1
|
Ho KH, Gu G, Kaverina I. Preparation of Whole-mount Mouse Islets on Vascular Extracellular Matrix for Live Islet Cell Microscopy. Bio Protoc 2023; 13:e4868. [PMID: 37969764 PMCID: PMC10632159 DOI: 10.21769/bioprotoc.4868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 11/17/2023] Open
Abstract
Pancreatic islet β cells preferentially secrete insulin toward the plasma membrane, making contact with the capillary extracellular matrix (ECM). Isolated islets separated from the exocrine acinar cells are the best system for cell biology studies of primary β cells, whereas isolated islets lose their capillary network during ex vivo culture. Providing the appropriate extracellular signaling by attaching islets to vascular ECM-coated surfaces can restore the polarized insulin secretion toward the ECM. The guided secretion toward ECM-coated glass coverslips provides a good model for recording insulin secretion in real time to study its regulation. Additionally, β cells attached to the ECM-coated coverslips are suitable for confocal live imaging of subcellular components including adhesion molecules, cytoskeleton, and ion channels. This procedure is also compatible for total internal reflection fluorescence (TIRF) microscopy, which provides optimal signal-to-noise ratio and high spatial precision of structures close to the plasma membrane. In this article, we describe the optimized protocol for vascular ECM-coating of glass coverslips and the process of attachment of isolated mouse islets on the coverslip. This preparation is compatible with any high-resolution microscopy of live primary β cells. Key features • Optimized coating procedure to attach isolated islets, compatible for both confocal and TIRF microscopy. • The ECM-coated glass coverslip functions as the artificial capillary surface to guide secretion toward the coated surface for optimal imaging of secretion events. • Shows the process of islets attachment to the ECM-coated surface in a 6-day ex vivo culture.
Collapse
Affiliation(s)
- Kung-Hsien Ho
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Guoqiang Gu
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Program in Developmental Biology and Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
| | - Irina Kaverina
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
2
|
Boyer CK, Bauchle CJ, Zhang J, Wang Y, Stephens SB. Synchronized proinsulin trafficking reveals delayed Golgi export accompanies β-cell secretory dysfunction in rodent models of hyperglycemia. Sci Rep 2023; 13:5218. [PMID: 36997560 PMCID: PMC10063606 DOI: 10.1038/s41598-023-32322-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 03/25/2023] [Indexed: 04/03/2023] Open
Abstract
The pancreatic islet β-cell's preference for release of newly synthesized insulin requires careful coordination of insulin exocytosis with sufficient insulin granule production to ensure that insulin stores exceed peripheral demands for glucose homeostasis. Thus, the cellular mechanisms regulating insulin granule production are critical to maintaining β-cell function. In this report, we utilized the synchronous protein trafficking system, RUSH, in primary β-cells to evaluate proinsulin transit through the secretory pathway leading to insulin granule formation. We demonstrate that the trafficking, processing, and secretion of the proinsulin RUSH reporter, proCpepRUSH, are consistent with current models of insulin maturation and release. Using both a rodent dietary and genetic model of hyperglycemia and β-cell dysfunction, we show that proinsulin trafficking is impeded at the Golgi and coincides with the decreased appearance of nascent insulin granules at the plasma membrane. Ultrastructural analysis of β-cells from diabetic leptin receptor deficient mice revealed gross morphological changes in Golgi structure, including shortened and swollen cisternae, and partial Golgi vesiculation, which are consistent with defects in secretory protein export. Collectively, this work highlights the utility of the proCpepRUSH reporter in studying proinsulin trafficking dynamics and suggests that altered Golgi export function contributes to β-cell secretory defects in the pathogenesis of Type 2 diabetes.
Collapse
Affiliation(s)
- Cierra K Boyer
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52246, USA
| | - Casey J Bauchle
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52246, USA
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa, Iowa City, IA, 52246, USA
| | - Jianchao Zhang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Yanzhuang Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
- Department of Neurology, School of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Samuel B Stephens
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52246, USA.
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa, Iowa City, IA, 52246, USA.
| |
Collapse
|
3
|
Yang L, Fye MA, Yang B, Tang Z, Zhang Y, Haigh S, Covington BA, Bracey K, Taraska JW, Kaverina I, Qu S, Chen W. Genome-wide CRISPR screen identified a role for commander complex mediated ITGB1 recycling in basal insulin secretion. Mol Metab 2022; 63:101541. [PMID: 35835371 PMCID: PMC9304790 DOI: 10.1016/j.molmet.2022.101541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVES Pancreatic beta cells secrete insulin postprandially and during fasting to maintain glucose homeostasis. Although glucose-stimulated insulin secretion (GSIS) has been extensively studied, much less is known about basal insulin secretion. Here, we performed a genome-wide CRISPR/Cas9 knockout screen to identify novel regulators of insulin secretion. METHODS To identify genes that cell autonomously regulate insulin secretion, we engineered a Cas9-expressing MIN6 subclone that permits irreversible fluorescence labeling of exocytic insulin granules. Using a fluorescence-activated cell sorting assay of exocytosis in low glucose and high glucose conditions in individual cells, we performed a genome-wide CRISPR/Cas9 knockout screen. RESULTS We identified several members of the COMMD family, a conserved family of proteins with central roles in intracellular membrane trafficking, as positive regulators of basal insulin secretion, but not GSIS. Mechanistically, we show that the Commander complex promotes insulin granules docking in basal state. This is mediated, at least in part, by its function in ITGB1 recycling. Defective ITGB1 recycling reduces its membrane distribution, the number of focal adhesions and cortical ELKS-containing complexes. CONCLUSIONS We demonstrated a previously unknown function of the Commander complex in basal insulin secretion. We showed that by ITGB1 recycling, Commander complex increases cortical adhesions, which enhances the assembly of the ELKS-containing complexes. The resulting increase in the number of insulin granules near the plasma membrane strengthens basal insulin secretion.
Collapse
Affiliation(s)
- Liu Yang
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Margret A Fye
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Bingyuan Yang
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Zihan Tang
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Yue Zhang
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sander Haigh
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Brittney A Covington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kai Bracey
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Justin W Taraska
- Biochemistry and Biophysics Center, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Irina Kaverina
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
4
|
Georgiadou E, Rutter GA. Age matters: Grading granule secretion in beta cells. J Biol Chem 2020; 295:8912-8913. [PMID: 32620690 PMCID: PMC7335800 DOI: 10.1074/jbc.h120.014586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Indexed: 12/25/2022] Open
Abstract
Insulin is stored in secretory granules to facilitate rapid release in response to rising glucose levels, but the mechanisms by which these granules are identified and prioritized for secretion remains unclear. Using a fluorescent timer and flow cytometry-assisted organelle sorting, Yau et al. develop an elegant approach to assess insulin secretion as a function of granule age in pancreatic islet beta cells. Their findings supply quantitative evidence supporting the age-dependent release of different granule pools and confirm earlier models of preferential release of younger granules.
Collapse
Affiliation(s)
- Eleni Georgiadou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Department of Metabolism, Reproduction, and Digestion, Imperial College London, London, United Kingdom
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Department of Metabolism, Reproduction, and Digestion, Imperial College London, London, United Kingdom,Lee Kong Chian School of Medicine, Nan Yang Technological University, Singapore, For correspondence: Guy Rutter,
| |
Collapse
|
5
|
Bearrows SC, Bauchle CJ, Becker M, Haldeman JM, Swaminathan S, Stephens SB. Chromogranin B regulates early-stage insulin granule trafficking from the Golgi in pancreatic islet β-cells. J Cell Sci 2019; 132:jcs.231373. [PMID: 31182646 DOI: 10.1242/jcs.231373] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/31/2019] [Indexed: 12/12/2022] Open
Abstract
Chromogranin B (CgB, also known as CHGB) is abundantly expressed in dense core secretory granules of multiple endocrine tissues and has been suggested to regulate granule biogenesis in some cell types, including the pancreatic islet β-cell, though the mechanisms are poorly understood. Here, we demonstrate a critical role for CgB in regulating secretory granule trafficking in the β-cell. Loss of CgB impairs glucose-stimulated insulin secretion, impedes proinsulin processing to yield increased proinsulin content, and alters the density of insulin-containing granules. Using an in situ fluorescent pulse-chase strategy to track nascent proinsulin, we show that loss of CgB impairs Golgi budding of proinsulin-containing secretory granules, resulting in a substantial delay in trafficking of nascent granules to the plasma membrane with an overall decrease in total plasma membrane-associated granules. These studies demonstrate that CgB is necessary for efficient trafficking of secretory proteins into the budding granule, which impacts the availability of insulin-containing secretory granules for exocytic release.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Shelby C Bearrows
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52246, USA.,Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52246, USA
| | - Casey J Bauchle
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52246, USA.,Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52246, USA
| | - McKenzie Becker
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52246, USA.,Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52246, USA
| | - Jonathan M Haldeman
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Svetha Swaminathan
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52246, USA.,Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52246, USA
| | - Samuel B Stephens
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52246, USA .,Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52246, USA
| |
Collapse
|
6
|
Taneera J, Prasad RB, Dhaiban S, Mohammed AK, Haataja L, Arvan P, Hamad M, Groop L, Wollheim CB. Silencing of the FTO gene inhibits insulin secretion: An in vitro study using GRINCH cells. Mol Cell Endocrinol 2018; 472:10-17. [PMID: 29890211 PMCID: PMC6559235 DOI: 10.1016/j.mce.2018.06.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/14/2018] [Accepted: 06/07/2018] [Indexed: 01/01/2023]
Abstract
Expression of fat mass and obesity-associated gene (FTO) and ADP-ribosylation factor-like 15 (ARL15) in human islets is inversely correlated with HbA1c. However, their impact on insulin secretion is still ambiguous. Here in, we investigated the role of FTO and ARL15 using GRINCH (Glucose-Responsive Insulin-secreting C-peptide-modified Human proinsulin) clonal rat β-cells. GRINCH cells have inserted GFP into the human C-peptide insulin gene. Hence, secreted CpepGFP served to monitor insulin secretion. mRNA silencing of FTO in GRINCH cells showed a significant reduction in glucose but not depolarization-stimulated insulin secretion, whereas ARL15 silencing had no effect. A significant down-regulation of insulin mRNA was observed in FTO knockdown cells. Type-2 Diabetic islets revealed a reduced expression of FTO mRNA. In conclusion, our data suggest that fluorescent CpepGFP released from GRINCH cells may serve as a convenient marker for insulin secretion. Silencing of FTO expression, but not ARL15, inhibits insulin secretion by affecting metabolic signaling.
Collapse
Affiliation(s)
- Jalal Taneera
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates; Lund University Diabetes Center, Malmoe, Lund University, Sweden.
| | - Rashmi B Prasad
- Lund University Diabetes Center, Malmoe, Lund University, Sweden
| | - Sarah Dhaiban
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Abdul Khader Mohammed
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Leena Haataja
- Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, USA
| | - Peter Arvan
- Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, USA
| | - Mawieh Hamad
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Leif Groop
- Lund University Diabetes Center, Malmoe, Lund University, Sweden; Finnish Institute for Molecular Medicine (FIMM), Helsinki University, Finland
| | - Claes B Wollheim
- Lund University Diabetes Center, Malmoe, Lund University, Sweden; Department of Cell Physiology and Metabolism, University Medical Center. Geneva, Switzerland
| |
Collapse
|
7
|
Liberati D, Wyatt RC, Brigatti C, Marzinotto I, Ferrari M, Bazzigaluppi E, Bosi E, Gillard BT, Gillespie KM, Gorus F, Weets I, Balti E, Piemonti L, Achenbach P, Williams AJK, Lampasona V. A novel LIPS assay for insulin autoantibodies. Acta Diabetol 2018; 55:263-270. [PMID: 29305766 DOI: 10.1007/s00592-017-1082-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 11/20/2017] [Indexed: 12/26/2022]
Abstract
AIMS Insulin autoantibodies (IAA) are often the first marker of autoimmunity detected in children in the preclinical phase of type 1 diabetes (T1D). Currently, the vast majority of laboratories adopt the radiobinding micro-assay (RBA) for measuring IAA. Our aim was to replace RBA with a novel non-radioactive IAA Luciferase Immuno Precipitation System (LIPS) assay with improved performance. METHODS We developed (pro)insulin antigens with alternative placements of a NanoLuc™ luciferase reporter (NLuc). Performance in LIPS was evaluated by testing sera from new onset T1D (n = 80), blood donors (n = 123), schoolchildren (n = 186), first-degree relatives (FDRs) from the Bart's Oxford family study (n = 53) and from the Belgian Diabetes Registry (n = 136), coded sera from the Islet Autoantibody Standardization Program (IASP) (T1D n = 50, blood donors n = 90). RESULTS IAA LIPS based on B chain-NLuc proinsulin or B chain-NLuc insulin, in which NLuc was fused at the C-terminus of the insulin B chain, required only 2 μL of serum and a short incubation time, showed high concordance with RBA (Spearman r = 0.866 and 0.833, respectively), high assay performance (B chain-NLuc proinsulin ROC-AUC = 0.894 and B chain-NLuc insulin ROC-AUC = 0.916), and an adjusted sensitivity at 95% specificity ranking on par with the best assays submitted to the two most recent IASP workshops. In FDRs, the IAA LIPS showed improved discrimination of progressors to T1D compared to RBA. CONCLUSIONS We established a novel high-performance non-radioactive IAA LIPS that might replace the current gold standard RBA and find wide application in the study of the IAA response in T1D.
Collapse
Affiliation(s)
- Daniela Liberati
- Human Pathology Genomic Diagnostics Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rebecca C Wyatt
- Diabetes and Metabolism Unit, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Cristina Brigatti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Marzinotto
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maurizio Ferrari
- Human Pathology Genomic Diagnostics Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Elena Bazzigaluppi
- Department of Laboratory Medicine, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Emanuele Bosi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Ben T Gillard
- Diabetes and Metabolism Unit, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Kathleen M Gillespie
- Diabetes and Metabolism Unit, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Frans Gorus
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Clinical Chemistry, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ilse Weets
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Clinical Chemistry, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Eric Balti
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Clinical Chemistry, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, Neuherberg, Germany
| | - Alistair J K Williams
- Diabetes and Metabolism Unit, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Vito Lampasona
- Human Pathology Genomic Diagnostics Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
8
|
Kalwat MA, Wichaidit C, Nava Garcia AY, McCoy MK, McGlynn K, Hwang IH, MacMillan JB, Posner BA, Cobb MH. Insulin promoter-driven Gaussia luciferase-based insulin secretion biosensor assay for discovery of β-cell glucose-sensing pathways. ACS Sens 2016; 1:1208-1212. [PMID: 27819058 DOI: 10.1021/acssensors.6b00433] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
High throughput screening of insulin secretion is intractable with current methods. We developed a secreted insulin-luciferase system (Ins-GLuc) in β cells that is rapid, inexpensive, and amenable to 96- and 384-well formats. We treated stable Ins-GLuc-expressing MIN6 cells overnight with 6298 marine natural product fractions. The cells were then washed to remove media and chemicals, followed by stimulation with glucose in the diazoxide paradigm. These conditions allowed the discovery of many insulin secretion suppressors and potentiators. The mechanisms of action of these natural products must be long-lasting given the continuance of secretory phenotypes in the absence of chemical treatment. We anticipate that these natural products and their target pathways will lead to a greater understanding of glucose-stimulated insulin secretion.
Collapse
Affiliation(s)
- Michael A. Kalwat
- Department of Phamacology and ‡Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75235, United States
| | - Chonlarat Wichaidit
- Department of Phamacology and ‡Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75235, United States
| | - Alejandra Y. Nava Garcia
- Department of Phamacology and ‡Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75235, United States
| | - Melissa K. McCoy
- Department of Phamacology and ‡Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75235, United States
| | - Kathleen McGlynn
- Department of Phamacology and ‡Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75235, United States
| | - In Hyun Hwang
- Department of Phamacology and ‡Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75235, United States
| | - John B. MacMillan
- Department of Phamacology and ‡Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75235, United States
| | - Bruce A. Posner
- Department of Phamacology and ‡Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75235, United States
| | - Melanie H. Cobb
- Department of Phamacology and ‡Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75235, United States
| |
Collapse
|
9
|
Gorasia DG, Dudek NL, Safavi-Hemami H, Perez RA, Schittenhelm RB, Saunders PM, Wee S, Mangum JE, Hubbard MJ, Purcell AW. A prominent role of PDIA6 in processing of misfolded proinsulin. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:715-723. [PMID: 26947243 DOI: 10.1016/j.bbapap.2016.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 02/22/2016] [Accepted: 03/02/2016] [Indexed: 11/17/2022]
Abstract
Despite its critical role in maintaining glucose homeostasis, surprisingly little is known about proinsulin folding in the endoplasmic reticulum. In this study we aimed to understand the chaperones involved in the maturation and degradation of proinsulin. We generated pancreatic beta cell lines expressing FLAG-tagged proinsulin. Several chaperones (including BiP, PDIA6, calnexin, calreticulin, GRP170, Erdj3 and ribophorin II) co-immunoprecipitated with proinsulin suggesting a role for these proteins in folding. To investigate the chaperones responsible for targeting misfolded proinsulin for degradation, we also created a beta cell line expressing FLAG-tagged proinsulin carrying the Akita mutation (Cys96Tyr). All chaperones found to be associated with wild type proinsulin also co-immunoprecipitated with Akita proinsulin. However, one additional protein, namely P58(IPK), specifically precipitated with Akita proinsulin and approximately ten fold more PDIA6, but not other PDI family members, was bound to Akita proinsulin. The latter suggests that PDIA6 may act as a key reductase and target misfolded proinsulin to the ER-degradation pathway. The preferential association of PDIA6 to Akita proinsulin was also confirmed in another beta cell line (βTC-6). Furthermore, for the first time, a physiologically relevant substrate for PDIA6 has been evidenced. Thus, this study has identified several chaperones/foldases that associated with wild type proinsulin and has also provided a comprehensive interactome for Akita misfolded proinsulin.
Collapse
Affiliation(s)
- Dhana G Gorasia
- Department of Biochemistry and Molecular Biology, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
| | - Nadine L Dudek
- Department of Biochemistry and Molecular Biology, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia; Infection and Immunity Program, Biomolecular Discovery Institute and Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| | - Helena Safavi-Hemami
- Department of Biochemistry and Molecular Biology, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
| | - Rochelle Ayala Perez
- Infection and Immunity Program, Biomolecular Discovery Institute and Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| | - Ralf B Schittenhelm
- Infection and Immunity Program, Biomolecular Discovery Institute and Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| | - Philippa M Saunders
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia
| | - Sheena Wee
- Department of Biochemistry and Molecular Biology, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
| | - Jon E Mangum
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michael J Hubbard
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Anthony W Purcell
- Department of Biochemistry and Molecular Biology, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia; Infection and Immunity Program, Biomolecular Discovery Institute and Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
10
|
Fang J, Liu M, Zhang X, Sakamoto T, Taatjes DJ, Jena BP, Sun F, Woods J, Bryson T, Kowluru A, Zhang K, Chen X. COPII-Dependent ER Export: A Critical Component of Insulin Biogenesis and β-Cell ER Homeostasis. Mol Endocrinol 2015; 29:1156-69. [PMID: 26083833 DOI: 10.1210/me.2015-1012] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic β-cells possess a highly active protein synthetic and export machinery in the endoplasmic reticulum (ER) to accommodate the massive production of proinsulin. ER homeostasis is vital for β-cell functions and is maintained by the delicate balance between protein synthesis, folding, export, and degradation. Disruption of ER homeostasis by diabetes-causing factors leads to β-cell death. Among the 4 components to maintain ER homeostasis in β-cells, the role of ER export in insulin biogenesis is the least understood. To address this knowledge gap, the present study investigated the molecular mechanism of proinsulin ER export in MIN6 cells and primary islets. Two inhibitory mutants of the secretion-associated RAS-related protein (Sar)1 small GTPase, known to specifically block coat protein complex II (COPII)-dependent ER export, were overexpressed in β-cells using recombinant adenoviruses. Results from this approach, as well as small interfering RNA-mediated Sar1 knockdown, demonstrated that defective Sar1 function blocked proinsulin ER export and abolished its conversion to mature insulin in MIN6 cells, isolated mouse, and human islets. It is further revealed, using an in vitro vesicle formation assay, that proinsulin was packaged into COPII vesicles in a GTP- and Sar1-dependent manner. Blockage of COPII-dependent ER exit by Sar1 mutants strongly induced ER morphology change, ER stress response, and β-cell apoptosis. These responses were mediated by the PKR (double-stranded RNA-dependent kinase)-like ER kinase (PERK)/eukaryotic translation initiation factor 2α (p-eIF2α) and inositol-requiring protein 1 (IRE1)/x-box binding protein 1 (Xbp1) pathways but not via activating transcription factor 6 (ATF6). Collectively, results from the study demonstrate that COPII-dependent ER export plays a vital role in insulin biogenesis, ER homeostasis, and β-cell survival.
Collapse
Affiliation(s)
- Jingye Fang
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Ming Liu
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Xuebao Zhang
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Takeshi Sakamoto
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Douglas J Taatjes
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Bhanu P Jena
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Fei Sun
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - James Woods
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Tim Bryson
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Anjaneyulu Kowluru
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Kezhong Zhang
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Xuequn Chen
- Department of Physiology (J.F., B.P.J., F.S., J.W., T.B., X.C.) and Center for Molecular Medicine and Genetics (X.Z., K.Z.), School of Medicine, Department of Physics and Astronomy (T.S.), College of Liberal Arts and Sciences, and Department of Pharmaceutical Sciences (A.K.), Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, and John D. Dingell VA Medical Center (A.K.), Detroit, Michigan 48201; Department of Internal Medicine (M.L.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Pathology (D.J.T.), Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, Vermont 05405
| |
Collapse
|
11
|
Stasiuk GJ, Minuzzi F, Sae-Heng M, Rivas C, Juretschke HP, Piemonti L, Allegrini PR, Laurent D, Duckworth AR, Beeby A, Rutter GA, Long NJ. Dual-modal magnetic resonance/fluorescent zinc probes for pancreatic β-cell mass imaging. Chemistry 2015; 21:5023-33. [PMID: 25736590 PMCID: PMC4464533 DOI: 10.1002/chem.201406008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Indexed: 11/11/2022]
Abstract
Despite the contribution of changes in pancreatic β-cell mass to the development of all forms of diabetes mellitus, few robust approaches currently exist to monitor these changes prospectively in vivo. Although magnetic-resonance imaging (MRI) provides a potentially useful technique, targeting MRI-active probes to the β cell has proved challenging. Zinc ions are highly concentrated in the secretory granule, but they are relatively less abundant in the exocrine pancreas and in other tissues. We have therefore developed functional dual-modal probes based on transition-metal chelates capable of binding zinc. The first of these, Gd⋅1, binds Zn(II) directly by means of an amidoquinoline moiety (AQA), thus causing a large ratiometric Stokes shift in the fluorescence from λem =410 to 500 nm with an increase in relaxivity from r1 =4.2 up to 4.9 mM(-1) s(-1) . The probe is efficiently accumulated into secretory granules in β-cell-derived lines and isolated islets, but more poorly by non-endocrine cells, and leads to a reduction in T1 in human islets. In vivo murine studies of Gd⋅1 have shown accumulation of the probe in the pancreas with increased signal intensity over 140 minutes.
Collapse
Affiliation(s)
- Graeme J Stasiuk
- Department of Chemistry, Imperial College LondonSouth Kensington Campus, London SW7 2AZ (UK)
| | - Florencia Minuzzi
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College London, Hammersmith Hospital, London W12 0NN (UK)
| | - Myra Sae-Heng
- Department of Chemistry, Imperial College LondonSouth Kensington Campus, London SW7 2AZ (UK)
| | - Charlotte Rivas
- Department of Chemistry, Imperial College LondonSouth Kensington Campus, London SW7 2AZ (UK)
| | - Hans-Paul Juretschke
- Sanofi-Aventis Deutschland GmbH, R&D DSAR/BiomakersBiom & Biol Ass, FF, Industriepark Hoechst, Building H825, 65926 Frankfurt (Germany)
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific InstituteVia Olgettina 60, 20132 Milano (Italy)
| | | | - Didier Laurent
- Novartis Institute for Biomedical Research, Fabrikstrasse10-2.40.4, 4056, Basel (Switzerland)
| | - Andrew R Duckworth
- Department of Chemistry, Durham University, South RoadDurham, DH1 3LE (UK)
| | - Andrew Beeby
- Department of Chemistry, Durham University, South RoadDurham, DH1 3LE (UK)
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College London, Hammersmith Hospital, London W12 0NN (UK)
| | - Nicholas J Long
- Department of Chemistry, Imperial College LondonSouth Kensington Campus, London SW7 2AZ (UK)
| |
Collapse
|
12
|
Burns SM, Vetere A, Walpita D, Dančík V, Khodier C, Perez J, Clemons PA, Wagner BK, Altshuler D. High-throughput luminescent reporter of insulin secretion for discovering regulators of pancreatic Beta-cell function. Cell Metab 2015; 21:126-37. [PMID: 25565210 DOI: 10.1016/j.cmet.2014.12.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 10/14/2014] [Accepted: 12/13/2014] [Indexed: 12/16/2022]
Abstract
Defects in insulin secretion play a central role in the pathogenesis of type 2 diabetes, yet the mechanisms driving beta-cell dysfunction remain poorly understood, and therapies to preserve glucose-dependent insulin release are inadequate. We report a luminescent insulin secretion assay that enables large-scale investigations of beta-cell function, created by inserting Gaussia luciferase into the C-peptide portion of proinsulin. Beta-cell lines expressing this construct cosecrete luciferase and insulin in close correlation, under both standard conditions or when stressed by cytokines, fatty acids, or ER toxins. We adapted the reporter for high-throughput assays and performed a 1,600-compound pilot screen, which identified several classes of drugs inhibiting secretion, as well as glucose-potentiated secretagogues that were confirmed to have activity in primary human islets. Requiring 40-fold less time and expense than the traditional ELISA, this assay may accelerate the identification of pathways governing insulin secretion and compounds that safely augment beta-cell function in diabetes.
Collapse
Affiliation(s)
- Sean M Burns
- Diabetes Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Medical and Population Genetics Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Amedeo Vetere
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Deepika Walpita
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Vlado Dančík
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Carol Khodier
- Center for the Development of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Jose Perez
- Center for the Development of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Paul A Clemons
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Bridget K Wagner
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - David Altshuler
- Diabetes Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Medical and Population Genetics Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| |
Collapse
|
13
|
Park S, Alfa RW, Topper SM, Kim GES, Kockel L, Kim SK. A genetic strategy to measure circulating Drosophila insulin reveals genes regulating insulin production and secretion. PLoS Genet 2014; 10:e1004555. [PMID: 25101872 PMCID: PMC4125106 DOI: 10.1371/journal.pgen.1004555] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/20/2014] [Indexed: 11/19/2022] Open
Abstract
Insulin is a major regulator of metabolism in metazoans, including the fruit fly Drosophila melanogaster. Genome-wide association studies (GWAS) suggest a genetic basis for reductions of both insulin sensitivity and insulin secretion, phenotypes commonly observed in humans with type 2 diabetes mellitus (T2DM). To identify molecular functions of genes linked to T2DM risk, we developed a genetic tool to measure insulin-like peptide 2 (Ilp2) levels in Drosophila, a model organism with superb experimental genetics. Our system permitted sensitive quantification of circulating Ilp2, including measures of Ilp2 dynamics during fasting and re-feeding, and demonstration of adaptive Ilp2 secretion in response to insulin receptor haploinsufficiency. Tissue specific dissection of this reduced insulin signaling phenotype revealed a critical role for insulin signaling in specific peripheral tissues. Knockdown of the Drosophila orthologues of human T2DM risk genes, including GLIS3 and BCL11A, revealed roles of these Drosophila genes in Ilp2 production or secretion. Discovery of Drosophila mechanisms and regulators controlling in vivo insulin dynamics should accelerate functional dissection of diabetes genetics.
Collapse
Affiliation(s)
- Sangbin Park
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Ronald W. Alfa
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
- Neuroscience Program, Stanford University School of Medicine, Stanford, California, United States of America
| | - Sydni M. Topper
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Grace E. S. Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Lutz Kockel
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Seung K. Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine (Oncology Division) Stanford University School of Medicine, Stanford, California, United States of America
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
14
|
Ivanova A, Kalaidzidis Y, Dirkx R, Sarov M, Gerlach M, Schroth-Diez B, Müller A, Liu Y, Andree C, Mulligan B, Münster C, Kurth T, Bickle M, Speier S, Anastassiadis K, Solimena M. Age-dependent labeling and imaging of insulin secretory granules. Diabetes 2013; 62:3687-96. [PMID: 23929935 PMCID: PMC3806613 DOI: 10.2337/db12-1819] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Insulin is stored within the secretory granules of pancreatic β-cells, and impairment of its release is the hallmark of type 2 diabetes. Preferential exocytosis of newly synthesized insulin suggests that granule aging is a key factor influencing insulin secretion. Here, we illustrate a technology that enables the study of granule aging in insulinoma cells and β-cells of knock-in mice through the conditional and unequivocal labeling of insulin fused to the SNAP tag. This approach, which overcomes the limits encountered with previous strategies based on radiolabeling or fluorescence timer proteins, allowed us to formally demonstrate the preferential release of newly synthesized insulin and reveal that the motility of cortical granules significantly changes over time. Exploitation of this approach may enable the identification of molecular signatures associated with granule aging and unravel possible alterations of granule turnover in diabetic β-cells. Furthermore, the method is of general interest for the study of membrane traffic and aging.
Collapse
Affiliation(s)
- Anna Ivanova
- Molecular Diabetology, Paul Langerhans Institute Dresden, Dresden University of Technology, Dresden, Germany
- International Max Planck Research School, Dresden, Germany
| | - Yannis Kalaidzidis
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- A. N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | - Ronald Dirkx
- Molecular Diabetology, Paul Langerhans Institute Dresden, Dresden University of Technology, Dresden, Germany
| | - Mihail Sarov
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Michael Gerlach
- Islet Cell Regeneration, Paul Langerhans Institute Dresden, Dresden University of Technology, Dresden, Germany
- Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany
| | - Britta Schroth-Diez
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Andreas Müller
- Molecular Diabetology, Paul Langerhans Institute Dresden, Dresden University of Technology, Dresden, Germany
| | - Yanmei Liu
- Molecular Diabetology, Paul Langerhans Institute Dresden, Dresden University of Technology, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Cordula Andree
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Bernard Mulligan
- Molecular Diabetology, Paul Langerhans Institute Dresden, Dresden University of Technology, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Carla Münster
- Molecular Diabetology, Paul Langerhans Institute Dresden, Dresden University of Technology, Dresden, Germany
| | - Thomas Kurth
- Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany
| | - Marc Bickle
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Stephan Speier
- Islet Cell Regeneration, Paul Langerhans Institute Dresden, Dresden University of Technology, Dresden, Germany
- Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany
| | | | - Michele Solimena
- Molecular Diabetology, Paul Langerhans Institute Dresden, Dresden University of Technology, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Corresponding author: Michele Solimena,
| |
Collapse
|
15
|
Yang L, Ji W, Xue Y, Chen L. Imaging beta-cell mass and function in situ and in vivo. J Mol Med (Berl) 2013; 91:929-38. [PMID: 23700217 DOI: 10.1007/s00109-013-1056-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 05/07/2013] [Accepted: 05/15/2013] [Indexed: 01/16/2023]
Abstract
Glucose-stimulated insulin secretion (GSIS) from pancreatic beta-cells is critical to the maintenance of blood glucose homeostasis in animals. Both decrease in pancreatic beta-cell mass and defects in beta-cell function contribute to the onset of diabetes, although the underlying mechanisms remain largely unknown. Molecular imaging techniques can help beta-cell study in a number of ways. High-resolution fluorescence imaging techniques provide novel insights into the fundamental mechanisms underlying GSIS in isolated beta-cells or in situ in pancreatic islets, and dynamic changes of beta-cell mass and function can be noninvasively monitored in vivo by imaging techniques such as positron emission tomography and single-photon emission computed tomography. All these techniques will contribute to the better understanding of the progression of diabetes and the search for the optimized therapeutic measures that reverse deficits in beta-cell mass and function.
Collapse
Affiliation(s)
- Lu Yang
- The State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Molecular Medicine, Peking University and National Center for Nanoscience and Technology, Beijing, China.
| | | | | | | |
Collapse
|
16
|
Ying Y, Li L, Cao W, Yan D, Zeng Q, Kong X, Lu L, Yan M, Xu X, Qu J, Su Q, Ma X. The microtubule associated protein syntabulin is required for glucose-stimulated and cAMP-potentiated insulin secretion. FEBS Lett 2012; 586:3674-80. [PMID: 22975310 DOI: 10.1016/j.febslet.2012.08.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 08/23/2012] [Accepted: 08/25/2012] [Indexed: 12/23/2022]
Abstract
Syntabulin is a microtubule-associated protein that mediates anterograde transport of vesicles to neuronal processes. Here, we found that syntabulin was expressed in mouse pancreas and insulin-secreting β-cells, and that it partially co-localized with microtubule and insulin-containing granules. The association of syntabulin with these organelles increased upon glucose stimulation. Knock-down of syntabulin by shRNA reduced both basal and glucose-stimulated insulin secretion, and diminished cAMP-Epac2 and cAMP-PKA potentiated insulin secretion. Additionally, syntabulin was preferentially phosphorylated by the Epac2 agonist 8-pCPT-2'-O-Me-cAMP, suggesting that syntabulin could be a novel effector of Epac2 and play a critical role in cAMP-enhanced insulin secretion.
Collapse
Affiliation(s)
- Ying Ying
- Dept. of Physiology, School of Medicine, Shenzhen University, Shenzhen 518060, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
de Backer MWA, Garner KM, Luijendijk MCM, Adan RAH. Recombinant adeno-associated viral vectors. Methods Mol Biol 2012; 789:357-76. [PMID: 21922421 DOI: 10.1007/978-1-61779-310-3_24] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Recombinant adeno-associated viral (rAAV) vectors can be used to locally or systemically enhance or silence gene expression. They are relatively nonimmunogenic and can transduce dividing and nondividing cells, and different rAAV serotypes may transduce diverse cell types. Therefore, rAAV vectors are excellent tools to study the function of neuropeptides in local brain areas. In this chapter, we describe a protocol to produce high-titer, in vivo grade, rAAV vector stocks. The protocol includes an Iodixanol gradient, an anion exchange column and a desalting/concentration step and can be used for every serotype. In addition, a short protocol for rAAV injections into the brain and directions on how to detect and localize transduced cells are given.
Collapse
Affiliation(s)
- Marijke W A de Backer
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, Utrecht University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | | | | |
Collapse
|
18
|
High-throughput subcellular protein localization using transfected-cell arrays. Subcellular protein localization using cell arrays. Methods Mol Biol 2011; 706:53-72. [PMID: 21104054 DOI: 10.1007/978-1-61737-970-3_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Knowledge of protein localization within the cellular environment is critical for understanding the function of the protein and its regulatory networks. Protein localization data, however, have traditionally been accumulated from single or small-scale experiments. Transfected-cell arrays (TCAs) represent a robust alternative for the high-throughput analysis of gene/protein functions in mammalian cells. For protein localization studies, TCAs not only allow for the transfection and expression of over 1,000 genes in a single experiment but also make it feasible for simultaneous co-localization analyses of different subcellular compartments. In this chapter, we have described a protein co-localization protocol using transfected human cell arrays for a large set of cellular compartments, including the nucleus, ER, Golgi apparatus, mitochondrion, lysosome, peroxisome, and the microtubules, intermediate filaments and actin filaments. The application of these "organelle-co-localized cell arrays" facilitates the precise determination of the localizations of numerous recombinant proteins in a single experiment, making it currently the most efficient technique for high-throughput protein co-localization screening.
Collapse
|
19
|
de Backer MWA, Brans MAD, Luijendijk MCM, Garner KM, van den Heuvel DMA, Pasterkamp RJ, Adan RAH. Neuropeptide delivery to the brain: a von Willebrand factor signal peptide to direct neuropeptide secretion. BMC Neurosci 2010; 11:94. [PMID: 20701764 PMCID: PMC2928777 DOI: 10.1186/1471-2202-11-94] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Accepted: 08/11/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Multiple neuropeptides, sometimes with opposing functions, can be produced from one precursor gene. To study the roles of the different neuropeptides encoded by one large precursor we developed a method to overexpress minigenes and establish local secretion. RESULTS We fused the signal peptide from the Von Willebrand Factor (VWF) to a furin site followed by a processed form of the Agouti related protein (AgRP), AgRP(83-132) or alpha-melanocyte stimulating hormone. In vitro, these minigenes were secreted and biologically active. Additionally, the proteins of the minigenes were not transported into projections of primary neurons, thereby ensuring local release. In vivo administration of VWF-AgRP(83-132), using an adeno-associated viral vector as a delivery vehicle, into the paraventricular hypothalamus increased body weight and food intake of these rats compared to rats which received a control vector. CONCLUSIONS This study demonstrated that removal of the N-terminal part of full length AgRP and addition of a VWF signal peptide is a successful strategy to deliver neuropeptide minigenes to the brain and establish local neuropeptide secretion.
Collapse
Affiliation(s)
- Marijke W A de Backer
- Rudolf Magnus Institute of Neuroscience, Department of Neuroscience and Pharmacology, Utrecht University Medical Centre Utrecht, Utrecht, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
20
|
Lin G, Wang G, Liu G, Yang LJ, Chang LJ, Lue TF, Lin CS. Treatment of type 1 diabetes with adipose tissue-derived stem cells expressing pancreatic duodenal homeobox 1. Stem Cells Dev 2010; 18:1399-406. [PMID: 19245309 DOI: 10.1089/scd.2009.0010] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Due to the limited supply of donor pancreas, it is imperative that we identify alternative cell sources that can be used to treat diabetes mellitus (DM). Multipotent adipose tissue-derived stem cells (ADSC) can be abundantly and safely isolated for autologous transplantation and therefore are an ideal candidate. Here, we report the derivation of insulin-producing cells from human or rat ADSC by transduction with the pancreatic duodenal homeobox 1 (Pdx1) gene. RT-PCR analyses showed that native ADSC expressed insulin, glucagon, and NeuroD genes that were up-regulated following Pdx1 transduction. ELISA analyses showed that the transduced cells secreted increasing amount of insulin in response to increasing concentration of glucose. Transplantation of these cells under the renal capsule of streptozotocin-induced diabetic rats resulted in lowered blood glucose, higher glucose tolerance, smoother fur, and less cataract. Histological examination showed that the transplanted cells formed tissue-like structures and expressed insulin. Thus, ADSC-expressing Pdx1 appear to be suitable for treatment of DM.
Collapse
Affiliation(s)
- Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California 94143-0738, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Griffiths EJ, Rutter GA. Mitochondrial calcium as a key regulator of mitochondrial ATP production in mammalian cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2009; 1787:1324-33. [PMID: 19366607 DOI: 10.1016/j.bbabio.2009.01.019] [Citation(s) in RCA: 272] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Revised: 01/23/2009] [Accepted: 01/27/2009] [Indexed: 12/30/2022]
Abstract
Mitochondrial Ca(2+) transport was initially considered important only in buffering of cytosolic Ca(2+) by acting as a "sink" under conditions of Ca(2+) overload. The main regulator of ATP production was considered to be the relative concentrations of high energy phosphates. However, work by Denton and McCormack in the 1970s and 1980s showed that free intramitochondrial Ca(2+) ([Ca(2+)](m)) activated dehydrogenase enzymes in mitochondria, leading to increased NADH and hence ATP production. This leads them to propose a scheme, subsequently termed a "parallel activation model" whereby increases in energy demand, such as hormonal stimulation or increased workload in muscle, produced an increase in cytosolic [Ca(2+)] that was relayed by the mitochondrial Ca(2+) transporters into the matrix to give an increase in [Ca(2+)](m). This then stimulated energy production to meet the increased energy demand. With the development of methods for measuring [Ca(2+)](m) in living cells that proved [Ca(2+)](m) changed over a dynamic physiological range rather than simply soaking up excess cytosolic [Ca(2+)], this model has now gained widespread acceptance. However, work by ourselves and others using targeted probes to measure changes in both [Ca(2+)] and [ATP] in different cell compartments has revealed variations in the interrelationships between these two in different tissues, suggesting that metabolic regulation by Ca(2+) is finely tuned to the demands and function of the individual organ.
Collapse
Affiliation(s)
- Elinor J Griffiths
- Department of Biochemistry, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, UK.
| | | |
Collapse
|
22
|
Nunes P, Hasler U, McKee M, Lu HAJ, Bouley R, Brown D. A fluorimetry-based ssYFP secretion assay to monitor vasopressin-induced exocytosis in LLC-PK1 cells expressing aquaporin-2. Am J Physiol Cell Physiol 2008; 295:C1476-87. [PMID: 18799651 DOI: 10.1152/ajpcell.00344.2008] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vasopressin (VP)-induced exocytosis was dissected in native and aquaporin-2 (AQP2)-expressing renal LLC-PK(1) cells by a fluorimetric exocytosis assay based on soluble secreted yellow fluorescent protein (ssYFP). YFP was targeted to the secretory pathway by addition of an 18-amino acid signal peptide from hen egg white lysozyme. Immunofluorescence labeling, together with analysis of Alexa 555-dextran internalization, revealed that ssYFP is exclusively located in the secretory pathway. Immunofluorescence and immunogold electron microscopy showed significant colocalization of ssYFP and AQP2. Fluorimetry and Western blot analysis demonstrated similar constitutive ssYFP secretion in native LLC-PK(1) and AQP2-expressing cells. In AQP2-expressing cells, a twofold increase in ssYFP secretion was observed within 15 min of VP stimulation. This transient burst of ssYFP secretion was abolished by the PKA inhibitor H-89 and was not observed in native cells. The endocytotic inhibitor methyl-beta-cyclodextrin, which also promotes membrane accumulation of AQP2, had no effect on ssYFP secretion. Although cells expressing phosphorylation-deficient AQP2-S256A showed significantly lower baseline levels of constitutive secretion, VP induced a significant increase in exocytosis. Our data indicate that 1) this assay can monitor exocytosis in cultured epithelial cells, 2) VP has an acute stimulatory effect on ssYFP secretion in AQP2-expressing, but not native, cells, and 3) phosphorylation of AQP2 at S256 may be involved in the regulation of constitutive AQP2 exocytosis and play only a minor role in the VP-induced burst. These results support the idea that, in addition to its role in reducing AQP2 endocytosis, VP increases AQP2 exocytosis.
Collapse
Affiliation(s)
- Paula Nunes
- Massachusetts General Hospital, Boston, MA 02114, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Liu M, Hodish I, Rhodes CJ, Arvan P. Proinsulin maturation, misfolding, and proteotoxicity. Proc Natl Acad Sci U S A 2007; 104:15841-6. [PMID: 17898179 PMCID: PMC2000385 DOI: 10.1073/pnas.0702697104] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
As a tool to explore proinsulin (PI) trafficking, a human PI cDNA has been constructed with GFP fused within the C peptide. In regulated secretory cells containing appropriate prohormone convertases, the hProCpepGFP construct undergoes endoproteolytic processing to CpepGFP and native human insulin, which are specifically detected and cosecreted in parallel with endogenous insulin. Expression of C(A7)Y mutant PI results in autosomal dominant diabetes in Akita mice. We directly identify the misfolded PI in Akita islets and also show that C(A7)Y mutant PI, either in the context of the hProCpepGFP chimera or not, engages directly in protein complexes with nonmutant PI, impairing the trafficking and recovery of nonmutant PI. This trapping mechanism decreases insulin production in beta cells. Thereafter we observe a loss of beta cell viability. The data imply that PI misfolding leading to impaired endoplasmic reticulum exit of nonmutant PI may be a key early step in a chain reaction of beta cell dysfunction and demise leading to onset and progression of diabetes.
Collapse
Affiliation(s)
- Ming Liu
- *Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical Center, Ann Arbor, MI 48109; and
| | - Israel Hodish
- *Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical Center, Ann Arbor, MI 48109; and
| | - Christopher J. Rhodes
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago, Chicago, IL 60637
| | - Peter Arvan
- *Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical Center, Ann Arbor, MI 48109; and
- To whom correspondence should be addressed at:
Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, 5560 Medical Science Research Building II, 1150 West Medical Center Drive, Ann Arbor, MI 48109-0678. E-mail:
| |
Collapse
|
24
|
Baroukh N, Ravier MA, Loder MK, Hill EV, Bounacer A, Scharfmann R, Rutter GA, Van Obberghen E. MicroRNA-124a regulates Foxa2 expression and intracellular signaling in pancreatic beta-cell lines. J Biol Chem 2007; 282:19575-88. [PMID: 17462994 DOI: 10.1074/jbc.m611841200] [Citation(s) in RCA: 251] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that have been implicated in fine-tuning gene regulation, although the precise roles of many are still unknown. Pancreatic development is characterized by the complex sequential expression of a gamut of transcription factors. We have performed miRNA expression profiling at two key stages of mouse embryonic pancreas development, e14.5 and e18.5. miR-124a2 expression was strikingly increased at e18.5 compared with e14.5, suggesting a possible role in differentiated beta-cells. Among the potential miR-124a gene targets identified by biocomputation, Foxa2 is known to play a role in beta-cell differentiation. To evaluate the impact of miR-124a2 on gene expression, we overexpressed or down-regulated miR-124a2 in MIN6 beta-cells. As predicted, miR-124a2 regulated Foxa2 gene expression, and that of its downstream target, pancreatic duodenum homeobox-1 (Pdx-1). Foxa2 has been described as a master regulator of pancreatic development and also of genes involved in glucose metabolism and insulin secretion, including the ATP-sensitive K(+) (K(ATP)) channel subunits, Kir6.2 and Sur-1. Correspondingly, miR-124a2 overexpression decreased, and anti-miR-124a2 increased Kir6.2 and Sur-1 mRNA levels. Moreover, miR-124a2 modified basal and glucose- or KCl-stimulated intracellular free Ca(2+) concentrations in single MIN6 and INS-1 (832/13) beta-cells, without affecting the secretion of insulin or co-transfected human growth hormone, consistent with an altered sensitivity of the beta-cell exocytotic machinery to Ca(2+). In conclusion, whereas the precise role of microRNA-124a2 in pancreatic development remains to be deciphered, we identify it as a regulator of a key transcriptional protein network in beta-cells responsible for modulating intracellular signaling.
Collapse
|
25
|
MacDonald PE, Rorsman P. The Ins and Outs of Secretion from Pancreatic β-Cells: Control of Single-Vesicle Exo- and Endocytosis. Physiology (Bethesda) 2007; 22:113-21. [PMID: 17420302 DOI: 10.1152/physiol.00047.2006] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exocytosis of insulin-containing secretory vesicles in pancreatic β-cells is crucial to maintenance of plasma glucose levels. They fuse with the plasma membrane in a regulated manner to release their contents and are subsequently recaptured either intact or through conventional clathrin-mediated endocytosis. Here, we discuss these mechanisms in β-cells at the single-vesicle level.
Collapse
Affiliation(s)
- Patrick E MacDonald
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada.
| | | |
Collapse
|
26
|
Michael DJ, Cai H, Xiong W, Ouyang J, Chow RH. Mechanisms of peptide hormone secretion. Trends Endocrinol Metab 2006; 17:408-15. [PMID: 17084640 DOI: 10.1016/j.tem.2006.10.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2006] [Revised: 10/06/2006] [Accepted: 10/24/2006] [Indexed: 11/21/2022]
Abstract
According to the classical view, peptide hormones are stored in large dense-core vesicles that release all of their cargo rapidly and completely when they fuse with and flatten into the plasma membrane. However, recent imaging studies suggest that this view is too simple. Even after vesicles fuse with the plasma membrane, cells might control the rate of dispersal of vesicle cargo - either by modulating the properties of the fusion pore that connects the vesicle lumen to the extracellular solution or by storing cargo in states that disperse slowly in the extracellular space. Understanding these mechanisms is important, owing to the increasing prevalence of diseases, such as type 2 diabetes mellitus, which arise from insufficient secretion of peptide hormones.
Collapse
Affiliation(s)
- Darren J Michael
- Department of Physiology and Biophysics, Keck School of Medicine Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90089, USA
| | | | | | | | | |
Collapse
|
27
|
Sobota JA, Ferraro F, Bäck N, Eipper BA, Mains RE. Not all secretory granules are created equal: Partitioning of soluble content proteins. Mol Biol Cell 2006; 17:5038-52. [PMID: 17005911 PMCID: PMC1761688 DOI: 10.1091/mbc.e06-07-0626] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Secretory granules carrying fluorescent cargo proteins are widely used to study granule biogenesis, maturation, and regulated exocytosis. We fused the soluble secretory protein peptidylglycine alpha-hydroxylating monooxygenase (PHM) to green fluorescent protein (GFP) to study granule formation. When expressed in AtT-20 or GH3 cells, the PHM-GFP fusion protein partitioned from endogenous hormone (adrenocorticotropic hormone, growth hormone) into separate secretory granule pools. Both exogenous and endogenous granule proteins were stored and released in response to secretagogue. Importantly, we found that segregation of content proteins is not an artifact of overexpression nor peculiar to GFP-tagged proteins. Neither luminal acidification nor cholesterol-rich membrane microdomains play essential roles in soluble content protein segregation. Our data suggest that intrinsic biophysical properties of cargo proteins govern their differential sorting, with segregation occurring during the process of granule maturation. Proteins that can self-aggregate are likely to partition into separate granules, which can accommodate only a few thousand copies of any content protein; proteins that lack tertiary structure are more likely to distribute homogeneously into secretory granules. Therefore, a simple "self-aggregation default" theory may explain the little acknowledged, but commonly observed, tendency for both naturally occurring and exogenous content proteins to segregate from each other into distinct secretory granules.
Collapse
Affiliation(s)
- Jacqueline A. Sobota
- *Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030-3401; and
| | - Francesco Ferraro
- *Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030-3401; and
| | - Nils Bäck
- Department of Anatomy, Institute of Biomedicine, University of Helsinki, FIN-00014, Helsinki, Finland
| | - Betty A. Eipper
- *Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030-3401; and
| | - Richard E. Mains
- *Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030-3401; and
| |
Collapse
|
28
|
Varadi A, Grant A, McCormack M, Nicolson T, Magistri M, Mitchell KJ, Halestrap AP, Yuan H, Schwappach B, Rutter GA. Intracellular ATP-sensitive K+ channels in mouse pancreatic beta cells: against a role in organelle cation homeostasis. Diabetologia 2006; 49:1567-77. [PMID: 16752175 DOI: 10.1007/s00125-006-0257-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2006] [Accepted: 02/28/2006] [Indexed: 11/30/2022]
Abstract
AIMS/HYPOTHESIS ATP-sensitive K(+) (K(ATP)) channels located on the beta cell plasma membrane play a critical role in regulating insulin secretion and are targets for the sulfonylurea class of antihyperglycaemic drugs. Recent reports suggest that these channels may also reside on insulin-containing dense-core vesicles and mitochondria. The aim of this study was to explore these possibilities and to test the hypothesis that vesicle-resident channels play a role in the control of organellar Ca(2+) concentration or pH. METHODS To quantify the subcellular distribution of the pore-forming subunit Kir6.2 and the sulfonylurea binding subunit SUR1 in isolated mouse islets and clonal pancreatic MIN6 beta cells, we used four complementary techniques: immunoelectron microscopy, density gradient fractionation, vesicle immunopurification and fluorescence-activated vesicle isolation. Intravesicular and mitochondrial concentrations of free Ca(2+) were measured in intact or digitonin-permeabilised MIN6 cells using recombinant, targeted aequorins, and intravesicular pH was measured with the recombinant fluorescent probe pHluorin. RESULTS SUR1 and Kir6.2 immunoreactivity were concentrated on dense-core vesicles and on vesicles plus the endoplasmic reticulum/Golgi network, respectively, in both islets and MIN6 cells. Reactivity to neither subunit was detected on mitochondria. Glibenclamide, tolbutamide and diazoxide all failed to affect Ca(2+) uptake into mitochondria, and K(ATP) channel regulators had no significant effect on intravesicular free Ca(2+) concentrations or vesicular pH. CONCLUSIONS/INTERPRETATION A significant proportion of Kir6.2 and SUR1 subunits reside on insulin-secretory vesicles and the distal secretory pathway in mouse beta cells but do not influence intravesicular ion homeostasis. We propose that dense-core vesicles may serve instead as sorting stations for the delivery of channels to the plasma membrane.
Collapse
Affiliation(s)
- A Varadi
- Henry Wellcome Laboratories for Integrated Cell Signalling and Department of Biochemistry, School of Medical Sciences, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Tsuboi T, Ravier MA, Parton LE, Rutter GA. Sustained exposure to high glucose concentrations modifies glucose signaling and the mechanics of secretory vesicle fusion in primary rat pancreatic beta-cells. Diabetes 2006; 55:1057-65. [PMID: 16567529 DOI: 10.2337/diabetes.55.04.06.db05-1577] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The mechanism(s) by which chronic hyperglycemia impairs glucose-stimulated insulin secretion is poorly defined. Here, we compare the "nanomechanics" of single exocytotic events in primary rat pancreatic beta-cells cultured for 48 h at optimal (10 mmol/l) or elevated (30 mmol/l) glucose concentrations. Cargo release was imaged by total internal reflection fluorescence microscopy of lumen-targeted probes (neuropeptide Y [NPY]-pH-insensitive yellow fluorescent protein [NPY-Venus] or NPY-monomeric red fluorescent protein), while the fate of the vesicle membrane was reported simultaneously with phosphatase-on-the-granule-of-insulinoma-enhanced green fluorescent protein. Under all conditions studied, exocytosis proceeded via a "cavity recapture" mechanism in which the vesicle and plasma membranes fused transiently. While essentially complete release of NPY-Venus was observed in 24 +/- 1% of glucose-stimulated exocytotic events in cells maintained at 10 mmol/l glucose, this value was reduced reversibly to 5 +/- 2% of events by culture at 30 mmol/l glucose, in line with decreases in Glut2 and glucokinase gene expression, and attenuated glucose-stimulated increases in NADPH and intracellular [Ca2+]. Since vesicle release in response to cell depolarization with KCl was not affected by culture at 30 mmol/l glucose, we conclude that hyperglycemia causes the abnormal termination of individual insulin release events principally by inhibiting glucose signaling.
Collapse
Affiliation(s)
- Takashi Tsuboi
- Department of Biochemistry, School of Medical Sciences, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | | | | | | |
Collapse
|
30
|
Insulin secretion in health and disease: genomics, proteomics and single vesicle dynamics. Biochem Soc Trans 2006. [DOI: 10.1042/bst0340247] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Defective insulin secretion from pancreatic islet β-cells is a sine qua non of Type II (non-insulin-dependent) diabetes. Digital imaging analysis of the nanomechanics of individual exocytotic events, achieved using total internal reflection fluorescence microscopy, has allowed us to demonstrate that insulin is released via transient or ‘cavicapture’ events whereby the vesicle and plasma membranes fuse transiently and reversibly. Such studies reveal that an increase in the number of abortive fusion events contributes to defective insulin secretion in in vitro models of Type II diabetes. Complementary analyses of genome-wide changes in β-cell gene expression, at both the mRNA and protein levels, are now facilitating the identification of key molecular players whose altered expression may contribute to the secretory defects in the diabetic β-cell.
Collapse
|
31
|
Abstract
Accomplishment of the human and mouse genome projects resulted in accumulation of extensive gene sequence information. However, the information about the biological functions of the identified genes remains a bottleneck of the post-genomic era. Hence, assays providing simple functional information, such as localization of the protein within the cell, can be very helpful in the elucidation of its function. Transfected cell arrays offer a robust platform for protein localization studies. Open reading frames of unknown genes can be linked to a His6-tag or GFP (green fluorescent protein) reporter in expression vectors and subsequently transfected using the cell array. Cellular localization of the transfected proteins is detected either by specific anti-His-tag antibodies or directly by fluorescence of the GFP fusion protein and by counterstaining with organelle-specific dyes. The high throughput of the method in terms of information provided for every single experiment makes this approach superior to classical immunohistological methods for protein localization.
Collapse
|
32
|
Kjaerulff S, Jensen MR. Comparison of different signal peptides for secretion of heterologous proteins in fission yeast. Biochem Biophys Res Commun 2005; 336:974-82. [PMID: 16157307 DOI: 10.1016/j.bbrc.2005.08.195] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2005] [Accepted: 08/25/2005] [Indexed: 11/21/2022]
Abstract
In the fission yeast Schizosaccharomyces pombe, there are relatively few signal peptides available and most reports of their activity have not been comparative. Using sequence information from the S. pombe genome database we have identified three putative signal peptides, designated Cpy, Amy and Dpp, and compared their ability to support secretion of green fluorescent protein (GFP). In the comparison we also included the two well-described secretion signals derived from the precursors of, respectively, the Saccharomyces cerevisiae alpha-factor and the S. pombe P-factor. The capability of the tested signal peptides to direct secretion of GFP varied greatly. The alpha-factor signal did not confer secretion to GFP and all the produced GFP was trapped intracellular. In contrast, the Cpy signal peptide supported efficient secretion of GFP with yields approximating 10 mg/L. We also found that the use of an attenuated version of the S. cerevisiae URA3 marker substantially increases vector copy number and expression yield in fission yeast.
Collapse
|
33
|
Garcia AL, Han SK, Janssen WG, Khaing ZZ, Ito T, Glucksman MJ, Benson DL, Salton SRJ. A prohormone convertase cleavage site within a predicted alpha-helix mediates sorting of the neuronal and endocrine polypeptide VGF into the regulated secretory pathway. J Biol Chem 2005; 280:41595-608. [PMID: 16221685 DOI: 10.1074/jbc.m509122200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Distinct intracellular pathways are involved in regulated and constitutive protein secretion from neuronal and endocrine cells, yet the peptide signals and molecular mechanisms responsible for targeting and retention of soluble proteins in secretory granules are incompletely understood. By using confocal microscopy and subcellular fractionation, we examined trafficking of the neuronal and endocrine peptide precursor VGF that is stored in large dense core vesicles and undergoes regulated secretion. VGF cofractionated with secretory vesicle membranes but was not detected in detergent-resistant lipid rafts. Deletional analysis using epitope-tagged VGF suggested that the C-terminal 73-amino acid fragment of VGF, containing two predicted alpha-helical loops and four potential prohormone convertase (PC) cleavage sites, was necessary and sufficient with an N-terminal signal peptide-containing domain, for large dense core vesicle sorting and regulated secretion from PC12 and INS-1 cells. Further transfection analysis identified the sorting sequence as a compact C-terminal alpha-helix and embedded 564RRR566 PC cleavage site; mutation of the 564RRR566 PC site in VGF-(1-65): GFP:VGF-(545-617) blocked regulated secretion, whereas disruption of the alpha-helix had no effect. Mutation of the adjacent 567HFHH570 motif, a charged region that might enhance PC cleavage in acidic environments, also blocked regulated release. Finally, inhibition of PC cleavage in PC12 cells using the membrane-permeable synthetic peptide chloromethyl ketone (decanoyl-RVKR-CMK) blocked regulated secretion of VGF. Our studies define a critical RRR-containing C-terminal domain that targets VGF into the regulated pathway in neuronal PC12 and endocrine INS-1 cells, providing additional support for the proposed role that PCs and their cleavage sites play in regulated peptide secretion.
Collapse
Affiliation(s)
- Angelo L Garcia
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Huang D, Shusta EV. Secretion and surface display of green fluorescent protein using the yeast Saccharomyces cerevisiae. Biotechnol Prog 2005; 21:349-57. [PMID: 15801770 DOI: 10.1021/bp0497482] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Green fluorescent protein (GFP) continues to be a very useful tool in biotechnology, but soluble production of GFP and GFP-protein fusions has been difficult. In this study, we have produced yeast-enhanced green fluorescent protein (yEGFP) in Saccharomyces cerevisiae as a soluble, secreted product with a purified level of 6 mg/L. Expression was directed by the inducible GAL1-10 promoter and synthetic prepro leader sequence. The secretion of yEGFP by yeast was strongly dependent on temperature, with 20 degrees C induction being optimal. Use of 2 micro multicopy expression constructs elevated yields over a low-copy CEN-based system by approximately 2-fold. Yeast-enhanced GFP was also expressed as a fusion to the Aga2p mating agglutinin in order to test the secretory processing fidelity of yEGFP-protein fusions. When the cell surface anchoring protein, Aga1p, was co-overexpressed with the Aga2p-yEGFP fusion, the Aga2p-yEGFP protein was tethered to the yeast cell surface. Flow cytometry and fluorescence microscopy analysis indicated that the fusion was displayed on the yeast cell surface at high levels. In the absence of high level Aga1p expression, the Aga2p-yEGFP fusion protein was instead secreted in its entirety with no detectable surface display. These findings reveal that yeast is a suitable host for secretion of GFP and GFP-protein fusions and thus could enable a wide range of biochemistry and biotechnology applications.
Collapse
Affiliation(s)
- Dagang Huang
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Wisconsin 53706, USA
| | | |
Collapse
|
35
|
Ravier MA, Rutter GA. Glucose or insulin, but not zinc ions, inhibit glucagon secretion from mouse pancreatic alpha-cells. Diabetes 2005; 54:1789-97. [PMID: 15919801 DOI: 10.2337/diabetes.54.6.1789] [Citation(s) in RCA: 219] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The mechanisms by which hypoglycemia stimulates glucagon release are still poorly understood. In particular, the relative importance of direct metabolic coupling versus paracrine regulation by beta-cell secretory products is unresolved. Here, we compare the responses to glucose of 1) alpha-cells within the intact mouse islet, 2) dissociated alpha-cells, and 3) clonal alphaTC1-9 cells. Free cytosolic concentrations of ATP ([ATP](c)) or Ca(2+) ([Ca(2+)](c)) were imaged using alpha-cell-targeted firefly luciferase or a green fluorescent protein-based Ca(2+) probe ("pericam"), respectively. Consistent with a direct effect of glucose on alpha-cell oxidative metabolism, an increase in glucose concentration (from 0 or 3 mmol/l to 20 mmol/l) increased [ATP](c) by 7-9% in alpha-cells within the intact islet and by approximately 4% in alphaTC1-9 cells. Moreover, glucose also dose-dependently decreased the frequency of [Ca(2+)](c) oscillations in both dissociated alpha-cells and alphaTC1-9 cells. Although the effects of glucose were mimicked by exogenous insulin, they were preserved when insulin signaling was blocked with wortmannin. Addition of ZnCl(2) slightly increased the frequency of [Ca(2+)](c) oscillations but failed to affect glucagon release from either islets or alphaTC1-9 cells under most conditions. We conclude that glucose and insulin, but not Zn(2+) ions, independently suppress glucagon secretion in the mouse.
Collapse
Affiliation(s)
- Magalie A Ravier
- Department of Biochemistry, School of Medical Sciences, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | | |
Collapse
|
36
|
Nakamura M, Maruyama M, Yamashita F, Takakura Y, Hashida M, Watanabe Y. Expression and visualization of a human interferon-beta-enhanced green fluorescent protein chimeric molecule in cultured cells. Biol Pharm Bull 2005; 27:411-4. [PMID: 14993812 DOI: 10.1248/bpb.27.411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have constructed a recombinant cDNA encoding the chimeric protein between human IFN-beta (HuIFN-beta) and enhanced green fluorescent protein (EGFP) to elucidate the intracellular localization of IFN-beta. Transient expression of the chimeric molecule, HuIFN-beta-EGFP, in L cells demonstrated that the chimeric molecule secreted from the cells had an intact biological activity as far as antiviral effect was concerned. Immunostaining of the transfected cells using anti-HuIFN-beta antibody demonstrated that green-fluorescence was co-localized with the IFN signal and its profile was similar to IFN signals in the cells transfected with HuIFN-beta expressing plasmid DNA. These results indicate that the HuIFN-beta-EGFP chimeric gene was expressed as a chimeric protein and the chimera was transported via the regular secretory pathway in the cells. In other cell types, the fluorescence derived from the chimeric protein was also seen on cytoplasmic vesicular structures. These results suggest that HuIFN-beta-EGFP will be a useful tool to investigate the intracellular trafficking processes of HuIFN-beta in a variety of cell types.
Collapse
Affiliation(s)
- Masaru Nakamura
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Venkatesh B, Arifuzzaman M, Mori H, Suzuki S, Taguchi T, Ohmiya Y. Use of GFP tags to monitor localization of different luciferases in E. coli. Photochem Photobiol Sci 2005; 4:740-3. [PMID: 16121286 DOI: 10.1039/b416747c] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The utility of the green fluorescent protein (GFP) as a probe to monitor protein localization in living cells is gaining a great deal of attention. In this study, to understand the localization of luciferases in E. coli, we have attached GFP tags at both the N- and the C-terminus of firefly luciferase (FF-Luc)(from Pyrocoelia miyako) and of red (RE-Luc) and green (GR-Luc) bioluminescence-emitting luciferases (from Phrixothrix railroad-worms), respectively. There was no significant change in the bioluminescence emission spectrum for any of the three luciferases following the tagging with GFP at either the N- or C-terminus, confirming the absence of energy transfer between one another. Using confocal imaging microscopy, we observed that all three luciferases expressed in the E.coli cultured at 37 degrees C tend to aggregate and are seen to localize in the poles, thus confirming their poor folding properties. In contrast, in the E.coli cultured at 18 degrees C FF-Luc was found to be highly expressed in the soluble form when compared to RE-Luc and GR-Luc. These results support our previous finding that the folding properties of FF-Luc and RE/GR-Luc are totally different.
Collapse
Affiliation(s)
- Balan Venkatesh
- Cell Dynamics Research Group, National Institute of Advanced Industrial Science and Technology, Midorigaoka Ikeda, Osaka 563-8577, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Tsuboi T, McMahon HT, Rutter GA. Mechanisms of dense core vesicle recapture following "kiss and run" ("cavicapture") exocytosis in insulin-secreting cells. J Biol Chem 2004; 279:47115-24. [PMID: 15331588 DOI: 10.1074/jbc.m408179200] [Citation(s) in RCA: 152] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The molecular mechanisms underlying "kiss and run" or "cavicapture" exocytosis of dense core secretory vesicles are presently unclear. Although dynamin-1 has previously been implicated in the recapture process in neurons, the recruitment of this fission protein to a single exocytosing vesicle has not been examined in real time during peptide release from pancreatic beta-cells. Imaged simultaneously in clonal insulin-secreting cells by dual color total internal reflection fluorescence microscopy, monomeric red fluorescent protein (mRFP)-tagged neuropeptide Y and green fluorescent protein (GFP)-tagged synaptotagmin-1 or synaptobrevin-2 rapidly diffused from sites of exocytosis, whereas the vesicle membrane protein phogrin and tissue plasminogen activator (tPA) were retained, consistent with fusion pore closure. Vesicle recovery frequently involved the recruitment of enhanced GFP-tagged dynamin-1, and GTPase-defective dynamin-1(K44E) increased the dwell time of tPA-mRFP at the plasma membrane. By contrast, recruitment of GFP chimeras of clathrin, epsin, and amphiphysin was not observed. Expression of dynamin-1(K535A), mutated in the pleckstrin homology domain, caused the apparent full fusion of vesicles, as reported by the additional release of tPA-mRFP (15-nm diameter) and enhanced GFP-tagged phogrin. We conclude that re-uptake of vesicles after peptide release by cavicapture corresponds to a novel form of endocytosis in which dynamin-1 stabilizes and eventually closes the fusion pore, with no requirement for "classical" endocytosis for retreat from the plasma membrane.
Collapse
Affiliation(s)
- Takashi Tsuboi
- Henry Wellcome Laboratories for Integrated Cell Signalling and the Department of Biochemistry, School of Medical Sciences, University Walk, University of Bristol, Bristol BS8 1TD, United Kingdom
| | | | | |
Collapse
|
39
|
Ohara-Imaizumi M, Nishiwaki C, Kikuta T, Nagai S, Nakamichi Y, Nagamatsu S. TIRF imaging of docking and fusion of single insulin granule motion in primary rat pancreatic beta-cells: different behaviour of granule motion between normal and Goto-Kakizaki diabetic rat beta-cells. Biochem J 2004; 381:13-8. [PMID: 15128287 PMCID: PMC1133756 DOI: 10.1042/bj20040434] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2004] [Revised: 05/04/2004] [Accepted: 05/05/2004] [Indexed: 11/17/2022]
Abstract
We imaged and analysed the motion of single insulin secretory granules near the plasma membrane in live pancreatic beta-cells, from normal and diabetic Goto-Kakizaki (GK) rats, using total internal reflection fluorescence microscopy (TIRFM). In normal rat primary beta-cells, the granules that were fusing during the first phase originate from previously docked granules, and those during the second phase originate from 'newcomers'. In diabetic GK rat beta-cells, the number of fusion events from previously docked granules were markedly reduced, and, in contrast, the fusion from newcomers was still preserved. The dynamic change in the number of docked insulin granules showed that, in GK rat beta-cells, the total number of docked insulin granules was markedly decreased to 35% of the initial number after glucose stimulation. Immunohistochemistry with anti-insulin antibody observed by TIRFM showed that GK rat beta-cells had a marked decline of endogenous insulin granules docked to the plasma membrane. Thus our results indicate that the decreased number of docked insulin granules accounts for the impaired insulin release during the first phase of insulin release in diabetic GK rat beta-cells.
Collapse
Affiliation(s)
- Mica Ohara-Imaizumi
- Department of Biochemistry (II), Kyorin University School of Medicine, Shinkawa 6-20-2, Mitaka, Tokyo 181-8611, Japan
| | - Chiyono Nishiwaki
- Department of Biochemistry (II), Kyorin University School of Medicine, Shinkawa 6-20-2, Mitaka, Tokyo 181-8611, Japan
| | - Toshiteru Kikuta
- Department of Biochemistry (II), Kyorin University School of Medicine, Shinkawa 6-20-2, Mitaka, Tokyo 181-8611, Japan
| | - Shintaro Nagai
- Department of Biochemistry (II), Kyorin University School of Medicine, Shinkawa 6-20-2, Mitaka, Tokyo 181-8611, Japan
| | - Yoko Nakamichi
- Department of Biochemistry (II), Kyorin University School of Medicine, Shinkawa 6-20-2, Mitaka, Tokyo 181-8611, Japan
| | - Shinya Nagamatsu
- Department of Biochemistry (II), Kyorin University School of Medicine, Shinkawa 6-20-2, Mitaka, Tokyo 181-8611, Japan
- To whom correspondence should be addressed (e-mail )
| |
Collapse
|
40
|
Rutter GA. Visualising insulin secretion. The Minkowski Lecture 2004. Diabetologia 2004; 47:1861-72. [PMID: 15551048 DOI: 10.1007/s00125-004-1541-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2004] [Accepted: 09/04/2004] [Indexed: 01/09/2023]
Abstract
Insulin secretion from pancreatic islet beta cells is a tightly regulated process, under the close control of blood glucose concentrations, neural inputs and circulating hormones. Defects in glucose-triggered insulin secretion, possibly exacerbated by a decrease in beta cell mass, are ultimately responsible for the development of type 2 diabetes. A full understanding of the mechanisms by which glucose and other nutrients trigger insulin secretion will probably be essential to allow for the development of new therapies of type 2 diabetes and for the derivation of "artificial" beta cells from embryonic stem cells as a treatment for type 1 diabetes. I focus here on recent developments in our understanding of beta cell glucose sensing, achieved in part through the development of recombinant targeted probes (luciferase, green fluorescent protein) that allow islet beta cell metabolism and Ca(2+) handling to be imaged in situ in the intact islet with single cell resolution. Combined with classical biochemistry, these techniques show that the beta cell is uniquely poised, thanks to the expression of low levels of lactate dehydrogenase and plasma membrane lactate/monocarboxylate transporters, to channel glucose carbons towards oxidative metabolism, ATP synthesis and inhibition of AMP-activated protein kinase, a newly defined regulator of insulin release. I also discuss the molecular basis of the recruitment of secretory vesicles to the cell surface, analysed by the use of new imaging techniques including total internal reflection of fluorescence, as well as the "nanomechanics" of the exocytotic event itself.
Collapse
Affiliation(s)
- G A Rutter
- Henry Wellcome Laboratories for Integrated Cell Signalling, School of Medical Sciences, University of Bristol, Bristol, UK.
| |
Collapse
|
41
|
Eiden-Plach A, Zagorc T, Heintel T, Carius Y, Breinig F, Schmitt MJ. Viral preprotoxin signal sequence allows efficient secretion of green fluorescent protein by Candida glabrata, Pichia pastoris, Saccharomyces cerevisiae, and Schizosaccharomyces pombe. Appl Environ Microbiol 2004; 70:961-6. [PMID: 14766577 PMCID: PMC348933 DOI: 10.1128/aem.70.2.961-966.2004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Besides its importance as model organism in eukaryotic cell biology, yeast species have also developed into an attractive host for the expression, processing, and secretion of recombinant proteins. Here we investigated foreign protein secretion in four distantly related yeasts (Candida glabrata, Pichia pastoris, Saccharomyces cerevisiae, and Schizosaccharomyces pombe) by using green fluorescent protein (GFP) as a reporter and a viral secretion signal sequence derived from the K28 preprotoxin (pptox), the precursor of the yeast K28 virus toxin. In vivo expression of GFP fused to the N-terminal pptox leader sequence and/or expression of the entire pptox gene was driven either from constitutive (PGK1 and TPI1) or from inducible and/or repressible (GAL1, AOX1, and NMT1) yeast promoters. In each case, GFP entered the secretory pathway of the corresponding host cell; confocal fluorescence microscopy as well as sodium dodecyl sulfate-polyacrylamide gel electrophoresis and Western analysis of cell-free culture supernatants confirmed that GFP was efficiently secreted into the culture medium. In addition to the results seen with GFP, the full-length viral pptox was correctly processed in all four yeast genera, leading to the secretion of a biologically active virus toxin. Taken together, our data indicate that the viral K28 pptox signal sequence has the potential for being used as a unique tool in recombinant protein production to ensure efficient protein secretion in yeast.
Collapse
Affiliation(s)
- Antje Eiden-Plach
- Angewandte Molekularbiologie, Universität des Saarlandes, D-66041 Saarbrücken, Germany
| | | | | | | | | | | |
Collapse
|
42
|
Tsuboi T, da Silva Xavier G, Leclerc I, Rutter GA. 5'-AMP-activated protein kinase controls insulin-containing secretory vesicle dynamics. J Biol Chem 2003; 278:52042-51. [PMID: 14532293 DOI: 10.1074/jbc.m307800200] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Changes in 5'-AMP-activated protein kinase (AMPK) activity have recently been implicated in the control of insulin secretion by glucose (da Silva Xavier, G., Leclerc, I., Varadi, A., Tsuboi, T., Moule, S. K., and Rutter, G. A. (2003) Biochem. J. 371, 761-774). Here, we examine the possibility that activation of AMPK may regulate distal steps in insulin secretion, including vesicle movement and fusion with the plasma membrane. Vesicle dynamics were imaged in single pancreatic MIN6 beta-cells expressing lumen-targeted pH-insensitive yellow fluorescent protein, neuropeptide Y.Venus, or monomeric red fluorescent protein by total internal reflection fluorescence and Nipkow disc confocal microscopy. Overexpression of a truncated, constitutively active form of AMPK (AMPKalpha1, 1-312, T172D; AMPK CA), inhibited glucose-stimulated (30 versus 3.0 mM) vesicle movements, and decreased the number of vesicles docked or fusing at the plasma membrane, while having no effect on the kinetics of individual secretory events. Expression of the activated form of AMPK also prevented dispersal of the cortical actin network at high glucose concentrations. Monitored in permeabilized cells, where the effects of AMPK CA on glucose metabolism and ATP synthesis were bypassed, AMPK CA inhibited Ca2+ and ATP-induced insulin secretion, and decreased ATP-dependent vesicle movements. These findings suggest that components of the vesicle transport network, including vesicle-associated motor proteins, may be targets of AMPK in beta-cells, dephosphorylation of which is required for vesicle mobilization at elevated glucose concentrations.
Collapse
Affiliation(s)
- Takashi Tsuboi
- Henry Wellcome Laboratories for Integrated Cell Signalling and Department of Biochemistry, School of Medical Sciences, University Walk, University of Bristol, Bristol BS8 1TD, United Kingdom
| | | | | | | |
Collapse
|
43
|
Aoki T, Koch KS, Leffert HL, Watabe H. Application of green fluorescent protein-protein A fusion protein to western blotting. Methods Enzymol 2003; 302:264-72. [PMID: 12876778 DOI: 10.1016/s0076-6879(99)02025-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Affiliation(s)
- T Aoki
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | | | | | | |
Collapse
|
44
|
Osamura RY, Miyai S, Egashira N, Takekoshi S, Yamazaki M, Sanno N, Teramoto A. Application of Genetic Engineering Technologies for the Study of Pituitary Development and Neoplasms. Acta Histochem Cytochem 2003. [DOI: 10.1267/ahc.36.249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | - Shunsuke Miyai
- Department of Pathology, Tokai University School of Medicine
| | - Noboru Egashira
- Department of Pathology, Tokai University School of Medicine
| | | | | | - Naoko Sanno
- Department of Neurosurgery, Nippon Medical School
| | | |
Collapse
|
45
|
Lavoie C, Meerloo T, Lin P, Farquhar MG. Calnuc, an EF-hand Ca(2+)-binding protein, is stored and processed in the Golgi and secreted by the constitutive-like pathway in AtT20 cells. Mol Endocrinol 2002; 16:2462-74. [PMID: 12403836 DOI: 10.1210/me.2002-0079] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Calnuc is an ubiquitous, EF-hand Ca(2+) binding protein found in the cytoplasm where it binds to Galphai3, in the Golgi lumen where it constitutes a Ca(2+) storage pool, and secreted outside the cell. Here we investigated the pathway of secretion of calnuc in AtT20 cells. We found by pulse-chase experiments that calnuc is synthesized in the endoplasmic reticulum, transported to the Golgi where it remains greater than 12 h and undergoes posttranslational modification (O-glycosylation and sulfation) followed by secretion into the culture medium. We examined if calnuc is secreted by the constitutive or regulated secretory pathway in AtT20 cells. By immunofluorescence and immunogold labeling, endogenous calnuc is found in immature secretion granules (ISG) but not mature regulated secretory granules (RSG), whereas overexpressed calnuc-green fluorescent protein (GFP) is found in both ISG and RSG, where it colocalizes with ACTH. Neither calnuc nor calnuc-GFP are released by the regulated secretory pathway, suggesting that endogenous calnuc and calnuc-GFP are progressively removed from ISG and RSG during granule maturation. We conclude that calnuc is secreted via the constitutive-like pathway and represents a useful endogenous marker for this pathway in AtT20 cells. Together, these observations indicate that calnuc has a unique itinerary as it is retained in the Golgi and is then constitutively secreted extracellularly where it may influence cell behavior via its Ca(2+)-binding properties.
Collapse
Affiliation(s)
- Christine Lavoie
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California 92093-0651, USA
| | | | | | | |
Collapse
|
46
|
Li J, Xu H, Herber WK, Bentley WE, Rao G. Integrated bioprocessing in Saccharomyces cerevisiae using green fluorescent protein as a fusion partner. Biotechnol Bioeng 2002; 79:682-93. [PMID: 12209816 DOI: 10.1002/bit.10331] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In this study, we examine the use of green fluorescent protein (GFP) for monitoring a hexokinase (HXK)-GFP fusion protein in Saccharomyces cerevisiae for various events including expression, degradation, purification, and localization. The fusion, HXK-EK-GFP-6 x His, was constructed where the histidine tag (6 x His) would allow for convenient affinity purification, and the enterokinase (EK) cleavage site would be used for separation of HXK from GFP after affinity purification. Our results showed that both HXK and GFP remained active in the fusion and, more importantly, that there was a linear correlation between HXK activity and GFP fluorescence. Enterokinase cleavage studies revealed that both GFP fluorescence intensity and HXK activity remained unchanged after separation of the fusion proteins, which indicated that fusion of GFP did not cause structural alteration of HXK and thus did not affect the enzymatic activity of HXK. We also found that degradation of the fusion protein occurred, and that degradation was limited to HXK with GFP remaining intact in the fusion. Confocal microscopy studies showed that while GFP was distributed evenly in the yeast cytosol, HXK-GFP fusion followed the correct localization of HXK, which resulted in a di-localization of both cytosol and the nucleus. GFP proved to be a useful fusion partner that may lead to the possibility of integrating the bioprocesses by quantitatively following the entire process visually.
Collapse
Affiliation(s)
- Jincai Li
- Department of Chemical and Biochemical Engineering, University of Maryland, Baltimore County, Maryland 21250, USA
| | | | | | | | | |
Collapse
|
47
|
Rizzo MA, Magnuson MA, Drain PF, Piston DW. A functional link between glucokinase binding to insulin granules and conformational alterations in response to glucose and insulin. J Biol Chem 2002; 277:34168-75. [PMID: 12101177 DOI: 10.1074/jbc.m112478200] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glucokinase (GK) activity is essential for the physiological regulation of insulin secretion by glucose. Because the enzyme exerts nearly total control over glucose metabolism in the beta-cell, even small changes in GK activity exert effects on glucose-stimulated insulin secretion and, consequently, the blood glucose concentration. Using quantitative imaging of multicolor fluorescent proteins fused to GK, we found that the association of GK with insulin granules is regulated by glucose in the beta-cell. Glucose stimulation increased the rate of fluorescence recovery after photobleaching of GK to insulin granules, indicating that GK is released into the cytoplasm after glucose stimulation. Changes in fluorescence resonance energy transfer between two different fluorescent protein variants inserted on opposing ends of GK were observed after glucose stimulation and correlated with increased enzyme activity. Furthermore, glucose-stimulated changes in GK regulation were blocked by two inhibitors of insulin secretion. Insulin treatment restored GK regulation in inhibited cells and stimulated GK translocation and activation by itself. Together, these data support a model for post-translational regulation of GK whereby insulin regulates both the association of GK with secretory granules and the activity of the enzyme within the pancreatic beta-cell.
Collapse
Affiliation(s)
- Megan A Rizzo
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | |
Collapse
|
48
|
Koticha DK, McCarthy EE, Baldini G. Plasma membrane targeting of SNAP-25 increases its local concentration and is necessary for SNARE complex formation and regulated exocytosis. J Cell Sci 2002; 115:3341-51. [PMID: 12140265 DOI: 10.1242/jcs.115.16.3341] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
SNAP-25 is an integral protein of the plasma membrane involved in neurotransmission and hormone secretion. The cysteine-rich domain of SNAP-25 is essential for membrane binding and plasma-membrane targeting. However, this domain is not required for SNARE complex formation and fusion of membranes in vitro. In this paper, we describe an `intact-cell'-based system designed to compare the effect of similar amounts of membrane-bound and soluble SNAP-25 proteins on regulated exocytosis. In transfected neuroblastoma cells,Botulinum neurotoxin E (BoNT/E), a protease that cleaves SNAP-25, blocks regulated release of hormone. However, hormone release is rescued by expressing a wild-type SNAP-25 protein resistant to the toxin. BoNT/E-resistant SNAP-25 proteins lacking the cysteine-rich domain or with all the cysteines substituted by alanines do not form SNARE complexes or rescue regulated exocytosis when expressed at the same level as membrane-bound SNAP-25, which is approximately four-fold higher than the endogenous protein. We conclude that the cysteine-rich domain of SNAP-25 is essential for Ca2+-dependent hormone release because, by targeting SNAP-25 to the plasma membrane, it increases its local concentration, leading to the formation of enough SNARE complexes to support exocytosis.
Collapse
Affiliation(s)
- Darshan K Koticha
- Department of Anatomy and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | |
Collapse
|
49
|
Tompkins LS, Nullmeyer KD, Murphy SM, Weber CS, Lynch RM. Regulation of secretory granule pH in insulin-secreting cells. Am J Physiol Cell Physiol 2002; 283:C429-37. [PMID: 12107052 DOI: 10.1152/ajpcell.01066.2000] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Luminal acidification is important for the maturation of secretory granules, yet little is known regarding the regulation of pH within them. A pH-sensitive green fluorescent protein (EGFP) was targeted to secretory granules in RIN1046-38 insulinoma cells by using a construct in which the EGFP gene was preceded by the nucleotide sequence for human growth hormone. Stimulatory levels of glucose doubled EGFP secretion from cell cultures, and potentiators of glucose-induced insulin secretion enhanced EGFP release. Thus this targeted EGFP is useful for population measurements of secretion. However, less than ~4% of total cell EGFP was released after 1.5 h of stimulation. Consequently, when analyzed in single cells, fluorescence of the targeted EGFP acts as an indicator of pH within secretory granules. Glucose elicited a decrease in granule pH, whereas inhibitors of the V-type H(+)-ATPase increased pH and blocked the glucose effect. Granule pH also was modified by effectors of the protein kinase A pathway, with activation eliciting granule alkalinization, suggesting that potentiation of peptide release by cAMP may involve regulated changes in secretory granule pH.
Collapse
Affiliation(s)
- Linda S Tompkins
- Department of Physiology, University of Arizona, Health Sciences Center, Tucson, Arizona 85718, USA
| | | | | | | | | |
Collapse
|
50
|
Watkins S, Geng X, Li L, Papworth G, Robbins PD, Drain P. Imaging secretory vesicles by fluorescent protein insertion in propeptide rather than mature secreted peptide. Traffic 2002; 3:461-71. [PMID: 12047554 DOI: 10.1034/j.1600-0854.2002.30703.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We combined confocal and live-cell imaging with a novel molecular strategy aimed at revealing mechanisms underlying glucose-regulated insulin vesicle secretion. The 'Ins-C-GFP' reporter monitors secretory peptide targeting, trafficking, and exocytosis without directly tagging the mature secreted peptide. We trapped a green fluorescent protein (GFP) reporter in equimolar quantity within the secretory vesicle by fusing it within the C peptide of proinsulin which only after nascent vesicle sealing and acidification is cleaved from the mature secreted A and B chains of insulin. Ins-C-GFP expression in mouse islets without fail exhibited punctate distribution of green fluorescence by confocal microscopy. Ins-C-GFP colocalized GFP with insulin at vesicle dense cores by immuno-electron microscopy. Glucose stimulation decreased vesicle fluorescence coordinately with enhanced secretion from islets of C-GFP detected by anti-GFP Western blots, and of insulin detected by anti-insulin radioimmunoassay. An insulin secretagogue with a red fluorescent label, glibenclamide BODIPY TR, was applied to islets expressing Ins-C-GFP. The stimulus response was imaged as a rise in red secretagogue leading to marked loss in green granules. Since neuropeptides as well as peptide hormones are processed from propeptides after sealing of secretory granules, vesicle trapping likely is widely applicable for studies on targeting, trafficking, and regulated release of secretory peptides.
Collapse
Affiliation(s)
- Simon Watkins
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | |
Collapse
|