1
|
Lithovius V, Lahdenpohja S, Ibrahim H, Saarimäki-Vire J, Uusitalo L, Montaser H, Mikkola K, Yim CB, Keller T, Rajander J, Balboa D, Barsby T, Solin O, Nuutila P, Grönroos TJ, Otonkoski T. Non-invasive quantification of stem cell-derived islet graft size and composition. Diabetologia 2024; 67:1912-1929. [PMID: 38871836 PMCID: PMC11410899 DOI: 10.1007/s00125-024-06194-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/16/2024] [Indexed: 06/15/2024]
Abstract
AIMS/HYPOTHESIS Stem cell-derived islets (SC-islets) are being used as cell replacement therapy for insulin-dependent diabetes. Non-invasive long-term monitoring methods for SC-islet grafts, which are needed to detect misguided differentiation in vivo and to optimise their therapeutic effectiveness, are lacking. Positron emission tomography (PET) has been used to monitor transplanted primary islets. We therefore aimed to apply PET as a non-invasive monitoring method for SC-islet grafts. METHODS We implanted different doses of human SC-islets, SC-islets derived using an older protocol or a state-of-the-art protocol and SC-islets genetically rendered hyper- or hypoactive into mouse calf muscle to yield different kinds of grafts. We followed the grafts with PET using two tracers, glucagon-like peptide 1 receptor-binding [18F]F-dibenzocyclooctyne-exendin-4 ([18F]exendin) and the dopamine precursor 6-[18F]fluoro-L-3,4-dihydroxyphenylalanine ([18F]FDOPA), for 5 months, followed by histological assessment of graft size and composition. Additionally, we implanted a kidney subcapsular cohort with different SC-islet doses to assess the connection between C-peptide and stem cell-derived beta cell (SC-beta cell) mass. RESULTS Small but pure and large but impure grafts were derived from SC-islets. PET imaging allowed detection of SC-islet grafts even <1 mm3 in size, [18F]exendin having a better detection rate than [18F]FDOPA (69% vs 44%, <1 mm3; 96% vs 85%, >1 mm3). Graft volume quantified with [18F]exendin (r2=0.91) and [18F]FDOPA (r2=0.86) strongly correlated with actual graft volume. [18F]exendin PET delineated large cystic structures and its uptake correlated with graft SC-beta cell proportion (r2=0.68). The performance of neither tracer was affected by SC-islet graft hyper- or hypoactivity. C-peptide measurements under fasted or glucose-stimulated conditions did not correlate with SC-islet graft volume or SC-beta cell mass, with C-peptide under hypoglycaemia having a weak correlation with SC-beta cell mass (r2=0.52). CONCLUSIONS/INTERPRETATION [18F]exendin and [18F]FDOPA PET enable non-invasive assessment of SC-islet graft size and aspects of graft composition. These methods could be leveraged for optimising SC-islet cell replacement therapy in diabetes.
Collapse
Affiliation(s)
- Väinö Lithovius
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | | | - Hazem Ibrahim
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jonna Saarimäki-Vire
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Hossam Montaser
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kirsi Mikkola
- Turku PET Centre, University of Turku, Turku, Finland
- Medicity Research Laboratories, University of Turku, Turku, Finland
| | - Cheng-Bin Yim
- Turku PET Centre, University of Turku, Turku, Finland
| | - Thomas Keller
- Turku PET Centre, University of Turku, Turku, Finland
| | - Johan Rajander
- Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Turku, Finland
| | - Diego Balboa
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tom Barsby
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olof Solin
- Turku PET Centre, University of Turku, Turku, Finland
- Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Turku, Finland
- Department of Chemistry, University of Turku, Turku, Finland
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
- The Wellbeing Services County of Southwest Finland, Turku, Finland
| | - Tove J Grönroos
- Turku PET Centre, University of Turku, Turku, Finland
- Medicity Research Laboratories, University of Turku, Turku, Finland
| | - Timo Otonkoski
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Children's Hospital, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
2
|
Sun Y, Li G, Hong H, Zhu L, Kung HF, Zhang Y, Zhu J. Serotonin transporter imaging agent as a probe for β-cells of pancreas. Nucl Med Biol 2024; 130-131:108894. [PMID: 38422917 DOI: 10.1016/j.nucmedbio.2024.108894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVE Diabetes mellitus (DM) is one of the major diseases in the world. Nuclear medicine imaging may be able to detect functional status of pancreatic β cells in vivo, which might elucidate the pathological mechanisms of diabetes and develop individualized treatment plans. In this study, we evaluated the ability of [125I]ADAM, a serotonin transporter (SERT) imaging agent, as a probe for detecting pancreatic β-cell mass (BCM). METHODS In vitro cell studies were evaluated in INS-1 cells (rat islet β cell line). Biodistribution studies were performed in male normal Sprague-Dawley rats and alloxan-induced type 1 diabetes mellitus (T1DM) rats. Distribution and expression of SERT protein in pancreas of rats were also measured by immunofluorescence staining and Western blot. RESULTS In vitro cell studies showed that the concentration of [125I]ADAM associated with the INS-1 cells was increased gradually with incubation time, and the SERT specific inhibitor, escitalopram, exhibited the inhibitory effect on this interaction. Biodistribution studies also showed that the uptake of [125I]ADAM in the pancreas of normal rats was decreased in the presence of escitalopram. However, in the T1DM rat model with a significant β cells reduction, the uptake of pancreas was increased when compared with the control. Through immunofluorescence staining and Western blot, it was found that both the endocrine and exocrine cells of the normal pancreas expressed SERT protein, and the level of SERT protein in the exocrine cells was higher than islets. In the diabetic state, the expression of SERT in the exocrine cells was further increased. CONCLUSIONS The SERT imaging agent, [125I]ADAM, at the present form will not be suitable for imaging β cells, specifically because there were extraordinarily high non-specific signals contributing from the exocrine cells of pancreas. In addition, we noticed that the level of SERT expression was abnormally elevated in the diabetic state, which might provide an unexpected target for studying the pathological mechanisms of diabetes.
Collapse
Affiliation(s)
- Yuli Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University, Beijing 100069, China
| | - Guangwen Li
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University, Beijing 100069, China
| | - Haiyan Hong
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Lin Zhu
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Hank F Kung
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yan Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University, Beijing 100069, China.
| | - Jinxia Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
3
|
El-Kawy OA, Ibrahim IT, Shewatah HA, Attalah KM. Preparation and evaluation of radiolabeled gliclazide parenteral nanoemulsion as a new tracer for pancreatic β-cells mass. Int J Radiat Biol 2023; 99:1738-1748. [PMID: 37071445 DOI: 10.1080/09553002.2023.2204914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/20/2023] [Indexed: 04/19/2023]
Abstract
PURPOSE The present investigation aims to develop and evaluate a radiopharmaceutical for targeting and assessing β-cells mass based on gliclazide, an antidiabetic drug that specifically binds the sulfonylurea receptor unique to the β-cells of the pancreas. METHODS Conditions were optimized to radiolabel gliclazide with radioiodine via electrophilic substitution reaction. Then, it was formulated as a nanoemulsion system using olive oil and egg lecithin by hot homogenization followed by ultrasonication. The system was assessed for its suitability for parenteral administration and drug release. Then, the tracer was evaluated in silico and in vivo in normal and diabetic rats. RESULTS AND CONCLUSIONS The labeled compound was obtained with a high radiochemical yield (99.3 ± 1.1%) and good stability (>48 h). The radiolabeled nanoemulsion showed an average droplet size of 24.7 nm, a polydispersity index of 0.21, a zeta potential of -45.3 mV, pH 7.4, an osmolality of 285.3 mOsm/kg, and viscosity of 1.24 mPa.s, indicating suitability for parenteral administration. In silico assessment suggested that the labeling did not affect the biological activity of gliclazide. The suggestion was further supported by the in vivo blocking study. Following intravenous administration of nanoemulsion, the pancreas uptake was highest in normal rats (19.57 ± 1.16 and 12 ± 0.13% ID) compared to diabetic rats (8.51 ± 0.16 and 5 ± 0.13% ID) at 1 and 4 h post-injection, respectively. All results supported the feasibility of radioiodinated gliclazide nanoemulsion as a tracer for pancreatic β-cells.
Collapse
Affiliation(s)
- O A El-Kawy
- Egyptian Atomic Energy Authority, Cairo, Egypt
| | - I T Ibrahim
- Egyptian Atomic Energy Authority, Cairo, Egypt
| | | | - K M Attalah
- Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
4
|
Bini J, Carson RE, Cline GW. Noninvasive Quantitative PET Imaging in Humans of the Pancreatic Beta-Cell Mass Biomarkers VMAT2 and Dopamine D2/D3 Receptors In Vivo. Methods Mol Biol 2023; 2592:61-74. [PMID: 36507985 DOI: 10.1007/978-1-0716-2807-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Noninvasive quantitative imaging of beta-cells can provide information on changes in cellular transporters, receptors, and signaling proteins that may affect function and/or loss of mass, both of which contribute to the loss of insulin secretion and glucose regulation of patients with type 1 or type 2 diabetes (T1D/T2D). We have developed and optimized the use of two positron emission tomography (PET) radioligands, [18F]FP-(+)-DTBZ and [11C](+)-PHNO, targeting beta-cell VMAT2 and dopamine (D2/D3) receptors, respectively. Here we describe our optimized methodology for the clinical use of these two tracers for quantitative PET imaging of beta-cell biomarkers in vivo. We also briefly discuss our previous results and their implications and value towards extending the use of PET radioligand beyond the original goal of quantitative imaging of beta-cell mass to the potential to provide insight into the biology of beta-cell loss of mass and/or function and to evaluate the efficacy of therapeutics to prevent or restore functional beta-cell mass.
Collapse
Affiliation(s)
- Jason Bini
- PET Center, Yale University School of Medicine, New Haven, CT, USA.
| | - Richard E Carson
- PET Center, Yale University School of Medicine, New Haven, CT, USA
| | - Gary W Cline
- Department of Internal Medicine, Division of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
5
|
Zhang Y, Li G, Sun Y, Hong H, Li L, Luo Y, Wang R, Zhu L, Kung HF, Zhu J. In vivo and in vitro binding of [ 125 I]I-R-(+)-TISCH: A dopamine D 1 receptor ligand for studying pancreatic β-cell mass. J Labelled Comp Radiopharm 2022; 65:354-360. [PMID: 36261868 DOI: 10.1002/jlcr.4005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/16/2022] [Accepted: 10/17/2022] [Indexed: 12/31/2022]
Abstract
Diabetes mellitus (DM) and insulinoma are mainly affected by the status of pancreatic β-cell mass (BCM). Development of imaging agents for BCM allows to study pancreatic β cells and the relationship between β cells and DM or insulinoma. In this study, we investigated the density of dopamine D1 receptor on the β cells and measured BCM by statistical image processing. The pancreatic uptakes of [125 I]I-R-(+)-7-chloro-8-hydroxy-1-(3'-iodopheny1)-3-methyl-2,3,4,5-tetrahydro-1H-3-benzazepine ([125 I]I-R-(+)-TISCH), dopamine D1 receptor tracer, in normal and diabetic rats displayed significant differences at 30 min (1.11 ± 0.08% ID/g vs. 0.63 ± 0.09% ID/g, p < 0.0001). In the presence of SCH23390, the pancreatic uptake of [125 I]I-R-(+)-TISCH at 30 min in normal rats was lower (1.01 ± 0.04% ID/g, p < 0.05). Although the blocking was not complete, [125 I]I-R-(+)-TISCH showed specific binding signals to the pancreas. Furthermore, the uptakes of [125 I]I-R-(+)-TISCH in INS-1 cells were reduced in the presence of SCH23390 at different concentrations. [125 I]I-R-(+)-TISCH displayed a respectable uptake in insulinoma. Overall, [125 I]I-R-(+)-TISCH provided specific binding signals to pancreatic β cells. Although the specific signal may not be sufficient for imaging in vivo, the dopamine D1 receptor can still be considered as a potential target for studying BCM. Further investigation will be required to optimize the ligand.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Guangwen Li
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Yuli Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Haiyan Hong
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Linlin Li
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Yang Luo
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Ran Wang
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Lin Zhu
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Hank F Kung
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jinxia Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Chen K, Zhang J, Huang Y, Tian X, Yang Y, Dong A. Single-cell RNA-seq transcriptomic landscape of human and mouse islets and pathological alterations of diabetes. iScience 2022; 25:105366. [PMID: 36339258 PMCID: PMC9626680 DOI: 10.1016/j.isci.2022.105366] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/13/2022] [Accepted: 10/12/2022] [Indexed: 11/15/2022] Open
Abstract
Single-cell RNA sequencing has paved the way for delineating the pancreatic islet cell atlas and identifying hallmarks of diabetes. However, pathological alterations of type 2 diabetes (T2D) remain unclear. We isolated pancreatic islets from control and T2D mice for single-cell RNA sequencing (scRNA-seq) and retrieved multiple datasets from the open databases. The complete islet cell landscape and robust marker genes and transcription factors of each endocrine cell type were identified. GLRA1 was restricted to beta cells, and beta cells exhibited obvious heterogeneity. The beta subcluster in the T2D mice remarkably decreased the expression of Slc2a2, G6pc2, Mafa, Nkx6-1, Pdx1, and Ucn3 and had higher unfolded protein response (UPR) scores than in the control mice. Moreover, we developed a Web-based interactive tool, creating new opportunities for the data mining of pancreatic islet scRNA-seq datasets. In conclusion, our work provides a valuable resource for a deeper understanding of the pathological mechanism underlying diabetes. Cross-species scRNA-seq reveals the complete cell landscape of the islets of Langerhans We identify the robust marker genes and TFs of each endocrine and exocrine cell type Pathological alterations of beta cells in type 2 diabetes are explored A Web-based interactive tool is established for pancreatic islet scRNA-seq datasets
Collapse
|
7
|
Van Simaeys D, De La Fuente A, Zilio S, Zoso A, Kuznetsova V, Alcazar O, Buchwald P, Grilli A, Caroli J, Bicciato S, Serafini P. RNA aptamers specific for transmembrane p24 trafficking protein 6 and Clusterin for the targeted delivery of imaging reagents and RNA therapeutics to human β cells. Nat Commun 2022; 13:1815. [PMID: 35383192 PMCID: PMC8983715 DOI: 10.1038/s41467-022-29377-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/08/2022] [Indexed: 12/20/2022] Open
Abstract
The ability to detect and target β cells in vivo can substantially refine how diabetes is studied and treated. However, the lack of specific probes still hampers a precise characterization of human β cell mass and the delivery of therapeutics in clinical settings. Here, we report the identification of two RNA aptamers that specifically and selectively recognize mouse and human β cells. The putative targets of the two aptamers are transmembrane p24 trafficking protein 6 (TMED6) and clusterin (CLUS). When given systemically in immune deficient mice, these aptamers recognize the human islet graft producing a fluorescent signal proportional to the number of human islets transplanted. These aptamers cross-react with endogenous mouse β cells and allow monitoring the rejection of mouse islet allografts. Finally, once conjugated to saRNA specific for X-linked inhibitor of apoptosis (XIAP), they can efficiently transfect non-dissociated human islets, prevent early graft loss, and improve the efficacy of human islet transplantation in immunodeficient in mice.
Collapse
Affiliation(s)
- Dimitri Van Simaeys
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Adriana De La Fuente
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Serena Zilio
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Alessia Zoso
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Victoria Kuznetsova
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Oscar Alcazar
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Andrea Grilli
- Center for Genome Research, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Jimmy Caroli
- Center for Genome Research, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvio Bicciato
- Center for Genome Research, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Paolo Serafini
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA. .,Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, USA. .,Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
8
|
Cheung P, Eriksson O. The Current State of Beta-Cell-Mass PET Imaging for Diabetes Research and Therapies. Biomedicines 2021; 9:1824. [PMID: 34944640 PMCID: PMC8698817 DOI: 10.3390/biomedicines9121824] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/28/2021] [Accepted: 11/30/2021] [Indexed: 12/03/2022] Open
Abstract
Diabetes is a chronic metabolic disease affecting over 400 million people worldwide and one of the leading causes of death, especially in developing nations. The disease is characterized by chronic hyperglycemia, caused by defects in the insulin secretion or action pathway. Current diagnostic methods measure metabolic byproducts of the disease such as glucose level, glycated hemoglobin (HbA1c), insulin or C-peptide levels, which are indicators of the beta-cell function. However, they inaccurately reflect the disease progression and provide poor longitudinal information. Beta-cell mass has been suggested as an alternative approach to study disease progression in correlation to beta-cell function, as it behaves differently in the diabetes physiopathology. Study of the beta-cell mass, however, requires highly invasive and potentially harmful procedures such as pancreatic biopsies, making diagnosis and monitoring of the disease tedious. Nuclear medical imaging techniques using radiation emitting tracers have been suggested as strong non-invasive tools for beta-cell mass. A highly sensitive and high-resolution technique, such as positron emission tomography, provides an ideal solution for the visualization of beta-cell mass, which is particularly essential for better characterization of a disease such as diabetes, and for estimating treatment effects towards regeneration of the beta-cell mass. Development of novel, validated biomarkers that are aimed at beta-cell mass imaging are thus highly necessary and would contribute to invaluable breakthroughs in the field of diabetes research and therapies. This review aims to describe the various biomarkers and radioactive probes currently available for positron emission tomography imaging of beta-cell mass, as well as highlight the need for precise quantification and visualization of the beta-cell mass for designing new therapy strategies and monitoring changes in the beta-cell mass during the progression of diabetes.
Collapse
Affiliation(s)
- Pierre Cheung
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, SE-75183 Uppsala, Sweden;
| | | |
Collapse
|
9
|
Zhou B, Tsai YJ, Chen X, Duncan JS, Liu C. MDPET: A Unified Motion Correction and Denoising Adversarial Network for Low-Dose Gated PET. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:3154-3164. [PMID: 33909561 PMCID: PMC8588635 DOI: 10.1109/tmi.2021.3076191] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
In positron emission tomography (PET), gating is commonly utilized to reduce respiratory motion blurring and to facilitate motion correction methods. In application where low-dose gated PET is useful, reducing injection dose causes increased noise levels in gated images that could corrupt motion estimation and subsequent corrections, leading to inferior image quality. To address these issues, we propose MDPET, a unified motion correction and denoising adversarial network for generating motion-compensated low-noise images from low-dose gated PET data. Specifically, we proposed a Temporal Siamese Pyramid Network (TSP-Net) with basic units made up of 1.) Siamese Pyramid Network (SP-Net), and 2.) a recurrent layer for motion estimation among the gates. The denoising network is unified with our motion estimation network to simultaneously correct the motion and predict a motion-compensated denoised PET reconstruction. The experimental results on human data demonstrated that our MDPET can generate accurate motion estimation directly from low-dose gated images and produce high-quality motion-compensated low-noise reconstructions. Comparative studies with previous methods also show that our MDPET is able to generate superior motion estimation and denoising performance. Our code is available at https://github.com/bbbbbbzhou/MDPET.
Collapse
|
10
|
Kahn SE, Chen YC, Esser N, Taylor AJ, van Raalte DH, Zraika S, Verchere CB. The β Cell in Diabetes: Integrating Biomarkers With Functional Measures. Endocr Rev 2021; 42:528-583. [PMID: 34180979 PMCID: PMC9115372 DOI: 10.1210/endrev/bnab021] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Indexed: 02/08/2023]
Abstract
The pathogenesis of hyperglycemia observed in most forms of diabetes is intimately tied to the islet β cell. Impairments in propeptide processing and secretory function, along with the loss of these vital cells, is demonstrable not only in those in whom the diagnosis is established but typically also in individuals who are at increased risk of developing the disease. Biomarkers are used to inform on the state of a biological process, pathological condition, or response to an intervention and are increasingly being used for predicting, diagnosing, and prognosticating disease. They are also proving to be of use in the different forms of diabetes in both research and clinical settings. This review focuses on the β cell, addressing the potential utility of genetic markers, circulating molecules, immune cell phenotyping, and imaging approaches as biomarkers of cellular function and loss of this critical cell. Further, we consider how these biomarkers complement the more long-established, dynamic, and often complex measurements of β-cell secretory function that themselves could be considered biomarkers.
Collapse
Affiliation(s)
- Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - Yi-Chun Chen
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Nathalie Esser
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - Austin J Taylor
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Daniël H van Raalte
- Department of Internal Medicine, Amsterdam University Medical Center (UMC), Vrije Universiteit (VU) University Medical Center, 1007 MB Amsterdam, The Netherlands.,Department of Experimental Vascular Medicine, Amsterdam University Medical Center (UMC), Academic Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Sakeneh Zraika
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - C Bruce Verchere
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| |
Collapse
|
11
|
Li J, Chen P, Bao Y, Sun Y, He J, Liu X. PET Imaging of Vesicular Monoamine Transporter 2 in Early Diabetic Retinopathy Using [ 18F]FP-(+)-DTBZ. Mol Imaging Biol 2021; 22:1161-1169. [PMID: 31650482 DOI: 10.1007/s11307-019-01443-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Diabetic retinopathy (DR) is characterized by dopaminergic neuron loss in the retina of the eyes. [18F]fluoropropyl-(+)-dihydrotetrabenazine ([18F]FP-(+)-DTBZ) positron emission tomography (PET) has been shown to detect dopaminergic neuron loss. The study is to investigate the feasibility of PET imaging with [18F]FP-(+)-DTBZ for early diagnosis of diabetic retinopathy (DR) in diabetes mellitus (DM) rat models. METHODS The DM rat model was established by a single intraperitoneal injection of streptozotocin (STZ) (65 mg/kg). After 4 weeks, 8 weeks, and 12 weeks of STZ injection, the retinas of the rats were evaluated by electroretinogram (ERG), color fundus photography (CFP), fundus fluorescein angiography (FFA), and small animal PET scan with [18F]FP-(+)-DTBZ by targeting vesicular monoamine transporter 2 (VMAT2). [18F]FP-(+)-DTBZ uptake in retina was quantified as standardized uptake value (SUV). Immunofluorescence staining and Western blot were also performed to confirm the expression level of VMAT2 in retina. RESULTS ERG dysfunction was observed at 8 weeks in STZ-diabetic rats, evidenced by smaller amplitudes of oscillatory potentials (OPs) when compared with OPs in normal rats. CFP and FFA showed no significant difference in vascular leakage and neovascularization between STZ-diabetic retinas and normal ones until 8 weeks. PET imaging revealed that the SUV of [18F]FP-(+)-DTBZ was significantly lower in the STZ-diabetic retinas compared with the normal ones as early as of week 4. The results from immunofluorescence staining and Western blots confirmed the early findings in PET imaging studies. CONCLUSIONS Early DR can be non-invasively detected with PET imaging using [18F]FP-(+)-DTBZ targeting VMAT2. The expression level of VMAT2 in retina may act as a new biomarker for early DR diagnosis.
Collapse
Affiliation(s)
- Jun Li
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, No. 12 Urumchi Middle Road, Jing'an District, Shanghai, 200040, China
- Department of Radiology and Medical imaging, University of Virginia, Charlottesville, VA, 22908, USA
| | - Ping Chen
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, No. 12 Urumchi Middle Road, Jing'an District, Shanghai, 200040, China
| | - Yong Bao
- Department of Pharmacology and Biochemistry, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yu Sun
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, No. 12 Urumchi Middle Road, Jing'an District, Shanghai, 200040, China
| | - Jiang He
- Department of Radiology and Medical imaging, University of Virginia, Charlottesville, VA, 22908, USA
| | - Xingdang Liu
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, No. 12 Urumchi Middle Road, Jing'an District, Shanghai, 200040, China.
| |
Collapse
|
12
|
Bini J, Norcross M, Cheung M, Duffy A. The Role of Positron Emission Tomography in Bariatric Surgery Research: a Review. Obes Surg 2021; 31:4592-4606. [PMID: 34304378 DOI: 10.1007/s11695-021-05576-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/24/2021] [Accepted: 06/30/2021] [Indexed: 12/15/2022]
Abstract
Bariatric surgery, initially understood as restricting or bypassing the amount of food that reaches the stomach to reduce food intake and/or increase malabsorption of food to promote weight loss, is now recognized to also affect incretin signaling in the gut and promote improvements in system-wide metabolism. Positron emission tomography (PET) is an imaging technique whereby patients are injected with picomolar concentrations of radioactive molecules, below the threshold of having physiological effects, to measure spatial distributions of blood flow, metabolism, receptor, and enzyme pharmacology. Recent advances in both whole-body PET imaging and radioligand development will allow for novel research that may help clarify the roles of peripheral and central receptor/enzyme systems in treating obesity with bariatric surgery.
Collapse
Affiliation(s)
- Jason Bini
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, USA.
| | | | - Maija Cheung
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Andrew Duffy
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
13
|
Dai Y, Sa R, Guan F, Wang Q, Li Y, Zhao H. A Purification Method of 18F-FP-(+)-DTBZ via Solid-Phase Extraction With Combined Cartridges. Front Med (Lausanne) 2021; 8:693632. [PMID: 34307421 PMCID: PMC8298858 DOI: 10.3389/fmed.2021.693632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 06/17/2021] [Indexed: 11/13/2022] Open
Abstract
Background: To optimize [18F] 9-fluoropropyl-(+)-dihydrotetrabenazine (18F-FP-(+)-DTBZ) purification via solid-phase extraction (SPE) with combined cartridges to facilitate its widespread clinical application. Methods: A modified SPE purification method, employing Sep-Pak PS-2 and Sep-Pak C18 cartridges, was used for the preparation of 18F-FP-(+)-DTBZ. This method was compared to the purification method of high-pressure liquid chromatography (HPLC) and SPE with one cartridge, following quality control test and positron emission tomography (PET) imaging in healthy volunteers and patients with parkinsn's disease (PD). Results: A SPE purification method integrating Sep-Pak PS-2 and Sep-Pak C18 cartridges was implemented successfully. The retention time of 18F-FP-(+)-DTBZ purified by HPLC, SPE with Sep-Pak PS-2, SPE with Sep-Pak C18, and SPE with combined use of Sep-Pak PS-2 and Sep-Pak C18 cartridges was 8.7, 8.8, 8.7, and 8.9 min, respectively. Fewest impurity peak was detected in 18F-FP-(+)-DTBZ purified by the SPE with combined use of Sep-Pak PS-2 and Sep-Pak C18 cartridges. This modified SPE purification method provided a satisfactory radiochemical yield of 29 ± 1.8% with radiochemical purity >99% and shortened synthesis time to 27 min. The brain uptake of 18F-FP-(+)-DTBZ purified by the modified SPE was comparable to that purified by HPLC in both healthy volunteers and PD patients. Conclusions: A SPE method integrating Sep-Pak PS-2 and Sep-Pak C18 cartridges for purification of 18F-FP-(+)-DTBZ may be highly suited to automatic synthesis for routine clinical applications, as it provides excellent radiochemical purity, high yield as well as operational simplicity.
Collapse
Affiliation(s)
- Yuyin Dai
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China
| | - Ri Sa
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China
| | - Feng Guan
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China
| | - Qi Wang
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yinghua Li
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China
| | - Hongguang Zhao
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Roberts S, Khera E, Choi C, Navaratna T, Grimm J, Thurber GM, Reiner T. Optoacoustic Imaging of Glucagon-like Peptide-1 Receptor with a Near-Infrared Exendin-4 Analog. J Nucl Med 2021; 62:839-848. [PMID: 33097631 PMCID: PMC8729860 DOI: 10.2967/jnumed.120.252262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/18/2020] [Indexed: 11/16/2022] Open
Abstract
Limitations in current imaging tools have long challenged the imaging of small pancreatic islets in animal models. Here, we report the first development and in vivo validation testing of a broad-spectrum and high-absorbance near-infrared optoacoustic contrast agent, E4x12-Cy7. Our near-infrared tracer is based on the amino acid sequence of exendin-4 and targets the glucagon-like peptide-1 receptor (GLP-1R). Cell assays confirmed that E4x12-Cy7 has a high-binding affinity (dissociation constant, Kd, 4.6 ± 0.8 nM). Using the multispectral optoacoustic tomography, we imaged E4x12-Cy7 and optoacoustically visualized β-cell insulinoma xenografts in vivo for the first time. In the future, similar optoacoustic tracers that are specific for β-cells and combines optoacoustic and fluorescence imaging modalities could prove to be important tools for monitoring the pancreas for the progression of diabetes.
Collapse
Affiliation(s)
- Sheryl Roberts
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eshita Khera
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Crystal Choi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Tejas Navaratna
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Jan Grimm
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Program of Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
- Pharmacology Program, Weill Cornell Medical College, New York, New York
| | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan; and
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
15
|
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
|
16
|
Joosten L, Boss M, Jansen T, Brom M, Buitinga M, Aarntzen E, Eriksson O, Johansson L, de Galan B, Gotthardt M. Molecular Imaging of Diabetes. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00041-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
17
|
Eriksson O, Långström B, Antoni G. News ways of understanding the complex biology of diabetes using PET. Nucl Med Biol 2021; 92:65-71. [PMID: 32387114 DOI: 10.1016/j.nucmedbio.2020.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/27/2020] [Accepted: 04/15/2020] [Indexed: 11/22/2022]
Abstract
The understanding of metabolic disease and diabetes on a molecular level has increased significantly due to the recent advances in molecular biology and biotechnology. However, in vitro studies and animal models do not always translate to the human disease, perhaps illustrated by the failure of many drug candidates in the clinical phase. Non-invasive biomedical imaging techniques such as Positron Emission Tomography (PET) offer tools for direct visualization and quantification of molecular processes in humans. Developments in this area potentially enable longitudinal in vivo studies of receptors and processes involved in diabetes guiding drug development and diagnosis in the near future. This mini-review focuses on describing the overall perspective of how PET can be used to increase our understanding and improve treatment of diabetes. The methodological aspects and future developments and challenges are highlighted.
Collapse
Affiliation(s)
- O Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden; Antaros Medical AB, Mölndal, Sweden
| | - B Långström
- Department of Chemistry, Uppsala University, Uppsala, Sweden
| | - G Antoni
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
18
|
Fujimoto H, Fujita N, Hamamatsu K, Murakami T, Nakamoto Y, Saga T, Ishimori T, Shimizu Y, Watanabe H, Sano K, Harada N, Nakamura H, Toyoda K, Kimura H, Nakagawa S, Hirai M, Murakami A, Ono M, Togashi K, Saji H, Inagaki N. First-in-Human Evaluation of Positron Emission Tomography/Computed Tomography With [ 18F]FB(ePEG12)12-Exendin-4: A Phase 1 Clinical Study Targeting GLP-1 Receptor Expression Cells in Pancreas. Front Endocrinol (Lausanne) 2021; 12:717101. [PMID: 34489868 PMCID: PMC8417326 DOI: 10.3389/fendo.2021.717101] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/27/2021] [Indexed: 01/09/2023] Open
Abstract
Pancreatic β-cell mass (BCM) has a central importance in the pathophysiology of diabetes mellitus. Recently, pancreatic β-cell-specific imaging, especially positron emission tomography (PET) with exendin-based probes, has emerged for non-invasive evaluation of BCM. We developed a novel exendin-based probe labeled with fluorine-18, [18F]FB(ePEG12)12-exendin-4 (18F-Ex4) for PET imaging. We subsequently conducted a first-in-human phase 1 study of 18F-Ex4 PET/computed tomography (CT) and investigated the safety and utility for visualizing the pancreas. Six healthy male subjects were enrolled in this study. A low dose (37.0 MBq) of 18F-Ex4 PET/CT was administered (first cohort: n = 2), and subsequently a higher dose (74.0 MBq) was administered (second cohort: n = 4). In the first and second cohorts, 38.6 ± 4.8 and 71.1 ± 4.8 MBq of 18F-Ex4 were administered, respectively. No serious adverse events were observed in both groups. Only one participant in the first cohort showed transient hypoglycemia during the PET scans. 18F-Ex4 PET/CT successfully visualized the pancreas in all participants. The mean standardized uptake value of the pancreas was found to be higher than that in the surrounding organs, except for the bladder and kidney, during the observation. Dosimetry analyses revealed the effective systemic doses of 18F-Ex4 as 0.0164 ± 0.0019 mSv/MBq (first cohort) and 0.0173 ± 0.0020 mSv/MBq (second cohort). 18F-Ex4 PET/CT demonstrated the safety and utility for non-invasive visualization of the pancreas in healthy male subjects. 18F-Ex4 is promising for clinical PET imaging targeting pancreatic β cells.
Collapse
Affiliation(s)
- Hiroyuki Fujimoto
- Radioisotope Research Center, Agency of Health, Safety and Environment, Kyoto University, Kyoto, Japan
| | - Naotaka Fujita
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keita Hamamatsu
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takaaki Murakami
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tsuneo Saga
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takayoshi Ishimori
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoichi Shimizu
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Kohei Sano
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Norio Harada
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Kentaro Toyoda
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Kimura
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Shunsaku Nakagawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Mitsuharu Hirai
- Research and Development Division, Arkray, Inc., Kyoto, Japan
| | | | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Kaori Togashi
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideo Saji
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- *Correspondence: Nobuya Inagaki,
| |
Collapse
|
19
|
Murakami T, Fujimoto H, Inagaki N. Non-invasive Beta-cell Imaging: Visualization, Quantification, and Beyond. Front Endocrinol (Lausanne) 2021; 12:714348. [PMID: 34248856 PMCID: PMC8270651 DOI: 10.3389/fendo.2021.714348] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 06/14/2021] [Indexed: 01/07/2023] Open
Abstract
Pancreatic beta (β)-cell dysfunction and reduced mass play a central role in the development and progression of diabetes mellitus. Conventional histological β-cell mass (BCM) analysis is invasive and limited to cross-sectional observations in a restricted sampling area. However, the non-invasive evaluation of BCM remains elusive, and practical in vivo and clinical techniques for β-cell-specific imaging are yet to be established. The lack of such techniques hampers a deeper understanding of the pathophysiological role of BCM in diabetes, the implementation of personalized BCM-based diabetes management, and the development of antidiabetic therapies targeting BCM preservation and restoration. Nuclear medical techniques have recently triggered a major leap in this field. In particular, radioisotope-labeled probes using exendin peptides that include glucagon-like peptide-1 receptor (GLP-1R) agonist and antagonist have been employed in positron emission tomography and single-photon emission computed tomography. These probes have demonstrated high specificity to β cells and provide clear images accurately showing uptake in the pancreas and transplanted islets in preclinical in vivo and clinical studies. One of these probes, 111indium-labeled exendin-4 derivative ([Lys12(111In-BnDTPA-Ahx)]exendin-4), has captured the longitudinal changes in BCM during the development and progression of diabetes and under antidiabetic therapies in various mouse models of type 1 and type 2 diabetes mellitus. GLP-1R-targeted imaging is therefore a promising tool for non-invasive BCM evaluation. This review focuses on recent advances in non-invasive in vivo β-cell imaging for BCM evaluation in the field of diabetes; in particular, the exendin-based GLP-1R-targeted nuclear medicine techniques.
Collapse
Affiliation(s)
- Takaaki Murakami
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroyuki Fujimoto
- Radioisotope Research Center, Agency of Health, Safety and Environment, Kyoto University, Kyoto, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
- *Correspondence: Nobuya Inagaki,
| |
Collapse
|
20
|
Lubberink M, Eriksson O. [ 11C]5-Hydroxy-tryptophan model for quantitative assessment of in vivo serotonin biosynthesis, retention and degradation in the endocrine pancreas. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2020; 10:226-234. [PMID: 33224618 PMCID: PMC7675115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/08/2020] [Indexed: 06/11/2023]
Abstract
[11C]5-Hydroxy-tryptophan ([11C]5-HTP) is a Positron Emission Tomography marker for serotonergic biosynthesis and degradation, with use in imaging of neuroendocrine tumors and recently also the endocrine pancreas in diabetes. In order to further develop [11C]5-HTP as a quantitative in vivo tool for understanding the mechanisms of serotonin signaling in human pancreas, we aimed to develop a kinetic modeling approach sensitive for changes in serotonin biosynthesis, retention and degradation. Cynomolgus monkeys were examined by [11C]5-HTP PET/CT, either at baseline (n=9) or following intravenous pretreatment with 3 mg/kg carbidopa (Dopa Decarboxylase inhibitor, n=3) or 2 mg/kg clorgyline (Monoamine Oxidase-A inhibitor, n=5). The dynamic tissue uptake was analysed by a 2-tissue compartment model including an efflux mechanism from the second tissue compartment (2TC kloss), which theoretically reproduces the known processing of 5-HTP in neuroendocrine cells. The 2TC kloss model could accurately describe all three modes of tissue kinetics depending on the pretreatment regiment. Rate constant k3 (corresponding to DDC activity) and the macro-parameter Flux (Ki) was decreased (P<0.05) by carbidopa pretreatment, while k2 (corresponding to cellular washout of intact [11C]5-HTP) was increased (P<0.05). The efflux parameter kloss (corresponding to MAO-A activity) was decreased (P<0.05) by pretreatment of clorgyline, while the macro-parameter Flux/Efflux ratio (Ki/kloss) was increased (P<0.0001). We present a compartment model analysis method that can quantitatively assess in vivo pharmacological interactions with several of the key enzymatic steps of the serotonergic biosynthesis in pancreas.
Collapse
Affiliation(s)
- Mark Lubberink
- Department of Surgical Sciences, Uppsala UniversityUppsala, Sweden
| | - Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala UniversityUppsala, Sweden
| |
Collapse
|
21
|
Current Progress and Perspective: Clinical Imaging of Islet Transplantation. Life (Basel) 2020; 10:life10090213. [PMID: 32961769 PMCID: PMC7555367 DOI: 10.3390/life10090213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Islet transplantation has great potential as a cure for type 1 diabetes. At present; the lack of a clinically validated non-invasive imaging method to track islet grafts limits the success of this treatment. Some major clinical imaging modalities and various molecular probes, which have been studied for non-invasive monitoring of transplanted islets, could potentially fulfill the goal of understanding pathophysiology of the functional status and viability of the islet grafts. In this current review, we summarize the recent clinical studies of a variety of imaging modalities and molecular probes for non-invasive imaging of transplanted beta cell mass. This review also includes discussions on in vivo detection of endogenous beta cell mass using clinical imaging modalities and various molecular probes, which will be useful for longitudinally detecting the status of islet transplantation in Type 1 diabetic patients. For the conclusion and perspectives, we highlight the applications of multimodality and novel imaging methods in islet transplantation.
Collapse
|
22
|
Cong GZ, Ghosh KK, Mishra S, Gulyás M, Kovács T, Máthé D, Padmanabhan P, Gulyás B. Targeted pancreatic beta cell imaging for early diagnosis. Eur J Cell Biol 2020; 99:151110. [PMID: 33070042 DOI: 10.1016/j.ejcb.2020.151110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 06/29/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic beta cells are important in blood glucose level regulation. As type 1 and 2 diabetes are getting prevalent worldwide, we need to explore new methods for early detection of beta cell-related afflictions. Using bioimaging techniques to measure beta cell mass is crucial because a decrease in beta cell density is seen in diseases such as diabetes and thus can be a new way of diagnosis for such diseases. We also need to appraise beta cell purity in transplanted islets for type 1 diabetes patients. Sufficient amount of functional beta cells must also be determined before being transplanted to the patients. In this review, indirect imaging of beta cells will be discussed. This includes membrane protein on pancreatic beta cells whereby specific probes are designed for different imaging modalities mainly magnetic resonance imaging, positron emission tomography and fluorescence imaging. Direct imaging of insulin is also explored though probes synthesized for such function are relatively fewer. The path for successful pancreatic beta cell imaging is fraught with challenges like non-specific binding, lack of beta cell-restricted targets, the requirement of probes to cross multiple lipid layers to bind to intracellular insulin. Hence, there is an urgent need to develop new imaging techniques and innovative probing constructs in the entire imaging chain of bioengineering to provide early detection of beta cell-related pathology.
Collapse
Affiliation(s)
- Goh Zheng Cong
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Krishna Kanta Ghosh
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Sachin Mishra
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Miklós Gulyás
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskölds väg 20, Uppsala Se-751 85, Sweden
| | - Tibor Kovács
- Institute of Radiochemistry and Radioecology, University of Pannonia, Egyetem u. 10, H-8200 Veszprém, Hungary
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Semmelweis University Faculty of Medicine, Tűzoltó u. 37-47, Budapest H-1094, Hungary
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore.
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore.
| |
Collapse
|
23
|
Kong Y, Zhou H, Feng H, Zhuang J, Wen T, Zhang C, Sun B, Wang J, Guan Y. Elucidating the Relationship Between Diabetes Mellitus and Parkinson's Disease Using 18F-FP-(+)-DTBZ, a Positron-Emission Tomography Probe for Vesicular Monoamine Transporter 2. Front Neurosci 2020; 14:682. [PMID: 32760240 PMCID: PMC7372188 DOI: 10.3389/fnins.2020.00682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 06/03/2020] [Indexed: 01/25/2023] Open
Abstract
Diabetes mellitus (DM) and Parkinson’s disease (PD) have been and will continue to be two common chronic diseases globally that are difficult to diagnose during the prodromal phase. Current molecular genetics, cell biological, and epidemiological evidences have shown the correlation between PD and DM. PD shares the same pathogenesis pathways and pathological factors with DM. In addition, β-cell reduction, which can cause hyperglycemia, is a striking feature of DM. Recent studies indicated that hyperglycemia is highly relevant to the pathologic changes in PD. However, further correlation between DM and PD remains to be investigated. Intriguingly, polycystic monoamine transporter 2 (VMAT2), which is co-expressed in dopaminergic neurons and β cells, is responsible for taking up dopamine into the presynaptic vesicles and can specifically bind to the β cells. Furthermore, we have summarized the specific molecular and diagnostic functions of VMAT2 for the two diseases reported in this review. Therefore, VMAT2 can be applied as a target probe for positron emission tomography (PET) imaging to detect β-cell and dopamine level changes, which can contribute to the diagnosis of DM and PD during the prodromal phase. Targeting VMAT2 with the molecular probe 18F-FP-(+)-DTBZ can be an entry point for the β cell mass (BCM) changes in DM at the molecular level, to clarify the potential relationship between DM and PD. VMAT2 has promising clinical significance in investigating the pathogenesis, early diagnosis, and treatment evaluation of the two diseases.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Haicong Zhou
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Hu Feng
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Junyi Zhuang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Tieqiao Wen
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Chencheng Zhang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bomin Sun
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Pan W, Zheng X, Chen G, Su L, Luo S, Wang W, Ye S, Weng J, Min Y. Nanotechnology's application in Type 1 diabetes. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1645. [PMID: 32558337 DOI: 10.1002/wnan.1645] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/17/2020] [Accepted: 04/17/2020] [Indexed: 12/16/2022]
Abstract
Type 1 diabetes mellitus (T1D) is an autoimmune disease caused by the immune system attacking islet cells. T1D, with a long prediabetes period, and the incidence of T1D increases with age during childhood and peaks at 10-14 years. And once it gets overt, it requires lifelong insulin replace treatment. Therefore, the diagnosis of early-stage T1D and effective treatments are important for the management of T1D patients. The imaging methods, such as magnetic resonance imaging (MRI) and so on, were applied in diagnosis of the early stage T1D and its development tracking. The addition of nanomaterials, especially in MRI, can improve the quality of T1D imaging for the diagnosis of T1D at early stage and cause less harm to human body. Meantime, among various treatment options, islet transplantation and immunotherapy are promising, effective, and less independent on insulin. The addition of nanotechnology can effectively reduce the attack of the immune system on drugs and cells, making the therapeutic drug more targeted in the body and prolonging the action time between drugs and cells, thus its addition makes these therapy safer and more efficient. In this review, we attempt to summarize the recent advances in the development of nanotechnology advances of T1D including using nanomaterials for the diagnosis and immunological imaging of T1D, protecting the transplanted islet cells from immune system attack, and delivering relevant molecules to targeted immunocytes. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Emerging Technologies Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement.
Collapse
Affiliation(s)
- Wen Pan
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, China.,CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, China.,Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Xueying Zheng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, China
| | - Guiyuan Chen
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, China.,CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, China.,Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Lanhong Su
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, China.,CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, China.,Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Sihui Luo
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, China
| | - Shandong Ye
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, China
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, China.,Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuanzeng Min
- Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, China.,CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, China.,Department of Chemistry, University of Science and Technology of China, Hefei, China.,Department of Bio-X Interdisciplinary Science at Hefei National Laboratory (HFNL) for Physical Science at the Microscale, University of Science and Technology of China, Hefei, China
| |
Collapse
|
25
|
Jiang D, Kong Y, Ren S, Cai H, Zhang Z, Huang Z, Peng F, Hua F, Guan Y, Xie F. Decreased striatal vesicular monoamine transporter 2 (VMAT2) expression in a type 1 diabetic rat model: A longitudinal study using micro-PET/CT. Nucl Med Biol 2020; 82-83:89-95. [PMID: 32120243 DOI: 10.1016/j.nucmedbio.2020.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/02/2020] [Accepted: 02/18/2020] [Indexed: 02/05/2023]
Abstract
AIMS Diabetes mellitus is a risk factor for Parkinson's disease. These diseases share similar pathogenic pathways, such as mitochondrial dysfunction, inflammation, and altered metabolism. Despite these similarities, the pathogenic relationship between these two diseases is unclear. [18F]FP-(+)-DTBZ is a promising radiotracer targeting VMAT2, which has been used to measure β-cell mass and to diagnose Parkinson's disease. The aim of this study was to examine the effect of type 1 diabetes on VMAT2 expression in the striatum using [18F]FP-(+)-DTBZ. MATERIALS AND METHODS A longitudinal study of type 1 diabetic rats was established by intraperitoneally injecting male Wistar rats with streptozotocin. Rats injected with saline were used as the control group. Glucose level, body weight, and [18F]FP-(+)-DTBZ uptake in the striatum and pancreas were evaluated at 0.5, 1, 4, 6 and 12 months after STZ or saline injection. RESULTS At one-half month post-STZ injection, the glucose levels in these rats increased and then returned to a normal level at 6 months. Along with increased glucose levels, body weight was also decreased significantly and returned slowly to a normal level. β-Cell mass and striatal [18F]FP-(+)-DTBZ uptake were impaired significantly at 2 weeks post-STZ injection in type 1 diabetic rats and returned to a normal level at 6 and 4 months post-STZ injection. CONCLUSIONS Due to increased glucose levels and decreased β-cell mass, decreased [18F]FP-(+)-DTBZ uptake in the striatum was observed in type 1 diabetic rats. Decreased BCM and increased glucose levels were correlated with VMAT2 expression in the striatum which indicated DM is a risk factor for PD.
Collapse
Affiliation(s)
- Donglang Jiang
- PET Center, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Shuhua Ren
- PET Center, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Huawei Cai
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, 610041 Chengdu, China
| | - Zhengwei Zhang
- PET Center, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Zheming Huang
- PET Center, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Fangyu Peng
- Department of Radiology, University of Texas Southwestern Medical Center, 75390 Dallas, TX, USA
| | - Fengchun Hua
- PET Center, Huashan Hospital, Fudan University, 200040 Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, 200040 Shanghai, China.
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, 200040 Shanghai, China.
| |
Collapse
|
26
|
Kang NY, Lee JY, Lee SH, Song IH, Hwang YH, Kim MJ, Phue WH, Agrawalla BK, Wan SYD, Lalic J, Park SJ, Kim JJ, Kwon HY, Im SH, Bae MA, Ahn JH, Lim CS, Teo AKK, Park S, Kim SE, Lee BC, Lee DY, Chang YT. Multimodal Imaging Probe Development for Pancreatic β Cells: From Fluorescence to PET. J Am Chem Soc 2020; 142:3430-3439. [PMID: 32040300 DOI: 10.1021/jacs.9b11173] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pancreatic β cells are responsible for insulin secretion and are important for glucose regulation in a healthy body and diabetic disease patient without prelabeling of islets. While the conventional biomarkers for diabetes have been glucose and insulin concentrations in the blood, the direct determination of the pancreatic β cell mass would provide critical information for the disease status and progression. By combining fluorination and diversity-oriented fluorescence library strategy, we have developed a multimodal pancreatic β cell probe PiF for both fluorescence and for PET (positron emission tomography). By simple tail vein injection, PiF stains pancreatic β cells specifically and allows intraoperative fluorescent imaging of pancreatic islets. PiF-injected pancreatic tissue even facilitated an antibody-free islet analysis within 2 h, dramatically accelerating the day-long histological procedure without any fixing and dehydration step. Not only islets in the pancreas but also the low background of PiF in the liver allowed us to monitor the intraportal transplanted islets, which is the first in vivo visualization of transplanted human islets without a prelabeling of the islets. Finally, we could replace the built-in fluorine atom in PiF with radioactive 18F and successfully demonstrate in situ PET imaging for pancreatic islets.
Collapse
Affiliation(s)
- Nam-Young Kang
- Laboratory of Bioimaging Probe Development , Singapore Bioimaging Consortium, Agency for Science, Technology and Research , Singapore 138667 , Singapore
| | - Jung Yeol Lee
- New Drug Discovery Center, DGMIF , Daegu 41061 , Republic of Korea
| | - Sang Hee Lee
- Department of Nuclear Medicine , Seoul National University College of Medicine, Seoul National University Bundang Hospital , Seongnam 13620 , Republic of Korea.,Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology , Seoul National University , Seoul 08826 , Republic of Korea
| | - In Ho Song
- Department of Nuclear Medicine , Seoul National University College of Medicine, Seoul National University Bundang Hospital , Seongnam 13620 , Republic of Korea
| | - Yong Hwa Hwang
- Department of Bioengineering, College of Engineering, and BK21 PLUS Future Biopharmaceutical Human Resources Training and Research Team, and Institute of Nano Science & Technology (INST) , Hanyang University , Seoul 04763 , Republic of Korea
| | - Min Jun Kim
- Department of Bioengineering, College of Engineering, and BK21 PLUS Future Biopharmaceutical Human Resources Training and Research Team, and Institute of Nano Science & Technology (INST) , Hanyang University , Seoul 04763 , Republic of Korea
| | - Wut Hmone Phue
- Laboratory of Bioimaging Probe Development , Singapore Bioimaging Consortium, Agency for Science, Technology and Research , Singapore 138667 , Singapore
| | | | - Si Yan Diana Wan
- Laboratory of Bioimaging Probe Development , Singapore Bioimaging Consortium, Agency for Science, Technology and Research , Singapore 138667 , Singapore
| | - Janise Lalic
- Laboratory of Bioimaging Probe Development , Singapore Bioimaging Consortium, Agency for Science, Technology and Research , Singapore 138667 , Singapore
| | - Sung-Jin Park
- Laboratory of Bioimaging Probe Development , Singapore Bioimaging Consortium, Agency for Science, Technology and Research , Singapore 138667 , Singapore
| | - Jong-Jin Kim
- Center for Self-Assembly and Complexity , Institute for Basic Science (IBS) , Pohang 37673 , Republic of Korea
| | - Haw-Young Kwon
- Center for Self-Assembly and Complexity , Institute for Basic Science (IBS) , Pohang 37673 , Republic of Korea
| | - So Hee Im
- Bio &Drug Discovery Division , Korea Research Institute of Chemical Technology Yuseong-Gu , Gajeongro 141 , Daejeon 34114 , Republic of Korea
| | - Myung Ae Bae
- Bio &Drug Discovery Division , Korea Research Institute of Chemical Technology Yuseong-Gu , Gajeongro 141 , Daejeon 34114 , Republic of Korea
| | - Jin Hee Ahn
- Department of Chemistry , Gwangju Institute of Science and Technology (GIST) , Gwangju 61005 , Republic of Korea
| | - Chang Siang Lim
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB) , Agency for Science, Technology and Research (A*STAR) , Singapore 138673 , Singapore
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB) , Agency for Science, Technology and Research (A*STAR) , Singapore 138673 , Singapore.,Department of Biochemistry and Department of Medicine, Yong Loo Lin School of Medicine , National University of Singapore , Singapore 117597 , Singapore
| | - Sunyou Park
- New Drug Discovery Center, DGMIF , Daegu 41061 , Republic of Korea
| | - Sang Eun Kim
- Department of Nuclear Medicine , Seoul National University College of Medicine, Seoul National University Bundang Hospital , Seongnam 13620 , Republic of Korea.,Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology , Seoul National University , Seoul 08826 , Republic of Korea.,Center for Nanomolecular Imaging and Innovative Drug Development , Advanced Institutes of Convergence Technology , Suwon 16229 , Republic of Korea
| | - Byung Chul Lee
- Department of Nuclear Medicine , Seoul National University College of Medicine, Seoul National University Bundang Hospital , Seongnam 13620 , Republic of Korea.,Center for Nanomolecular Imaging and Innovative Drug Development , Advanced Institutes of Convergence Technology , Suwon 16229 , Republic of Korea
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, and BK21 PLUS Future Biopharmaceutical Human Resources Training and Research Team, and Institute of Nano Science & Technology (INST) , Hanyang University , Seoul 04763 , Republic of Korea
| | - Young-Tae Chang
- Laboratory of Bioimaging Probe Development , Singapore Bioimaging Consortium, Agency for Science, Technology and Research , Singapore 138667 , Singapore.,Center for Self-Assembly and Complexity , Institute for Basic Science (IBS) , Pohang 37673 , Republic of Korea
| |
Collapse
|
27
|
Ren S, Laub P, Lu Y, Naganawa M, Carson RE. Atlas-Based Multiorgan Segmentation for Dynamic Abdominal PET. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2020. [DOI: 10.1109/trpms.2019.2926889] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
28
|
Zheng L, Wang Y, Yang B, Zhang B, Wu Y. Islet Transplantation Imaging in vivo. Diabetes Metab Syndr Obes 2020; 13:3301-3311. [PMID: 33061492 PMCID: PMC7520574 DOI: 10.2147/dmso.s263253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 07/29/2020] [Indexed: 12/31/2022] Open
Abstract
Although islet transplantation plays an effective and powerful role in the treatment of diabetes, a large amount of islet grafts are lost at an early stage due to instant blood-mediated inflammatory reactions, immune rejection, and β-cell toxicity resulting from immunosuppressive agents. Timely intervention based on the viability and function of the transplanted islets at an early stage is crucial. Various islet transplantation imaging techniques are available for monitoring the conditions of post-transplanted islets. Due to the development of various imaging modalities and the continuous study of contrast agents, non-invasive islet transplantation imaging in vivo has made great progress. The tracing and functional evaluation of transplanted islets in vivo have thus become possible. However, most studies on contrast agent and imaging modalities are limited to animal experiments, and long-term toxicity and stability need further evaluation. Accordingly, the clinical application of the current achievements still requires a large amount of effort. In this review, we discuss the contrast agents for MRI, SPECT/PET, BLI/FI, US, MPI, PAI, and multimodal imaging. We further summarize the advantages and limitations of various molecular imaging methods.
Collapse
Affiliation(s)
- Lei Zheng
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310000, People’s Republic of China
| | - Yinghao Wang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310000, People’s Republic of China
| | - Bin Yang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310000, People’s Republic of China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310000, People’s Republic of China
| | - Bo Zhang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310000, People’s Republic of China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310000, People’s Republic of China
- Correspondence: Bo Zhang; Yulian Wu Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310000, People’s Republic of China Tel/Fax +86 571 87783563 Email ;
| | - Yulian Wu
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310000, People’s Republic of China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310000, People’s Republic of China
| |
Collapse
|
29
|
Nucleic acid-based theranostics in type 1 diabetes. Transl Res 2019; 214:50-61. [PMID: 31491371 DOI: 10.1016/j.trsl.2019.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/01/2019] [Accepted: 08/17/2019] [Indexed: 12/12/2022]
Abstract
Application of RNAi interference for type 1 diabetes (T1D) therapy bears tremendous potential. This review will discuss vehicles for oligonucleotide delivery, imaging modalities used for delivery monitoring, therapeutic targets, and different theranostic strategies that can be applied for T1D treatment.
Collapse
|
30
|
Fujita N, Fujimoto H, Hamamatsu K, Murakami T, Kimura H, Toyoda K, Saji H, Inagaki N. Noninvasive longitudinal quantification of β-cell mass with [ 111In]-labeled exendin-4. FASEB J 2019; 33:11836-11844. [PMID: 31370679 PMCID: PMC6902711 DOI: 10.1096/fj.201900555rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/09/2019] [Indexed: 01/09/2023]
Abstract
Currently, quantifying β-cell mass (BCM) requires harvesting the pancreas. In this study, we investigated a potential noninvasive method to quantify BCM changes longitudinally using [Lys12(111In-BnDTPA-Ahx)]exendin-4 ([111In]-Ex4) and single-photon emission computed tomography (SPECT). We used autoradiography and transgenic mice expressing green fluorescent protein under the control of mouse insulin 1 gene promotor to evaluate the specificity of [111In]-Ex4 toward β cells. Using nonobese diabetic (NOD) mice, we injected [111In]-Ex4 (3.0 MBq) intravenously and performed SPECT 30 min later, repeating this at a 2-wk interval. After the second scan, we harvested the pancreas and calculated BCM from immunohistochemically stained pancreatic sections. Specific accumulation of [111In]-Ex4 in β cells was confirmed by autoradiography, with a significant correlation (r = 0.94) between the fluorescent and radioactive signal intensities. The radioactive signal from the pancreas in the second SPECT scan significantly correlated (r = 0.89) with BCM calculated from the immunostained pancreatic sections. We developed a regression formula to estimate BCM from the radioactive signals from the pancreas in SPECT scans. BCM can be quantified longitudinally and noninvasively by SPECT imaging with [111In]-Ex4. This technique successfully demonstrated longitudinal changes in BCM in NOD mice before and after onset of hyperglycemia.-Fujita, N., Fujimoto, H., Hamamatsu, K., Murakami, T., Kimura, H., Toyoda, K., Saji, H., Inagaki, N. Noninvasive longitudinal quantification of β-cell mass with [111In]-labeled exendin-4.
Collapse
Affiliation(s)
- Naotaka Fujita
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Fujimoto
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Radioisotope Research Center, Agency for Health, Safety, and Environment, Kyoto University, Kyoto, Japan
| | - Keita Hamamatsu
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takaaki Murakami
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Kimura
- Department of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kentaro Toyoda
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideo Saji
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
31
|
Ren S, Lu Y, Bertolli O, Thielemans K, Carson RE. Event-by-event non-rigid data-driven PET respiratory motion correction methods: comparison of principal component analysis and centroid of distribution. ACTA ACUST UNITED AC 2019; 64:165014. [DOI: 10.1088/1361-6560/ab0bc9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
32
|
Abstract
PURPOSE OF REVIEW Quantitative markers for beta-cell mass (BCM) in human pancreas are currently lacking. Medical imaging using positron emission tomography (PET) markers for beta-cell restricted targets may provide an accurate and non-invasive measurement of BCM, to assist diagnosis and treatment of metabolic disease. GPR44 was recently discovered as a putative marker for beta cells and this review summarizes the developments so far. RECENT FINDINGS Several small molecule binders targeting GPR44 have been radiolabeled for PET imaging and evaluated in vitro and in small and large animal models. 11C-AZ12204657 and 11C-MK-7246 displayed a dose-dependent and GPR44-mediated binding to beta cells both in vitro and in vivo, with negligible uptake in exocrine pancreas. GPR44 represents an attractive target for visualization of BCM. Further progress in radioligand development including clinical testing is expected to clarify the role of GPR44 as a surrogate marker for BCM in humans.
Collapse
Affiliation(s)
- Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Dag Hammarskjölds väg 14C, 3tr, SE-752 37, Uppsala, Sweden.
- Antaros Medical AB, Mölndal, Sweden.
| |
Collapse
|
33
|
Wu MR, Hsiao JK, Liu HM, Huang YY, Tseng YJ, Chou PT, Weng TI, Yang CY. In vivo imaging of insulin-secreting human pancreatic ductal cells using MRI reporter gene technique: A feasibility study. Magn Reson Med 2019; 82:763-774. [PMID: 30957300 DOI: 10.1002/mrm.27749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE The purpose of this study was to investigate the feasibility of in vivo imaging of human pancreatic ductal cells by OATP1B3 reporter gene under MRI. METHODS A human cell line (PANC-1) derived from the pancreatic ductal epithelium was used in this study. After transduction of OATP1B3, the cellular physiological functions and the ability of intracellular uptake of the MRI contrast medium (Gd-EOB-DTPA) were examined. Induced differentiation of the PANC-1 cells into hormone-secreting cells were performed to simulate pancreatic β-like cells. The hormone-secreting cells were implanted into rats and in vivo MRI was evaluated. RESULTS The mRNA and proteins of OATP1B3 were highly expressed. No significant change of cellular physiological functions was found after the expression. After induced differentiation, the hormone secretion capacities of the OATP1B3-expressing PANC-1 cells were confirmed. Intra-cellular uptake of Gd-EOB-DTPA was determined in vitro by inductively coupled plasma mass spectrometry and MRI. In vivo MRI of the OATP1B3-expressing xenograft revealed an increased signal intensity after contrast enhancement. CONCLUSION OATP1B3 can be used as a safe and feasible in vivo MRI gene reporter for human pancreatic ductal cells.
Collapse
Affiliation(s)
- Menq-Rong Wu
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan.,Department of Medical Imaging, Buddhist Tzu Chi General Hospital, Taipei Branch, New Taipei city, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Buddhist Tzu Chi General Hospital, Taipei Branch, New Taipei city, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Hon-Man Liu
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan.,Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Radiology and Medical Imaging, Fu-Jen Catholic University and Hospital, New Taipei City, Taiwan
| | - Yi-You Huang
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Yu-Jui Tseng
- Department of Chemistry and Center for Emerging Material and Advanced Devices, National Taiwan University, Taipei, Taiwan
| | - Pi-Tai Chou
- Department of Chemistry and Center for Emerging Material and Advanced Devices, National Taiwan University, Taipei, Taiwan
| | - Te-I Weng
- Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Yi Yang
- Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Medical Imaging, E-Da Hospital, I-Shou University, Kaohsiung City, Taiwan.,School of Medicine for International Students, I-Shou University, Kaohsiung City, Taiwan
| |
Collapse
|
34
|
Naganawa M, Lim K, Nabulsi NB, Lin SF, Labaree D, Ropchan J, Herold KC, Huang Y, Harris P, Ichise M, Cline GW, Carson RE. Evaluation of Pancreatic VMAT2 Binding with Active and Inactive Enantiomers of [ 18F]FP-DTBZ in Healthy Subjects and Patients with Type 1 Diabetes. Mol Imaging Biol 2019; 20:835-845. [PMID: 29468404 DOI: 10.1007/s11307-018-1170-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Previous studies demonstrated the utility of [18F]fluoropropyl-(+)-dihydrotetrabenazine ([18F]FP-(+)-DTBZ) as a positron emission tomography (PET) radiotracer for the vesicular monoamine transporter type 2 (VMAT2) to quantify beta cell mass in healthy control (HC) and type 1 diabetes mellitus (T1DM) groups. Quantification of specific binding requires measurement of non-displaceable uptake. Our goal was to identify a reference tissue (renal cortex or spleen) to quantify pancreatic non-specific binding of [18F]FP-(+)-DTBZ with the inactive enantiomer, [18F]FP-(-)-DTBZ. This was the first human study of [18F]FP-(-)-DTBZ. PROCEDURES Six HCs and four T1DM patients were scanned on separate days after injection of [18F]FP-(+)-DTBZ or [18F]FP-(-)-DTBZ. Distribution volumes (VT) and standardized uptake values (SUVs) were compared between groups. Three methods for calculation of non-displaceable uptake (VND) or reference SUV were applied: (1) use of [18F]FP-(+)-DTBZ reference VT as VND, assuming VND is uniform across organs; (2) use of [18F]FP-(-)-DTBZ pancreatic VT as VND, assuming that VND is uniform between enantiomers in the pancreas; and (3) use of a scaled [18F]FP-(+)-DTBZ reference VT as VND, assuming that a ratio of non-displaceable uptake between organs is uniform between enantiomers. Group differences in VT (or SUV), binding potential (BPND), or SUV ratio (SUVR) were estimated using these three methods. RESULTS [18F]FP-(-)-DTBZ VT values were different among organs, and VT(+) and VT(-) were also different in the renal cortex and spleen. Method 3 with the spleen to estimate VND (or reference SUV) gave the highest non-displaceable uptake and the largest HC vs. T1DM group differences. Significant group differences were also observed in VT (or SUV) with method 1 using spleen. SUV was affected by differences in the input function between groups and between enantiomers. CONCLUSIONS Non-displaceable uptake was different among organs and between enantiomers. Use of scaled spleen VT values for VND is a suitable method for quantification of VMAT2 in the pancreas.
Collapse
Affiliation(s)
- Mika Naganawa
- Yale University, P.O. Box 208048, New Haven, CT, 06520-8048, USA.
| | - Keunpoong Lim
- Yale University, P.O. Box 208048, New Haven, CT, 06520-8048, USA
| | - Nabeel B Nabulsi
- Yale University, P.O. Box 208048, New Haven, CT, 06520-8048, USA
| | - Shu-Fei Lin
- Yale University, P.O. Box 208048, New Haven, CT, 06520-8048, USA
| | - David Labaree
- Yale University, P.O. Box 208048, New Haven, CT, 06520-8048, USA
| | - Jim Ropchan
- Yale University, P.O. Box 208048, New Haven, CT, 06520-8048, USA
| | - Kevan C Herold
- Yale University, P.O. Box 208048, New Haven, CT, 06520-8048, USA
| | - Yiyun Huang
- Yale University, P.O. Box 208048, New Haven, CT, 06520-8048, USA
| | | | | | - Gary W Cline
- Yale University, P.O. Box 208048, New Haven, CT, 06520-8048, USA
| | - Richard E Carson
- Yale University, P.O. Box 208048, New Haven, CT, 06520-8048, USA
| |
Collapse
|
35
|
Abstract
The clinical onset of type 1 diabetes is characterized by the destruction of the insulin-producing β cells of the pancreas and is caused by autoantigen-induced inflammation (insulitis) of the islets of Langerhans. The current standard of care for type 1 diabetes mellitus patients allows for management of the disease with exogenous insulin, but patients eventually succumb to many chronic complications such as limb amputation, blindness, and kidney failure. New therapeutic approaches now on the horizon are looking beyond glycemic management and are evaluating new strategies from protecting and regenerating endogenous islets to treating the underlying autoimmunity through selective modulation of key immune cell populations. Currently, there are no effective treatments for the autoimmunity that causes the disease, and strategies that aim to delay or prevent the onset of the disease will play an important role in the future of diabetes research. In this review, we summarize many of the key efforts underway that utilize molecular approaches to selectively modulate this disease and look at new therapeutic paradigms that can transform clinical treatment.
Collapse
Affiliation(s)
- Daniel Sheehy
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Sean Quinnell
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Arturo J. Vegas
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| |
Collapse
|
36
|
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
|
37
|
Cline GW, Naganawa M, Chen L, Chidsey K, Carvajal-Gonzalez S, Pawlak S, Rossulek M, Zhang Y, Bini J, McCarthy TJ, Carson RE, Calle RA. Decreased VMAT2 in the pancreas of humans with type 2 diabetes mellitus measured in vivo by PET imaging. Diabetologia 2018; 61:2598-2607. [PMID: 29721633 DOI: 10.1007/s00125-018-4624-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/29/2018] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS The progressive loss of beta cell function is part of the natural history of type 2 diabetes. Autopsy studies suggest that this is, in part, due to loss of beta cell mass (BCM), but this has not been confirmed in vivo. Non-invasive methods to quantify BCM may contribute to a better understanding of type 2 diabetes pathophysiology and the development of therapeutic strategies. In humans, the localisation of vesicular monoamine transporter type 2 (VMAT2) in beta cells and pancreatic polypeptide cells, with minimal expression in other exocrine or endocrine pancreatic cells, has led to its development as a measure of BCM. We used the VMAT2 tracer [18F]fluoropropyl-(+)-dihydrotetrabenazine to quantify BCM in humans with impaired glucose tolerance (prediabetes) or type 2 diabetes, and in healthy obese volunteers (HOV). METHODS Dynamic positron emission tomography (PET) data were obtained for 4 h with metabolite-corrected arterial blood measurement in 16 HOV, five prediabetic and 17 type 2 diabetic participants. Eleven participants (six HOV and five with type 2 diabetes) underwent two abdominal PET/computed tomography (CT) scans for the assessment of test-retest variability. Standardised uptake value ratio (SUVR) was calculated in pancreatic subregions (head, body and tail), with the spleen as a reference region to determine non-specific tracer uptake at 3-4 h. The outcome measure SUVR minus 1 (SUVR-1) accounts for non-specific tracer uptake. Functional beta cell capacity was assessed by C-peptide release following standard (arginine stimulus test [AST]) and acute insulin response to the glucose-enhanced AST (AIRargMAX). Pearson correlation analysis was performed between the binding variables and the C-peptide AUC post-AST and post-AIRargMAX. RESULTS Absolute test-retest variability (aTRV) was ≤15% for all regions. Variability and overlap of SUVR-1 was measured in all groups; HOV and participants with prediabetes and with type 2 diabetes. SUVR-1 showed significant positive correlations with AIRargMAX (all groups) in all pancreas subregions (whole pancreas p = 0.009 and pancreas head p = 0.009; body p = 0.019 and tail p = 0.023). SUVR-1 inversely correlated with HbA1c (all groups) in the whole pancreas (p = 0.033) and pancreas head (p = 0.008). SUVR-1 also inversely correlated with years since diagnosis of type 2 diabetes in the pancreas head (p = 0.049) and pancreas tail (p = 0.035). CONCLUSIONS/INTERPRETATION The observed correlations of VMAT2 density in the pancreas and pancreas regions with years since diagnosis of type 2 diabetes, glycaemic control and beta cell function suggest that loss of BCM contributes to deficient insulin secretion in humans with type 2 diabetes.
Collapse
Affiliation(s)
- Gary W Cline
- Yale University, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520, USA.
| | - Mika Naganawa
- Yale University, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520, USA
| | | | | | | | | | | | | | - Jason Bini
- Yale University, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520, USA
| | | | - Richard E Carson
- Yale University, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520, USA
| | | |
Collapse
|
38
|
Cline GW, McCarthy TJ, Carson RE, Calle RA. Clinical and scientific value in the pursuit of quantification of beta cells in the pancreas by PET imaging. Diabetologia 2018; 61:2671-2673. [PMID: 30136144 PMCID: PMC6219921 DOI: 10.1007/s00125-018-4718-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Gary W Cline
- Yale University, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520, USA.
| | | | - Richard E Carson
- Yale University, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520, USA
| | | |
Collapse
|
39
|
Roh E, Kim KM, Park KS, Kim YJ, Chun EJ, Choi SH, Park KS, Jang HC, Lim S. Comparison of pancreatic volume and fat amount linked with glucose homeostasis between healthy Caucasians and Koreans. Diabetes Obes Metab 2018; 20:2642-2652. [PMID: 29934972 DOI: 10.1111/dom.13447] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/13/2018] [Accepted: 06/20/2018] [Indexed: 02/06/2023]
Abstract
AIM To compare pancreatic volume and fat amount, and their associations with glucose homeostasis, in a Korean and a white population. MATERIALS AND METHODS In 43 healthy Korean and 43 healthy white people, matched for age (±3 years) and body mass index (BMI; ±1 kg/m2 ), we measured pancreatic volume and fat amount in the pancreas and abdomen using computed tomography. Pancreatic β-cell function and insulin resistance were estimated according to biochemical characteristics and a 75-g oral glucose tolerance test. Body composition and resting energy expenditure (REE) were examined using bioimpedance and indirect calorimetry, respectively. RESULTS The mean ±SD age of the participants was 29.9 ± 5.9 years and 30.0 ± 5.2 years, and BMI was 24.0 ±3.7 and 24.1 ±3.2 kg/m2 in the white participants and the Korean participants, respectively. Pancreatic volume in the white participants was greater than that in Korean participants (77.8 ±11.6 vs 68.2 ±12.1 cm3 ; P < .001). Pancreatic fat content in Korean participants was 22.8% higher than in white participants (P = .051). Insulinogenic index, disposition index, muscle mass and REE were significantly lower in Korean participants. Pancreatic volume was positively associated with indices linked to β-cell function; fat content in the pancreas was negatively associated with such indices, and positively with insulin resistance after adjusting for relevant variables including REE. CONCLUSIONS A smaller pancreas and higher fat deposition might be crucial determinants of vulnerability to diabetes in Korean people compared with white people with similar BMI and body fat levels.
Collapse
Affiliation(s)
- Eun Roh
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, South Korea
| | - Kyoung M Kim
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Kyeong S Park
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine, Daerim Saint Mary's Hospital, Seoul, South Korea
| | - Yoon J Kim
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine, Mediplex Sejong Hospital, Incheon, South Korea
| | - Eun J Chun
- Department of Radiology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Sung H Choi
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Kyong S Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Hak C Jang
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, South Korea
| |
Collapse
|
40
|
Templin AT, Meier DT, Willard JR, Wolden-Hanson T, Conway K, Lin YG, Gillespie PJ, Bokvist KB, Attardo G, Kahn SE, Scheuner D, Hull RL. Use of the PET ligand florbetapir for in vivo imaging of pancreatic islet amyloid deposits in hIAPP transgenic mice. Diabetologia 2018; 61:2215-2224. [PMID: 30046852 PMCID: PMC6119478 DOI: 10.1007/s00125-018-4695-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/19/2018] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS Islet amyloid deposits contribute to beta cell dysfunction and death in most individuals with type 2 diabetes but non-invasive methods to determine the presence of these pathological protein aggregates are currently not available. Therefore, we examined whether florbetapir, a radiopharmaceutical agent used for detection of amyloid-β deposits in the brain, also allows identification of islet amyloid in the pancreas. METHODS Saturation binding assays were used to determine the affinity of florbetapir for human islet amyloid polypeptide (hIAPP) aggregates in vitro. Islet amyloid-prone transgenic mice that express hIAPP in their beta cells and amyloid-free non-transgenic control mice were used to examine the ability of florbetapir to detect islet amyloid deposits in vitro, in vivo and ex vivo. Mice or mouse pancreases were subjected to autoradiographic, histochemical and/or positron emission tomography (PET) analyses to assess the utility of florbetapir in identifying islet amyloid. RESULTS In vitro, florbetapir bound synthetic hIAPP fibrils with a dissociation constant of 7.9 nmol/l. Additionally, florbetapir bound preferentially to amyloid-containing hIAPP transgenic vs amyloid-free non-transgenic mouse pancreas sections in vitro, as determined by autoradiography (16,475 ± 5581 vs 5762 ± 575 density/unit area, p < 0.05). In hIAPP transgenic and non-transgenic mice fed a high-fat diet for 1 year, intravenous administration of florbetapir followed by PET scanning showed that the florbetapir signal was significantly higher in amyloid-laden hIAPP transgenic vs amyloid-free non-transgenic pancreases in vivo during the first 5 min of the scan (36.83 ± 2.22 vs 29.34 ± 2.03 standardised uptake value × min, p < 0.05). Following PET, pancreases were excised and florbetapir uptake was determined ex vivo by γ counting. Pancreatic uptake of florbetapir was significantly correlated with the degree of islet amyloid deposition, the latter assessed by histochemistry (r = 0.74, p < 0.001). CONCLUSIONS/INTERPRETATION Florbetapir binds to islet amyloid deposits in a specific and quantitative manner. In the future, florbetapir may be useful as a non-invasive tool to identify islet amyloid deposits in humans.
Collapse
Affiliation(s)
- Andrew T Templin
- Department of Medicine, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA.
- University of Washington, Seattle, WA, USA.
| | - Daniel T Meier
- Department of Medicine, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA
- University of Washington, Seattle, WA, USA
| | - Joshua R Willard
- Department of Medicine, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA
- University of Washington, Seattle, WA, USA
| | - Tami Wolden-Hanson
- Department of Medicine, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - Kelly Conway
- Avid Radiopharmaceuticals, Inc., Philadelphia, PA, USA
| | - Yin-Guo Lin
- Avid Radiopharmaceuticals, Inc., Philadelphia, PA, USA
| | | | | | | | - Steven E Kahn
- Department of Medicine, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA
- University of Washington, Seattle, WA, USA
| | | | - Rebecca L Hull
- Department of Medicine, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA
- University of Washington, Seattle, WA, USA
| |
Collapse
|
41
|
Brom M, Joosten L, Frielink C, Peeters H, Bos D, van Zanten M, Boerman O, Gotthardt M. Validation of 111In-Exendin SPECT for the Determination of the β-Cell Mass in BioBreeding Diabetes-Prone Rats. Diabetes 2018; 67:2012-2018. [PMID: 30045920 DOI: 10.2337/db17-1312] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 07/12/2018] [Indexed: 11/13/2022]
Abstract
The changes in β-cell mass (BCM) during the development and progression of diabetes could potentially be measured by radionuclide imaging using radiolabeled exendin. In this study, we investigated the potential of 111In-diethylenetriaminepentaacetic acid-exendin-3 (111In-exendin) in a rat model that closely mimics the development of type 1 diabetes (T1D) in humans: BioBreeding diabetes-prone (BBDP) rats. BBDP rats of 4-18 weeks of age were injected intravenously with 111In-exendin, and single-photon emission computed tomography (SPECT) images were acquired. The accumulation of the radiotracer was measured as well as the BCM and grade of insulitis by histology. 111In-exendin accumulated specifically in the islets, resulting in a linear correlation with the BCM (%) (Pearson r = 0.89, P < 0.0001, and r = 0.64 for SPECT). Insulitis did not have an influence on this correlation. These results indicate that 111In-exendin is a promising tracer to determine the BCM during the development of T1D, irrespective of the degree of insulitis.
Collapse
Affiliation(s)
- Maarten Brom
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lieke Joosten
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cathelijne Frielink
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hanneke Peeters
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Desirée Bos
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Monica van Zanten
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Otto Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Martin Gotthardt
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
42
|
What Can Be and What Cannot Be Accomplished With PET: Rectifying Ongoing Misconceptions. Clin Nucl Med 2018; 42:603-605. [PMID: 28570374 DOI: 10.1097/rlu.0000000000001695] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
43
|
Lu Y, Fontaine K, Mulnix T, Onofrey JA, Ren S, Panin V, Jones J, Casey ME, Barnett R, Kench P, Fulton R, Carson RE, Liu C. Respiratory Motion Compensation for PET/CT with Motion Information Derived from Matched Attenuation-Corrected Gated PET Data. J Nucl Med 2018; 59:1480-1486. [PMID: 29439015 DOI: 10.2967/jnumed.117.203000] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/25/2018] [Indexed: 11/16/2022] Open
Abstract
Respiratory motion degrades the detection and quantification capabilities of PET/CT imaging. Moreover, mismatch between a fast helical CT image and a time-averaged PET image due to respiratory motion results in additional attenuation correction artifacts and inaccurate localization. Current motion compensation approaches typically have 3 limitations: the mismatch among respiration-gated PET images and the CT attenuation correction (CTAC) map can introduce artifacts in the gated PET reconstructions that can subsequently affect the accuracy of the motion estimation; sinogram-based correction approaches do not correct for intragate motion due to intracycle and intercycle breathing variations; and the mismatch between the PET motion compensation reference gate and the CT image can cause an additional CT-mismatch artifact. In this study, we established a motion correction framework to address these limitations. Methods: In the proposed framework, the combined emission-transmission reconstruction algorithm was used for phase-matched gated PET reconstructions to facilitate the motion model building. An event-by-event nonrigid respiratory motion compensation method with correlations between internal organ motion and external respiratory signals was used to correct both intracycle and intercycle breathing variations. The PET reference gate was automatically determined by a newly proposed CT-matching algorithm. We applied the new framework to 13 human datasets with 3 different radiotracers and 323 lesions and compared its performance with CTAC and non-attenuation correction (NAC) approaches. Validation using 4-dimensional CT was performed for one lung cancer dataset. Results: For the 10 18F-FDG studies, the proposed method outperformed (P < 0.006) both the CTAC and the NAC methods in terms of region-of-interest-based SUVmean, SUVmax, and SUV ratio improvements over no motion correction (SUVmean: 19.9% vs. 14.0% vs. 13.2%; SUVmax: 15.5% vs. 10.8% vs. 10.6%; SUV ratio: 24.1% vs. 17.6% vs. 16.2%, for the proposed, CTAC, and NAC methods, respectively). The proposed method increased SUV ratios over no motion correction for 94.4% of lesions, compared with 84.8% and 86.4% using the CTAC and NAC methods, respectively. For the 2 18F-fluoropropyl-(+)-dihydrotetrabenazine studies, the proposed method reduced the CT-mismatch artifacts in the lower lung where the CTAC approach failed and maintained the quantification accuracy of bone marrow where the NAC approach failed. For the 18F-FMISO study, the proposed method outperformed both the CTAC and the NAC methods in terms of motion estimation accuracy at 2 lung lesion locations. Conclusion: The proposed PET/CT respiratory event-by-event motion-correction framework with motion information derived from matched attenuation-corrected PET data provides image quality superior to that of the CTAC and NAC methods for multiple tracers.
Collapse
Affiliation(s)
- Yihuan Lu
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Kathryn Fontaine
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Tim Mulnix
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - John A Onofrey
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Silin Ren
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | | | - Judson Jones
- Siemens Medical Solutions, Knoxville, Tennessee; and
| | | | - Robert Barnett
- Discipline of Medical Radiation Sciences, Faculty of Health Sciences, University of Sydney, Sydney, Australia
| | - Peter Kench
- Discipline of Medical Radiation Sciences, Faculty of Health Sciences, University of Sydney, Sydney, Australia
| | - Roger Fulton
- Discipline of Medical Radiation Sciences, Faculty of Health Sciences, University of Sydney, Sydney, Australia
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut.,Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Chi Liu
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut.,Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| |
Collapse
|
44
|
Chan C, Onofrey J, Jian Y, Germino M, Papademetris X, Carson RE, Liu C. Non-Rigid Event-by-Event Continuous Respiratory Motion Compensated List-Mode Reconstruction for PET. IEEE TRANSACTIONS ON MEDICAL IMAGING 2018; 37:504-515. [PMID: 29028189 PMCID: PMC7304524 DOI: 10.1109/tmi.2017.2761756] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Respiratory motion during positron emission tomography (PET)/computed tomography (CT) imaging can cause significant image blurring and underestimation of tracer concentration for both static and dynamic studies. In this paper, with the aim to eliminate both intra-cycle and inter-cycle motions, and apply to dynamic imaging, we developed a non-rigid event-by-event (NR-EBE) respiratory motion-compensated list-mode reconstruction algorithm. The proposed method consists of two components: the first component estimates a continuous non-rigid motion field of the internal organs using the internal-external motion correlation. This continuous motion field is then incorporated into the second component, non-rigid MOLAR (NR-MOLAR) reconstruction algorithm to deform the system matrix to the reference location where the attenuation CT is acquired. The point spread function (PSF) and time-of-flight (TOF) kernels in NR-MOLAR are incorporated in the system matrix calculation, and therefore are also deformed according to motion. We first validated NR-MOLAR using a XCAT phantom with a simulated respiratory motion. NR-EBE motion-compensated image reconstruction using both the components was then validated on three human studies injected with 18F-FPDTBZ and one with 18F-fluorodeoxyglucose (FDG) tracers. The human results were compared with conventional non-rigid motion correction using discrete motion field (NR-discrete, one motion field per gate) and a previously proposed rigid EBE motion-compensated image reconstruction (R-EBE) that was designed to correct for rigid motion on a target lesion/organ. The XCAT results demonstrated that NR-MOLAR incorporating both PSF and TOF kernels effectively corrected for non-rigid motion. The 18F-FPDTBZ studies showed that NR-EBE out-performed NR-Discrete, and yielded comparable results with R-EBE on target organs while yielding superior image quality in other regions. The FDG study showed that NR-EBE clearly improved the visibility of multiple moving lesions in the liver where some of them could not be discerned in other reconstructions, in addition to improving quantification. These results show that NR-EBE motion-compensated image reconstruction appears to be a promising tool for lesion detection and quantification when imaging thoracic and abdominal regions using PET.
Collapse
|
45
|
Bini J, Naganawa M, Nabulsi N, Huang Y, Ropchan J, Lim K, Najafzadeh S, Herold KC, Cline GW, Carson RE. Evaluation of PET Brain Radioligands for Imaging Pancreatic β-Cell Mass: Potential Utility of 11C-(+)-PHNO. J Nucl Med 2018; 59:1249-1254. [PMID: 29371405 DOI: 10.2967/jnumed.117.197285] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 12/07/2017] [Indexed: 12/21/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is characterized by a loss of β-cells in the islets of Langerhans of the pancreas and subsequent deficient insulin secretion in response to hyperglycemia. Development of an in vivo test to measure β-cell mass (BCM) would greatly enhance the ability to track diabetes therapies. β-cells and neurologic tissues have common cellular receptors and transporters, therefore, we screened brain radioligands for their ability to identify β-cells. Methods: We examined a β-cell gene atlas for endocrine pancreas receptor targets and cross-referenced these targets with brain radioligands that were available at our institution. Twelve healthy control subjects and 2 T1DM subjects underwent dynamic PET/CT scans with 6 tracers. Results: The D2/D3 receptor agonist radioligand 11C-(+)-4-propyl-9-hydroxynaphthoxazine (PHNO) was the only radioligand to demonstrate sustained uptake in the pancreas with high contrast versus abdominal organs such as the kidneys, liver, and spleen, based on the first 30 min of data. Mean SUV from 20 to 30 min demonstrated high uptake of 11C-(+)-PHNO in healthy controls (SUV, 13.8) with a 71% reduction in a T1DM subject with undetectable levels of C-peptide (SUV, 4.0) and a 20% reduction in a T1DM subject with fasting C-peptide level of 0.38 ng/mL (SUV, 11.0). SUV in abdominal organs outside the pancreas did not show measurable differences between the control and T1DM subjects, suggesting that the changes in SUV of 11C-(+)-PHNO may be specific to changes in the pancreas between healthy controls and T1DM subjects. When D3 and D2 antagonists were used in nonhuman primates, specific pancreatic binding (SUVR-1) of 11C-PHNO was reduced by 57% and 38%, respectively. Conclusion:11C-(+)-PHNO is a potential marker of BCM, with 2:1 binding of D3 receptors over D2 receptors. Further in vitro and in vivo studies to establish D2/D3 receptor specificity to β-cells is warranted to characterize 11C-(+)-PHNO as a candidate for clinical measurement of BCM in healthy control and diabetic subjects.
Collapse
Affiliation(s)
- Jason Bini
- PET Center, Yale University School of Medicine, New Haven, CT .,Department of Biomedical Engineering, Yale University, New Haven, CT; and
| | - Mika Naganawa
- PET Center, Yale University School of Medicine, New Haven, CT
| | - Nabeel Nabulsi
- PET Center, Yale University School of Medicine, New Haven, CT
| | - Yiyun Huang
- PET Center, Yale University School of Medicine, New Haven, CT
| | - Jim Ropchan
- PET Center, Yale University School of Medicine, New Haven, CT
| | - Keunpoong Lim
- PET Center, Yale University School of Medicine, New Haven, CT
| | | | - Kevan C Herold
- Department of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Gary W Cline
- Department of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Richard E Carson
- PET Center, Yale University School of Medicine, New Haven, CT.,Department of Biomedical Engineering, Yale University, New Haven, CT; and
| |
Collapse
|
46
|
Liu F, Choi SR, Zha Z, Ploessl K, Zhu L, Kung HF. Deuterated 18F-9-O-hexadeutero-3-fluoropropoxyl-(+)-dihydrotetrabenazine (D6-FP-(+)-DTBZ): A vesicular monoamine transporter 2 (VMAT2) imaging agent. Nucl Med Biol 2017; 57:42-49. [PMID: 29306111 DOI: 10.1016/j.nucmedbio.2017.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/21/2017] [Accepted: 11/28/2017] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Vesicular monoamine transporters 2 (VMAT2) in the brain serve as transporter for packaging monoamine in vesicles for normal CNS neurotransmission. Several VMAT2 imaging agents, [11C]-(+)-DTBZ, dihydrotetrabenazine and [18F]FP-(+)-DTBZ (9-O-fluoropropyl-(+)-dihydro tetrabenazine, a.k.a. [18F]AV-133), are useful for studying the changes in brain function related to monoamine transmission by in vivo imaging. Deuterated analogs have been reported targeting VMAT2 binding sites. METHODS A novel deuterated [18F]9-O-hexaduterofluoropropyl-(+)-dihydrotetrabenazine, [18F]D6-FP-(+)-DTBZ, [18F]1, was prepared as a VMAT2 imaging agent. This 18F agent which targeted VMAT2 was evaluated by in vitro binding, in vivo biodistribution and microPET imaging studies in rodents. RESULTS The one step radiolabeling reaction led to the desired [18F]D6-FP-(+)-DTBZ, [18F]1, which showed excellent binding affinity to VMAT2 (Ki=0.32±0.07nM) comparable to that of FP-(+)-DTBZ (Ki=0.33±0.02nM) using [18F]FP-(+)-DTBZ and rat striatum membrane homogenates. In vivo biodistribution in normal rats showed that 1, exhibited excellent brain uptake and comparable high ratio of striatum to cerebellum (target/background) ratio at 1h after injection (ratio of 6.05±0.43 vs 5.66±0.72 for [18F]FP-(+)-DTBZ vs [18F]1, respectively). MicroPET imaging studies in rats further confirm that the striatum with high VMAT2 concentration was clearly delineated in normal rat brain after iv injection of [18F]1. We observed minor changes of metabolism in rat plasma between these two agents; however, the changes showed little effect on regional brain uptake and retention. CONCLUSIONS The results reported here lend support for using [18F]D6-FP-(+)-DTBZ, [18F]1, as in vivo PET imaging agent for VMAT2 binding in the brain.
Collapse
Affiliation(s)
- Futao Liu
- Key Laboratory of Radiopharmaceuticals, Beijing Normal University, Ministry of Education, Beijing 100875, PR China; Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seok Rye Choi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Five Eleven Pharma Inc., Philadelphia, PA 19104, USA
| | - Zhihao Zha
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karl Ploessl
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Five Eleven Pharma Inc., Philadelphia, PA 19104, USA
| | - Lin Zhu
- Key Laboratory of Radiopharmaceuticals, Beijing Normal University, Ministry of Education, Beijing 100875, PR China; Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hank F Kung
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Five Eleven Pharma Inc., Philadelphia, PA 19104, USA.
| |
Collapse
|
47
|
Yang CT, Ghosh KK, Padmanabhan P, Langer O, Liu J, Halldin C, Gulyás BZ. PET probes for imaging pancreatic islet cells. Clin Transl Imaging 2017. [DOI: 10.1007/s40336-017-0251-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
48
|
Kimura H, Fujita N, Kanbe K, Matsuda H, Watanabe H, Arimitsu K, Fujimoto H, Hamamatsu K, Yagi Y, Ono M, Inagaki N, Saji H. Synthesis and biological evaluation of an 111In-labeled exendin-4 derivative as a single-photon emission computed tomography probe for imaging pancreatic β-cells. Bioorg Med Chem 2017; 25:5772-5778. [PMID: 28927802 DOI: 10.1016/j.bmc.2017.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 11/19/2022]
Abstract
A non-invasive method of pancreatic β-cell mass measurement is needed to enhance our understanding of the pathogenesis of diabetes, facilitate the early diagnosis of this disease, and promote the development of novel therapeutics. Here, we described the synthesis of a novel indium-111 (111In) exendin-4 derivative, [Lys12(In-BnDTPA-Ahx)]exendin-4, through a process involving isothiocyanate-benzyl-DTPA (BnDTPA) and 6-aminohexanoic acid (Ahx) attached to an ɛ-amino group at the lysine-12 residue. We further evaluated the potential use of this derivative as a SPECT probe for pancreatic β-cell imaging. An in vitro binding assay revealed that [Lys12(natIn-BnDTPA-Ahx)]exendin-4 has a high affinity for GLP-1 receptors (IC50=0.43nM). In biodistribution experiments involving normal mice, high [Lys12(111In-BnDTPA-Ahx)]exendin-4 uptake was observed in the pancreas (21.8 ± 4.0%ID/g) and was maintained for 2h after injection. Pre-injection of excess exendin(9-39) markedly reduced the pancreatic uptake of [Lys12(111In-BnDTPA-Ahx)]exendin-4 (95.2%), indicating that the uptake of this tracer is specific and mediated by GLP-1 receptors. Ex vivo autoradiography experiments involving pancreatic sections from MIP-GFP mice confirmed the accumulation of [Lys12(111In-BnDTPA-Ahx)]exendin-4 in pancreatic β-cells. Finally, in mice, [Lys12(111In-BnDTPA-Ahx)]exendin-4 SPECT/CT yielded clear images of the pancreas at 30min post-injection. In conclusion, SPECT with [Lys12(111In-BnDTPA-Ahx)]exendin-4 enables to visualize β-cells in vivo non-invasively.
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.
| | - Naotaka Fujita
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kaori Kanbe
- Department of Patho-Functional Bioanalysis, Kyoto University Graduate School of Pharmaceutical Sciences, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hirokazu Matsuda
- Department of Patho-Functional Bioanalysis, Kyoto University Graduate School of Pharmaceutical Sciences, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan; Research & Development Division, Arkray, Inc., Yousuien-nai, 59 Gansuin-cho, Kamigyo-ku, Kyoto 602-0008, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Kyoto University Graduate School of Pharmaceutical Sciences, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, 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
| | - Hiroyuki Fujimoto
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Keita Hamamatsu
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto University, 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
| | - 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
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto University, 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
|
49
|
Chen C, Cohrs CM, Stertmann J, Bozsak R, Speier S. Human beta cell mass and function in diabetes: Recent advances in knowledge and technologies to understand disease pathogenesis. Mol Metab 2017; 6:943-957. [PMID: 28951820 PMCID: PMC5605733 DOI: 10.1016/j.molmet.2017.06.019] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/31/2017] [Accepted: 06/07/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Plasma insulin levels are predominantly the product of the morphological mass of insulin producing beta cells in the pancreatic islets of Langerhans and the functional status of each of these beta cells. Thus, deficiency in either beta cell mass or function, or both, can lead to insufficient levels of insulin, resulting in hyperglycemia and diabetes. Nonetheless, the precise contribution of beta cell mass and function to the pathogenesis of diabetes as well as the underlying mechanisms are still unclear. In the past, this was largely due to the restricted number of technologies suitable for studying the scarcely accessible human beta cells. However, in recent years, a number of new platforms have been established to expand the available techniques and to facilitate deeper insight into the role of human beta cell mass and function as cause for diabetes and as potential treatment targets. SCOPE OF REVIEW This review discusses the current knowledge about contribution of human beta cell mass and function to different stages of type 1 and type 2 diabetes pathogenesis. Furthermore, it highlights standard and newly developed technological platforms for the study of human beta cell biology, which can be used to increase our understanding of beta cell mass and function in human glucose homeostasis. MAJOR CONCLUSIONS In contrast to early disease models, recent studies suggest that in type 1 and type 2 diabetes impairment of beta cell function is an early feature of disease pathogenesis while a substantial decrease in beta cell mass occurs more closely to clinical manifestation. This suggests that, in addition to beta cell mass replacement for late stage therapies, the development of novel strategies for protection and recovery of beta cell function could be most promising for successful diabetes treatment and prevention. The use of today's developing and wide range of technologies and platforms for the study of human beta cells will allow for a more detailed investigation of the underlying mechanisms and will facilitate development of treatment approaches to specifically target human beta cell mass and function.
Collapse
Affiliation(s)
- Chunguang Chen
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, München-Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Christian M. Cohrs
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, München-Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Julia Stertmann
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, München-Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Robert Bozsak
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, München-Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Stephan Speier
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, München-Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
50
|
Ren S, Jin X, Chan C, Jian Y, Mulnix T, Liu C, Carson RE. Data-driven event-by-event respiratory motion correction using TOF PET list-mode centroid of distribution. Phys Med Biol 2017; 62:4741-4755. [PMID: 28520558 DOI: 10.1088/1361-6560/aa700c] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Data-driven respiratory gating techniques were developed to correct for respiratory motion in PET studies, without the help of external motion tracking systems. Due to the greatly increased image noise in gated reconstructions, it is desirable to develop a data-driven event-by-event respiratory motion correction method. In this study, using the Centroid-of-distribution (COD) algorithm, we established a data-driven event-by-event respiratory motion correction technique using TOF PET list-mode data, and investigated its performance by comparing with an external system-based correction method. Ten human scans with the pancreatic β-cell tracer 18F-FP-(+)-DTBZ were employed. Data-driven respiratory motions in superior-inferior (SI) and anterior-posterior (AP) directions were first determined by computing the centroid of all radioactive events during each short time frame with further processing. The Anzai belt system was employed to record respiratory motion in all studies. COD traces in both SI and AP directions were first compared with Anzai traces by computing the Pearson correlation coefficients. Then, respiratory gated reconstructions based on either COD or Anzai traces were performed to evaluate their relative performance in capturing respiratory motion. Finally, based on correlations of displacements of organ locations in all directions and COD information, continuous 3D internal organ motion in SI and AP directions was calculated based on COD traces to guide event-by-event respiratory motion correction in the MOLAR reconstruction framework. Continuous respiratory correction results based on COD were compared with that based on Anzai, and without motion correction. Data-driven COD traces showed a good correlation with Anzai in both SI and AP directions for the majority of studies, with correlation coefficients ranging from 63% to 89%. Based on the determined respiratory displacements of pancreas between end-expiration and end-inspiration from gated reconstructions, there was no significant difference between COD-based and Anzai-based methods. Finally, data-driven COD-based event-by-event respiratory motion correction yielded comparable results to that based on Anzai respiratory traces, in terms of contrast recovery and reduced motion-induced blur. Data-driven event-by-event respiratory motion correction using COD showed significant image quality improvement compared with reconstructions with no motion correction, and gave comparable results to the Anzai-based method.
Collapse
Affiliation(s)
- Silin Ren
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States of America
| | | | | | | | | | | | | |
Collapse
|