1:100125. [PMID: 33205075 PMCID: PMC7659558 DOI: 10.1016/j.xcrm.2020.100125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/06/2020] [Accepted: 09/22/2020] [Indexed: 12/17/2022]
Abstract
Enteroviruses are suspected to contribute to insulin-producing β cell loss and hyperglycemia-induced diabetes. However, mechanisms are not fully defined. Here, we show that coxsackievirus B type 4 (CVB4) infection in human islet-engrafted mice and in rat insulinoma cells displays loss of unconventional prefoldin RPB5 interactor (URI) and PDX1, affecting β cell function and identity. Genetic URI ablation in the mouse pancreas causes PDX1 depletion in β cells. Importantly, diabetic PDX1 heterozygous mice overexpressing URI in β cells are more glucose tolerant. Mechanistically, URI loss triggers estrogen receptor nuclear translocation leading to DNA methyltransferase 1 (DNMT1) expression, which induces Pdx1 promoter hypermethylation and silencing. Consequently, demethylating agent procainamide-mediated DNMT1 inhibition reinstates PDX1 expression and protects against diabetes in pancreatic URI-depleted mice . Finally, the β cells of human diabetes patients show correlations between viral protein 1 and URI, PDX1, and DNMT1 levels. URI and DNMT1 expression and PDX1 silencing provide a causal link between enterovirus infection and diabetes. Coxsackievirus B type 4 infection downregulates URI and affects β cell function Genetic URI ablation in mouse pancreas recapitulates diabetes URI controls Pdx1 methylation via ERα-activating DNMT1 Coxsackievirus B type 4, URI, PDX1, and DNMT1 expression correlate in human pancreata
Collapse
MESH Headings
- Animals
- Capsid Proteins/genetics
- Capsid Proteins/metabolism
- Coxsackievirus Infections/genetics
- Coxsackievirus Infections/metabolism
- Coxsackievirus Infections/pathology
- Coxsackievirus Infections/virology
- DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors
- DNA (Cytosine-5-)-Methyltransferase 1/genetics
- DNA (Cytosine-5-)-Methyltransferase 1/metabolism
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/virology
- Disease Models, Animal
- Enterovirus B, Human/genetics
- Enterovirus B, Human/metabolism
- Enterovirus B, Human/pathogenicity
- Female
- Gene Expression Regulation
- Glucose/metabolism
- Glucose/pharmacology
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Insulin-Secreting Cells/transplantation
- Male
- Mice
- Mice, Transgenic
- Procainamide/pharmacology
- Rats
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Hugo Bernard
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid 28029, Spain
| | - Ana Teijeiro
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid 28029, Spain
| | - Almudena Chaves-Pérez
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid 28029, Spain
| | - Cristian Perna
- Department of Pathology, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid 28034, Spain
| | - Basanthi Satish
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Anna Novials
- IDIBAPS, August Pi i Sunyer Biomedical Research Institute and, CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Barcelona, Spain
| | - Jennifer P. Wang
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Nabil Djouder
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid 28029, Spain
- Corresponding author
| |
Collapse
|
26
|
The role of Ca2+ signalling in the physiology and pathophysiology of exocrine pancreas. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.07.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
27
|
Egozi A, Bahar Halpern K, Farack L, Rotem H, Itzkovitz S. Zonation of Pancreatic Acinar Cells in Diabetic Mice. Cell Rep 2020; 32:108043. [PMID: 32814046 PMCID: PMC7443616 DOI: 10.1016/j.celrep.2020.108043] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/02/2020] [Accepted: 07/24/2020] [Indexed: 12/25/2022] Open
Abstract
The islets of Langerhans are dynamic structures that can change in size, number of cells, and molecular function in response to physiological and pathological stress. Molecular cues originating from the surrounding "peri-islet" acinar cells that could facilitate this plasticity have not been explored. Here, we combine single-molecule transcript imaging in the intact pancreas and transcriptomics to identify spatial heterogeneity of acinar cell gene expression. We find that peri-islet acinar cells exhibit a distinct molecular signature in db/db diabetic mice that includes upregulation of trypsin family genes and elevated mTOR activity. This zonated expression program seems to be induced by CCK that is secreted from islet cells. Elevated peri-islet trypsin secretion could facilitate the islet expansion observed in this model via modulation of the islet capsule matrix components. Our study highlights a molecular axis of communication between the pancreatic exocrine and endocrine compartments that may be relevant to islet expansion.
Collapse
Affiliation(s)
- Adi Egozi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Keren Bahar Halpern
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Lydia Farack
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Hagar Rotem
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Shalev Itzkovitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
28
|
De Faveri F, Chvanov M, Voronina S, Moore D, Pollock L, Haynes L, Awais M, Beckett AJ, Mayer U, Sutton R, Criddle DN, Prior IA, Wileman T, Tepikin AV. LAP-like non-canonical autophagy and evolution of endocytic vacuoles in pancreatic acinar cells. Autophagy 2020; 16:1314-1331. [PMID: 31651224 PMCID: PMC7469629 DOI: 10.1080/15548627.2019.1679514] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 08/30/2019] [Accepted: 10/07/2019] [Indexed: 12/20/2022] Open
Abstract
Activation of trypsinogen (formation of trypsin) inside the pancreas is an early pathological event in the development of acute pancreatitis. In our previous studies we identified the activation of trypsinogen within endocytic vacuoles (EVs), cellular organelles that appear in pancreatic acinar cells treated with the inducers of acute pancreatitis. EVs are formed as a result of aberrant compound exocytosis and subsequent internalization of post-exocytic structures. These organelles can be up to 12 μm in diameter and can be actinated (i.e. coated with F-actin). Notably, EVs can undergo intracellular rupture and fusion with the plasma membrane, providing trypsin with access to cytoplasmic and extracellular targets. Unraveling the mechanisms involved in cellular processing of EVs is an interesting cell biological challenge with potential benefits for understanding acute pancreatitis. In this study we have investigated autophagy of EVs and discovered that it involves a non-canonical LC3-conjugation mechanism, reminiscent in its properties to LC3-associated phagocytosis (LAP); in both processes LC3 was recruited to single, outer organellar membranes. Trypsinogen activation peptide was observed in approximately 55% of LC3-coated EVs indicating the relevance of the described process to the early cellular events of acute pancreatitis. We also investigated relationships between actination and non-canonical autophagy of EVs and concluded that these processes represent sequential steps in the evolution of EVs. Our study expands the known roles of LAP and indicates that, in addition to its well-established functions in phagocytosis and macropinocytosis, LAP is also involved in the processing of post-exocytic organelles in exocrine secretory cells. ABBREVIATIONS AP: acute pancreatitis; CCK: cholecystokinin; CLEM: correlative light and electron microscopy; DPI: diphenyleneiodonium; EV: endocytic vacuole; LAP: LC3-associate phagocytosis; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; PACs: pancreatic acinar cells; PFA: paraformaldehyde; PtdIns3K: phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol 3-phosphate; Res: resveratrol; TAP: trypsinogen activation peptide; TEM: transmission electron microscopy; TLC-S: taurolithocholic acid 3-sulfate; TRD: Dextran Texas Red 3000 MW Neutral; ZGs: zymogen granules.
Collapse
Affiliation(s)
- Francesca De Faveri
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Michael Chvanov
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Svetlana Voronina
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Danielle Moore
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Liam Pollock
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Lee Haynes
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Muhammad Awais
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Alison J. Beckett
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Ulrike Mayer
- Bio-Medical Research Centre, Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Robert Sutton
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - David N. Criddle
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Ian A. Prior
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Tom Wileman
- Bio-Medical Research Centre, Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Alexei V. Tepikin
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| |
Collapse
|
29
|
Qadir MMF, Álvarez-Cubela S, Weitz J, Panzer JK, Klein D, Moreno-Hernández Y, Cechin S, Tamayo A, Almaça J, Hiller H, Beery M, Kusmartseva I, Atkinson M, Speier S, Ricordi C, Pugliese A, Caicedo A, Fraker CA, Pastori RL, Domínguez-Bendala J. Long-term culture of human pancreatic slices as a model to study real-time islet regeneration. Nat Commun 2020; 11:3265. [PMID: 32601271 PMCID: PMC7324563 DOI: 10.1038/s41467-020-17040-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 06/04/2020] [Indexed: 01/02/2023] Open
Abstract
The culture of live pancreatic tissue slices is a powerful tool for the interrogation of physiology and pathology in an in vitro setting that retains near-intact cytoarchitecture. However, current culture conditions for human pancreatic slices (HPSs) have only been tested for short-term applications, which are not permissive for the long-term, longitudinal study of pancreatic endocrine regeneration. Using a culture system designed to mimic the physiological oxygenation of the pancreas, we demonstrate high viability and preserved endocrine and exocrine function in HPS for at least 10 days after sectioning. This extended lifespan allowed us to dynamically lineage trace and quantify the formation of insulin-producing cells in HPS from both non-diabetic and type 2 diabetic donors. This technology is expected to be of great impact for the conduct of real-time regeneration/developmental studies in the human pancreas.
Collapse
Affiliation(s)
- Mirza Muhammad Fahd Qadir
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Jonathan Weitz
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Julia K Panzer
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Dagmar Klein
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Yaisa Moreno-Hernández
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Universidad Francisco de Vitoria, Madrid, Spain
| | - Sirlene Cechin
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alejandro Tamayo
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Joana Almaça
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Helmut Hiller
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Maria Beery
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Irina Kusmartseva
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Mark Atkinson
- nPOD Laboratory, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32611, USA
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Stephan Speier
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
- Faculty of Medicine, Institute of Physiology, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München, Neuherberg, Germany
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alberto Pugliese
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Alejandro Caicedo
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Christopher A Fraker
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Biomedical Engineering, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Ricardo Luis Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Juan Domínguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
30
|
Chung KM, Singh J, Lawres L, Dorans KJ, Garcia C, Burkhardt DB, Robbins R, Bhutkar A, Cardone R, Zhao X, Babic A, Vayrynen SA, Dias Costa A, Nowak JA, Chang DT, Dunne RF, Hezel AF, Koong AC, Wilhelm JJ, Bellin MD, Nylander V, Gloyn AL, McCarthy MI, Kibbey RG, Krishnaswamy S, Wolpin BM, Jacks T, Fuchs CS, Muzumdar MD. Endocrine-Exocrine Signaling Drives Obesity-Associated Pancreatic Ductal Adenocarcinoma. Cell 2020; 181:832-847.e18. [PMID: 32304665 PMCID: PMC7266008 DOI: 10.1016/j.cell.2020.03.062] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/13/2020] [Accepted: 03/27/2020] [Indexed: 12/30/2022]
Abstract
Obesity is a major modifiable risk factor for pancreatic ductal adenocarcinoma (PDAC), yet how and when obesity contributes to PDAC progression is not well understood. Leveraging an autochthonous mouse model, we demonstrate a causal and reversible role for obesity in early PDAC progression, showing that obesity markedly enhances tumorigenesis, while genetic or dietary induction of weight loss intercepts cancer development. Molecular analyses of human and murine samples define microenvironmental consequences of obesity that foster tumorigenesis rather than new driver gene mutations, including significant pancreatic islet cell adaptation in obesity-associated tumors. Specifically, we identify aberrant beta cell expression of the peptide hormone cholecystokinin (Cck) in response to obesity and show that islet Cck promotes oncogenic Kras-driven pancreatic ductal tumorigenesis. Our studies argue that PDAC progression is driven by local obesity-associated changes in the tumor microenvironment and implicate endocrine-exocrine signaling beyond insulin in PDAC development.
Collapse
Affiliation(s)
| | - Jaffarguriqbal Singh
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Lauren Lawres
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | | | - Cathy Garcia
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Daniel B Burkhardt
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Rebecca Robbins
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA
| | - Arjun Bhutkar
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA
| | - Rebecca Cardone
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xiaojian Zhao
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02114, USA
| | - Sara A Vayrynen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02114, USA
| | - Andressa Dias Costa
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02114, USA
| | - Jonathan A Nowak
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel T Chang
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford, CA 94305, USA
| | - Richard F Dunne
- Division of Hematology and Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Aram F Hezel
- Division of Hematology and Oncology, Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Albert C Koong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joshua J Wilhelm
- Schulze Diabetes Institute and Department of Surgery, University of Minnesota Medical Center, Minneapolis, MN 55454, USA
| | - Melena D Bellin
- Schulze Diabetes Institute and Department of Surgery, University of Minnesota Medical Center, Minneapolis, MN 55454, USA; Department of Pediatrics, University of Minnesota Medical Center, Minneapolis, MN 55454, USA
| | - Vibe Nylander
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Anna L Gloyn
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK; Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford OX3 7LE, UK
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK; Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford OX3 7LE, UK
| | - Richard G Kibbey
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Smita Krishnaswamy
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02114, USA
| | - Tyler Jacks
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Charles S Fuchs
- Yale Cancer Center, Smilow Cancer Hospital, New Haven, CT 06511, USA
| | - Mandar Deepak Muzumdar
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA; Yale Cancer Center, Smilow Cancer Hospital, New Haven, CT 06511, USA.
| |
Collapse
|
31
|
Takahashi T, Miao Y, Kang F, Dolai S, Gaisano HY. Susceptibility Factors and Cellular Mechanisms Underlying Alcoholic Pancreatitis. Alcohol Clin Exp Res 2020; 44:777-789. [PMID: 32056245 DOI: 10.1111/acer.14304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/03/2020] [Indexed: 12/16/2022]
Abstract
Alcohol is a major cause of acute and chronic pancreatitis. There have been some recent advances in the understanding of the mechanisms underlying alcoholic pancreatitis, which include perturbation in mitochondrial function and autophagy and ectopic exocytosis, with some of these cellular events involving membrane fusion soluble N-ethylmaleimide-sensitive factor receptor protein receptor proteins. Although new insights have been unraveled recently, the precise mechanisms remain complex, and their finer details have yet to be established. The overall pathophysiology of pancreatitis involves not only the pancreatic acinar cells but also the stellate cells and duct cells. Why only some are more susceptible to pancreatitis and with increased severity, while others are not, would suggest that there may be undefined protective factors or mechanisms that enhance recovery and regeneration after injury. Furthermore, there are confounding influences of lifestyle factors such as smoking and diet, and genetic background. Whereas alcohol and smoking cessation and a generally healthy lifestyle are intuitively the advice given to these patients afflicted with alcoholic pancreatitis in order to reduce disease recurrence and progression, there is as yet no specific treatment. A more complete understanding of the pathogenesis of pancreatitis from which novel therapeutic targets could be identified will have a great impact, particularly with the stubbornly high fatality (>30%) of severe pancreatitis. This review focuses on the susceptibility factors and underlying cellular mechanisms of alcohol injury on the exocrine pancreas.
Collapse
Affiliation(s)
- Toshimasa Takahashi
- From the, Departments of Medicine and Physiology, University of Toronto, Toronto, ON, Canada
| | - Yifan Miao
- From the, Departments of Medicine and Physiology, University of Toronto, Toronto, ON, Canada
| | - Fei Kang
- From the, Departments of Medicine and Physiology, University of Toronto, Toronto, ON, Canada
| | - Subhankar Dolai
- From the, Departments of Medicine and Physiology, University of Toronto, Toronto, ON, Canada
| | - Herbert Y Gaisano
- From the, Departments of Medicine and Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
Egberts JH, Raza GS, Wilgus C, Teyssen S, Kiehne K, Herzig KH. Release of Cholecystokinin from Rat Intestinal Mucosal Cells and the Enteroendocrine Cell Line STC-1 in Response to Maleic and Succinic Acid, Fermentation Products of Alcoholic Beverages. Int J Mol Sci 2020; 21:ijms21020589. [PMID: 31963306 PMCID: PMC7013850 DOI: 10.3390/ijms21020589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 02/06/2023] Open
Abstract
Alcoholic beverages stimulate pancreatic enzyme secretions by inducing cholecystokinin (CCK) release. CCK is the major stimulatory hormone of pancreatic exocrine secretions, secreted from enteroendocrine I-cells of the intestine. Fermentation products of alcoholic beverages, such as maleic and succinic acids, influence gastric acid secretions. We hypothesize that maleic and succinic acids stimulate pancreatic exocrine secretions during beer and wine ingestion by increasing CCK secretions. Therefore, the effects of maleic and succinic acids on CCK release were studied in duodenal mucosal cells and the enteroendocrine cell line STC-1. Mucosal cells were perfused for 30 min with 5 min sampling intervals, STC-1 cells were studied under static incubation for 15 min, and supernatants were collected for CCK measurements. Succinate and maleate-induced CCK release were investigated. Succinate and maleate doses dependently stimulated CCK secretions from mucosal cells and STC-1 cells. Diltiazem, a calcium channel blocker, significantly inhibited succinate and maleate-induced CCK secretions from mucosal cells and STC-1 cells. Maleate and succinate did not show cytotoxicity in STC-1 cells. Our results indicate that succinate and maleate are novel CCK-releasing factors in fermented alcoholic beverages and could contribute to pancreatic exocrine secretions and their pathophysiology.
Collapse
Affiliation(s)
- Jan-Hendrik Egberts
- University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany; (J.-H.E.); (C.W.); (K.K.)
| | - Ghulam Shere Raza
- Research Unit of Biomedicine, University of Oulu, 90014 Oulu, Finland;
| | - Cornelia Wilgus
- University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany; (J.-H.E.); (C.W.); (K.K.)
| | | | - Karlheinz Kiehne
- University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany; (J.-H.E.); (C.W.); (K.K.)
| | - Karl-Heinz Herzig
- University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany; (J.-H.E.); (C.W.); (K.K.)
- Research Unit of Biomedicine, University of Oulu, 90014 Oulu, Finland;
- Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, 61-701 Poznan, Poland
- Medical Research Center Oulu and Oulu University Hospital, 90014 Oulu, Finland
- Correspondence:
| |
Collapse
|
33
|
Peters L, Posgai A, Brusko TM. Islet-immune interactions in type 1 diabetes: the nexus of beta cell destruction. Clin Exp Immunol 2019; 198:326-340. [PMID: 31309537 DOI: 10.1111/cei.13349] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2019] [Indexed: 12/12/2022] Open
Abstract
Recent studies in Type 1 Diabetes (T1D) support an emerging model of disease pathogenesis that involves intrinsic β-cell fragility combined with defects in both innate and adaptive immune cell regulation. This combination of defects induces systematic changes leading to organ-level atrophy and dysfunction of both the endocrine and exocrine portions of the pancreas, ultimately culminating in insulin deficiency and β-cell destruction. In this review, we discuss the animal model data and human tissue studies that have informed our current understanding of the cross-talk that occurs between β-cells, the resident stroma, and immune cells that potentiate T1D. Specifically, we will review the cellular and molecular signatures emerging from studies on tissues derived from organ procurement programs, focusing on in situ defects occurring within the T1D islet microenvironment, many of which are not yet detectable by standard peripheral blood biomarkers. In addition to improved access to organ donor tissues, various methodological advances, including immune receptor repertoire sequencing and single-cell molecular profiling, are poised to improve our understanding of antigen-specific autoimmunity during disease development. Collectively, the knowledge gains from these studies at the islet-immune interface are enhancing our understanding of T1D heterogeneity, likely to be an essential component for instructing future efforts to develop targeted interventions to restore immune tolerance and preserve β-cell mass and function.
Collapse
Affiliation(s)
- L Peters
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - A Posgai
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - T M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| |
Collapse
|
34
|
Gál E, Dolenšek J, Stožer A, Pohorec V, Ébert A, Venglovecz V. A Novel in situ Approach to Studying Pancreatic Ducts in Mice. Front Physiol 2019; 10:938. [PMID: 31396104 PMCID: PMC6668154 DOI: 10.3389/fphys.2019.00938] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/09/2019] [Indexed: 12/19/2022] Open
Abstract
Introduction: The tissue slice technique offers several benefits compared to isolated cells and cell clusters that help us understand the (patho)physiology of several organs in situ. The most prominent features are preserved architecture and function, with intact homotypic and heterotypic interactions between cells in slices. In the pancreas, this technique has been utilized successfully to study acinar and endocrine islet cells. However, it has never been used to investigate ductal function. Since pancreatic ductal epithelial cells (PDECs) play an essential role in the physiology of the pancreas, our aim was to use this technique to study PDEC structure and function in situ. Materials and methods: Eight- to sixteen weeks old C57BL/6 mice were used for preparation of pancreas tissue slices. Low melting point agarose was injected into the common bile duct and the whole organ was extracted. For morphological studies, pieces of tissue were embedded in agarose and cryosectioned to obtain 15 μm thick slices. In order to visualize pancreatic ducts, (i) the Giemsa dye was added to the agarose and visualized using light microscopy or (ii) immunostaining for the cystic fibrosis transmembrane conductance regulator (CFTR) was performed. For functional characterization, agarose-embedded tissue was immediately cut to 140 μm thick tissue slices that were loaded with the cell permeant form of the Oregon Green 488 BAPTA-1 dye and used for confocal calcium imaging. Results: Giemsa staining has shown that the injected agarose reaches the head and body of the pancreas to a greater extent than the tail, without disrupting the tissue architecture. Strong CFTR expression was detected at the apical membranes of PDECs and acinar cells, whereas islet cells were completely negative for CFTR. Stimulation with chenodeoxycholic acid (CDCA, 1 mM) resulted in a robust transient increase in intracellular calcium concentration that was readily visible in >40 ductal cells per slice. Conclusion: Our results confirm that the acutely-isolated pancreas tissue slice technique is suitable for structural and functional investigation of PDECs and their relationship with other cell types, such as acini and endocrine cells in situ. In combination with different genetic, pharmacological or dietary approaches it could become a method of choice in the foreseeable future.
Collapse
Affiliation(s)
- Eleonóra Gál
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Jurij Dolenšek
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.,Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Viljem Pohorec
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Attila Ébert
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Viktória Venglovecz
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| |
Collapse
|
35
|
Bharmal SH, Pendharkar SA, Singh RG, Petrov MS. Associations between gastrointestinal humoral factors and pancreatic proteolytic enzymes in alcohol-related versus non-alcohol-related pancreatitis. Alcohol 2019; 76:1-10. [PMID: 30529016 DOI: 10.1016/j.alcohol.2018.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 06/25/2018] [Accepted: 06/25/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alcohol-related pancreatitis is common and the gastrointestinal tract plays an important role in the regulation of pancreatic exocrine function. While the relationship between pancreatic proteolytic enzymes and insulin (as well as other pancreatic hormones) has been investigated in detail, little is known about the relationship between pancreatic proteolytic enzymes and gastrointestinal humoral factors. The aim of this study was to study the associations between trypsin, chymotrypsin, and a panel of gastrointestinal humoral factors in patients after an episode of alcohol-related versus non-alcohol-related pancreatitis. METHODS Fasting venous blood samples were analyzed for trypsin, chymotrypsin, cholecystokinin, gastrin, ghrelin, gastrin-related peptide, neuropeptide Y, peptide YY, secretin, and vasoactive intestinal peptide. Linear regression analysis was used in three statistical models, adjusting for covariates (age, sex, ethnicity, smoking, exercise, body mass index, dysglycemia, recurrence of pancreatitis, duration of pancreatitis, and severity of pancreatitis). RESULTS The study included 21 patients with alcohol-related pancreatitis and 72 with non-alcohol-related pancreatitis. Gastrin, cholecystokinin, and vasoactive intestinal peptide were significantly associated with chymotrypsin in all three statistical models and resulted in a 1.06, 1.98, and 2.74 times higher chymotrypsin level in alcohol-related pancreatitis, respectively. Ghrelin was significantly associated with trypsin in all three statistical models and resulted in a 2.64 times higher trypsin level in alcohol-related pancreatitis. Other associations did not demonstrate a consistent significant pattern. CONCLUSION In alcohol-related pancreatitis, several gut-related peptides are significantly associated with pancreatic exocrine function. Further studies to investigate the effect of alcohol on the interaction between cholecystokinin (as well as gastrin, ghrelin, and vasoactive intestinal peptide) and pancreatic exocrine function are warranted.
Collapse
|
36
|
Gukovskaya AS, Gorelick FS, Groblewski GE, Mareninova OA, Lugea A, Antonucci L, Waldron RT, Habtezion A, Karin M, Pandol SJ, Gukovsky I. Recent Insights Into the Pathogenic Mechanism of Pancreatitis: Role of Acinar Cell Organelle Disorders. Pancreas 2019; 48:459-470. [PMID: 30973461 PMCID: PMC6461375 DOI: 10.1097/mpa.0000000000001298] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [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
Acute pancreatitis (AP) is a potentially lethal inflammatory disease that lacks specific therapy. Damaged pancreatic acinar cells are believed to be the site of AP initiation. The primary function of these cells is the synthesis, storage, and export of digestive enzymes. Beginning in the endoplasmic reticulum and ending with secretion of proteins stored in zymogen granules, distinct pancreatic organelles use ATP produced by mitochondria to move and modify nascent proteins through sequential vesicular compartments. Compartment-specific accessory proteins concentrate cargo and promote vesicular budding, targeting, and fusion. The autophagy-lysosomal-endosomal pathways maintain acinar cell homeostasis by removing damaged/dysfunctional organelles and recycling cell constituents for substrate and energy. Here, we discuss studies in experimental and genetic AP models, primarily from our groups, which show that acinar cell injury is mediated by distinct mechanisms of organelle dysfunction involved in protein synthesis and trafficking, secretion, energy generation, and autophagy. These early AP events (often first manifest by abnormal cytosolic Ca signaling) in the acinar cell trigger the inflammatory and cell death responses of pancreatitis. Manifestations of acinar cell organelle disorders are also prominent in human pancreatitis. Our findings suggest that targeting specific mediators of organelle dysfunction could reduce disease severity.
Collapse
Affiliation(s)
- Anna S. Gukovskaya
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles
- Department of Medicine, West Los Angeles VA Healthcare Center, Los Angeles, CA
| | - Fred S. Gorelick
- Department of Cell Biology Yale University School of Medicine, New Haven, CT
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Guy E. Groblewski
- Department of Nutritional Sciences, University of Wisconsin, Madison, WI
| | - Olga A. Mareninova
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles
- Department of Medicine, West Los Angeles VA Healthcare Center, Los Angeles, CA
| | - Aurelia Lugea
- Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Laura Antonucci
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine, La Jolla, CA
| | - Richard T. Waldron
- Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Aida Habtezion
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine, La Jolla, CA
| | - Stephen J. Pandol
- Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Ilya Gukovsky
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles
- Department of Medicine, West Los Angeles VA Healthcare Center, Los Angeles, CA
| |
Collapse
|
37
|
Williams JA. Cholecystokinin (CCK) Regulation of Pancreatic Acinar Cells: Physiological Actions and Signal Transduction Mechanisms. Compr Physiol 2019; 9:535-564. [PMID: 30873601 DOI: 10.1002/cphy.c180014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pancreatic acinar cells synthesize and secrete about 20 digestive enzymes and ancillary proteins with the processes that match the supply of these enzymes to their need in digestion being regulated by a number of hormones (CCK, secretin and insulin), neurotransmitters (acetylcholine and VIP) and growth factors (EGF and IGF). Of these regulators, one of the most important and best studied is the gastrointestinal hormone, cholecystokinin (CCK). Furthermore, the acinar cell has become a model for seven transmembrane, heterotrimeric G protein coupled receptors to regulate multiple processes by distinct signal transduction cascades. In this review, we briefly describe the chemistry and physiology of CCK and then consider the major physiological effects of CCK on pancreatic acinar cells. The majority of the review is devoted to the physiologic signaling pathways activated by CCK receptors and heterotrimeric G proteins and the functions they affect. The pathways covered include the traditional second messenger pathways PLC-IP3-Ca2+ , DAG-PKC, and AC-cAMP-PKA/EPAC that primarily relate to secretion. Then there are the protein-protein interaction pathways Akt-mTOR-S6K, the three major MAPK pathways (ERK, JNK, and p38 MAPK), and Ca2+ -calcineurin-NFAT pathways that primarily regulate non-secretory processes including biosynthesis and growth, and several miscellaneous pathways that include the Rho family small G proteins, PKD, FAK, and Src that may regulate both secretory and nonsecretory processes but are not as well understood. © 2019 American Physiological Society. Compr Physiol 9:535-564, 2019.
Collapse
Affiliation(s)
- John A Williams
- University of Michigan, Departments of Molecular & Integrative Physiology and Internal Medicine (Gastroenterology), Ann Arbor, Michigan, USA
| |
Collapse
|
38
|
Domínguez-Bendala J, Qadir MMF, Pastori RL. Pancreatic Progenitors: There and Back Again. Trends Endocrinol Metab 2019; 30:4-11. [PMID: 30502039 PMCID: PMC6354578 DOI: 10.1016/j.tem.2018.10.002] [Citation(s) in RCA: 19] [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: 08/20/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 02/06/2023]
Abstract
Adult pancreatic regeneration is one of the most contentious topics in modern biology. The long-held view that the islets of Langerhans can be replenished throughout adult life through the reactivation of ductal progenitor cells has been replaced over the past decade by the now prevailing notion that regeneration does not involve progenitors and occurs only through the duplication of pre-existing mature cells. Here we dissect the limitations of lineage tracing (LT) to draw categorical conclusions about pancreatic regeneration, especially in view of emerging evidence that traditional lineages are less homogeneous and cell fates more dynamic than previously thought. This new evidence further suggests that the two competing hypotheses about regeneration are not mutually exclusive.
Collapse
Affiliation(s)
- Juan Domínguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Mirza Muhammad Fahd Qadir
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ricardo Luis Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
39
|
Obafemi TF, Yu P, Li J, Davis JM, Liu K, Cheng B, Zhao X, Shen Q, Younes M, Ko TC, Cao Y. Comparable Responses in Male and Female Mice to Cerulein-Induced Chronic Pancreatic Injury and Recovery. JOP : JOURNAL OF THE PANCREAS 2018; 19:236-243. [PMID: 30636940 PMCID: PMC6327960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
OBJECTIVE The cerulein-induced mouse pancreatitis model is a well-established, commonly used representation of human chronic pancreatitis pathology. Although studies report sex-dependent differences in human chronic pancreatitis, there are no studies in this model directly comparing sex response to pancreatic injury and recovery. Therefore, we designed a study to investigate whether sex- dependent differences in chronic pancreatitis injury and recovery exist in the cerulein-induced pancreatitis model. METHODS Adult male and female C57BL/6 mice were administered cerulein (50 μg/kg, 5 hourly intraperitoneal injections/day, 3 days/week) for 4 weeks to induce chronic pancreatitis; control mice received normal saline injections. Pancreata and blood were harvested at 4 days (as injury group) or 4 weeks (as recovery group) after the last injection. Amylase secretion was measured from the serum. Acinar injury was scored on H&E sections. Fibrosis was assessed by Sirius Red and collagen immunofluorescence staining. RESULTS Compared to time-matched controls, injury group displayed decreased body and pancreas weight, and increased acinar injury and fibrosis, with no significant differences between males and females. Recovery group demonstrated recovery of body weight, partial recovery of pancreas weight, reversal of acinar injury, and partial reversal of fibrosis, with no significant differences between males and females. Amylase secretion/body weight was similar across all groups. CONCLUSIONS Male and female mice of the cerulein-induced chronic pancreatitis demonstrate similar responses to chronic pancreatitis injury and recovery. Although this model may not sufficiently emulate sex-dependent responses in human chronic pancreatitis, our study supports that both sexes of mice from this model can be used for the study of chronic pancreatitis.
Collapse
Affiliation(s)
- Tolulope F Obafemi
- Department of Surgery, UTHealth, 6431 Fannin Street, Houston, TX 77030, USA
| | - Peter Yu
- Department of Surgery, UTHealth, 6431 Fannin Street, Houston, TX 77030, USA
| | - Jing Li
- Department of Surgery, UTHealth, 6431 Fannin Street, Houston, TX 77030, USA
- Department of Clinical Laboratory Science, The Affiliated Hospital of Qingdao University, 19 Jiangsu Road, Qingdao, Shandong 266003, China
| | - Joy M Davis
- Department of Surgery, UTHealth, 6431 Fannin Street, Houston, TX 77030, USA
| | - Ka Liu
- Department of Surgery, UTHealth, 6431 Fannin Street, Houston, TX 77030, USA
| | - Binglu Cheng
- Department of Surgery, UTHealth, 6431 Fannin Street, Houston, TX 77030, USA
| | - Xiurong Zhao
- Department of Neurology, UTHealth, 6431 Fannin Street, Houston, TX 77030, USA
| | - Qiang Shen
- Department of Clinical Cancer Prevention, Division of Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Mamoun Younes
- Department of Pathology & Laboratory Medicine, UTHealth, 6431 Fannin Street, Houston, TX 77030, USA
| | - Tien C Ko
- Department of Surgery, UTHealth, 6431 Fannin Street, Houston, TX 77030, USA
| | - Yanna Cao
- Department of Surgery, UTHealth, 6431 Fannin Street, Houston, TX 77030, USA
| |
Collapse
|
40
|
Chvanov M, De Faveri F, Moore D, Sherwood MW, Awais M, Voronina S, Sutton R, Criddle DN, Haynes L, Tepikin AV. Intracellular rupture, exocytosis and actin interaction of endocytic vacuoles in pancreatic acinar cells: initiating events in acute pancreatitis. J Physiol 2018; 596:2547-2564. [PMID: 29717784 PMCID: PMC6023832 DOI: 10.1113/jp275879] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/20/2018] [Indexed: 12/18/2022] Open
Abstract
Key points Giant trypsin‐containing endocytic vacuoles are formed in pancreatic acinar cells stimulated with inducers of acute pancreatitis. F‐actin envelops endocytic vacuoles and regulates their properties. Endocytic vacuoles can rupture and release their content into the cytosol of acinar cells. Endocytic vacuoles can fuse with the plasma membrane of acinar cells and exocytose their content.
Abstract Intrapancreatic activation of trypsinogen is an early event in and hallmark of the development of acute pancreatitis. Endocytic vacuoles, which form by disconnection and transport of large post‐exocytic structures, are the only resolvable sites of the trypsin activity in live pancreatic acinar cells. In the present study, we characterized the dynamics of endocytic vacuole formation induced by physiological and pathophysiological stimuli and visualized a prominent actin coat that completely or partially surrounded endocytic vacuoles. An inducer of acute pancreatitis taurolithocholic acid 3‐sulphate and supramaximal concentrations of cholecystokinin triggered the formation of giant (more than 2.5 μm in diameter) endocytic vacuoles. We discovered and characterized the intracellular rupture of endocytic vacuoles and the fusion of endocytic vacuoles with basal and apical regions of the plasma membrane. Experiments with specific protease inhibitors suggest that the rupture of endocytic vacuoles is probably not induced by trypsin or cathepsin B. Perivacuolar filamentous actin (observed on the surface of ∼30% of endocytic vacuoles) may play a stabilizing role by preventing rupture of the vacuoles and fusion of the vacuoles with the plasma membrane. The rupture and fusion of endocytic vacuoles allow trypsin to escape the confinement of a membrane‐limited organelle, gain access to intracellular and extracellular targets, and initiate autodigestion of the pancreas, comprising a crucial pathophysiological event. Giant trypsin‐containing endocytic vacuoles are formed in pancreatic acinar cells stimulated with inducers of acute pancreatitis. F‐actin envelops endocytic vacuoles and regulates their properties. Endocytic vacuoles can rupture and release their content into the cytosol of acinar cells. Endocytic vacuoles can fuse with the plasma membrane of acinar cells and exocytose their content.
Collapse
Affiliation(s)
- Michael Chvanov
- Department of Cellular and Molecular Physiology and NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Liverpool, UK
| | - Francesca De Faveri
- Department of Cellular and Molecular Physiology and NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Liverpool, UK
| | - Danielle Moore
- Department of Cellular and Molecular Physiology and NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Liverpool, UK
| | - Mark W Sherwood
- Department of Cellular and Molecular Physiology and NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Liverpool, UK
| | - Muhammad Awais
- Department of Cellular and Molecular Physiology and NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Liverpool, UK
| | - Svetlana Voronina
- Department of Cellular and Molecular Physiology and NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Liverpool, UK
| | - Robert Sutton
- Department of Cellular and Molecular Physiology and NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Liverpool, UK
| | - David N Criddle
- Department of Cellular and Molecular Physiology and NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Liverpool, UK
| | - Lee Haynes
- Department of Cellular and Molecular Physiology and NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Liverpool, UK
| | - Alexei V Tepikin
- Department of Cellular and Molecular Physiology and NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Liverpool, UK
| |
Collapse
|
41
|
Dolai S, Liang T, Orabi AI, Holmyard D, Xie L, Greitzer-Antes D, Kang Y, Xie H, Javed TA, Lam PP, Rubin DC, Thorn P, Gaisano HY. Pancreatitis-Induced Depletion of Syntaxin 2 Promotes Autophagy and Increases Basolateral Exocytosis. Gastroenterology 2018; 154:1805-1821.e5. [PMID: 29360461 PMCID: PMC6461447 DOI: 10.1053/j.gastro.2018.01.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 12/13/2017] [Accepted: 01/08/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Pancreatic acinar cells are polarized epithelial cells that store enzymes required for digestion as inactive zymogens, tightly packed at the cell apex. Stimulation of acinar cells causes the zymogen granules to fuse with the apical membrane, and the cells undergo exocytosis to release proteases into the intestinal lumen. Autophagy maintains homeostasis of pancreatic acini. Syntaxin 2 (STX2), an abundant soluble N-ethyl maleimide sensitive factor attachment protein receptor in pancreatic acini, has been reported to mediate apical exocytosis. Using human pancreatic tissues and STX2-knockout (KO) mice, we investigated the functions of STX2 in zymogen granule-mediated exocytosis and autophagy. METHODS We obtained pancreatic tissues from 5 patients undergoing surgery for pancreatic cancer and prepared 80-μm slices; tissues were exposed to supramaximal cholecystokinin octapeptide (CCK-8) or ethanol and a low concentration of CCK-8 and analyzed by immunoblot and immunofluorescence analyses. STX2-KO mice and syntaxin 2+/+ C57BL6 mice (controls) were given intraperitoneal injections of supramaximal caerulein (a CCK-8 analogue) or fed ethanol and then given a low dose of caerulein to induce acute pancreatitis, or saline (controls); pancreata were isolated and analyzed by histology and immunohistochemistry. Acini were isolated from mice, incubated with CCK-8, and analyzed by immunofluorescence microscopy or used in immunoprecipitation experiments. Exocytosis was quantified using live-cell exocytosis and Ca2+ imaging analyses and based on formation of exocytotic soluble N-ethyl maleimide sensitive factor attachment protein receptor complexes. Dysregulations in autophagy were identified using markers, electron and immunofluorescence microscopy, and protease activation assays. RESULTS Human pancreatic tissues and dispersed pancreatic acini from control mice exposed to CCK-8 or ethanol plus CCK-8 were depleted of STX2. STX2-KO developed more severe pancreatitis after administration of supramaximal caerulein or a 6-week ethanol diet compared with control. Acini from STX2-KO mice had increased apical exocytosis after exposure to CCK-8, as well as increased basolateral exocytosis, which led to ectopic release of proteases. These increases in apical and basolateral exocytosis required increased formation of fusogenic soluble N-ethyl maleimide sensitive factor attachment protein receptor complexes, mediated by STX3 and STX4. STX2 bound ATG16L1 and prevented it from binding clathrin. Deletion of STX2 from acini increased binding of AT16L1 to clathrin, increasing formation of pre-autophagosomes and inducing autophagy. Induction of autophagy promoted the CCK-8-induced increase in autolysosome formation and the activation of trypsinogen. CONCLUSIONS In studies of human pancreatic tissues and pancreata from STX2-KO and control mice, we found STX2 to block STX3- and STX4-mediated fusion of zymogen granules with the plasma membrane and exocytosis and prevent binding of ATG16L1 to clathrin, which contributes to induction of autophagy. Exposure of pancreatic tissues to CCK-8 or ethanol depletes acinar cells of STX2, increasing basolateral exocytosis and promoting autophagy induction, leading to activation of trypsinogen.
Collapse
Affiliation(s)
- Subhankar Dolai
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Tao Liang
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Abrahim I Orabi
- Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Douglas Holmyard
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Li Xie
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Youhou Kang
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Huanli Xie
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tanveer A Javed
- Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Patrick P Lam
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Deborah C Rubin
- Division of Gastroenterology, Departments of Medicine, and Developmental Biology, Washington University School of Medicine, St Louis, Missouri
| | - Peter Thorn
- University of Sydney, Sydney, New South Wales, Australia
| | - Herbert Y Gaisano
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
42
|
Gryshchenko O, Gerasimenko JV, Peng S, Gerasimenko OV, Petersen OH. Calcium signalling in the acinar environment of the exocrine pancreas: physiology and pathophysiology. J Physiol 2018; 596:2663-2678. [PMID: 29424931 PMCID: PMC6046068 DOI: 10.1113/jp275395] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 01/25/2018] [Indexed: 12/12/2022] Open
Abstract
Key points Ca2+ signalling in different cell types in exocrine pancreatic lobules was monitored simultaneously and signalling responses to various stimuli were directly compared. Ca2+ signals evoked by K+‐induced depolarization were recorded from pancreatic nerve cells. Nerve cell stimulation evoked Ca2+ signals in acinar but not in stellate cells. Stellate cells are not electrically excitable as they, like acinar cells, did not generate Ca2+ signals in response to membrane depolarization. The responsiveness of the stellate cells to bradykinin was markedly reduced in experimental alcohol‐related acute pancreatitis, but they became sensitive to stimulation with trypsin. Our results provide fresh evidence for an important role of stellate cells in acute pancreatitis. They seem to be a critical element in a vicious circle promoting necrotic acinar cell death. Initial trypsin release from a few dying acinar cells generates Ca2+ signals in the stellate cells, which then in turn damage more acinar cells causing further trypsin liberation.
Abstract Physiological Ca2+ signals in pancreatic acinar cells control fluid and enzyme secretion, whereas excessive Ca2+ signals induced by pathological agents induce destructive processes leading to acute pancreatitis. Ca2+ signals in the peri‐acinar stellate cells may also play a role in the development of acute pancreatitis. In this study, we explored Ca2+ signalling in the different cell types in the acinar environment of the pancreatic tissue. We have, for the first time, recorded depolarization‐evoked Ca2+ signals in pancreatic nerves and shown that whereas acinar cells receive a functional cholinergic innervation, there is no evidence for functional innervation of the stellate cells. The stellate, like the acinar, cells are not electrically excitable as they do not generate Ca2+ signals in response to membrane depolarization. The principal agent evoking Ca2+ signals in the stellate cells is bradykinin, but in experimental alcohol‐related acute pancreatitis, these cells become much less responsive to bradykinin and then acquire sensitivity to trypsin. Our new findings have implications for our understanding of the development of acute pancreatitis and we propose a scheme in which Ca2+ signals in stellate cells provide an amplification loop promoting acinar cell death. Initial release of the proteases kallikrein and trypsin from dying acinar cells can, via bradykinin generation and protease‐activated receptors, induce Ca2+ signals in stellate cells which can then, possibly via nitric oxide generation, damage more acinar cells and thereby cause additional release of proteases, generating a vicious circle. Ca2+ signalling in different cell types in exocrine pancreatic lobules was monitored simultaneously and signalling responses to various stimuli were directly compared. Ca2+ signals evoked by K+‐induced depolarization were recorded from pancreatic nerve cells. Nerve cell stimulation evoked Ca2+ signals in acinar but not in stellate cells. Stellate cells are not electrically excitable as they, like acinar cells, did not generate Ca2+ signals in response to membrane depolarization. The responsiveness of the stellate cells to bradykinin was markedly reduced in experimental alcohol‐related acute pancreatitis, but they became sensitive to stimulation with trypsin. Our results provide fresh evidence for an important role of stellate cells in acute pancreatitis. They seem to be a critical element in a vicious circle promoting necrotic acinar cell death. Initial trypsin release from a few dying acinar cells generates Ca2+ signals in the stellate cells, which then in turn damage more acinar cells causing further trypsin liberation.
Collapse
Affiliation(s)
- Oleksiy Gryshchenko
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK.,Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine
| | | | - Shuang Peng
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK.,Department of Physiology, Medical College, Jinan University, Guangzhou 510632, China
| | | | - Ole H Petersen
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK.,Systems Immunity Research Institute, Cardiff University, Cardiff, CF14 4XN, UK
| |
Collapse
|
43
|
Jiang HN, Li Y, Jiang WY, Cui ZJ. Cholecystokinin 1 Receptor - A Unique G Protein- Coupled Receptor Activated by Singlet Oxygen ( GPCR-ABSO). Front Physiol 2018; 9:497. [PMID: 29867546 PMCID: PMC5953346 DOI: 10.3389/fphys.2018.00497] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/18/2018] [Indexed: 02/05/2023] Open
Abstract
Plasma membrane-delimited generation of singlet oxygen by photodynamic action with photosensitizer sulfonated aluminum phthalocyanine (SALPC) activates cholecystokinin 1 receptor (CCK1R) in pancreatic acini. Whether CCK1R retains such photooxidative singlet oxygen activation properties in other environments is not known. Genetically encoded protein photosensitizers KillerRed or mini singlet oxygen generator (miniSOG) were expressed in pancreatic acinar tumor cell line AR4-2J, CCK1R, KillerRed or miniSOG were expressed in HEK293 or CHO-K1 cells. Cold light irradiation (87 mW⋅cm-2) was applied to photosensitizer-expressing cells to examine photodynamic activation of CCK1R by Fura-2 fluorescent calcium imaging. When CCK1R was transduced into HEK293 cells which lack endogenous CCK1R, photodynamic action with SALPC was found to activate CCK1R in CCK1R-HEK293 cells. When KillerRed or miniSOG were transduced into AR4-2J which expresses endogenous CCK1R, KillerRed or miniSOG photodynamic action at the plasma membrane also activated CCK1R. When fused KillerRed-CCK1R was transduced into CHO-K1 cells, light irradiation activated the fused CCK1R leading to calcium oscillations. Therefore KillerRed either expressed independently, or fused with CCK1R can both activate CCK1R photodynamically. It is concluded that photodynamic singlet oxygen activation is an intrinsic property of CCK1R, independent of photosensitizer used, or CCK1R-expressing cell types. Photodynamic singlet oxygen CCK1R activation after transduction of genetically encoded photosensitizer in situ may provide a convenient way to verify intrinsic physiological functions of CCK1R in multiple CCK1R-expressing cells and tissues, or to actuate CCK1R function in CCK1R-expressing and non-expressing cell types after transduction with fused KillerRed-CCK1R.
Collapse
|
44
|
Dolai S, Liang T, Orabi AI, Xie L, Holmyard D, Javed TA, Fernandez NA, Xie H, Cattral MS, Thurmond DC, Thorn P, Gaisano HY. Depletion of the membrane-fusion regulator Munc18c attenuates caerulein hyperstimulation-induced pancreatitis. J Biol Chem 2017; 293:2510-2522. [PMID: 29284677 DOI: 10.1074/jbc.ra117.000792] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/21/2017] [Indexed: 12/26/2022] Open
Abstract
Epithelial pancreatic acinar cells perform crucial functions in food digestion, and acinar cell homeostasis required for secretion of digestive enzymes relies on SNARE-mediated exocytosis. The ubiquitously expressed Sec1/Munc18 protein mammalian uncoordinated-18c (Munc18c) regulates membrane fusion by activating syntaxin-4 (STX-4) to bind cognate SNARE proteins to form a SNARE complex that mediates exocytosis in many cell types. However, in the acinar cell, Munc18c's functions in exocytosis and homeostasis remain inconclusive. Here, we found that pancreatic acini from Munc18c-depleted mice (Munc18c+/-) and human pancreas (lenti-Munc18c-shRNA-treated) exhibit normal apical exocytosis of zymogen granules (ZGs) in response to physiologic stimulation with the intestinal hormone cholecystokinin (CCK-8). However, when stimulated with supraphysiologic CCK-8 levels to mimic pancreatitis, Munc18c-depleted (Munc18c+/-) mouse acini exhibited a reduction in pathological basolateral exocytosis of ZGs resulting from a decrease in fusogenic STX-4 SNARE complexes. This reduced basolateral exocytosis in part explained the less severe pancreatitis observed in Munc18c+/- mice after hyperstimulation with the CCK-8 analog caerulein. Likely as a result of this secretory blockade, Munc18c-depleted acini unexpectedly activated a component of the endoplasmic reticulum (ER) stress response that contributed to autophagy induction, resulting in downstream accumulation of autophagic vacuoles and autolysosomes. We conclude that Munc18c's role in mediating ectopic basolateral membrane fusion of ZGs contributes to the initiation of CCK-induced pancreatic injury, and that blockade of this secretory process could increase autophagy induction.
Collapse
Affiliation(s)
- Subhankar Dolai
- From the Departments of Medicine and .,Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tao Liang
- From the Departments of Medicine and.,Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Abrahim I Orabi
- Division of Pediatric Gastroenterology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Li Xie
- From the Departments of Medicine and.,Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Douglas Holmyard
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Tanveer A Javed
- Division of Pediatric Gastroenterology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | | | | | - Mark S Cattral
- Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario M5G 2N2, Canada
| | - Debbie C Thurmond
- Beckman Research Institute of the City of Hope, Duarte, California 91010, and
| | - Peter Thorn
- School of Biomedical Sciences,University of Sydney, Sydney, New South Wales 2050, Australia
| | - Herbert Y Gaisano
- From the Departments of Medicine and .,Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
45
|
Acinar injury and early cytokine response in human acute biliary pancreatitis. Sci Rep 2017; 7:15276. [PMID: 29127325 PMCID: PMC5681596 DOI: 10.1038/s41598-017-15479-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 10/27/2017] [Indexed: 12/16/2022] Open
Abstract
Clinical acute pancreatitis (AP) is marked by an early phase of systemic inflammatory response syndrome (SIRS) with multiorgan dysfunction (MODS), and a late phase characterized by sepsis with MODS. However, the mechanisms of acinar injury in human AP and the associated systemic inflammation are not clearly understood. This study, for the first time, evaluated the early interactions of bile acid induced human pancreatic acinar injury and the resulting cytokine response. We exposed freshly procured resected human pancreata to taurolithocolic acid (TLCS) and evaluated for acinar injury, cytokine release and interaction with peripheral blood mononuclear cells (PBMCs). We observed autophagy in acinar cells in response to TLCS exposure. There was also time-dependent release of IL-6, IL-8 and TNF-α from the injured acini that resulted in activation of PBMCs. We also observed that cytokines secreted by activated PBMCs resulted in acinar cell apoptosis and further cytokine release from them. Our data suggests that the earliest immune response in human AP originates within the acinar cell itself, which subsequently activates circulating PBMCs leading to SIRS. These findings need further detailed evaluation so that specific therapeutic targets to curb SIRS and resulting early adverse outcomes could be identified and tested.
Collapse
|
46
|
Lugea A, Waldron RT, Mareninova OA, Shalbueva N, Deng N, Su HY, Thomas DD, Jones EK, Messenger SW, Yang J, Hu C, Gukovsky I, Liu Z, Groblewski GE, Gukovskaya AS, Gorelick FS, Pandol SJ. Human Pancreatic Acinar Cells: Proteomic Characterization, Physiologic Responses, and Organellar Disorders in ex Vivo Pancreatitis. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2726-2743. [PMID: 28935577 DOI: 10.1016/j.ajpath.2017.08.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 06/30/2017] [Accepted: 08/03/2017] [Indexed: 12/17/2022]
Abstract
Knowledge of the molecular mechanisms of acute pancreatitis is largely based on studies using rodents. To assess similar mechanisms in humans, we performed ex vivo pancreatitis studies in human acini isolated from cadaveric pancreata from organ donors. Because data on these human acinar preparations are sparse, we assessed their functional integrity and cellular and organellar morphology using light, fluorescence, and electron microscopy; and their proteome by liquid chromatography-tandem mass spectrometry. Acinar cell responses to the muscarinic agonist carbachol (CCh) and the bile acid taurolithocholic acid 3-sulfate were also analyzed. Proteomic analysis of acini from donors of diverse ethnicity showed similar profiles of digestive enzymes and proteins involved in translation, secretion, and endolysosomal function. Human acini preferentially expressed the muscarinic acetylcholine receptor M3 and maintained physiological responses to CCh for at least 20 hours. As in rodent acini, human acini exposed to toxic concentrations of CCh and taurolithocholic acid 3-sulfate responded with trypsinogen activation, decreased cell viability, organelle damage manifest by mitochondrial depolarization, disordered autophagy, and pathological endoplasmic reticulum stress. Human acini also secreted inflammatory mediators elevated in acute pancreatitis patients, including IL-6, tumor necrosis factor-α, IL-1β, chemokine (C-C motif) ligands 2 and 3, macrophage inhibitory factor, and chemokines mediating neutrophil and monocyte infiltration. In conclusion, human cadaveric pancreatic acini maintain physiological functions and have similar pathological responses and organellar disorders with pancreatitis-causing treatments as observed in rodent acini.
Collapse
Affiliation(s)
- Aurelia Lugea
- Department of Medicine and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California; Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California; Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California.
| | - Richard T Waldron
- Department of Medicine and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California; Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California; Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California
| | - Olga A Mareninova
- Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California; Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California
| | - Natalia Shalbueva
- Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California; Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California
| | - Nan Deng
- Department of Biostatistics and Bioinformatics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Hsin-Yuan Su
- Department of Medicine and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Diane D Thomas
- Department of Nutritional Sciences, University of Wisconsin, Madison, Wisconsin
| | - Elaina K Jones
- Department of Nutritional Sciences, University of Wisconsin, Madison, Wisconsin
| | - Scott W Messenger
- Department of Nutritional Sciences, University of Wisconsin, Madison, Wisconsin
| | - Jiayue Yang
- Department of Medicine and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Cheng Hu
- Department of Medicine and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ilya Gukovsky
- Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California; Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California
| | - Zhenqiu Liu
- Department of Biostatistics and Bioinformatics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Guy E Groblewski
- Department of Nutritional Sciences, University of Wisconsin, Madison, Wisconsin
| | - Anna S Gukovskaya
- Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California; Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California
| | - Fred S Gorelick
- Departments of Internal Medicine and Cell Biology, Yale University School of Medicine, New Haven, Connecticut; Veterans Administration Connecticut Healthcare, West Haven, Connecticut
| | - Stephen J Pandol
- Department of Medicine and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California; Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California; Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California
| |
Collapse
|
47
|
Gerasimenko JV, Peng S, Tsugorka T, Gerasimenko OV. Ca 2+ signalling underlying pancreatitis. Cell Calcium 2017; 70:95-101. [PMID: 28552244 DOI: 10.1016/j.ceca.2017.05.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 12/13/2022]
Abstract
In spite of significant scientific progress in recent years, acute pancreatitis (AP) is still a dangerous and in up to 5% of cases deadly disease with no specific cure. It is self-resolved in the majority of cases, but could result in chronic pancreatitis (CP) and increased risk of pancreatic cancer (PC). One of the early events in AP is premature activation of digestive pro-enzymes, including trypsinogen, inside pancreatic acinar cells (PACs) due to an excessive rise in the cytosolic Ca2+ concentration, which is the result of Ca2+ release from internal stores followed by Ca2+ entry through the store operated Ca2+ channels in the plasma membrane. The leading causes of AP are high alcohol intake and biliary disease with gallstones obstruction leading to bile reflux into the pancreatic duct. Recently attention in this area of research turned to another cause of AP - Asparaginase based drugs - which have been used quite successfully in treatments of childhood acute lymphoblastic leukaemia (ALL). Unfortunately, Asparaginase is implicated in triggering AP in 5-10% of cases as a side effect of the anti-cancer therapy. The main features of Asparaginase-elicited AP (AAP) were found to be remarkably similar to AP induced by alcohol metabolites and bile acids. Several potential therapeutic avenues in counteracting AAP have been suggested and could also be useful for dealing with AP induced by other causes. Another interesting development in this field includes recent research related to pancreatic stellate cells (PSCs) that are much less studied in their natural environment but nevertheless critically involved in AP, CP and PC. This review will attempt to evaluate developments, approaches and potential therapies for AP and discuss links to other relevant diseases.
Collapse
Affiliation(s)
- J V Gerasimenko
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, Wales, UK.
| | - S Peng
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, Wales, UK; Department of Physiology, Medical College, Jinan University, Guangzhou 510632, China
| | - T Tsugorka
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, Wales, UK
| | - O V Gerasimenko
- Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, Wales, UK.
| |
Collapse
|