1
|
Fairley LH, Lai KO, Grimm A, Eckert A, Barron AM. The mitochondrial translocator protein (TSPO) in Alzheimer's disease: Therapeutic and immunomodulatory functions. Biochimie 2024; 224:120-131. [PMID: 38971458 DOI: 10.1016/j.biochi.2024.07.003] [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: 01/12/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
The translocator protein (TSPO) has been widely investigated as a PET-imaging biomarker of neuroinflammation and, more recently, as a therapeutic target for the treatment of neurodegenerative disease. TSPO ligands have been shown to exert neuroprotective effects in vivo and in vitro models of Alzheimer's disease (AD), by reducing toxic beta amyloid peptides, and attenuating brain atrophy. Recent transcriptomic and proteomic analyses, and the generation of TSPO-KO mice, have enabled new insights into the mechanistic function of TSPO in AD. Using a multi-omics approach in both TSPO-KO- and TSPO ligand-treated mice, we have demonstrated a key role for TSPO in microglial respiratory metabolism and phagocytosis in AD. In this review, we discuss emerging evidence for therapeutic and immunomodulatory functions of TSPO in AD, and new tools for studying TSPO in the brain.
Collapse
Affiliation(s)
- Lauren H Fairley
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 308232, Singapore
| | - Kei Onn Lai
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 308232, Singapore
| | - Amandine Grimm
- Transfaculty Research Platform, Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland; Psychiatric University Clinics, Basel, Switzerland
| | - Anne Eckert
- Transfaculty Research Platform, Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland; Psychiatric University Clinics, Basel, Switzerland
| | - Anna M Barron
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 308232, Singapore.
| |
Collapse
|
2
|
Mahemuti Y, Kadeer K, Su R, Abula A, Aili Y, Maimaiti A, Abulaiti S, Maimaitituerxun M, Miao T, Jiang S, Axier A, Aisha M, Wang Y, Cheng X. TSPO exacerbates acute cerebral ischemia/reperfusion injury by inducing autophagy dysfunction. Exp Neurol 2023; 369:114542. [PMID: 37717810 DOI: 10.1016/j.expneurol.2023.114542] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/19/2023]
Abstract
Autophagy is considered a double-edged sword, with a role in the regulation of the pathophysiological processes of the central nervous system (CNS) after cerebral ischemia-reperfusion injury (CIRI). The 18-kDa translocator protein (TSPO) is a highly conserved protein, with its expression level in the nervous system closely associated with the regulation of pathophysiological processes. In addition, the ligand of TSPO reduces neuroinflammation in brain diseases, but the potential role of TSPO in CIRI is largely undiscovered. On this basis, we investigated whether TSPO regulates neuroinflammatory response by affecting autophagy in microglia. In our study, increased expression of TSPO was detected in rat brain tissues with transient middle cerebral artery occlusion (tMCAO) and in BV2 microglial cells exposed to oxygen-glucose deprivation or reoxygenation (OGD/R) treatment, respectively. In addition, we confirmed that autophagy was over-activated during CIRI by increased expression of autophagy activation related proteins with Beclin-1 and LC3B, while the expression of p62 was decreased. The degradation process of autophagy was inhibited, while the expression levels of LAMP-1 and Cathepsin-D were significantly reduced. Results of confocal laser microscopy and transmission electron microscopy (TEM) indicated that autophagy flux was disordered. In contrast, inhibition of TSPO prevented autophagy over-activation both in vivo and in vitro. Interestingly, suppression of TSPO alleviated nerve cell damage by reducing reactive oxygen species (ROS) and pro-inflammatory factors, including TNF-α and IL-6 in microglia cells. In summary, these results indicated that TSPO might affect CIRI by mediating autophagy dysfunction and thus might serve as a potential target for ischemic stroke treatment.
Collapse
Affiliation(s)
- Yusufu Mahemuti
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China; School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, PR China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, Zhejiang, PR China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, Zhejiang, PR China
| | - Kaheerman Kadeer
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Riqing Su
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Abudureheman Abula
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Yirizhati Aili
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Aierpati Maimaiti
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Subinuer Abulaiti
- Department of Epidemiology and Biostatistics, Institute of Public Health, Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | | | - Tong Miao
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Shihao Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Aximujiang Axier
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Maimaitili Aisha
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Yongxin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China
| | - Xiaojiang Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China.
| |
Collapse
|
3
|
Adinolfi A, Di Sante G, Rivignani Vaccari L, Tredicine M, Ria F, Bonvissuto D, Corvino V, Sette C, Geloso MC. Regionally restricted modulation of Sam68 expression and Arhgef9 alternative splicing in the hippocampus of a murine model of multiple sclerosis. Front Mol Neurosci 2023; 15:1073627. [PMID: 36710925 PMCID: PMC9878567 DOI: 10.3389/fnmol.2022.1073627] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/21/2022] [Indexed: 01/15/2023] Open
Abstract
Multiple sclerosis (MS) and its preclinical models are characterized by marked changes in neuroplasticity, including excitatory/inhibitory imbalance and synaptic dysfunction that are believed to underlie the progressive cognitive impairment (CI), which represents a significant clinical hallmark of the disease. In this study, we investigated several parameters of neuroplasticity in the hippocampus of the experimental autoimmune encephalomyelitis (EAE) SJL/J mouse model, characterized by rostral inflammatory and demyelinating lesions similar to Relapsing-Remitting MS. By combining morphological and molecular analyses, we found that the hippocampus undergoes extensive inflammation in EAE-mice, more pronounced in the CA3 and dentate gyrus (DG) subfields than in the CA1, associated with changes in GABAergic circuitry, as indicated by the increased expression of the interneuron marker Parvalbumin selectively in CA3. By laser-microdissection, we investigated the impact of EAE on the alternative splicing of Arhgef9, a gene encoding a post-synaptic protein playing an essential role in GABAergic synapses and whose mutations have been related to CI and epilepsy. Our results indicate that EAE induces a specific increase in inclusion of the alternative exon 11a only in the CA3 and DG subfields, in line with the higher local levels of inflammation. Consistently, we found a region-specific downregulation of Sam68, a splicing-factor that represses this splicing event. Collectively, our findings confirm a regionalized distribution of inflammation in the hippocampus of EAE-mice. Moreover, since neuronal circuit rearrangement and dynamic remodeling of structural components of the synapse are key processes that contribute to neuroplasticity, our study suggests potential new molecular players involved in EAE-induced hippocampal dysfunction.
Collapse
Affiliation(s)
- Annalisa Adinolfi
- Section of Human Anatomy, Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gabriele Di Sante
- Section of Human, Clinic and Forensic Anatomy, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Luca Rivignani Vaccari
- Section of Human Anatomy, Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Tredicine
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Ria
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Davide Bonvissuto
- Section of Human Anatomy, Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Corvino
- Section of Human Anatomy, Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudio Sette
- Section of Human Anatomy, Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy,GSTEP-Organoids Core Facility, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy,*Correspondence: Claudio Sette, ✉
| | - Maria Concetta Geloso
- Section of Human Anatomy, Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy,Maria Concetta Geloso, ✉
| |
Collapse
|
4
|
18F-Radiolabeled Translocator Protein (TSPO) PET Tracers: Recent Development of TSPO Radioligands and Their Application to PET Study. Pharmaceutics 2022; 14:pharmaceutics14112545. [PMID: 36432736 PMCID: PMC9697781 DOI: 10.3390/pharmaceutics14112545] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Translocator protein 18 kDa (TSPO) is a transmembrane protein in the mitochondrial membrane, which has been identified as a peripheral benzodiazepine receptor. TSPO is generally present at high concentrations in steroid-producing cells and plays an important role in steroid synthesis, apoptosis, and cell proliferation. In the central nervous system, TSPO expression is relatively modest under normal physiological circumstances. However, some pathological disorders can lead to changes in TSPO expression. Overexpression of TSPO is associated with several diseases, such as neurodegenerative diseases, neuroinflammation, brain injury, and cancers. TSPO has therefore become an effective biomarker of related diseases. Positron emission tomography (PET), a non-invasive molecular imaging technique used for the clinical diagnosis of numerous diseases, can detect diseases related to TSPO expression. Several radiolabeled TSPO ligands have been developed for PET. In this review, we describe recent advances in the development of TSPO ligands, and 18F-radiolabeled TSPO in particular, as PET tracers. This review covers pharmacokinetic studies, preclinical and clinical trials of 18F-labeled TSPO PET ligands, and the synthesis of TSPO ligands.
Collapse
|
5
|
Van Camp N, Lavisse S, Roost P, Gubinelli F, Hillmer A, Boutin H. TSPO imaging in animal models of brain diseases. Eur J Nucl Med Mol Imaging 2021; 49:77-109. [PMID: 34245328 PMCID: PMC8712305 DOI: 10.1007/s00259-021-05379-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/25/2021] [Indexed: 12/19/2022]
Abstract
Over the last 30 years, the 18-kDa TSPO protein has been considered as the PET imaging biomarker of reference to measure increased neuroinflammation. Generally assumed to image activated microglia, TSPO has also been detected in endothelial cells and activated astrocytes. Here, we provide an exhaustive overview of the recent literature on the TSPO-PET imaging (i) in the search and development of new TSPO tracers and (ii) in the understanding of acute and chronic neuroinflammation in animal models of neurological disorders. Generally, studies testing new TSPO radiotracers against the prototypic [11C]-R-PK11195 or more recent competitors use models of acute focal neuroinflammation (e.g. stroke or lipopolysaccharide injection). These studies have led to the development of over 60 new tracers during the last 15 years. These studies highlighted that interpretation of TSPO-PET is easier in acute models of focal lesions, whereas in chronic models with lower or diffuse microglial activation, such as models of Alzheimer's disease or Parkinson's disease, TSPO quantification for detection of neuroinflammation is more challenging, mirroring what is observed in clinic. Moreover, technical limitations of preclinical scanners provide a drawback when studying modest neuroinflammation in small brains (e.g. in mice). Overall, this review underlines the value of TSPO imaging to study the time course or response to treatment of neuroinflammation in acute or chronic models of diseases. As such, TSPO remains the gold standard biomarker reference for neuroinflammation, waiting for new radioligands for other, more specific targets for neuroinflammatory processes and/or immune cells to emerge.
Collapse
Affiliation(s)
- Nadja Van Camp
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Sonia Lavisse
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Pauline Roost
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Francesco Gubinelli
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Ansel Hillmer
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, CT, USA
| | - Hervé Boutin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Brain and Mental Health, University of Manchester, M13 9PL, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, 27 Palatine Road, M20 3LJ, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
| |
Collapse
|
6
|
Daïen C, Tan J, Audo R, Mielle J, Quek L, Krycer J, Angelatos A, Duraes M, Pinget G, Ni D, Robert R, Alam M, Amian M, Sierro F, Parmar A, Perkins G, Hoque S, Gosby A, Simpson S, Ribeiro R, Mackay C, Macia L. Gut-derived acetate promotes B10 cells with antiinflammatory effects. JCI Insight 2021; 6:144156. [PMID: 33729999 PMCID: PMC8119207 DOI: 10.1172/jci.insight.144156] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
Autoimmune diseases are characterized by a breakdown of immune tolerance partly due to environmental factors. The short-chain fatty acid acetate, derived mostly from gut microbial fermentation of dietary fiber, promotes antiinflammatory Tregs and protects mice from type 1 diabetes, colitis, and allergies. Here, we show that the effects of acetate extend to another important immune subset involved in tolerance, the IL-10-producing regulatory B cells (B10 cells). Acetate directly promoted B10 cell differentiation from mouse B1a cells both in vivo and in vitro. These effects were linked to metabolic changes through the increased production of acetyl-coenzyme A, which fueled the TCA cycle and promoted posttranslational lysine acetylation. Acetate also promoted B10 cells from human blood cells through similar mechanisms. Finally, we identified that dietary fiber supplementation in healthy individuals was associated with increased blood-derived B10 cells. Direct delivery of acetate or indirect delivery via diets or bacteria that produce acetate might be a promising approach to restore B10 cells in noncommunicable diseases.
Collapse
MESH Headings
- Acetates/blood
- Acetates/metabolism
- Acetates/pharmacology
- Acetyl Coenzyme A/metabolism
- Acetylation
- Animals
- Arthritis, Experimental/immunology
- Arthritis, Experimental/therapy
- B-Lymphocytes, Regulatory/drug effects
- B-Lymphocytes, Regulatory/physiology
- B-Lymphocytes, Regulatory/transplantation
- Cell Differentiation/drug effects
- Dietary Fiber/pharmacology
- Fatty Acids, Volatile/metabolism
- Fatty Acids, Volatile/pharmacology
- Female
- Humans
- Interleukin-10
- Male
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Neutrophils/cytology
- Neutrophils/drug effects
- Receptors, G-Protein-Coupled/genetics
- Mice
Collapse
Affiliation(s)
- C.I. Daïen
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- Faculty of Medicine and Health, The University of Sydney School of Medicine, New South Wales, Sydney, Australia
- Department of Rheumatology, Montpellier Hospital, University of Montpellier, Montpellier, France
- Institute of Molecular Genetics of Montpellier, UMR5535, University of Montpellier, Montpellier, France
| | - J. Tan
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- Faculty of Medicine and Health, The University of Sydney School of Medicine, New South Wales, Sydney, Australia
- Human Health, Nuclear Science & Technology and Landmark Infrastructure (NSTLI) Australian Nuclear Science and Technology Organisation, New South Wales, Sydney, Australia
| | - R. Audo
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- Faculty of Medicine and Health, The University of Sydney School of Medicine, New South Wales, Sydney, Australia
- Department of Rheumatology, Montpellier Hospital, University of Montpellier, Montpellier, France
- Institute of Molecular Genetics of Montpellier, UMR5535, University of Montpellier, Montpellier, France
| | - J. Mielle
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- Faculty of Medicine and Health, The University of Sydney School of Medicine, New South Wales, Sydney, Australia
- Institute of Molecular Genetics of Montpellier, UMR5535, University of Montpellier, Montpellier, France
| | - L.E. Quek
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- School of Mathematics and Statistics and
| | - J.R. Krycer
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- School of Life and Environmental Sciences, The University of Sydney, New South Wales, Sydney, Australia
| | - A. Angelatos
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- Faculty of Medicine and Health, The University of Sydney School of Medicine, New South Wales, Sydney, Australia
| | - M. Duraes
- Department of Gynecology, Montpellier Hospital, University of Montpellier, Montpellier, France
| | - G. Pinget
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- Faculty of Medicine and Health, The University of Sydney School of Medicine, New South Wales, Sydney, Australia
| | - D. Ni
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- Faculty of Medicine and Health, The University of Sydney School of Medicine, New South Wales, Sydney, Australia
| | | | - M.J. Alam
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - M.C.B. Amian
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- Faculty of Medicine and Health, The University of Sydney School of Medicine, New South Wales, Sydney, Australia
- School of Life and Environmental Sciences, The University of Sydney, New South Wales, Sydney, Australia
| | - F. Sierro
- Faculty of Medicine and Health, The University of Sydney School of Medicine, New South Wales, Sydney, Australia
- Human Health, Nuclear Science & Technology and Landmark Infrastructure (NSTLI) Australian Nuclear Science and Technology Organisation, New South Wales, Sydney, Australia
| | - A. Parmar
- Human Health, Nuclear Science & Technology and Landmark Infrastructure (NSTLI) Australian Nuclear Science and Technology Organisation, New South Wales, Sydney, Australia
- Brain and Mind Centre, The University of Sydney, New South Wales, Sydney, Australia
| | - G. Perkins
- Biosciences platform, NSTLI Australian Nuclear Science and Technology Organisation, New South Wales, Sydney, Australia
| | - S. Hoque
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- School of Mathematics and Statistics and
| | - A.K. Gosby
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- School of Life and Environmental Sciences, The University of Sydney, New South Wales, Sydney, Australia
| | - S.J. Simpson
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- School of Life and Environmental Sciences, The University of Sydney, New South Wales, Sydney, Australia
| | - R.V. Ribeiro
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- School of Life and Environmental Sciences, The University of Sydney, New South Wales, Sydney, Australia
| | | | - L. Macia
- Charles Perkins Centre, The University of Sydney, New South Wales, Sydney, Australia
- Faculty of Medicine and Health, The University of Sydney School of Medicine, New South Wales, Sydney, Australia
| |
Collapse
|
7
|
Behrangi N, Lorenz P, Kipp M. Oligodendrocyte Lineage Marker Expression in eGFP-GFAP Transgenic Mice. J Mol Neurosci 2020; 71:2237-2248. [PMID: 33346907 PMCID: PMC8585802 DOI: 10.1007/s12031-020-01771-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022]
Abstract
Oligodendrocytes, the myelinating cells of the central nervous system, orchestrate several key cellular functions in the brain and spinal cord, including axon insulation, energy transfer to neurons, and, eventually, modulation of immune responses. There is growing interest for obtaining reliable markers that can specifically label oligodendroglia and their progeny. In many studies, anti-CC1 antibodies, presumably recognizing the protein adenomatous polyposis coli (APC), are used to label mature, myelinating oligodendrocytes. However, it has been discussed whether anti-CC1 antibodies could recognize as well, under pathological conditions, other cell populations, particularly astrocytes. In this study, we used transgenic mice in which astrocytes are labeled by the enhanced green fluorescent protein (eGFP) under the control of the human glial fibrillary acidic protein (GFAP) promoter. By detailed co-localization studies we were able to demonstrate that a significant proportion of eGFP-expressing cells co-express markers of the oligodendrocyte lineage, such as the transcription factor Oligodendrocyte Transcription Factor 2 (OLIG2); the NG2 proteoglycan, also known as chrondroitin sulfate proteoglycan 4 (CSPG4); or APC. The current finding that the GFAP promoter drives transgene expression in cells of the oligodendrocyte lineage should be considered when interpreting results from co-localization studies.
Collapse
Affiliation(s)
- Newshan Behrangi
- Institute of Anatomy, Rostock University Medical Center, 18057, Rostock, Germany.,Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336, Munich, Germany
| | - Peter Lorenz
- Institute of Immunology, Rostock University Medical Center, 18057, Rostock, Germany
| | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, 18057, Rostock, Germany. .,Center for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Center, Gelsheimer Strasse 20, 18147, Rostock, Germany.
| |
Collapse
|
8
|
Hermes DJ, Jacobs IR, Key MC, League AF, Yadav-Samudrala BJ, Xu C, McLane VD, Nass SR, Jiang W, Meeker RB, Ignatowska-Jankowska BM, Lichtman AH, Li Z, Wu Z, Yuan H, Knapp PE, Hauser KF, Fitting S. Escalating morphine dosing in HIV-1 Tat transgenic mice with sustained Tat exposure reveals an allostatic shift in neuroinflammatory regulation accompanied by increased neuroprotective non-endocannabinoid lipid signaling molecules and amino acids. J Neuroinflammation 2020; 17:345. [PMID: 33208151 PMCID: PMC7672881 DOI: 10.1186/s12974-020-01971-6] [Citation(s) in RCA: 14] [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: 05/18/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus type-1 (HIV-1) and opiates cause long-term inflammatory insult to the central nervous system (CNS) and worsen disease progression and HIV-1-related neuropathology. The combination of these proinflammatory factors reflects a devastating problem as opioids have high abuse liability and continue to be prescribed for certain patients experiencing HIV-1-related pain. METHODS Here, we examined the impact of chronic (3-month) HIV-1 transactivator of transcription (Tat) exposure to short-term (8-day), escalating morphine in HIV-1 Tat transgenic mice that express the HIV-1 Tat protein in a GFAP promoter-regulated, doxycycline (DOX)-inducible manner. In addition to assessing morphine-induced tolerance in nociceptive responses organized at spinal (i.e., tail-flick) and supraspinal (i.e., hot-plate) levels, we evaluated neuroinflammation via positron emission tomography (PET) imaging using the [18F]-PBR111 ligand, immunohistochemistry, and cytokine analyses. Further, we examined endocannabinoid (eCB) levels, related non-eCB lipids, and amino acids via mass spectrometry. RESULTS: Tat-expressing [Tat(+)] transgenic mice displayed antinociceptive tolerance in the tail withdrawal and hot-plate assays compared to control mice lacking Tat [Tat(-)]. This tolerance was accompanied by morphine-dependent increases in Iba-1 ± 3-nitrotryosine immunoreactive microglia, and alterations in pro- and anti-inflammatory cytokines, and chemokines in the spinal cord and striatum, while increases in neuroinflammation were absent by PET imaging of [18F]-PBR111 uptake. Tat and morphine exposure differentially affected eCB levels, non-eCB lipids, and specific amino acids in a region-dependent manner. In the striatum, non-eCB lipids were significantly increased by short-term, escalating morphine exposure, including peroxisome proliferator activator receptor alpha (PPAR-α) ligands N-oleoyl ethanolamide (OEA) and N-palmitoyl ethanolamide (PEA), as well as the amino acids phenylalanine and proline. In the spinal cord, Tat exposure increased amino acids leucine and valine, while morphine decreased levels of tyrosine and valine but did not affect eCBs or non-eCB lipids. CONCLUSION Overall results demonstrate that 3 months of Tat exposure increased morphine tolerance and potentially innate immune tolerance evidenced by reductions in specific cytokines (e.g., IL-1α, IL-12p40) and microglial reactivity. In contrast, short-term, escalating morphine exposure acted as a secondary stressor revealing an allostatic shift in CNS baseline inflammatory responsiveness from sustained Tat exposure.
Collapse
Affiliation(s)
- Douglas J Hermes
- Department of Psychology & Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Ian R Jacobs
- Department of Psychology & Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Megan C Key
- Department of Psychology & Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Alexis F League
- Department of Psychology & Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | | | - Changqing Xu
- Department of Psychology & Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Virginia D McLane
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Sara R Nass
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Rick B Meeker
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | | | - Aron H Lichtman
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Zibo Li
- Department of Radiology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Zhanhong Wu
- Department of Radiology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Hong Yuan
- Department of Radiology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Pamela E Knapp
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Kurt F Hauser
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Sylvia Fitting
- Department of Psychology & Neuroscience, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
9
|
Banati RB, Wilcox P, Xu R, Yin G, Si E, Son ET, Shimizu M, Holsinger RMD, Parmar A, Zahra D, Arthur A, Middleton RJ, Liu GJ, Charil A, Graeber MB. Selective, high-contrast detection of syngeneic glioblastoma in vivo. Sci Rep 2020; 10:9968. [PMID: 32561881 PMCID: PMC7305160 DOI: 10.1038/s41598-020-67036-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 05/19/2020] [Indexed: 01/14/2023] Open
Abstract
Glioblastoma is a highly malignant, largely therapy-resistant brain tumour. Deep infiltration of brain tissue by neoplastic cells represents the key problem of diffuse glioma. Much current research focuses on the molecular makeup of the visible tumour mass rather than the cellular interactions in the surrounding brain tissue infiltrated by the invasive glioma cells that cause the tumour’s ultimately lethal outcome. Diagnostic neuroimaging that enables the direct in vivo observation of the tumour infiltration zone and the local host tissue responses at a preclinical stage are important for the development of more effective glioma treatments. Here, we report an animal model that allows high-contrast imaging of wild-type glioma cells by positron emission tomography (PET) using [18 F]PBR111, a selective radioligand for the mitochondrial 18 kDa Translocator Protein (TSPO), in the Tspo−/− mouse strain (C57BL/6-Tspotm1GuMu(GuwiyangWurra)). The high selectivity of [18 F]PBR111 for the TSPO combined with the exclusive expression of TSPO in glioma cells infiltrating into null-background host tissue free of any TSPO expression, makes it possible, for the first time, to unequivocally and with uniquely high biological contrast identify peri-tumoral glioma cell invasion at preclinical stages in vivo. Comparison of the in vivo imaging signal from wild-type glioma cells in a null background with the signal in a wild-type host tissue, where the tumour induces the expected TSPO expression in the host’s glial cells, illustrates the substantial extent of the peritumoral host response to the growing tumour. The syngeneic tumour (TSPO+/+) in null background (TSPO−/−) model is thus well suited to study the interaction of the tumour front with the peri-tumoral tissue, and the experimental evaluation of new therapeutic approaches targeting the invasive behaviour of glioblastoma.
Collapse
Affiliation(s)
- Richard B Banati
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia. .,Medical Imaging, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia.
| | - Paul Wilcox
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Ran Xu
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Grace Yin
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Emily Si
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Eric Taeyoung Son
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Mauricio Shimizu
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - R M Damian Holsinger
- Molecular Neuroscience, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Arvind Parmar
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - David Zahra
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Andrew Arthur
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Ryan J Middleton
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia.,Medical Imaging, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Arnaud Charil
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Manuel B Graeber
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
10
|
Coda AR, Anzilotti S, Boscia F, Greco A, Panico M, Gargiulo S, Gramanzini M, Zannetti A, Albanese S, Pignataro G, Annunziato L, Salvatore M, Brunetti A, De Berardinis P, Quarantelli M, Palma G, Pappatà S. In vivo imaging of CNS microglial activation/macrophage infiltration with combined [ 18F]DPA-714-PET and SPIO-MRI in a mouse model of relapsing remitting experimental autoimmune encephalomyelitis. Eur J Nucl Med Mol Imaging 2020; 48:40-52. [PMID: 32378022 PMCID: PMC7835304 DOI: 10.1007/s00259-020-04842-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 04/27/2020] [Indexed: 12/26/2022]
Abstract
Purpose To evaluate the feasibility and sensitivity of multimodality PET/CT and MRI imaging for non-invasive characterization of brain microglial/macrophage activation occurring during the acute phase in a mouse model of relapsing remitting multiple sclerosis (RR-MS) using [18F]DPA-714, a selective radioligand for the 18-kDa translocator protein (TSPO), superparamagnetic iron oxide particles (SPIO), and ex vivo immunohistochemistry. Methods Experimental autoimmune encephalomyelitis (EAE) was induced in female SJL/J mice by immunization with PLP139–151. Seven symptomatic EAE mice and five controls underwent both PET/CT and MRI studies between 11 and 14 days post-immunization. SPIO was injected i.v. in the same animals immediately after [18F]DPA-714 and MRI acquisition was performed after 24 h. Regional brain volumes were defined according to a mouse brain atlas on co-registered PET and SPIO-MRI images. [18F]DPA-714 standardized uptake value (SUV) ratios (SUVR), with unaffected neocortex as reference, and SPIO fractional volumes (SPIO-Vol) were generated. Both SUVR and SPIO-Vol values were correlated with the clinical score (CS) and among them. Five EAE and four control mice underwent immunohistochemical analysis with the aim of identifying activated microglia/macrophage and TSPO expressions. Results SUVR and SPIO-Vol values were significantly increased in EAE compared with controls in the hippocampus (p < 0.01; p < 0.02, respectively), thalamus (p < 0.02; p < 0.05, respectively), and cerebellum and brainstem (p < 0.02), while only SPIO-Vol was significantly increased in the caudate/putamen (p < 0.05). Both SUVR and SPIO-Vol values were positively significantly correlated with CS and among them in the same regions. TSPO/Iba1 and F4/80/Prussian blue staining immunohistochemistry suggests that increased activated microglia/macrophages underlay TSPO expression and SPIO uptake in symptomatic EAE mice. Conclusions These preliminary results suggest that both activated microglia and infiltrated macrophages are present in vulnerable brain regions during the acute phase of PLP-EAE and contribute to disease severity. Both [18F]DPA-714-PET and SPIO-MRI appear suitable modalities for preclinical study of neuroinflammation in MS mice models.
Collapse
Affiliation(s)
- A R Coda
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy
| | - S Anzilotti
- IRCCS SDN, Via E. Gianturco 113, 80143, Naples, Italy
| | - F Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, University "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - A Greco
- Department of Advanced Biomedical Sciences, University "Federico II", Via S. Pansini 5, 80131, Naples, Italy
- Ceinge Biotecnologie Avanzate s. c. a. r. l., Via G. Salvatore 486, 80145, Naples, Italy
| | - M Panico
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy
| | - S Gargiulo
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy
| | - M Gramanzini
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy
| | - A Zannetti
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy
| | - S Albanese
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy
| | - G Pignataro
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, University "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - L Annunziato
- IRCCS SDN, Via E. Gianturco 113, 80143, Naples, Italy
| | - M Salvatore
- IRCCS SDN, Via E. Gianturco 113, 80143, Naples, Italy
| | - A Brunetti
- Department of Advanced Biomedical Sciences, University "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - P De Berardinis
- Institute of Biochemistry and Cell Biology, National Research Council, Via P. Castellino 111, 80131, Naples, Italy
| | - Mario Quarantelli
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy.
| | - G Palma
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy
| | - Sabina Pappatà
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy.
| |
Collapse
|
11
|
Laquinimod ameliorates secondary brain inflammation. Neurobiol Dis 2019; 134:104675. [PMID: 31731041 DOI: 10.1016/j.nbd.2019.104675] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/21/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence suggests that a degenerative processes within the brain can trigger the formation of new, focal inflammatory lesions in Multiple Sclerosis (MS). Here, we used a novel pre-clinical MS animal model to test whether the amelioration of degenerative brain events reduces the secondary recruitment of peripheral immune cells and, in consequence, inflammatory lesion development. Neural degeneration was induced by a 3 weeks cuprizone intoxication period. To mitigate the cuprizone-induced pathology, animals were treated with Laquinimod (25 mg/kg) during the cuprizone-intoxication period. At the beginning of week 6, encephalitogenic T cell development in peripheral lymphoid organs was induced by the immunization with myelin oligodendrocyte glycoprotein 35-55 peptide (i.e., Cup/EAE). Demyelination, axonal injury and reactive gliosis were determined by immunohistochemistry. Positron emission tomography (PET) imaging was performed to analyze glia activation in vivo. Vehicle-treated cuprizone mice displayed extensive callosal demyelination, glia activation and enhanced TSPO-ligand binding. This cuprizone-induced pathology was profoundly ameliorated in mice treated with Laquinimod. In vehicle-treated Cup/EAE mice, the cuprizone-induced pathology triggered massive peripheral immune cell recruitment into the forebrain, evidenced by multifocal perivascular inflammation, glia activation and neuro-axonal injury. While anti myelin oligodendrocyte glycoprotein 35-55 peptide immune responses were comparable in vehicle- and Laquinimod-treated Cup/EAE mice, the cuprizone-triggered immune cell recruitment was ameliorated by the Laquinimod treatment. This study clearly illustrates that amelioration of a primary brain-intrinsic degenerative process secondary halts peripheral immune cell recruitment and, in consequence, inflammatory lesion development. These findings have important consequences for the interpretation of the results of clinical studies.
Collapse
|
12
|
Nack A, Brendel M, Nedelcu J, Daerr M, Nyamoya S, Beyer C, Focke C, Deussing M, Hoornaert C, Ponsaerts P, Schmitz C, Bartenstein P, Rominger A, Kipp M. Expression of Translocator Protein and [18F]-GE180 Ligand Uptake in Multiple Sclerosis Animal Models. Cells 2019; 8:cells8020094. [PMID: 30696113 PMCID: PMC6406715 DOI: 10.3390/cells8020094] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/16/2019] [Accepted: 01/23/2019] [Indexed: 12/19/2022] Open
Abstract
Positron emission tomography (PET) ligands targeting the translocator protein (TSPO) represent promising tools to visualize neuroinflammation in multiple sclerosis (MS). Although it is known that TSPO is expressed in the outer mitochondria membrane, its cellular localization in the central nervous system under physiological and pathological conditions is not entirely clear. The purpose of this study was to assess the feasibility of utilizing PET imaging with the TSPO tracer, [18F]-GE180, to detect histopathological changes during experimental demyelination, and to determine which cell types express TSPO. C57BL/6 mice were fed with cuprizone for up to 5 weeks to induce demyelination. Groups of mice were investigated by [18F]-GE180 PET imaging at week 5. Recruitment of peripheral immune cells was triggered by combining cuprizone intoxication with MOG35–55 immunization (i.e., Cup/EAE). Immunofluorescence double-labelling and transgene mice were used to determine which cell types express TSPO. [18F]-GE180-PET reliably detected the cuprizone-induced pathology in various white and grey matter regions, including the corpus callosum, cortex, hippocampus, thalamus and caudoputamen. Cuprizone-induced demyelination was paralleled by an increase in TSPO expression, glia activation and axonal injury. Most of the microglia and around one-third of the astrocytes expressed TSPO. TSPO expression induction was more severe in the white matter corpus callosum compared to the grey matter cortex. Although mitochondria accumulate at sites of focal axonal injury, these mitochondria do not express TSPO. In Cup/EAE mice, both microglia and recruited monocytes contribute to the TSPO expressing cell populations. These findings support the notion that TSPO is a valuable marker for the in vivo visualization and quantification of neuropathological changes in the MS brain. The pathological substrate of an increase in TSPO-ligand binding might be diverse including microglia activation, peripheral monocyte recruitment, or astrocytosis, but not axonal injury.
Collapse
MESH Headings
- Animals
- Astrocytes/pathology
- Astrocytes/ultrastructure
- Axons/metabolism
- Axons/ultrastructure
- Biomarkers/metabolism
- Carbazoles/metabolism
- Cuprizone
- Demyelinating Diseases/diagnostic imaging
- Demyelinating Diseases/pathology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Inflammation/pathology
- Ligands
- Mice, Inbred C57BL
- Mitochondria/metabolism
- Mitochondria/ultrastructure
- Monocytes/metabolism
- Multiple Sclerosis/diagnostic imaging
- Neuroglia/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, GABA/genetics
- Receptors, GABA/metabolism
Collapse
Affiliation(s)
- Anne Nack
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
- Department of Anatomy, 39071 Rostock University Medical Center, Rostock, Germany.
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, 80336 Munich, Germany.
| | - Julia Nedelcu
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
- Department of Anatomy, 39071 Rostock University Medical Center, Rostock, Germany.
| | - Markus Daerr
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
- Department of Anatomy, 39071 Rostock University Medical Center, Rostock, Germany.
| | - Stella Nyamoya
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
- Department of Anatomy, 39071 Rostock University Medical Center, Rostock, Germany.
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
| | - Carola Focke
- Department of Nuclear Medicine, University Hospital, LMU Munich, 80336 Munich, Germany.
| | - Maximilian Deussing
- Department of Nuclear Medicine, University Hospital, LMU Munich, 80336 Munich, Germany.
| | - Chloé Hoornaert
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.
| | - Christoph Schmitz
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, 80336 Munich, Germany.
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital, LMU Munich, 80336 Munich, Germany.
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland.
| | - Markus Kipp
- Department of Anatomy, 39071 Rostock University Medical Center, Rostock, Germany.
| |
Collapse
|
13
|
Betlazar C, Harrison-Brown M, Middleton RJ, Banati R, Liu GJ. Cellular Sources and Regional Variations in the Expression of the Neuroinflammatory Marker Translocator Protein (TSPO) in the Normal Brain. Int J Mol Sci 2018; 19:ijms19092707. [PMID: 30208620 PMCID: PMC6163555 DOI: 10.3390/ijms19092707] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 09/09/2018] [Accepted: 09/09/2018] [Indexed: 02/07/2023] Open
Abstract
The inducible expression of the mitochondrial translocator protein 18 kDa (TSPO) by activated microglia is a prominent, regular feature of acute and chronic-progressive brain pathology. This expression is also the rationale for the continual development of new TSPO binding molecules for the diagnosis of "neuroinflammation" by molecular imaging. However, there is in the normal brain an ill-defined, low-level constitutive expression of TSPO. Taking advantage of healthy TSPO knockout mouse brain tissue to validate TSPO antibody specificity, this study uses immunohistochemistry to determine the regional distribution and cellular sources of TSPO in the normal mouse brain. Fluorescence microscopy revealed punctate TSPO immunostaining in vascular endothelial cells throughout the brain. In the olfactory nerve layers and glomeruli of the olfactory bulb, choroid plexus and ependymal layers, we confirm constitutive TSPO expression levels similar to peripheral organs, while some low TSPO expression is present in regions of known neurogenesis, as well as cerebellar Purkinje cells. The distributed-sparse expression of TSPO in endothelial mitochondria throughout the normal brain can be expected to give rise to a low baseline signal in TSPO molecular imaging studies. Finally, our study emphasises the need for valid and methodologically robust verification of the selectivity of TSPO ligands through the use of TSPO knockout tissues.
Collapse
Affiliation(s)
- Calina Betlazar
- Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia.
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW 2050, Australia.
| | - Meredith Harrison-Brown
- Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia.
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW 2050, Australia.
| | - Ryan J Middleton
- Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia.
| | - Richard Banati
- Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia.
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW 2050, Australia.
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia.
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW 2050, Australia.
| |
Collapse
|
14
|
Jiemy WF, Heeringa P, Kamps JA, van der Laken CJ, Slart RH, Brouwer E. Positron emission tomography (PET) and single photon emission computed tomography (SPECT) imaging of macrophages in large vessel vasculitis: Current status and future prospects. Autoimmun Rev 2018; 17:715-726. [PMID: 29729443 DOI: 10.1016/j.autrev.2018.02.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 02/07/2018] [Indexed: 12/21/2022]
|
15
|
Selvaraj S, Bloomfield PS, Cao B, Veronese M, Turkheimer F, Howes OD. Brain TSPO imaging and gray matter volume in schizophrenia patients and in people at ultra high risk of psychosis: An [ 11C]PBR28 study. Schizophr Res 2018; 195:206-214. [PMID: 28893493 PMCID: PMC6027955 DOI: 10.1016/j.schres.2017.08.063] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 08/31/2017] [Accepted: 08/31/2017] [Indexed: 12/12/2022]
Abstract
Patients with schizophrenia show whole brain and cortical gray matter (GM) volume reductions which are progressive early in their illness. Microglia, the resident immune cells in the CNS, phagocytose neurons and synapses. Some post mortem and in vivo studies in schizophrenia show evidence for elevated microglial activation compared to matched controls. However, it is currently unclear how these results relate to changes in cortical structure. METHODS Fourteen patients with schizophrenia and 14 ultra high risk for psychosis (UHR) subjects alongside two groups of age and genotype matched healthy controls received [11C]PBR28 PET scans to index TSPO expression, a marker of microglial activation and a 3T MRI scan. We investigated the relationship between the volume changes of cortical regions and microglial activation in cortical GM (as indexed by [11C]PBR28 distribution volume ratio (DVR). RESULTS The total cortical GM volume was significantly lower in SCZ than the controls [mean (SD)/cm3: SCZ=448.83 (39.2) and controls=499.6 (59.2) (p=0.02) but not in UHR (mean (SD)=503.06 (57.9) and controls=524.46 (45.3) p=0.3). Regression model fitted the total cortical GM DVR values with the cortical regional volumes in SCZ (r=0.81; p<0.001) and in UHR (r=0.63; p=0.02). We found a significant negative correlation between the TSPO signal and total cortical GM volume in SCZ with the highest absolute correlation coefficient in the right superior-parietal cortex (r=-0.72; p=0.006). CONCLUSIONS These findings suggest that microglial activity is related to the altered cortical volume seen in schizophrenia. Longitudinal investigations are required to determine whether microglial activation leads to cortical gray matter loss.
Collapse
Affiliation(s)
- Sudhakar Selvaraj
- Department of Psychiatry and Behavioural Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; Psychiatric Imaging Group, MRC Clinical Sciences Centre, Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK.
| | - Peter S Bloomfield
- Psychiatric Imaging Group, MRC Clinical Sciences Centre, Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Bo Cao
- Department of Psychiatry and Behavioural Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, IoPPN, King's College London, Box PO89, De Crespigny Park, London SE5 8AF, UK
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, IoPPN, King's College London, Box PO89, De Crespigny Park, London SE5 8AF, UK
| | - Oliver D Howes
- Psychiatric Imaging Group, MRC Clinical Sciences Centre, Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| |
Collapse
|
16
|
Belloli S, Zanotti L, Murtaj V, Mazzon C, Di Grigoli G, Monterisi C, Masiello V, Iaccarino L, Cappelli A, Poliani PL, Politi LS, Moresco RM. 18F-VC701-PET and MRI in the in vivo neuroinflammation assessment of a mouse model of multiple sclerosis. J Neuroinflammation 2018; 15:33. [PMID: 29402285 PMCID: PMC5800080 DOI: 10.1186/s12974-017-1044-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 12/15/2017] [Indexed: 11/17/2022] Open
Abstract
Background Positron emission tomography (PET) using translocator protein (TSPO) ligands has been used to detect neuroinflammatory processes in neurological disorders, including multiple sclerosis (MS). The aim of this study was to evaluate neuroinflammation in a mouse MS model (EAE) using TSPO-PET with 18F-VC701, in combination with magnetic resonance imaging (MRI). Methods MOG35-55/CFA and pertussis toxin protocol was used to induce EAE in C57BL/6 mice. Disease progression was monitored daily, whereas MRI evaluation was performed at 1, 2, and 4 weeks post-induction. Microglia activation was assessed in vivo by 18F-VC701 PET at the time of maximum disease score and validated by radioligand ex vivo distribution and immunohistochemistry at 2 and 4 weeks post-immunization. Results In vivo and ex vivo analyses show that 18F-VC701 significantly accumulates within the central nervous system (CNS), particularly in the cortex, striatum, hippocampus, cerebellum, and cervical spinal cord of EAE compared to control mice, at 2 weeks post-immunization. MRI confirmed the presence of focal brain lesions at 2 weeks post-immunization in both T1-weighted and T2 images. Of note, MRI abnormalities attenuated in later post-immunization phase. Neuropathological analysis confirmed the presence of microglial activation in EAE mice, consistent with the in vivo increase of 18F-VC701 uptake. Conclusion Increase of 18F-VC701 uptake in EAE mice is strongly associated with the presence of microglia activation in the acute phase of the disease. The combined use of TSPO-PET and MRI provided complementary evidence on the ongoing disease process, thus representing an attractive new tool to investigate neuronal damage and neuroinflammation at preclinical levels. Electronic supplementary material The online version of this article (10.1186/s12974-017-1044-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sara Belloli
- IBFM-CNR, Segrate, Italy.,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Milan Center for Neuroscience (NeuroMI) University of Milano-Bicocca, Milan, Italy
| | - Lucia Zanotti
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Murtaj
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.,PhD Program in Neuroscience, University of Milan-Bicocca, Monza, Italy.,Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Cristina Mazzon
- Humanitas Clinical and Research Centre, Rozzano, Italy.,Biomedical Sciences Department, University of Padua, Padua, Italy
| | - Giuseppe Di Grigoli
- IBFM-CNR, Segrate, Italy.,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Monterisi
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Valeria Masiello
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Leonardo Iaccarino
- Vita-Salute San Raffaele University and In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Cappelli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Pietro Luigi Poliani
- Department of Molecular and Translational Medicine, Pathology Unit, University of Brescia, Brescia, Italy
| | - Letterio Salvatore Politi
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Advanced MRI Center, University of Massachusetts Medical School, Worcester, MA, USA.,Neuroimaging Research, Boston Children's Hospital, Boston, MA, USA
| | - Rosa Maria Moresco
- IBFM-CNR, Segrate, Italy. .,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy. .,Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy.
| |
Collapse
|
17
|
Mouse models of neurodegenerative disease: preclinical imaging and neurovascular component. Brain Imaging Behav 2017; 12:1160-1196. [PMID: 29075922 DOI: 10.1007/s11682-017-9770-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases represent great challenges for basic science and clinical medicine because of their prevalence, pathologies, lack of mechanism-based treatments, and impacts on individuals. Translational research might contribute to the study of neurodegenerative diseases. The mouse has become a key model for studying disease mechanisms that might recapitulate in part some aspects of the corresponding human diseases. Neurodegenerative disorders are very complicated and multifactorial. This has to be taken in account when testing drugs. Most of the drugs screening in mice are very difficult to be interpretated and often useless. Mouse models could be condiderated a 'pathway models', rather than as models for the whole complicated construct that makes a human disease. Non-invasive in vivo imaging in mice has gained increasing interest in preclinical research in the last years thanks to the availability of high-resolution single-photon emission computed tomography (SPECT), positron emission tomography (PET), high field Magnetic resonance, Optical Imaging scanners and of highly specific contrast agents. Behavioral test are useful tool to characterize different animal models of neurodegenerative pathology. Furthermore, many authors have observed vascular pathological features associated to the different neurodegenerative disorders. Aim of this review is to focus on the different existing animal models of neurodegenerative disorders, describe behavioral tests and preclinical imaging techniques used for diagnose and describe the vascular pathological features associated to these diseases.
Collapse
|
18
|
Vállez García D, Doorduin J, de Paula Faria D, Dierckx RAJO, de Vries EFJ. Effect of Preventive and Curative Fingolimod Treatment Regimens on Microglia Activation and Disease Progression in a Rat Model of Multiple Sclerosis. J Neuroimmune Pharmacol 2017; 12:521-530. [PMID: 28361437 PMCID: PMC5527053 DOI: 10.1007/s11481-017-9741-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/23/2017] [Indexed: 11/29/2022]
Abstract
Fingolimod was the first oral drug approved for multiple sclerosis treatment. Its principal mechanism of action is blocking of lymphocyte trafficking. In addition, recent studies have shown its capability to diminish microglia activation. The effect of preventive and curative fingolimod treatment on the time-course of neuroinflammation was investigated in the experimental autoimmune encephalomyelitis rat model for multiple sclerosis. Neuroinflammatory progression was followed in Dark Agouti female rats after immunization. Positron-Emission tomography (PET) imaging with (R)-[11C]PK11195 was performed on day 11, 15, 19, 27, 29 and 34 during normal disease progression, preventive and curative treatments with fingolimod (1 mg/kg/day). Additionally, bodyweight and clinical symptoms were determined. Preventive treatment diminished bodyweight loss and inhibited the appearance of neurological symptoms. In non-treated rats, PET showed that neuroinflammation peaked in the brainstem at day 19, whereas the imaging signal was decreased in cortical regions. Both preventive and curative treatment reduced neuroinflammation in the brainstem at day 19. Eight days after treatment withdrawal, neuroinflammation had flared-up, especially in cortical regions. Preventive treatment with fingolimod suppressed clinical symptoms and neuroinflammation in the brainstem. After treatment withdrawal, clinical symptoms reappeared together with neuroinflammation in cortical regions, suggesting a different pathway of disease progression.
Collapse
Affiliation(s)
- David Vállez García
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Daniele de Paula Faria
- Department of Radiology and Oncology, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, 9700 RB Groningen, The Netherlands.
| |
Collapse
|
19
|
Poutiainen P, Jaronen M, Quintana FJ, Brownell AL. Precision Medicine in Multiple Sclerosis: Future of PET Imaging of Inflammation and Reactive Astrocytes. Front Mol Neurosci 2016; 9:85. [PMID: 27695400 PMCID: PMC5023680 DOI: 10.3389/fnmol.2016.00085] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 08/30/2016] [Indexed: 12/29/2022] Open
Abstract
Non-invasive molecular imaging techniques can enhance diagnosis to achieve successful treatment, as well as reveal underlying pathogenic mechanisms in disorders such as multiple sclerosis (MS). The cooperation of advanced multimodal imaging techniques and increased knowledge of the MS disease mechanism allows both monitoring of neuronal network and therapeutic outcome as well as the tools to discover novel therapeutic targets. Diverse imaging modalities provide reliable diagnostic and prognostic platforms to better achieve precision medicine. Traditionally, magnetic resonance imaging (MRI) has been considered the golden standard in MS research and diagnosis. However, positron emission tomography (PET) imaging can provide functional information of molecular biology in detail even prior to anatomic changes, allowing close follow up of disease progression and treatment response. The recent findings support three major neuroinflammation components in MS: astrogliosis, cytokine elevation, and significant changes in specific proteins, which offer a great variety of specific targets for imaging purposes. Regardless of the fact that imaging of astrocyte function is still a young field and in need for development of suitable imaging ligands, recent studies have shown that inflammation and astrocyte activation are related to progression of MS. MS is a complex disease, which requires understanding of disease mechanisms for successful treatment. PET is a precise non-invasive imaging method for biochemical functions and has potential to enhance early and accurate diagnosis for precision therapy of MS. In this review we focus on modulation of different receptor systems and inflammatory aspect of MS, especially on activation of glial cells, and summarize the recent findings of PET imaging in MS and present the most potent targets for new biomarkers with the main focus on experimental MS research.
Collapse
Affiliation(s)
- Pekka Poutiainen
- Athinoula A Martinos Biomedical Imaging Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, MA, USA
| | - Merja Jaronen
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical SchoolBoston, MA, USA
| | - Francisco J. Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical SchoolBoston, MA, USA
| | - Anna-Liisa Brownell
- Athinoula A Martinos Biomedical Imaging Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, MA, USA
| |
Collapse
|
20
|
Colasanti A, Guo Q, Giannetti P, Wall MB, Newbould RD, Bishop C, Onega M, Nicholas R, Ciccarelli O, Muraro PA, Malik O, Owen DR, Young AH, Gunn RN, Piccini P, Matthews PM, Rabiner EA. Hippocampal Neuroinflammation, Functional Connectivity, and Depressive Symptoms in Multiple Sclerosis. Biol Psychiatry 2016; 80:62-72. [PMID: 26809249 PMCID: PMC4918731 DOI: 10.1016/j.biopsych.2015.11.022] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 11/04/2015] [Accepted: 11/25/2015] [Indexed: 01/03/2023]
Abstract
BACKGROUND Depression, a condition commonly comorbid with multiple sclerosis (MS), is associated more generally with elevated inflammatory markers and hippocampal pathology. We hypothesized that neuroinflammation in the hippocampus is responsible for depression associated with MS. We characterized the relationship between depressive symptoms and hippocampal microglial activation in patients with MS using the 18-kDa translocator protein radioligand [(18)F]PBR111. To evaluate pathophysiologic mechanisms, we explored the relationships between hippocampal neuroinflammation, depressive symptoms, and hippocampal functional connectivities defined by resting-state functional magnetic resonance imaging. METHODS The Beck Depression Inventory (BDI) was administered to 11 patients with MS and 22 healthy control subjects before scanning with positron emission tomography and functional magnetic resonance imaging. We tested for higher [(18)F]PBR111 uptake in the hippocampus of patients with MS relative to healthy control subjects and examined the correlations between [(18)F]PBR111 uptake, BDI scores, and hippocampal functional connectivities in the patients with MS. RESULTS Patients with MS had an increased hippocampal [(18)F]PBR111 distribution volume ratio relative to healthy control subjects (p = .024), and the hippocampal distribution volume ratio was strongly correlated with the BDI score in patients with MS (r = .86, p = .006). Hippocampal functional connectivities to the subgenual cingulate and prefrontal and parietal regions correlated with BDI scores and [(18)F]PBR111 distribution volume ratio. CONCLUSIONS Our results provide evidence that hippocampal microglial activation in MS impairs the brain functional connectivities in regions contributing to maintenance of a normal affective state. Our results suggest a rationale for the responsiveness of depression in some patients with MS to effective control of brain neuroinflammation. Our findings also lend support to further investigation of the role of inflammatory processes in the pathogenesis of depression more generally.
Collapse
Affiliation(s)
- Alessandro Colasanti
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom; Centre for Affective Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Imanova Centre for Imaging Sciences, London, United Kingdom.
| | - Qi Guo
- Imanova Centre for Imaging Sciences, London, United Kingdom
| | - Paolo Giannetti
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Matthew B Wall
- Imanova Centre for Imaging Sciences, London, United Kingdom
| | | | | | - Mayca Onega
- Imanova Centre for Imaging Sciences, London, United Kingdom
| | - Richard Nicholas
- Imperial College Healthcare National Health Service Trust, London, United Kingdom
| | - Olga Ciccarelli
- Department of Neuroinflammation, University College London Institute of Neurology, London, United Kingdom; National Institute of Health Research Biomedical Research Centre at University College London Hospitals, London, United Kingdom
| | - Paolo A Muraro
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Omar Malik
- Imperial College Healthcare National Health Service Trust, London, United Kingdom
| | - David R Owen
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Allan H Young
- Centre for Affective Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Roger N Gunn
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom; Imanova Centre for Imaging Sciences, London, United Kingdom
| | - Paola Piccini
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Paul M Matthews
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Eugenii A Rabiner
- Psychological Medicine, and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Imanova Centre for Imaging Sciences, London, United Kingdom
| |
Collapse
|
21
|
In Vivo Detection of Age- and Disease-Related Increases in Neuroinflammation by 18F-GE180 TSPO MicroPET Imaging in Wild-Type and Alzheimer's Transgenic Mice. J Neurosci 2016; 35:15716-30. [PMID: 26609163 DOI: 10.1523/jneurosci.0996-15.2015] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Alzheimer's disease (AD) is the most common cause of dementia. Neuroinflammation appears to play an important role in AD pathogenesis. Ligands of the 18 kDa translocator protein (TSPO), a marker for activated microglia, have been used as positron emission tomography (PET) tracers to reflect neuroinflammation in humans and mouse models. Here, we used the novel TSPO-targeted PET tracer (18)F-GE180 (flutriciclamide) to investigate differences in neuroinflammation between young and old WT and APP/PS1dE9 transgenic (Tg) mice. In vivo PET scans revealed an overt age-dependent elevation in whole-brain uptake of (18)F-GE180 in both WT and Tg mice, and a significant increase in whole-brain uptake of (18)F-GE180 (peak-uptake and retention) in old Tg mice compared with young Tg mice and all WT mice. Similarly, the (18)F-GE180 binding potential in hippocampus was highest to lowest in old Tg > old WT > young Tg > young WT mice using MRI coregistration. Ex vivo PET and autoradiography analysis further confirmed our in vivo PET results: enhanced uptake and specific binding (SUV75%) of (18)F-GE180 in hippocampus and cortex was highest in old Tg mice followed by old WT, young Tg, and finally young WT mice. (18)F-GE180 specificity was confirmed by an in vivo cold tracer competition study. We also examined (18)F-GE180 metabolites in 4-month-old WT mice and found that, although total radioactivity declined over 2 h, of the remaining radioactivity, ∼90% was due to parent (18)F-GE180. In conclusion, (18)F-GE180 PET scans may be useful for longitudinal monitoring of neuroinflammation during AD progression and treatment. SIGNIFICANCE STATEMENT Microglial activation, a player in Alzheimer's disease (AD) pathogenesis, is thought to reflect neuroinflammation. Using in vivo microPET imaging with a novel TSPO radioligand, (18)F-GE180, we detected significantly enhanced neuroinflammation during normal aging in WT mice and in response to AD-associated pathology in APP/PS1dE9 Tg mice, an AD mouse model. Increased uptake and specific binding of (18)F-GE180 in whole brain and hippocampus were confirmed by ex vivo PET and autoradiography. The binding specificity and stability of (18)F-GE180 was further confirmed by a cold tracer competition study and a metabolite study, respectively. Therefore, (18)F-GE180 PET imaging may be useful for longitudinal monitoring of neuroinflammation during AD progression and treatment and may also be useful for other neurodegenerative diseases.
Collapse
|
22
|
Martín A, Vázquez-Villoldo N, Gómez-Vallejo V, Padro D, Soria FN, Szczupak B, Plaza-García S, Arrieta A, Reese T, Llop J, Domercq M, Matute C. In vivo imaging of system xc- as a novel approach to monitor multiple sclerosis. Eur J Nucl Med Mol Imaging 2015; 43:1124-38. [PMID: 26659901 DOI: 10.1007/s00259-015-3275-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/27/2015] [Indexed: 01/24/2023]
Abstract
PURPOSE Glutamate excitotoxicity contributes to oligodendroglial and axonal damage in multiple sclerosis pathology. Extracellular glutamate concentration in the brain is controlled by cystine/glutamate antiporter (system xc-), a membrane antiporter that imports cystine and releases glutamate. Despite this, the system xc(-) activity and its connection to the inflammatory reaction in multiple sclerosis (MS) is largely unknown. METHODS Longitudinal in vivo magnetic resonance (MRI) and positron emission tomography (PET) imaging studies with 2-[(18)F]Fluoro-2-deoxy-D-glucose ([(18)F]FDG), [(11)C]-(R)-(1-(2-chlorophenyl)-N-methyl-N-1(1-methylpropyl)-3-isoquinolinecarboxamide ([(11)C]PK11195) and (4S)-4-(3-(18)F-fluoropropyl)-L-glutamate ([(18)F]FSPG) were carried out during the course of experimental autoimmune encephalomyelitis (EAE) induction in rats. RESULTS [(18)F]FSPG showed a significant increase of system xc(-) function in the lumbar section of the spinal cord at 14 days post immunization (dpi) that stands in agreement with the neurological symptoms and ventricle edema formation at this time point. Likewise, [(18)F]FDG did not show significant changes in glucose metabolism throughout central nervous system and [(11)C]PK11195 evidenced a significant increase of microglial/macrophage activation in spinal cord and cerebellum 2 weeks after EAE induction. Therefore, [(18)F]FSPG showed a major capacity to discriminate regions of the central nervous system affected by the MS in comparison to [(18)F]FDG and [(11)C]PK11195. Additionally, clodronate-treated rats showed a depletion in microglial population and [(18)F]FSPG PET signal in spinal cord confirming a link between neuroinflammatory reaction and cystine/glutamate antiporter activity in EAE rats. CONCLUSIONS Altogether, these results suggest that in vivo PET imaging of system xc(-) could become a valuable tool for the diagnosis and treatment evaluation of MS.
Collapse
Affiliation(s)
- Abraham Martín
- Molecular Imaging Unit, CIC biomaGUNE, P° Miramon 182, San Sebastian, Spain.
| | - Nuria Vázquez-Villoldo
- Department of Neurosciences, University of the Basque Country, Barrio Sarriena s/n, 48940, Leioa, Spain.,Achucarro Basque Center for Neuroscience, UPV/EHU, 48170, Zamudio, Spain.,Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 48940, Leioa, Spain
| | - Vanessa Gómez-Vallejo
- Molecular Imaging Unit, CIC biomaGUNE, P° Miramon 182, San Sebastian, Spain.,Radiochemistry and Nuclear Imaging, CIC biomaGUNE, P° Miramon 182, San Sebastian, Spain
| | - Daniel Padro
- Molecular Imaging Unit, CIC biomaGUNE, P° Miramon 182, San Sebastian, Spain.,Magnetic Resonance Imaging, CIC biomaGUNE, P° Miramon 182, San Sebastian, Spain
| | - Federico N Soria
- Department of Neurosciences, University of the Basque Country, Barrio Sarriena s/n, 48940, Leioa, Spain.,Achucarro Basque Center for Neuroscience, UPV/EHU, 48170, Zamudio, Spain.,Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 48940, Leioa, Spain
| | - Boguslaw Szczupak
- Molecular Imaging Unit, CIC biomaGUNE, P° Miramon 182, San Sebastian, Spain
| | - Sandra Plaza-García
- Molecular Imaging Unit, CIC biomaGUNE, P° Miramon 182, San Sebastian, Spain.,Magnetic Resonance Imaging, CIC biomaGUNE, P° Miramon 182, San Sebastian, Spain
| | - Ander Arrieta
- Molecular Imaging Unit, CIC biomaGUNE, P° Miramon 182, San Sebastian, Spain
| | - Torsten Reese
- Molecular Imaging Unit, CIC biomaGUNE, P° Miramon 182, San Sebastian, Spain.,Magnetic Resonance Imaging, CIC biomaGUNE, P° Miramon 182, San Sebastian, Spain
| | - Jordi Llop
- Molecular Imaging Unit, CIC biomaGUNE, P° Miramon 182, San Sebastian, Spain.,Radiochemistry and Nuclear Imaging, CIC biomaGUNE, P° Miramon 182, San Sebastian, Spain
| | - Maria Domercq
- Department of Neurosciences, University of the Basque Country, Barrio Sarriena s/n, 48940, Leioa, Spain.,Achucarro Basque Center for Neuroscience, UPV/EHU, 48170, Zamudio, Spain.,Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 48940, Leioa, Spain
| | - Carlos Matute
- Department of Neurosciences, University of the Basque Country, Barrio Sarriena s/n, 48940, Leioa, Spain. .,Achucarro Basque Center for Neuroscience, UPV/EHU, 48170, Zamudio, Spain. .,Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 48940, Leioa, Spain.
| |
Collapse
|
23
|
Janssen B, Vugts DJ, Funke U, Molenaar GT, Kruijer PS, van Berckel BNM, Lammertsma AA, Windhorst AD. Imaging of neuroinflammation in Alzheimer's disease, multiple sclerosis and stroke: Recent developments in positron emission tomography. Biochim Biophys Acta Mol Basis Dis 2015; 1862:425-41. [PMID: 26643549 DOI: 10.1016/j.bbadis.2015.11.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/09/2015] [Accepted: 11/19/2015] [Indexed: 12/13/2022]
Abstract
Neuroinflammation is thought to play a pivotal role in many diseases affecting the brain, including Alzheimer's disease, multiple sclerosis and stroke. Neuroinflammation is characterised predominantly by microglial activation, which can be visualised using positron emission tomography (PET). Traditionally, translocator protein 18kDa (TSPO) is the target for imaging of neuroinflammation using PET. In this review, recent preclinical and clinical research using PET in Alzheimer's disease, multiple sclerosis and stroke is summarised. In addition, new molecular targets for imaging of neuroinflammation, such as monoamine oxidases, adenosine receptors and cannabinoid receptor type 2, are discussed. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
Affiliation(s)
- Bieneke Janssen
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands.
| | - Danielle J Vugts
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Uta Funke
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; BV Cyclotron VU, Amsterdam, The Netherlands
| | - Ger T Molenaar
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands; BV Cyclotron VU, Amsterdam, The Netherlands
| | | | - Bart N M van Berckel
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
24
|
Gentile A, De Vito F, Fresegna D, Musella A, Buttari F, Bullitta S, Mandolesi G, Centonze D. Exploring the role of microglia in mood disorders associated with experimental multiple sclerosis. Front Cell Neurosci 2015; 9:243. [PMID: 26161070 PMCID: PMC4479791 DOI: 10.3389/fncel.2015.00243] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/15/2015] [Indexed: 01/01/2023] Open
Abstract
Microglia is increasingly recognized to play a crucial role in the pathogenesis of psychiatric diseases. In particular, microglia may be the cellular link between inflammation and behavioral alterations: by releasing a number of soluble factors, among which pro-inflammatory cytokines, that can regulate synaptic activity, thereby leading to perturbation of behavior. In multiple sclerosis (MS), the most common neuroinflammatory disorder affecting young adults, microglia activation and dysfunction may account for mood symptoms, like depression and anxiety, that are often diagnosed in patients even in the absence of motor disability. Behavioral studies in experimental autoimmune encephalomyelitis (EAE), the animal model of MS, have shown that emotional changes occur early in the disease and in correlation to inflammatory mediator and neurotransmitter level alterations. However, such studies lack a full and comprehensive analysis of the role played by microglia in EAE-behavioral syndrome. We review the experimental studies addressing behavioral symptoms in EAE, and propose the study of neuron-glia interaction as a powerful but still poorly explored tool to investigate the burden of microglia in mood alterations associated to MS.
Collapse
Affiliation(s)
- Antonietta Gentile
- Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC) Rome, Italy ; Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata Rome, Italy
| | - Francesca De Vito
- Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC) Rome, Italy ; Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata Rome, Italy
| | - Diego Fresegna
- Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC) Rome, Italy ; Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata Rome, Italy
| | - Alessandra Musella
- Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC) Rome, Italy
| | - Fabio Buttari
- Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata Rome, Italy ; IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed Pozzilli, Italy
| | - Silvia Bullitta
- Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC) Rome, Italy
| | - Georgia Mandolesi
- Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC) Rome, Italy
| | - Diego Centonze
- Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata Rome, Italy ; IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed Pozzilli, Italy
| |
Collapse
|
25
|
Mattner F, Quinlivan M, Greguric I, Pham T, Liu X, Jackson T, Berghofer P, Fookes CJR, Dikic B, Gregoire MC, Dolle F, Katsifis A. Radiosynthesis, In Vivo Biological Evaluation, and Imaging of Brain Lesions with [123I]-CLINME, a New SPECT Tracer for the Translocator Protein. DISEASE MARKERS 2015; 2015:729698. [PMID: 26199457 PMCID: PMC4496498 DOI: 10.1155/2015/729698] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 06/10/2015] [Indexed: 11/17/2022]
Abstract
The high affinity translocator protein (TSPO) ligand 6-chloro-2-(4'-iodophenyl)-3-(N,N-methylethyl)imidazo[1,2-a]pyridine-3-acetamide (CLINME) was radiolabelled with iodine-123 and assessed for its sensitivity for the TSPO in rodents. Moreover neuroinflammatory changes on a unilateral excitotoxic lesion rat model were detected using SPECT imaging. [(123)I]-CLINME was prepared in 70-80% radiochemical yield. The uptake of [(123)I]-CLINME was evaluated in rats by biodistribution, competition, and metabolite studies. The unilateral excitotoxic lesion was performed by injection of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid unilaterally into the striatum. The striatum lesion was confirmed and correlated with TSPO expression in astrocytes and activated microglia by immunohistochemistry and autoradiography. In vivo studies with [(123)I]-CLINME indicated a biodistribution pattern consistent with TPSO distribution and the competition studies with PK11195 and Ro 5-4864 showed that [(123)I]-CLINME is selective for this site. The metabolite study showed that the extractable radioactivity was unchanged [(123)I]-CLINME in organs which expresses TSPO. SPECT/CT imaging on the unilateral excitotoxic lesion indicated that the mean ratio uptake in striatum (lesion:nonlesion) was 2.2. Moreover, TSPO changes observed by SPECT imaging were confirmed by immunofluorescence, immunochemistry, and autoradiography. These results indicated that [(123)I]-CLINME is a promising candidate for the quantification and visualization of TPSO expression in activated astroglia using SPECT.
Collapse
Affiliation(s)
- F. Mattner
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
- Department of Molecular Imaging, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - M. Quinlivan
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - I. Greguric
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - T. Pham
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - X. Liu
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - T. Jackson
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - P. Berghofer
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - C. J. R. Fookes
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - B. Dikic
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - M.-C. Gregoire
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - F. Dolle
- CEA, DSV/I2BM, Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401 Orsay, France
| | - A. Katsifis
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
- Department of Molecular Imaging, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| |
Collapse
|
26
|
Matthews PM, Datta G. Positron-emission tomography molecular imaging of glia and myelin in drug discovery for multiple sclerosis. Expert Opin Drug Discov 2015; 10:557-70. [PMID: 25843125 DOI: 10.1517/17460441.2015.1032240] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Therapies acting on glial cells are being explored for new drug development for multiple sclerosis. Molecular imaging using positron-emission tomography (PET) could address relevant questions in early phase clinical trials. AREAS COVERED In this article, the authors critically review human PET methods that can be applied in specialised centres for imaging activated microglia and astrocytes and myelin. EXPERT OPINION Strengths of PET lie in the molecular selectivity, sensitivity and potential for absolute quantitation. Even now, translocator protein PET radioligands could be used in exploratory studies for interventions targeting brain microglial activation. The clinical and neuropathological meaningfulness of signal from PET radioligands reporting on astrocyte activation through cellular expression of either monoamine oxidase B or the I2-imidazoline receptor or metabolism of [(11)C]acetate can now explored. [(11)C] N-methyl-4,4'-diaminostilbene, a PET marker for myelin, could soon enter first human trials. However, use of any of these PET glial markers demands a well-focused hypothesis and a commitment to validation in the context of use. Enhanced access to these radioligands, standardisation of analyses and lowering the costs of using them are needed if their full promise is to be realised.
Collapse
Affiliation(s)
- Paul M Matthews
- Imperial College London, Division of Brain Sciences, Department of Medicine , E515, Burlington Danes Building, Du Cane Road, W12 0NN London , UK +44 02075942612 ; +44 02075946548 ;
| | | |
Collapse
|
27
|
Banati RB, Middleton RJ, Chan R, Hatty CR, Wai-Ying Kam W, Quin C, Graeber MB, Parmar A, Zahra D, Callaghan P, Fok S, Howell NR, Gregoire M, Szabo A, Pham T, Davis E, Liu GJ. Positron emission tomography and functional characterization of a complete PBR/TSPO knockout. Nat Commun 2014; 5:5452. [PMID: 25406832 PMCID: PMC4263137 DOI: 10.1038/ncomms6452] [Citation(s) in RCA: 189] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 10/01/2014] [Indexed: 12/17/2022] Open
Abstract
The evolutionarily conserved peripheral benzodiazepine receptor (PBR), or 18-kDa translocator protein (TSPO), is thought to be essential for cholesterol transport and steroidogenesis, and thus life. TSPO has been proposed as a biomarker of neuroinflammation and a new drug target in neurological diseases ranging from Alzheimer's disease to anxiety. Here we show that global C57BL/6-Tspo(tm1GuWu(GuwiyangWurra))-knockout mice are viable with normal growth, lifespan, cholesterol transport, blood pregnenolone concentration, protoporphyrin IX metabolism, fertility and behaviour. However, while the activation of microglia after neuronal injury appears to be unimpaired, microglia from (GuwiyangWurra)TSPO knockouts produce significantly less ATP, suggesting reduced metabolic activity. Using the isoquinoline PK11195, the ligand originally used for the pharmacological and structural characterization of the PBR/TSPO, and the imidazopyridines CLINDE and PBR111, we demonstrate the utility of (GuwiyangWurra)TSPO knockouts to provide robust data on drug specificity and selectivity, both in vitro and in vivo, as well as the mechanism of action of putative TSPO-targeting drugs.
Collapse
Affiliation(s)
- Richard B. Banati
- Life Sciences, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
- Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
- Medical Imaging & Radiation Sciences, Faculty of Health Science and Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
- National Imaging Facility, Sydney, Camperdown, New South Wales 2006, Australia
| | - Ryan J. Middleton
- Life Sciences, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
| | - Ronald Chan
- Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
- Medical Imaging & Radiation Sciences, Faculty of Health Science and Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Claire R. Hatty
- Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
- Medical Imaging & Radiation Sciences, Faculty of Health Science and Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Winnie Wai-Ying Kam
- Life Sciences, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
- Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
- Medical Imaging & Radiation Sciences, Faculty of Health Science and Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Candice Quin
- Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
- Medical Imaging & Radiation Sciences, Faculty of Health Science and Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Manuel B. Graeber
- Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
- Medical Imaging & Radiation Sciences, Faculty of Health Science and Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Arvind Parmar
- Life Sciences, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
| | - David Zahra
- Life Sciences, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
| | - Paul Callaghan
- Life Sciences, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
| | - Sandra Fok
- Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Nicholas R. Howell
- Life Sciences, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
| | - Marie Gregoire
- Life Sciences, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
| | - Alexander Szabo
- Life Sciences, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
- Centre for Translational Neuroscience, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Tien Pham
- Life Sciences, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
| | - Emma Davis
- Life Sciences, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
| | - Guo-Jun Liu
- Life Sciences, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
- Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
- Medical Imaging & Radiation Sciences, Faculty of Health Science and Brain & Mind Research Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
28
|
Liu G, Middleton RJ, Hatty CR, Kam WW, Chan R, Pham T, Harrison‐Brown M, Dodson E, Veale K, Banati RB. The 18 kDa translocator protein, microglia and neuroinflammation. Brain Pathol 2014; 24:631-53. [PMID: 25345894 PMCID: PMC8029074 DOI: 10.1111/bpa.12196] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 08/19/2014] [Indexed: 12/17/2022] Open
Abstract
The 18 kDa translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor, is expressed in the injured brain. It has become known as an imaging marker of "neuroinflammation" indicating active disease, and is best interpreted as a nondiagnostic biomarker and disease staging tool that refers to histopathology rather than disease etiology. The therapeutic potential of TSPO as a drug target is mostly based on the understanding that it is an outer mitochondrial membrane protein required for the translocation of cholesterol, which thus regulates the rate of steroid synthesis. This pivotal role together with the evolutionary conservation of TSPO has underpinned the belief that any loss or mutation of TSPO should be associated with significant physiological deficits or be outright incompatible with life. However, against prediction, full Tspo knockout mice are viable and across their lifespan do not show the phenotype expected if cholesterol transport and steroid synthesis were significantly impaired. Thus, the "translocation" function of TSPO remains to be better substantiated. Here, we discuss the literature before and after the introduction of the new nomenclature for TSPO and review some of the newer findings. In light of the controversy surrounding the function of TSPO, we emphasize the continued importance of identifying compounds with confirmed selectivity and suggest that TSPO expression is analyzed within specific disease contexts rather than merely equated with the reified concept of "neuroinflammation."
Collapse
Affiliation(s)
- Guo‐Jun Liu
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Ryan J. Middleton
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
| | - Claire R. Hatty
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Winnie Wai‐Ying Kam
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Ronald Chan
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Tien Pham
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
| | - Meredith Harrison‐Brown
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Eoin Dodson
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
| | - Kelly Veale
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Richard B. Banati
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
- National Imaging Facility and Ramaciotti Brain Imaging CentreSydneyNSWAustralia
| |
Collapse
|
29
|
de Paula Faria D, Vlaming ML, Copray SC, Tielen F, Anthonijsz HJ, Sijbesma JW, Buchpiguel CA, Dierckx RA, van der Hoorn JW, de Vries EF. PET Imaging of Disease Progression and Treatment Effects in the Experimental Autoimmune Encephalomyelitis Rat Model. J Nucl Med 2014; 55:1330-5. [DOI: 10.2967/jnumed.114.137216] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 04/17/2014] [Indexed: 12/13/2022] Open
|
30
|
Colasanti A, Guo Q, Muhlert N, Giannetti P, Onega M, Newbould RD, Ciccarelli O, Rison S, Thomas C, Nicholas R, Muraro PA, Malik O, Owen DR, Piccini P, Gunn RN, Rabiner EA, Matthews PM. In Vivo Assessment of Brain White Matter Inflammation in Multiple Sclerosis with (18)F-PBR111 PET. J Nucl Med 2014; 55:1112-8. [PMID: 24904112 DOI: 10.2967/jnumed.113.135129] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 03/18/2014] [Indexed: 01/02/2023] Open
Abstract
UNLABELLED PET radioligand binding to the 18-kD translocator protein (TSPO) in the brains of patients with multiple sclerosis (MS) primarily reflects activated microglia and macrophages. We previously developed genetic stratification for accurate quantitative estimation of TSPO using second-generation PET radioligands. In this study, we used (18)F-PBR111 PET and MR imaging to measure relative binding in the lesional, perilesional, and surrounding normal-appearing white matter of MS patients, as an index of the innate immune response. METHODS (18)F-PBR111 binding was quantified in 11 MS patients and 11 age-matched healthy volunteers, stratified according to the rs6971 TSPO gene polymorphism. Fluid-attenuated inversion recovery and magnetization transfer ratio (MTR) MR imaging were used to segment the white matter in MS patients as lesions, perilesional volumes, nonlesional white matter with reduced MTR, and nonlesional white matter with normal MTR. RESULTS (18)F-PBR111 binding was higher in the white matter lesions and perilesional volumes of MS patients than in white matter of healthy controls (P < 0.05). Although there was substantial heterogeneity in binding between different lesions, a within-subject analysis showed higher (18)F-PBR111 binding in MS lesions (P < 0.05) and in perilesional (P < 0.05) and nonlesional white matter with reduced MTR (P < 0.005) than in nonlesional white matter with a normal MTR. A positive correlation was observed between the mean (18)F-PBR111 volume of distribution increase in lesions relative to nonlesional white matter with a normal MTR and the MS severity score (Spearman ρ = 0.62, P < 0.05). CONCLUSION This study demonstrates that quantitative TSPO PET with a second-generation radioligand can be used to characterize innate immune responses in MS in vivo and provides further evidence supporting an association between the white matter TSPO PET signal in lesions and disease severity. Our approach is practical for extension to studies of the role of the innate immune response in MS for differentiation of antiinflammatory effects of new medicines and their longer term impact on clinical outcome.
Collapse
Affiliation(s)
- Alessandro Colasanti
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom Imanova Centre for Imaging Sciences, London, United Kingdom
| | - Qi Guo
- Imanova Centre for Imaging Sciences, London, United Kingdom Centre for Neuroimaging Sciences, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Nils Muhlert
- UCL Institute of Neurology, London, United Kingdom School of Psychology and Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, United Kingdom
| | - Paolo Giannetti
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Mayca Onega
- Imanova Centre for Imaging Sciences, London, United Kingdom
| | | | | | - Stuart Rison
- Imperial College Healthcare NHS Trust, London, United Kingdom; and
| | - Charlotte Thomas
- Imperial College Healthcare NHS Trust, London, United Kingdom; and
| | - Richard Nicholas
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom Imperial College Healthcare NHS Trust, London, United Kingdom; and
| | - Paolo A Muraro
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Omar Malik
- Imperial College Healthcare NHS Trust, London, United Kingdom; and
| | - David R Owen
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Paola Piccini
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Roger N Gunn
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom Imanova Centre for Imaging Sciences, London, United Kingdom
| | - Eugenii A Rabiner
- Imanova Centre for Imaging Sciences, London, United Kingdom Centre for Neuroimaging Sciences, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Paul M Matthews
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom Neurosciences, GlaxoSmithKline, Brentford, United Kingdom
| |
Collapse
|
31
|
Faria DDP, Copray S, Buchpiguel C, Dierckx R, de Vries E. PET imaging in multiple sclerosis. J Neuroimmune Pharmacol 2014; 9:468-82. [PMID: 24809810 DOI: 10.1007/s11481-014-9544-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 04/21/2014] [Indexed: 01/03/2023]
Abstract
Positron emission tomography (PET) is a non-invasive technique for quantitative imaging of biochemical and physiological processes in animals and humans. PET uses probes labeled with a radioactive isotope, called PET tracers, which can bind to or be converted by a specific biological target and thus can be applied to detect and monitor different aspects of diseases. The number of applications of PET imaging in multiple sclerosis is still limited. Clinical studies using PET are basically focused on monitoring changes in glucose metabolism and the presence of activated microglia/macrophages in sclerotic lesions. In preclinical studies, PET imaging of targets for other processes, like demyelination and remyelination, has been investigated and may soon be translated to clinical applications. Moreover, more PET tracers that could be relevant for MS are available now, but have not been studied in this context yet. In this review, we summarize the PET imaging studies performed in multiple sclerosis up to now. In addition, we will identify potential applications of PET imaging of processes or targets that are of interest to MS research, but have yet remained largely unexplored.
Collapse
Affiliation(s)
- Daniele de Paula Faria
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
32
|
Dickens AM, Vainio S, Marjamäki P, Johansson J, Lehtiniemi P, Rokka J, Rinne J, Solin O, Haaparanta-Solin M, Jones PA, Trigg W, Anthony DC, Airas L. Detection of microglial activation in an acute model of neuroinflammation using PET and radiotracers 11C-(R)-PK11195 and 18F-GE-180. J Nucl Med 2014; 55:466-72. [PMID: 24516258 DOI: 10.2967/jnumed.113.125625] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED It remains unclear how different translocator protein (TSPO) ligands reflect the spatial extent of astrocyte or microglial activation in various neuroinflammatory conditions. Here, we use a reproducible lipopolysaccharide (LPS)-induced model of acute central nervous system inflammation to compare the binding performance of a new TSPO ligand (18)F-GE-180 with (11)C-(R)-PK11195. Using immunohistochemistry, we also explore the ability of the TSPO ligands to detect activated microglial cells and astrocytes. METHODS Lewis rats (n = 30) were microinjected with LPS (1 or 10 μg) or saline (1 μL) into the left striatum. The animals were imaged in vivo at 16 h after the injection using PET radiotracers (18)F-GE-180 or (11)C-(R)-PK11195 (n = 3 in each group) and were killed afterward for autoradiography of the brain. Immunohistochemical assessment of OX-42 and glial fibrillary acidic protein (GFAP) was performed to identify activated microglial cells and reactive astrocytes. RESULTS In vivo PET imaging revealed an increase in the ipsilateral TSPO binding, compared with binding in the contralateral hemisphere, after the microinjection of 10 μg of LPS. No increase was observed with vehicle. By autoradiography, the TSPO radiotracer binding potential in the injected hemisphere was increased after striatal injection of 1 or 10 μg of LPS. However, the significant increase was observed only when using (18)F-GE-180. The area of CD11b-expressing microglial cells extended beyond that of enhanced GFAP staining and mapped more closely to the extent of (18)F-GE-180 binding than to (11)C-(R)-PK11195 binding. The signal from either PET ligand was significantly increased in regions of increased GFAP immunoreactivity and OX-42 colocalization, meaning that the presence of both activated microglia and astrocytes in a given area leads to increased binding of the TSPO radiotracers. CONCLUSION (18)F-GE-180 is able to reveal sites of activated microglia in both gray and white matter. However, the signal is increased by the presence of activated astrocytes. Therefore, (18)F-GE-180 is a promising new fluorinated longer-half-life tracer that reveals the presence of activated microglia in a manner that is superior to (11)C-(R)-PK11195 due to the higher binding potential observed for this ligand.
Collapse
Affiliation(s)
- Alex M Dickens
- Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Turku, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Neuroinflammation plays a central role in a variety of neurological diseases, including stroke, multiple sclerosis, Alzheimer’s disease, and malignant CNS neoplasms, among many other. Different cell types and molecular mediators participate in a cascade of events in the brain that is ultimately aimed at control, regeneration and repair, but leads to damage of brain tissue under pathological conditions. Non-invasive molecular imaging of key players in the inflammation cascade holds promise for identification and quantification of the disease process before it is too late for effective therapeutic intervention. In this review, we focus on molecular imaging techniques that target inflammatory cells and molecules that are of interest in neuroinflammation, especially those with high translational potential. Over the past decade, a plethora of molecular imaging agents have been developed and tested in animal models of (neuro)inflammation, and a few have been translated from bench to bedside. The most promising imaging techniques to visualize neuroinflammation include MRI, positron emission tomography (PET), single photon emission computed tomography (SPECT), and optical imaging methods. These techniques enable us to image adhesion molecules to visualize endothelial cell activation, assess leukocyte functions such as oxidative stress, granule release, and phagocytosis, and label a variety of inflammatory cells for cell tracking experiments. In addition, several cell types and their activation can be specifically targeted in vivo, and consequences of neuroinflammation such as neuronal death and demyelination can be quantified. As we continue to make progress in utilizing molecular imaging technology to study and understand neuroinflammation, increasing efforts and investment should be made to bring more of these novel imaging agents from the “bench to bedside.”
Collapse
Affiliation(s)
- Benjamin Pulli
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| | - John W Chen
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| |
Collapse
|