1
|
Yanai H, Adachi H, Hakoshima M, Katsuyama H. Pathology and Treatments of Alzheimer's Disease Based on Considering Changes in Brain Energy Metabolism Due to Type 2 Diabetes. Molecules 2024; 29:5936. [PMID: 39770025 PMCID: PMC11677283 DOI: 10.3390/molecules29245936] [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: 10/22/2024] [Revised: 11/22/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with cognitive dysfunction, memory decline, and behavioral disturbance, and it is pathologically characterized by the accumulation of amyloid plaques and neurofibrillary tangles in the brain. Although various hypotheses have been proposed to explain the pathogenesis of AD, including the amyloid beta hypothesis, oxidative stress hypothesis, and abnormal phosphorylation of tau proteins, the exact pathogenic mechanisms underlying AD remain largely undefined. Furthermore, effective curative treatments are very limited. Epidemiologic studies provide convincing evidence for a significant association between type 2 diabetes and AD. Here, we showed energy metabolism using glucose, lactate, ketone bodies, and lipids as energy substrates in a normal brain, and changes in such energy metabolism due to type 2 diabetes. We also showed the influences of such altered energy metabolism due to type 2 diabetes on the pathology of AD. Furthermore, we comprehensively searched for risk factors related with type 2 diabetes for AD and showed possible therapeutic interventions based on considering risk factors and altered brain energy metabolism due to type 2 diabetes for the development of AD.
Collapse
Affiliation(s)
- Hidekatsu Yanai
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Ichikawa 272-8516, Chiba, Japan; (H.A.); (M.H.); (H.K.)
| | | | | | | |
Collapse
|
2
|
Gujral J, Gandhi OH, Singh SB, Ahmed M, Ayubcha C, Werner TJ, Revheim ME, Alavi A. PET, SPECT, and MRI imaging for evaluation of Parkinson's disease. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2024; 14:371-390. [PMID: 39840378 PMCID: PMC11744359 DOI: 10.62347/aicm8774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025]
Abstract
This review assesses the primary neuroimaging techniques used to evaluate Parkinson's disease (PD) - a neurological condition characterized by gradual dopamine-producing nerve cell degeneration. The neuroimaging techniques explored include positron emission tomography (PET), single-photon emission computed tomography (SPECT), and magnetic resonance imaging (MRI). These modalities offer varying degrees of insights into PD pathophysiology, diagnostic accuracy, specificity by way of exclusion of other Parkinsonian syndromes, and monitoring of disease progression. Neuroimaging is thus crucial for diagnosing and managing PD, with integrated multimodal approaches and novel techniques further enhancing early detection and treatment evaluation.
Collapse
Affiliation(s)
- Jaskeerat Gujral
- Department of Radiology, University of PennsylvaniaPhiladelphia, PA 19104, USA
| | - Om H Gandhi
- Department of Radiology, University of PennsylvaniaPhiladelphia, PA 19104, USA
| | - Shashi B Singh
- Stanford University School of MedicineStanford, CA 94305, USA
| | - Malia Ahmed
- Department of Radiology, University of PennsylvaniaPhiladelphia, PA 19104, USA
| | - Cyrus Ayubcha
- Harvard Medical SchoolBoston, MA 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public HealthBoston, MA 02115, USA
| | - Thomas J Werner
- Department of Radiology, University of PennsylvaniaPhiladelphia, PA 19104, USA
| | - Mona-Elisabeth Revheim
- The Intervention Center, Rikshopitalet, Division of Technology and Innovation, Oslo University HospitalOslo 0372, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of OsloOslo 0315, Norway
| | - Abass Alavi
- Department of Radiology, University of PennsylvaniaPhiladelphia, PA 19104, USA
| |
Collapse
|
3
|
White AM, Craig AJ, Richie DL, Corley C, Sadek SM, Barton HN, Gipson CD. Nicotine is an Immunosuppressant: Implications for Women's Health and Disease. J Neuroimmunol 2024; 397:578468. [PMID: 39461120 DOI: 10.1016/j.jneuroim.2024.578468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/04/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
A plethora of evidence supports that nicotine, the primary alkaloid in tobacco products that is generally accepted for maintaining use, is immunoregulatory and may function as an immunosuppressant. Women have unique experiences with use of nicotine-containing products and also undergo significant reproductive transitions throughout their lifespan which may be impacted by nicotine use. Within the extant literature, there is conflicting evidence that nicotine may confer beneficial health effects in specific disease states (e.g., in ulcerative colitis). Use prevalence of nicotine-containing products is exceptionally high in individuals presenting with some comorbid disease states that impact immune system health and can be a risk factor for the development of diseases which disproportionately impact women; however, the mechanisms underlying these relationships are largely unclear. Further, little is known regarding the impacts of nicotine's immunosuppressive effects on women's health during the menopausal transition, which is arguably an inflammatory event characterized by a pro-inflammatory peri-menopause period. Given that post-menopausal women are at a higher risk than men for the development of neurodegenerative diseases such as Alzheimer's disease and are also more vulnerable to negative health effects associated with diseases such as HIV-1 infection, it is important to understand how use of nicotine-containing products may impact the immune milieu in women. In this review, we define instances in which nicotine use confers immunosuppressive, anti-inflammatory, or pro-inflammatory effects in the context of comorbid disease states, and focus on how nicotine impacts neuroimmune signaling to maintain use. We posit that regardless of potential health benefits, nicotine use cessation should be a priority in the clinical care of women. The synthesis of this review demonstrates the importance of systematically defining the relationships between volitional nicotine use, immune system function, and comorbid disease states in women to better understand how nicotine impacts women's health and disease.
Collapse
Affiliation(s)
- Ashley M White
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ashley J Craig
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Daryl L Richie
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Christa Corley
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Safiyah M Sadek
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Heather N Barton
- Beebe Health, Gastroenterology and Internal Medicine, Lewes, Delaware, USA
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
4
|
Emvalomenos GM, Kang JWM, Jupp B, Mychasiuk R, Keay KA, Henderson LA. Recent developments and challenges in positron emission tomography imaging of gliosis in chronic neuropathic pain. Pain 2024; 165:2184-2199. [PMID: 38713812 DOI: 10.1097/j.pain.0000000000003247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/05/2024] [Indexed: 05/09/2024]
Abstract
ABSTRACT Understanding the mechanisms that underpin the transition from acute to chronic pain is critical for the development of more effective and targeted treatments. There is growing interest in the contribution of glial cells to this process, with cross-sectional preclinical studies demonstrating specific changes in these cell types capturing targeted timepoints from the acute phase and the chronic phase. In vivo longitudinal assessment of the development and evolution of these changes in experimental animals and humans has presented a significant challenge. Recent technological advances in preclinical and clinical positron emission tomography, including the development of specific radiotracers for gliosis, offer great promise for the field. These advances now permit tracking of glial changes over time and provide the ability to relate these changes to pain-relevant symptomology, comorbid psychiatric conditions, and treatment outcomes at both a group and an individual level. In this article, we summarize evidence for gliosis in the transition from acute to chronic pain and provide an overview of the specific radiotracers available to measure this process, highlighting their potential, particularly when combined with ex vivo / in vitro techniques, to understand the pathophysiology of chronic neuropathic pain. These complementary investigations can be used to bridge the existing gap in the field concerning the contribution of gliosis to neuropathic pain and identify potential targets for interventions.
Collapse
Affiliation(s)
- Gaelle M Emvalomenos
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - James W M Kang
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Bianca Jupp
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Kevin A Keay
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Luke A Henderson
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| |
Collapse
|
5
|
Ohshima M, Moriguchi T, Enmi JI, Kawashima H, Koshino K, Zeniya T, Tsuji M, Iida H. [ 123I]CLINDE SPECT as a neuroinflammation imaging approach in a rat model of stroke. Exp Neurol 2024; 378:114843. [PMID: 38823675 DOI: 10.1016/j.expneurol.2024.114843] [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: 01/22/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/03/2024]
Abstract
Poststroke neuroinflammation exacerbates disease progression. [11C]PK11195-positron emission tomography (PET) imaging has been used to visualize neuroinflammation; however, its short half-life of 20 min limits its clinical use. [123I]CLINDE has a longer half-life (13h); therefore, [123I]CLINDE-single-photon emission computed tomography (SPECT) imaging is potentially more practical than [11C]PK11195-PET imaging in clinical settings. The objectives of this study were to 1) validate neuroinflammation imaging using [123I]CLINDE and 2) investigate the mechanisms underlying stroke in association with neuroinflammation using multimodal techniques, including magnetic resonance imaging (MRI), gas-PET, and histological analysis, in a rat model of ischemic stroke, that is, permanent middle cerebral artery occlusion (pMCAo). At 6 days post-pMCAo, [123I]CLINDE-SPECT considerably corresponded to the immunohistochemical images stained with the CD68 antibody (a marker for microglia/microphages), comparable to the level observed in [11C]PK11195-PET images. In addition, the [123I]CLINDE-SPECT images corresponded well with autoradiography images. Rats with severe infarcts, as defined by MRI, exhibited marked neuroinflammation in the peri-infarct area and less neuroinflammation in the ischemic core, accompanied by a substantial reduction in the cerebral metabolic rate of oxygen (CMRO2) in 15O-gas-PET. Rats with moderate-to-mild infarcts exhibited neuroinflammation in the ischemic core, where CMRO2 levels were mildly reduced. This study demonstrates that [123I]CLINDE-SPECT imaging is suitable for neuroinflammation imaging and that the distribution of neuroinflammation varies depending on the severity of infarction.
Collapse
Affiliation(s)
- Makiko Ohshima
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Department of Neurobiology, Care Sciences & Society, Karolinska Institute, Visionsgatan 4, Solna 171 64, Sweden
| | - Tetsuaki Moriguchi
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Institute of Physics, University of Tsukuba, Ibaraki 305-8571, Japan
| | - Jun-Ichiro Enmi
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Center for Information and Neural Networks (CiNet), Advanced ICT Research Institute, National Institute of Information and Communications Technology (NICT), 1-4 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hidekazu Kawashima
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Radioisotope Research Center, Kyoto Pharmaceutical University, 1 Misasagi-Shichono-cho, Yamashina-ku, Kyoto 607-8412, Japan
| | - Kazuhiro Koshino
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Department of Systems and Informatics, Hokkaido Information University, 59-2 Nishi-nopporo, Ebetsu, Hokkaido, Japan
| | - Tsutomu Zeniya
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Graduate School of Science and Technology, Hirosaki University, 3 Bunkyo-cho, Hirosaki, Aomori 036-8561, Japan
| | - Masahiro Tsuji
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Department of Food and Nutrition, Kyoto Women's University, 35 Kitahiyoshi-cho, Imakumano, Higashiyama-ku, Kyoto 605-8501, Japan.
| | - Hidehiro Iida
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Faculty of Medicine, University of Turku, and Turku PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, 20520 Turku, Finland
| |
Collapse
|
6
|
Cao Y, Xu W, Liu Q. Alterations of the blood-brain barrier during aging. J Cereb Blood Flow Metab 2024; 44:881-895. [PMID: 38513138 PMCID: PMC11318406 DOI: 10.1177/0271678x241240843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 02/19/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024]
Abstract
The blood-brain barrier (BBB) is a complex and dynamic interface that regulates the exchange of molecules and cells between the blood and the central nervous system. It undergoes structural and functional changes during aging, which may compromise its integrity and contribute to the pathogenesis of neurodegenerative diseases. In recent years, advances in microscopy and high-throughput bioinformatics have allowed a more in-depth investigation of the aging mechanisms of BBB. This review summarizes age-related alterations of the BBB structure and function from six perspectives: endothelial cells, astrocytes, pericytes, basement membrane, microglia and perivascular macrophages, and fibroblasts, ranging from the molecular level to the human multi-system level. These basic components are essential for the proper functioning of the BBB. Recent imaging methods of BBB were also reviewed. Elucidation of age-associated BBB changes may offer insights into BBB homeostasis and may provide effective therapeutic strategies to protect it during aging.
Collapse
Affiliation(s)
- Yufan Cao
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weihai Xu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Liu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Poxleitner M, Hoffmann SHL, Berezhnoy G, Ionescu TM, Gonzalez-Menendez I, Maier FC, Seyfried D, Ehrlichmann W, Quintanilla-Martinez L, Schmid AM, Reischl G, Trautwein C, Maurer A, Pichler BJ, Herfert K, Beziere N. Western diet increases brain metabolism and adaptive immune responses in a mouse model of amyloidosis. J Neuroinflammation 2024; 21:129. [PMID: 38745337 PMCID: PMC11092112 DOI: 10.1186/s12974-024-03080-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/29/2024] [Indexed: 05/16/2024] Open
Abstract
Diet-induced increase in body weight is a growing health concern worldwide. Often accompanied by a low-grade metabolic inflammation that changes systemic functions, diet-induced alterations may contribute to neurodegenerative disorder progression as well. This study aims to non-invasively investigate diet-induced metabolic and inflammatory effects in the brain of an APPPS1 mouse model of Alzheimer's disease. [18F]FDG, [18F]FTHA, and [18F]GE-180 were used for in vivo PET imaging in wild-type and APPPS1 mice. Ex vivo flow cytometry and histology in brains complemented the in vivo findings. 1H- magnetic resonance spectroscopy in the liver, plasma metabolomics and flow cytometry of the white adipose tissue were used to confirm metaflammatory condition in the periphery. We found disrupted glucose and fatty acid metabolism after Western diet consumption, with only small regional changes in glial-dependent neuroinflammation in the brains of APPPS1 mice. Further ex vivo investigations revealed cytotoxic T cell involvement in the brains of Western diet-fed mice and a disrupted plasma metabolome. 1H-magentic resonance spectroscopy and immunological results revealed diet-dependent inflammatory-like misbalance in livers and fatty tissue. Our multimodal imaging study highlights the role of the brain-liver-fat axis and the adaptive immune system in the disruption of brain homeostasis in amyloid models of Alzheimer's disease.
Collapse
Affiliation(s)
- Marilena Poxleitner
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Sabrina H L Hoffmann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Georgy Berezhnoy
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Tudor M Ionescu
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Irene Gonzalez-Menendez
- Department of Pathology and Neuropathology, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Florian C Maier
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Dominik Seyfried
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Walter Ehrlichmann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Leticia Quintanilla-Martinez
- Department of Pathology and Neuropathology, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Andreas M Schmid
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Gerald Reischl
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Kristina Herfert
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany.
| | - Nicolas Beziere
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany.
- Cluster of Excellence CMFI (EXC 2124) "Controlling Microbes to Fight Infections", Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
8
|
Ottoy J, De Picker L, Kang MS. Microglial Positron Emission Tomography Imaging In Vivo : Positron Emission Tomography Radioligands: Utility in Research and Clinical Practice. ADVANCES IN NEUROBIOLOGY 2024; 37:579-589. [PMID: 39207714 DOI: 10.1007/978-3-031-55529-9_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia, the resident immune cells of the central nervous system (CNS) play a key role in regulating and maintaining homeostasis in the brain. However, the CNS is also vulnerable to infections and inflammatory processes. In response to CNS perturbations, microglia become reactive, notably with expression of the translocator protein (TSPO), primarily on their outer mitochondrial membrane. Despite TSPO being commonly used as a marker for microglia, it is also present in other cell types such as astrocytes. Positron emission tomography (PET) ligands that target the TSPO enable the noninvasive detection and quantification of glial reactivity. While some limitations were raised, TSPO PET remains an attractive biomarker of CNS infection and inflammation. This book chapter delves into the development and application of microglial PET imaging with a focus on the TSPO PET. First, we provide an overview of the evolution of TSPO PET radioligands from first-generation to second-generation ligands and their applications in studying neuroinflammation (or CNS inflammation). Subsequently, we discuss the limitations and challenges associated with TSPO PET. Then we go on to explore non-TSPO targets for microglial PET imaging. Finally, we conclude with future directions for research and clinical practice in this field.
Collapse
Affiliation(s)
- Julie Ottoy
- Dr. Sandra E. Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Livia De Picker
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium
- University Psychiatric Hospital Campus Duffel, Duffel, Belgium
| | - Min Su Kang
- Dr. Sandra E. Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Bartos LM, Kirchleitner SV, Kolabas ZI, Quach S, Beck A, Lorenz J, Blobner J, Mueller SA, Ulukaya S, Hoeher L, Horvath I, Wind-Mark K, Holzgreve A, Ruf VC, Gold L, Kunze LH, Kunte ST, Beumers P, Park HE, Antons M, Zatcepin A, Briel N, Hoermann L, Schaefer R, Messerer D, Bartenstein P, Riemenschneider MJ, Lindner S, Ziegler S, Herms J, Lichtenthaler SF, Ertürk A, Tonn JC, von Baumgarten L, Albert NL, Brendel M. Deciphering sources of PET signals in the tumor microenvironment of glioblastoma at cellular resolution. SCIENCE ADVANCES 2023; 9:eadi8986. [PMID: 37889970 PMCID: PMC10610915 DOI: 10.1126/sciadv.adi8986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023]
Abstract
Various cellular sources hamper interpretation of positron emission tomography (PET) biomarkers in the tumor microenvironment (TME). We developed an approach of immunomagnetic cell sorting after in vivo radiotracer injection (scRadiotracing) with three-dimensional (3D) histology to dissect the cellular allocation of PET signals in the TME. In mice with implanted glioblastoma, translocator protein (TSPO) radiotracer uptake per tumor cell was higher compared to tumor-associated microglia/macrophages (TAMs), validated by protein levels. Translation of in vitro scRadiotracing to patients with glioma immediately after tumor resection confirmed higher single-cell TSPO tracer uptake of tumor cells compared to immune cells. Across species, cellular radiotracer uptake explained the heterogeneity of individual TSPO-PET signals. In consideration of cellular tracer uptake and cell type abundance, tumor cells were the main contributor to TSPO enrichment in glioblastoma; however, proteomics identified potential PET targets highly specific for TAMs. Combining cellular tracer uptake measures with 3D histology facilitates precise allocation of PET signals and serves to validate emerging novel TAM-specific radioligands.
Collapse
Affiliation(s)
- Laura M. Bartos
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Zeynep Ilgin Kolabas
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital of Munich, LMU Munich, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Stefanie Quach
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Alexander Beck
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Julia Lorenz
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | - Jens Blobner
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Stephan A. Mueller
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
| | - Selin Ulukaya
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- Faculty of Biology, Master of Science Program in Molecular and Cellular Biology, Ludwig-Maximilians-Universität München, Planegg, Germany
| | - Luciano Hoeher
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Izabela Horvath
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- School of Computation, Information and Technology (CIT), TUM, Boltzmannstr. 3, 85748 Garching, Germany
| | - Karin Wind-Mark
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Viktoria C. Ruf
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Lukas Gold
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Lea H. Kunze
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Sebastian T. Kunte
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Philipp Beumers
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Ha Eun Park
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Melissa Antons
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Artem Zatcepin
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
| | - Nils Briel
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
| | - Leonie Hoermann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Rebecca Schaefer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Denise Messerer
- Department of Cardiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Stefan F. Lichtenthaler
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital of Munich, LMU Munich, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joerg C. Tonn
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nathalie L. Albert
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
10
|
Larrea A, Elexpe A, Díez-Martín E, Torrecilla M, Astigarraga E, Barreda-Gómez G. Neuroinflammation in the Evolution of Motor Function in Stroke and Trauma Patients: Treatment and Potential Biomarkers. Curr Issues Mol Biol 2023; 45:8552-8585. [PMID: 37998716 PMCID: PMC10670324 DOI: 10.3390/cimb45110539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/25/2023] Open
Abstract
Neuroinflammation has a significant impact on different pathologies, such as stroke or spinal cord injury, intervening in their pathophysiology: expansion, progression, and resolution. Neuroinflammation involves oxidative stress, damage, and cell death, playing an important role in neuroplasticity and motor dysfunction by affecting the neuronal connection responsible for motor control. The diagnosis of this pathology is performed using neuroimaging techniques and molecular diagnostics based on identifying and measuring signaling molecules or specific markers. In parallel, new therapeutic targets are being investigated via the use of bionanomaterials and electrostimulation to modulate the neuroinflammatory response. These novel diagnostic and therapeutic strategies have the potential to facilitate the development of anticipatory patterns and deliver the most beneficial treatment to improve patients' quality of life and directly impact their motor skills. However, important challenges remain to be solved. Hence, the goal of this study was to review the implication of neuroinflammation in the evolution of motor function in stroke and trauma patients, with a particular focus on novel methods and potential biomarkers to aid clinicians in diagnosis, treatment, and therapy. A specific analysis of the strengths, weaknesses, threats, and opportunities was conducted, highlighting the key challenges to be faced in the coming years.
Collapse
Affiliation(s)
- Ane Larrea
- Research and Development Division, IMG Pharma Biotech, 48170 Zamudio, Spain; (A.L.); (A.E.); (E.D.-M.); (E.A.)
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
| | - Ane Elexpe
- Research and Development Division, IMG Pharma Biotech, 48170 Zamudio, Spain; (A.L.); (A.E.); (E.D.-M.); (E.A.)
| | - Eguzkiñe Díez-Martín
- Research and Development Division, IMG Pharma Biotech, 48170 Zamudio, Spain; (A.L.); (A.E.); (E.D.-M.); (E.A.)
- Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
| | - María Torrecilla
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
| | - Egoitz Astigarraga
- Research and Development Division, IMG Pharma Biotech, 48170 Zamudio, Spain; (A.L.); (A.E.); (E.D.-M.); (E.A.)
| | - Gabriel Barreda-Gómez
- Research and Development Division, IMG Pharma Biotech, 48170 Zamudio, Spain; (A.L.); (A.E.); (E.D.-M.); (E.A.)
| |
Collapse
|
11
|
Finze A, Biechele G, Rauchmann BS, Franzmeier N, Palleis C, Katzdobler S, Weidinger E, Guersel S, Schuster S, Harris S, Schmitt J, Beyer L, Gnörich J, Lindner S, Albert NL, Wetzel CH, Rupprecht R, Rominger A, Danek A, Burow L, Kurz C, Tato M, Utecht J, Papazov B, Zaganjori M, Trappmann LK, Goldhardt O, Grimmer T, Haeckert J, Janowitz D, Buerger K, Keeser D, Stoecklein S, Dietrich O, Morenas-Rodriguez E, Barthel H, Sabri O, Bartenstein P, Simons M, Haass C, Höglinger GU, Levin J, Perneczky R, Brendel M. Individual regional associations between Aβ-, tau- and neurodegeneration (ATN) with microglial activation in patients with primary and secondary tauopathies. Mol Psychiatry 2023; 28:4438-4450. [PMID: 37495886 PMCID: PMC10827660 DOI: 10.1038/s41380-023-02188-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/27/2023] [Accepted: 07/10/2023] [Indexed: 07/28/2023]
Abstract
β-amyloid (Aβ) and tau aggregation as well as neuronal injury and atrophy (ATN) are the major hallmarks of Alzheimer's disease (AD), and biomarkers for these hallmarks have been linked to neuroinflammation. However, the detailed regional associations of these biomarkers with microglial activation in individual patients remain to be elucidated. We investigated a cohort of 55 patients with AD and primary tauopathies and 10 healthy controls that underwent TSPO-, Aβ-, tau-, and perfusion-surrogate-PET, as well as structural MRI. Z-score deviations for 246 brain regions were calculated and biomarker contributions of Aβ (A), tau (T), perfusion (N1), and gray matter atrophy (N2) to microglial activation (TSPO, I) were calculated for each individual subject. Individual ATN-related microglial activation was correlated with clinical performance and CSF soluble TREM2 (sTREM2) concentrations. In typical and atypical AD, regional tau was stronger and more frequently associated with microglial activation when compared to regional Aβ (AD: βT = 0.412 ± 0.196 vs. βA = 0.142 ± 0.123, p < 0.001; AD-CBS: βT = 0.385 ± 0.176 vs. βA = 0.131 ± 0.186, p = 0.031). The strong association between regional tau and microglia reproduced well in primary tauopathies (βT = 0.418 ± 0.154). Stronger individual associations between tau and microglial activation were associated with poorer clinical performance. In patients with 4RT, sTREM2 levels showed a positive association with tau-related microglial activation. Tau pathology has strong regional associations with microglial activation in primary and secondary tauopathies. Tau and Aβ related microglial response indices may serve as a two-dimensional in vivo assessment of neuroinflammation in neurodegenerative diseases.
Collapse
Grants
- EXC 2145 SyNergy - ID 390857198 Deutsche Forschungsgemeinschaft (German Research Foundation)
- EXC 2155 - project number 39087428 Deutsche Forschungsgemeinschaft (German Research Foundation)
- HO2402/18-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
- FOR-2858 project numbers 403161218, 421887978 and 422188432 Deutsche Forschungsgemeinschaft (German Research Foundation)
- 19063p Alzheimer Forschung Initiative (Alzheimer Forschung Initiative e.V.)
- GUH was additionally funded by the German Federal Ministry of Education and Research (BMBF, 01KU1403A EpiPD; 01EK1605A HitTau; 01DH18025 TauTherapy); European Joint Programme on Rare Diseases (Improve-PSP); VolkswagenStiftung (Niedersächsisches Vorab); Petermax-Müller Foundation (Etiology and Therapy of Synucleinopathies and Tauopathies). The Lüneburg Heritage and Friedrich-Baur-Stiftung have supported the work of CP. The Hirnliga e.V. supported recruitment and imaging of the ActiGliA cohort (Manfred-Strohscheer-Stiftung) by a grant to BSR and MB.
- TG received consulting fees from AbbVie, Alector, Anavex, Biogen, Eli Lilly, Functional Neuromodulation, Grifols, Iqvia, Noselab, Novo Nordisk, NuiCare, Orphanzyme, Roche Diagnostics, Roche Pharma, UCB, and Vivoryon; lecture fees from Grifols, Medical Tribune, Novo Nordisk, Roche Pharma, and Schwabe; and has received grants to his institution from Roche Diagnostics.
- CH collaborates with Denali Therapeutics. CH is chief advisor of ISAR Bioscience and a member of the advisory board of AviadoBio.
- Günter Höglinger participated in industry-sponsored research projects from Abbvie, Biogen, Biohaven, Novartis, Roche, Sanofi, UCB; serves as a consultant for Abbvie, Alzprotect, Aprineua, Asceneuron, Bial, Biogen, Biohaven, Kyowa Kirin, Lundbeck, Novartis, Retrotope, Roche, Sanofi, UCB; received honoraria for scientific presentations from Abbvie, Bayer Vital, Bial, Biogen, Bristol Myers Squibb, Kyowa Kirin, Roche, Teva, UCB, Zambon; holds a patent on Treatment of Synucleinopathies. United States Patent No.: US 10,918,628 B2: EP 17 787 904.6-1109 / 3 525 788; received publication royalties from Academic Press, Kohlhammer, and Thieme.
Collapse
Affiliation(s)
- Anika Finze
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Gloria Biechele
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
- NeuroImaging Core Unit Munich (NICUM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Carla Palleis
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sabrina Katzdobler
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Endy Weidinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Selim Guersel
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Schuster
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Stefanie Harris
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Julia Schmitt
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University Regensburg, Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University Regensburg, Regensburg, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Nuclear Medicine, University Hospital, Inselspital Bern, Bern, Switzerland
| | - Adrian Danek
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lena Burow
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Carolin Kurz
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Maia Tato
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Julia Utecht
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Boris Papazov
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
- NeuroImaging Core Unit Munich (NICUM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Mirlind Zaganjori
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lena-Katharina Trappmann
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Oliver Goldhardt
- Department of Psychiatry and Psychotherapy, Klinikum Rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum Rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Jan Haeckert
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, Augsburg, Germany
| | | | | | - Daniel Keeser
- NeuroImaging Core Unit Munich (NICUM), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sophia Stoecklein
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Olaf Dietrich
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mikael Simons
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Christian Haass
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Johannes Levin
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
- Sheffield Institute for Translational Neurosciences (SITraN), University of Sheffield, Sheffield, UK
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| |
Collapse
|
12
|
Lin Q, Wang Y, Zhao W, Yao S. Visualization of Thromboinflammation by 18F-DPA-714 PET in a Stroke Patient. Clin Nucl Med 2023; 48:e477-e479. [PMID: 37682615 DOI: 10.1097/rlu.0000000000004803] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
ABSTRACT A 44-year-old man who presented with progressive right limb weakness was diagnosed with ischemic stroke. He was referred for 18F-DPA-714 PET/CT for evaluation of the disease. 18F-DPA-714 PET/CT showed increased uptake of the intracranial thrombus. This DPA-714-avid thrombus highly suggested the involvement of immune cells in the extension of the clot resulting in neurological deterioration. This present case suggested that 18F-DPA-714 PET might be a promising tracer in visualizing thromboinflammation in vivo.
Collapse
Affiliation(s)
- Qianqian Lin
- From the Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University
| | - Youliang Wang
- From the Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University
| | - Wenlong Zhao
- From the Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University
| | | |
Collapse
|
13
|
De Picker LJ, Morrens M, Branchi I, Haarman BCM, Terada T, Kang MS, Boche D, Tremblay ME, Leroy C, Bottlaender M, Ottoy J. TSPO PET brain inflammation imaging: A transdiagnostic systematic review and meta-analysis of 156 case-control studies. Brain Behav Immun 2023; 113:415-431. [PMID: 37543251 DOI: 10.1016/j.bbi.2023.07.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 06/26/2023] [Accepted: 07/30/2023] [Indexed: 08/07/2023] Open
Abstract
INTRODUCTION The 18-kDa translocator protein (TSPO) is increasingly recognized as a molecular target for PET imaging of inflammatory responses in various central nervous system (CNS) disorders. However, the reported sensitivity and specificity of TSPO PET to identify brain inflammatory processes appears to vary greatly across disorders, disease stages, and applied quantification methods. To advance TSPO PET as a potential biomarker to evaluate brain inflammation and anti-inflammatory therapies, a better understanding of its applicability across disorders is needed. We conducted a transdiagnostic systematic review and meta-analysis of all in vivo human TSPO PET imaging case-control studies in the CNS. Specifically, we investigated the direction, strength, and heterogeneity associated with the TSPO PET signal across disorders in pre-specified brain regions, and explored the demographic and methodological sources of heterogeneity. METHODS We searched for English peer-reviewed articles that reported in vivo human case-control TSPO PET differences. We extracted the demographic details, TSPO PET outcomes, and technical variables of the PET procedure. A random-effects meta-analysis was applied to estimate case-control standardized mean differences (SMD) of the TSPO PET signal in the lobar/whole-brain cortical grey matter (cGM), thalamus, and cortico-limbic circuitry between different illness categories. Heterogeneity was evaluated with the I2 statistic and explored using subgroup and meta-regression analyses for radioligand generation, PET quantification method, age, sex, and publication year. Significance was set at the False Discovery Rate (FDR)-corrected P < 0.05. RESULTS 156 individual case-control studies were included in the systematic review, incorporating data for 2381 healthy controls and 2626 patients. 139 studies documented meta-analysable data and were grouped into 11 illness categories. Across all the illness categories, we observed a significantly higher TSPO PET signal in cases compared to controls for the cGM (n = 121 studies, SMD = 0.358, PFDR < 0.001, I2 = 68%), with a significant difference between the illness categories (P = 0.004). cGM increases were only significant for Alzheimer's disease (SMD = 0.693, PFDR < 0.001, I2 = 64%) and other neurodegenerative disorders (SMD = 0.929, PFDR < 0.001, I2 = 73%). Cortico-limbic increases (n = 97 studies, SMD = 0.541, P < 0.001, I2 = 67%) were most prominent for Alzheimer's disease, mild cognitive impairment, other neurodegenerative disorders, mood disorders and multiple sclerosis. Thalamic involvement (n = 79 studies, SMD = 0.393, P < 0.001, I2 = 71%) was observed for Alzheimer's disease, other neurodegenerative disorders, multiple sclerosis, and chronic pain and functional disorders (all PFDR < 0.05). Main outcomes for systemic immunological disorders, viral infections, substance use disorders, schizophrenia and traumatic brain injury were not significant. We identified multiple sources of between-study variance to the TSPO PET signal including a strong transdiagnostic effect of the quantification method (explaining 25% of between-study variance; VT-based SMD = 0.000 versus reference tissue-based studies SMD = 0.630; F = 20.49, df = 1;103, P < 0.001), patient age (9% of variance), and radioligand generation (5% of variance). CONCLUSION This study is the first overarching transdiagnostic meta-analysis of case-control TSPO PET findings in humans across several brain regions. We observed robust increases in the TSPO signal for specific types of disorders, which were widespread or focal depending on illness category. We also found a large and transdiagnostic horizontal (positive) shift of the effect estimates of reference tissue-based compared to VT-based studies. Our results can support future studies to optimize experimental design and power calculations, by taking into account the type of disorder, brain region-of-interest, radioligand, and quantification method.
Collapse
Affiliation(s)
- Livia J De Picker
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Campus Duffel, Duffel, Belgium.
| | - Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Campus Duffel, Duffel, Belgium
| | - Igor Branchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Roma, Italy
| | - Bartholomeus C M Haarman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tatsuhiro Terada
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Min Su Kang
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences School, Faculty of Medicine, University of Southampton, UK
| | - Marie-Eve Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, BC, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Claire Leroy
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay (BioMaps), Orsay, France
| | - Michel Bottlaender
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay (BioMaps), Orsay, France; Université Paris-Saclay, UNIACT, Neurospin, CEA, Gif-sur-Yvette, France
| | - Julie Ottoy
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Chassé M, Pees A, Lindberg A, Liang SH, Vasdev N. Spirocyclic Iodonium Ylides for Fluorine-18 Radiolabeling of Non-Activated Arenes: From Concept to Clinical Research. CHEM REC 2023; 23:e202300072. [PMID: 37183954 DOI: 10.1002/tcr.202300072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/27/2023] [Indexed: 05/16/2023]
Abstract
Positron emission tomography (PET) is a powerful imaging tool for drug discovery, clinical diagnosis, and monitoring of disease progression. Fluorine-18 is the most common radionuclide used for PET, but advances in radiotracer development have been limited by the historical lack of methodologies and precursors amenable to radiolabeling with fluorine-18. Radiolabeling of electron-rich (hetero)aromatic rings remains a long-standing challenge in the production of PET radiopharmaceuticals. In this personal account, we discuss the history of spirocyclic iodonium ylide precursors, from inception to applications in clinical research, for the incorporation of fluorine-18 into complex non-activated (hetero)aromatic rings.
Collapse
Affiliation(s)
- Melissa Chassé
- Institute of Medical Science, University of Toronto, 1 Kings College Circle, Toronto, ON M5S 1A8, Canada
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, ON M5T 1R8, Canada
| | - Anna Pees
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, ON M5T 1R8, Canada
| | - Anton Lindberg
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, ON M5T 1R8, Canada
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Neil Vasdev
- Institute of Medical Science, University of Toronto, 1 Kings College Circle, Toronto, ON M5S 1A8, Canada
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, Toronto, ON M5T 1R8, Canada
| |
Collapse
|
15
|
Neumann KD, Broshek DK, Newman BT, Druzgal TJ, Kundu BK, Resch JE. Concussion: Beyond the Cascade. Cells 2023; 12:2128. [PMID: 37681861 PMCID: PMC10487087 DOI: 10.3390/cells12172128] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
Sport concussion affects millions of athletes each year at all levels of sport. Increasing evidence demonstrates clinical and physiological recovery are becoming more divergent definitions, as evidenced by several studies examining blood-based biomarkers of inflammation and imaging studies of the central nervous system (CNS). Recent studies have shown elevated microglial activation in the CNS in active and retired American football players, as well as in active collegiate athletes who were diagnosed with a concussion and returned to sport. These data are supportive of discordance in clinical symptomology and the inflammatory response in the CNS upon symptom resolution. In this review, we will summarize recent advances in the understanding of the inflammatory response associated with sport concussion and broader mild traumatic brain injury, as well as provide an outlook for important research questions to better align clinical and physiological recovery.
Collapse
Affiliation(s)
- Kiel D. Neumann
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Donna K. Broshek
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA 22903, USA;
| | - Benjamin T. Newman
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (B.T.N.); (T.J.D.); (B.K.K.)
| | - T. Jason Druzgal
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (B.T.N.); (T.J.D.); (B.K.K.)
| | - Bijoy K. Kundu
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (B.T.N.); (T.J.D.); (B.K.K.)
| | - Jacob E. Resch
- Department of Kinesiology, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
16
|
Vicente-Rodríguez M, Mancuso R, Peris-Yague A, Simmons C, Gómez-Nicola D, Perry VH, Turkheimer F, Lovestone S, Parker CA, Cash D. Pharmacological modulation of TSPO in microglia/macrophages and neurons in a chronic neurodegenerative model of prion disease. J Neuroinflammation 2023; 20:92. [PMID: 37032328 PMCID: PMC10084680 DOI: 10.1186/s12974-023-02769-y] [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: 05/10/2022] [Accepted: 03/20/2023] [Indexed: 04/11/2023] Open
Abstract
Neuroinflammation is an important component of many neurodegenerative diseases, whether as a primary cause or a secondary outcome. For that reason, either as diagnostic tools or to monitor progression and/or pharmacological interventions, there is a need for robust biomarkers of neuroinflammation in the brain. Mitochondrial TSPO (18 kDa Translocator protein) is one of few available biomarkers of neuroinflammation for which there are clinically available PET imaging agents. In this study, we further characterised neuroinflammation in a mouse model of prion-induced chronic neurodegeneration (ME7) including a pharmacological intervention via a CSF1R inhibitor. This was achieved by autoradiographic binding of the second-generation TSPO tracer, [3H]PBR28, along with a more comprehensive examination of the cellular contributors to the TSPO signal changes by immunohistochemistry. We observed regional increases of TSPO in the ME7 mouse brains, particularly in the hippocampus, cortex and thalamus. This increased TSPO signal was detected in the cells of microglia/macrophage lineage as well as in astrocytes, endothelial cells and neurons. Importantly, we show that the selective CSF1R inhibitor, JNJ-40346527 (JNJ527), attenuated the disease-dependent increase in TSPO signal, particularly in the dentate gyrus of the hippocampus, where JNJ527 attenuated the number of Iba1+ microglia and neurons, but not GFAP+ astrocytes or endothelial cells. These findings suggest that [3H]PBR28 quantitative autoradiography in combination with immunohistochemistry are important translational tools for detecting and quantifying neuroinflammation, and its treatments, in neurodegenerative disease. Furthermore, we demonstrate that although TSPO overexpression in the ME7 brains was driven by various cell types, the therapeutic effect of the CSF1R inhibitor was primarily to modulate TSPO expression in microglia and neurons, which identifies an important route of biological action of this particular CSF1R inhibitor and provides an example of a cell-specific effect of this type of therapeutic agent on the neuroinflammatory process.
Collapse
Affiliation(s)
- Marta Vicente-Rodríguez
- Department of Neuroimaging, BRAIN Centre (Biomarker Research and Imaging for Neuroscience), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK.
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain.
| | - Renzo Mancuso
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, UK
| | - Alba Peris-Yague
- Department of Neuroimaging, BRAIN Centre (Biomarker Research and Imaging for Neuroscience), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Camilla Simmons
- Department of Neuroimaging, BRAIN Centre (Biomarker Research and Imaging for Neuroscience), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
| | - Diego Gómez-Nicola
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
- Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, UK
| | - V Hugh Perry
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
- Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, UK
| | - Federico Turkheimer
- Department of Neuroimaging, BRAIN Centre (Biomarker Research and Imaging for Neuroscience), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
| | - Simon Lovestone
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
- Janssen Medical Ltd, High Wycombe, UK
| | - Christine A Parker
- Department of Neuroimaging, BRAIN Centre (Biomarker Research and Imaging for Neuroscience), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
- GlaxoSmithKline, Stevenage, London, UK
| | - Diana Cash
- Department of Neuroimaging, BRAIN Centre (Biomarker Research and Imaging for Neuroscience), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
| |
Collapse
|
17
|
Tan Z, Haider A, Zhang S, Chen J, Wei J, Liao K, Li G, Wei H, Dong C, Ran W, Li Y, Li Y, Rong J, Li Y, Liang SH, Xu H, Wang L. Quantitative assessment of translocator protein (TSPO) in the non-human primate brain and clinical translation of [ 18F]LW223 as a TSPO-targeted PET radioligand. Pharmacol Res 2023; 189:106681. [PMID: 36746361 DOI: 10.1016/j.phrs.2023.106681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/12/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Translocator protein 18 kDa (TSPO) positron emission tomography (PET) can be harnessed for the non-invasive detection of macrophage-driven inflammation. [18F]LW223, a newly reported TSPO PET tracer which was insensitive to rs6971 polymorphism, showed favorable performance characteristics in a recent imaging study involving a rat myocardial infarction model. To enable quantitative neuroimaging with [18F]LW223, we conducted kinetic analysis in the non-human primate (NHP) brain. Further, we sought to assess the utility of [18F]LW223-based TSPO imaging in a first-in-human study. METHODS Radiosynthesis of [18F]LW223 was accomplished on an automated module, whereas molar activities, stability in formulation, lipophilicity and unbound free fraction (fu) of the probe were measured. Brain penetration and target specificity of [18F]LW223 in NHPs were corroborated by PET-MR imaging under baseline and pre-blocking conditions using the validated TSPO inhibitor, (R)-PK11195, at doses ranging from 5 to 10 mg/kg. Kinetic modeling was performed using one-tissue compartment model (1TCM), two-tissue compartment model (2TCM) and Logan graphical analyses, using dynamic PET data acquisition, arterial blood collection and metabolic stability testing. Clinical PET scans were performed in two healthy volunteers (HVs). Regional brain standard uptake value ratio (SUVr) was assessed for different time intervals. RESULTS [18F]LW223 was synthesized in non-decay corrected radiochemical yields (n.d.c. RCYs) of 33.3 ± 6.5% with molar activities ranging from 1.8 ± 0.7 Ci/µmol (n = 11). [18F]LW223 was stable in formulation for up to 4 h and LogD7.4 of 2.31 ± 0.13 (n = 6) and fu of 5.80 ± 1.42% (n = 6) were determined. [18F]LW223 exhibited good brain penetration in NHPs, with a peak SUV value of ca. 1.79 in the whole brain. Pre-treatment with (R)-PK11195 substantially accelerated the washout and attenuated the area under the time-activity curve, indicating in vivo specificity of [18F]LW223 towards TSPO. Kinetic modeling demonstrated that 2TCM was the most suitable model for [18F]LW223-based neuroimaging. Global transfer rate constants (K1) and total volumes of distribution (VT) were found to be 0.10 ± 0.01 mL/cm3/min and 2.30 ± 0.17 mL/cm3, respectively. Dynamic PET data analyses across distinct time windows revealed that the VT values were relatively stable after 60 min post-injection. In a preliminary clinical study with two healthy volunteers, [18F]LW223 exhibited good brain uptake and considerable tracer retention across all analyzed brain regions. Of note, an excellent correlation between SUVr with VT was obtained when assessing the time interval from 20 to 40 min post tracer injection (SUVr(20-40 min), R2 = 0.94, p < 0.0001), suggesting this time window may be suitable to estimate specific binding to TSPO in human brain. CONCLUSION Our findings indicate that [18F]LW223 is suitable for quantitative TSPO-targeted PET imaging in higher species. Employing state-of-the-art kinetic modeling, we found that [18F]LW223 was effective in mapping TSPO throughout the NHP brain, with best model fits obtained from 2TCM and Logan graphical analyses. Overall, our results indicate that [18F]LW223 exhibits favorable tracer performance characteristics in higher species, and this novel imaging tool may hold promise to provide effective neuroinflammation imaging in patients with neurological disease.
Collapse
Affiliation(s)
- Zhiqiang Tan
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Achi Haider
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Shaojuan Zhang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jiahui Chen
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Junjie Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Kai Liao
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Guocong Li
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Huiyi Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Chenchen Dong
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Wenqing Ran
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Ying Li
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yuefeng Li
- Guangdong Landau Biotechnology Co. Ltd., Guangzhou 510555, China
| | - Jian Rong
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Yinlong Li
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA 30322, USA.
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| |
Collapse
|
18
|
Mohammadi Z, Alizadeh H, Marton J, Cumming P. The Sensitivity of Tau Tracers for the Discrimination of Alzheimer's Disease Patients and Healthy Controls by PET. Biomolecules 2023; 13:290. [PMID: 36830659 PMCID: PMC9953528 DOI: 10.3390/biom13020290] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/12/2023] [Accepted: 01/25/2023] [Indexed: 02/09/2023] Open
Abstract
Hyperphosphorylated tau aggregates, also known as neurofibrillary tangles, are a hallmark neuropathological feature of Alzheimer's disease (AD). Molecular imaging of tau by positron emission tomography (PET) began with the development of [18F]FDDNP, an amyloid β tracer with off-target binding to tau, which obtained regional specificity through the differing distributions of amyloid β and tau in AD brains. A concerted search for more selective and affine tau PET tracers yielded compounds belonging to at least eight structural categories; 18F-flortaucipir, known variously as [18F]-T807, AV-1451, and Tauvid®, emerged as the first tau tracer approved by the American Food and Drug Administration. The various tau tracers differ concerning their selectivity over amyloid β, off-target binding at sites such as monoamine oxidase and neuromelanin, and degree of uptake in white matter. While there have been many reviews of molecular imaging of tau in AD and other conditions, there has been no systematic comparison of the fitness of the various tracers for discriminating between AD patient and healthy control (HC) groups. In this narrative review, we endeavored to compare the binding properties of the various tau tracers in vitro and the effect size (Cohen's d) for the contrast by PET between AD patients and age-matched HC groups. The available tracers all gave good discrimination, with Cohen's d generally in the range of two-three in culprit brain regions. Overall, Cohen's d was higher for AD patient groups with more severe illness. Second-generation tracers, while superior concerning off-target binding, do not have conspicuously higher sensitivity for the discrimination of AD and HC groups. We suppose that available pharmacophores may have converged on a maximal affinity for tau fibrils, which may limit the specific signal imparted in PET studies.
Collapse
Affiliation(s)
- Zohreh Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - Hadi Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - János Marton
- ABX Advanced Biochemical Compounds Biomedizinische Forschungsreagenzien GmbH, Heinrich-Glaeser-Straße 10-14, D-01454 Radeberg, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Freiburgstraße 18, CH-3010 Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD 4059, Australia
| |
Collapse
|
19
|
Lopresti BJ, Royse SK, Mathis CA, Tollefson SA, Narendran R. Beyond monoamines: I. Novel targets and radiotracers for Positron emission tomography imaging in psychiatric disorders. J Neurochem 2023; 164:364-400. [PMID: 35536762 DOI: 10.1111/jnc.15615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
With the emergence of positron emission tomography (PET) in the late 1970s, psychiatry had access to a tool capable of non-invasive assessment of human brain function. Early applications in psychiatry focused on identifying characteristic brain blood flow and metabolic derangements using radiotracers such as [15 O]H2 O and [18 F]FDG. Despite the success of these techniques, it became apparent that more specific probes were needed to understand the neurochemical bases of psychiatric disorders. The first neurochemical PET imaging probes targeted sites of action of neuroleptic (dopamine D2 receptors) and psychoactive (serotonin receptors) drugs. Based on the centrality of monoamine dysfunction in psychiatric disorders and the measured success of monoamine-enhancing drugs in treating them, the next 30 years witnessed the development of an armamentarium of PET radiopharmaceuticals and imaging methodologies for studying monoamines. Continued development of monoamine-enhancing drugs over this time however was less successful, realizing only modest gains in efficacy and tolerability. As patent protection for many widely prescribed and profitable psychiatric drugs lapsed, drug development pipelines shifted away from monoamines in search of novel targets with the promises of improved efficacy, or abandoned altogether. Over this period, PET radiopharmaceutical development activities closely paralleled drug development priorities resulting in the development of new PET imaging agents for non-monoamine targets. Part one of this review will briefly survey novel PET imaging targets with relevance to the field of psychiatry, which include the metabotropic glutamate receptor type 5 (mGluR5), purinergic P2 X7 receptor, type 1 cannabinoid receptor (CB1 ), phosphodiesterase 10A (PDE10A), and describe radiotracers developed for these and other targets that have matured to human subject investigations. Current limitations of the targets and techniques will also be discussed.
Collapse
Affiliation(s)
- Brian J Lopresti
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sarah K Royse
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Chester A Mathis
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Savannah A Tollefson
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rajesh Narendran
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Departments of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
20
|
Bach P, de Timary P, Gründer G, Cumming P. Molecular Imaging Studies of Alcohol Use Disorder. Curr Top Behav Neurosci 2023. [PMID: 36639552 DOI: 10.1007/7854_2022_414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Alcohol use disorder (AUD) is a serious public health problem in many countries, bringing a gamut of health risks and impairments to individuals and a great burden to society. Despite the prevalence of a disease model of AUD, the current pharmacopeia does not present reliable treatments for AUD; approved treatments are confined to a narrow spectrum of medications engaging inhibitory γ-aminobutyric acid (GABA) neurotransmission and possibly excitatory N-methyl-D-aspartate (NMDA) receptors, and opioid receptor antagonists. Molecular imaging with positron emission tomography (PET) and single-photon emission computed tomography (SPECT) can open a window into the living brain and has provided diverse insights into the pathology of AUD. In this narrative review, we summarize the state of molecular imaging findings on the pharmacological action of ethanol and the neuropathological changes associated with AUD. Laboratory and preclinical imaging results highlight the interactions between ethanol and GABA A-type receptors (GABAAR), but the interpretation of such results is complicated by subtype specificity. An abundance of studies with the glucose metabolism tracer fluorodeoxyglucose (FDG) concur in showing cerebral hypometabolism after ethanol challenge, but there is relatively little data on long-term changes in AUD. Alcohol toxicity evokes neuroinflammation, which can be tracked using PET with ligands for the microglial marker translocator protein (TSPO). Several PET studies show reversible increases in TSPO binding in AUD individuals, and preclinical results suggest that opioid-antagonists can rescue from these inflammatory responses. There are numerous PET/SPECT studies showing changes in dopaminergic markers, generally consistent with an impairment in dopamine synthesis and release among AUD patients, as seen in a number of other addictions; this may reflect the composite of an underlying deficiency in reward mechanisms that predisposes to AUD, in conjunction with acquired alterations in dopamine signaling. There is little evidence for altered serotonin markers in AUD, but studies with opioid receptor ligands suggest a specific up-regulation of the μ-opioid receptor subtype. Considerable heterogeneity in drinking patterns, gender differences, and the variable contributions of genetics and pre-existing vulnerability traits present great challenges for charting the landscape of molecular imaging in AUD.
Collapse
Affiliation(s)
- Patrick Bach
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.
| | - Philippe de Timary
- Department of Adult Psychiatry, Cliniques Universitaires Saint-Luc and Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Gerhard Gründer
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, Australia
- International Centre for Education and Research in Neuropsychiatry (ICERN), Samara State Medical University, Samara, Russia
| |
Collapse
|
21
|
Bartolo ND, Mortimer N, Manter MA, Sanchez N, Riley M, O'Malley TT, Hooker JM. Identification and Prioritization of PET Neuroimaging Targets for Microglial Phenotypes Associated with Microglial Activity in Alzheimer's Disease. ACS Chem Neurosci 2022; 13:3641-3660. [PMID: 36473177 DOI: 10.1021/acschemneuro.2c00607] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Activation of microglial cells accompanies the progression of many neurodegenerative disorders, including Alzheimer's disease (AD). Development of molecular imaging tools specific to microglia can help elucidate the mechanism through which microglia contribute to the pathogenesis and progression of neurodegenerative disorders. Through analysis of published genetic, transcriptomic, and proteomic data sets, we identified 19 genes with microglia-specific expression that we then ranked based on association with the AD characteristics, change in expression, and potential druggability of the target. We believe that the process we used to identify and rank microglia-specific genes is broadly applicable to the identification and evaluation of targets in other disease areas and for applications beyond molecular imaging.
Collapse
Affiliation(s)
- Nicole D Bartolo
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| | - Niall Mortimer
- Human Biology and Data Science, Eisai Center for Genetics Guided Dementia Discovery, 35 Cambridgepark Drive, Cambridge, Massachusetts 02140, United States
| | - Mariah A Manter
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| | - Nicholas Sanchez
- Human Biology and Data Science, Eisai Center for Genetics Guided Dementia Discovery, 35 Cambridgepark Drive, Cambridge, Massachusetts 02140, United States
| | - Misha Riley
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| | - Tiernan T O'Malley
- Human Biology and Data Science, Eisai Center for Genetics Guided Dementia Discovery, 35 Cambridgepark Drive, Cambridge, Massachusetts 02140, United States
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
22
|
Murtaj V, Penati S, Belloli S, Foti M, Coliva A, Papagna A, Gotti C, Toninelli E, Chiaffarelli R, Mantero S, Pucci S, Matteoli M, Malosio ML, Moresco RM. Brain sex-dependent alterations after prolonged high fat diet exposure in mice. Commun Biol 2022; 5:1276. [PMID: 36414721 PMCID: PMC9681749 DOI: 10.1038/s42003-022-04214-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 11/02/2022] [Indexed: 11/23/2022] Open
Abstract
We examined effects of exposing female and male mice for 33 weeks to 45% or 60% high fat diet (HFD). Males fed with either diet were more vulnerable than females, displaying higher and faster increase in body weight and more elevated cholesterol and liver enzymes levels. Higher glucose metabolism was revealed by PET in the olfactory bulbs of both sexes. However, males also displayed altered anterior cortex and cerebellum metabolism, accompanied by a more prominent brain inflammation relative to females. Although both sexes displayed reduced transcripts of neuronal and synaptic genes in anterior cortex, only males had decreased protein levels of AMPA and NMDA receptors. Oppositely, to anterior cortex, cerebellum of HFD-exposed mice displayed hypometabolism and transcriptional up-regulation of neuronal and synaptic genes. These results indicate that male brain is more susceptible to metabolic changes induced by HFD and that the anterior cortex versus cerebellum display inverse susceptibility to HFD.
Collapse
Affiliation(s)
- Valentina Murtaj
- grid.7563.70000 0001 2174 1754PhD Program in Neuroscience, University of Milano-Bicocca, Monza (MB), Italy ,grid.18887.3e0000000417581884Department of Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy ,grid.18887.3e0000000417581884Present Address: Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Hospital and Vita Salute San Raffaele University, Milan, Italy, 20132 Milan, Italy
| | - Silvia Penati
- Institute of Neuroscience, National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Via Manzoni 56, 20089 Rozzano (MI), Italy ,grid.417728.f0000 0004 1756 8807Laboratory of Pharmacology and Brain Pathology, Neuro Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano (MI), Italy ,grid.4367.60000 0001 2355 7002Present Address: Department of Pathology and Immunology, Washington Univerisity School of Medicine, St. Louis, MO 63110 USA
| | - Sara Belloli
- grid.18887.3e0000000417581884Department of Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy ,grid.428490.30000 0004 1789 9809Institute of Molecular Bioimaging and Physiology, CNR, 20090 Segrate (MI), Italy
| | - Maria Foti
- grid.7563.70000 0001 2174 1754Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy
| | - Angela Coliva
- grid.18887.3e0000000417581884Department of Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Angela Papagna
- grid.7563.70000 0001 2174 1754Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy
| | - Cecilia Gotti
- grid.5326.20000 0001 1940 4177Institute of Neuroscience, National Research Council of Italy (CNR) c/o Università di Milano-Bicocca, Via R. Follereau 3, 20854 Vedano al Lambro (MB), Italy
| | - Elisa Toninelli
- grid.18887.3e0000000417581884Department of Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Remy Chiaffarelli
- grid.18887.3e0000000417581884Department of Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy ,grid.7563.70000 0001 2174 1754Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy ,grid.10392.390000 0001 2190 1447Present Address: Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
| | - Stefano Mantero
- grid.5326.20000 0001 1940 4177Institute for Genetic and Biomedical Research, National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Via Manzoni 56, 20089 Rozzano (MI), Italy ,grid.5326.20000 0001 1940 4177Present Address: DCSR, National Research Council of Italy (CNR), Via A. Corti 12, 20133 Milan, Italy
| | - Susanna Pucci
- grid.5326.20000 0001 1940 4177Institute of Neuroscience, National Research Council of Italy (CNR) c/o Università di Milano-Bicocca, Via R. Follereau 3, 20854 Vedano al Lambro (MB), Italy
| | - Michela Matteoli
- Institute of Neuroscience, National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Via Manzoni 56, 20089 Rozzano (MI), Italy ,grid.417728.f0000 0004 1756 8807Laboratory of Pharmacology and Brain Pathology, Neuro Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano (MI), Italy
| | - Maria Luisa Malosio
- Institute of Neuroscience, National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Via Manzoni 56, 20089 Rozzano (MI), Italy ,grid.417728.f0000 0004 1756 8807Laboratory of Pharmacology and Brain Pathology, Neuro Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano (MI), Italy
| | - Rosa Maria Moresco
- grid.18887.3e0000000417581884Department of Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy ,grid.428490.30000 0004 1789 9809Institute of Molecular Bioimaging and Physiology, CNR, 20090 Segrate (MI), Italy ,grid.7563.70000 0001 2174 1754Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy
| |
Collapse
|
23
|
Martinez-Orengo N, Tahmazian S, Lai J, Wang Z, Sinharay S, Schreiber-Stainthorp W, Basuli F, Maric D, Reid W, Shah S, Hammoud DA. Assessing organ-level immunoreactivity in a rat model of sepsis using TSPO PET imaging. Front Immunol 2022; 13:1010263. [PMID: 36439175 PMCID: PMC9685400 DOI: 10.3389/fimmu.2022.1010263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
There is current need for new approaches to assess/measure organ-level immunoreactivity and ensuing dysfunction in systemic inflammatory response syndrome (SIRS) and sepsis, in order to protect or recover organ function. Using a rat model of systemic sterile inflammatory shock (intravenous LPS administration), we performed PET imaging with a translocator protein (TSPO) tracer, [18F]DPA-714, as a biomarker for reactive immunoreactive changes in the brain and peripheral organs. In vivo dynamic PET/CT scans showed increased [18F]DPA-714 binding in the brain, lungs, liver and bone marrow, 4 hours after LPS injection. Post-LPS mean standard uptake values (SUVmean) at equilibrium were significantly higher in those organs compared to baseline. Changes in spleen [18F]DPA-714 binding were variable but generally decreased after LPS. SUVmean values in all organs, except the spleen, positively correlated with several serum cytokines/chemokines. In vitro measures of TSPO expression and immunofluorescent staining validated the imaging results. Noninvasive molecular imaging with [18F]DPA-714 PET in a rat model of systemic sterile inflammatory shock, along with in vitro measures of TSPO expression, showed brain, liver and lung inflammation, spleen monocytic efflux/lymphocytic activation and suggested increased bone marrow hematopoiesis. TSPO PET imaging can potentially be used to quantify SIRS and sepsis-associated organ-level immunoreactivity and assess the effectiveness of therapeutic and preventative approaches for associated organ failures, in vivo.
Collapse
Affiliation(s)
- Neysha Martinez-Orengo
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Sarine Tahmazian
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Jianhao Lai
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Zeping Wang
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Sanhita Sinharay
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - William Schreiber-Stainthorp
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, United States
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - William Reid
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Swati Shah
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Dima A. Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Dima A. Hammoud,
| |
Collapse
|
24
|
Eggerstorfer B, Kim JH, Cumming P, Lanzenberger R, Gryglewski G. Meta-analysis of molecular imaging of translocator protein in major depression. Front Mol Neurosci 2022; 15:981442. [PMID: 36226319 PMCID: PMC9549359 DOI: 10.3389/fnmol.2022.981442] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Molecular neuroimaging studies provide mounting evidence that neuroinflammation plays a contributory role in the pathogenesis of major depressive disorder (MDD). This has been the focus of a number of positron emission tomography (PET) studies of the 17-kDa translocator protein (TSPO), which is expressed by microglia and serves as a marker of neuroinflammation. In this meta-analysis, we compiled and analyzed all available molecular imaging studies comparing cerebral TSPO binding in MDD patients with healthy controls. Our systematic literature search yielded eight PET studies encompassing 238 MDD patients and 164 healthy subjects. The meta-analysis revealed relatively increased TSPO binding in several cortical regions (anterior cingulate cortex: Hedges' g = 0.6, 95% CI: 0.36, 0.84; hippocampus: g = 0.54, 95% CI: 0.26, 0.81; insula: g = 0.43, 95% CI: 0.17, 0.69; prefrontal cortex: g = 0.36, 95% CI: 0.14, 0.59; temporal cortex: g = 0.39, 95% CI: -0.04, 0.81). While the high range of effect size in the temporal cortex might reflect group-differences in body mass index (BMI), exploratory analyses failed to reveal any relationship between elevated TSPO availability in the other four brain regions and depression severity, age, BMI, radioligand, or the binding endpoint used, or with treatment status at the time of scanning. Taken together, this meta-analysis indicates a widespread ∼18% increase of TSPO availability in the brain of MDD patients, with effect sizes comparable to those in earlier molecular imaging studies of serotonin transporter availability and monoamine oxidase A binding.
Collapse
Affiliation(s)
- Benjamin Eggerstorfer
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Jong-Hoon Kim
- Department of Psychiatry, Gachon University College of Medicine, Gil Medical Center, Neuroscience Research Institute, GAIHST, Gachon University, Incheon, South Korea
| | - Paul Cumming
- Department of Nuclear Medicine, Inselspital, Bern University, Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, Australia
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Gregor Gryglewski
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| |
Collapse
|
25
|
Jamar F, Gormsen LC, Yildiz H, Slart RH, van der Geest KS, Gheysens O. The role of PET/CT in large vessel vasculitis and related disorders: diagnosis, extent evaluation and assessment of therapy response. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2022; 66:182-193. [PMID: 36066110 DOI: 10.23736/s1824-4785.22.03465-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Large vessel vasculitides (LVV) are defined as chronic inflammatory disorders that affect the arteries with two major variants being distinguished: giant cell arteritis (GCA) and Takayasu's arteritis (TAK). These often present with nonspecific constitutional symptoms which makes an accurate diagnosis often challenging. Nevertheless, timely diagnosis is of utmost importance to initiate treatment and to avoid potential life-threatening complications. [18F]FDG-PET/CT is nowadays widely accepted as useful tool to aid in the diagnosis of large vessel vasculitis. However, its role to monitor disease activity and to predict disease relapse during follow-up is less obvious since vascular [18F]FDG uptake can be detected in the absence of clinical or biochemical signs of disease activity. In addition to the two major variants, [18F]FDG-PET/CT has shown promise in (peri-)aortitis and related disorders. This article aims to provide an update on the current knowledge and limitations of [18F]FDG-PET/CT for the diagnosis and assessment of treatment response in LVV. Furthermore, other radiopharmaceuticals targeting key components of the vascular immune system are being discussed which could provide an interesting alternative to image vascular inflammation in LVV.
Collapse
Affiliation(s)
- François Jamar
- Department of Nuclear Medicine, Saint-Luc University Clinics and Institute of Clinical and Experimental Research (IREC), Catholic University of Louvain (UCLouvain), Brussels, Belgium -
| | - Lars C Gormsen
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Halil Yildiz
- Department of Internal Medicine and Infectious Diseases, Saint-Luc University Clinics, Brussels, Belgium
| | - Riemer H Slart
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center of Groningen, University of Groningen, Groningen, the Netherlands
- Department of Biomedical Photonic Imaging, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| | - Kornelis S van der Geest
- Department of Rheumatology and Clinical Immunology, University Medical Center of Groningen, University of Groningen, Groningen, the Netherlands
| | - Olivier Gheysens
- Department of Nuclear Medicine, Saint-Luc University Clinics and Institute of Clinical and Experimental Research (IREC), Catholic University of Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
26
|
18F-GE180, a failed tracer for translocator protein, has no place in child abuse imaging. Pediatr Radiol 2022; 52:1015-1016. [PMID: 34837109 PMCID: PMC9035024 DOI: 10.1007/s00247-021-05248-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/16/2021] [Accepted: 11/15/2021] [Indexed: 10/19/2022]
|
27
|
Candelario-Jalil E, Dijkhuizen RM, Magnus T. Neuroinflammation, Stroke, Blood-Brain Barrier Dysfunction, and Imaging Modalities. Stroke 2022; 53:1473-1486. [PMID: 35387495 PMCID: PMC9038693 DOI: 10.1161/strokeaha.122.036946] [Citation(s) in RCA: 310] [Impact Index Per Article: 103.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Maintaining blood-brain barrier (BBB) integrity is crucial for the homeostasis of the central nervous system. Structurally comprising the BBB, brain endothelial cells interact with pericytes, astrocytes, neurons, microglia, and perivascular macrophages in the neurovascular unit. Brain ischemia unleashes a profound neuroinflammatory response to remove the damaged tissue and prepare the brain for repair. However, the intense neuroinflammation occurring during the acute phase of stroke is associated with BBB breakdown, neuronal injury, and worse neurological outcomes. Here, we critically discuss the role of neuroinflammation in ischemic stroke pathology, focusing on the BBB and the interactions between central nervous system and peripheral immune responses. We highlight inflammation-driven injury mechanisms in stroke, including oxidative stress, increased MMP (matrix metalloproteinase) production, microglial activation, and infiltration of peripheral immune cells into the ischemic tissue. We provide an updated overview of imaging techniques for in vivo detection of BBB permeability, leukocyte infiltration, microglial activation, and upregulation of cell adhesion molecules following ischemic brain injury. We discuss the possibility of clinical implementation of imaging modalities to assess stroke-associated neuroinflammation with the potential to provide image-guided diagnosis and treatment. We summarize the results from several clinical studies evaluating the efficacy of anti-inflammatory interventions in stroke. Although convincing preclinical evidence suggests that neuroinflammation is a promising target for ischemic stroke, thus far, translating these results into the clinical setting has proved difficult. Due to the dual role of inflammation in the progression of ischemic damage, more research is needed to mechanistically understand when the neuroinflammatory response begins the transition from injury to repair. This could have important implications for ischemic stroke treatment by informing time- and context-specific therapeutic interventions.
Collapse
Affiliation(s)
- Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville (E.C-J)
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, the Netherlands (R.M.D.)
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Germany (T.M.)
| |
Collapse
|
28
|
Moran C, Than S, Callisaya M, Beare R, Srikanth V. New Horizons-Cognitive Dysfunction Associated With Type 2 Diabetes. J Clin Endocrinol Metab 2022; 107:929-942. [PMID: 34788847 DOI: 10.1210/clinem/dgab797] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Indexed: 11/19/2022]
Abstract
The prevalence of type 2 diabetes (T2D) and cognitive dysfunction increases with age. As society ages, clinicians will be increasingly tasked with managing older people who have both T2D and cognitive dysfunction. T2D is associated with an increased risk of cognitive dysfunction and hence there is increasing interest in whether T2D is a causal factor in the pathogenesis of cognitive decline and dementia. Recent advances in the use of sensitive measures of in vivo brain dysfunction in life-course studies can help understand potential mechanistic pathways and also help guide recommendations for clinical practice. In this article we will describe new horizons in the understanding of cognitive dysfunction associated with T2D. Coming from a clinical perspective, we discuss potential mechanisms and pathways linking the 2 conditions and the contribution of multimodal neuroimaging and study designs to advancing understanding in the field. We also highlight the important issues on the horizon that will need addressing in clinical identification, management, and risk reduction for people with coexistent T2D and cognitive dysfunction.
Collapse
Affiliation(s)
- Chris Moran
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Department of Geriatric Medicine, Peninsula Health, Melbourne, 3199 Victoria, Australia
- Department of Geriatric Medicine, Alfred Health, Melbourne, 3004 Victoria, Australia
| | - Stephanie Than
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Department of Geriatric Medicine, Peninsula Health, Melbourne, 3199 Victoria, Australia
| | - Michele Callisaya
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, 7000 Tasmania, Australia
| | - Richard Beare
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, 3052 Victoria, Australia
| | - Velandai Srikanth
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Department of Geriatric Medicine, Peninsula Health, Melbourne, 3199 Victoria, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, 7000 Tasmania, Australia
| |
Collapse
|
29
|
Weiss F, Labrador-Garrido A, Dzamko N, Halliday G. Immune responses in the Parkrtdinson's disease brain. Neurobiol Dis 2022; 168:105700. [DOI: 10.1016/j.nbd.2022.105700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/15/2022] Open
|
30
|
Varlow C, Knight AC, McQuade P, Vasdev N. Characterization of neuroinflammatory positron emission tomography biomarkers in chronic traumatic encephalopathy. Brain Commun 2022; 4:fcac019. [PMID: 35198978 PMCID: PMC8856182 DOI: 10.1093/braincomms/fcac019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/10/2021] [Accepted: 02/01/2022] [Indexed: 11/12/2022] Open
Abstract
Chronic traumatic encephalopathy is a neurological disorder associated with head trauma and is confirmed upon autopsy. PET imaging of chronic traumatic encephalopathy may provide a means to move towards ante-mortem diagnosis and therapeutic intervention following brain injuries. Characterization of the neuroinflammatory PET biomarkers, 18 kDa translocator protein and monoamine oxidase-B was conducted using [3H]PBR-28 and [3H]L-deprenyl, respectively, in post-mortem chronic traumatic encephalopathy brain tissue. [3H]PBR-28 displayed high specific binding in both chronic traumatic encephalopathy (95.40 ± 1.87%; n = 11 cases) and healthy controls (89.89 ± 8.52%, n = 3 cases). Cell-type expression of the 18 kDa translocator protein was confirmed by immunofluorescence to microglia, astrocyte and macrophage markers. [3H]L-deprenyl also displayed high specific binding in chronic traumatic encephalopathy (96.95 ± 1.43%; n = 12 cases) and healthy controls (93.24 ± 0.43%; n = 2 cases), with the distribution co-localized to astrocytes by immunofluorescence. Saturation analysis was performed to quantify the target density of the 18 kDa translocator protein and monoamine oxidase-B in both chronic traumatic encephalopathy and healthy control tissue. Using [3H]PBR-28, the target density of the 18 kDa translocator protein in healthy controls was 177.91 ± 56.96 nM (n = 7 cases; mean ± standard deviation); however, a highly variable target density (345.84 ± 372.42 nM; n = 11 cases; mean ± standard deviation) was measured in chronic traumatic encephalopathy. [3H]L-deprenyl quantified a monoamine oxidase-B target density of 304.23 ± 115.93 nM (n = 8 cases; mean ± standard deviation) in healthy control tissue and is similar to the target density in chronic traumatic encephalopathy tissues (365.80 ± 128.55 nM; n = 12 cases; mean ± standard deviation). A two-sample t-test determined no significant difference in the target density values of the 18 kDa translocator protein and monoamine oxidase-B between healthy controls and chronic traumatic encephalopathy (P > 0.05), albeit a trend towards increased expression of both targets was observed in chronic traumatic encephalopathy. To our knowledge, this work represents the first in vitro characterization of 18 kDa translocator protein and monoamine oxidase-B in chronic traumatic encephalopathy and reveals the variability in neuroinflammatory pathology following brain injuries. These preliminary findings will be considered when designing PET imaging studies after brain injury and for the ultimate goal of imaging chronic traumatic encephalopathy in vivo.
Collapse
Affiliation(s)
- Cassis Varlow
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada M5T 1R8
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Ashley C. Knight
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada M5T 1R8
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Paul McQuade
- Takeda Pharmaceutical Company, Cambridge, MA 02139, USA
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada M5T 1R8
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada M5S 1A8
| |
Collapse
|
31
|
Gouilly D, Saint-Aubert L, Ribeiro MJ, Salabert AS, Tauber C, Péran P, Arlicot N, Pariente J, Payoux P. Neuroinflammation PET imaging of the translocator protein (TSPO) in Alzheimer's disease: an update. Eur J Neurosci 2022; 55:1322-1343. [PMID: 35083791 DOI: 10.1111/ejn.15613] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/28/2022]
Abstract
Neuroinflammation is a significant contributor to Alzheimer's disease (AD). Until now, PET imaging of the translocator protein (TSPO) has been widely used to depict the neuroimmune endophenotype of AD. The aim of this review was to provide an update to the results from 2018 and to advance the characterization of the biological basis of TSPO imaging in AD by re-examining TSPO function and expression and the methodological aspects of interest. Although the biological basis of the TSPO PET signal is obviously related to microglia and astrocytes in AD, the observed process remains uncertain and might not be directly related to neuroinflammation. Further studies are required to re-examine the cellular significance underlying a variation in the PET signal in AD and how it can be impacted by a disease-modifying treatment.
Collapse
Affiliation(s)
- Dominique Gouilly
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Laure Saint-Aubert
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Maria-Joao Ribeiro
- Department of Nuclear Medicine, CHU, Tours, France.,UMR 1253, iBrain, Université de Tours, France.,Inserm CIC 1415, CHRU, Tours, France
| | - Anne-Sophie Salabert
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.,Department of Nuclear Medicine, CHU, Toulouse, France
| | | | - Patrice Péran
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, France.,Inserm CIC 1415, CHRU, Tours, France
| | - Jérémie Pariente
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.,Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU, Toulouse, France.,Center of Clinical Investigations (CIC1436), CHU, Toulouse, France
| | - Pierre Payoux
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.,Department of Nuclear Medicine, CHU, Toulouse, France
| |
Collapse
|
32
|
Moraga-Amaro R, Guerrin CGJ, Reali Nazario L, Lima Giacobbo B, J O Dierckx RA, Stehberg J, de Vries EFJ, Doorduin J. A single dose of ketamine cannot prevent protracted stress-induced anhedonia and neuroinflammation in rats. Stress 2022; 25:145-155. [PMID: 35384793 DOI: 10.1080/10253890.2022.2045269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Worldwide, millions of people suffer from treatment-resistant depression. Ketamine, a glutamatergic receptor antagonist, can have a rapid antidepressant effect even in treatment-resistant patients. A proposed mechanism for the antidepressant effect of ketamine is the reduction of neuroinflammation. To further explore this hypothesis, we investigated whether a single dose of ketamine can modulate protracted neuroinflammation in a repeated social defeat (RSD) stress rat model, which resembles features of depression. To this end, male animals exposed to RSD were injected with ketamine (20 mg/kg) or vehicle. A combination of behavioral analyses and PET scans of the inflammatory marker TSPO in the brain were performed. Rats submitted to RSD showed anhedonia-like behavior in the sucrose preference test, decreased weight gain, and increased TSPO levels in the insular and entorhinal cortices, as observed by [11C]-PK11195 PET. Whole brain TSPO levels correlated with corticosterone levels in several brain regions of RSD exposed animals, but not in controls. Ketamine injection 1 day after RSD disrupted the correlation between TSPO levels and serum corticosterone levels, but had no effect on depressive-like symptoms, weight gain or the protracted RSD-induced increase in TSPO expression in male rats. These results suggest that ketamine does not exert its effect on the hypothalamic-pituitary-adrenal axis by modulation of neuroinflammation.
Collapse
Affiliation(s)
- Rodrigo Moraga-Amaro
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| | - Cyprien G J Guerrin
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| | - Luiza Reali Nazario
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| | - Bruno Lima Giacobbo
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| | - Jimmy Stehberg
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Erik F J de Vries
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| |
Collapse
|
33
|
Joo YH, Lee MW, Son YD, Chang KA, Yaqub M, Kim HK, Cumming P, Kim JH. In Vivo Cerebral Translocator Protein (TSPO) Binding and Its Relationship with Blood Adiponectin Levels in Treatment-Naïve Young Adults with Major Depression: A [ 11C]PK11195 PET Study. Biomedicines 2021; 10:biomedicines10010034. [PMID: 35052718 PMCID: PMC8773340 DOI: 10.3390/biomedicines10010034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/05/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022] Open
Abstract
Adiponectin is an adipokine that mediates cellular cholesterol efflux and plays important roles in neuroinflammatory processes. In this study, we undertook positron emission tomography (PET) with the translocator protein (TSPO) ligand [11C]PK11195 and measured serum adiponectin levels in groups of treatment-naïve young adult patients with major depressive disorder (MDD) and matched healthy controls. Thirty treatment-naïve MDD patients (median age: 24 years) and twenty-three healthy controls underwent [11C]PK11195 PET. We quantified TSPO availability in brain as the [11C]PK11195 binding potential (BPND) using a reference tissue model in conjunction with the supervised cluster analysis (SVCA4) algorithm. Age, sex distribution, body mass index, and serum adiponectin levels did not differ between the groups. Between-group analysis using a region-of-interest approach showed significantly higher [11C]PK11195 BPND in the left anterior and right posterior cingulate cortices in MDD patients than in controls. Serum adiponectin levels had significant negative correlations with [11C]PK11195 BPND in the bilateral hippocampus in MDD patients, but significant positive correlations in the bilateral hippocampus in the control group. Our results indicate significantly higher TSPO binding in the anterior and posterior cingulate cortices in treatment-naïve young MDD patients, suggesting microglial activation in these limbic regions, which are involved in cognitive and emotional processing. The opposite correlations between [11C]PK11195 BPND in the hippocampus with serum adiponectin levels in MDD and control groups suggest that microglial activation in the hippocampus may respond differentially to adiponectin signaling in MDD and healthy subjects, possibly with respect to microglial phenotype.
Collapse
Affiliation(s)
- Yo-Han Joo
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (M.-W.L.); (Y.-D.S.); (K.-A.C.); (H.-K.K.)
| | - Min-Woo Lee
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (M.-W.L.); (Y.-D.S.); (K.-A.C.); (H.-K.K.)
| | - Young-Don Son
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (M.-W.L.); (Y.-D.S.); (K.-A.C.); (H.-K.K.)
- Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon 21936, Korea
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
| | - Keun-A Chang
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (M.-W.L.); (Y.-D.S.); (K.-A.C.); (H.-K.K.)
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
- Department of Pharmacology, Gachon University College of Medicine, Gachon University, Incheon 21936, Korea
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands;
| | - Hang-Keun Kim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (M.-W.L.); (Y.-D.S.); (K.-A.C.); (H.-K.K.)
- Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon 21936, Korea
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
| | - Paul Cumming
- Department of Nuclear Medicine, Inselspital, Bern University, CH-3010 Bern, Switzerland;
- School of Psychology and Counselling, Queensland University of Technology, Brisbane 4059, Australia
| | - Jong-Hoon Kim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (M.-W.L.); (Y.-D.S.); (K.-A.C.); (H.-K.K.)
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
- Department of Psychiatry, Gachon University College of Medicine, Gil Medical Center, Gachon University, Incheon 21565, Korea
- Correspondence: ; Tel.: +82-32-460-2696
| |
Collapse
|
34
|
Ramakrishnan NK, Hird M, Thompson S, Williamson DJ, Qiao L, Owen DR, Brooks AF, Scott PJH, Bacallado S, O'Brien JT, Aigbirhio FI. Preclinical evaluation of (S)-[ 18F]GE387, a novel 18-kDa translocator protein (TSPO) PET radioligand with low binding sensitivity to human polymorphism rs6971. Eur J Nucl Med Mol Imaging 2021; 49:125-136. [PMID: 34405276 PMCID: PMC8712295 DOI: 10.1007/s00259-021-05495-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 07/08/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE Positron emission tomography (PET) studies with radioligands for 18-kDa translocator protein (TSPO) have been instrumental in increasing our understanding of the complex role neuroinflammation plays in disorders affecting the brain. However, (R)-[11C]PK11195, the first and most widely used TSPO radioligand has limitations, while the next-generation TSPO radioligands have suffered from high interindividual variability in binding due to a genetic polymorphism in the TSPO gene (rs6971). Herein, we present the biological evaluation of the two enantiomers of [18F]GE387, which we have previously shown to have low sensitivity to this polymorphism. METHODS Dynamic PET scans were conducted in male Wistar rats and female rhesus macaques to investigate the in vivo behaviour of (S)-[18F]GE387 and (R)-[18F]GE387. The specific binding of (S)-[18F]GE387 to TSPO was investigated by pre-treatment with (R)-PK11195. (S)-[18F]GE387 was further evaluated in a rat model of lipopolysaccharide (LPS)-induced neuroinflammation. Sensitivity to polymorphism of (S)-GE387 was evaluated in genotyped human brain tissue. RESULTS (S)-[18F]GE387 and (R)-[18F]GE387 entered the brain in both rats and rhesus macaques. (R)-PK11195 blocked the uptake of (S)-[18F]GE387 in healthy olfactory bulb and peripheral tissues constitutively expressing TSPO. A 2.7-fold higher uptake of (S)-[18F]GE387 was found in the inflamed striatum of LPS-treated rodents. In genotyped human brain tissue, (S)-GE387 was shown to bind similarly in low affinity binders (LABs) and high affinity binders (HABs) with a LAB to HAB ratio of 1.8. CONCLUSION We established that (S)-[18F]GE387 has favourable kinetics in healthy rats and non-human primates and that it can distinguish inflamed from normal brain regions in the LPS model of neuroinflammation. Crucially, we have reconfirmed its low sensitivity to the TSPO polymorphism on genotyped human brain tissue. Based on these factors, we conclude that (S)-[18F]GE387 warrants further evaluation with studies on human subjects to assess its suitability as a TSPO PET radioligand for assessing neuroinflammation.
Collapse
Affiliation(s)
- Nisha K Ramakrishnan
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK.
| | - Matthew Hird
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Stephen Thompson
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - David J Williamson
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Luxi Qiao
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - David R Owen
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Allen F Brooks
- Division of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, 1301 Catherine Street, Ann Arbor, MI, 48109, USA
| | - Peter J H Scott
- Division of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, 1301 Catherine Street, Ann Arbor, MI, 48109, USA
| | - Sergio Bacallado
- Statistical Laboratory, Centre for the Mathematical Sciences, University of Cambridge, Wilberforce Rd., Cambridge, CB3 0WB, UK
| | - John T O'Brien
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Franklin I Aigbirhio
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Biomedical Campus, Cambridge, CB2 0SZ, UK
| |
Collapse
|
35
|
Dirks M, Buchert R, Wirries AK, Pflugrad H, Grosse GM, Petrusch C, Schütze C, Wilke F, Mamach M, Hamann L, Langer LBN, Ding XQ, Barg-Hock H, Klempnauer J, Wetzel CH, Lukacevic M, Janssen E, Kessler M, Bengel FM, Geworski L, Rupprecht R, Ross TL, Berding G, Weissenborn K. Reduced microglia activity in patients with long-term immunosuppressive therapy after liver transplantation. Eur J Nucl Med Mol Imaging 2021; 49:234-245. [PMID: 33978829 PMCID: PMC8712291 DOI: 10.1007/s00259-021-05398-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/02/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE Calcineurin inhibitors (CNI) can cause long-term impairment of brain function. Possible pathomechanisms include alterations of the cerebral immune system. This study used positron emission tomography (PET) imaging with the translocator protein (TSPO) ligand 18F-GE-180 to evaluate microglial activation in liver-transplanted patients under different regimens of immunosuppression. METHODS PET was performed in 22 liver-transplanted patients (3 CNI free, 9 with low-dose CNI, 10 with standard-dose CNI immunosuppression) and 9 healthy controls. The total distribution volume (VT) estimated in 12 volumes-of-interest was analyzed regarding TSPO genotype, CNI therapy, and cognitive performance. RESULTS In controls, VT was about 80% higher in high affinity binders (n = 5) compared to mixed affinity binders (n = 3). Mean VT corrected for TSPO genotype was significantly lower in patients compared to controls, especially in patients in whom CNI dose had been reduced because of nephrotoxic side effect. CONCLUSION Our results provide evidence of chronic suppression of microglial activity in liver-transplanted patients under CNI therapy especially in patients with high sensitivity to CNI toxicity.
Collapse
Affiliation(s)
- Meike Dirks
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- Integrated Research and Treatment Centre Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany.
| | - Ralph Buchert
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ann-Katrin Wirries
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Henning Pflugrad
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Integrated Research and Treatment Centre Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
| | - Gerrit M Grosse
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Carlotta Petrusch
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Christian Schütze
- Department of Medical Physics and Radiation Protection, Hannover Medical School, Hannover, Germany
| | - Florian Wilke
- Department of Medical Physics and Radiation Protection, Hannover Medical School, Hannover, Germany
| | - Martin Mamach
- Department of Medical Physics and Radiation Protection, Hannover Medical School, Hannover, Germany
| | - Linda Hamann
- Department of Medical Physics and Radiation Protection, Hannover Medical School, Hannover, Germany
| | - Laura B N Langer
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Xiao-Qi Ding
- Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Hannelore Barg-Hock
- General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Jürgen Klempnauer
- General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Mario Lukacevic
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Eike Janssen
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Mariella Kessler
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Lilli Geworski
- Department of Medical Physics and Radiation Protection, Hannover Medical School, Hannover, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Georg Berding
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Karin Weissenborn
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Integrated Research and Treatment Centre Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
| |
Collapse
|
36
|
Chauveau F, Becker G, Boutin H. Have (R)-[ 11C]PK11195 challengers fulfilled the promise? A scoping review of clinical TSPO PET studies. Eur J Nucl Med Mol Imaging 2021; 49:201-220. [PMID: 34387719 PMCID: PMC8712292 DOI: 10.1007/s00259-021-05425-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE The prototypical TSPO radiotracer (R)-[11C]PK11195 has been used in humans for more than thirty years to visualize neuroinflammation in several pathologies. Alternative radiotracers have been developed to improve signal-to-noise ratio and started to be tested clinically in 2008. Here we examined the scientific value of these "(R)-[11C]PK11195 challengers" in clinical research to determine if they could supersede (R)-[11C]PK11195. METHODS A systematic MEDLINE (PubMed) search was performed (up to end of year 2020) to extract publications reporting TSPO PET in patients with identified pathologies, excluding studies in healthy subjects and methodological studies. RESULTS Of the 288 publications selected, 152 used 13 challengers, and 142 used (R)-[11C]PK11195. Over the last 20 years, the number of (R)-[11C]PK11195 studies remained stable (6 ± 3 per year), but was surpassed by the total number of challenger studies for the last 6 years. In total, 3914 patients underwent a TSPO PET scan, and 47% (1851 patients) received (R)-[11C]PK11195. The 2 main challengers were [11C]PBR28 (24%-938 patients) and [18F]FEPPA (11%-429 patients). Only one-in-ten patients (11%-447) underwent 2 TSPO scans, among whom 40 (1%) were scanned with 2 different TSPO radiotracers. CONCLUSIONS Generally, challengers confirmed disease-specific initial (R)-[11C]PK11195 findings. However, while their better signal-to-noise ratio seems particularly useful in diseases with moderate and widespread neuroinflammation, most challengers present an allelic-dependent (Ala147Thr polymorphism) TSPO binding and genetic stratification is hindering their clinical implementation. As new challengers, insensitive to TSPO human polymorphism, are about to enter clinical evaluation, we propose this systematic review to be regularly updated (living review).
Collapse
Affiliation(s)
- Fabien Chauveau
- University of Lyon, Lyon Neuroscience Research Center (CRNL), CNRS UMR5292, INSERM U1028, University Lyon 1, Lyon, France.
| | - Guillaume Becker
- GIGA - CRC In Vivo Imaging, University Liege, Liege, Belgium
- University of Lyon, CarMeN Laboratory, INSERM U1060, University Lyon 1, Hospices Civils Lyon, Lyon, France
| | - Hervé Boutin
- Faculty of Biology Medicine and Health, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
| |
Collapse
|
37
|
Wimberley C, Lavisse S, Hillmer A, Hinz R, Turkheimer F, Zanotti-Fregonara P. Kinetic modeling and parameter estimation of TSPO PET imaging in the human brain. Eur J Nucl Med Mol Imaging 2021; 49:246-256. [PMID: 33693967 PMCID: PMC8712306 DOI: 10.1007/s00259-021-05248-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/07/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE Translocator protein 18-kDa (TSPO) imaging with positron emission tomography (PET) is widely used in research studies of brain diseases that have a neuro-immune component. Quantification of TSPO PET images, however, is associated with several challenges, such as the lack of a reference region, a genetic polymorphism affecting the affinity of the ligand for TSPO, and a strong TSPO signal in the endothelium of the brain vessels. These challenges have created an ongoing debate in the field about which type of quantification is most useful and whether there is an appropriate simplified model. METHODS This review focuses on the quantification of TSPO radioligands in the human brain. The various methods of quantification are summarized, including the gold standard of compartmental modeling with metabolite-corrected input function as well as various alternative models and non-invasive approaches. Their advantages and drawbacks are critically assessed. RESULTS AND CONCLUSIONS Researchers employing quantification methods for TSPO should understand the advantages and limitations associated with each method. Suggestions are given to help researchers choose between these viable alternative methods.
Collapse
Affiliation(s)
| | - Sonia Lavisse
- CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Université Paris-Saclay, 92265, Fontenay-aux-Roses, France
| | - Ansel Hillmer
- Departments of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Departments of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, USA
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, M20 3LJ, UK
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Centre for Neuroimaging Sciences, King's College London, De Crespigny Park, London, SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE1 1UL, UK
| | - Paolo Zanotti-Fregonara
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
38
|
Betlazar C, Middleton RJ, Howell N, Storer B, Davis E, Davies J, Banati R, Liu GJ. Mitochondrial Translocator Protein (TSPO) Expression in the Brain After Whole Body Gamma Irradiation. Front Cell Dev Biol 2021; 9:715444. [PMID: 34760884 PMCID: PMC8573390 DOI: 10.3389/fcell.2021.715444] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/29/2021] [Indexed: 01/04/2023] Open
Abstract
The brain's early response to low dose ionizing radiation, as may be encountered during diagnostic procedures and space exploration, is not yet fully characterized. In the brain parenchyma, the mitochondrial translocator protein (TSPO) is constitutively expressed at low levels by endothelial cells, and can therefore be used to assess the integrity of the brain's vasculature. At the same time, the inducible expression of TSPO in activated microglia, the brain's intrinsic immune cells, is a regularly observed early indicator of subtle or incipient brain pathology. Here, we explored the use of TSPO as a biomarker of brain tissue injury following whole body irradiation. Post-radiation responses were measured in C57BL/6 wild type (Tspo +/+) and TSPO knockout (Tspo -/-) mice 48 h after single whole body gamma irradiations with low doses 0, 0.01, and 0.1 Gy and a high dose of 2 Gy. Additionally, post-radiation responses of primary microglial cell cultures were measured at 1, 4, 24, and 48 h at an irradiation dose range of 0 Gy-2 Gy. TSPO mRNA and protein expression in the brain showed a decreased trend after 0.01 Gy relative to sham-irradiated controls, but remained unchanged after higher doses. Immunohistochemistry confirmed subtle decreases in TSPO expression after 0.01 Gy in vascular endothelial cells of the hippocampal region and in ependymal cells, with no detectable changes following higher doses. Cytokine concentrations in plasma after whole body irradiation showed differential changes in IL-6 and IL-10 with some variations between Tspo-/- and Tspo +/+ animals. The in vitro measurements of TSPO in primary microglial cell cultures showed a significant reduction 1 h after low dose irradiation (0.01 Gy). In summary, acute low and high doses of gamma irradiation up to 2 Gy reduced TSPO expression in the brain's vascular compartment without de novo induction of TSPO expression in parenchymal microglia, while TSPO expression in directly irradiated, isolated, and thus highly activated microglia, too, was reduced after low dose irradiation. The potential link between TSPO, its role in mitochondrial energy metabolism and the selective radiation sensitivity, notably of cells with constitutive TSPO expression such as vascular endothelial cells, merits further exploration.
Collapse
Affiliation(s)
- Calina Betlazar
- Australian Nuclear Science and Technology Organisation, Sydney, NSW, Australia
- Discipline of Medical Imaging and Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Ryan J. Middleton
- Australian Nuclear Science and Technology Organisation, Sydney, NSW, Australia
| | - Nicholas Howell
- Australian Nuclear Science and Technology Organisation, Sydney, NSW, Australia
| | - Ben Storer
- Australian Nuclear Science and Technology Organisation, Sydney, NSW, Australia
| | - Emma Davis
- Australian Nuclear Science and Technology Organisation, Sydney, NSW, Australia
| | - Justin Davies
- Australian Nuclear Science and Technology Organisation, Sydney, NSW, Australia
| | - Richard Banati
- Australian Nuclear Science and Technology Organisation, Sydney, NSW, Australia
- Discipline of Medical Imaging and Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation, Sydney, NSW, Australia
- Discipline of Medical Imaging and Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
39
|
Khan W, Corben LA, Bilal H, Vivash L, Delatycki MB, Egan GF, Harding IH. Neuroinflammation in the Cerebellum and Brainstem in Friedreich Ataxia: An [ 18 F]-FEMPA PET Study. Mov Disord 2021; 37:218-224. [PMID: 34643298 DOI: 10.1002/mds.28825] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/08/2021] [Accepted: 09/21/2021] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Neuroinflammation is proposed to accompany, or even contribute to, neuropathology in Friedreich ataxia (FRDA), with implications for disease treatment and tracking. OBJECTIVES To examine brain glial activation and systemic immune dysfunction in people with FRDA and quantify their relationship with symptom severity, duration, and onset age. METHODS Fifteen individuals with FRDA and 13 healthy controls underwent brain positron emission tomography using the translocator protein (TSPO) radioligand [18 F]-FEMPA, a marker of glial activation, together with the quantification of blood plasma inflammatory cytokines. RESULTS [18 F]-FEMPA binding was significantly increased in the dentate nuclei (d = 0.67), superior cerebellar peduncles (d = 0.74), and midbrain (d = 0.87), alongside increased plasma interleukin-6 (IL-6) (d = 0.73), in individuals with FRDA compared to controls. Increased [18 F]-FEMPA binding in the dentate nuclei, brainstem, and cerebellar anterior lobe correlated with earlier age of symptom onset (controlling for the genetic triplet repeat expansion length; all rpart < -0.6), and in the pons and anterior lobe with shorter disease duration (r = -0.66; -0.73). CONCLUSIONS Neuroinflammation is evident in brain regions implicated in FRDA neuropathology. Increased neuroimmune activity may be related to earlier disease onset and attenuate over the course of the illness. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Wasim Khan
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Louise A Corben
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.,Turner Institute for Brain and Mental Health and School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Hiba Bilal
- Turner Institute for Brain and Mental Health and School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Lucy Vivash
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Martin B Delatycki
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.,Victorian Clinical Genetics Service, Melbourne, Victoria, Australia
| | - Gary F Egan
- Turner Institute for Brain and Mental Health and School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia.,Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| | - Ian H Harding
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
40
|
Lim TX, Ahamed M, Reutens DC. The aryl hydrocarbon receptor: A diagnostic and therapeutic target in glioma. Drug Discov Today 2021; 27:422-435. [PMID: 34624509 DOI: 10.1016/j.drudis.2021.09.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 07/29/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022]
Abstract
Glioblastoma multiforme (GBM) is a deadly disease; 5-year survival rates have shown little improvement over the past 30 years. In vivo positron emission tomography (PET) imaging is an important method of identifying potential diagnostic and therapeutic molecular targets non-invasively. The aryl hydrocarbon receptor (AhR) is a transcription factor that regulates multiple genes involved in immune response modulation and tumorigenesis. The AhR is an attractive potential drug target and studies have shown that its activation by small molecules can modulate innate and adaptive immunity beneficially and prevent AhR-mediated tumour promotion in several cancer types. In this review, we provide an overview of the role of the AhR in glioma tumorigenesis and highlight its potential as an emerging biomarker for glioma therapies targeting the tumour immune response and PET diagnostics.
Collapse
Affiliation(s)
- Ting Xiang Lim
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Muneer Ahamed
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - David C Reutens
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
41
|
Direct Comparison of [ 18F]F-DPA with [ 18F]DPA-714 and [ 11C]PBR28 for Neuroinflammation Imaging in the same Alzheimer's Disease Model Mice and Healthy Controls. Mol Imaging Biol 2021; 24:157-166. [PMID: 34542805 PMCID: PMC8760190 DOI: 10.1007/s11307-021-01646-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/22/2021] [Accepted: 08/23/2021] [Indexed: 11/02/2022]
Abstract
PURPOSE In this study we compared the recently developed TSPO tracer [18F]F-DPA, with [18F]DPA-714 and [11C]PBR28 by performing in vivo PET imaging on the same Alzheimer's disease mouse model APP/PS1-21 (TG) and wild-type (WT) mice with all three radiotracers. PROCEDURES To compare the radiotracer uptake, percentage of injected dose/mL (%ID/mL), standardized uptake value ratios to cerebellum (SUVRCB), and voxel-wise analyses were performed. RESULTS The peak uptake of [18F]F-DPA was higher than 4.3% ID/mL, while [18F]DPA-714 reached just over 3% ID/mL, and [11C]PBR28 was over 4% ID/mL in only one brain region in the WT mice. The peak/60-min uptake ratios of [18F]F-DPA were significantly higher (p < 0.001) than those of [18F]DPA-714 and [11C]PBR28. The differences in [18F]F-DPA SUVRCB between WT and TG mice were highly significant (p < 0.001) in the three studied time periods after injection. [18F]DPA-714 uptake was significantly higher in TG mice starting in the 20-40-min timeframe and increased thereafter, whereas [11C]PBR28 uptake became significant at 10-20 min (p < 0.05). The voxel-wise analysis confirmed the differences between the radiotracers. CONCLUSIONS [18F]F-DPA displays higher brain uptake, higher TG-to-WT SUVRCB ratios, and faster clearance than [18F]DPA-714 and [11C]PBR28, and could prove useful for detecting low levels of inflammation and allow for shorter dynamic PET scans.
Collapse
|
42
|
Biechele G, Blume T, Deussing M, Zott B, Shi Y, Xiang X, Franzmeier N, Kleinberger G, Peters F, Ochs K, Focke C, Sacher C, Wind K, Schmidt C, Lindner S, Gildehaus FJ, Eckenweber F, Beyer L, von Ungern-Sternberg B, Bartenstein P, Baumann K, Dorostkar MM, Rominger A, Cumming P, Willem M, Adelsberger H, Herms J, Brendel M. Pre-therapeutic microglia activation and sex determine therapy effects of chronic immunomodulation. Theranostics 2021; 11:8964-8976. [PMID: 34522221 PMCID: PMC8419052 DOI: 10.7150/thno.64022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/06/2021] [Indexed: 12/21/2022] Open
Abstract
Modulation of the innate immune system is emerging as a promising therapeutic strategy against Alzheimer's disease (AD). However, determinants of a beneficial therapeutic effect are ill-understood. Thus, we investigated the potential of 18 kDa translocator protein positron-emission-tomography (TSPO-PET) for assessment of microglial activation in mouse brain before and during chronic immunomodulation. Methods: Serial TSPO-PET was performed during five months of chronic microglia modulation by stimulation of the peroxisome proliferator-activated receptor (PPAR)-γ with pioglitazone in two different mouse models of AD (PS2APP, AppNL-G-F ). Using mixed statistical models on longitudinal TSPO-PET data, we tested for effects of therapy and sex on treatment response. We tested correlations of baseline with longitudinal measures of TSPO-PET, and correlations between PET results with spatial learning performance and β-amyloid accumulation of individual mice. Immunohistochemistry was used to determine the molecular source of the TSPO-PET signal. Results: Pioglitazone-treated female PS2APP and AppNL-G-F mice showed attenuation of the longitudinal increases in TSPO-PET signal when compared to vehicle controls, whereas treated male AppNL-G-F mice showed the opposite effect. Baseline TSPO-PET strongly predicted changes in microglial activation in treated mice (R = -0.874, p < 0.0001) but not in vehicle controls (R = -0.356, p = 0.081). Reduced TSPO-PET signal upon pharmacological treatment was associated with better spatial learning despite higher fibrillar β-amyloid accumulation. Immunohistochemistry confirmed activated microglia to be the source of the TSPO-PET signal (R = 0.952, p < 0.0001). Conclusion: TSPO-PET represents a sensitive biomarker for monitoring of immunomodulation and closely reflects activated microglia. Sex and pre-therapeutic assessment of baseline microglial activation predict individual immunomodulation effects and may serve for responder stratification.
Collapse
Affiliation(s)
- Gloria Biechele
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Tanja Blume
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Maximilian Deussing
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Benedikt Zott
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Yuan Shi
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Xianyuan Xiang
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Finn Peters
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Katharina Ochs
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Carola Focke
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Christian Sacher
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Karin Wind
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Claudio Schmidt
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Simon Lindner
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Franz-Josef Gildehaus
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Florian Eckenweber
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Leonie Beyer
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Peter Bartenstein
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- SyNergy, University of Munich, Munich, Germany
| | - Karlheinz Baumann
- Roche Pharma Research and Early Development, Neuroscience Discovery, Roche, Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Mario M. Dorostkar
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Axel Rominger
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- SyNergy, University of Munich, Munich, Germany
- Dept. of Nuclear Medicine, Inselspital Bern, Bern, Switzerland
| | - Paul Cumming
- Dept. of Nuclear Medicine, Inselspital Bern, Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, Australia
| | - Michael Willem
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Jochen Herms
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
- SyNergy, University of Munich, Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Matthias Brendel
- Dept. of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- SyNergy, University of Munich, Munich, Germany
| |
Collapse
|
43
|
Wang J, Beecher K. TSPO: an emerging role in appetite for a therapeutically promising biomarker. Open Biol 2021; 11:210173. [PMID: 34343461 PMCID: PMC8331234 DOI: 10.1098/rsob.210173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
There is accumulating evidence that an obesogenic Western diet causes neuroinflammatory damage to the brain, which then promotes further appetitive behaviour. Neuroinflammation has been extensively studied by analysing the translocator protein of 18 kDa (TSPO), a protein that is upregulated in the inflamed brain following a damaging stimulus. As a result, there is a rich supply of TSPO-specific agonists, antagonists and positron emission tomography ligands. One TSPO ligand, etifoxine, is also currently used clinically for the treatment of anxiety with a minimal side-effect profile. Despite the neuroinflammatory pathogenesis of diet-induced obesity, and the translational potential of targeting TSPO, there is sparse literature characterizing the effect of TSPO on appetite. Therefore, in this review, the influence of TSPO on appetite is discussed. Three putative mechanisms for TSPO's appetite-modulatory effect are then characterized: the TSPO–allopregnanolone–GABAAR signalling axis, glucosensing in tanycytes and association with the synaptic protein RIM-BP1. We highlight that, in addition to its plethora of functions, TSPO is a regulator of appetite. This review ultimately suggests that the appetite-modulating function of TSPO should be further explored due to its potential therapeutic promise.
Collapse
Affiliation(s)
- Joshua Wang
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kate Beecher
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
44
|
Ji B, Ono M, Yamasaki T, Fujinaga M, Zhang MR, Seki C, Aoki I, Kito S, Sawada M, Suhara T, Sahara N, Higuchi M. Detection of Alzheimer's disease-related neuroinflammation by a PET ligand selective for glial versus vascular translocator protein. J Cereb Blood Flow Metab 2021; 41:2076-2089. [PMID: 33557690 PMCID: PMC8327108 DOI: 10.1177/0271678x21992457] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A substantial and constitutive expression of translocator protein (TSPO) in cerebral blood vessels hampers the sensitive detection of neuroinflammation characterized by greatly induced TSPO expression in activated glia. Here, we conducted in vivo positron emission tomography (PET) and in vitro autoradiographic imaging of normal and TSPO-deficient mouse brains to compare the binding properties of 18F-FEBMP, a relatively novel TSPO radioligand developed for human studies based on its insensitivity to a common polymorphism, with 11C-PK11195, as well as other commonly used TSPO radioligands including 11C-PBR28, 11C-Ac5216 and 18F-FEDAA1106. TSPO in cerebral vessels of normal mice was found to provide a major binding site for 11C-PK11195, 11C-PBR28 and 18F-FEDAA1106, in contrast to no overt specific binding of 18F-FEBMP and 11C-Ac5216 to this vascular component. In addition, 18F-FEBMP yielded PET images of microglial TSPO with a higher contrast than 11C-PK11195 in a tau transgenic mouse modeling Alzheimer's disease (AD) and allied neurodegenerative tauopathies. Moreover, TSPO expression examined by immunoblotting was significantly increased in AD brains compared with healthy controls, and was well correlated with the autoradiographic binding of 18F-FEBMP but not 11C-PK11195. Our findings support the potential advantage of comparatively glial TSPO-selective radioligands such as 18F-FEBMP for PET imaging of inflammatory glial cells.
Collapse
Affiliation(s)
- Bin Ji
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Maiko Ono
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Tomoteru Yamasaki
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Masayuki Fujinaga
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Ming-Rong Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Chie Seki
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Ichio Aoki
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Seiji Kito
- Research, Development and Support Center, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Makoto Sawada
- Department of Brain Function, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Tetsuya Suhara
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Naruhiko Sahara
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
45
|
Veronese M, Tuosto M, Marques TR, Howes O, Pascual B, Yu M, Masdeu JC, Turkheimer F, Bertoldo A, Zanotti-Fregonara P. Parametric Mapping for TSPO PET Imaging with Spectral Analysis Impulsive Response Function. Mol Imaging Biol 2021; 23:560-571. [PMID: 33475944 PMCID: PMC8277653 DOI: 10.1007/s11307-020-01575-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/27/2020] [Accepted: 12/21/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim of this study was to investigate the use of spectral analysis (SA) for voxel-wise analysis of TSPO PET imaging studies. TSPO PET quantification is methodologically complicated by the heterogeneity of TSPO expression and its cell-dependent modulation during neuroinflammatory response. Compartmental models to account for this complexity exist, but they are unreliable at the high noise typical of voxel data. On the contrary, SA is noise-robust for parametric mapping and provides useful information about tracer kinetics with a free compartmental structure. PROCEDURES SA impulse response function (IRF) calculated at 90 min after tracer injection was used as main parameter of interest in 3 independent PET imaging studies to investigate its sensitivity to (1) a TSPO genetic polymorphism (rs6971) known to affect tracer binding in a cross-sectional analysis of healthy controls scanned with [11C]PBR28 PET; (2) TSPO density with [11C]PBR28 in a competitive blocking study with a TSPO blocker, XBD173; and (3) the higher affinity of a second radiotracer for TSPO, by using data from a head-to-head comparison between [11C]PBR28 and [11C]ER176 scans. RESULTS SA-IRF produced parametric maps of visually good quality. These were sensitive to TSPO genotype (mean relative difference between high- and mixed-affinity binders = 25 %) and TSPO availability (mean signal displacement after 90 mg oral administration of XBD173 = 39 %). Regional averages of voxel-wise IRF estimates were strongly associated with regional total distribution volume (VT) estimated with a 2-tissue compartmental model with vascular compartment (Pearson's r = 0.86 ± 0.11) but less strongly with standard 2TCM-VT (Pearson's r = 0.76 ± 0.32). Finally, SA-IRF estimates for [11C]ER176 were significantly higher than [11C]PBR28 ones, consistent with the higher amount of specific binding of the former tracer. CONCLUSIONS SA-IRF can be used for voxel-wise quantification of TSPO PET data because it generates high-quality parametric maps, it is sensitive to TSPO availability and genotype, and it accounts for the complexity of TSPO tracer kinetics with no additional assumptions.
Collapse
Affiliation(s)
- Mattia Veronese
- Department of Neuroimaging, IoPPN, King's College London, London, UK.
| | - Marcello Tuosto
- Department of Information Engineering, Padova University, Padova, Italy
| | - Tiago Reis Marques
- Department of Psychosis Studies, IoPPN, King's College London, London, UK
| | - Oliver Howes
- Department of Psychosis Studies, IoPPN, King's College London, London, UK
- MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Belen Pascual
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Meixiang Yu
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | | | - Alessandra Bertoldo
- Department of Information Engineering, Padova University, Padova, Italy
- Padova Neuroscience Centre, Padova University, Padova, Italy
| | - Paolo Zanotti-Fregonara
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| |
Collapse
|
46
|
MRI and PET of Brain Tumor Neuroinflammation in the Era of Immunotherapy, From the AJR Special Series on Inflammation. AJR Am J Roentgenol 2021; 218:582-596. [PMID: 34259035 DOI: 10.2214/ajr.21.26159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
With the emergence of immune-modulating therapies, brain tumors present significant diagnostic imaging challenges. These challenges include planning personalized treatment and adjudicating accurate monitoring approaches and therapeutically specific response criteria. This has been due, in part, to the reliance on nonspecific imaging metrics, such as gadolinium-contrast-enhanced MRI or FDG PET, and rapidly evolving biologic understanding of neuroinflammation. The importance of the tumor-immune interaction and ability to therapeutically augment inflammation to improve clinical outcomes necessitates that the radiologist develop a working knowledge of the immune system and its role in clinical neuroimaging. In this article, we review relevant biologic concepts of the tumor microenvironment of primary and metastatic brain tumors, these tumors' interactions with the immune system, and MRI and PET methods for imaging inflammatory elements associated with these malignancies. Recognizing the growing fields of immunotherapeutics and precision oncology, we highlight clinically translatable imaging metrics for the diagnosis and monitoring of brain tumor neuroinflammation. Practical guidance is provided for implementing iron nanoparticle imaging, including imaging indications, protocol, interpretation, and pitfalls. A comprehensive understanding of the inflammatory mechanisms within brain tumors and their imaging features will facilitate the development of innovative non-invasive prognostic and predictive imaging strategies for precision oncology.
Collapse
|
47
|
Giordani A, Menziani MC, Moresco RM, Matarrese M, Paolino M, Saletti M, Giuliani G, Anzini M, Cappelli A. Exploring Translocator Protein (TSPO) Medicinal Chemistry: An Approach for Targeting Radionuclides and Boron Atoms to Mitochondria. J Med Chem 2021; 64:9649-9676. [PMID: 34254805 DOI: 10.1021/acs.jmedchem.1c00379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Translocator protein 18 kDa [TSPO or peripheral-type benzodiazepine receptor (PBR)] was identified in the search of binding sites for benzodiazepine anxiolytic drugs in peripheral regions. In these areas, binding sites for TSPO ligands were recognized in steroid-producing tissues. TSPO plays an important role in many cellular functions, and its coding sequence is highly conserved across species. TSPO is located predominantly on the membrane of mitochondria and is overexpressed in several solid cancers. TSPO basal expression in the CNS is low, but it becomes high in neurodegenerative conditions. Thus, TSPO constitutes not only as an outstanding drug target but also as a valuable marker for the diagnosis of a number of diseases. The aim of the present article is to show the lesson we have learned from our activity in TSPO medicinal chemistry and in approaching the targeted delivery to mitochondria by means of TSPO ligands.
Collapse
Affiliation(s)
- Antonio Giordani
- Rottapharm Biotech S.p.A., Via Valosa di Sopra 9, 20900 Monza, Italy
| | - Maria Cristina Menziani
- Dipartimento di Scienze Chimiche e Geologiche, Università di Modena e Reggio Emilia, Via Campi 103, 41121 Modena, Italy
| | - Rosa Maria Moresco
- Department of Medicine and Surgery, University of Milan-Bicocca, Nuclear Medicine Department, San Raffaele Scientific Institute, IBFM-CNR, Via Olgettina 60, 20132 Milano, Italy
| | - Mario Matarrese
- Department of Medicine and Surgery, University of Milan-Bicocca, Nuclear Medicine Department, San Raffaele Scientific Institute, IBFM-CNR, Via Olgettina 60, 20132 Milano, Italy
| | - Marco Paolino
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Mario Saletti
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Germano Giuliani
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Maurizio Anzini
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Andrea Cappelli
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
48
|
Patkar OL, Mohamed AZ, Narayanan A, Mardon K, Cowin G, Bhalla R, Stimson DHR, Kassiou M, Beecher K, Belmer A, Alvarez Cooper I, Morgan M, Hume DA, Irvine KM, Bartlett SE, Nasrallah F, Cumming P. A binge high sucrose diet provokes systemic and cerebral inflammation in rats without inducing obesity. Sci Rep 2021; 11:11252. [PMID: 34045616 PMCID: PMC8160215 DOI: 10.1038/s41598-021-90817-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
While the dire cardiometabolic consequences of the hypercaloric modern 'Western' diet are well known, there is not much information on the health impact of a high sucrose diet not inducing weight gain. Here, we tested the hypothesis that rats reared with intermittent binge access to sucrose in addition to normal chow would develop an inflammatory response in brain. To test this hypothesis, we undertook serial PET/MRI scans with the TSPO ligand [18F]DPA714 in a group of (n=9) rats at baseline and again after voluntarily consuming 5% sucrose solution three days a week for three months. Compared to a control group fed with normal chow (n=9), the sucrose rats indeed showed widespread increases in the availability of cerebral binding sites for the microglial marker, despite normal weight gain compared to the control diet group. Subsequent immunofluorescence staining of the brains confirmed the PET findings, showing a widespread 20% increase in the abundance of IBA-1-positive microglia with characteristic 'semi-activated' morphology in the binge sucrose rats, which had 23% lower density of microglial endpoints and 25% lower mean process length compared to microglia in the control rats with ordinary feeding. GFAP immunofluorescence showed no difference in astroglial coverage in the sucrose rats, except for a slight reduction in hypothalamus. The binge sucrose diet-induced neuroinflammation was associated with a significant elevation of white blood cell counts. Taking these results together, we find that long-term intake of sucrose in a binge paradigm, similar in sucrose content to the contemporary Western diet, triggered a low-grade systemic and central inflammation in non-obese rats. The molecular mechanism of this phenomenon remains to be established.
Collapse
Affiliation(s)
- Omkar L Patkar
- Macrophage Biology Group, Mater Research, Translational Research Institute, Brisbane, QLD, Australia
- Department of Nuclear Medicine, Inselspital, Bern University, Bern, Switzerland
| | - Abdalla Z Mohamed
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Ashwin Narayanan
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Karine Mardon
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Gary Cowin
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Rajiv Bhalla
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Damion H R Stimson
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Michael Kassiou
- School of Chemistry, University of Sydney, Sydney, NSW, 2006, Australia
| | - Kate Beecher
- Queensland University of Technology, Translational Research Institute, Brisbane, QLD, Australia
| | - Arnauld Belmer
- Queensland University of Technology, Translational Research Institute, Brisbane, QLD, Australia
| | - Ignatius Alvarez Cooper
- Queensland University of Technology, Translational Research Institute, Brisbane, QLD, Australia
| | - Michael Morgan
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Australia
| | - David A Hume
- Macrophage Biology Group, Mater Research, Translational Research Institute, Brisbane, QLD, Australia
| | - Katharine M Irvine
- Macrophage Biology Group, Mater Research, Translational Research Institute, Brisbane, QLD, Australia
| | - Selena E Bartlett
- Queensland University of Technology, Translational Research Institute, Brisbane, QLD, Australia
| | - Fatima Nasrallah
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Paul Cumming
- Department of Nuclear Medicine, Inselspital, Bern University, Bern, Switzerland.
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
49
|
Potential Effects of Leukotriene Receptor Antagonist Montelukast in Treatment of Neuroinflammation in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22115606. [PMID: 34070609 PMCID: PMC8198163 DOI: 10.3390/ijms22115606] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/16/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder where misfolded alpha-synuclein-enriched aggregates called Lewy bodies are central in pathogenesis. No neuroprotective or disease-modifying treatments are currently available. Parkinson’s disease is considered a multifactorial disease and evidence from multiple patient studies and animal models has shown a significant immune component during the course of the disease, highlighting immunomodulation as a potential treatment strategy. The immune changes occur centrally, involving microglia and astrocytes but also peripherally with changes to the innate and adaptive immune system. Here, we review current understanding of different components of the PD immune response with a particular emphasis on the leukotriene pathway. We will also describe evidence of montelukast, a leukotriene receptor antagonist, as a possible anti-inflammatory treatment for PD.
Collapse
|
50
|
Schollhammer R, Lepreux S, Barthe N, Vimont D, Rullier A, Sibon I, Berard X, Zhang A, Kimura Y, Fujita M, Innis RB, Zanotti-Fregonara P, Morgat C. In vitro and pilot in vivo imaging of 18 kDa translocator protein (TSPO) in inflammatory vascular disease. EJNMMI Res 2021; 11:45. [PMID: 33950298 PMCID: PMC8099943 DOI: 10.1186/s13550-021-00786-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/27/2021] [Indexed: 11/25/2022] Open
Abstract
Background Inflammatory vascular disease of the arteries, such as inflamed atheromatous plaques or arteritis, may cause aneurysms or ischemic strokes. In this context, using positron emission tomography (PET) to image inflammation may help select patients who would benefit from appropriate therapeutic interventions. This study sought to assess the usefulness of the 18 kDa translocator protein (TSPO) tracers [11C]-PBR28 and [18F]-PBR06 for imaging inflammatory vascular disease in vitro and in vivo. Immunohistochemistry for macrophage infiltration as well as autoradiography with [18F]-PBR06 were performed on eight paraffin-embedded, formalin-fixed atherosclerosis plaques prospectively collected after carotid endarterectomy of eight patients affected by ischemic stroke. Six different patients, one of whom was also included in the in vitro study, underwent PET imaging. Two patients with carotid stenosis associated with ischemic stroke were imaged with [18F]-PBR06 PET/CT, and four other patients (three with large vessel vasculitis and one with bilateral carotid stenosis but without stroke) were imaged with [11C]-PBR28. Results All in vitro sections showed specific binding of [18F]-PBR06, which co-localized with immunohistochemistry markers for inflammation. However, in vivo TSPO imaging with either [11C]-PBR28 or [18F]-PBR06 was negative in all participants. Conclusion Despite good uptake on surgical samples in vitro, [11C]-PBR28 and [18F]-PBR06 are not viable clinical tools for imaging inflammatory vascular disease. Trial registration: NCT02513589, registered 31 July 2015 and NCT00547976, registered 23 October 2007. https://clinicaltrials.gov.
Collapse
Affiliation(s)
- Romain Schollhammer
- Nuclear Medicine Department, University Hospital of Bordeaux, 33076, Bordeaux, France. .,University of Bordeaux, INCIA, UMR5287, 33400, Talence, France. .,CNRS, INCIA, UMR5287, 33400, Talence, France. .,Nuclear Medicine Department, University Hospital of Bordeaux, Place Amélie Raba Léon, 33000, Bordeaux, France.
| | | | | | - Delphine Vimont
- University of Bordeaux, INCIA, UMR5287, 33400, Talence, France.,CNRS, INCIA, UMR5287, 33400, Talence, France
| | - Anne Rullier
- Histologic Department, University Hospital of Bordeaux, 33076, Bordeaux, France
| | - Igor Sibon
- Neurology Department, University Hospital of Bordeaux, 33076, Bordeaux, France
| | - Xavier Berard
- Vascular Surgery Department, University Hospital of Bordeaux, 33076, Bordeaux, France
| | - Andrea Zhang
- Molecular Imaging Branch, NIMH, Bethesda, MD, USA
| | | | | | | | | | - Clément Morgat
- Nuclear Medicine Department, University Hospital of Bordeaux, 33076, Bordeaux, France.,University of Bordeaux, INCIA, UMR5287, 33400, Talence, France.,CNRS, INCIA, UMR5287, 33400, Talence, France
| |
Collapse
|