1
|
Kamat V, Sweet IR. Hypertonicity during a rapid rise in D-glucose mediates first-phase insulin secretion. Front Endocrinol (Lausanne) 2024; 15:1395028. [PMID: 38989001 PMCID: PMC11233695 DOI: 10.3389/fendo.2024.1395028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Introduction Biphasic insulin secretion is an intrinsic characteristic of the pancreatic islet and has clinical relevance due to the loss of first-phase in patients with Type 2 diabetes. As it has long been shown that first-phase insulin secretion only occurs in response to rapid changes in glucose, we tested the hypothesis that islet response to an increase in glucose is a combination of metabolism plus an osmotic effect where hypertonicity is driving first-phase insulin secretion. Methods Experiments were performed using perifusion analysis of rat, mouse, and human islets. Insulin secretion rate (ISR) and other parameters associated with its regulation were measured in response to combinations of D-glucose and membrane-impermeable carbohydrates (L-glucose or mannitol) designed to dissect the effect of hypertonicity from that of glucose metabolism. Results Remarkably, the appearance of first-phase responses was wholly dependent on changes in tonicity: no first-phase in NAD(P)H, cytosolic calcium, cAMP secretion rate (cAMP SR), or ISR was observed when increased D-glucose concentration was counterbalanced by decreases in membrane-impermeable carbohydrates. When D-glucose was greater than 8 mM, rapid increases in L-glucose without any change in D-glucose resulted in first-phase responses in all measured parameters that were kinetically similar to D-glucose. First-phase ISR was completely abolished by H89 (a non-specific inhibitor of protein kinases) without affecting first-phase calcium response. Defining first-phase ISR as the difference between glucose-stimulated ISR with and without a change in hypertonicity, the peak of first-phase ISR occurred after second-phase ISR had reached steady state, consistent with the well-established glucose-dependency of mechanisms that potentiate glucose-stimulated ISR. Discussion The data collected in this study suggests a new model of glucose-stimulated biphasic ISR where first-phase ISR derives from (and after) a transitory amplification of second-phase ISR and driven by hypertonicity-induced rise in H89-inhibitable kinases likely driven by first-phase responses in cAMP, calcium, or a combination of both.
Collapse
Affiliation(s)
| | - Ian R. Sweet
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States
| |
Collapse
|
2
|
Kamat V, Grumbine MK, Bao K, Mokate K, Khalil G, Cook D, Clearwater B, Hirst R, Harman J, Boeck M, Fu Z, Smith LEH, Goswami M, Wubben TJ, Walker EM, Zhu J, Soleimanpour SA, Scarlett JM, Robbings BM, Hass D, Hurley JB, Sweet IR. A versatile pumpless multi-channel fluidics system for maintenance and real-time functional assessment of tissue and cells. CELL REPORTS METHODS 2023; 3:100642. [PMID: 37963464 PMCID: PMC10694526 DOI: 10.1016/j.crmeth.2023.100642] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/24/2023] [Accepted: 10/20/2023] [Indexed: 11/16/2023]
Abstract
To address the needs of the life sciences community and the pharmaceutical industry in pre-clinical drug development to both maintain and continuously assess tissue metabolism and function with simple and rapid systems, we improved on the initial BaroFuse to develop it into a fully functional, pumpless, scalable multi-channel fluidics instrument that continuously measures changes in oxygen consumption and other endpoints in response to test compounds. We and several other laboratories assessed it with a wide range of tissue types including retina, pancreatic islets, liver, and hypothalamus with both aqueous and gaseous test compounds. The setup time was less than an hour for all collaborating groups, and there was close agreement between data obtained from the different laboratories. This easy-to-use system reliably generates real-time metabolic and functional data from tissue and cells in response to test compounds that will address a critical need in basic and applied research.
Collapse
Affiliation(s)
- Varun Kamat
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA
| | | | - Khang Bao
- EnTox Sciences, Inc., Mercer Island, WA 98040, USA
| | - Kedar Mokate
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA
| | - Gamal Khalil
- EnTox Sciences, Inc., Mercer Island, WA 98040, USA
| | - Daniel Cook
- EnTox Sciences, Inc., Mercer Island, WA 98040, USA
| | | | - Richard Hirst
- Technical Assembly Service Corporation, Seattle, WA 98109, USA
| | - Jarrod Harman
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Myriam Boeck
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Moloy Goswami
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Thomas J Wubben
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Emily M Walker
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 98195, USA
| | - Jie Zhu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 98195, USA
| | - Scott A Soleimanpour
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 98195, USA
| | - Jarrad M Scarlett
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA 98145, USA
| | - Brian M Robbings
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Biochemistry, University of Washington, Seattle, WA 98109, USA
| | - Daniel Hass
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA
| | - James B Hurley
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA
| | - Ian R Sweet
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; EnTox Sciences, Inc., Mercer Island, WA 98040, USA.
| |
Collapse
|
3
|
Grumbine MK, Kamat V, Bao K, Crupi T, Mokate K, Lim R, Chao JR, Robbings BM, Hass DT, Hurley JB, Sweet IR. Maintaining and Assessing Various Tissue and Cell Types of the Eye Using a Novel Pumpless Fluidics System. J Vis Exp 2023:10.3791/65399. [PMID: 37522735 PMCID: PMC10791547 DOI: 10.3791/65399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023] Open
Abstract
Many in vitro models used to investigate tissue function and cell biology require a flow of media to provide adequate oxygenation and optimal cell conditions required for the maintenance of function and viability. Toward this end, we have developed a multi-channel flow culture system to maintain tissue and cells in culture and continuously assess function and viability by either in-line sensors and/or collection of outflow fractions. The system combines 8-channel, continuous optical sensing of oxygen consumption rate with a built-in fraction collector to simultaneously measure production rates of metabolites and hormone secretion. Although it is able to maintain and assess a wide range of tissue and cell models, including islets, muscle, and hypothalamus, here we describe its operating principles and the experimental preparations/protocols that we have used to investigate bioenergetic regulation of isolated mouse retina, mouse retinal pigment epithelium (RPE)-choroid-sclera, and cultured human RPE cells. Innovations in the design of the system, such as pumpless fluid flow, have produced a greatly simplified operation of a multi-channel flow system. Videos and images are shown that illustrate how to assemble, prepare the instrument for an experiment, and load the different tissue/cell models into the perifusion chambers. In addition, guidelines for selecting conditions for protocol- and tissue-specific experiments are delineated and discussed, including setting the correct flow rate to tissue ratio to obtain consistent and stable culture conditions and accurate determinations of consumption and production rates. The combination of optimal tissue maintenance and real-time assessment of multiple parameters yields highly informative data sets that will have great utility for research in the physiology of the eye and drug discovery for the treatment of impaired vision.
Collapse
Affiliation(s)
| | - Varun Kamat
- UW Medicine Diabetes Institute, University of Washington
| | | | | | - Kedar Mokate
- UW Medicine Diabetes Institute, University of Washington
| | - Rayne Lim
- Department of Ophthalmology, University of Washington
| | | | | | - Daniel T Hass
- Department of Biochemistry, University of Washington
| | | | - Ian R Sweet
- EnTox Sciences, Inc; UW Medicine Diabetes Institute, University of Washington;
| |
Collapse
|
4
|
Chichura KS, Elfers CT, Salameh TS, Kamat V, Chepurny OG, McGivney A, Milliken BT, Holz GG, Applebey SV, Hayes MR, Sweet IR, Roth CL, Doyle RP. A peptide triple agonist of GLP-1, neuropeptide Y1, and neuropeptide Y2 receptors promotes glycemic control and weight loss. Sci Rep 2023; 13:9554. [PMID: 37308546 PMCID: PMC10261008 DOI: 10.1038/s41598-023-36178-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 05/24/2023] [Indexed: 06/14/2023] Open
Abstract
Mechanisms underlying long-term sustained weight loss and glycemic normalization after obesity surgery include changes in gut hormone levels, including glucagon-like peptide 1 (GLP-1) and peptide YY (PYY). We demonstrate that two peptide biased agonists (GEP44 and GEP12) of the GLP-1, neuropeptide Y1, and neuropeptide Y2 receptors (GLP-1R, Y1-R, and Y2-R, respectively) elicit Y1-R antagonist-controlled, GLP-1R-dependent stimulation of insulin secretion in both rat and human pancreatic islets, thus revealing the counteracting effects of Y1-R and GLP-1R agonism. These agonists also promote insulin-independent Y1-R-mediated glucose uptake in muscle tissue ex vivo and more profound reductions in food intake and body weight than liraglutide when administered to diet-induced obese rats. Our findings support a role for Y1-R signaling in glucoregulation and highlight the therapeutic potential of simultaneous receptor targeting to achieve long-term benefits for millions of patients.
Collapse
Affiliation(s)
- Kylie S Chichura
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA
| | - Clinton T Elfers
- Seattle Children's Research Institute, 1900 Ninth Ave, Seattle, WA, 98101, USA
| | - Therese S Salameh
- Seattle Children's Research Institute, 1900 Ninth Ave, Seattle, WA, 98101, USA
| | - Varun Kamat
- Diabetes Research Institute and Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, 98195, USA
| | - Oleg G Chepurny
- Department of Medicine, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Aelish McGivney
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA
| | - Brandon T Milliken
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA
| | - George G Holz
- Department of Medicine, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Sarah V Applebey
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ian R Sweet
- Diabetes Research Institute and Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, 98195, USA
| | - Christian L Roth
- Seattle Children's Research Institute, 1900 Ninth Ave, Seattle, WA, 98101, USA.
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, 98105, USA.
| | - Robert P Doyle
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA.
- Department of Medicine, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA.
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
5
|
Fauzi M, Murakami T, Fujimoto H, Botagarova A, Sakaki K, Kiyobayashi S, Ogura M, Inagaki N. Preservation effect of imeglimin on pancreatic β-cell mass: Noninvasive evaluation using 111In-exendin-4 SPECT/CT imaging and the perspective of mitochondrial involvements. Front Endocrinol (Lausanne) 2022; 13:1010825. [PMID: 36246910 PMCID: PMC9559817 DOI: 10.3389/fendo.2022.1010825] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/15/2022] [Indexed: 11/20/2022] Open
Abstract
Progressive loss of β-cell mass (BCM) has a pernicious influence on type 2 diabetes mellitus (T2DM); evaluation of BCM has conventionally required an invasive method that provides only cross-sectional data. However, a noninvasive approach to longitudinal assessment of BCM in living subjects using an indium 111-labeled exendin-4 derivative ([Lys12(111In-BnDTPA-Ahx)]exendin-4) (111In-exendin-4) has been developed recently. Imeglimin is a novel antidiabetic agent that is reported to improve glycemic control and glucose-stimulated insulin secretion (GSIS) via augmentation of mitochondrial function. However, the influence of imeglimin on BCM is not fully understood. We have investigated the effects of imeglimin on BCM in vivo in prediabetic db/db mice using a noninvasive 111In-exendin-4 single-photon emission computed tomography/computed tomography (SPECT/CT) technique. During the 5-week study period, imeglimin treatment attenuated the progression of glucose intolerance, and imeglimin-treated mice retained greater BCM than control, which was consistent with the results of 111In-exendin-4 SPECT/CT scans. Furthermore, immunohistochemical analysis revealed reduced β-cell apoptosis in the imeglimin-treated db/db mice, and also lowered release of cytosolic cytochrome c protein in the β cells. Furthermore, electron microscopy observation and membrane potential measurement revealed improved structural integrity and membrane potential of the mitochondria of imeglimin-treated islets, respectively. These results demonstrate attenuation of progression of BCM loss in prediabetic db/db mice partly via inhibition of mitochondria-mediated apoptosis.
Collapse
Affiliation(s)
- Muhammad Fauzi
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takaaki Murakami
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Fujimoto
- Radioisotope Research Center, Agency of Health, Safety, and Environment, Kyoto University, Kyoto, Japan
| | - Ainur Botagarova
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kentaro Sakaki
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sakura Kiyobayashi
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahito Ogura
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- *Correspondence: Nobuya Inagaki,
| |
Collapse
|
6
|
Kamat V, Robbings BM, Jung SR, Kelly J, Hurley JB, Bube KP, Sweet IR. Fluidics system for resolving concentration-dependent effects of dissolved gases on tissue metabolism. eLife 2021; 10:e66716. [PMID: 34734803 PMCID: PMC8660022 DOI: 10.7554/elife.66716] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 11/01/2021] [Indexed: 12/15/2022] Open
Abstract
Oxygen (O2) and other dissolved gases such as the gasotransmitters H2S, CO, and NO affect cell metabolism and function. To evaluate effects of dissolved gases on processes in tissue, we developed a fluidics system that controls dissolved gases while simultaneously measuring parameters of electron transport, metabolism, and secretory function. We use pancreatic islets, retina, and liver from rodents to highlight its ability to assess effects of O2 and H2S. Protocols aimed at emulating hypoxia-reperfusion conditions resolved a previously unrecognized transient spike in O2 consumption rate (OCR) following replenishment of O2, and tissue-specific recovery of OCR following hypoxia. The system revealed both inhibitory and stimulatory effects of H2S on insulin secretion rate from isolated islets. The unique ability of this new system to quantify metabolic state and cell function in response to precise changes in dissolved gases provides a powerful platform for cell physiologists to study a wide range of disease states.
Collapse
Affiliation(s)
- Varun Kamat
- University of Washington Medicine Diabetes Institute, University of WashingtonSeattleUnited States
| | - Brian M Robbings
- University of Washington Medicine Diabetes Institute, University of WashingtonSeattleUnited States
- Department of Biochemistry, University of WashingtonSeattleUnited States
| | - Seung-Ryoung Jung
- University of Washington Medicine Diabetes Institute, University of WashingtonSeattleUnited States
| | | | - James B Hurley
- Department of Biochemistry, University of WashingtonSeattleUnited States
| | - Kenneth P Bube
- Department of Mathematics, University of WashingtonSeattleUnited States
| | - Ian R Sweet
- University of Washington Medicine Diabetes Institute, University of WashingtonSeattleUnited States
| |
Collapse
|
7
|
Schulze T, Mattern K, Erfle P, Brüning D, Scherneck S, Dietzel A, Rustenbeck I. A Parallel Perifusion Slide From Glass for the Functional and Morphological Analysis of Pancreatic Islets. Front Bioeng Biotechnol 2021; 9:615639. [PMID: 33763408 PMCID: PMC7982818 DOI: 10.3389/fbioe.2021.615639] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/12/2021] [Indexed: 12/15/2022] Open
Abstract
An islet-on-chip system in the form of a completely transparent microscope slide optically accessible from both sides was developed. It is made from laser-structured borosilicate glass and enables the parallel perifusion of five microchannels, each containing one islet precisely immobilized in a pyramidal well. The islets can be in inserted via separate loading windows above each pyramidal well. This design enables a gentle, fast and targeted insertion of the islets and a reliable retention in the well while at the same time permitting a sufficiently fast exchange of the media. In addition to the measurement of the hormone content in the fractionated efflux, parallel live cell imaging of the islet is possible. By programmable movement of the microscopic stage imaging of five wells can be performed. The current chip design ensures sufficient time resolution to characterize typical parameters of stimulus-secretion coupling. This was demonstrated by measuring the reaction of the islets to stimulation by glucose and potassium depolarization. After the perifusion experiment islets can be removed for further analysis. The live-dead assay of the removed islets confirmed that the process of insertion and removal was not detrimental to islet structure and viability. In conclusion, the present islet-on-chip design permits the practical implementation of parallel perifusion experiments on a single and easy to load glass slide. For each immobilized islet the correlation between secretion, signal transduction and morphology is possible. The slide concept allows the scale-up to even higher degrees of parallelization.
Collapse
Affiliation(s)
- Torben Schulze
- Institute of Pharmacology and Toxicology, Technische Universität Braunschweig, Braunschweig, Germany.,Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Braunschweig, Germany
| | - Kai Mattern
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Braunschweig, Germany.,Institute of Microtechnology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Per Erfle
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Braunschweig, Germany.,Institute of Microtechnology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Dennis Brüning
- Institute of Pharmacology and Toxicology, Technische Universität Braunschweig, Braunschweig, Germany.,Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Braunschweig, Germany
| | - Stephan Scherneck
- Institute of Pharmacology and Toxicology, Technische Universität Braunschweig, Braunschweig, Germany.,Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Braunschweig, Germany
| | - Andreas Dietzel
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Braunschweig, Germany.,Institute of Microtechnology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Ingo Rustenbeck
- Institute of Pharmacology and Toxicology, Technische Universität Braunschweig, Braunschweig, Germany.,Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
8
|
Postnatal maturation of calcium signaling in islets of Langerhans from neonatal mice. Cell Calcium 2020; 94:102339. [PMID: 33422769 DOI: 10.1016/j.ceca.2020.102339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 01/02/2023]
Abstract
Pancreatic islet cells develop mature physiological responses to glucose and other fuels postnatally. In this study, we used fluorescence imaging techniques to measure changes in intracellular calcium ([Ca2+]i) to compare islets isolated from mice on postnatal days 0, 4, and 12 with islets from adult CD-1 mice. In addition, we used publicly available RNA-sequencing data to compare expression levels of key genes in β-cell physiology with [Ca2+]i data across these ages. We show that islets isolated from mice on postnatal day 0 displayed elevated [Ca2+]i in basal glucose (≤4 mM) but lower [Ca2+]i responses to stimulation by 12-20 mM glucose compared to adult. Neonatal islets displayed more adult-like [Ca2+]i in basal glucose by day 4 but continued to show lower [Ca2+]i responses to 16 and 20 mM glucose stimulation up to at least day 12. A right shift in glucose sensing (EC50) correlated with lower fragment-per-kilobase-of-transcript-per-million-reads-mapped (FPKM) of Slc2a2 (glut2) and Actn3 and increased FPKM for Galk1 and Nupr1. Differences in [Ca2+]i responses to additional stimuli were also observed. Calcium levels in the endoplasmic reticulum were elevated on day 0 but became adult-like by day 4, which corresponded with reduced expression in Atp2a2 (SERCA2) and novel K+-channel Ktd17, increased expression of Pml, Wfs1, Thada, and Herpud1, and basal [Ca2+]i maturing to adult levels. Ion-channel activity also matured rapidly, but RNA sequencing data mining did not yield strong leads. In conclusion, the maturation of islet [Ca2+]i signaling is complex and multifaceted; several possible gene targets were identified that may participate in this process.
Collapse
|
9
|
Heterogeneous expression of CFTR in insulin-secreting β-cells of the normal human islet. PLoS One 2020; 15:e0242749. [PMID: 33264332 PMCID: PMC7710116 DOI: 10.1371/journal.pone.0242749] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
Cystic fibrosis (CF) is due to mutations in the CF-transmembrane conductance regulator (CFTR) and CF-related diabetes (CFRD) is its most common co-morbidity, affecting ~50% of all CF patients, significantly influencing pulmonary function and longevity. Yet, the complex pathogenesis of CFRD remains unclear. Two non-mutually exclusive underlying mechanisms have been proposed in CFRD: i) damage of the endocrine cells secondary to the severe exocrine pancreatic pathology and ii) intrinsic β-cell impairment of the secretory response in combination with other factors. The later has proven difficult to determine due to low expression of CFTR in β-cells, which results in the general perception that this Cl−channel does not participate in the modulation of insulin secretion or the development of CFRD. The objective of the present work is to demonstrate CFTR expression at the molecular and functional levels in insulin-secreting β-cells in normal human islets, where it seems to play a role. Towards this end, we have used immunofluorescence confocal and immunofluorescence microscopy, immunohistochemistry, RT-qPCR, Western blotting, pharmacology, electrophysiology and insulin secretory studies in normal human, rat and mouse islets. Our results demonstrate heterogeneous CFTR expression in human, mouse and rat β-cells and provide evidence that pharmacological inhibition of CFTR influences basal and stimulated insulin secretion in normal mouse islets but not in islets lacking this channel, despite being detected by electrophysiological means in ~30% of β-cells. Therefore, our results demonstrate a potential role for CFTR in the pancreatic β-cell secretory response suggesting that intrinsic β-cell dysfunction may also participate in the pathogenesis of CFRD.
Collapse
|
10
|
Bax and Bak jointly control survival and dampen the early unfolded protein response in pancreatic β-cells under glucolipotoxic stress. Sci Rep 2020; 10:10986. [PMID: 32620813 PMCID: PMC7335194 DOI: 10.1038/s41598-020-67755-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/12/2020] [Indexed: 12/31/2022] Open
Abstract
ER stress and apoptosis contribute to the loss of pancreatic β-cells under pro-diabetic conditions of glucolipotoxicity. Although activation of canonical intrinsic apoptosis is known to require pro-apoptotic Bcl-2 family proteins Bax and Bak, their individual and combined involvement in glucolipotoxic β-cell death are not known. It has also remained an open question if Bax and Bak in β-cells have non-apoptotic roles in mitochondrial function and ER stress signaling, as suggested in other cell types. Using mice with individual or combined β-cell deletion of Bax and Bak, we demonstrated that glucolipotoxic β-cell death in vitro occurs by both non-apoptotic and apoptotic mechanisms, and the apoptosis could be triggered by either Bax or Bak alone. In contrast, they had non-redundant roles in mediating staurosporine-induced apoptosis. We further established that Bax and Bak do not affect normal glucose-stimulated β-cell Ca2+ responses, insulin secretion, or in vivo glucose tolerance. Finally, our experiments revealed that combined deletion of Bax and Bak amplified the unfolded protein response in islets during the early stages of chemical- or glucolipotoxicity-induced ER stress. These findings shed new light on roles of the core apoptosis machinery in β-cell survival and stress signals of importance for the pathobiology of diabetes.
Collapse
|
11
|
Cardenas-Diaz FL, Leavens KF, Kishore S, Osorio-Quintero C, Chen YJ, Stanger BZ, Wang P, French D, Gadue P. A Dual Reporter EndoC-βH1 Human β-Cell Line for Efficient Quantification of Calcium Flux and Insulin Secretion. Endocrinology 2020; 161:bqaa005. [PMID: 31960055 PMCID: PMC7028009 DOI: 10.1210/endocr/bqaa005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 01/07/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
Abstract
Human in vitro model systems of diabetes are critical to both study disease pathophysiology and offer a platform for drug testing. We have generated a set of tools in the human β-cell line EndoC-βH1 that allows the efficient and inexpensive characterization of β-cell physiology and phenotypes driven by disruption of candidate genes. First, we generated a dual reporter line that expresses a preproinsulin-luciferase fusion protein along with GCaMP6s. This reporter line allows the quantification of insulin secretion by measuring luciferase activity and calcium flux, a critical signaling step required for insulin secretion, via fluorescence microscopy. Using these tools, we demonstrate that the generation of the reporter human β-cell line was highly efficient and validated that luciferase activity could accurately reflect insulin secretion. Second, we used a lentiviral vector carrying the CRISPR-Cas9 system to generate candidate gene disruptions in the reporter line. We also show that we can achieve gene disruption in ~90% of cells using a CRISPR-Cas9 lentiviral system. As a proof of principle, we disrupt the β-cell master regulator, PDX1, and show that mutant EndoC-βH1 cells display impaired calcium responses and fail to secrete insulin when stimulated with high glucose. Furthermore, we show that PDX1 mutant EndoC-βH1 cells exhibit decreased expression of the β-cell-specific genes MAFA and NKX6.1 and increased GCG expression. The system presented here provides a platform to quickly and easily test β-cell functionality in wildtype and cells lacking a gene of interest.
Collapse
Affiliation(s)
- Fabian L Cardenas-Diaz
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Karla F Leavens
- Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Siddharth Kishore
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Cell and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Catherine Osorio-Quintero
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Yi-Ju Chen
- Department of Cell and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ben Z Stanger
- Department of Cell and Molecular Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Gastroenterology Division, Department of Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia
| | - Pei Wang
- Departments of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Deborah French
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Paul Gadue
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
12
|
Kuok IT, Rountree AM, Jung SR, Sweet IR. Palmitate is not an effective fuel for pancreatic islets and amplifies insulin secretion independent of calcium release from endoplasmic reticulum. Islets 2019; 11:51-64. [PMID: 31084524 PMCID: PMC6548485 DOI: 10.1080/19382014.2019.1601490] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The aim of the study was to determine the acute contribution of fuel oxidation in mediating the increase in insulin secretion rate (ISR) in response to fatty acids. Measures of mitochondrial metabolism, as reflected by oxygen consumption rate (OCR) and cytochrome c reduction, calcium signaling, and ISR by rat islets were used to evaluate processes stimulated by acute exposure to palmitic acid (PA). The contribution of mitochondrial oxidation of PA was determined in the presence and absence of a blocker of mitochondrial transport of fatty acids (etomoxir) at different glucose concentrations. Subsequent to increasing glucose from 3 to 20 mM, PA caused small increases in OCR and cytosolic calcium (about 20% of the effect of glucose). In contrast, the effect of PA on ISR was almost 3 times that by glucose, suggesting that the metabolism of PA is not the dominant mechanism mediating PA's effect on ISR. This was further supported by lack of inhibition of PA-stimulated OCR and ISR when blocking entry of PA into mitochondria (with etomoxir), and PA's lack of stimulation of reduced cytochrome c in the presence of high glucose. Consistent with the lack of metabolic stimulation by PA, an inhibitor of calcium release from the endoplasmic reticulum, but not a blocker of L-type calcium channels, abolished the PA-induced elevation of cytosolic calcium. Notably, ISR was unaffected by thapsigargin showing the dissociation of endoplasmic reticulum calcium release and second phase insulin secretion. In conclusion, stimulation of ISR by PA was mediated by mechanisms largely independent of the oxidation of the fuel.
Collapse
Affiliation(s)
- Iok Teng Kuok
- University of Washington Diabetes Research Institute, University of Washington, Seattle, WA, USA
| | - Austin M. Rountree
- University of Washington Diabetes Research Institute, University of Washington, Seattle, WA, USA
| | - Seung-Ryoung Jung
- University of Washington Diabetes Research Institute, University of Washington, Seattle, WA, USA
| | - Ian R. Sweet
- University of Washington Diabetes Research Institute, University of Washington, Seattle, WA, USA
- CONTACT Ian R. Sweet UW Diabetes Institute, University of Washington, Box 358062, 750 Republican Street, Seattle, WA 98195-8062
| |
Collapse
|
13
|
Ahn C, Kang HS, Lee JH, Hong EJ, Jung EM, Yoo YM, Jeung EB. Bisphenol A and octylphenol exacerbate type 1 diabetes mellitus by disrupting calcium homeostasis in mouse pancreas. Toxicol Lett 2018; 295:162-172. [PMID: 29935216 DOI: 10.1016/j.toxlet.2018.06.1071] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 05/14/2018] [Accepted: 06/17/2018] [Indexed: 12/17/2022]
Abstract
In pancreatic β cells, which produce and secrete insulin, Ca2+ signals contribute to insulin production and secretion. Bisphenol A (BPA) and octylphenol (OP) are reported to increase plasma insulin levels and insulin transcription factors, but regulation of plasma glucose levels did not decrease proportionally to the insulin increase. We hypothesized that BPA and OP disrupt calcium homeostasis resulting in insulin resistance through induction of endoplasmic reticulum (ER) stress. BPA and OP treatment leads to survival of pancreatic β cells against streptozotocin, but despite an increased insulin level, serum glucose regulation is not properly regulated. The expression of genes involved in transporting calcium ions to the cytosol and ER decreased while the expression of those affecting the removal of calcium from the cytosol and ER increased. Depletion of calcium from the ER leads to ER stress and can induce insulin resistance. Insulin resistance is also confirmed by insulin-responsive gene, such as glucose transporter 4 (GLUT4) and IRS2, expression. Taken together, these results imply that disruption of calcium homeostasis by BPA and OP induces ER stress and leads to insulin resistance, especially in a streptozotocin (STZ) -induced type 1 diabetes mellitus model.
Collapse
Affiliation(s)
- Changhwan Ahn
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Hong-Seok Kang
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Jae-Hwan Lee
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Eui-Ju Hong
- Laboratory of Veterinary Biochemistry, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134 Republic of Korea
| | - Eui-Man Jung
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Yeong-Min Yoo
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea.
| |
Collapse
|
14
|
Abstract
Pancreatic islet β cells secrete insulin in response to nutrient secretagogues, like glucose, dependent on calcium influx and nutrient metabolism. One of the most intriguing qualities of β cells is their ability to use metabolism to amplify the amount of secreted insulin independent of further alterations in intracellular calcium. Many years studying this amplifying process have shaped our current understanding of β cell stimulus-secretion coupling; yet, the exact mechanisms of amplification have been elusive. Recent studies utilizing metabolomics, computational modeling, and animal models have progressed our understanding of the metabolic amplifying pathway of insulin secretion from the β cell. New approaches will be discussed which offer in-roads to a more complete model of β cell function. The development of β cell therapeutics may be aided by such a model, facilitating the targeting of aspects of the metabolic amplifying pathway which are unique to the β cell.
Collapse
Affiliation(s)
- Michael A Kalwat
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| | - Melanie H Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
15
|
Kursan S, McMillen TS, Beesetty P, Dias-Junior E, Almutairi MM, Sajib AA, Kozak JA, Aguilar-Bryan L, Di Fulvio M. The neuronal K +Cl - co-transporter 2 (Slc12a5) modulates insulin secretion. Sci Rep 2017; 7:1732. [PMID: 28496181 PMCID: PMC5431760 DOI: 10.1038/s41598-017-01814-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 04/03/2017] [Indexed: 11/09/2022] Open
Abstract
Intracellular chloride concentration ([Cl-]i) in pancreatic β-cells is kept above electrochemical equilibrium due to the predominant functional presence of Cl- loaders such as the Na+K+2Cl- co-transporter 1 (Slc12a2) over Cl-extruders of unidentified nature. Using molecular cloning, RT-PCR, Western blotting, immunolocalization and in vitro functional assays, we establish that the "neuron-specific" K+Cl- co-transporter 2 (KCC2, Slc12a5) is expressed in several endocrine cells of the pancreatic islet, including glucagon secreting α-cells, but particularly in insulin-secreting β-cells, where we provide evidence for its role in the insulin secretory response. Three KCC2 splice variants were identified: the formerly described KCC2a and KCC2b along with a novel one lacking exon 25 (KCC2a-S25). This new variant is undetectable in brain or spinal cord, the only and most abundant known sources of KCC2. Inhibition of KCC2 activity in clonal MIN6 β-cells increases basal and glucose-stimulated insulin secretion and Ca2+ uptake in the presence of glibenclamide, an inhibitor of the ATP-dependent potassium (KATP)-channels, thus suggesting a possible mechanism underlying KCC2-dependent insulin release. We propose that the long-time considered "neuron-specific" KCC2 co-transporter is expressed in pancreatic islet β-cells where it modulates Ca2+-dependent insulin secretion.
Collapse
Affiliation(s)
- Shams Kursan
- Department of Pharmacology and Toxicology, Wright State University, School of Medicine, Dayton, OH, 45435, USA
| | | | - Pavani Beesetty
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, School of Medicine, Dayton, OH, 45435, USA
| | - Eduardo Dias-Junior
- Department of Pharmacology and Toxicology, Wright State University, School of Medicine, Dayton, OH, 45435, USA
| | - Mohammed M Almutairi
- Department of Pharmacology and Toxicology, Wright State University, School of Medicine, Dayton, OH, 45435, USA
| | - Abu A Sajib
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - J Ashot Kozak
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, School of Medicine, Dayton, OH, 45435, USA
| | | | - Mauricio Di Fulvio
- Department of Pharmacology and Toxicology, Wright State University, School of Medicine, Dayton, OH, 45435, USA.
| |
Collapse
|
16
|
A method for high-throughput functional imaging of single cells within heterogeneous cell preparations. Sci Rep 2016; 6:39319. [PMID: 27982116 PMCID: PMC5159830 DOI: 10.1038/srep39319] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 11/17/2016] [Indexed: 01/25/2023] Open
Abstract
Functional characterization of individual cells within heterogeneous tissue preparations is challenging. Here, we report the development of a versatile imaging method that assesses single cell responses of various endpoints in real time, while identifying the individual cell types. Endpoints that can be measured include (but are not limited to) ionic flux (calcium, sodium, potassium and hydrogen), metabolic responsiveness (NAD(P)H, mitochondrial membrane potential), and signal transduction (H2O2 and cAMP). Subsequent to fluorescent imaging, identification of cell types using immunohistochemistry allows for mapping of cell type to their respective functional real time responses. To validate the utility of this method, NAD(P)H responses to glucose of islet alpha versus beta cells generated from dispersed pancreatic islets, followed by the construction of frequency distributions characterizing the variability in the magnitude of each individual cell responses were compared. As expected, no overlap between the glucose response frequency distributions for beta cells versus alpha cells was observed, thereby establishing both the high degree of fidelity and low rate of both false-negatives and false-positives in this approach. This novel method has the ability not only to resolve single cell level functional differences between cell types, but also to characterize functional heterogeneity within a given cell type.
Collapse
|
17
|
Tennant BR, Chen J, Shih AZL, Luciani DS, Hoffman BG. Myt3 Mediates Laminin-V/Integrin-β1-Induced Islet-Cell Migration via Tgfbi. Mol Endocrinol 2015; 29:1254-68. [PMID: 26177052 PMCID: PMC5414683 DOI: 10.1210/me.2014-1387] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 07/10/2015] [Indexed: 12/17/2022] Open
Abstract
Myt3 is a prosurvival factor in pancreatic islets; however, its role in islet-cell development is not known. Here, we demonstrate that myelin transcription factor 3 (Myt3) is expressed in migrating islet cells in the developing and neonatal pancreas and thus sought to determine whether Myt3 plays a role in this process. Using an ex vivo model of islet-cell migration, we demonstrate that Myt3 suppression significantly inhibits laminin-V/integrin-β1-dependent α- and β-cell migration onto 804G, and impaired 804G-induced F-actin and E-cadherin redistribution. Exposure of islets to proinflammatory cytokines, which suppress Myt3 expression, had a similar effect, whereas Myt3 overexpression partially rescued the migratory ability of the islet cells. We show that loss of islet-cell migration, due to Myt3 suppression or cytokine exposure, is independent of effects on islet-cell survival or proliferation. Myt3 suppression also had no effect on glucose-induced calcium influx, F-actin remodeling or insulin secretion by β-cells. RNA-sequencing (RNA-seq) analysis of transduced islets showed that Myt3 suppression results in the up-regulation of Tgfbi, a secreted diabetogenic factor thought to impair cellular adhesion. Exposure of islets to exogenous transforming growth factor β-induced (Tgfbi) impaired islet-cell migration similar to Myt3 suppression. Taken together, these data suggest a model by which cytokine-induced Myt3 suppression leads to Tgfbi de-repression and subsequently to impaired islet-cell migration, revealing a novel role for Myt3 in regulating islet-cell migration.
Collapse
Affiliation(s)
- Bryan R Tennant
- Child and Family Research Institute (B.R.T., J.C., A.Z.L.S., D.S.L., B.G.H.), British Columbia Children's Hospital and Sunny Hill Health Centre, Vancouver, British Columbia, Canada V5Z 4H4; and Department of Surgery (D.S.L., B.G.H.), University of British Columbia, Vancouver, British Columbia, Canada V5Z 4E3
| | - Jenny Chen
- Child and Family Research Institute (B.R.T., J.C., A.Z.L.S., D.S.L., B.G.H.), British Columbia Children's Hospital and Sunny Hill Health Centre, Vancouver, British Columbia, Canada V5Z 4H4; and Department of Surgery (D.S.L., B.G.H.), University of British Columbia, Vancouver, British Columbia, Canada V5Z 4E3
| | - Alexis Z L Shih
- Child and Family Research Institute (B.R.T., J.C., A.Z.L.S., D.S.L., B.G.H.), British Columbia Children's Hospital and Sunny Hill Health Centre, Vancouver, British Columbia, Canada V5Z 4H4; and Department of Surgery (D.S.L., B.G.H.), University of British Columbia, Vancouver, British Columbia, Canada V5Z 4E3
| | - Dan S Luciani
- Child and Family Research Institute (B.R.T., J.C., A.Z.L.S., D.S.L., B.G.H.), British Columbia Children's Hospital and Sunny Hill Health Centre, Vancouver, British Columbia, Canada V5Z 4H4; and Department of Surgery (D.S.L., B.G.H.), University of British Columbia, Vancouver, British Columbia, Canada V5Z 4E3
| | - Brad G Hoffman
- Child and Family Research Institute (B.R.T., J.C., A.Z.L.S., D.S.L., B.G.H.), British Columbia Children's Hospital and Sunny Hill Health Centre, Vancouver, British Columbia, Canada V5Z 4H4; and Department of Surgery (D.S.L., B.G.H.), University of British Columbia, Vancouver, British Columbia, Canada V5Z 4E3
| |
Collapse
|