1
|
Uo T, Ojo KK, Sprenger CC, Epilepsia KS, Perera BGK, Damodarasamy M, Sun S, Kim S, Hogan HH, Hulverson MA, Choi R, Whitman GR, Barrett LK, Michaels SA, Xu LH, Sun VL, Arnold SLM, Pang HJ, Nguyen MM, Vigil ALBG, Kamat V, Sullivan LB, Sweet IR, Vidadala R, Maly DJ, Van Voorhis WC, Plymate SR. A Compound that Inhibits Glycolysis in Prostate Cancer Controls Growth of Advanced Prostate Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.01.547355. [PMID: 37461469 PMCID: PMC10350011 DOI: 10.1101/2023.07.01.547355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Purpose Metastatic castration-resistant prostate cancer remains incurable regardless of recent therapeutic advances. Prostate cancer tumors display highly glycolytic phenotypes as the cancer progresses. Non-specific inhibitors of glycolysis have not been utilized successfully for chemotherapy, because of their penchant to cause systemic toxicity. This study reports the preclinical activity, safety, and pharmacokinetics of a novel small molecule preclinical candidate, BKIDC-1553, with antiglycolytic activity. Experimental design We tested a large battery of prostate cancer cell lines for inhibition of cell proliferation, in vitro. Cell cycle, metabolic and enzymatic assays were used to demonstrate their mechanism of action. A human PDX model implanted in mice and a human organoid were studied for sensitivity to our BKIDC preclinical candidate. A battery of pharmacokinetic experiments, absorption, distribution, metabolism, and excretion experiments, and in vitro and in vivo toxicology experiments were carried out to assess readiness for clinical trials. Results We demonstrate a new class of small molecule inhibitors where antiglycolytic activity in prostate cancer cell lines is mediated through inhibition of hexokinase 2. These compounds display selective growth inhibition across multiple prostate cancer models. We describe a lead BKIDC-1553 that demonstrates promising activity in a preclinical xenograft model of advanced prostate cancer, equivalent to that of enzalutamide. BKIDC-1553 demonstrates safety and pharmacologic properties consistent with a compound that can be taken into human studies with expectations of a good safety margin and predicted dosing for efficacy. Conclusion This work supports testing BKIDC-1553 and its derivatives in clinical trials for patients with advanced prostate cancer.
Collapse
|
2
|
Pharaoh G, Kamat V, Kannan S, Stuppard RS, Whitson J, Martín-Pérez M, Qian WJ, MacCoss MJ, Villén J, Rabinovitch P, Campbell MD, Sweet IR, Marcinek DJ. The mitochondrially targeted peptide elamipretide (SS-31) improves ADP sensitivity in aged mitochondria by increasing uptake through the adenine nucleotide translocator (ANT). GeroScience 2023; 45:3529-3548. [PMID: 37462785 PMCID: PMC10643647 DOI: 10.1007/s11357-023-00861-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/23/2023] [Indexed: 07/28/2023] Open
Abstract
Aging muscle experiences functional decline in part mediated by impaired mitochondrial ADP sensitivity. Elamipretide (ELAM) rapidly improves physiological and mitochondrial function in aging and binds directly to the mitochondrial ADP transporter ANT. We hypothesized that ELAM improves ADP sensitivity in aging leading to rescued physiological function. We measured the response to ADP stimulation in young and old muscle mitochondria with ELAM treatment, in vivo heart and muscle function, and compared protein abundance, phosphorylation, and S-glutathionylation of ADP/ATP pathway proteins. ELAM treatment increased ADP sensitivity in old muscle mitochondria by increasing uptake of ADP through the ANT and rescued muscle force and heart systolic function. Protein abundance in the ADP/ATP transport and synthesis pathway was unchanged, but ELAM treatment decreased protein s-glutathionylation incuding of ANT. Mitochondrial ADP sensitivity is rapidly modifiable. This research supports the hypothesis that ELAM improves ANT function in aging and links mitochondrial ADP sensitivity to physiological function. ELAM binds directly to ANT and ATP synthase and ELAM treatment improves ADP sensitivity, increases ATP production, and improves physiological function in old muscles. ADP (adenosine diphosphate), ATP (adenosine triphosphate), VDAC (voltage-dependent anion channel), ANT (adenine nucleotide translocator), H+ (proton), ROS (reactive oxygen species), NADH (nicotinamide adenine dinucleotide), FADH2 (flavin adenine dinucleotide), O2 (oxygen), ELAM (elamipretide), -SH (free thiol), -SSG (glutathionylated protein).
Collapse
Affiliation(s)
- Gavin Pharaoh
- Department of Radiology, University of Washington, Seattle, WA, 98195, USA
| | - Varun Kamat
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Sricharan Kannan
- Department of Radiology, University of Washington, Seattle, WA, 98195, USA
| | - Rudolph S Stuppard
- Department of Radiology, University of Washington, Seattle, WA, 98195, USA
| | - Jeremy Whitson
- Department of Biology, High Point University, High Point, NC, 27268, USA
| | - Miguel Martín-Pérez
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, 08028, Barcelona, Spain
| | - Wei-Jun Qian
- Integrative Omics Group, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Judit Villén
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Peter Rabinovitch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Matthew D Campbell
- Department of Radiology, University of Washington, Seattle, WA, 98195, USA
| | - Ian R Sweet
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - David J Marcinek
- Department of Radiology, University of Washington, Seattle, WA, 98195, USA.
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA.
| |
Collapse
|
3
|
Nishi K, Yoshii A, Abell L, Zhou B, Frausto R, Ritterhoff J, McMillen TS, Sweet I, Wang Y, Gao C, Tian R. Branched-chain keto acids inhibit mitochondrial pyruvate carrier and suppress gluconeogenesis in hepatocytes. Cell Rep 2023; 42:112641. [PMID: 37310861 PMCID: PMC10592489 DOI: 10.1016/j.celrep.2023.112641] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/06/2023] [Accepted: 05/29/2023] [Indexed: 06/15/2023] Open
Abstract
Branched-chain amino acid (BCAA) metabolism is linked to glucose homeostasis, but the underlying signaling mechanisms are unclear. We find that gluconeogenesis is reduced in mice deficient of Ppm1k, a positive regulator of BCAA catabolism, which protects against obesity-induced glucose intolerance. Accumulation of branched-chain keto acids (BCKAs) inhibits glucose production in hepatocytes. BCKAs suppress liver mitochondrial pyruvate carrier (MPC) activity and pyruvate-supported respiration. Pyruvate-supported gluconeogenesis is selectively suppressed in Ppm1k-deficient mice and can be restored with pharmacological activation of BCKA catabolism by BT2. Finally, hepatocytes lack branched-chain aminotransferase that alleviates BCKA accumulation via reversible conversion between BCAAs and BCKAs. This renders liver MPC most susceptible to circulating BCKA levels hence a sensor of BCAA catabolism.
Collapse
Affiliation(s)
- Kiyoto Nishi
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican Street, Seattle, WA 98109, USA; Department of Pharmacology, Shiga University of Medical Science, Otsu, Shiga 520-2182, Japan
| | - Akira Yoshii
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Lauren Abell
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Bo Zhou
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Ricardo Frausto
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Julia Ritterhoff
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Timothy S McMillen
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Ian Sweet
- University of Washington Medicine Diabetes Institute, University of Washington, 750 Republican Street, Seattle, WA 98109, USA
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; Signature Program in Cardiovascular and Metabolic Diseases, Duke-NUS School of Medicine, Singapore, Singapore
| | - Chen Gao
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH 45267-0575, USA.
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican Street, Seattle, WA 98109, USA.
| |
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: 3] [Impact Index Per Article: 3.0] [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
|
Pharaoh G, Kamat V, Kannan S, Stuppard RS, Whitson J, Martin-Perez M, Qian WJ, MacCoss MJ, Villen J, Rabinovitch P, Campbell MD, Sweet IR, Marcinek DJ. Elamipretide Improves ADP Sensitivity in Aged Mitochondria by Increasing Uptake through the Adenine Nucleotide Translocator (ANT). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.525989. [PMID: 36778398 PMCID: PMC9915686 DOI: 10.1101/2023.02.01.525989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aging muscle experiences functional decline in part mediated by impaired mitochondrial ADP sensitivity. Elamipretide (ELAM) rapidly improves physiological and mitochondrial function in aging and binds directly to the mitochondrial ADP transporter ANT. We hypothesized that ELAM improves ADP sensitivity in aging leading to rescued physiological function. We measured the response to ADP stimulation in young and old muscle mitochondria with ELAM treatment, in vivo heart and muscle function, and compared protein abundance, phosphorylation, and S-glutathionylation of ADP/ATP pathway proteins. ELAM treatment increased ADP sensitivity in old muscle mitochondria by increasing uptake of ADP through the ANT and rescued muscle force and heart systolic function. Protein abundance in the ADP/ATP transport and synthesis pathway was unchanged, but ELAM treatment decreased protein s-glutathionylation incuding of ANT. Mitochondrial ADP sensitivity is rapidly modifiable. This research supports the hypothesis that ELAM improves ANT function in aging and links mitochondrial ADP sensitivity to physiological function. Abstract Figure
Collapse
Affiliation(s)
- Gavin Pharaoh
- Department of Radiology, University of Washington, Seattle, Washington, 98195, USA
| | - Varun Kamat
- Department of Medicine, University of Washington, Seattle, Washington, 98195, USA
| | - Sricharan Kannan
- Department of Radiology, University of Washington, Seattle, Washington, 98195, USA
| | - Rudolph S. Stuppard
- Department of Radiology, University of Washington, Seattle, Washington, 98195, USA
| | - Jeremy Whitson
- Department of Biology, High Point University, High Point, NC, 27268, USA
| | - Miguel Martin-Perez
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, 08028, Spain
| | - Wei-Jun Qian
- Integrative Omics Group, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Michael J. MacCoss
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Judit Villen
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Peter Rabinovitch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Matthew D. Campbell
- Department of Radiology, University of Washington, Seattle, Washington, 98195, USA
| | - Ian R. Sweet
- Department of Medicine, University of Washington, Seattle, Washington, 98195, USA
| | - David J. Marcinek
- Department of Radiology, University of Washington, Seattle, Washington, 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA
| |
Collapse
|
6
|
Hass DT, Bisbach CM, Robbings BM, Sadilek M, Sweet IR, Hurley JB. Succinate metabolism in the retinal pigment epithelium uncouples respiration from ATP synthesis. Cell Rep 2022; 39:110917. [PMID: 35675773 PMCID: PMC9251713 DOI: 10.1016/j.celrep.2022.110917] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/08/2022] [Accepted: 05/12/2022] [Indexed: 11/28/2022] Open
Abstract
Fumarate can be a surrogate for O2 as a terminal electron acceptor in the electron transport chain. Reduction of fumarate produces succinate, which can be exported. It is debated whether intact tissues can import and oxidize succinate produced by other tissues. In a previous report, we showed that mitochondria in retinal pigment epithelium (RPE)-choroid preparations can use succinate to reduce O2 to H2O. However, cells in that preparation could have been disrupted during tissue isolation. We now use multiple strategies to quantify intactness of the isolated RPE-choroid tissue. We find that exogenous 13C4-succinate is oxidized by intact cells then exported as fumarate or malate. Unexpectedly, we also find that oxidation of succinate is different from oxidation of other substrates because it uncouples electron transport from ATP synthesis. Retinas produce and export succinate. Our findings imply that retina succinate may substantially increase O2 consumption by uncoupling adjacent RPE mitochondria. The retina releases succinate, a source of reducing power for mitochondria. Hass et al. outline a pathway by which retina succinate can enter intact RPE-choroid cells and stimulate mitochondrial respiration that is uncoupled from ATP synthesis. Rapid RPE succinate oxidation may limit O2 levels in the retina.
Collapse
Affiliation(s)
- Daniel T Hass
- Biochemistry Department, The University of Washington, Seattle, WA 98195, USA
| | - Celia M Bisbach
- Biochemistry Department, The University of Washington, Seattle, WA 98195, USA; Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
| | - Brian M Robbings
- Biochemistry Department, The University of Washington, Seattle, WA 98195, USA; Diabetes Institute, The University of Washington, Seattle, WA 98109, USA
| | - Martin Sadilek
- Chemistry Department, The University of Washington, Seattle, WA 98195, USA
| | - Ian R Sweet
- Diabetes Institute, The University of Washington, Seattle, WA 98109, USA; Division of Metabolism, Endocrinology and Nutrition, The University of Washington, Seattle, WA 98195, USA
| | - James B Hurley
- Biochemistry Department, The University of Washington, Seattle, WA 98195, USA; Opthalmology Department, The University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
7
|
Zeng N, Wang Y, Cheng Y, Huang Z, Song B. Imaging evaluation of the pancreas in diabetic patients. Abdom Radiol (NY) 2022; 47:715-726. [PMID: 34786594 DOI: 10.1007/s00261-021-03340-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 02/05/2023]
Abstract
Diabetes mellitus (DM) is becoming a global epidemic and its diagnosis and monitoring are based on laboratory testing which sometimes have limitations. The pancreas plays a key role in metabolism and is involved in the pathogenesis of DM. It has long been known through cadaver biopsies that pancreas volume is decreased in patients with DM. With the development of different imaging modalities over the last two decades, many studies have attempted to determine whether there other changes occurred in the pancreas of diabetic patients. This review summarizes current knowledge about the use of different imaging approaches (such as CT, MR, and US) and radiomics for exploring pancreatic changes in diabetic patients. Imaging studies are expected to produce reliable information regarding DM, and radiomics could provide increasingly valuable information to identify some undetectable features and help diagnose and predict the occurrence of diabetes through pancreas imaging.
Collapse
Affiliation(s)
- Ni Zeng
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Yi Wang
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Yue Cheng
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Zixing Huang
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
8
|
Kahraman S, Manna D, Dirice E, Maji B, Small J, Wagner BK, Choudhary A, Kulkarni RN. Harnessing reaction-based probes to preferentially target pancreatic β-cells and β-like cells. Life Sci Alliance 2021; 4:4/4/e202000840. [PMID: 33514654 PMCID: PMC7898467 DOI: 10.26508/lsa.202000840] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 01/10/2023] Open
Abstract
Highly sensitive approaches to target insulin-expressing cells would allow more effective imaging, sorting, and analysis of pancreatic β-cells. Here, we introduce the use of a reaction-based probe, diacetylated Zinpyr1 (DA-ZP1), to image pancreatic β-cells and β-like cells derived from human pluripotent stem cells. We harness the high intracellular zinc concentration of β-cells to induce a fluorescence signal in cells after administration of DA-ZP1. Given its specificity and rapid uptake by cells, we used DA-ZP1 to purify live stem cell-derived β-like cells as confirmed by immunostaining analysis. We tested the ability of DA-ZP1 to image transplanted human islet grafts and endogenous mouse pancreatic islets in vivo after its systemic administration into mice. Thus, DA-ZP1 enables purification of insulin-secreting β-like cells for downstream applications, such as functional studies, gene-expression, and cell-cell interaction analyses and can be used to label engrafted human islets and endogenous mouse islets in vivo.
Collapse
Affiliation(s)
- Sevim Kahraman
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Debasish Manna
- Chemical Biology and Therapeutics Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA.,Divisions of Renal Medicine and Engineering, Brigham and Women's Hospital, Boston, MA, USA
| | - Ercument Dirice
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Basudeb Maji
- Chemical Biology and Therapeutics Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA.,Divisions of Renal Medicine and Engineering, Brigham and Women's Hospital, Boston, MA, USA
| | - Jonnell Small
- Chemical Biology and Therapeutics Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Chemical Biology Program, Harvard University, Cambridge, MA, USA
| | - Bridget K Wagner
- Chemical Biology and Therapeutics Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amit Choudhary
- Chemical Biology and Therapeutics Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA .,Department of Medicine, Harvard Medical School, Boston, MA, USA.,Divisions of Renal Medicine and Engineering, Brigham and Women's Hospital, Boston, MA, USA.,Chemical Biology Program, Harvard University, Cambridge, MA, USA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Molecular Imaging of Autoimmune Diseases. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00055-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
10
|
Garg PK, Putegnat B, Truong L, Reynolds C, Sanchez I, Nedrelow JK, Uffman J, Lokitz SJ, Nazih R, Garg S, Thornton PS. Visual interpretation, not SUV ratios, is the ideal method to interpret 18F-DOPA PET scans to aid in the cure of patients with focal congenital hyperinsulinism. PLoS One 2020; 15:e0241243. [PMID: 33108363 PMCID: PMC7591017 DOI: 10.1371/journal.pone.0241243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/11/2020] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Congenital hyperinsulinism is characterized by abnormal regulation of insulin secretion from the pancreas causing profound hypoketotic hypoglycemia and is the leading cause of persistent hypoglycemia in infants and children. The main objective of this study is to highlight the different mechanisms to interpret the 18F-DOPA PET scans and how this can influence outcomes. MATERIALS AND METHODS After 18F-Fluoro-L-DOPA was injected intravenously into 50 subjects' arm at a dose of 2.96-5.92 MBq/kg, three to four single-bed position PET scans were acquired at 20, 30, 40 and 50-minute post injection. The radiologist interpreted the scans for focal and diffuse hyperinsulinism using a visual interpretation method, as well as determining the Standard Uptake Value ratios with varying cut-offs. RESULTS Visual interpretation had the combination of the best sensitivity and positive prediction values. CONCLUSIONS In patients with focal disease, SUV ratios are not as accurate in identifying the focal lesion as visual inspection, and cases of focal disease may be missed by those relying on SUV ratios, thereby denying the patients a chance of cure. We recommend treating patients with diazoxide-resistant hyperinsulinism in centers with dedicated multidisciplinary team comprising of at least a pediatric endocrinologist with a special interest in hyperinsulinism, a radiologist experienced in interpretation of 18F-Fluoro-L-DOPA PET/CT scans, a histopathologist with experience in frozen section analysis of the pancreas and a pancreatic surgeon experienced in partial pancreatectomies in patients with hyperinsulinism.
Collapse
Affiliation(s)
- Pradeep K. Garg
- Center for Molecular Imaging and Therapy, Biomedical Research Foundation, Shreveport, Louisiana, United States of America
- * E-mail:
| | - Burton Putegnat
- Cook Children’s Medical Center, Fort Worth, Texas, United States of America
| | - Lisa Truong
- Cook Children’s Medical Center, Fort Worth, Texas, United States of America
| | - Courtney Reynolds
- Cook Children’s Medical Center, Fort Worth, Texas, United States of America
| | - Irene Sanchez
- Cook Children’s Medical Center, Fort Worth, Texas, United States of America
| | | | - John Uffman
- Cook Children’s Medical Center, Fort Worth, Texas, United States of America
| | - Stephen J. Lokitz
- Center for Molecular Imaging and Therapy, Biomedical Research Foundation, Shreveport, Louisiana, United States of America
| | - Rachid Nazih
- Center for Molecular Imaging and Therapy, Biomedical Research Foundation, Shreveport, Louisiana, United States of America
| | - Sudha Garg
- Center for Molecular Imaging and Therapy, Biomedical Research Foundation, Shreveport, Louisiana, United States of America
| | - Paul S. Thornton
- Cook Children’s Medical Center, Fort Worth, Texas, United States of America
| |
Collapse
|
11
|
Neo CWY, Ciaramicoli LM, Soetedjo AAP, Teo AKK, Kang NY. A new perspective of probe development for imaging pancreatic beta cell in vivo. Semin Cell Dev Biol 2020; 103:3-13. [PMID: 32057664 DOI: 10.1016/j.semcdb.2020.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 01/20/2020] [Accepted: 01/28/2020] [Indexed: 12/23/2022]
Abstract
Beta cells assume a fundamental role in maintaining blood glucose homeostasis through the secretion of insulin, which is contingent on both beta cell mass and function, in response to elevated blood glucose levels or secretagogues. For this reason, evaluating beta cell mass and function, as well as scrutinizing how they change over time in a diabetic state, are essential prerequisites in elucidating diabetes pathophysiology. Current clinical methods to measure human beta cell mass and/or function are largely lacking, indirect and sub-optimal, highlighting the continued need for noninvasive in vivo beta cell imaging technologies such as optical imaging techniques. While numerous probes have been developed and evaluated for their specificity to beta cells, most of them are more suited to visualize beta cell mass rather than function. In this review, we highlight the distinction between beta cell mass and function, and the importance of developing more probes to measure beta cell function. Additionally, we also explore various existing probes that can be employed to measure beta cell mass and function in vivo, as well as the caveats in probe development for in vivo beta cell imaging.
Collapse
Affiliation(s)
- Claire Wen Ying Neo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, 138673, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Larissa Miasiro Ciaramicoli
- Department of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Andreas Alvin Purnomo Soetedjo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, 138673, Singapore
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, 138673, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore.
| | - Nam-Young Kang
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, C5 Building, Room 203, Pohang, Kyungbuk, 37673, Republic of Korea.
| |
Collapse
|
12
|
Saunders DC, Brissova M, Phillips N, Shrestha S, Walker JT, Aramandla R, Poffenberger G, Flaherty DK, Weller KP, Pelletier J, Cooper T, Goff MT, Virostko J, Shostak A, Dean ED, Greiner DL, Shultz LD, Prasad N, Levy SE, Carnahan RH, Dai C, Sévigny J, Powers AC. Ectonucleoside Triphosphate Diphosphohydrolase-3 Antibody Targets Adult Human Pancreatic β Cells for In Vitro and In Vivo Analysis. Cell Metab 2019; 29:745-754.e4. [PMID: 30449685 PMCID: PMC6402969 DOI: 10.1016/j.cmet.2018.10.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/15/2018] [Accepted: 10/19/2018] [Indexed: 01/09/2023]
Abstract
Identification of cell-surface markers specific to human pancreatic β cells would allow in vivo analysis and imaging. Here we introduce a biomarker, ectonucleoside triphosphate diphosphohydrolase-3 (NTPDase3), that is expressed on the cell surface of essentially all adult human β cells, including those from individuals with type 1 or type 2 diabetes. NTPDase3 is expressed dynamically during postnatal human pancreas development, appearing first in acinar cells at birth, but several months later its expression declines in acinar cells while concurrently emerging in islet β cells. Given its specificity and membrane localization, we utilized an NTPDase3 antibody for purification of live human β cells as confirmed by transcriptional profiling, and, in addition, for in vivo imaging of transplanted human β cells. Thus, NTPDase3 is a cell-surface biomarker of adult human β cells, and the antibody directed to this protein should be a useful new reagent for β cell sorting, in vivo imaging, and targeting.
Collapse
Affiliation(s)
- Diane C Saunders
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37240, USA
| | - Marcela Brissova
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Neil Phillips
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Shristi Shrestha
- HudsonAlpha Institute of Biotechnology, Huntsville, AL 35806, USA
| | - John T Walker
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37240, USA
| | - Radhika Aramandla
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Greg Poffenberger
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - David K Flaherty
- Flow Cytometry Shared Resource, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kevin P Weller
- Flow Cytometry Shared Resource, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Julie Pelletier
- Centre de recherche du CHU de Québec - Université Laval, Québec City, QC G1V 4G2, Canada
| | - Tracy Cooper
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Matt T Goff
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John Virostko
- Department of Diagnostic Medicine, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA
| | - Alena Shostak
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - E Danielle Dean
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Dale L Greiner
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | - Nripesh Prasad
- HudsonAlpha Institute of Biotechnology, Huntsville, AL 35806, USA
| | - Shawn E Levy
- HudsonAlpha Institute of Biotechnology, Huntsville, AL 35806, USA
| | - Robert H Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Chunhua Dai
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jean Sévigny
- Centre de recherche du CHU de Québec - Université Laval, Québec City, QC G1V 4G2, Canada; Départment de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37240, USA; Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA; VA Tennessee Valley Healthcare, Nashville, TN 37212, USA.
| |
Collapse
|
13
|
Wei W, Ehlerding EB, Lan X, Luo QY, Cai W. Molecular imaging of β-cells: diabetes and beyond. Adv Drug Deliv Rev 2019; 139:16-31. [PMID: 31378283 DOI: 10.1016/j.addr.2018.06.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/27/2018] [Accepted: 06/26/2018] [Indexed: 02/09/2023]
Abstract
Since diabetes is becoming a global epidemic, there is a great need to develop early β-cell specific diagnostic techniques for this disorder. There are two types of diabetes (i.e., type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM)). In T1DM, the destruction of pancreatic β-cells leads to reduced insulin production or even absolute insulin deficiency, which consequently results in hyperglycemia. Actually, a central issue in the pathophysiology of all types of diabetes is the relative reduction of β-cell mass (BCM) and/or impairment of the function of individual β-cells. In the past two decades, scientists have been trying to develop imaging techniques for noninvasive measurement of the viability and mass of pancreatic β-cells. Despite intense scientific efforts, only two tracers for positron emission tomography (PET) and one contrast agent for magnetic resonance (MR) imaging are currently under clinical evaluation. β-cell specific imaging probes may also allow us to precisely and specifically visualize transplanted β-cells and to improve transplantation outcomes, as transplantation of pancreatic islets has shown promise in treating T1DM. In addition, some of these probes can be applied to the preoperative detection of hidden insulinomas as well. In the present review, we primarily summarize potential tracers under development for imaging β-cells with a focus on tracers for PET, SPECT, MRI, and optical imaging. We will discuss the advantages and limitations of the various imaging probes and extend an outlook on future developments in the field.
Collapse
|
14
|
Hart NJ, Weber C, Papas KK, Limesand SW, Vagner J, Lynch RM. Multivalent activation of GLP-1 and sulfonylurea receptors modulates β-cell second-messenger signaling and insulin secretion. Am J Physiol Cell Physiol 2018; 316:C48-C56. [PMID: 30404557 DOI: 10.1152/ajpcell.00209.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Linking two pharmacophores that bind different cell surface receptors into a single molecule can enhance cell-targeting specificity to cells that express the complementary receptor pair. In this report, we developed and tested a synthetic multivalent ligand consisting of glucagon-like peptide-1 (GLP-1) linked to glibenclamide (Glb) (GLP-1/Glb) for signaling efficacy in β-cells. Expression of receptors for these ligands, as a combination, is relatively specific to the β-cell in the pancreas. The multivalent GLP-1/Glb increased both intracellular cAMP and Ca2+, although Ca2+ responses were significantly depressed compared with the monomeric Glb. Moreover, GLP-1/Glb increased glucose-stimulated insulin secretion in a dose-dependent manner. However, unlike the combined monomers, GLP-1/Glb did not augment insulin secretion at nonstimulatory glucose concentrations in INS 832/13 β-cells or human islets of Langerhans. These data suggest that linking two binding elements, such as GLP-1 and Glb, into a single bivalent ligand can provide a unique functional agent targeted to β-cells.
Collapse
Affiliation(s)
| | - Craig Weber
- Department of Physiology, University of Arizona , Tucson, Arizona
| | | | - Sean W Limesand
- School of Animal and Comparative Biomedical Sciences, University of Arizona , Tucson, Arizona.,BIO5 Institute, University of Arizona , Tucson, Arizona
| | - Josef Vagner
- BIO5 Institute, University of Arizona , Tucson, Arizona
| | - Ronald M Lynch
- Department of Physiology, University of Arizona , Tucson, Arizona.,Department of Pharmacology, University of Arizona , Tucson, Arizona.,BIO5 Institute, University of Arizona , Tucson, Arizona
| |
Collapse
|
15
|
Kimura H, Ogawa Y, Fujimoto H, Mukai E, Kawashima H, Arimitsu K, Toyoda K, Fujita N, Yagi Y, Hamamatsu K, Murakami T, Murakami A, Ono M, Nakamoto Y, Togashi K, Inagaki N, Saji H. Evaluation of 18F-labeled exendin(9-39) derivatives targeting glucagon-like peptide-1 receptor for pancreatic β-cell imaging. Bioorg Med Chem 2017; 26:463-469. [PMID: 29273416 DOI: 10.1016/j.bmc.2017.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 11/30/2017] [Accepted: 12/03/2017] [Indexed: 01/09/2023]
Abstract
β-cell mass (BCM) is known to be decreased in subjects with type-2 diabetes (T2D). Quantitative analysis for BCM would be useful for understanding how T2D progresses and how BCM affects treatment efficacy and for earlier diagnosis of T2D and development of new therapeutic strategies. However, a noninvasive method to measure BCM has not yet been developed. We developed four 18F-labeled exendin(9-39) derivatives for β-cell imaging by PET: [18F]FB9-Ex(9-39), [18F]FB12-Ex(9-39), [18F]FB27-Ex(9-39), and [18F]FB40-Ex(9-39). Affinity to the glucagon-like peptide-1 receptor (GLP-1R) was evaluated with dispersed islet cells of ddY mice. Uptake of exendin(9-39) derivatives in the pancreas as well as in other organs was evaluated by a biodistribution study. Small-animal PET study was performed after injecting [18F]FB40-Ex(9-39). FB40-Ex(9-39) showed moderate affinity to the GLP-1R. Among all of the derivatives, [18F]FB40-Ex(9-39) resulted in the highest uptake of radioactivity in the pancreas 30 min after injection. Moreover, it showed significantly less radioactivity accumulated in the liver and kidney, resulting in an overall increase in the pancreas-to-organ ratio. In the PET imaging study, pancreas was visualized at 30 min after injection of [18F]FB40-Ex(9-39). [18F]FB40-Ex(9-39) met the basic requirements for an imaging probe for GLP-1R in pancreatic β-cells. Further enhancement of pancreatic uptake and specific binding to GLP-1R will lead to a clear visualization of pancreatic β-cells.
Collapse
Affiliation(s)
- Hiroyuki Kimura
- Department of Patho-Functional Bioanalysis, Kyoto University Graduate School of Pharmaceutical Sciences, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan; Department of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan.
| | - Yu Ogawa
- Department of Patho-Functional Bioanalysis, Kyoto University Graduate School of Pharmaceutical Sciences, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Fujimoto
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Eri Mukai
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hidekazu Kawashima
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Radioisotope Research Center, Kyoto Pharmaceutical University, 1 Misasagi-shichono-cho, Yamashina-ku, Kyoto 607-8412, Japan
| | - Kenji Arimitsu
- Department of Patho-Functional Bioanalysis, Kyoto University Graduate School of Pharmaceutical Sciences, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan; Department of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Kentaro Toyoda
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Naotaka Fujita
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yusuke Yagi
- Department of Patho-Functional Bioanalysis, Kyoto University Graduate School of Pharmaceutical Sciences, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan; Department of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Keita Hamamatsu
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takaaki Murakami
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Atsushi Murakami
- Research & Development Division, Arkray, Inc., Yousuien-nai, 59 Gansuin-cho, Kamigyo-ku, Kyoto 602-0008, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Kyoto University Graduate School of Pharmaceutical Sciences, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kaori Togashi
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hideo Saji
- Department of Patho-Functional Bioanalysis, Kyoto University Graduate School of Pharmaceutical Sciences, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
16
|
Suarez Castellanos I, Singh T, Balteanu B, Bhowmick DC, Jeremic A, Zderic V. Calcium-dependent ultrasound stimulation of secretory events from pancreatic beta cells. J Ther Ultrasound 2017; 5:30. [PMID: 29214024 PMCID: PMC5715497 DOI: 10.1186/s40349-017-0108-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 11/09/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Our previous studies have indicated that ultrasound can stimulate the release of insulin from pancreatic beta cells, providing a potential novel treatment for type 2 diabetes. The purpose of this study was to explore the temporal dynamics and Ca2+-dependency of ultrasound-stimulated secretory events from dopamine-loaded pancreatic beta cells in an in vitro setup. METHODS Carbon fiber amperometry was used to detect secretion from INS-1832/13 beta cells in real time. The levels of released insulin were also measured in response to ultrasound treatment using insulin-specific ELISA kit. Beta cells were exposed to continuous wave 800 kHz ultrasound at intensities of 0.1 W/cm2, 0.5 W/cm2 and 1 W/cm2 for several seconds. Cell viability tests were done with trypan blue dye exclusion test and MTT analysis. RESULTS Carbon fiber amperometry experiments showed that application of 800 kHz ultrasound at intensities of 0.5 and 1 W/cm2 was capable of stimulating secretory events for durations lasting as long as the duration of the stimulus. Furthermore, the amplitude of the detected peaks was reduced by 64% (p < 0.01) when extracellular Ca2+ was chelated with 10 mM EGTA in cells exposed to ultrasound intensity of 0.5 W/cm2. Measurements of released insulin in response to ultrasound stimulation showed complete inhibition of insulin secretion by chelating extracellular Ca2+ with 10 mM EGTA (p < 0.01). Viability studies showed that 800 kHz, 0.5 W/cm2 ultrasound did not cause any significant effects on viability and metabolic activity in cells exposed to ultrasound as compared to sham-treated cells. CONCLUSIONS Our results demonstrated that application of ultrasound was capable of stimulating the release of insulin from pancreatic beta cells in a safe, controlled and Ca2+-dependent manner.
Collapse
Affiliation(s)
- Ivan Suarez Castellanos
- Department of Biomedical Engineering, The George Washington University, 800 22nd St. NW rm 5290, Washington, District of Columbia 20052 USA
| | - Tania Singh
- Department of Biomedical Engineering, The George Washington University, 800 22nd St. NW rm 5290, Washington, District of Columbia 20052 USA
| | - Bogdan Balteanu
- Department of Biomedical Engineering, The George Washington University, 800 22nd St. NW rm 5290, Washington, District of Columbia 20052 USA
| | - Diti Chatterjee Bhowmick
- Department of Biological Sciences, The George Washington University, Washington, District of Columbia USA
| | - Aleksandar Jeremic
- Department of Biological Sciences, The George Washington University, Washington, District of Columbia USA
| | - Vesna Zderic
- Department of Biomedical Engineering, The George Washington University, 800 22nd St. NW rm 5290, Washington, District of Columbia 20052 USA
| |
Collapse
|
17
|
99mTc-labeled glimepiride as a tracer for targeting pancreatic β-cells mass: preparation and preclinical evaluation. J Radioanal Nucl Chem 2017. [DOI: 10.1007/s10967-017-5615-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
18
|
Zhang L, Thurber GM. Quantitative Impact of Plasma Clearance and Down-regulation on GLP-1 Receptor Molecular Imaging. Mol Imaging Biol 2016; 18:79-89. [PMID: 26194012 DOI: 10.1007/s11307-015-0880-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE Quantitative molecular imaging of beta cell mass (BCM) would enable early detection and treatment monitoring of type 1 diabetes. The glucagon-like peptide-1 (GLP-1) receptor is an attractive target due to its beta cell specificity and cell surface location. We quantitatively investigated the impact of plasma clearance and receptor internalization on targeting efficiency in healthy B6 mice. PROCEDURES Four exenatide-based probes were synthesized that varied in molecular weight, binding affinity, and plasma clearance. The GLP-1 receptor internalization rate and in vivo receptor expression were quantified. RESULTS Receptor internalization (54,000 receptors/cell in vivo) decreased significantly within minutes, reducing the benefit of a slower-clearing agent. The multimers and albumin binding probes had higher kidney and liver uptake, respectively. CONCLUSIONS Slow plasma clearance is beneficial for GLP-1 receptor peptide therapeutics. However, for exendin-based imaging of islets, down-regulation of the GLP-1 receptor and non-specific background uptake result in a higher target-to-background ratio for fast-clearing agents.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA. .,Department of Biomedical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI, 48109, USA.
| |
Collapse
|
19
|
Li J, Karunananthan J, Pelham B, Kandeel F. Imaging pancreatic islet cells by positron emission tomography. World J Radiol 2016; 8:764-774. [PMID: 27721939 PMCID: PMC5039672 DOI: 10.4329/wjr.v8.i9.764] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 04/15/2016] [Accepted: 08/08/2016] [Indexed: 02/06/2023] Open
Abstract
It was estimated that every year more than 30000 persons in the United States - approximately 80 people per day - are diagnosed with type 1 diabetes (T1D). T1D is caused by autoimmune destruction of the pancreatic islet (β cells) cells. Islet transplantation has become a promising therapy option for T1D patients, while the lack of suitable tools is difficult to directly evaluate of the viability of the grafted islet over time. Positron emission tomography (PET) as an important non-invasive methodology providing high sensitivity and good resolution, is able to accurate detection of the disturbed biochemical processes and physiological abnormality in living organism. The successful PET imaging of islets would be able to localize the specific site where transplanted islets engraft in the liver, and to quantify the level of islets remain alive and functional over time. This information would be vital to establishing and evaluating the efficiency of pancreatic islet transplantation. Many novel imaging agents have been developed to improve the sensitivity and specificity of PET islet imaging. In this article, we summarize the latest developments in carbon-11, fluorine-18, copper-64, and gallium-68 labeled radioligands for the PET imaging of pancreatic islet cells.
Collapse
|
20
|
Christoffersson G, von Herrath MG. A Deeper Look into Type 1 Diabetes - Imaging Immune Responses during Onset of Disease. Front Immunol 2016; 7:313. [PMID: 27574523 PMCID: PMC4983548 DOI: 10.3389/fimmu.2016.00313] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 08/02/2016] [Indexed: 12/28/2022] Open
Abstract
Cytotoxic T lymphocytes execute the killing of insulin-producing beta cells during onset of type 1 diabetes mellitus (T1D). The research community has come far in dissecting the major events in the development of this disease, but still the trigger and high-resolved information of the immunological events leading up to beta cell loss are missing. During the past decades, intravital imaging of immune responses has led to significant scientific breakthroughs in diverse models of disease, including T1D. Dynamic imaging of immune cells at the pancreatic islets during T1D onset has been made possible through the development of both advanced microscopes, and animal models that allow long-term immobilization of the pancreas. The use of these modalities has revealed a milling microenvironment at the pancreatic islets during disease onset with a plethora of active players. Clues to answering the remaining questions in this disease may lie in intravital imaging, including how key immune cells traffic to and from the pancreas, and how cells interact at this target tissue. This review highlights and discusses recent studies, models, and techniques focused to understand the immune responses during T1D onset through intravital imaging.
Collapse
Affiliation(s)
- Gustaf Christoffersson
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology , La Jolla, CA , USA
| | - Matthias G von Herrath
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA; Novo Nordisk Diabetes Research and Development Center, Seattle, WA, USA
| |
Collapse
|
21
|
Laurent D, Vinet L, Lamprianou S, Daval M, Filhoulaud G, Ktorza A, Wang H, Sewing S, Juretschke HP, Glombik H, Meda P, Boisgard R, Nguyen DL, Stasiuk GJ, Long NJ, Montet X, Hecht P, Kramer W, Rutter GA, Hecksher-Sørensen J. Pancreatic β-cell imaging in humans: fiction or option? Diabetes Obes Metab 2016; 18:6-15. [PMID: 26228188 DOI: 10.1111/dom.12544] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 07/02/2015] [Accepted: 07/28/2015] [Indexed: 01/02/2023]
Abstract
Diabetes mellitus is a growing worldwide epidemic disease, currently affecting 1 in 12 adults. Treatment of disease complications typically consumes ∼10% of healthcare budgets in developed societies. Whilst immune-mediated destruction of insulin-secreting pancreatic β cells is responsible for Type 1 diabetes, both the loss and dysfunction of these cells underly the more prevalent Type 2 diabetes. The establishment of robust drug development programmes aimed at β-cell restoration is still hampered by the absence of means to measure β-cell mass prospectively in vivo, an approach which would provide new opportunities for understanding disease mechanisms and ultimately assigning personalized treatments. In the present review, we describe the progress towards this goal achieved by the Innovative Medicines Initiative in Diabetes, a collaborative public-private consortium supported by the European Commission and by dedicated resources of pharmaceutical companies. We compare several of the available imaging methods and molecular targets and provide suggestions as to the likeliest to lead to tractable approaches. Furthermore, we discuss the simultaneous development of animal models that can be used to measure subtle changes in β-cell mass, a prerequisite for validating the clinical potential of the different imaging tracers.
Collapse
Affiliation(s)
- D Laurent
- Biomarker Department, Clinical Imaging, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - L Vinet
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - S Lamprianou
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - M Daval
- Metabolic Diseases Department, Servier Research Institute, Suresnes, France
| | - G Filhoulaud
- Metabolic Diseases Department, Servier Research Institute, Suresnes, France
| | - A Ktorza
- Metabolic Diseases Department, Servier Research Institute, Suresnes, France
| | - H Wang
- Roche Pharma Research and Early Development, Innovation Center Basel, Basel, Switzerland
| | - S Sewing
- Roche Pharma Research and Early Development, Innovation Center Basel, Basel, Switzerland
| | - H-P Juretschke
- Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany
| | - H Glombik
- Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany
| | - P Meda
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - R Boisgard
- Commissariat à l'Energie Atomique, Equipe d'Imagerie Moléculaire Expérimentale, Orsay, France
| | - D L Nguyen
- Commissariat à l'Energie Atomique, Equipe d'Imagerie Moléculaire Expérimentale, Orsay, France
| | - G J Stasiuk
- Department of Chemistry, Imperial College London, London, UK
| | - N J Long
- Department of Chemistry, Imperial College London, London, UK
| | - X Montet
- Department of Radiology, Geneva University Hospital, Geneva, Switzerland
| | - P Hecht
- IMIDIA Project Office, Graz, Austria
| | - W Kramer
- Scientific Consultant for Sanofi Deutschland GmbH, Frankfurt am Main, Germany
| | - G A Rutter
- Section of Cell Biology and Functional Genomics, Department of Medicine, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | | |
Collapse
|
22
|
Babič A, Lamprianou S, Vinet L, Stransky-Heilkron N, Xayaphoummine C, Campo MA, Glombik H, Schulte A, Juretschke HP, Montet X, Meda P, Lange N. Multivalent glibenclamide to generate islet specific imaging probes. Biomaterials 2015; 75:1-12. [PMID: 26474038 DOI: 10.1016/j.biomaterials.2015.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 09/29/2015] [Accepted: 10/01/2015] [Indexed: 12/16/2022]
Abstract
The monitoring of diabetes mellitus, as it develops and becomes clinically evident, remains a major challenge for diagnostic imaging in clinical practice. Here we present a novel approach to beta-cell imaging by targeting the sulphonylurea receptor subtype 1 (SUR1), using multivalent derivatives of the anti-diabetic drug glibenclamide. Since glibenclamide has a high affinity for SUR1 but does not contain a suitable functional group to be linked to an imaging probe, we have synthesized 11 glibenclamide derivatives and evaluated their affinity to SUR1 in MIN6 cells. The most promising compound has been used to obtain multivalent glibenclamide-polyamidoamine (PAMAM) derivatives, containing up to 15 sulphonylurea moieties per dendrimer. The remaining functional groups on the dendrimers can consecutively be used for labeling with reporter groups for different imaging modalities, thus allowing for multifunctional imaging, and for the modification of pharmacokinetic properties. We synthesized fluorochrome-labeled multivalent probes, that demonstrate in cellular assays affinities to SUR1 in the nanomolar range, superior to native glibenclamide. The probes specifically label MIN6 cells, but not HeLa or PANC-1 cells which do not express SUR1. A very low cytotoxicity of the multivalent probes is demonstrated by the persistent release of insulin from MIN6 cells exposed to high glucose concentrations. Furthermore, the probes display positive labeling of beta-cells of primary mouse and human islet-cells ex vivo and of islets of Langerhans in vivo. The data document that multivalent probes based on glibenclamide derivatives provide a suitable platform for further developments of cell-specific probes, and can be adapted for multiple imaging modalities, including those that are now used in the clinics.
Collapse
Affiliation(s)
- Andrej Babič
- School of Pharmaceutical Sciences Geneva-Lausanne, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Smaragda Lamprianou
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Geneva, Switzerland
| | - Laurent Vinet
- Department of Radiology, Geneva University Hospital, Geneva, Switzerland
| | - Nathalie Stransky-Heilkron
- School of Pharmaceutical Sciences Geneva-Lausanne, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Celine Xayaphoummine
- School of Pharmaceutical Sciences Geneva-Lausanne, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Marino A Campo
- School of Pharmaceutical Sciences Geneva-Lausanne, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Heiner Glombik
- Sanofi Germany, Industriepark Höchst, Frankfurt, Germany
| | - Anke Schulte
- Sanofi Germany, Industriepark Höchst, Frankfurt, Germany
| | | | - Xavier Montet
- Department of Radiology, Geneva University Hospital, Geneva, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Geneva, Switzerland
| | - Norbert Lange
- School of Pharmaceutical Sciences Geneva-Lausanne, University of Geneva, University of Lausanne, Geneva, Switzerland.
| |
Collapse
|
23
|
Signore A, Capriotti G, Chianelli M, Bonanno E, Galli F, Catalano C, Quintero AM, De Toma G, Manfrini S, Pozzilli P. Detection of insulitis by pancreatic scintigraphy with 99mTc-labeled IL-2 and MRI in patients with LADA (Action LADA 10). Diabetes Care 2015; 38:652-8. [PMID: 25665813 DOI: 10.2337/dc14-0580] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Pancreatic scintigraphy with interleukin-2 radiolabeled with (99m)Tc ((99m)Tc-IL-2) is a technique used to image chronic inflammatory-mediated disorders. We used this method to detect a signal consistent with the presence of insulitis in patients with autoimmune diabetes. Positive and negative controls (patients with pancreatic carcinoma and type 2 diabetes, respectively) also were studied. RESEARCH DESIGN AND METHODS We examined 25 patients with autoimmune diabetes (16 with recently diagnosed type 1 diabetes, 9 with latent autoimmune diabetes in adults [LADA]), 6 with type 2 diabetes, and 7 with pancreatic carcinoma (the latter two groups were used as negative and positive controls, respectively). All patients underwent (99m)Tc-IL-2 scintigraphy and contrast-enhanced MRI of the pancreas. To validate positive controls, samples were taken from patients with pancreatic carcinoma during surgery for histological and immunohistochemical investigations. RESULTS Pancreatic accumulation of (99m)Tc-IL-2 was detected in patients with autoimmune diabetes (61% positive) and, notably, in 6 of 9 patients with LADA; semiquantitative evaluation of pancreatic uptake of (99m)Tc-IL-2 showed higher values in patients with autoimmune diabetes (both childhood and LADA) and pancreatic carcinoma than in those with type 2 diabetes (4.45 ± 1.99, 4.79 ± 1.1, and 4.54 ± 1.62 vs. 2.81 ± 0.63; P = 0.06, P = 0.01, and P = 0.04, respectively). In patients with pancreatic carcinoma, pancreatic interleukin-2 receptor expression correlated with pancreatic (99m)Tc-IL-2 uptake (r = 0.8; P = 0.01). In patients with LADA, (99m)Tc-IL-2 uptake inversely correlated with duration of disease (r = 0.7; P = 0.03). CONCLUSIONS Autoimmune diabetes in adults is associated with increased pancreatic (99m)Tc-IL-2 uptake, indicating the presence of insulitis, particularly within 1 year of the beginning of insulin therapy, similar to type 1 diabetes at diagnosis.
Collapse
Affiliation(s)
- Alberto Signore
- Nuclear Medicine Unit, Faculty of Medicine and Psychology, University "Sapienza," Rome, Italy Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Gabriela Capriotti
- Nuclear Medicine Unit, Faculty of Medicine and Psychology, University "Sapienza," Rome, Italy
| | - Marco Chianelli
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands Regina Apostolorum Hospital, Albano, Rome, Italy
| | - Elena Bonanno
- Department of Biopathology and Diagnostic Imaging, University "Tor Vergata," Rome, Italy
| | - Filippo Galli
- Nuclear Medicine Unit, Faculty of Medicine and Psychology, University "Sapienza," Rome, Italy
| | - Carlo Catalano
- Department of Radiological Science, University "Sapienza," Rome, Italy
| | - Ana M Quintero
- Department of Radiology, Clínica Colsanitas, Bogotà, Colombia
| | - Giorgio De Toma
- Department of Surgery Science, University "Sapienza," Rome, Italy
| | - Silvia Manfrini
- Department of Endocrinology and Diabetes, University Campus Bio-Medico, Rome, Italy
| | - Paolo Pozzilli
- Department of Endocrinology and Diabetes, University Campus Bio-Medico, Rome, Italy Centre of Diabetes, St. Bartholomew's and The London School of Medicine, Queen Mary, University of London, London, U.K.
| | | |
Collapse
|
24
|
Steyn LV, Ananthakrishnan K, Anderson MJ, Patek R, Kelly A, Vagner J, Lynch RM, Limesand SW. A Synthetic Heterobivalent Ligand Composed of Glucagon-Like Peptide 1 and Yohimbine Specifically Targets β Cells Within the Pancreas. Mol Imaging Biol 2015; 17:461-70. [PMID: 25604385 DOI: 10.1007/s11307-014-0817-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 12/11/2014] [Accepted: 12/18/2014] [Indexed: 12/15/2022]
Abstract
PURPOSE β Cell specificity for a heterobivalent ligand composed of glucagon-like peptide-1 (GLP-1) linked to yohimbine (GLP-1/Yhb) was evaluated to determine its utility as a noninvasive imaging agent. PROCEDURES Competition binding assays were performed on βTC3 cells and isolated rat islets. Immunostaining for insulin was used to co-localized intravenously injected Cy5-labeled GLP-1/Yhb in β cells of Sprague-Dawley rats. Rats were intravenously injected with In-111-labeled GLP-1/Yhb to determine clearance rates and tissue biodistribution. Tissue-specific binding was confirmed by competition with pre-administration of unlabeled GLP-1/Yhb and in Streptozotocin-induced diabetic rats. RESULTS In βTC3 cells, high affinity binding of GLP-1/Yhb required interactions with both receptors because monovalent competition or receptor knockdown with RNAi lowered specificity and avidity of the heterobivalent ligand. Binding specificity for isolated islets was 2.6-fold greater than that of acinar tissue or islets pre-incubated with excess unlabeled GLP-1/Yhb. Immunofluorescent localization of Cy5-labeled GLP-1/Yhb was restricted to pancreatic islets. Within 30 min, ~90% of the In-111-labeled GLP-1/Yhb was cleared from blood. Tissue-specific accumulation of radiolabeled ligand was apparent in the pancreas, but not in other tissues within the abdominal imaging field. Pancreas specificity was lost in Streptozotocin-induced diabetic rats. CONCLUSIONS The GLP-1/Yhb exhibits high specificity for β cells, rapid blood clearance rates, and low non-specific uptake by other tissues within the abdominal imaging field. These characteristics of GLP-1/Yhb are desirable for application to β cell imaging in vivo and provide a basis for developing additional multivalent β cell-specific targeting agents to aid in the management of type 1 diabetes.
Collapse
Affiliation(s)
- Leah V Steyn
- School of Animal and Comparative Biomedical Sciences, William J. Parker Agricultural Research Center, The University of Arizona, 4101 N Campbell Ave, Tucson, AZ, 85719, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Rountree AM, Neal AS, Lisowski M, Rizzo N, Radtke J, White S, Luciani DS, Kim F, Hampe CS, Sweet IR. Control of insulin secretion by cytochrome C and calcium signaling in islets with impaired metabolism. J Biol Chem 2014; 289:19110-9. [PMID: 24841202 DOI: 10.1074/jbc.m114.556050] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The aim of the study was to assess the relative control of insulin secretion rate (ISR) by calcium influx and signaling from cytochrome c in islets where, as in diabetes, the metabolic pathways are impaired. This was achieved either by culturing isolated islets at low (3 mm) glucose or by fasting rats prior to the isolation of the islets. Culture in low glucose greatly reduced the glucose response of cytochrome c reduction and translocation and ISR, but did not affect the response to the mitochondrial fuel α-ketoisocaproate. Unexpectedly, glucose-stimulated calcium influx was only slightly reduced in low glucose-cultured islets and was not responsible for the impairment in glucose-stimulated ISR. A glucokinase activator acutely restored cytochrome c reduction and translocation and ISR, independent of effects on calcium influx. Islets from fasted rats had reduced ISR and cytochrome c reduction in response to both glucose and α-ketoisocaproate despite normal responses of calcium. Our data are consistent with the scenario where cytochrome c reduction and translocation are essential signals in the stimulation of ISR, the loss of which can result in impaired ISR even when calcium response is normal.
Collapse
Affiliation(s)
- Austin M Rountree
- From the Diabetes and Obesity Center, Department of Medicine, University of Washington, Seattle, Washington 98195 and
| | - Adam S Neal
- From the Diabetes and Obesity Center, Department of Medicine, University of Washington, Seattle, Washington 98195 and
| | - Mark Lisowski
- From the Diabetes and Obesity Center, Department of Medicine, University of Washington, Seattle, Washington 98195 and
| | - Norma Rizzo
- From the Diabetes and Obesity Center, Department of Medicine, University of Washington, Seattle, Washington 98195 and
| | - Jared Radtke
- From the Diabetes and Obesity Center, Department of Medicine, University of Washington, Seattle, Washington 98195 and
| | - Sarah White
- the Department of Surgery, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Dan S Luciani
- the Department of Surgery, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Francis Kim
- From the Diabetes and Obesity Center, Department of Medicine, University of Washington, Seattle, Washington 98195 and
| | - Christiane S Hampe
- From the Diabetes and Obesity Center, Department of Medicine, University of Washington, Seattle, Washington 98195 and
| | - Ian R Sweet
- From the Diabetes and Obesity Center, Department of Medicine, University of Washington, Seattle, Washington 98195 and
| |
Collapse
|
26
|
Brom M, Woliner-van der Weg W, Joosten L, Frielink C, Bouckenooghe T, Rijken P, Andralojc K, Göke BJ, de Jong M, Eizirik DL, Béhé M, Lahoutte T, Oyen WJG, Tack CJ, Janssen M, Boerman OC, Gotthardt M. Non-invasive quantification of the beta cell mass by SPECT with ¹¹¹In-labelled exendin. Diabetologia 2014; 57:950-9. [PMID: 24488022 DOI: 10.1007/s00125-014-3166-3] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 12/23/2013] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS A reliable method for in vivo quantification of pancreatic beta cell mass (BCM) could lead to further insight into the pathophysiology of diabetes. The glucagon-like peptide 1 receptor, abundantly expressed on beta cells, may be a suitable target for imaging. We investigated the potential of radiotracer imaging with the GLP-1 analogue exendin labelled with indium-111 for determination of BCM in vivo in a rodent model of beta cell loss and in patients with type 1 diabetes and healthy individuals. METHODS The targeting of (111)In-labelled exendin was examined in a rat model of alloxan-induced beta cell loss. Rats were injected with 15 MBq (111)In-labelled exendin and single photon emission computed tomography (SPECT) acquisition was performed 1 h post injection, followed by dissection, biodistribution and ex vivo autoradiography studies of pancreatic sections. BCM was determined by morphometric analysis after staining with an anti-insulin antibody. For clinical evaluation SPECT was acquired 4, 24 and 48 h after injection of 150 MBq (111)In-labelled exendin in five patients with type 1 diabetes and five healthy individuals. The tracer uptake was determined by quantitative analysis of the SPECT images. RESULTS In rats, (111)In-labelled exendin specifically targets the beta cells and pancreatic uptake is highly correlated with BCM. In humans, the pancreas was visible in SPECT images and the pancreatic uptake showed high interindividual variation with a substantially lower uptake in patients with type 1 diabetes. CONCLUSIONS/INTERPRETATION These studies indicate that (111)In-labelled exendin may be suitable for non-invasive quantification of BCM. TRIAL REGISTRATION ClinicalTrials.gov NCT01825148, EudraCT: 2012-000619-10.
Collapse
Affiliation(s)
- Maarten Brom
- Department of Radiology and Nuclear Medicine, Radboud university medical center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Pandey S, Venugopal A, Kant R, Coleman R, Mukherjee J. ¹²⁴I-Epidepride: a PET radiotracer for extended imaging of dopamine D2/D3 receptors. Nucl Med Biol 2014; 41:426-31. [PMID: 24602412 DOI: 10.1016/j.nucmedbio.2014.01.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 01/15/2014] [Accepted: 01/23/2014] [Indexed: 11/26/2022]
Abstract
OBJECTIVES A new radiotracer, ¹²⁴I-epidepride, has been developed for the imaging of dopamine D2/3 receptors (D2/3Rs). ¹²⁴I-Epidepride (half-life of ¹²⁴I=4.2 days) allows imaging over extended periods compared to (18)F-fallypride (half-life of ¹⁸F=0.076 days) and may maximize visualization of D2/3Rs in the brain and pancreas (allowing clearance from adjacent organs). D2/3 Rs are also present in pancreatic islets where they co-localize with insulin to produce granules and may serve as a surrogate marker for imaging diabetes. METHODS ¹²⁴I-Epidepride was synthesized using N-[[(2S)-1-ethylpyrrolidin-2-yl]methyl]-5-tributyltin-2,3-dimethoxybenzamide and ¹²⁴I-iodide under no carrier added condition. Rats were used for in vitro and in vivo imaging. Brain slices were incubated with (124)I-epidepride (0.75 μCi/cc) and nonspecific binding measured with 10 μM haloperidol. Autoradiograms were analyzed by OptiQuant. ¹²⁴I-Epidepride (0.2 to 0.3 mCi, iv) was administered to rats and brain uptake at 3 hours, 24 hours, and 48 hours post injection was evaluated. RESULTS ¹²⁴I-Epidepride was obtained with 50% radiochemical yield and high radiochemical purity (>95%). (124)I-Epidepride localized in the striatum with a striatum to cerebellum ratio of 10. Binding was displaced by dopamine and haloperidol. Brain slices demonstrated localization of ¹²⁴I-epidepride up until 48 hours in the striatum. However, the extent of binding was reduced significantly. CONCLUSIONS ¹²⁴I-Epidepride is a new radiotracer suitable for extended imaging of dopamine D2/3 receptors and may have applications in imaging of receptors in the brain and monitoring pancreatic islet cell grafting.
Collapse
Affiliation(s)
- Suresh Pandey
- Preclinical Imaging, Department of Radiological Sciences, University of California - Irvine, Irvine, CA 92697, USA
| | - Archana Venugopal
- Preclinical Imaging, Department of Radiological Sciences, University of California - Irvine, Irvine, CA 92697, USA
| | - Ritu Kant
- Preclinical Imaging, Department of Radiological Sciences, University of California - Irvine, Irvine, CA 92697, USA
| | - Robert Coleman
- Preclinical Imaging, Department of Radiological Sciences, University of California - Irvine, Irvine, CA 92697, USA
| | - Jogeshwar Mukherjee
- Preclinical Imaging, Department of Radiological Sciences, University of California - Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
28
|
McClure J, Margineantu DH, Sweet IR, Polyak SJ. Inhibition of HIV by Legalon-SIL is independent of its effect on cellular metabolism. Virology 2014; 449:96-103. [PMID: 24418542 PMCID: PMC3909448 DOI: 10.1016/j.virol.2013.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 09/22/2013] [Accepted: 11/01/2013] [Indexed: 01/18/2023]
Abstract
In this report, we further characterized the effects of silibinin (SbN), derived from milk thistle extract, and Legalon-SIL (SIL), a water-soluble derivative of SbN, on T cell metabolism and HIV infection. We assessed the effects of SbN and SIL on peripheral blood mononuclear cells (PBMC) and CEM-T4 cells in terms of cellular growth, ATP content, metabolism, and HIV infection. SIL and SbN caused a rapid and reversible (upon removal) decrease in cellular ATP levels, which was associated with suppression of mitochondrial respiration and glycolysis. SbN, but not SIL inhibited glucose uptake. Exposure of T cells to SIL (but not SbN or metabolic inhibitors) during virus adsorption blocked HIV infection. Thus, both SbN and SIL rapidly perturb T cell metabolism in vitro, which may account for its anti-inflammatory and anti-proliferative effects that arise with prolonged exposure of cells. However, the metabolic effects are not involved in SIL's unique ability to block HIV entry.
Collapse
Affiliation(s)
- Janela McClure
- Department of Laboratory Medicine, University of Washington, Seattle, WA, United States
| | - Daciana H Margineantu
- Department of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Ian R Sweet
- Department of Medicine (Division of Metabolism, Endocrinology, and Nutrition), University of Washington, Seattle, WA, United States
| | - Stephen J Polyak
- Department of Laboratory Medicine, University of Washington, Seattle, WA, United States; Department of Global Health, University of Washington, Seattle, WA, United States.
| |
Collapse
|
29
|
Hart NJ, Chung WJ, Weber C, Ananthakrishnan K, Anderson M, Patek R, Zhang Z, Limesand SW, Vagner J, Lynch RM. Hetero-bivalent GLP-1/glibenclamide for targeting pancreatic β-cells. Chembiochem 2013; 15:135-45. [PMID: 24259278 DOI: 10.1002/cbic.201300375] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Indexed: 01/15/2023]
Abstract
G protein-coupled receptor (GPCR) cell signalling cascades are initiated upon binding of a specific agonist ligand to its cell surface receptor. Linking multiple heterologous ligands that simultaneously bind and potentially link different receptors on the cell surface is a unique approach to modulate cell responses. Moreover, if the target receptors are selected based on analysis of cell-specific expression of a receptor combination, then the linked binding elements might provide enhanced specificity of targeting the cell type of interest, that is, only to cells that express the complementary receptors. Two receptors whose expression is relatively specific (in combination) to insulin-secreting pancreatic β-cells are the sulfonylurea-1 (SUR1) and the glucagon-like peptide-1 (GLP-1) receptors. A heterobivalent ligand was assembled from the active fragment of GLP-1 (7-36 GLP-1) and glibenclamide, a small organic ligand for SUR1. The synthetic construct was labelled with Cy5 or europium chelated in DTPA to evaluate binding to β-cells, by using fluorescence microscopy or time-resolved saturation and competition binding assays, respectively. Once the ligand binds to β-cells, it is rapidly capped and presumably removed from the cell surface by endocytosis. The bivalent ligand had an affinity approximately fivefold higher than monomeric europium-labelled GLP-1, likely a result of cooperative binding to the complementary receptors on the βTC3 cells. The high-affinity binding was lost in the presence of either unlabelled monomer, thus demonstrating that interaction with both receptors is required for the enhanced binding at low concentrations. Importantly, bivalent enhancement was accomplished in a cell system with physiological levels of expression of the complementary receptors, thus indicating that this approach might be applicable for β-cell targeting in vivo.
Collapse
Affiliation(s)
- Nathaniel J Hart
- Department of Physiological Sciences, University of Arizona, 1656 E. Mabel St., Tucson, AZ 85721 (USA)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Blomberg BA, Codreanu I, Cheng G, Werner TJ, Alavi A. Beta-cell imaging: call for evidence-based and scientific approach. Mol Imaging Biol 2013; 15:123-30. [PMID: 23413090 DOI: 10.1007/s11307-013-0620-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Advances in positron emission tomography (PET) imaging have provided opportunities to develop radiotracers specific for imaging insulin-producing pancreatic β-cells. However, a host of lingering questions should be addressed before these radiotracers are advocated for noninvasive quantification of β-cell mass (BCM) in vivo in the native pancreas. METHOD We provide an overview of tetrabenazine-based PET tracers developed to image and quantify BCM and discuss several theoretical, technical, and biological limitations of applying these tracers in clinical practice. DISCUSSION VMAT2, a transporter protein expressed on pancreatic β-cells, has been advocated as a promising target for PET imaging tracers, such as dihydrotetrabenazine. However, the lack of radiotracer specificity for these proteins hampers their clinical application. Another important argument against their use is a striking discrepancy between radiotracer uptake and BCM in subjects with type I diabetes mellitus and healthy controls. Additionally, technical issues, such as the finite spatial resolution of PET, partial volume effects, and movement of the pancreas during respiration, impede PET imaging as a viable option for BCM quantification in the foreseeable future. CONCLUSION The assertion that BCM can be accurately quantified by tetrabenazine derived β-cell-specific radiotracers as density per unit volume of pancreatic tissue is not justifiable at this time. The fallacy of these claims can be explained by technical as well as biological facts that have been disregarded and ignored in the literature.
Collapse
Affiliation(s)
- Björn A Blomberg
- Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | | | | | | |
Collapse
|
31
|
6-[18F]fluoro-L-DOPA uptake in the rat pancreas is dependent on the tracer metabolism. Mol Imaging Biol 2013; 16:403-11. [PMID: 24217945 DOI: 10.1007/s11307-013-0701-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 10/08/2013] [Accepted: 10/17/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE 6-[(18)F]fluoro-L-3,4-dihydroxyphenyl alanine ([(18)F]FDOPA) positron emission tomography (PET) is a diagnostic tool which can detect malignancies of the pancreas. We aimed to study whether the manipulation of the [(18)F]FDOPA metabolic pathway would change the (18)F-behavior to provide a biochemical foundation for PET imaging of rat pancreas with [(18)F]FDOPA. PROCEDURES Inhibitors of aromatic amino acid decarboxylase, catechol-O-methyltransferase, monoamine oxidases A and B, or their combinations on [(18)F]FDOPA uptake, metabolism, and the regional distribution in the rat pancreas was evaluated using in vivo PET/computed tomography imaging, chromatographic metabolite analyses, and autoradiography. RESULTS Enzyme inhibition generally increased the uptake of [(18)F]FDOPA derived (18)F-radioactivity in rat pancreas. Dependent on which enzymatic pathway is blocked (or a combination of pathways), different radiolabeled metabolites in pancreas are responsible for this increase in uptake. CONCLUSIONS Altering the metabolism of [(18)F]FDOPA by using various enzymatic inhibitors increased the radioactivity uptake and changed the radiometabolic profile in the pancreas allowing better discrimination between pancreas and surrounding tissues of rat. However, these manipulations did not separate islets from the exocrine pancreas. Elucidating the metabolic behavior of [(18)F]FDOPA provides a biochemical foundation of PET imaging of the rat pancreas.
Collapse
|
32
|
Positron emission tomography study on pancreatic somatostatin receptors in normal and diabetic rats with 68Ga-DOTA-octreotide: a potential PET tracer for beta cell mass measurement. Biochem Biophys Res Commun 2013; 442:79-84. [PMID: 24220338 DOI: 10.1016/j.bbrc.2013.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 11/02/2013] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus (DM) is a metabolic disorder characterized by hyperglycemia, and the loss or dysfunction of pancreatic beta cells has been reported before the appearance of clinical symptoms and hyperglycemia. To evaluate beta cell mass (BCM) for improving the detection and treatment of DM at earlier stages, we focused on somatostatin receptors that are highly expressed in the pancreatic beta cells, and developed a positron emission tomography (PET) probe derived from octreotide, a metabolically stable somatostatin analog. Octreotide was conjugated with 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA), a chelating agent, and labeled with (68)Gallium ((68)Ga). After intravenous injection of (68)Ga-DOTA-octreotide, a 90-min emission scan of the abdomen was performed in normal and DM model rats. The PET studies showed that (68)Ga-DOTA-octreotide radioactivity was highly accumulated in the pancreas of normal rats and that the pancreatic accumulation was significantly reduced in the rats administered with an excess amount of unlabeled octreotide or after treatment with streptozotocin, which was used for the chemical induction of DM in rats. These results were in good agreement with the ex vivo biodistribution data. These results indicated that the pancreatic accumulation of (68)Ga-DOTA-octreotide represented specific binding to the somatostatin receptors and reflected BCM. Therefore, PET imaging with (68)Ga-DOTA-octreotide could be a potential tool for evaluating BCM.
Collapse
|
33
|
Du J, Cleghorn WM, Contreras L, Lindsay K, Rountree AM, Chertov AO, Turner SJ, Sahaboglu A, Linton J, Sadilek M, Satrústegui J, Sweet IR, Paquet-Durand F, Hurley JB. Inhibition of mitochondrial pyruvate transport by zaprinast causes massive accumulation of aspartate at the expense of glutamate in the retina. J Biol Chem 2013; 288:36129-40. [PMID: 24187136 DOI: 10.1074/jbc.m113.507285] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transport of pyruvate into mitochondria by the mitochondrial pyruvate carrier is crucial for complete oxidation of glucose and for biosynthesis of amino acids and lipids. Zaprinast is a well known phosphodiesterase inhibitor and lead compound for sildenafil. We found Zaprinast alters the metabolomic profile of mitochondrial intermediates and amino acids in retina and brain. This metabolic effect of Zaprinast does not depend on inhibition of phosphodiesterase activity. By providing (13)C-labeled glucose and glutamine as fuels, we found that the metabolic profile of the Zaprinast effect is nearly identical to that of inhibitors of the mitochondrial pyruvate carrier. Both stimulate oxidation of glutamate and massive accumulation of aspartate. Moreover, Zaprinast inhibits pyruvate-driven O2 consumption in brain mitochondria and blocks mitochondrial pyruvate carrier in liver mitochondria. Inactivation of the aspartate glutamate carrier in retina does not attenuate the metabolic effect of Zaprinast. Our results show that Zaprinast is a potent inhibitor of mitochondrial pyruvate carrier activity, and this action causes aspartate to accumulate at the expense of glutamate. Our findings show that Zaprinast is a specific mitochondrial pyruvate carrier (MPC) inhibitor and may help to elucidate the roles of MPC in amino acid metabolism and hypoglycemia.
Collapse
|
34
|
Sakata N, Yoshimatsu G, Tsuchiya H, Aoki T, Mizuma M, Motoi F, Katayose Y, Kodama T, Egawa S, Unno M. Imaging of transplanted islets by positron emission tomography, magnetic resonance imaging, and ultrasonography. Islets 2013; 5:179-87. [PMID: 24231367 PMCID: PMC4010569 DOI: 10.4161/isl.26980] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
While islet transplantation is considered a useful therapeutic option for severe diabetes mellitus (DM), the outcome of this treatment remains unsatisfactory. This is largely due to the damage and loss of islets in the early transplant stage. Thus, it is important to monitor the condition of the transplanted islets, so that a treatment can be selected to rescue the islets from damage if needed. Recently, numerous trials have been performed to investigate the efficacy of different imaging modalities for visualizing transplanted islets. Positron emission tomography (PET) and magnetic resonance imaging (MRI) are the most commonly used imaging modalities for this purpose. Some groups, including ours, have also tried to visualize transplanted islets by ultrasonography (US). In this review article, we discuss the recent progress in islet imaging.
Collapse
Affiliation(s)
- Naoaki Sakata
- Division of Hepato-Biliary-Pancreatic Surgery; Department of Surgery; Tohoku University Graduate School of Medicine; Sendai, Japan
- Correspondence to: Naoaki Sakata,
| | - Gumpei Yoshimatsu
- Division of Hepato-Biliary-Pancreatic Surgery; Department of Surgery; Tohoku University Graduate School of Medicine; Sendai, Japan
| | - Haruyuki Tsuchiya
- Division of Hepato-Biliary-Pancreatic Surgery; Department of Surgery; Tohoku University Graduate School of Medicine; Sendai, Japan
| | - Takeshi Aoki
- Division of Hepato-Biliary-Pancreatic Surgery; Department of Surgery; Tohoku University Graduate School of Medicine; Sendai, Japan
| | - Masamichi Mizuma
- Division of Hepato-Biliary-Pancreatic Surgery; Department of Surgery; Tohoku University Graduate School of Medicine; Sendai, Japan
| | - Fuyuhiko Motoi
- Division of Hepato-Biliary-Pancreatic Surgery; Department of Surgery; Tohoku University Graduate School of Medicine; Sendai, Japan
| | - Yu Katayose
- Division of Hepato-Biliary-Pancreatic Surgery; Department of Surgery; Tohoku University Graduate School of Medicine; Sendai, Japan
- Division of Integrated Surgery and Oncology; Tohoku University Graduate School of Medicine; Sendai, Japan
| | - Tetsuya Kodama
- Department of Biomedical Engineering; Graduate School of Biomedical Engineering; Tohoku University; Sendai, Japan
| | - Shinichi Egawa
- Division of International Cooperation for Disaster Medicine; International Research Institute of Disaster Science; Tohoku University; Sendai, Japan
| | - Michiaki Unno
- Division of Hepato-Biliary-Pancreatic Surgery; Department of Surgery; Tohoku University Graduate School of Medicine; Sendai, Japan
| |
Collapse
|
35
|
Tuomela J, Forsback S, Haavisto L, Vahlberg T, Grönroos TJ, Solin O, Haaparanta-Solin M. Enzyme inhibition of dopamine metabolism alters 6-[18F]FDOPA uptake in orthotopic pancreatic adenocarcinoma. EJNMMI Res 2013; 3:18. [PMID: 23497589 PMCID: PMC3618317 DOI: 10.1186/2191-219x-3-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 02/24/2013] [Indexed: 02/04/2023] Open
Abstract
Background An unknown location hampers removal of pancreatic tumours. We studied the effects of enzyme inhibitors on the uptake of 6-[18F]fluoro-l-3,4-dihydroxyphenylalanine ([18F]FDOPA) in the pancreas, aiming at improved imaging of pancreatic adenocarcinoma. Methods Mice bearing orthotopic BxPC3 pancreatic adenocarcinoma were injected with 2-deoxy-2-[18F]fluoro-d-glucose ([18F]FDG) and scanned with positron emission tomography/computed tomography (PET/CT). For [18F]FDOPA studies, tumour-bearing mice and sham-operated controls were pretreated with enzyme inhibitors of aromatic amino acid decarboxylase (AADC), catechol-O-methyl transferase (COMT), monoamine oxidase A (MAO-A) or a combination of COMT and MAO-A. Mice were injected with [18F]FDOPA and scanned with PET/CT. The absolute [18F]FDOPA uptake was determined from selected tissues using a gamma counter. The intratumoural biodistribution of [18F]FDOPA was recorded by autoradiography. The main [18F]FDOPA metabolites present in the pancreata were determined with radio-high-performance liquid chromatography. Results [18F]FDG uptake was high in pancreatic tumours, while [18F]FDOPA uptake was highest in the healthy pancreas and significantly lower in tumours. [18F]FDOPA uptake in the pancreas was lowest with vehicle pretreatment and highest with pretreatment with the inhibitor of AADC. When mice received COMT + MAO-A inhibitors, the uptake was high in the healthy pancreas but low in the tumour-bearing pancreas. Conclusions Combined use of [18F]FDG and [18F]FDOPA is suitable for imaging pancreatic tumours. Unequal pancreatic uptake after the employed enzyme inhibitors is due to the blockade of metabolism and therefore increased availability of [18F]FDOPA metabolites, in which uptake differs from that of [18F]FDOPA. Pretreatment with COMT + MAO-A inhibitors improved the differentiation of pancreas from the surrounding tissue and healthy pancreas from tumour. Similar advantage was not achieved using AADC enzyme inhibitor, carbidopa.
Collapse
Affiliation(s)
- Johanna Tuomela
- MediCity/PET Preclinical Imaging, Turku PET Centre, University of Turku, Turku, 20520, Finland.
| | | | | | | | | | | | | |
Collapse
|
36
|
Di Gialleonardo V, de Vries EFJ, Di Girolamo M, Quintero AM, Dierckx RAJO, Signore A. Imaging of β-cell mass and insulitis in insulin-dependent (Type 1) diabetes mellitus. Endocr Rev 2012; 33:892-919. [PMID: 22889646 DOI: 10.1210/er.2011-1041] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Insulin-dependent (type 1) diabetes mellitus is a metabolic disease with a complex multifactorial etiology and a poorly understood pathogenesis. Genetic and environmental factors cause an autoimmune reaction against pancreatic β-cells, called insulitis, confirmed in pancreatic samples obtained at autopsy. The possibility to noninvasively quantify β-cell mass in vivo would provide important biological insights and facilitate aspects of diagnosis and therapy, including follow-up of islet cell transplantation. Moreover, the availability of a noninvasive tool to quantify the extent and severity of pancreatic insulitis could be useful for understanding the natural history of human insulin-dependent (type 1) diabetes mellitus, to early diagnose children at risk to develop overt diabetes, and to select patients to be treated with immunotherapies aimed at blocking the insulitis and monitoring the efficacy of these therapies. In this review, we outline the imaging techniques currently available for in vivo, noninvasive detection of β-cell mass and insulitis. These imaging techniques include magnetic resonance imaging, ultrasound, computed tomography, bioluminescence and fluorescence imaging, and the nuclear medicine techniques positron emission tomography and single-photon emission computed tomography. Several approaches and radiopharmaceuticals for imaging β-cells and lymphocytic insulitis are reviewed in detail.
Collapse
Affiliation(s)
- Valentina Di Gialleonardo
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9700 AB, Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|
37
|
Botsikas D, Terraz S, Vinet L, Lamprianou S, Becker C, Bosco D, Meda P, Montet X. Pancreatic magnetic resonance imaging after manganese injection distinguishes type 2 diabetic and normoglycemic patients. Islets 2012; 4:243-8. [PMID: 22722479 PMCID: PMC3442822 DOI: 10.4161/isl.20857] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A non-invasive method to image the mass and/or function of human pancreatic islets is needed to monitor the progression of diabetes, and the effect of therapeutic interventions. As yet, no method is available for this purpose, which could be applied to in situ human islets. Animal and in vitro studies have documented that manganese infusion could improve the magnetic resonance imaging (MRI) of the endocrine pancreas. Here, we have tested whether a similar approach could discriminate diabetic and non-diabetic patients. In vitro, human isolated islets readily incorporated manganese. In vivo, 243 manganese-enhanced magnetic resonance imaging (MEMRI) examinations were reviewed, including 41 examinations which were run on 24 patients with type 2 diabetes and 202 examinations which were run on 119 normoglycemic patients. The results show that MEMRI discriminates type 2 diabetics from non-diabetic patients, based on the signal enhancement of pancreas.
Collapse
Affiliation(s)
- Diomidis Botsikas
- Department of Radiology; Geneva University Hospital; Geneva, Switzerland
| | - Sylvain Terraz
- Department of Radiology; Geneva University Hospital; Geneva, Switzerland
| | - Laurent Vinet
- Department of Cell Physiology and Metabolism; Geneva University; Geneva, Switzerland
| | - Smaragda Lamprianou
- Department of Cell Physiology and Metabolism; Geneva University; Geneva, Switzerland
| | - Christoph Becker
- Department of Radiology; Geneva University Hospital; Geneva, Switzerland
| | - Domenico Bosco
- Department of Surgery; Geneva University Hospital; Geneva, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism; Geneva University; Geneva, Switzerland
| | - Xavier Montet
- Department of Radiology; Geneva University Hospital; Geneva, Switzerland
- Correspondence to: Xavier Montet,
| |
Collapse
|
38
|
Andralojc K, Srinivas M, Brom M, Joosten L, de Vries IJM, Eizirik DL, Boerman OC, Meda P, Gotthardt M. Obstacles on the way to the clinical visualisation of beta cells: looking for the Aeneas of molecular imaging to navigate between Scylla and Charybdis. Diabetologia 2012; 55:1247-57. [PMID: 22358499 PMCID: PMC3328679 DOI: 10.1007/s00125-012-2491-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 01/09/2012] [Indexed: 12/25/2022]
Abstract
For more than a decade, researchers have been trying to develop non-invasive imaging techniques for the in vivo measurement of viable pancreatic beta cells. However, in spite of intense research efforts, only one tracer for positron emission tomography (PET) imaging is currently under clinical evaluation. To many diabetologists it may remain unclear why the imaging world struggles to develop an effective method for non-invasive beta cell imaging (BCI), which could be useful for both research and clinical purposes. Here, we provide a concise overview of the obstacles and challenges encountered on the way to such BCI, in both native and transplanted islets. We discuss the major difficulties posed by the anatomical and cell biological features of pancreatic islets, as well as the chemical and physical limits of the main imaging modalities, with special focus on PET, SPECT and MRI. We conclude by indicating new avenues for future research in the field, based on several remarkable recent results.
Collapse
Affiliation(s)
- K. Andralojc
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - M. Srinivas
- Department of Tumour Immunology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - M. Brom
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - L. Joosten
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - I. J. M. de Vries
- Department of Tumour Immunology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - D. L. Eizirik
- Laboratory of Experimental Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - O. C. Boerman
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - P. Meda
- Deparment of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - M. Gotthardt
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| |
Collapse
|
39
|
Suckow AT, Zhang C, Egodage S, Comoletti D, Taylor P, Miller MT, Sweet IR, Chessler SD. Transcellular neuroligin-2 interactions enhance insulin secretion and are integral to pancreatic β cell function. J Biol Chem 2012; 287:19816-26. [PMID: 22528485 DOI: 10.1074/jbc.m111.280537] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Normal glucose-stimulated insulin secretion is dependent on interactions between neighboring β cells. Elucidation of the reasons why this cell-to-cell contact is essential will probably yield critical insights into β cell maturation and function. In the central nervous system, transcellular protein interactions (i.e. interactions between proteins on the surfaces of different cells) involving neuroligins are key mediators of synaptic functional development. We previously demonstrated that β cells express neuroligin-2 and that insulin secretion is affected by changes in neuroligin-2 expression. Here we show that the effect of neuroligin-2 on insulin secretion is mediated by transcellular interactions. Neuroligin-2 binds with nanomolar affinity to a partner on the β cell surface and contributes to the increased insulin secretion brought about by β cell-to-β cell contact. It does so in a manner seemingly independent of interactions with neurexin, a known binding partner. As in the synapse, transcellular neuroligin-2 interactions enhance the functioning of the submembrane exocytic machinery. Also, as in the synapse, neuroligin-2 clustering is important. Neuroligin-2 in soluble form, rather than presented on a cell surface, decreases insulin secretion by rat islets and MIN-6 cells, most likely by interfering with endogenous neuroligin interactions. Prolonged contact with neuroligin-2-expressing cells increases INS-1 β cell proliferation and insulin content. These results extend the known parallels between the synaptic and β cell secretory machineries to extracellular interactions. Neuroligin-2 interactions are one of the few transcellular protein interactions thus far identified that directly enhance insulin secretion. Together, these results indicate a significant role for transcellular neuroligin-2 interactions in the establishment of β cell function.
Collapse
Affiliation(s)
- Arthur T Suckow
- Department of Medicine and Pediatric Diabetes Research Center, UCSD School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Multimodal image coregistration and inducible selective cell ablation to evaluate imaging ligands. Proc Natl Acad Sci U S A 2011; 108:20719-24. [PMID: 22143775 DOI: 10.1073/pnas.1109480108] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We combined multimodal imaging (bioluminescence, X-ray computed tomography, and PET), tomographic reconstruction of bioluminescent sources, and two unique, complementary models to evaluate three previously synthesized PET radiotracers thought to target pancreatic beta cells. The three radiotracers {[(18)F]fluoropropyl-(+)-dihydrotetrabenazine ([(18)F]FP-DTBZ), [(18)F](+)-2-oxiranyl-3-isobutyl-9-(3-fluoropropoxy)-10-methoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]isoquinoline ((18)F-AV-266), and (2S,3R,11bR)-9-(3-fluoropropoxy)-2-(hydroxymethyl)-3-isobutyl-10-methoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]isoquinolin-2-ol ((18)F-AV-300)} bind vesicular monoamine transporter 2. Tomographic reconstruction of the bioluminescent signal in mice expressing luciferase only in pancreatic beta cells was used to delineate the pancreas and was coregistered with PET and X-ray computed tomography images. This strategy enabled unambiguous identification of the pancreas on PET images, permitting accurate quantification of the pancreatic PET signal. We show here that, after conditional, specific, and rapid mouse beta-cell ablation, beta-cell loss was detected by bioluminescence imaging but not by PET imaging, given that the pancreatic signal provided by three PET radiotracers was not altered. To determine whether these ligands bound human beta cells in vivo, we imaged mice transplanted with luciferase-expressing human islets. The human islets were imaged by bioluminescence but not with the PET ligands, indicating that these vesicular monoamine transporter 2-directed ligands did not specifically bind beta cells. These data demonstrate the utility of coregistered multimodal imaging as a platform for evaluation and validation of candidate ligands for imaging islets.
Collapse
|
41
|
Wang Y, Lim K, Normandin M, Zhao X, Cline GW, Ding YS. Synthesis and evaluation of [18F]exendin (9-39) as a potential biomarker to measure pancreatic β-cell mass. Nucl Med Biol 2011; 39:167-76. [PMID: 22033026 DOI: 10.1016/j.nucmedbio.2011.07.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2010] [Revised: 06/24/2011] [Accepted: 07/15/2011] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Glucagon-like peptide 1 (GLP-1) is released in response to food intake and plays an important role in maintaining blood glucose homeostasis. Exendin (9-39), a potent glucagon-like peptide 1 receptor antagonist, has been labeled with In-111 for SPECT imaging. We report here the first radiosynthesis of [(18)F]exendin (9-39) ([(18)F]Ex(9-39)) and an evaluation of its potential as a biomarker for in vivo positron emission tomography (PET) imaging of pancreatic β-cell mass (BCM) in rats. METHODS F-18 label was introduced by conjugation of [(18)F]4-fluorobenzaldehyde with an Ex(9-39) derivative containing a 6-hydrazinonicotinyl group on the ε-amine of Lys27. Positron emission tomography imaging was carried out in Sprague-Dawley rats (five control and five streptozotocin-induced diabetic) and BioBreeding diabetes-prone rats (three at 7 weeks and three at 12 weeks) using the high-resolution research tomograph (HRRT) after 0.187 ± 0.084 mCi [(18)F]Ex(9-39) administration. Time-activity curves were obtained from pancreas, liver and kidney. Pancreases were assayed for insulin content after the imaging study. RESULTS Site-specifically labeled [(18)F]Ex(9-39) was purified on a G15 open column with radiochemical and chemical purities >98%. Positron emission tomography imaging showed pancreatic standardized uptake value (SUV) peaked at 10 min and plateaued by 50 min to the end of scan (240 min). No correlations of pancreatic SUV with postmortem measures of insulin content were seen. CONCLUSIONS [(18)F]Ex(9-39) was successfully prepared and used for PET imaging for the first time to measure pancreatic BCM. The results suggest that derivatization of the Lys27 residue might reduce binding affinity, as evidenced by the absence of specific binding. Exendin analogues radiolabeled at other sites may elucidate the active site required for binding.
Collapse
Affiliation(s)
- Yi Wang
- Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | |
Collapse
|
42
|
Molecular imaging: a promising tool to monitor islet transplantation. J Transplant 2011; 2011:202915. [PMID: 22013504 PMCID: PMC3195545 DOI: 10.1155/2011/202915] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 07/29/2011] [Indexed: 12/18/2022] Open
Abstract
Replacement of insulin production by pancreatic islet transplantation has great potential as a therapy for type 1 diabetes mellitus. At present, the lack of an effective approach to islet grafts assessment limits the success of this treatment. The development of molecular imaging techniques has the potential to fulfill the goal of real-time noninvasive monitoring of the functional status and viability of the islet grafts. We review the application of a variety of imaging modalities for detecting endogenous and transplanted beta-cell mass. The review also explores the various molecular imaging strategies for assessing islet delivery, the metabolic effects on the islet grafts as well as detection of immunorejection. Here, we highlight the use of combined imaging and therapeutic interventions in islet transplantation and the in vivo monitoring of stem cells differentiation into insulin-producing cells.
Collapse
|
43
|
Eriksson O, Alavi A. Imaging the islet graft by positron emission tomography. Eur J Nucl Med Mol Imaging 2011; 39:533-42. [PMID: 21932118 DOI: 10.1007/s00259-011-1928-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 08/22/2011] [Indexed: 10/17/2022]
Abstract
Clinical islet transplantation is being investigated as a permanent cure for type 1 diabetes mellitus (T1DM). Currently, intraportal infusion of islets is the favoured procedure, but several novel implantation sites have been suggested. Noninvasive longitudinal methodologies are an increasingly important tool for assessing the fate of transplanted islets, their mass, function and early signs of rejection. This article reviews the approaches available for islet graft imaging by positron emission tomography and progress in the field, as well as future challenges and opportunities.
Collapse
Affiliation(s)
- Olof Eriksson
- Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.
| | | |
Collapse
|
44
|
Islet-selectivity of G-protein coupled receptor ligands evaluated for PET imaging of pancreatic β-cell mass. Biochem Biophys Res Commun 2011; 412:413-8. [DOI: 10.1016/j.bbrc.2011.07.077] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 07/20/2011] [Indexed: 11/18/2022]
|
45
|
Accurate measurement of pancreatic islet beta-cell mass using a second-generation fluorescent exendin-4 analog. Proc Natl Acad Sci U S A 2011; 108:12815-20. [PMID: 21768367 DOI: 10.1073/pnas.1109859108] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The hallmark of type 1 diabetes is autoimmune destruction of the insulin-producing β-cells of the pancreatic islets. Autoimmune diabetes has been difficult to study or treat because it is not usually diagnosed until substantial β-cell loss has already occurred. Imaging agents that permit noninvasive visualization of changes in β-cell mass remain a high-priority goal. We report on the development and testing of a near-infrared fluorescent β-cell imaging agent. Based on the amino acid sequence of exendin-4, we created a neopeptide via introduction of an unnatural amino acid at the K(12) position, which could subsequently be conjugated to fluorophores via bioorthogonal copper-catalyzed click-chemistry. Cell assays confirmed that the resulting fluorescent probe (E4(×12)-VT750) had a high binding affinity (~3 nM). Its in vivo properties were evaluated using high-resolution intravital imaging, histology, whole-pancreas visualization, and endoscopic imaging. According to intravital microscopy, the probe rapidly bound to β-cells and, as demonstrated by confocal microscopy, it was internalized. Histology of the whole pancreas showed a close correspondence between fluorescence and insulin staining, and there was an excellent correlation between imaging signals and β-cell mass in mice treated with streptozotocin, a β-cell toxin. Individual islets could also be visualized by endoscopic imaging. In short, E4(×12)-VT750 showed strong and selective binding to glucose-like peptide-1 receptors and permitted accurate measurement of β-cell mass in both diabetic and nondiabetic mice. This near-infrared imaging probe, as well as future radioisotope-labeled versions of it, should prove to be important tools for monitoring diabetes, progression, and treatment in both experimental and clinical contexts.
Collapse
|
46
|
Garcia A, Mirbolooki MR, Constantinescu C, Pan ML, Sevrioukov E, Milne N, Wang PH, Lakey J, Chandy KG, Mukherjee J. 18F-Fallypride PET of pancreatic islets: in vitro and in vivo rodent studies. J Nucl Med 2011; 52:1125-32. [PMID: 21680697 DOI: 10.2967/jnumed.111.088583] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Islet cell loss in the pancreas results in diabetes. A noninvasive method that measures islet cell loss and also tracks the fate of transplanted islets would facilitate the development of novel therapeutics and improve the management of diabetes. We describe a novel dopamine D(2)/D(3) receptor (D(2)/D(3)R)-based PET method to study islet cells in the rat pancreas and in islet cell transplantation. METHODS (18)F-fallypride binding to isolated rat islets and pancreas was evaluated in the absence and presence of the D(2)/D(3)R inhibitor haloperidol. After intravenous (18)F-fallypride (28-37 MBq) administration, normal rats and rats pretreated with haloperidol were imaged in a PET/CT scanner and subsequently studied ex vivo for (18)F-fallypride localization in the pancreas. A streptozotocin-treated diabetic rat model was used to study localization of (18)F-fallypride in the pancreas, in vitro and ex vivo. Rat islet cells were transplanted into the spleen and visualized using (18)F-fallypride PET. RESULTS (18)F-fallypride bound to isolated islet cells and pancreatic sections with an endocrine or exocrine selectivity of approximately 4; selectivity was reduced by haloperidol, suggesting that binding was D(2)/D(3)R-specific. Chemical destruction of islets by streptozotocin decreased (18)F-fallypride binding in pancreas by greater than 50%, paralleling the decrease in insulin immunostaining. Uptake of (18)F-fallypride in the pancreas was confirmed by radiochromatography and was 0.05% injected dose/cm(3) as measured by PET/CT. The ratio of (18)F-fallypride uptake in the pancreas to reference tissue (erector spinae muscle) was 5.5. Rat islets transplanted into the spleen were visualized in vivo by (18)F-fallypride and confirmed by immunostaining. The ratio of spleen-transplanted islets to erector spinae muscle was greater than 5, compared with a ratio of 2.8 in untransplanted rats. CONCLUSION These studies demonstrate the potential utility of (18)F-fallypride as a PET agent for islet cells.
Collapse
Affiliation(s)
- Adriana Garcia
- Preclinical Imaging Center, Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, California 92697, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Jung SR, Kuok ITD, Couron D, Rizzo N, Margineantu DH, Hockenbery DM, Kim F, Sweet IR. Reduced cytochrome C is an essential regulator of sustained insulin secretion by pancreatic islets. J Biol Chem 2011; 286:17422-34. [PMID: 21393241 DOI: 10.1074/jbc.m110.202820] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Influx of calcium is an essential but insufficient signal in sustained nutrient-stimulated insulin secretion, and increased metabolic rate of the beta cell is also required. The aim of the study was to test the hypothesis that the reduced state of cytochrome c is a metabolic co-factor necessary for insulin secretion, over and above its participation in the ATP-generating function of electron transport/oxidative phosphorylation. We found that nutrient stimulation of insulin secretion by isolated rat islets was strongly correlated with reduced cytochrome c, and agents that acutely and specifically reduced cytochrome c led to increased insulin secretion, even in the face of decreased oxygen consumption and calcium influx. In contrast, neither sites 1 nor 4 of the electron transport chain were both necessary and essential for the stimulation of insulin secretion to occur. Importantly, stimulation of islets with glucose, α-ketoisocaproate, or glyceraldehyde resulted in the appearance of cytochrome c in the cytosol, suggesting a pathway for the regulation of exocytotic machinery by reduction of cytochrome c. The data suggest that the metabolic factor essential for sustained calcium-stimulated insulin secretion to occur is linked to reduction and translocation of cytochrome c.
Collapse
Affiliation(s)
- Seung-Ryoung Jung
- Department of Medicine, Diabetes and Obesity Center, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Reiner T, Kohler RH, Liew CW, Hill JA, Gaglia J, Kulkarni RN, Weissleder R. Near-infrared fluorescent probe for imaging of pancreatic beta cells. Bioconjug Chem 2010; 21:1362-8. [PMID: 20583828 DOI: 10.1021/bc100184w] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The ability to image and ultimately quantitate beta-cell mass in vivo will likely have far reaching implications in the study of diabetes biology, in the monitoring of disease progression or response to treatment, and for drug development. Here, using animal models, we report on the synthesis, characterization, and intravital microscopic imaging properties of a near-infrared fluorescent exendin-4 analogue with specificity for the GLP-1 receptor on beta cells (E4(K12)-Fl). The agent demonstrated subnanomolar EC(50) binding concentrations, with high specificity and binding that could be inhibited by GLP-1R agonists. Following intravenous administration to mice, pancreatic islets were readily distinguishable from exocrine pancreas, achieving target-to-background ratios within the pancreas of 6:1, as measured by intravital microscopy. Serial imaging revealed rapid accumulation kinetics (with initial signal within the islets detectable within 3 min and peak fluorescence within 20 min of injection), making this an ideal agent for in vivo imaging.
Collapse
Affiliation(s)
- Thomas Reiner
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Wu Z, Kandeel F. Radionuclide probes for molecular imaging of pancreatic beta-cells. Adv Drug Deliv Rev 2010; 62:1125-38. [PMID: 20854861 DOI: 10.1016/j.addr.2010.09.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 09/09/2010] [Accepted: 09/13/2010] [Indexed: 12/16/2022]
Abstract
Islet transplantation is a promising treatment option for patients with type 1 diabetes (T1D); however, the fate of the graft over time remains difficult to follow, due to the lack of available tools capable of monitoring graft rejection and inflammation prior to islet graft loss. Due to the challenges imposed by the location of the pancreas and the sparsely dispersed beta-cell population within the pancreas, currently, the clinical verification of beta-cell abnormalities can only be obtained indirectly via metabolic studies, which typically is not possible until after a significant deterioration in islet function has already occurred. The development of non-invasive imaging methods for the assessment of the pancreatic beta-cells, however, offers the potential for the early detection of beta-cell dysfunction prior to the clinical onset of T1D and type 2 diabetes (T2D). Ideal islet imaging agents would have an acceptable residence time in the human body, be capable of providing high-resolution images with minimal uptake in surrounding tissues (e.g., the liver), would not be toxic to islets, and would not require pre-treatment of islets prior to transplantation. A variety of currently available imaging techniques, including magnetic resonance imaging (MRI), bioluminescence imaging (BLI), and nuclear imaging have been tested for the study of beta-cell diseases. In this article, we summarize the recent advances made in nuclear imaging techniques for non-invasive imaging of pancreatic beta-cells. The use of radioactive probes for islet imaging is also discussed.
Collapse
|
50
|
Ueberberg S, Ziegler D, Schechinger W, Dietrich JW, Akinturk S, Klein HH, Schneider S. In vitro phage display in a rat beta cell line: a simple approach for the generation of a single-chain antibody targeting a novel beta cell-specific epitope. Diabetologia 2010; 53:1384-94. [PMID: 20369222 DOI: 10.1007/s00125-010-1725-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 02/15/2010] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS The aim of the present study was to evaluate in vitro phage display in a beta cell line as a novel strategy for the isolation of beta cell-specific agents/biomarkers. METHODS A single-chain antibody (SCA) library was pre-incubated with AR42J cells in order to eliminate SCAs with exocrine binding properties. It was then panned against INS-1 cells to select beta cell-targeted antibodies. RESULTS By these means, we isolated a novel antibody, SCA B5, that binds rapidly (6.0 min) and with a 450-fold higher specificity to beta cells relative to exocrine cells. We estimated for SCA B5 a binding affinity in the low micromol/l range and 858 binding sites per beta cell. Confocal microscopy showed binding to the beta cell surface and confirmed subsequent internalisation. Moreover, staining of rat and human pancreatic tissue sections with SCA B5 suggests that the target epitope is presented in pancreatic beta cells of different origins. Infrared imaging revealed that labelling of beta cells with tracer SCA B5 is strictly dependent on beta cell mass. With competition assays we excluded insulin, glutamate decarboxylase, C-peptide and islet amyloid polypeptide as SCA B5 targets. In accordance with these predictions, SCA B5 homed in vivo highly selectively to normal beta cells and dysfunctional beta cells of diabetic rats. Moreover, accumulation of radioactively labelled SCA B5 in the pancreas was reduced by 80% after pre-injection with unlabelled SCA B5, thereby confirming the specific uptake in the pancreas. CONCLUSIONS/INTERPRETATION We report a simple strategy for the generation of an SCA targeting a novel beta cell-specific epitope.
Collapse
Affiliation(s)
- S Ueberberg
- Department of Internal Medicine I, Division of Endocrinology and Metabolism, University Hospital Bergmannsheil, Ruhr-University Bochum, Bürkle de la Camp Platz 1, 44789, Bochum, Germany
| | | | | | | | | | | | | |
Collapse
|