1
|
Kumari N, Adhikari A, Bhagat S, Mishra AK, Tiwari AK. Benzoxazolone-based FITC-conjugated fluorescent probe for locating in-vivo expression level of translocator protein (TSPO) during lung inflammation. Mol Divers 2025:10.1007/s11030-025-11192-9. [PMID: 40259117 DOI: 10.1007/s11030-025-11192-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/07/2025] [Indexed: 04/23/2025]
Abstract
Translocator protein 18 kDa (TSPO) has been a salient target for probing and monitoring inflammation in the central nervous system (CNS) and peripheral systems. Leveraging our previously developed, TSPO specific, modified acetamidobenzoxazolone derivative, the present work describes the synthesis and development of an optical probe for lung inflammation imaging: 2-(3,6-dihydroxy-9H-xanthen-9-yl)-5-(3-(3-(2-(methyl(phenyl)amino)-2-oxoethyl)-2-oxo-2,3-dihydrobenzo[d]oxazol-5-yl)thioureido)benzoic acid (FITC-MBP). The FITC-MBP is prepared through facile methodology by conjugating MBP to fluorophore dye FITC. Spectral properties remained equivalent to FITC dye with absorption and emission wavelength at 486 and 520 nm, respectively. Cellular uptake studies established overexpression of TSPO in lipopolysaccharide (LPS)-induced inflammation in H1299 lung cells. Reduced mean fluorescence intensity (MFI) during blocking experiments with PK11195 in flow cytometry suggests the specificity of the fluorescent probe towards TSPO. In-vivo optical imaging analysis on LPS-induced lung-inflamed balb/c mice revealed major sequestration of FITC-MBP in the lungs compared to control at 25 min post-injection that significantly decreased on pretreatment with PK11195 due to competitive binding to TSPO. On ground of these findings, we believe the novel fluorescent probe (FITC-MBP) might be utilized to visualize the overexpressed TSPO.
Collapse
Affiliation(s)
- Neelam Kumari
- Division of Cyclotron and Radiopharmaceutical Sciences (DCRS), Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research & Development Organization, Brig S K Mazumdar Road, Delhi, 110054, India
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi, India
| | - Anupriya Adhikari
- Department of Chemistry, Graphic Era Hill University, Dehradun, Uttarakhand, India
| | - Sunita Bhagat
- Organic Synthesis Research Laboratory, Department of Chemistry, A.R.S.D. College, University of Delhi, New Delhi, India
| | - Anil K Mishra
- Division of Cyclotron and Radiopharmaceutical Sciences (DCRS), Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research & Development Organization, Brig S K Mazumdar Road, Delhi, 110054, India
| | - Anjani K Tiwari
- Division of Cyclotron and Radiopharmaceutical Sciences (DCRS), Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research & Development Organization, Brig S K Mazumdar Road, Delhi, 110054, India.
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
2
|
Victorio CBL, Ganasarajah A, Novera W, Ong J, Msallam R, Chacko AM. Translocator protein (TSPO) is a biomarker of Zika virus (ZIKV) infection-associated neuroinflammation. Emerg Microbes Infect 2024; 13:2348528. [PMID: 38662785 PMCID: PMC11132733 DOI: 10.1080/22221751.2024.2348528] [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: 01/31/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024]
Abstract
Zika is a systemic inflammatory disease caused by infection with Zika virus (ZIKV). ZIKV infection in adults is associated with encephalitis marked by elevated expression of pro-inflammatory cytokines and chemokines, as well as increased brain infiltration of immune cells. In this study, we demonstrate that ZIKV encephalitis in a mouse infection model exhibits increased brain TSPO expression. TSPO expression on brain-resident and infiltrating immune cells in ZIKV infection correlates with disease and inflammation status in the brain. Brain TSPO expression can also be sensitively detected ex vivo and in vitro using radioactive small molecule probes that specifically bind to TSPO, such as [3H]PK11195. TSPO expression on brain-resident and infiltrating immune cells is a biomarker of ZIKV neuroinflammation, which can also be a general biomarker of acute viral neuroinflammatory disease.
Collapse
Affiliation(s)
- Carla Bianca Luena Victorio
- Laboratory for Translational and Molecular Imaging (LTMI), Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Arun Ganasarajah
- Laboratory for Translational and Molecular Imaging (LTMI), Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Wisna Novera
- Laboratory for Translational and Molecular Imaging (LTMI), Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Joanne Ong
- Laboratory for Translational and Molecular Imaging (LTMI), Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Rasha Msallam
- Laboratory for Translational and Molecular Imaging (LTMI), Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Ann-Marie Chacko
- Laboratory for Translational and Molecular Imaging (LTMI), Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
3
|
Wongso H, Kurniawan A, Setiadi Y, Kusumaningrum CE, Widyasari EM, Wibawa TH, Mahendra I, Febrian MB, Sriyani ME, Halimah I, Daruwati I, Gunawan R, Achmad A, Nugraha DH, Lesmana R, Nugraha AS. Translocator Protein 18 kDa (TSPO): A Promising Molecular Target for Image-Guided Surgery of Solid Cancers. Adv Pharm Bull 2024; 14:86-104. [PMID: 38585455 PMCID: PMC10997928 DOI: 10.34172/apb.2024.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/26/2023] [Accepted: 10/08/2023] [Indexed: 04/09/2024] Open
Abstract
The translocator protein 18-kDa (TSPO) is a mitochondrial membrane protein that is previously identified as the peripheral benzodiazepine receptor (PBR). Furthermore, it plays a significant role in a diverse range of biochemical processes, including steroidogenesis, mitochondrial cholesterol transport, cell survival and death, cell proliferation, and carcinogenesis. Several investigations also reported its roles in various types of cancers, including colorectal, brain, breast, prostate, and lung cancers, as well as melanoma. According to a previous study, the expression of TSPO was upregulated in cancer cells, which corresponds to an aggressive phenotype and/or poor prognosis. Consequently, the potential for crafting diagnostic and prognostic tools with a focus on TSPO holds great potential. In this context, several radioligands designed to target this protein have been identified, and some of the candidates have advanced to clinical trials. In recent years, the use of hybrid probes with radioactive and fluorescence molecules for image-guided surgery has exhibited promising results in animal and human studies. This indicates that the approach can serve as a valuable surgical navigator during cancer surgery. The current hybrid probes are built from various molecular platforms, including small molecules, nanoparticles, and antibodies. Although several TSPO-targeted imaging probes have been developed, their development for image-guided surgery of cancers is still limited. Therefore, this review aims to highlight recent findings on the involvement of TSPO in carcinogenesis, as well as provide a new perspective on the potential application of TSPO-targeted hybrid probes for image-guided surgery.
Collapse
Affiliation(s)
- Hendris Wongso
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
| | - Ahmad Kurniawan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Yanuar Setiadi
- Research Center for Environmental and Clean Technology, Research Organization for Life Sciences and Environment, National Research and Innovation Agency, Puspiptek, Banten 15314, Indonesia
| | - Crhisterra E. Kusumaningrum
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Eva M. Widyasari
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Teguh H.A. Wibawa
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Isa Mahendra
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
| | - Muhamad B. Febrian
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Maula E. Sriyani
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Iim Halimah
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Isti Daruwati
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
| | - Rudi Gunawan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
| | - Arifudin Achmad
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
- Department of Nuclear Medicine and Molecular Theranostics, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161
- Oncology and Stem Cells Working Group, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161
| | | | - Ronny Lesmana
- Department of Biomedical Science, Faculty of Medicine, Universitas Padjadjaran, Jatinangor 45363, Indonesia
- Physiology Molecular, Division of Biological Activity, Central Laboratory, Universitas Padjadjaran, Jatinangor 45363, Indonesia
- Laboratory of Sciences, Graduate School, Universitas Padjadjaran, Bandung, Indonesia
| | - Ari S. Nugraha
- Drug Utilisation and Discovery Research Group, Faculty of Pharmacy, Universitas Jember, Jember 68121, Indonesia
- School of Chemistry and Molecular Biosciences, Molecular Horizons, University of Wollongong, Wollongong, New South Wales, 2522, Australia
| |
Collapse
|
4
|
O'Neill J, Diaz MP, Alger JR, Pochon JB, Ghahremani D, Dean AC, Tyndale RF, Petersen N, Marohnic S, Karaiskaki A, London ED. Smoking, tobacco dependence, and neurometabolites in the dorsal anterior cingulate cortex. Mol Psychiatry 2023; 28:4756-4765. [PMID: 37749232 PMCID: PMC10914613 DOI: 10.1038/s41380-023-02247-0] [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: 03/31/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/27/2023]
Abstract
Cigarette smoking has a major impact on global health and morbidity, and positron emission tomographic research has provided evidence for reduced inflammation in the human brain associated with cigarette smoking. Given the consequences of inflammatory dysfunction for health, the question of whether cigarette smoking affects neuroinflammation warrants further investigation. The goal of this project therefore was to validate and extend evidence of hypoinflammation related to smoking, and to examine the potential contribution of inflammation to clinical features of smoking. Using magnetic resonance spectroscopy, we measured levels of neurometabolites that are putative neuroinflammatory markers. N-acetyl compounds (N-acetylaspartate + N-acetylaspartylglutamate), glutamate, creatine, choline-compounds (phosphocholine + glycerophosphocholine), and myo-inositol, have all been linked to neuroinflammation, but they have not been examined as such with respect to smoking. We tested whether people who smoke cigarettes have brain levels of these metabolites consistent with decreased neuroinflammation, and whether clinical features of smoking are associated with levels of these metabolites. The dorsal anterior cingulate cortex was chosen as the region-of-interest because of previous evidence linking it to smoking and related states. Fifty-four adults who smoked daily maintained overnight smoking abstinence before testing and were compared with 37 nonsmoking participants. Among the smoking participants, we tested for associations of metabolite levels with tobacco dependence, smoking history, craving, and withdrawal. Levels of N-acetyl compounds and glutamate were higher, whereas levels of creatine and choline compounds were lower in the smoking group as compared with the nonsmoking group. In the smoking group, glutamate and creatine levels correlated negatively with tobacco dependence, and creatine correlated negatively with lifetime smoking, but none of the metabolite levels correlated with craving or withdrawal. The findings indicate a link between smoking and a hypoinflammatory state in the brain, specifically in the dorsal anterior cingulate cortex. Smoking may thereby increase vulnerability to infection and brain injury.
Collapse
Affiliation(s)
- Joseph O'Neill
- Division of Child & Adolescent Psychiatry, Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
- Brain Research Institute, University of California at Los Angeles, Los Angeles, CA, USA
| | - Maylen Perez Diaz
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
- Biogen, Inc., Nashville, TN, USA
| | - Jeffry R Alger
- Department of Neurology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jean-Baptiste Pochon
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Dara Ghahremani
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Andrew C Dean
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Rachel F Tyndale
- Department of Pharmacology & Toxicology, and Department of Psychiatry, University of Toronto, and Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
| | - Nicole Petersen
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Shane Marohnic
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Andrea Karaiskaki
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Edythe D London
- Brain Research Institute, University of California at Los Angeles, Los Angeles, CA, USA.
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
5
|
D'Anna L, Searle G, Harvey K, Matthews PM, Veltkamp R. Time course of neuroinflammation after human stroke - a pilot study using co-registered PET and MRI. BMC Neurol 2023; 23:193. [PMID: 37193998 DOI: 10.1186/s12883-023-03178-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 03/22/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND Microglial activation contributes to both inflammatory damage and repair in experimental ischemic stroke. However, because of the logistical challenges, there have been few clinical imaging studies directly describing inflammatory activation and its resolution after stroke. The purpose of our pilot study was to describe the spatio-temporal profile of brain inflammation after stroke using 18kD translocator protein (TSPO) positron emission tomography (PET) with magnetic resonance (MR) co-registration in the subacute and chronic stage after stroke. METHODS Three patients underwent magnetic resonance imaging (MRI) and PET scans with TSPO ligand [11C]PBR28 15 ± 3 and 90 ± 7 days after an ischaemic stroke. Regions of interest (ROI) were defined on MRI images and applied to the dynamic PET data to derive regional time-activity curves. Regional uptake was quantified as standardised uptake values (SUV) over 60 to 90 min post-injection. ROI analysis was applied to identify binding in the infarct, and in frontal, temporal, parietal, and occipital lobes and cerebellum excluding the infarcted area. RESULTS The mean age of participants was 56 ± 20.4 years and mean infarct volume was 17.9 ± 18.1 ml. [11C]PBR28 showed increased tracer signal in the infarcted area compared to non-infarcted areas of the brain in the subacute phase of stroke (Patient 1 SUV 1.81; Patient 2 SUV 1.15; Patient 3 SUV 1.64). [11C]PBR28 uptake returned to the level of non-infarcted areas at 90 days Patient 1 SUV 0.99; Patient 3 SUV 0.80). No additional upregulation was detected elsewhere at either time point. CONCLUSIONS The neuroinflammatory reaction after ischaemic stroke is limited in time and circumscribed in space suggesting that post-ischaemic inflammation is tightly controlled but regulatory mechanisms.
Collapse
Affiliation(s)
- Lucio D'Anna
- Department of Stroke and Neuroscience, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | | | - Kirsten Harvey
- Department of Brain Sciences, Imperial College London, London, UK
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK
- Dementia Research Institute at Imperial College London, London, UK
| | - Roland Veltkamp
- Department of Brain Sciences, Imperial College London, London, UK.
- Department of Neurology, Alfried-Krupp Krankenhaus Essen, Essen, Germany.
- Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
6
|
Wongso H, Hendra R, Nugraha AS, Ritawidya R, Saptiama I, Kusumaningrum CE. Microbial metabolites diversity and their potential as molecular template for the discovery of new fluorescent and radiopharmaceutical probes. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
7
|
Viviano M, Barresi E, Siméon FG, Costa B, Taliani S, Da Settimo F, Pike VW, Castellano S. Essential Principles and Recent Progress in the Development of TSPO PET Ligands for Neuroinflammation Imaging. Curr Med Chem 2022; 29:4862-4890. [PMID: 35352645 PMCID: PMC10080361 DOI: 10.2174/0929867329666220329204054] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/21/2021] [Accepted: 01/25/2022] [Indexed: 11/22/2022]
Abstract
The translocator protein 18kDa (TSPO) is expressed in the outer mitochondrial membrane and is implicated in several functions, including cholesterol transport and steroidogenesis. Under normal physiological conditions, TSPO is present in very low concentrations in the human brain but is markedly upregulated in response to brain injury and inflammation. This upregulation is strongly associated with activated microglia. Therefore, TSPO is particularly suited for assessing active gliosis associated with brain lesions following injury or disease. For over three decades, TSPO has been studied as a biomarker. Numerous radioligands for positron emission tomography (PET) that target TSPO have been developed for imaging inflammatory progression in the brain. Although [11C]PK11195, the prototypical first-generation PET radioligand, is still widely used for in vivo studies, mainly now as its single more potent R-enantiomer, it has severe limitations, including low sensitivity and poor amenability to quantification. Second-generation radioligands are characterized by higher TSPO specific signals but suffer from other drawbacks, such as sensitivity to the TSPO single nucleotide polymorphism (SNP) rs6971. Therefore, their applications in human studies have the burden of needing to genotype subjects. Consequently, recent efforts are focused on developing improved radioligands that combine the optimal features of the second generation with the ability to overcome the differences in binding affinities across the population. This review presents essential principles in the design and development of TSPO PET ligands and discusses prominent examples among the main chemotypes.
Collapse
Affiliation(s)
- Monica Viviano
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| | | | - Fabrice G. Siméon
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | | | - Victor W. Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sabrina Castellano
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| |
Collapse
|
8
|
The mitochondrial translocator protein (TSPO): a key multifunctional molecule in the nervous system. Biochem J 2022; 479:1455-1466. [PMID: 35819398 DOI: 10.1042/bcj20220050] [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: 01/31/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 12/12/2022]
Abstract
Translocator protein (TSPO, 18 kDa), formerly known as peripheral benzodiazepine receptor, is an evolutionary well-conserved protein located on the outer mitochondrial membrane. TSPO is involved in a variety of fundamental physiological functions and cellular processes. Its expression levels are regulated under many pathological conditions, therefore, TSPO has been proposed as a tool for diagnostic imaging and an attractive therapeutic drug target in the nervous system. Several synthetic TSPO ligands have thus been explored as agonists and antagonists for innovative treatments as neuroprotective and regenerative agents. In this review, we provide state-of-the-art knowledge of TSPO functions in the brain and peripheral nervous system. Particular emphasis is placed on its contribution to important physiological functions such as mitochondrial homeostasis, energy metabolism and steroidogenesis. We also report how it is involved in neuroinflammation, brain injury and diseases of the nervous system.
Collapse
|
9
|
Wongso H, Yamasaki T, Kumata K, Ono M, Higuchi M, Zhang MR, Fulham MJ, Katsifis A, Keller PA. Design, Synthesis, and Biological Evaluation of Novel Fluorescent Probes Targeting the 18-kDa Translocator Protein. ChemMedChem 2021; 16:1902-1916. [PMID: 33631047 DOI: 10.1002/cmdc.202000984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/22/2021] [Indexed: 12/20/2022]
Abstract
A series of fluorescent probes from the 6-chloro-2-phenylimidazo[1,2-a]pyridine-3-yl acetamides ligands featuring the 7-nitro-2-oxa-1,3-diazol-4-yl (NBD) moiety has been synthesized and biologically evaluated for their fluorescence properties and for their binding affinity to the 18-kDa translocator protein (TSPO). Spectroscopic studies including UV/Vis absorption and fluorescence measurements showed that the synthesized fluorescent probes exhibit favorable spectroscopic properties, especially in nonpolar environments. In vitro fluorescence staining in brain sections from lipopolysaccharide (LPS)-injected mice revealed partial colocalization of the probes with the TSPO. The TSPO binding affinity of the probes was measured on crude mitochondrial fractions separated from rat brain homogenates in a [11 C]PK11195 radioligand binding assay. All the new fluorescent probes demonstrated moderate to high binding affinity to the TSPO, with affinity (Ki ) values ranging from 0.58 nM to 3.28 μM. Taking these data together, we propose that the new fluorescent probes could be used to visualize the TSPO.
Collapse
Affiliation(s)
- Hendris Wongso
- School of Chemistry and Molecular Bioscience, and Molecular Horizons, University of Wollongong, Wollongong, NSW, 2522, Australia.,Center for Applied Nuclear Science and Technology, National Nuclear Energy Agency, Bandung, 40132, Indonesia
| | - Tomoteru Yamasaki
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Chiba, 263-8555, Japan
| | - Katsushi Kumata
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Chiba, 263-8555, Japan
| | - Maiko Ono
- Department of Functional Brain Imaging Research, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging Research, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, Chiba, 263-8555, Japan
| | - Michael J Fulham
- Department of PET and Nuclear Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Andrew Katsifis
- Department of PET and Nuclear Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Paul A Keller
- School of Chemistry and Molecular Bioscience, and Molecular Horizons, University of Wollongong, Wollongong, NSW, 2522, Australia
| |
Collapse
|
10
|
Shteinfer-Kuzmine A, Verma A, Arif T, Aizenberg O, Paul A, Shoshan-Barmaz V. Mitochondria and nucleus cross-talk: Signaling in metabolism, apoptosis, and differentiation, and function in cancer. IUBMB Life 2021; 73:492-510. [PMID: 33179373 DOI: 10.1002/iub.2407] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/15/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022]
Abstract
The cross-talk between the mitochondrion and the nucleus regulates cellular functions, including differentiation and adaptation to stress. Mitochondria supply metabolites for epigenetic modifications and other nuclear-associated activities and certain mitochondrial proteins were found in the nucleus. The voltage-dependent anion channel 1 (VDAC1), localized at the outer mitochondrial membrane (OMM) is a central protein in controlling energy production, cell growth, Ca2+ homeostasis, and apoptosis. To alter the cross-talk between the mitochondria and the nucleus, we used specific siRNA to silence the expression of VDAC1 in glioblastoma (GBM) U87-MG and U118-MG cell-derived tumors, and then monitored the nuclear localization of mitochondrial proteins and the methylation and acetylation of histones. Depletion of VDAC1 from tumor cells reduced metabolism, leading to inhibition of tumor growth, and several tumor-associated processes and signaling pathways linked to cancer development. In addition, we demonstrate that certain mitochondrial pro-apoptotic proteins such as caspases 3, 8, and 9, and p53 were unexpectedly overexpressed in tumors, suggesting that they possess additional non-apoptotic functions. VDAC1 depletion and metabolic reprograming altered their expression levels and subcellular localization, specifically their translocation to the nucleus. In addition, VDAC1 depletion also leads to epigenetic modifications of histone acetylation and methylation, suggesting that the interchange between metabolism and cancer signaling pathways involves mitochondria-nucleus cross-talk. The mechanisms regulating mitochondrial protein trafficking into and out of the nucleus and the role these proteins play in the nucleus remain to be elucidated.
Collapse
Affiliation(s)
- Anna Shteinfer-Kuzmine
- Department of Life Sciences, Ben-Gurion University of the Negev and the National Institute for Biotechnology in the Negev, Beersheba, Israel
| | - Ankit Verma
- Department of Life Sciences, Ben-Gurion University of the Negev and the National Institute for Biotechnology in the Negev, Beersheba, Israel
| | - Tasleem Arif
- Department of Life Sciences, Ben-Gurion University of the Negev and the National Institute for Biotechnology in the Negev, Beersheba, Israel
- Department of Cell, Developmental, & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Or Aizenberg
- Department of Life Sciences, Ben-Gurion University of the Negev and the National Institute for Biotechnology in the Negev, Beersheba, Israel
| | - Avijit Paul
- Department of Life Sciences, Ben-Gurion University of the Negev and the National Institute for Biotechnology in the Negev, Beersheba, Israel
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Varda Shoshan-Barmaz
- Department of Life Sciences, Ben-Gurion University of the Negev and the National Institute for Biotechnology in the Negev, Beersheba, Israel
| |
Collapse
|
11
|
Zhang L, Hu K, Shao T, Hou L, Zhang S, Ye W, Josephson L, Meyer JH, Zhang MR, Vasdev N, Wang J, Xu H, Wang L, Liang SH. Recent developments on PET radiotracers for TSPO and their applications in neuroimaging. Acta Pharm Sin B 2021; 11:373-393. [PMID: 33643818 PMCID: PMC7893127 DOI: 10.1016/j.apsb.2020.08.006] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/15/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
The 18 kDa translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor, is predominately localized to the outer mitochondrial membrane in steroidogenic cells. Brain TSPO expression is relatively low under physiological conditions, but is upregulated in response to glial cell activation. As the primary index of neuroinflammation, TSPO is implicated in the pathogenesis and progression of numerous neuropsychiatric disorders and neurodegenerative diseases, including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), multiple sclerosis (MS), major depressive disorder (MDD) and obsessive compulsive disorder (OCD). In this context, numerous TSPO-targeted positron emission tomography (PET) tracers have been developed. Among them, several radioligands have advanced to clinical research studies. In this review, we will overview the recent development of TSPO PET tracers, focusing on the radioligand design, radioisotope labeling, pharmacokinetics, and PET imaging evaluation. Additionally, we will consider current limitations, as well as translational potential for future application of TSPO radiopharmaceuticals. This review aims to not only present the challenges in current TSPO PET imaging, but to also provide a new perspective on TSPO targeted PET tracer discovery efforts. Addressing these challenges will facilitate the translation of TSPO in clinical studies of neuroinflammation associated with central nervous system diseases.
Collapse
Key Words
- AD, Alzheimer's disease
- ALS, amyotrophic lateral sclerosis
- AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid
- ANT, adenine nucleotide transporter
- Am, molar activities
- BBB, blood‒brain barrier
- BMSC, bone marrow stromal cells
- BP, binding potential
- BPND, non-displaceable binding potential
- BcTSPO, Bacillus cereus TSPO
- CBD, corticobasal degeneration
- CNS disorders
- CNS, central nervous system
- CRAC, cholesterol recognition amino acid consensus sequence
- DLB, Lewy body dementias
- EP, epilepsy
- FTD, frontotemporal dementia
- HAB, high-affinity binding
- HD, Huntington's disease
- HSE, herpes simplex encephalitis
- IMM, inner mitochondrial membrane
- KA, kainic acid
- LAB, low-affinity binding
- LPS, lipopolysaccharide
- MAB, mixed-affinity binding
- MAO-B, monoamine oxidase B
- MCI, mild cognitive impairment
- MDD, major depressive disorder
- MMSE, mini-mental state examination
- MRI, magnetic resonance imaging
- MS, multiple sclerosis
- MSA, multiple system atrophy
- Microglial activation
- NAA/Cr, N-acetylaspartate/creatine
- Neuroinflammation
- OCD, obsessive compulsive disorder
- OMM, outer mitochondrial membrane
- P2X7R, purinergic receptor P2X7
- PAP7, RIa-associated protein
- PBR, peripheral benzodiazepine receptor
- PCA, posterior cortical atrophy
- PD, Parkinson's disease
- PDD, PD dementia
- PET, positron emission tomography
- PKA, protein kinase A
- PRAX-1, PBR-associated protein 1
- PSP, progressive supranuclear palsy
- Positron emission tomography (PET)
- PpIX, protoporphyrin IX
- QA, quinolinic acid
- RCYs, radiochemical yields
- ROS, reactive oxygen species
- RRMS, relapsing remitting multiple sclerosis
- SA, specific activity
- SAH, subarachnoid hemorrhage
- SAR, structure–activity relationship
- SCIDY, spirocyclic iodonium ylide
- SNL, selective neuronal loss
- SNR, signal to noise ratio
- SUV, standard uptake volume
- SUVR, standard uptake volume ratio
- TBAH, tetrabutyl ammonium hydroxide
- TBI, traumatic brain injury
- TLE, temporal lobe epilepsy
- TSPO
- TSPO, translocator protein
- VDAC, voltage-dependent anion channel
- VT, distribution volume
- d.c. RCYs, decay-corrected radiochemical yields
- dMCAO, distal middle cerebral artery occlusion
- fP, plasma free fraction
- n.d.c. RCYs, non-decay-corrected radiochemical yields
- p.i., post-injection
Collapse
|
12
|
de Souza AM, Pitombeira MS, de Souza LE, Marques FLN, Buchpiguel CA, Real CC, de Paula Faria D. 11C-PK11195 plasma metabolization has the same rate in multiple sclerosis patients and healthy controls: a cross-sectional study. Neural Regen Res 2021; 16:2494-2498. [PMID: 33907039 PMCID: PMC8374550 DOI: 10.4103/1673-5374.313062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
11C-PK11195 is a positron emitter tracer used for Positron Emission Tomography (PET) imaging of innate immune cell activation in studies of neuroinflammatory diseases. For the image quantitative analysis, it is necessary to quantify the intact fraction of this tracer in the arterial plasma during imaging acquisition (plasma intact fraction). Due to the complexity and costs involved in this analysis it is important to evaluate the real necessity of individual analysis in each 11C-PK11195 PET imaging acquisition. The purpose of this study is to compare 11C-PK11195 plasma metabolization rate between healthy controls and multiple sclerosis (MS) patients and evaluate the interference of sex, age, treatment, and disease phenotype in the tracer intact fraction measured in arterial plasma samples. 11C-PK11195 metabolization rate in arterial plasma was quantified by high performance liquid chromatography in samples from MS patients (n = 50) and healthy controls (n = 23) at 20, 45, and 60 minutes after 11C-PK11195 injection. Analyses were also stratified by sex, age, treatment type, and MS phenotype. The results showed no significant differences in the metabolization rate of healthy controls and MS patients, or in the stratified samples. In conclusion, 11C-PK11195 metabolization has the same rate in patients with MS and healthy controls, which is not affected by sex, age, treatment, and disease phenotype. Thus, these findings could contribute to exempting the necessity for tracer metabolization determination in all 11C-PK11195 PET imaging acquisition, by using a population metabolization rate average. The study procedures were approved by the Ethics Committee for Research Projects Analysis of the Hospital das Clinicas of the University of Sao Paulo Medical School (approval No. 624.065) on April 23, 2014.
Collapse
Affiliation(s)
- Aline Morais de Souza
- Laboratory of Nuclear Medicine (LIM43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Milena Sales Pitombeira
- Department of Neurology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Larissa Estessi de Souza
- Laboratory of Nuclear Medicine (LIM43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Fabio Luiz Navarro Marques
- Laboratory of Nuclear Medicine (LIM43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Carlos Alberto Buchpiguel
- Laboratory of Nuclear Medicine (LIM43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Caroline Cristiano Real
- Laboratory of Nuclear Medicine (LIM43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Daniele de Paula Faria
- Laboratory of Nuclear Medicine (LIM43), Department of Radiology and Oncology; Department of Neurology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
13
|
Ma Z, Guo X, Liu J. Translocator protein mediates olfactory repulsion. FASEB J 2020; 34:513-524. [PMID: 31914587 DOI: 10.1096/fj.201900528rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 10/14/2019] [Accepted: 10/21/2019] [Indexed: 11/11/2022]
Abstract
Translocator protein (TSPO, 18kDa), which was previously known as a peripheral-type benzodiazepine receptor, is associated with psychiatric disorders and acts as a neuroimaging biomarker. However, its function and mechanism in modulating behaviors are less well-known. Herein, we found that TSPO in migratory locusts shows conserved protein traits and is expressed at high levels in the brains. The expression levels of tspo mRNA and protein were higher in brains of solitary locusts than those in gregarious locusts, whereas the mRNA and protein expression levels remained stable during crowding and isolation, suggesting that the expression level of TSPO is potentially associated with behavioral phenotype of solitary locusts. Moreover, tspo RNAi knockdown in the brains of solitary locusts decreased their olfactory repulsion. After RNAi knockdown of tyramine receptor (TyR) in the brains of solitary locusts, RNA-seq analysis identified that a functional class of receptors, which included tspo, was downregulated significantly. Moreover, tspo mRNA and protein expression levels were downregulated and upregulated after TyR RNAi knockdown and activation, respectively. tspo RNAi knockdown in the brains of solitary locusts induced the attractive response and inhibited the function of tyramine (TA)-TyR in inducing olfactory repulsion. In gregarious locusts, tspo RNAi knockdown inhibited the function of TA-TyR inducing olfactory repulsion. This study confirms that TSPO acts as a crucial effector protein in TA-TyR signaling to modulate olfactory repulsion. Furthermore, this study provides a novel mechanism by which TSPO functionally connects a G-protein-coupled receptor and a mitochondria membrane protein in modulating olfactory repulsion.
Collapse
Affiliation(s)
- Zongyuan Ma
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Xiaojiao Guo
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Jipeng Liu
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
14
|
Shoshan-Barmatz V, Pittala S, Mizrachi D. VDAC1 and the TSPO: Expression, Interactions, and Associated Functions in Health and Disease States. Int J Mol Sci 2019; 20:ijms20133348. [PMID: 31288390 PMCID: PMC6651789 DOI: 10.3390/ijms20133348] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 12/12/2022] Open
Abstract
The translocator protein (TSPO), located at the outer mitochondrial membrane (OMM), serves multiple functions and contributes to numerous processes, including cholesterol import, mitochondrial metabolism, apoptosis, cell proliferation, Ca2+ signaling, oxidative stress, and inflammation. TSPO forms a complex with the voltage-dependent anion channel (VDAC), a protein that mediates the flux of ions, including Ca2+, nucleotides, and metabolites across the OMM, controls metabolism and apoptosis and interacts with many proteins. This review focuses on the two OMM proteins TSPO and VDAC1, addressing their structural interaction and associated functions. TSPO appears to be involved in the generation of reactive oxygen species, proposed to represent the link between TSPO activation and VDAC, thus playing a role in apoptotic cell death. In addition, expression of the two proteins in healthy brains and diseased states is considered, as is the relationship between TSPO and VDAC1 expression. Both proteins are over-expressed in in brains from Alzheimer’s disease patients. Finally, TSPO expression levels were proposed as a biomarker of some neuropathological settings, while TSPO-interacting ligands have been considered as a potential basis for drug development.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| | - Srinivas Pittala
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Dario Mizrachi
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| |
Collapse
|
15
|
Investigating targets for neuropharmacological intervention by molecular dynamics simulations. Biochem Soc Trans 2019; 47:909-918. [PMID: 31085614 DOI: 10.1042/bst20190048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/11/2019] [Accepted: 04/23/2019] [Indexed: 01/09/2023]
Abstract
Medical research has identified over 500 brain disorders. Among these, there are still only very few neuropathologies whose causes are fully understood and, consequently, very few drugs whose mechanism of action is known. No FDA drug has been identified for major neurodegenerative diseases, such as Alzheimer's and Parkinson's. We still lack effective treatments and strategies for modulating progression or even early neurodegenerative disease onset diagnostic tools. A great support toward the highly needed identification of neuroactive drugs comes from computer simulation methods and, in particular, from molecular dynamics (MD). This provides insight into structure-function relationship of a target and predicts structure, dynamics and energetics of ligand/target complexes under biologically relevant conditions like temperature and physiological saline concentration. Here, we present examples of the predictive power of MD for neuroactive ligands/target complexes. This brief survey from our own research shows the usefulness of partnerships between academia and industry, and from joint efforts between experimental and theoretical groups.
Collapse
|
16
|
Srivastava P, Kakkar D, Kumar P, Tiwari AK. Modified benzoxazolone (ABO‐AA) based single photon emission computed tomography (SPECT) probes for 18 kDa translocator protein. Drug Dev Res 2019; 80:741-749. [DOI: 10.1002/ddr.21547] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/09/2019] [Accepted: 05/11/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Pooja Srivastava
- Division of Cyclotron and Radiopharmaceutical SciencesInstitute of Nuclear Medicine and Allied Sciences Delhi India
- Molecular Neuroscience and Functional Genomic Laboratory, Department of BiotechnologyDelhi Technological University Delhi India
| | - Dipti Kakkar
- Division of Cyclotron and Radiopharmaceutical SciencesInstitute of Nuclear Medicine and Allied Sciences Delhi India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomic Laboratory, Department of BiotechnologyDelhi Technological University Delhi India
| | - Anjani Kumar Tiwari
- Division of Cyclotron and Radiopharmaceutical SciencesInstitute of Nuclear Medicine and Allied Sciences Delhi India
- Department of Chemistry, School of Physical & Decision Sciences (SPDS)Babasaheb Bhimrao Ambedkar Central University Lucknow UP India
| |
Collapse
|
17
|
Qiao L, Fisher E, McMurray L, Milicevic Sephton S, Hird M, Kuzhuppilly-Ramakrishnan N, Williamson DJ, Zhou X, Werry E, Kassiou M, Luthra S, Trigg W, Aigbirhio FI. Radiosynthesis of (R,S)-[ 18 F]GE387: A Potential PET Radiotracer for Imaging Translocator Protein 18 kDa (TSPO) with Low Binding Sensitivity to the Human Gene Polymorphism rs6971. ChemMedChem 2019; 14:982-993. [PMID: 30900397 PMCID: PMC6563049 DOI: 10.1002/cmdc.201900023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/26/2019] [Indexed: 12/14/2022]
Abstract
Translocator protein (TSPO) is a biomarker of neuroinflammation, which is a hallmark of many neurodegenerative diseases and has been exploited as a positron emission tomography (PET) target. Carbon-11-labelled PK11195 remains the most applied agent for imaging TSPO, despite its short-lived isotope and low brain permeability. Second-generation radiotracers show variance in affinity amongst subjects (low-, mixed-, and high-affinity binders) caused by the genetic polymorphism (rs6971) of the TSPO gene. To overcome these limitations, a new structural scaffold was explored based on the TSPO pharmacophore, and the analogue with a low-affinity binder/high-affinity binder (LAB/HAB) ratio similar (1.2 vs. 1.3) to that of (R)-[11 C]PK11195 was investigated. The synthesis of the reference compound was accomplished in six steps and 9 % overall yield, and the precursor was prepared in eight steps and 8 % overall yield. The chiral separation of the reference and precursor compounds was performed using supercritical fluid chromatography with >95 % ee. The absolute configuration was determined by circular dichroism. Optimisation of reaction conditions for manual radiolabelling revealed acetonitrile as a preferred solvent at 100 °C. Automation of this radiolabelling method provided R and S enantiomers in respective 21.3±16.7 and 25.6±7.1 % decay-corrected yields and molar activities of 55.8±35.6 and 63.5±39.5 GBq μmol-1 (n=3). Injection of the racemic analogue into a healthy rat confirmed passage through the blood-brain barrier.
Collapse
Affiliation(s)
- Luxi Qiao
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Emily Fisher
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Lindsay McMurray
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Selena Milicevic Sephton
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Matthew Hird
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Nisha Kuzhuppilly-Ramakrishnan
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - David J Williamson
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Xiouyun Zhou
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Eryn Werry
- School of Chemistry, The University of Sydney, Building F11, Eastern Avenue, Sydney, NSW, 2006, Australia
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, Building F11, Eastern Avenue, Sydney, NSW, 2006, Australia
| | | | | | - Franklin I Aigbirhio
- Molecular Imaging Chemical Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| |
Collapse
|
18
|
Novel Immunotherapeutic Approaches to Target Alpha-Synuclein and Related Neuroinflammation in Parkinson's Disease. Cells 2019; 8:cells8020105. [PMID: 30708997 PMCID: PMC6406239 DOI: 10.3390/cells8020105] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/23/2019] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
The etiology of Parkinson’s disease (PD) is significantly influenced by disease-causing changes in the protein alpha-Synuclein (aSyn). It can trigger and promote intracellular stress and thereby impair the function of dopaminergic neurons. However, these damage mechanisms do not only extend to neuronal cells, but also affect most glial cell populations, such as astroglia and microglia, but also T lymphocytes, which can no longer maintain the homeostatic CNS milieu because they produce neuroinflammatory responses to aSyn pathology. Through precise neuropathological examination, molecular characterization of biomaterials, and the use of PET technology, it has been clearly demonstrated that neuroinflammation is involved in human PD. In this review, we provide an in-depth overview of the pathomechanisms that aSyn elicits in models of disease and focus on the affected glial cell and lymphocyte populations and their interaction with pathogenic aSyn species. The interplay between aSyn and glial cells is analyzed both in the basic research setting and in the context of human neuropathology. Ultimately, a strong rationale builds up to therapeutically reduce the burden of pathological aSyn in the CNS. The current antibody-based approaches to lower the amount of aSyn and thereby alleviate neuroinflammatory responses is finally discussed as novel therapeutic strategies for PD.
Collapse
|
19
|
Srivastava P, Kumari N, Kakkar D, Kaul A, Kumar P, Tiwari AK. Comparative evaluation of 99mTc-MBIP-X/11[C] MBMP for visualization of 18 kDa translocator protein. NEW J CHEM 2019. [DOI: 10.1039/c9nj00180h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An elevated translocator protein (18 kDa, TSPO) density is observed during inflammation in the brain and peripheral organs making it a viable target for imaging.
Collapse
Affiliation(s)
- Pooja Srivastava
- Division of Cyclotron and Radiopharmaceutical Sciences
- Institute of Nuclear Medicine and Allied Sciences
- Delhi 110054
- India
- Molecular Neuroscience and Functional Genomics Laboratory
| | - Neelam Kumari
- Division of Cyclotron and Radiopharmaceutical Sciences
- Institute of Nuclear Medicine and Allied Sciences
- Delhi 110054
- India
| | - Dipti Kakkar
- Division of Cyclotron and Radiopharmaceutical Sciences
- Institute of Nuclear Medicine and Allied Sciences
- Delhi 110054
- India
| | - Ankur Kaul
- Division of Cyclotron and Radiopharmaceutical Sciences
- Institute of Nuclear Medicine and Allied Sciences
- Delhi 110054
- India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory
- Department of Biotechnology
- Delhi Technological University
- Delhi 110042
- India
| | - Anjani K. Tiwari
- Division of Cyclotron and Radiopharmaceutical Sciences
- Institute of Nuclear Medicine and Allied Sciences
- Delhi 110054
- India
- Department of Chemistry
| |
Collapse
|
20
|
Kwon YD, Kang S, Park H, Cheong IK, Chang KA, Lee SY, Jung JH, Lee BC, Lim ST, Kim HK. Novel potential pyrazolopyrimidine based translocator protein ligands for the evaluation of neuroinflammation with PET. Eur J Med Chem 2018; 159:292-306. [PMID: 30296688 DOI: 10.1016/j.ejmech.2018.09.069] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 09/06/2018] [Accepted: 09/28/2018] [Indexed: 01/06/2023]
Abstract
Translocator protein (TSPO) is an interesting biological target because TSPO overexpression is associated with microglial activation caused by neuronal damage or neuroinflammation, and these activated microglia are involved in several central nervous system diseases. Herein, novel fluorinated ligands (14a-c and 16a-c) based on a 2-phenylpyrazolo[1,5-a]pyrimidin-3-yl acetamide scaffold were synthesized, and in vitro characterization of each of the novel ligands was performed to elucidate structure activity relationships. All of the newly synthesized ligands displayed nano-molar affinity for TSPO. Particularly, an in vitro affinity study suggests that 2-(5,7-diethyl-2-(4-(3-fluoro-2-methylpropoxy)phenyl)pyrazolo[1,5-a]pyrimidin-3-yl)-N,N-diethylacetamide (14a), which exhibited high nano-molar affinity for TSPO and proper lipophilicity, was suitable for in vivo brain studies. Thus, radiosynthesis from tosylate precursor 13a using fluorine-18 was performed, and [18F]14a was obtained in a 31% radiochemical yield (decay-corrected). Dynamic positron emission tomography (PET) imaging studies were performed in a lipopolysaccharide (LPS)-induced neuroinflammation rat model using [18F]14a to identify the location of inflammation in the brain with a high target-to-background signal ratio. In addition, we validated that the locations of inflammatory lesions found by PET imaging were consistent with the locations observed by histological examination of dissected brains using antibodies. These results suggest that [18F]14a is a novel promising PET imaging agent for diagnosing neuroinflammation, and it may also prove to be applicable for diagnosing other diseases, including cancers associated with altered TSPO expression, using PET techniques.
Collapse
Affiliation(s)
- Young-Do Kwon
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Department of Nuclear Medicine, Molecular Imaging & Therapeutic Medicine Research Center, Chonbuk National University Medical School and Hospital, Jeonju, 54907, Republic of Korea
| | - Shinwoo Kang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, 21936, Republic of Korea; Neuroscience Research Institute, Gachon University, Incheon, 21565, Republic of Korea
| | - Hyunjun Park
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, 21936, Republic of Korea; Neuroscience Research Institute, Gachon University, Incheon, 21565, Republic of Korea; Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon, 21936, Republic of Korea
| | - Il-Koo Cheong
- Neuroscience Research Institute, Gachon University, Incheon, 21565, Republic of Korea; Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon, 21936, Republic of Korea
| | - Keun-A Chang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, 21936, Republic of Korea; Neuroscience Research Institute, Gachon University, Incheon, 21565, Republic of Korea; Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon, 21936, Republic of Korea.
| | - Sang-Yoon Lee
- Neuroscience Research Institute, Gachon University, Incheon, 21565, Republic of Korea; Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon, 21936, Republic of Korea; Department of Neuroscience, College of Medicine, Gachon University, Incheon, 21936, Republic of Korea
| | - Jae Ho Jung
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, 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
| | - Seok Tae Lim
- Department of Nuclear Medicine, Molecular Imaging & Therapeutic Medicine Research Center, Chonbuk National University Medical School and Hospital, Jeonju, 54907, Republic of Korea; Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, 54907, Republic of Korea
| | - Hee-Kwon Kim
- Department of Nuclear Medicine, Molecular Imaging & Therapeutic Medicine Research Center, Chonbuk National University Medical School and Hospital, Jeonju, 54907, Republic of Korea; Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, 54907, Republic of Korea.
| |
Collapse
|
21
|
Yu Z, Yang L, Yang Y, Chen S, Sun D, Xu H, Fan X. Epothilone B Benefits Nigral Dopaminergic Neurons by Attenuating Microglia Activation in the 6-Hydroxydopamine Lesion Mouse Model of Parkinson's Disease. Front Cell Neurosci 2018; 12:324. [PMID: 30323743 PMCID: PMC6172330 DOI: 10.3389/fncel.2018.00324] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 09/06/2018] [Indexed: 11/24/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by loss of dopamine (DA) neurons in the substantia nigra pars compacta (SNc) and a subsequent reduction in striatal DA levels. Recent studies have shown that systemic administration of subtoxic doses of epothilone B (EpoB), a microtubule stabilizing agent, enhances axonal regeneration. However, the underlying alterations in cellular mechanisms remain undetermined. In the present study, we investigated the neuroprotective effects of EpoB on DA neurons in mouse model of PD induced by 6-hydroxyDA (6-OHDA) and in vitro. The results indicated that EpoB improved behavioral deficits, protected the nigrostriatal dopaminergic projections and restored DA level in the striatum of mice exposed to 6-OHDA. Meanwhile, EpoB attenuated microglia activation in the SNc of PD mice. Furthermore, EpoB treatment ameliorated 6-OHDA induced cytotoxicity to MN9D dopaminergic cells in a co-culture transwell system of BV2/MN9D cells, and redistributed the cytoskeleton of microglial BV2 and caused the morphological transition, inhibited the polarization to the M1 phenotype by suppressing expression of pro-inflammatory factors including interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α. Overall, our study suggested that EpoB treatment protects nigral DA neurons and projections through limiting the cytotoxicity of activated microglia in 6-OHDA lesioned mice.
Collapse
Affiliation(s)
- Zhongyuan Yu
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Army Medical University, Chongqing, China.,Battalion 3 of Cadet Brigade, Third Military Medical University, Army Medical University, Chongqing, China
| | - Ling Yang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Army Medical University, Chongqing, China
| | - Yang Yang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
| | - Siyu Chen
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
| | - Dayu Sun
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
| | - Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Army Medical University, Chongqing, China
| |
Collapse
|
22
|
Zeng J, Guareschi R, Damre M, Cao R, Kless A, Neumaier B, Bauer A, Giorgetti A, Carloni P, Rossetti G. Structural Prediction of the Dimeric Form of the Mammalian Translocator Membrane Protein TSPO: A Key Target for Brain Diagnostics. Int J Mol Sci 2018; 19:E2588. [PMID: 30200318 PMCID: PMC6165245 DOI: 10.3390/ijms19092588] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/21/2018] [Accepted: 08/28/2018] [Indexed: 11/17/2022] Open
Abstract
Positron emission tomography (PET) radioligands targeting the human translocator membrane protein (TSPO) are broadly used for the investigations of neuroinflammatory conditions associated with neurological disorders. Structural information on the mammalian protein homodimers-the suggested functional state of the protein-is limited to a solid-state nuclear magnetic resonance (NMR) study and to a model based on the previously-deposited solution NMR structure of the monomeric mouse protein. Computational studies performed here suggest that the NMR-solved structure in the presence of detergents is not prone to dimer formation and is furthermore unstable in its native membrane environment. We, therefore, propose a new model of the functionally-relevant dimeric form of the mouse protein, based on a prokaryotic homologue. The model, fully consistent with solid-state NMR data, is very different from the previous predictions. Hence, it provides, for the first time, structural insights into this pharmaceutically-important target which are fully consistent with experimental data.
Collapse
Affiliation(s)
- Juan Zeng
- Institute for Advanced Simulations (IAS)-5/Institute for Neuroscience and Medicine (INM)-9, Forschungszentrum Jülich, 52428 Jülich, Germany.
- Laboratory of Computational Chemistry and Drug Design, Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, 518055 Shenzhen, China.
| | - Riccardo Guareschi
- Institute for Advanced Simulations (IAS)-5/Institute for Neuroscience and Medicine (INM)-9, Forschungszentrum Jülich, 52428 Jülich, Germany.
| | - Mangesh Damre
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134 Verona, Italy.
- Neurobiology, International School for Advanced Studies (SISSA), 34136 Trieste, Italy.
| | - Ruyin Cao
- Institute for Advanced Simulations (IAS)-5/Institute for Neuroscience and Medicine (INM)-9, Forschungszentrum Jülich, 52428 Jülich, Germany.
| | - Achim Kless
- Grünenthal Innovation, Translational Science & Intelligence, Grünenthal GmbH, 52078 Aachen, Germany.
| | - Bernd Neumaier
- Institute for Neuroscience and Medicine (INM)-5, Forschungszentrum Jülich, 52428 Jülich, Germany.
| | - Andreas Bauer
- Institute for Neuroscience and Medicine (INM)-2, Forschungszentrum Jülich, 52428 Jülich, Germany.
| | - Alejandro Giorgetti
- Institute for Advanced Simulations (IAS)-5/Institute for Neuroscience and Medicine (INM)-9, Forschungszentrum Jülich, 52428 Jülich, Germany.
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134 Verona, Italy.
| | - Paolo Carloni
- Institute for Advanced Simulations (IAS)-5/Institute for Neuroscience and Medicine (INM)-9, Forschungszentrum Jülich, 52428 Jülich, Germany.
- RWTH Aachen University, Department of Physics, 52078 Aachen, Germany.
| | - Giulia Rossetti
- Institute for Advanced Simulations (IAS)-5/Institute for Neuroscience and Medicine (INM)-9, Forschungszentrum Jülich, 52428 Jülich, Germany.
- Jülich Supercomputing Center (JSC), Forschungszentrum Jülich, 52428 Jülich, Germany.
- University Hospital Aachen, RWTH Aachen University, 52078 Aachen, Germany.
| |
Collapse
|
23
|
Chen Z, Jamadar SD, Li S, Sforazzini F, Baran J, Ferris N, Shah NJ, Egan GF. From simultaneous to synergistic MR-PET brain imaging: A review of hybrid MR-PET imaging methodologies. Hum Brain Mapp 2018; 39:5126-5144. [PMID: 30076750 DOI: 10.1002/hbm.24314] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/25/2018] [Accepted: 07/02/2018] [Indexed: 12/17/2022] Open
Abstract
Simultaneous Magnetic Resonance Imaging (MRI) and Positron Emission Tomography (PET) scanning is a recent major development in biomedical imaging. The full integration of the PET detector ring and electronics within the MR system has been a technologically challenging design to develop but provides capacity for simultaneous imaging and the potential for new diagnostic and research capability. This article reviews state-of-the-art MR-PET hardware and software, and discusses future developments focusing on neuroimaging methodologies for MR-PET scanning. We particularly focus on the methodologies that lead to an improved synergy between MRI and PET, including optimal data acquisition, PET attenuation and motion correction, and joint image reconstruction and processing methods based on the underlying complementary and mutual information. We further review the current and potential future applications of simultaneous MR-PET in both systems neuroscience and clinical neuroimaging research. We demonstrate a simultaneous data acquisition protocol to highlight new applications of MR-PET neuroimaging research studies.
Collapse
Affiliation(s)
- Zhaolin Chen
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Department of Electrical and Computer Systems Engineering, Monash University, Clayton, Victoria, Australia
| | - Sharna D Jamadar
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Clayton, Victoria, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Monash University, Clayton, Victoria, Australia
| | - Shenpeng Li
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Department of Electrical and Computer Systems Engineering, Monash University, Clayton, Victoria, Australia
| | | | - Jakub Baran
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Department of Biophysics, Faculty of Mathematics and Natural Sciences, University of Rzeszów, Rzeszów, Poland
| | - Nicholas Ferris
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Monash Imaging, Monash Health, Clayton, Victoria, Australia
| | - Nadim Jon Shah
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum, Jülich, Germany
| | - Gary F Egan
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Clayton, Victoria, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
24
|
Sakata M, Ishibashi K, Imai M, Wagatsuma K, Ishii K, Hatano K, Ishiwata K, Toyohara J. Assessment of safety, efficacy, and dosimetry of a novel 18-kDa translocator protein ligand, [ 11C]CB184, in healthy human volunteers. EJNMMI Res 2017; 7:26. [PMID: 28337723 PMCID: PMC5364125 DOI: 10.1186/s13550-017-0271-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 03/01/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND N,N-di-n-propyl-2-[2-(4-[11C]methoxyphenyl)-6,8-dichloroimidazol[1,2-a]pyridine-3-yl]acetamide ([11C]CB184) is a novel selective radioligand for the 18-kD translocator protein (TSPO), which is upregulated in activated microglia in the brain, and may be useful in positron emission tomography (PET). We examined the safety, radiation dosimetry, and initial brain imaging with [11C]CB184 in healthy human volunteers. RESULTS Dynamic [11C]CB184 PET scans (90 min) were performed in five healthy male subjects. During the scan, arterial blood was sampled at various time intervals, and the fraction of the parent compound in plasma was determined with high-performance liquid chromatography. No serious adverse events occurred in any of the subjects throughout the study period. [11C]CB184 was metabolized in the periphery: 36.7% ± 5.7% of the radioactivity in plasma was detected as the unchanged form after 60 min. The total distribution volume (V T) was estimated with a two-tissue compartment model. The V T of [11C]CB184 was highest in the thalamus (5.1 ± 0.4), followed by the cerebellar cortex (4.4 ± 0.2), and others. Although regional differences were small, the observed [11C]CB184 binding pattern was consistent with the TSPO distribution in the normal human brain. Radiation dosimetry was determined in three healthy male subjects using a serial whole-body PET scan acquired over 2 h after [11C]CB184 injection. [11C]CB184 PET demonstrated high uptake in the gallbladder at a later time (>60 min). In urine obtained approximately 100 min post-injection, 0.3% of the total injected radioactivity was recovered, indicating hepatobiliary excretion of radioactivity. The absorbed dose (μGy/MBq) was highest in the kidneys (21.0 ± 0.5) followed by the lungs (16.8 ± 2.7), spleen (16.6 ± 6.6), and pancreas (16.5 ± 2.2). The estimated effective dose for [11C]CB184 was 5.9 ± 0.6 μSv/MBq. CONCLUSIONS This initial evaluation indicated that [11C]CB184 is feasible for imaging of TSPO in the brain.
Collapse
Affiliation(s)
- Muneyuki Sakata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, 173-0015 Tokyo, Japan
| | - Kenji Ishibashi
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, 173-0015 Tokyo, Japan
| | - Masamichi Imai
- Department of Radiology, Toranomon Hospital, Tokyo, Japan
| | - Kei Wagatsuma
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, 173-0015 Tokyo, Japan
| | - Kenji Ishii
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, 173-0015 Tokyo, Japan
| | - Kentaro Hatano
- Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kiichi Ishiwata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, 173-0015 Tokyo, Japan
- Institute of Cyclotron and Drug Discovery Research, Southern TOHOKU Research Institute for Neuroscience, Koriyama, Japan
- Department of Biofunctional Imaging, Fukushima Medical University, Fukushima, Japan
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, 173-0015 Tokyo, Japan
| |
Collapse
|
25
|
Alam MM, Lee J, Lee SY. Recent Progress in the Development of TSPO PET Ligands for Neuroinflammation Imaging in Neurological Diseases. Nucl Med Mol Imaging 2017; 51:283-296. [PMID: 29242722 DOI: 10.1007/s13139-017-0475-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/08/2016] [Accepted: 02/06/2017] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammation is heavily associated with various neurological diseases including Alzheimer's disease, Parkinson's disease, multiple sclerosis, and stroke. It is strongly characterized by the activation of microglia which can be visualized using position emission tomography (PET). Traditionally, translocator protein 18 kDa (TSPO) has been the preferred target for imaging the inflammatory progression of the microglial component. TSPO is expressed in the outer mitochondrial membrane and present in very low concentrations in the healthy human brain, but is markedly upregulated in response to brain injury and inflammation. Due to its value as a marker of microglial activation and subsequent utility for evaluating neuroinflammation in CNS disorders, several classes of TSPO radioligands have been developed and evaluated. However, the application of these second-generation TSPO radiotracers has been subject to several limiting factors, including a polymorphism that affects TSPO binding. This review focuses on recent developments in TSPO imaging, as well as current limitations and suggestions for future directions from a medical imaging perspective.
Collapse
Affiliation(s)
- Md Maqusood Alam
- Neuroscience Research Institute, Gachon University, Incheon, 20565 South Korea
| | - Jihye Lee
- Neuroscience Research Institute, Gachon University, Incheon, 20565 South Korea
| | - Sang-Yoon Lee
- Neuroscience Research Institute, Gachon University, Incheon, 20565 South Korea.,Department of Neuroscience, College of Medicine, Gachon University, Incheon, 21936 South Korea
| |
Collapse
|
26
|
Li J, Smith JA, Dawson ES, Fu A, Nickels ML, Schulte ML, Manning HC. Optimized Translocator Protein Ligand for Optical Molecular Imaging and Screening. Bioconjug Chem 2017; 28:1016-1023. [PMID: 28156095 DOI: 10.1021/acs.bioconjchem.6b00711] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Translocator protein (TSPO) is a validated target for molecular imaging of a variety of human diseases and disorders. Given its involvement in cholesterol metabolism, TSPO expression is commonly elevated in solid tumors, including glioma, colorectal cancer, and breast cancer. TSPO ligands capable of detection by optical imaging are useful molecular tracers for a variety of purposes that range from quantitative biology to drug discovery. Leveraging our prior optimization of the pyrazolopyrimidine TSPO ligand scaffold for cancer imaging, we report herein a new generation of TSPO tracers with superior binding affinity and suitability for optical imaging and screening. In total, seven candidate TSPO tracers were synthesized and vetted in this study; the most promising tracer identified (29, Kd = 0.19 nM) was the result of conjugating a high-affinity TSPO ligand to a fluorophore used routinely in biological sciences (FITC) via a functional carbon linker of optimal length. Computational modeling suggested that an n-alkyl linker of eight carbons in length allows for positioning of the bulky fluorophore distal to the ligand binding domain and toward the solvent interface, minimizing potential ligand-protein interference. Probe 29 was found to be highly suitable for in vitro imaging of live TSPO-expressing cells and could be deployed as a ligand screening and discovery tool. Competitive inhibition of probe 29 quantified by fluorescence and 3H-PK11195 quantified by traditional radiometric detection resulted in equivalent affinity data for two previously reported TSPO ligands. This study introduces the utility of TSPO ligand 29 for in vitro imaging and screening and provides a structural basis for the development of future TSPO imaging ligands bearing bulky signaling moieties.
Collapse
Affiliation(s)
- Jun Li
- Interdisciplinary Materials Science Program, ∥Vanderbilt University Center for Structural Biology, and ■Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee 37232, United States.,Vanderbilt University Institute of Imaging Science (VUIIS), §Center for Molecular Probes, ⊥Department of Radiology and Radiological Sciences, #Department of Biochemistry, ¶Vanderbilt-Ingram Cancer Center (VICC), and ▽Department of Neurosurgery, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Jarrod A Smith
- Interdisciplinary Materials Science Program, ∥Vanderbilt University Center for Structural Biology, and ■Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee 37232, United States.,Vanderbilt University Institute of Imaging Science (VUIIS), §Center for Molecular Probes, ⊥Department of Radiology and Radiological Sciences, #Department of Biochemistry, ¶Vanderbilt-Ingram Cancer Center (VICC), and ▽Department of Neurosurgery, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Eric S Dawson
- Interdisciplinary Materials Science Program, ∥Vanderbilt University Center for Structural Biology, and ■Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee 37232, United States.,Vanderbilt University Institute of Imaging Science (VUIIS), §Center for Molecular Probes, ⊥Department of Radiology and Radiological Sciences, #Department of Biochemistry, ¶Vanderbilt-Ingram Cancer Center (VICC), and ▽Department of Neurosurgery, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Allie Fu
- Interdisciplinary Materials Science Program, ∥Vanderbilt University Center for Structural Biology, and ■Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee 37232, United States.,Vanderbilt University Institute of Imaging Science (VUIIS), §Center for Molecular Probes, ⊥Department of Radiology and Radiological Sciences, #Department of Biochemistry, ¶Vanderbilt-Ingram Cancer Center (VICC), and ▽Department of Neurosurgery, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Michael L Nickels
- Interdisciplinary Materials Science Program, ∥Vanderbilt University Center for Structural Biology, and ■Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee 37232, United States.,Vanderbilt University Institute of Imaging Science (VUIIS), §Center for Molecular Probes, ⊥Department of Radiology and Radiological Sciences, #Department of Biochemistry, ¶Vanderbilt-Ingram Cancer Center (VICC), and ▽Department of Neurosurgery, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Michael L Schulte
- Interdisciplinary Materials Science Program, ∥Vanderbilt University Center for Structural Biology, and ■Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee 37232, United States.,Vanderbilt University Institute of Imaging Science (VUIIS), §Center for Molecular Probes, ⊥Department of Radiology and Radiological Sciences, #Department of Biochemistry, ¶Vanderbilt-Ingram Cancer Center (VICC), and ▽Department of Neurosurgery, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - H Charles Manning
- Interdisciplinary Materials Science Program, ∥Vanderbilt University Center for Structural Biology, and ■Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee 37232, United States.,Vanderbilt University Institute of Imaging Science (VUIIS), §Center for Molecular Probes, ⊥Department of Radiology and Radiological Sciences, #Department of Biochemistry, ¶Vanderbilt-Ingram Cancer Center (VICC), and ▽Department of Neurosurgery, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| |
Collapse
|
27
|
Lee J, Jung JH, Lee BC, Lee SY. Design and Synthesis of Phenoxypyridyl Acetamide or Aryl-oxodihydropurine Derivatives for the Development of Novel PET Ligands Targeting the Translocator Protein 18 kDa (TSPO). B KOREAN CHEM SOC 2016. [DOI: 10.1002/bkcs.10979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Jihye Lee
- Neuroscience Research Institute; Gachon University; Incheon 21565 Korea
| | - Jae Ho Jung
- Department of Nuclear Medicine; Seoul National University College of Medicine, Seoul National University Bundang Hospital; Seongnam 13620 Korea
| | - Byung Chul Lee
- Department of Nuclear Medicine; Seoul National University College of Medicine, Seoul National University Bundang Hospital; Seongnam 13620 Korea
- Center for Nanomolecular Imaging and Innovative Drug Development; Advanced Institutes of Convergence Technology; Suwon 16229 Korea
| | - Sang-Yoon Lee
- Neuroscience Research Institute; Gachon University; Incheon 21565 Korea
- Department of Neuroscience, College of Medicine; Gachon University; Incheon 21936 Korea
| |
Collapse
|
28
|
Toyohara J, Sakata M, Hatano K, Yanai S, Endo S, Ishibashi K, Wagatsuma K, Ishii K, Ishiwata K. Preclinical and first-in-man studies of [(11)C]CB184 for imaging the 18-kDa translocator protein by positron emission tomography. Ann Nucl Med 2016; 30:534-543. [PMID: 27329083 DOI: 10.1007/s12149-016-1094-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/30/2016] [Indexed: 01/17/2023]
Abstract
OBJECTIVE We performed preclinical and first-in-man clinical positron emission tomography (PET) studies in human brain using N,N-di-n-propyl-2-[2-(4-[(11)C]methoxyphenyl)-6,8-dichloroimidazol[1,2-a]pyridine-3-yl]acetamide ([(11)C]CB184) to image the 18-kDa translocator protein (TSPO), which is overexpressed in activated microglia in neuroinflammatory conditions. METHODS In vitro selectivity of CB184 was characterized. The radiation absorbed dose by [(11)C]CB184 in humans was calculated from murine distribution data. Acute toxicity of CB184 hydrochloride in rats at a dose of 5.81 mg/kg body weight, which is >10,000-fold higher than the clinical equivalent dose of [(11)C]CB184, was evaluated. Acute toxicity of [(11)C]CB184 injection of a 400-fold dose to administer a postulated dose of 740 MBq [(11)C]CB184 was also evaluated after the decay-out of (11)C. The mutagenicity of CB184 was studied with a reverse mutation test (Ames test). The pharmacological effect of CB184 injection in mice was studied with an open field test. The first PET imaging of TSPO with [(11)C]CB184 in a normal human volunteer was performed. RESULTS A suitable preparation method for [(11)C]CB184 injection was established. CB184 showed low activity in a 28-standard receptor binding profile. The radiation absorbed dose by [(11)C]CB184 in humans was sufficiently low for clinical use, and no acute toxicity of CB184 or [(11)C]CB184 injection was found. No mutagenicity or apparent effect on locomotor activity or anxiety status was observed for CB184. We safely performed brain imaging with PET following administration of [(11)C]CB184 in a normal human volunteer. A 90-min dynamic scan showed rapid initial uptake of radioactivity in the brain followed by prompt clearance. [(11)C]CB184 was homogeneously distributed in the gray matter. The total distribution volume of [(11)C]CB184 was highest in the thalamus followed by the cerebellar cortex and elsewhere. Although regional differences were small, the observed [(11)C]CB184 binding pattern was consistent with the TSPO distribution in normal human brain. Peripherally, [(11)C]CB184 was metabolized in humans: 30 % of the radioactivity in plasma was detected as the unchanged form after 60 min. CONCLUSIONS [(11)C]CB184 is suitable for imaging TSPO in human brain and provides an acceptable radiation dose. Pharmacological safety was noted at the dose required for PET imaging.
Collapse
Affiliation(s)
- Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan.
| | - Muneyuki Sakata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Kentaro Hatano
- Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shuichi Yanai
- Research Team for Aging Neuroscience, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Shogo Endo
- Research Team for Aging Neuroscience, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Kenji Ishibashi
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Kei Wagatsuma
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Kenji Ishii
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Kiichi Ishiwata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- Institute of Cyclotron and Drug Discovery Research, Southern Tohoku Research Institute for Neuroscience, Koriyama, Japan
- Department of Biofunctional Imaging, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
29
|
Tripodo G, Trapani A, Torre ML, Giammona G, Trapani G, Mandracchia D. Hyaluronic acid and its derivatives in drug delivery and imaging: Recent advances and challenges. Eur J Pharm Biopharm 2016; 97:400-16. [PMID: 26614559 DOI: 10.1016/j.ejpb.2015.03.032] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/21/2015] [Accepted: 03/23/2015] [Indexed: 01/06/2023]
Abstract
Hyaluronic acid (HA) is a biodegradable, biocompatible, nontoxic, and non-immunogenic glycosaminoglycan used for various biomedical applications. The interaction of HA with the CD44 receptor, whose expression is elevated on the surface of many types of tumor cells, makes this polymer a promising candidate for intracellular delivery of imaging and anticancer agents exploiting a receptor-mediated active targeting strategy. Therefore, HA and its derivatives have been most investigated for the development of several carrier systems intended for cancer diagnosis and therapy. Nonetheless, different and important delivery applications of the polysaccharide have also been described, including gene and peptide/protein drugs delivery. The aim of this review was to provide an overview of the existing recent literature on the use of HA and its derivatives for drug delivery and imaging. Notable attention is given to nanotheranostic systems obtained after conjugation of HA to nanocarriers as quantum dots, carbon nanotubes and graphene. Meanwhile, attention is also paid to some challenging aspects that need to be addressed in order to allow translation of preclinical models based on HA and its derivatives for drug delivery and imaging purposes to clinical testing and further their development.
Collapse
Affiliation(s)
- Giuseppe Tripodo
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Adriana Trapani
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | - Maria Luisa Torre
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Gaetano Giammona
- Department of "Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF)", University of Palermo, via Archirafi 32, Palermo 90123, Italy
| | - Giuseppe Trapani
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy
| | - Delia Mandracchia
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", via Orabona 4, 70125 Bari, Italy.
| |
Collapse
|
30
|
Changes in Binding of [(123)I]CLINDE, a High-Affinity Translocator Protein 18 kDa (TSPO) Selective Radioligand in a Rat Model of Traumatic Brain Injury. Neuromolecular Med 2016; 18:158-69. [PMID: 26969181 DOI: 10.1007/s12017-016-8385-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/02/2016] [Indexed: 01/01/2023]
Abstract
After traumatic brain injury (TBI), secondary injuries develop, including neuroinflammatory processes that contribute to long-lasting impairments. These secondary injuries represent potential targets for treatment and diagnostics. The translocator protein 18 kDa (TSPO) is expressed in activated microglia cells and upregulated in response to brain injury and therefore a potential biomarker of the neuroinflammatory processes. Second-generation radioligands of TSPO, such as [(123)I]CLINDE, have a higher signal-to-noise ratio as the prototype ligand PK11195. [(123)I]CLINDE has been employed in human studies using single-photon emission computed tomography to image the neuroinflammatory response after stroke. In this study, we used the same tracer in a rat model of TBI to determine changes in TSPO expression. Adult Sprague-Dawley rats were subjected to moderate controlled cortical impact injury and sacrificed at 6, 24, 72 h and 28 days post surgery. TSPO expression was assessed in brain sections employing [(123)I]CLINDE in vitro autoradiography. From 24 h to 28 days post surgery, injured animals exhibited a marked and time-dependent increase in [(123)I]CLINDE binding in the ipsilateral motor, somatosensory and parietal cortex, as well as in the hippocampus and thalamus. Interestingly, binding was also significantly elevated in the contralateral M1 motor cortex following TBI. Craniotomy without TBI caused a less marked increase in [(123)I]CLINDE binding, restricted to the ipsilateral hemisphere. Radioligand binding was consistent with an increase in TSPO mRNA expression and CD11b immunoreactivity at the contusion site. This study demonstrates the applicability of [(123)I]CLINDE for detailed regional and quantitative assessment of glial activity in experimental models of TBI.
Collapse
|
31
|
Parente A, Feltes PK, Vállez García D, Sijbesma JW, Moriguchi Jeckel CM, Dierckx RA, de Vries EF, Doorduin J. Pharmacokinetic Analysis of 11C-PBR28 in the Rat Model of Herpes Encephalitis: Comparison with (R)-11C-PK11195. J Nucl Med 2016; 57:785-91. [DOI: 10.2967/jnumed.115.165019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/04/2015] [Indexed: 01/21/2023] Open
|
32
|
Magnetic Resonance Spectroscopy discriminates the response to microglial stimulation of wild type and Alzheimer's disease models. Sci Rep 2016; 6:19880. [PMID: 26813748 PMCID: PMC4728482 DOI: 10.1038/srep19880] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/18/2015] [Indexed: 11/26/2022] Open
Abstract
Microglia activation has emerged as a potential key factor in the pathogenesis of Alzheimer’s disease. Metabolite levels assessed by magnetic resonance spectroscopy (MRS) are used as markers of neuroinflammation in neurodegenerative diseases, but how they relate to microglial activation in health and chronic disease is incompletely understood. Using MRS, we monitored the brain metabolic response to lipopolysaccharides (LPS)-induced microglia activation in vivo in a transgenic mouse model of Alzheimer’s disease (APP/PS1) and healthy controls (wild-type (WT) littermates) over 4 hours. We assessed reactive gliosis by immunohistochemistry and correlated metabolic and histological measures. In WT mice, LPS induced a microglial phenotype consistent with activation, associated with a sustained increase in macromolecule and lipid levels (ML9). This effect was not seen in APP/PS1 mice, where LPS did not lead to a microglial response measured by histology, but induced a late increase in the putative inflammation marker myoinositol (mI) and metabolic changes in total creatine and taurine previously reported to be associated with amyloid load. We argue that ML9 and mI distinguish the response of WT and APP/PS1 mice to immune mediators. Lipid and macromolecule levels may represent a biomarker of activation of healthy microglia, while mI may not be a glial marker.
Collapse
|
33
|
Vivash L, O'Brien TJ. Imaging Microglial Activation with TSPO PET: Lighting Up Neurologic Diseases? J Nucl Med 2015; 57:165-8. [PMID: 26697963 DOI: 10.2967/jnumed.114.141713] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 12/22/2015] [Indexed: 11/16/2022] Open
Abstract
Neuroinflammation is implicated in the pathogenesis of a wide range of neurologic and neuropsychiatric diseases. For over 20 years, (11)C-PK11195 PET, which aims to image expression of the translocator protein (TSPO) on activated microglia in the brain, has been used in preclinical and clinical research to investigate neuroinflammation in vivo in patients with brain diseases. However, (11)C-PK11195 suffers from two major limitations: its low brain permeability and high nonspecific and plasma binding results in a low signal-to-noise ratio, and the use of (11)C restricts its use to PET research centers and hospitals with an on-site cyclotron. In recent years, there has been a great deal of work into the development of new TSPO-specific PET radiotracers. This work has focused on fluorinated radiotracers, which would enable wider use and improved signal-to-noise ratios. These radiotracers have been utilized in preclinical and clinical studies of several neurologic diseases with varying degrees of success. Unfortunately, the application of these second-generation TSPO radiotracers has revealed additional problems, including a polymorphism that affects TSPO binding. In this review, the developments in TSPO imaging are discussed, and current limitations and suggestions for future directions are explored.
Collapse
Affiliation(s)
- Lucy Vivash
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Royal Parade, Parkville, Victoria, Australia
| | - Terence J O'Brien
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Royal Parade, Parkville, Victoria, Australia
| |
Collapse
|
34
|
Abstract
The 3D structure of the 18-kDa transmembrane (TM) protein TSPO (translocator protein)/PBR (peripheral benzodiazepine receptor), which contains a binding site for benzodiazepines, is important to better understand its function and regulation by endogenous and synthetic ligands. We have recently determined the structure of mammalian TSPO/PBR in complex with the diagnostic ligand PK11195 [1-(2-chlorophenyl)-N-methyl-N-(1-methylpropyl)-3-isoquinolinecarboxamide; Jaremko et al. (2014) Science 343: , 1363-1366], providing for the first time atomic-level insight into the conformation of this protein, which is up-regulated in various pathological conditions including Alzheimer's disease and Parkinson's disease. Here, we review the studies which have probed the structural properties of mammalian TSPO/PBR as well as the homologues bacterial tryptophan-rich sensory proteins (TspOs) over the years and provide detailed insight into the 3D structure of mouse TSPO (mTSPO)/PBR in complex with PK11195.
Collapse
|
35
|
Targeting the 18-kDa translocator protein: recent perspectives for neuroprotection. Biochem Soc Trans 2015; 43:559-65. [DOI: 10.1042/bst20150028] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Indexed: 12/14/2022]
Abstract
The translocator protein (TSPO, 18 kDa), mainly localized in the outer mitochondrial membrane of steroidogenic tissues, is involved in several cellular functions. TSPO level alterations have been reported in a number of human disorders, particularly in cancer, psychiatric and neurological diseases. In the central nervous system (CNS), TSPO is usually expressed in glial cells, but also in some neuronal cell types. Interestingly, the expression of TSPO on glial cells rises after brain injury and increased TSPO expression is often observed in neurological disorders, gliomas, encephalitis and traumatic injury. Since TSPO is up-regulated in brain diseases, several structurally different classes of ligands targeting TSPO have been described as potential diagnostic or therapeutic agents. Recent researches have reported that TSPO ligands might be valuable in the treatment of brain diseases. This review focuses on currently available TSPO ligands, as useful tools for the treatment of neurodegeneration, neuro-inflammation and neurotrauma.
Collapse
|
36
|
Barresi E, Bruno A, Taliani S, Cosconati S, Da Pozzo E, Salerno S, Simorini F, Daniele S, Giacomelli C, Marini AM, La Motta C, Marinelli L, Cosimelli B, Novellino E, Greco G, Da Settimo F, Martini C. Deepening the Topology of the Translocator Protein Binding Site by Novel N,N-Dialkyl-2-arylindol-3-ylglyoxylamides. J Med Chem 2015; 58:6081-92. [PMID: 26177193 DOI: 10.1021/acs.jmedchem.5b00689] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
As a continuation of our studies on 2-phenylindol-3-ylglyoxylamides as potent and selective translocator protein (TSPO) ligands, two subsets of novel derivatives, featuring hydrophilic group (OH, NH2, COOH) at the para-position of the pendent 2-phenyl ring (8-16) or different 2-aryl moieties, namely, 3-thienyl, p-biphenyl, 2-naphthyl (23-35), were synthesized and biologically evaluated, some of them showing Ki values in the subnanomolar range and the 2-naphthyl group performance being the best. The resulting SARs confirmed the key role played by interactions taking place between ligands and the lipophilic L1 pocket of the TSPO binding site. Docking simulations were performed on the most potent compound of the present series (29) exploiting the recently available 3D structures of TSPO bound to its standard ligand (PK11195). Our theoretical model was fully consistent with SARs of the newly investigated as well of the previously reported 2-phenylindol-3-ylglyoxylamide derivatives.
Collapse
Affiliation(s)
- Elisabetta Barresi
- †Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Agostino Bruno
- ‡Dipartimento di Farmacia, Università di Napoli "Federico II", Via D. Montesano 49, 80131 Napoli, Italy
| | - Sabrina Taliani
- †Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Sandro Cosconati
- §DiSTABiF, Seconda Università di Napoli, Via Vivaldi 43, 81100 Caserta, Italy
| | - Eleonora Da Pozzo
- †Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Silvia Salerno
- †Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Francesca Simorini
- †Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Simona Daniele
- †Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Chiara Giacomelli
- †Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Anna Maria Marini
- †Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Concettina La Motta
- †Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Luciana Marinelli
- ‡Dipartimento di Farmacia, Università di Napoli "Federico II", Via D. Montesano 49, 80131 Napoli, Italy
| | - Barbara Cosimelli
- ‡Dipartimento di Farmacia, Università di Napoli "Federico II", Via D. Montesano 49, 80131 Napoli, Italy
| | - Ettore Novellino
- ‡Dipartimento di Farmacia, Università di Napoli "Federico II", Via D. Montesano 49, 80131 Napoli, Italy
| | - Giovanni Greco
- ‡Dipartimento di Farmacia, Università di Napoli "Federico II", Via D. Montesano 49, 80131 Napoli, Italy
| | - Federico Da Settimo
- †Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Claudia Martini
- †Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| |
Collapse
|
37
|
Enigmatic Translocator protein (TSPO) and cellular stress regulation. Trends Biochem Sci 2015; 40:497-503. [PMID: 26228316 DOI: 10.1016/j.tibs.2015.07.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/25/2015] [Accepted: 07/06/2015] [Indexed: 11/23/2022]
Abstract
Translocator proteins (TSPOs) are conserved, ubiquitous membrane proteins identified initially as benzodiazepine-binding proteins in mammalian cells. Recent genetic and biochemical studies have challenged the accepted model that TSPOs are essential and required for steroidogenesis in animal cells. Instead, evidence from different kingdoms of life suggests that TSPOs are encoded by nonessential genes that are temporally upregulated in cells encountering conditions of oxidative stress, including inflammation and tissue injury. Here we discuss how TSPOs may be involved in complex homeostasis signaling mechanisms. We suggest that the main physiological role of TSPOs may be to modulate oxidative stress, irrespective of the cell type or subcellular localization, in part through the subtle regulation of tetrapyrrole metabolism.
Collapse
|
38
|
Jaremko M, Jaremko Ł, Giller K, Becker S, Zweckstetter M. Structural Integrity of the A147T Polymorph of Mammalian TSPO. Chembiochem 2015; 16:1483-9. [PMID: 25974690 DOI: 10.1002/cbic.201500217] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Indexed: 01/27/2023]
Abstract
Ligands of the transmembrane protein TSPO are used for imaging of brain inflammation, but a common polymorphism in TSPO complicates their application to humans. Here we determined the three-dimensional structure and side-chain dynamics of the A147T polymorph of mammalian TSPO in complex with the first-generation ligand PK11195. We show that A147T TSPO is able to retain the same structural and dynamic profile as the wild-type protein and thus binds PK11195 with comparable affinity. Our study is important for the design of more potent diagnostic and therapeutic ligands of TSPO.
Collapse
Affiliation(s)
- Mariusz Jaremko
- Department for NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen (Germany)
| | - Łukasz Jaremko
- Department for NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen (Germany).,German Center for Neurodegenerative Diseases (DZNE), 37077 Göttingen (Germany)
| | - Karin Giller
- Department for NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen (Germany)
| | - Stefan Becker
- Department for NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen (Germany)
| | - Markus Zweckstetter
- Department for NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen (Germany). .,German Center for Neurodegenerative Diseases (DZNE), 37077 Göttingen (Germany). .,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, 37073 Göttingen (Germany).
| |
Collapse
|
39
|
Vállez Garcia D, de Vries EFJ, Toyohara J, Ishiwata K, Hatano K, Dierckx RAJO, Doorduin J. Evaluation of [(11)C]CB184 for imaging and quantification of TSPO overexpression in a rat model of herpes encephalitis. Eur J Nucl Med Mol Imaging 2015; 42:1106-1118. [PMID: 25771904 PMCID: PMC4424274 DOI: 10.1007/s00259-015-3021-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/16/2015] [Indexed: 02/04/2023]
Abstract
PURPOSE Evaluation of translocator protein (TSPO) overexpression is considered an attractive research tool for monitoring neuroinflammation in several neurological and psychiatric disorders. [(11)C]PK11195 PET imaging has been widely used for this purpose. However, it has a low sensitivity and a poor signal-to-noise ratio. For these reasons, [(11)C]CB184 was evaluated as a potentially more sensitive PET tracer. METHODS A model of herpes simplex encephalitis (HSE) was induced in male Wistar rats. On day 6 or 7 after virus inoculation, [(11)C]CB184 PET scans were acquired followed by ex vivo evaluation of biodistribution. In addition, [(11)C]CB184 and [(11)C]PK11195 PET scans with arterial blood sampling were acquired to generate input for pharmacokinetic modelling. Differences between the saline-treated control group and the virus-treated HSE group were explored using volumes of interest and voxel-based analysis. RESULTS The biodistribution study showed significantly higher [(11)C]CB184 uptake in the amygdala, olfactory bulb, medulla, pons and striatum (p < 0.05) in HSE rats than in control rats, and the voxel-based analysis showed higher bilateral uptake in the pons and medulla (p < 0.05, corrected at the cluster level). A high correlation was found between tracer uptake in the biodistribution study and on the PET scans (p < 0.001, r (2) = 0.71). Pretreatment with 5 mg/kg of unlabelled PK11195 effectively reduced (p < 0.001) [(11)C]CB184 uptake in the whole brain. Both, [(11)C]CB184 and [(11)C]PK11195, showed similar amounts of metabolites in plasma, and the binding potential (BPND) was not significantly different between the HSE rats and the control rats. In HSE rats BPND for [(11)C]CB184 was significantly higher (p < 0.05) in the amygdala, hypothalamus, medulla, pons and septum than in control rats, whereas higher uptake of [(11)C]PK11195 was only detected in the medulla. CONCLUSION [(11)C]CB184 showed nonspecific binding to healthy tissue comparable to that observed for [(11)C]PK11195, but it displayed significantly higher specific binding in those brain regions affected by the HSE. Our results suggest that [(11)C]CB184 PET is a good alternative for imaging of neuroinflammatory processes.
Collapse
Affiliation(s)
- David Vállez Garcia
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Erik F. J. de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, 173-0015 Japan
| | - Kiichi Ishiwata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, 173-0015 Japan
| | - Kentaro Hatano
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8522 Japan
| | - Rudi A. J. O. Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
40
|
Tiwari AK, Ji B, Yui J, Fujinaga M, Yamasaki T, Xie L, Luo R, Shimoda Y, Kumata K, Zhang Y, Hatori A, Maeda J, Higuchi M, Wang F, Zhang MR. [18F]FEBMP: Positron Emission Tomography Imaging of TSPO in a Model of Neuroinflammation in Rats, and in vitro Autoradiograms of the Human Brain. Am J Cancer Res 2015; 5:961-9. [PMID: 26155312 PMCID: PMC4493534 DOI: 10.7150/thno.12027] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/09/2015] [Indexed: 12/13/2022] Open
Abstract
We evaluated the efficacy of 2-[5-(4-[18F]fluoroethoxy-2-oxo-1,3-benzoxazol-3(2H)-yl)-N-methyl-N-phenylacetamide] ([18F]FEBMP) for positron emission tomography (PET) imaging of translocator protein (18 kDa, TSPO). Dissection was used to determine the distribution of [18F]FEBMP in mice, while small-animal PET and metabolite analysis were used for a rat model of focal cerebral ischemia. [18F]FEBMP showed high radioactivity uptake in mouse peripheral organs enriched with TSPO, and relatively high initial brain uptake (2.67 ± 0.12% ID/g). PET imaging revealed an increased accumulation of radioactivity in the infarcted striatum, with a maximum ratio of 3.20 ± 0.12, compared to non-injured striatum. Displacement with specific TSPO ligands lowered the accumulation levels in infarcts to those on the contralateral side. This suggests that the increased accumulation reflected TPSO-specific binding of [18F]FEBMP in vivo. Using a simplified reference tissue model, the binding potential on the infarcted area was 2.72 ± 0.27. Metabolite analysis in brain tissues showed that 83.2 ± 7.4% and 76.4 ± 2.1% of radioactivity was from intact [18F]FEBMP at 30 and 60 min, respectively, and that this ratio was higher than in plasma (8.6 ± 1.9% and 3.9 ± 1.1%, respectively). In vitro autoradiography on postmortem human brains showed that TSPO rs6971 polymorphism did not affect binding sites for [18F]FEBMP. These findings suggest that [18F]FEBMP is a promising new tool for visualization of neuroinflammation.
Collapse
|
41
|
Bosco A, Romero CO, Ambati BK, Vetter ML. In vivo dynamics of retinal microglial activation during neurodegeneration: confocal ophthalmoscopic imaging and cell morphometry in mouse glaucoma. J Vis Exp 2015:e52731. [PMID: 25992962 DOI: 10.3791/52731] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Microglia, which are CNS-resident neuroimmune cells, transform their morphology and size in response to CNS damage, switching to an activated state with distinct functions and gene expression profiles. The roles of microglial activation in health, injury and disease remain incompletely understood due to their dynamic and complex regulation in response to changes in their microenvironment. Thus, it is critical to non-invasively monitor and analyze changes in microglial activation over time in the intact organism. In vivo studies of microglial activation have been delayed by technical limitations to tracking microglial behavior without altering the CNS environment. This has been particularly challenging during chronic neurodegeneration, where long-term changes must be tracked. The retina, a CNS organ amenable to non-invasive live imaging, offers a powerful system to visualize and characterize the dynamics of microglia activation during chronic disorders. This protocol outlines methods for long-term, in vivo imaging of retinal microglia, using confocal ophthalmoscopy (cSLO) and CX3CR1(GFP/+) reporter mice, to visualize microglia with cellular resolution. Also, we describe methods to quantify monthly changes in cell activation and density in large cell subsets (200-300 cells per retina). We confirm the use of somal area as a useful metric for live tracking of microglial activation in the retina by applying automated threshold-based morphometric analysis of in vivo images. We use these live image acquisition and analyses strategies to monitor the dynamic changes in microglial activation and microgliosis during early stages of retinal neurodegeneration in a mouse model of chronic glaucoma. This approach should be useful to investigate the contributions of microglia to neuronal and axonal decline in chronic CNS disorders that affect the retina and optic nerve.
Collapse
Affiliation(s)
| | - Cesar O Romero
- Department of Neurobiology & Anatomy, University of Utah
| | | | | |
Collapse
|
42
|
Bosco A, Romero CO, Breen KT, Chagovetz AA, Steele MR, Ambati BK, Vetter ML. Neurodegeneration severity can be predicted from early microglia alterations monitored in vivo in a mouse model of chronic glaucoma. Dis Model Mech 2015; 8:443-55. [PMID: 25755083 PMCID: PMC4415894 DOI: 10.1242/dmm.018788] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/26/2015] [Indexed: 12/30/2022] Open
Abstract
Microglia serve key homeostatic roles, and respond to neuronal perturbation and decline with a high spatiotemporal resolution. The course of all chronic CNS pathologies is thus paralleled by local microgliosis and microglia activation, which begin at early stages of the disease. However, the possibility of using live monitoring of microglia during early disease progression to predict the severity of neurodegeneration has not been explored. Because the retina allows live tracking of fluorescent microglia in their intact niche, here we investigated their early changes in relation to later optic nerve neurodegeneration. To achieve this, we used the DBA/2J mouse model of inherited glaucoma, which develops progressive retinal ganglion cell degeneration of variable severity during aging, and represents a useful model to study pathogenic mechanisms of retinal ganglion cell decline that are similar to those in human glaucoma. We imaged CX3CR1(+/GFP) microglial cells in vivo at ages ranging from 1 to 5 months by confocal scanning laser ophthalmoscopy (cSLO) and quantified cell density and morphological activation. We detected early microgliosis at the optic nerve head (ONH), where axonopathy first manifests, and could track attenuation of this microgliosis induced by minocycline. We also observed heterogeneous and dynamic patterns of early microglia activation in the retina. When the same animals were aged and analyzed for the severity of optic nerve pathology at 10 months of age, we found a strong correlation with the levels of ONH microgliosis at 3 to 4 months. Our findings indicate that live imaging and monitoring the time course and levels of early retinal microgliosis and microglia activation in glaucoma could serve as indicators of future neurodegeneration severity.
Collapse
Affiliation(s)
- Alejandra Bosco
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
| | - Cesar O Romero
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
| | - Kevin T Breen
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
| | - Alexis A Chagovetz
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84132, USA
| | - Michael R Steele
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
| | - Balamurali K Ambati
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT 84132, USA
| | - Monica L Vetter
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
| |
Collapse
|
43
|
Banister SD, Beinat C, Wilkinson SM, Shen B, Bartoli C, Selleri S, Da Pozzo E, Martini C, Chin FT, Kassiou M. Ether analogues of DPA-714 with subnanomolar affinity for the translocator protein (TSPO). Eur J Med Chem 2015; 93:392-400. [DOI: 10.1016/j.ejmech.2015.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 02/04/2015] [Accepted: 02/05/2015] [Indexed: 12/13/2022]
|
44
|
Zhang P, Hong L, Li G, Wang R. Sodium Halides as Halogenating Reagents: Rhodium(III)-Catalyzed Versatile and Practical Halogenation of Aryl Compounds. Adv Synth Catal 2014. [DOI: 10.1002/adsc.201400590] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
45
|
Vicidomini C, Panico M, Greco A, Gargiulo S, Coda ARD, Zannetti A, Gramanzini M, Roviello GN, Quarantelli M, Alfano B, Tavitian B, Dollé F, Salvatore M, Brunetti A, Pappatà S. In vivo imaging and characterization of [(18)F]DPA-714, a potential new TSPO ligand, in mouse brain and peripheral tissues using small-animal PET. Nucl Med Biol 2014; 42:309-16. [PMID: 25537727 DOI: 10.1016/j.nucmedbio.2014.11.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 11/18/2014] [Accepted: 11/19/2014] [Indexed: 02/05/2023]
Abstract
INTRODUCTION The translocator protein 18 kDa (TSPO), a biochemical marker of neuroinflammation, is highly expressed in the brain activated microglia and it is also expressed by peripheral inflammatory cells and normal peripheral tissues. Thus, development of radioligands for the TSPO may contribute to further understanding the in vivo TSPO function in central and peripheral inflammatory processes and other pathologies. Here, we report the biodistribution, the specific binding and the radiometabolites of [(18)F]DPA-714, a promising fluorinated PET radiotracer, in normal mice using a microPET/CT scanner. METHODS The in vivo biodistribution and kinetics of [(18)F]DPA-714 were measured in mice brain and peripheral tissues. Specific binding to TSPO sites was assessed using pharmacological competitive studies by means of saturation experiments performed by i.v. injection of 1mg/kg of unlabeled DPA-714 or 3mg/kg of unlabeled PK11195. A region of interest analysis was performed to generate time-activity curves in the brain, heart, lung, kidney, spleen and liver. Metabolites assay was performed in the plasma and peripheral organs by radio-HPLC. RESULTS [(18)F]DPA-714 reached high concentration in lung, heart, kidney and spleen, tissues well known to be rich in TSPO sites. [(18)F]DPA-714 kinetics were faster in the lung and slower in the kidney. Pre-injection of unlabeled DPA-714 or PK11195 inhibited about 80% of [(18)F]DPA-714 uptake in the lung and heart (p<0.0005). The percentage of inhibition in the kidney was lower and achieved at later times only with DPA-714 (p<0.05) but not with PK11195. Sixty minutes after radiotracer injection only unmetabolized radioligand was found in the brain, lung, heart and spleen. CONCLUSION These results suggest that [(18)F]DPA-714 is a suitable PET ligand for imaging in mice brain and peripheral tissues since it binds with high specificity TSPO binding sites and it is almost unchanged at 60 minutes after radiotracer injection in the brain and TSPO-rich regions.
Collapse
Affiliation(s)
- Caterina Vicidomini
- Institute of Biostructure and Bioimaging, CNR, Naples, Italy; CEINGE, Biotecnologie Avanzate, s.c.a.r.l., Naples, Italy
| | - Mariarosaria Panico
- Institute of Biostructure and Bioimaging, CNR, Naples, Italy; CEINGE, Biotecnologie Avanzate, s.c.a.r.l., Naples, Italy
| | - Adelaide Greco
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy; CEINGE, Biotecnologie Avanzate, s.c.a.r.l., Naples, Italy
| | - Sara Gargiulo
- Institute of Biostructure and Bioimaging, CNR, Naples, Italy; CEINGE, Biotecnologie Avanzate, s.c.a.r.l., Naples, Italy
| | - Anna Rita Daniela Coda
- Institute of Biostructure and Bioimaging, CNR, Naples, Italy; CEINGE, Biotecnologie Avanzate, s.c.a.r.l., Naples, Italy
| | - Antonella Zannetti
- Institute of Biostructure and Bioimaging, CNR, Naples, Italy; CEINGE, Biotecnologie Avanzate, s.c.a.r.l., Naples, Italy
| | - Matteo Gramanzini
- Institute of Biostructure and Bioimaging, CNR, Naples, Italy; CEINGE, Biotecnologie Avanzate, s.c.a.r.l., Naples, Italy
| | | | | | - Bruno Alfano
- Institute of Biostructure and Bioimaging, CNR, Naples, Italy
| | - Bertrand Tavitian
- Inserm U970, PARCC, Université Paris Descartes, Hôpital Européen Georges Pompidou, Paris, France; CEA, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Frederic Dollé
- CEA, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Marco Salvatore
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Arturo Brunetti
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy; CEINGE, Biotecnologie Avanzate, s.c.a.r.l., Naples, Italy
| | - Sabina Pappatà
- Institute of Biostructure and Bioimaging, CNR, Naples, Italy.
| |
Collapse
|
46
|
Martinez FO, Gordon S. The evolution of our understanding of macrophages and translation of findings toward the clinic. Expert Rev Clin Immunol 2014; 11:5-13. [PMID: 25434688 DOI: 10.1586/1744666x.2015.985658] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
'There is at bottom only one genuinely scientific treatment for all diseases, and that is to stimulate the phagocytes,' so declaimed Sir Ralph Bloomfield Bonington in The Doctor's Dilemma, Act 1, by George Bernard Shaw (1906). More often nowadays, the need is to calm the phagocytes, given their role in inflammation and tissue damage. In spite of the growth of cellular and molecular information gained from studies in macrophage cell culture, mouse models and, to a lesser extent, human investigations, and the importance of macrophages in pathogenesis in a wide range of chronic disease processes, there is still a substantial shortfall in terms of clinical applications. In this review, we summarize concepts derived from macrophage studies and suggest possible properties suitable for diagnosis, prognosis and selective targeting of macrophage pathogenic functions.
Collapse
Affiliation(s)
- Fernando O Martinez
- Botnar Research Centre, University of Oxford, Windmill Road, Oxford, OX3 7LD, UK
| | | |
Collapse
|
47
|
Tiwari AK, Yui J, Fujinaga M, Kumata K, Shimoda Y, Yamasaki T, Xie L, Hatori A, Maeda J, Nengaki N, Zhang MR. Characterization of a novel acetamidobenzoxazolone-based PET ligand for translocator protein (18 kDa) imaging of neuroinflammation in the brain. J Neurochem 2014; 129:712-20. [PMID: 24484439 DOI: 10.1111/jnc.12670] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 01/11/2014] [Accepted: 01/29/2014] [Indexed: 01/20/2023]
Abstract
We developed the novel positron emission tomography (PET) ligand 2-[5-(4-[(11)C]methoxyphenyl)-2-oxo-1,3-benzoxazol-3(2H)-yl]-N-methyl-N-phenylacetamide ([(11)C]MBMP) for translocator protein (18 kDa, TSPO) imaging and evaluated its efficacy in ischemic rat brains. [(11)C]MBMP was synthesized by reacting desmethyl precursor (1) with [(11)C]CH3 I in radiochemical purity of ≥ 98% and specific activity of 85 ± 30 GBq/μmol (n = 18) at the end of synthesis. Biodistribution study on mice showed high accumulation of radioactivity in the TSPO-rich organs, e.g., the lungs, heart, kidneys, and adrenal glands. The metabolite analysis in mice brain homogenate showed 80.1 ± 2.7% intact [(11)C]MBMP at 60 min after injection. To determine the specific binding of [(11)C]MBMP with TSPO in the brain, in vitro autoradiography and PET studies were performed in an ischemic rat model. In vitro autoradiography indicated significantly increased binding on the ipsilateral side compared with that on the contralateral side of ischemic rat brains. This result was supported firmly by the contrast of radioactivity between the ipsilateral and contralateral sides in PET images. Displacement experiments with unlabelled MBMP or PK11195 minimized the difference in uptake between the two sides. In summary, [(11)C]MBMP is a potential PET imaging agent for TSPO and, consequently, for the up-regulation of microglia during neuroinflammation.
Collapse
Affiliation(s)
- Anjani K Tiwari
- Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan; Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Fairweather D, Coronado MJ, Garton AE, Dziedzic JL, Bucek A, Cooper LT, Brandt JE, Alikhan FS, Wang H, Endres CJ, Choi J, Pomper MG, Guilarte TR. Sex differences in translocator protein 18 kDa (TSPO) in the heart: implications for imaging myocardial inflammation. J Cardiovasc Transl Res 2014; 7:192-202. [PMID: 24402571 DOI: 10.1007/s12265-013-9538-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 12/18/2013] [Indexed: 12/17/2022]
Abstract
Myocarditis is more severe in men than in women and difficult to diagnose due to a lack of imaging modalities that directly detect myocardial inflammation. Translocator protein 18 kDa (TSPO) is used extensively to image brain inflammation due to its presence in CD11b(+) brain microglia. In this study, we examined expression of TSPO and CD11b in mice with coxsackievirus B3 (CVB3) myocarditis and biopsy sections from myocarditis patients in order to determine if it could be used to image myocarditis. We found that male mice with CVB3 myocarditis upregulated more genes associated with TSPO activation than female mice. TSPO expression was increased in the heart of male mice and men with myocarditis compared with female subjects due to testosterone, where it was expressed predominantly in CD11b(+) immune cells. We show that TSPO ligands detect myocardial inflammation using microSPECT, with increased uptake of [(125)I]-IodoDPA-713 in male mice with CVB3 myocarditis compared with undiseased controls.
Collapse
Affiliation(s)
- DeLisa Fairweather
- Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Goeders NE, Guerin GF, Schmoutz CD. The combination of metyrapone and oxazepam for the treatment of cocaine and other drug addictions. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 69:419-79. [PMID: 24484984 DOI: 10.1016/b978-0-12-420118-7.00011-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Although scientists have been investigating the neurobiology of psychomotor stimulant reward for many decades, there is still no FDA-approved treatment for cocaine or methamphetamine abuse. Research in our laboratory has focused on the relationship between stress, the subsequent activation of the hypothalamic-pituitary-adrenal (HPA) axis, and psychomotor stimulant reinforcement for almost 30 years. This research has led to the development of a combination of low doses of the cortisol synthesis inhibitor, metyrapone, and the benzodiazepine, oxazepam, as a potential pharmacological treatment for cocaine and other substance use disorders. In fact, we have conducted a pilot clinical trial that demonstrated that this combination can reduce cocaine craving and cocaine use. Our initial hypothesis underlying this effect was that the combination of metyrapone and oxazepam reduced cocaine seeking and taking by decreasing activity within the HPA axis. Even so, doses of the metyrapone and oxazepam combination that consistently reduced cocaine taking and seeking did not reliably alter plasma corticosterone (or cortisol in the pilot clinical trial). Furthermore, subsequent research has demonstrated that this drug combination is effective in adrenalectomized rats, suggesting that these effects must be mediated above the level of the adrenal gland. Our evolving hypothesis is that the combination of metyrapone and oxazepam produces its effects by increasing the levels of neuroactive steroids, most notably tetrahydrodeoxycorticosterone, in the medial prefrontal cortex and amygdala. Additional research will be necessary to confirm this hypothesis and may lead to the development of improved and specific pharmacotherapies for the treatment of psychomotor stimulant use.
Collapse
Affiliation(s)
- Nicholas E Goeders
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center, Shreveport, Louisiana, USA.
| | - Glenn F Guerin
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center, Shreveport, Louisiana, USA
| | - Christopher D Schmoutz
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center, Shreveport, Louisiana, USA
| |
Collapse
|
50
|
Affiliation(s)
- Lucia G Le Roux
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX; Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dawid Schellingerhout
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX; Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|