1
|
Aceves-Serrano L, Neva JL, Munro J, Vavasour IM, Parent M, Boyd LA, Doudet DJ. Evaluation of microglia activation related markers following a clinical course of TBS: A non-human primate study. PLoS One 2024; 19:e0301118. [PMID: 38753646 PMCID: PMC11098425 DOI: 10.1371/journal.pone.0301118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 03/11/2024] [Indexed: 05/18/2024] Open
Abstract
While the applicability and popularity of theta burst stimulation (TBS) paradigms remain, current knowledge of their neurobiological effects is still limited, especially with respect to their impact on glial cells and neuroinflammatory processes. We used a multimodal imaging approach to assess the effects of a clinical course of TBS on markers for microglia activation and tissue injury as an indirect assessment of neuroinflammatory processes. Healthy non-human primates received continuous TBS (cTBS), intermittent TBS (iTBS), or sham stimulation over the motor cortex at 90% of resting motor threshold. Stimulation was delivered to the awake subjects 5 times a week for 3-4 weeks. Translocator protein (TSPO) expression was evaluated using Positron Emission Tomography and [11C]PBR28, and myo-inositol (mI) and N-acetyl-aspartate (NAA) concentrations were assessed with Magnetic Resonance Spectroscopy. Animals were then euthanized, and immunofluorescence staining was performed using antibodies against TSPO. Paired t-tests showed no significant changes in [11C]PBR28 measurements after stimulation. Similarly, no significant changes in mI and NAA concentrations were found. Post-mortem TSPO evaluation showed comparable mean immunofluorescence intensity after active TBS and sham delivery. The current study suggests that in healthy brains a clinical course of TBS, as evaluated with in-vivo imaging techniques (PET and MRS), did not measurably modulate the expression of glia related markers and metabolite associated with neural viability.
Collapse
Affiliation(s)
- Lucero Aceves-Serrano
- Department of Medicine, Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jason L. Neva
- Faculté de Médecine, École de Kinésiologie et des Sciences de l’activité Physique, Université de Montréal, Montreal, Quebec, Canada
- Centre de Recherche de l’institut Universitaire de Gériatrie de Montréal, Montreal, QC, Canada
| | - Jonathan Munro
- CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Irene M. Vavasour
- Faculty of Medicine, UBC MRI Research Center, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin Parent
- CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Lara A. Boyd
- Faculty of Medicine, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada
- Faculty of Medicine, Graduate Program of Rehabilitation Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Doris J. Doudet
- Department of Medicine, Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
2
|
Guerrin CG, Prasad K, Vazquez-Matias DA, Zheng J, Franquesa-Mullerat M, Barazzuol L, Doorduin J, de Vries EF. Prenatal infection and adolescent social adversity affect microglia, synaptic density, and behavior in male rats. Neurobiol Stress 2023; 27:100580. [PMID: 37920548 PMCID: PMC10618826 DOI: 10.1016/j.ynstr.2023.100580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023] Open
Abstract
Maternal infection during pregnancy and childhood social trauma have been associated with neurodevelopmental and affective disorders, such as schizophrenia, autism spectrum disorders, bipolar disorder and depression. These disorders are characterized by changes in microglial cells, which play a notable role in synaptic pruning, and synaptic deficits. Here, we investigated the effect of prenatal infection and social adversity during adolescence - either alone or in combination - on behavior, microglia, and synaptic density. Male offspring of pregnant rats injected with poly I:C, mimicking prenatal infection, were exposed to repeated social defeat during adolescence. We found that maternal infection during pregnancy prevented the reduction in social behavior and increase in anxiety induced by social adversity during adolescence. Furthermore, maternal infection and social adversity, alone or in combination, induced hyperlocomotion in adulthood. Longitudinal in vivo imaging with [11C]PBR28 positron emission tomography revealed that prenatal infection alone and social adversity during adolescence alone induced a transient increase in translocator protein TSPO density, an indicator of glial reactivity, whereas their combination induced a long-lasting increase that remained until adulthood. Furthermore, only the combination of prenatal infection and social adversity during adolescence induced an increase in microglial cell density in the frontal cortex. Prenatal infection increased proinflammatory cytokine IL-1β protein levels in hippocampus and social adversity reduced anti-inflammatory cytokine IL-10 protein levels in hippocampus during adulthood. This reduction in IL-10 was prevented if rats were previously exposed to prenatal infection. Adult offspring exposed to prenatal infection or adolescent social adversity had a higher synaptic density in the frontal cortex, but not hippocampus, as evaluated by synaptophysin density. Interestingly, such an increase in synaptic density was not observed in rats exposed to the combination of prenatal infection and social adversity, perhaps due to the long-lasting increase in microglial density, which may lead to an increase in microglial synaptic pruning. These findings suggest that changes in microglia activity and cytokine release induced by prenatal infection and social adversity during adolescence may be related to a reduced synaptic pruning, resulting in a higher synaptic density and behavioral changes in adulthood.
Collapse
Affiliation(s)
- Cyprien G.J. Guerrin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Kavya Prasad
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Daniel A. Vazquez-Matias
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Jing Zheng
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Maria Franquesa-Mullerat
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Lara Barazzuol
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Erik F.J. de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| |
Collapse
|
3
|
Tuwar MN, Chen WH, Chiwaya AM, Yeh HL, Nguyen MH, Bai CH. Brain-Derived Neurotrophic Factor (BDNF) and Translocator Protein (TSPO) as Diagnostic Biomarkers for Acute Ischemic Stroke. Diagnostics (Basel) 2023; 13:2298. [PMID: 37443691 DOI: 10.3390/diagnostics13132298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/11/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) interacts with tropomyosin-related kinase B (TrkB) to promote neuronal growth, survival, differentiation, neurotransmitter release, and synaptic plasticity. The translocator protein (TSPO) is known to be found in arterial plaques, which are a symptom of atherosclerosis and a contributory cause of ischemic stroke. This study aims to determine the diagnostic accuracy of plasma BDNF and TSPO levels in discriminating new-onset acute ischemic stroke (AIS) patients from individuals without acute ischemic stroke. A total of 90 AIS patients (61% male, with a mean age of 67.7 ± 12.88) were recruited consecutively in a stroke unit, and each patient was paired with two age- and gender-matched controls. The sensitivity, specificity, and area of the curve between high plasma BDNF and TSPO and having AIS was determined using receiver operating characteristic curves. Furthermore, compared to the controls, AIS patients exhibited significantly higher levels of BDNF and TSPO, blood pressure, HbA1c, and white blood cells, as well as higher creatinine levels. The plasma levels of BDNF and TSPO can significantly discriminate AIS patients from healthy individuals (AUC 0.76 and 0.89, respectively). However, combining the two biomarkers provided little improvement in AUC (0.90). It may be possible to use elevated levels of TSPO as a diagnostic biomarker in patients with acute ischemic stroke upon admission.
Collapse
Affiliation(s)
- Mayuri N Tuwar
- School of Public Health, College of Public Health, Taipei Medical University, Taipei 106236, Taiwan
| | - Wei-Hung Chen
- Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111045, Taiwan
| | - Arthur M Chiwaya
- CLIME Group, Department of Biomedical Sciences, Division of Molecular Biology and Human Genetics, FMHS, Stellenbosch University, Francie Van Zijl Drive, Tygerberg, Cape Town 7505, South Africa
| | - Hsu-Ling Yeh
- Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111045, Taiwan
| | - Minh H Nguyen
- School of Preventive Medicine and Public Health, Hanoi Medical University, Hanoi 100000, Vietnam
| | - Chyi-Huey Bai
- School of Public Health, College of Public Health, Taipei Medical University, Taipei 106236, Taiwan
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 106236, Taiwan
| |
Collapse
|
4
|
Nwaubani P, Cercignani M, Colasanti A. In vivo quantitative imaging of hippocampal inflammation in autoimmune neuroinflammatory conditions: a systematic review. Clin Exp Immunol 2022; 210:24-38. [PMID: 35802780 PMCID: PMC9585553 DOI: 10.1093/cei/uxac058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 05/17/2022] [Accepted: 07/04/2022] [Indexed: 01/25/2023] Open
Abstract
The hippocampus is a morphologically complex region of the brain limbic system centrally involved in important cognitive, affective, and behavioural regulatory roles. It has exquisite vulnerability to neuroinflammatory processes, with some of its subregions found to be specific sites of neuroinflammatory pathology in ex-vivo studies. Optimizing neuroimaging correlates of hippocampal neuroinflammation would enable the direct study of functional consequences of hippocampal neuroinflammatory pathology, as well as the definition of therapeutic end-points for treatments targeting neuroinflammation, and their related affective or cognitive sequelae. However, in vivo traditional imaging of the hippocampus and its subregions is fraught with difficulties, due to methodological challenges deriving from its unique anatomical characteristics. The main objective of this review is to provide a current update on the characterization of quantitative neuroimaging correlates of hippocampal neuroinflammation by focusing on three prototypical autoimmune neuro-inflammatory conditions [multiple sclerosis (MS), systemic lupus erythematosus (SLE), and autoimmune encephalitis (AE)]. We focused on studies employing TSPO-targeting positron emission tomography (PET), quantitative magnetic resonance imaging (MRI), and spectroscopy techniques assumed to be sensitive to neuroinflammatory tissue changes. We found 18 eligible studies (14, 2, and 2 studies in MS, AE, and SLE, respectively). Across conditions, the largest effect was seen in TSPO PET and diffusion-weighted MRI studies. No study examined neuroinflammation-related changes at the hippocampal subfield level. Overall, results were largely inconsistent due to heterogeneous imaging methods, small sample sizes, and different population studies. We discuss how these data could inform future study design and conclude by suggesting further methodological directions aimed at improving the precision and sensitivity of neuroimaging techniques to characterize hippocampal neuroinflammatory pathology in the human brain.
Collapse
Affiliation(s)
- P Nwaubani
- Department of Clinical Neuroscience and Neuroimaging, Brighton and Sussex Medical School, University of Sussex, Falmer, Brighton, UK
| | - M Cercignani
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, UK
| | - A Colasanti
- Correspondence: Alessandro Colasanti, Department of Clinical Neuroscience and Neuroimaging, Brighton and Sussex Medical School, University of Sussex, Trafford Centre for Medical Research, University of Sussex, Falmer, Brighton, BN1 4RY, UK.
| |
Collapse
|
5
|
Badiuk SR, Thiessen JD, Maleki Vareki S, Foster PJ, Chen JZ, Wong E. Glial activation positron emission tomography imaging in radiation treatment of breast cancer brain metastases. Phys Imaging Radiat Oncol 2022; 21:115-122. [PMID: 35359488 PMCID: PMC8961463 DOI: 10.1016/j.phro.2022.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 02/18/2022] [Accepted: 02/20/2022] [Indexed: 11/20/2022] Open
Abstract
Brain metastases affect more breast cancer patients than ever before due to increased overall patient survival with improved molecularly targeted treatments. Approximately 25–34% of breast cancer patients develop brain metastases in their lifetime. Due to the blood–brain barrier (BBB), the standard treatment for breast cancer brain metastases (BCBM) is surgery, stereotactic radiosurgery (SRS) and/or whole brain radiation therapy (WBRT). At the cost of cognitive side effects, WBRT has proven efficacy in treating brain metastases when used with local therapies such as SRS and surgery. This review investigated the potential use of glial activation positron emission tomography (PET) imaging for radiation treatment of BCBM. In order to put these studies into context, we provided background on current radiation treatment approaches for BCBM, our current understanding of the brain microenvironment, its interaction with the peripheral immune system, and alterations in the brain microenvironment by BCBM and radiation. We summarized preclinical literature on the interactions between glial activation and cognition and clinical studies using translocator protein (TSPO) PET to image glial activation in the context of neurological diseases. TSPO-PET is not employed clinically in assessing and guiding cancer therapies. However, it has gained traction in preclinical studies where glial activation was investigated from primary brain cancer, metastases and radiation treatments. Novel glial activation PET imaging and its applications in preclinical studies using breast cancer models and glial immunohistochemistry are highlighted. Lastly, we discuss the potential clinical application of glial activation imaging to improve the therapeutic ratio of radiation treatments for BCBM.
Collapse
Affiliation(s)
- Sawyer Rhae Badiuk
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Jonathan D Thiessen
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 3K7, Canada
- Department of Medical Imaging, University of Western Ontario, London, ON N6A 3K7, Canada
- Imaging Program, Lawson Health Research Institute, London, ON N6A 5W9, Canada
| | - Saman Maleki Vareki
- Cancer Research Laboratory Program, Lawson Health Research Institute, London, ON N6A5 W9, Canada
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON N6A 3K7, Canada
- Department of Oncology, Division of Experimental Oncology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Paula J Foster
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 3K7, Canada
- Imaging Research Laboratories, Robarts Research Institute, London, ON N6A 5B7, Canada
| | - Jeff Z Chen
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 3K7, Canada
- London Regional Cancer Program, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Eugene Wong
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 3K7, Canada
- Department of Physics and Astronomy, University of Western Ontario, London, ON N6A 3K7, Canada
| |
Collapse
|
6
|
Delage C, Vignal N, Guerin C, Taib T, Barboteau C, Mamma C, Khacef K, Margaill I, Sarda-Mantel L, Rizzo-Padoin N, Hontonnou F, Marchand-Leroux C, Lerouet D, Hosten B, Besson V. From positron emission tomography to cell analysis of the 18-kDa Translocator Protein in mild traumatic brain injury. Sci Rep 2021; 11:24009. [PMID: 34907268 PMCID: PMC8671393 DOI: 10.1038/s41598-021-03416-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 11/17/2021] [Indexed: 11/24/2022] Open
Abstract
Traumatic brain injury (TBI) leads to a deleterious neuroinflammation, originating from microglial activation. Monitoring microglial activation is an indispensable step to develop therapeutic strategies for TBI. In this study, we evaluated the use of the 18-kDa translocator protein (TSPO) in positron emission tomography (PET) and cellular analysis to monitor microglial activation in a mild TBI mouse model. TBI was induced on male Swiss mice. PET imaging analysis with [18F]FEPPA, a TSPO radiotracer, was performed at 1, 3 and 7 days post-TBI and flow cytometry analysis on brain at 1 and 3 days post-TBI. PET analysis showed no difference in TSPO expression between non-operated, sham-operated and TBI mice. Flow cytometry analysis demonstrated an increase in TSPO expression in ipsilateral brain 3 days post-TBI, especially in microglia, macrophages, lymphocytes and neutrophils. Moreover, microglia represent only 58.3% of TSPO+ cells in the brain. Our results raise the question of the use of TSPO radiotracer to monitor microglial activation after TBI. More broadly, flow cytometry results point the lack of specificity of TSPO for microglia and imply that microglia contribute to the overall increase in TSPO in the brain after TBI, but is not its only contributor.
Collapse
Affiliation(s)
- Clément Delage
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France.
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France.
| | - Nicolas Vignal
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Médecine Nucléaire, Hôpital Lariboisière, Paris, France
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
| | - Coralie Guerin
- Université de Paris, Innovative Therapies in Haemostasis, Inserm, 75006, Paris, France
- Institut Curie, Cytometry Core, 75005, Paris, France
- Université de Paris, Inserm UMS 3612 CNRS - US25 Inserm -Faculté de Pharmacie de Paris, Paris, France
| | - Toufik Taib
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
| | - Clément Barboteau
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Célia Mamma
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
| | - Kahina Khacef
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
| | - Isabelle Margaill
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1140, Paris, France
| | - Laure Sarda-Mantel
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Médecine Nucléaire, Hôpital Lariboisière, Paris, France
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
| | - Nathalie Rizzo-Padoin
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
- CHU de Martinique, Service Pharmacie, Hôpital Pierre Zobda-Quitman, Fort-de-France, France
| | - Fortune Hontonnou
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
- Université de Paris, Inserm UMR-S 942, Hôpital Lariboisière, Paris, France
| | - Catherine Marchand-Leroux
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Dominique Lerouet
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Benoit Hosten
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service Pharmacie, Hôpital Saint-Louis, Paris, France
| | - Valérie Besson
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
| |
Collapse
|
7
|
Veronese M, Tuosto M, Marques TR, Howes O, Pascual B, Yu M, Masdeu JC, Turkheimer F, Bertoldo A, Zanotti-Fregonara P. Parametric Mapping for TSPO PET Imaging with Spectral Analysis Impulsive Response Function. Mol Imaging Biol 2021; 23:560-571. [PMID: 33475944 PMCID: PMC8277653 DOI: 10.1007/s11307-020-01575-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/27/2020] [Accepted: 12/21/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim of this study was to investigate the use of spectral analysis (SA) for voxel-wise analysis of TSPO PET imaging studies. TSPO PET quantification is methodologically complicated by the heterogeneity of TSPO expression and its cell-dependent modulation during neuroinflammatory response. Compartmental models to account for this complexity exist, but they are unreliable at the high noise typical of voxel data. On the contrary, SA is noise-robust for parametric mapping and provides useful information about tracer kinetics with a free compartmental structure. PROCEDURES SA impulse response function (IRF) calculated at 90 min after tracer injection was used as main parameter of interest in 3 independent PET imaging studies to investigate its sensitivity to (1) a TSPO genetic polymorphism (rs6971) known to affect tracer binding in a cross-sectional analysis of healthy controls scanned with [11C]PBR28 PET; (2) TSPO density with [11C]PBR28 in a competitive blocking study with a TSPO blocker, XBD173; and (3) the higher affinity of a second radiotracer for TSPO, by using data from a head-to-head comparison between [11C]PBR28 and [11C]ER176 scans. RESULTS SA-IRF produced parametric maps of visually good quality. These were sensitive to TSPO genotype (mean relative difference between high- and mixed-affinity binders = 25 %) and TSPO availability (mean signal displacement after 90 mg oral administration of XBD173 = 39 %). Regional averages of voxel-wise IRF estimates were strongly associated with regional total distribution volume (VT) estimated with a 2-tissue compartmental model with vascular compartment (Pearson's r = 0.86 ± 0.11) but less strongly with standard 2TCM-VT (Pearson's r = 0.76 ± 0.32). Finally, SA-IRF estimates for [11C]ER176 were significantly higher than [11C]PBR28 ones, consistent with the higher amount of specific binding of the former tracer. CONCLUSIONS SA-IRF can be used for voxel-wise quantification of TSPO PET data because it generates high-quality parametric maps, it is sensitive to TSPO availability and genotype, and it accounts for the complexity of TSPO tracer kinetics with no additional assumptions.
Collapse
Affiliation(s)
- Mattia Veronese
- Department of Neuroimaging, IoPPN, King's College London, London, UK.
| | - Marcello Tuosto
- Department of Information Engineering, Padova University, Padova, Italy
| | - Tiago Reis Marques
- Department of Psychosis Studies, IoPPN, King's College London, London, UK
| | - Oliver Howes
- Department of Psychosis Studies, IoPPN, King's College London, London, UK
- MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Belen Pascual
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Meixiang Yu
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | | | - Alessandra Bertoldo
- Department of Information Engineering, Padova University, Padova, Italy
- Padova Neuroscience Centre, Padova University, Padova, Italy
| | - Paolo Zanotti-Fregonara
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| |
Collapse
|
8
|
Fisher AS, Lanigan MT, Upton N, Lione LA. Preclinical Neuropathic Pain Assessment; the Importance of Translatability and Bidirectional Research. Front Pharmacol 2021; 11:614990. [PMID: 33628181 PMCID: PMC7897667 DOI: 10.3389/fphar.2020.614990] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/10/2020] [Indexed: 02/04/2023] Open
Abstract
For patients suffering with chronic neuropathic pain the need for suitable novel therapies is imperative. Over recent years a contributing factor for the lack of development of new analgesics for neuropathic pain has been the mismatch of primary neuropathic pain assessment endpoints in preclinical vs. clinical trials. Despite continuous forward translation failures across diverse mechanisms, reflexive quantitative sensory testing remains the primary assessment endpoint for neuropathic pain and analgesia in animals. Restricting preclinical evaluation of pain and analgesia to exclusively reflexive outcomes is over simplified and can be argued not clinically relevant due to the continued lack of forward translation and failures in the clinic. The key to developing new analgesic treatments for neuropathic pain therefore lies in the development of clinically relevant endpoints that can translate preclinical animal results to human clinical trials. In this review we discuss this mismatch of primary neuropathic pain assessment endpoints, together with clinical and preclinical evidence that supports how bidirectional research is helping to validate new clinically relevant neuropathic pain assessment endpoints. Ethological behavioral endpoints such as burrowing and facial grimacing and objective measures such as electroencephalography provide improved translatability potential together with currently used quantitative sensory testing endpoints. By tailoring objective and subjective measures of neuropathic pain the translatability of new medicines for patients suffering with neuropathic pain will hopefully be improved.
Collapse
Affiliation(s)
- Amy S. Fisher
- Transpharmation Ltd., The London Bioscience Innovation Centre, London, United Kingdom
| | - Michael T. Lanigan
- Transpharmation Ltd., The London Bioscience Innovation Centre, London, United Kingdom
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Neil Upton
- Transpharmation Ltd., The London Bioscience Innovation Centre, London, United Kingdom
| | - Lisa A. Lione
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| |
Collapse
|
9
|
Tonon MC, Vaudry H, Chuquet J, Guillebaud F, Fan J, Masmoudi-Kouki O, Vaudry D, Lanfray D, Morin F, Prevot V, Papadopoulos V, Troadec JD, Leprince J. Endozepines and their receptors: Structure, functions and pathophysiological significance. Pharmacol Ther 2020; 208:107386. [DOI: 10.1016/j.pharmthera.2019.06.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023]
|
10
|
Mota F, Ordonez AA, Firth G, Ruiz-Bedoya CA, Ma MT, Jain SK. Radiotracer Development for Bacterial Imaging. J Med Chem 2020; 63:1964-1977. [PMID: 32048838 DOI: 10.1021/acs.jmedchem.9b01623] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Bacterial infections remain a major threat to humanity and are a leading cause of death and disability. Antimicrobial resistance has been declared as one of the top ten threats to human health by the World Health Organization, and new technologies are urgently needed for the early diagnosis and monitoring of deep-seated and complicated infections in hospitalized patients. This review summarizes the radiotracers as applied to imaging of bacterial infections. We summarize the recent progress in the development of pathogen-specific imaging and the application of radiotracers in understanding drug pharmacokinetics as well as the local biology at the infection sites. We also highlight the opportunities for medicinal chemists in radiotracer development for bacterial infections, with an emphasis on target selection and radiosynthetic approaches. Imaging of infections is an emerging field. Beyond clinical applications, these technologies could provide unique insights into disease pathogenesis and expedite bench-to-bedside translation of new therapeutics.
Collapse
Affiliation(s)
- Filipa Mota
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - George Firth
- School of Biomedical Engineering and Imaging Sciences, St. Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Camilo A Ruiz-Bedoya
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Michelle T Ma
- School of Biomedical Engineering and Imaging Sciences, St. Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| |
Collapse
|
11
|
Lacapere JJ, Duma L, Finet S, Kassiou M, Papadopoulos V. Insight into the Structural Features of TSPO: Implications for Drug Development. Trends Pharmacol Sci 2020; 41:110-122. [PMID: 31864680 PMCID: PMC7021566 DOI: 10.1016/j.tips.2019.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/13/2019] [Accepted: 11/18/2019] [Indexed: 01/01/2023]
Abstract
The translocator protein (TSPO), an 18-kDa transmembrane protein primarily found in the outer mitochondrial membrane, is evolutionarily conserved and widely distributed across species. In mammals, TSPO has been described as a key member of a multiprotein complex involved in many putative functions and, over the years, several classes of ligand have been developed to modulate these functions. In this review, we consider the currently available atomic structures of mouse and bacterial TSPO and propose a rationale for the development of new ligands for the protein. We provide a review of TSPO monomeric and oligomeric states and their conformational flexibility, together with ligand-binding site and interaction mechanisms. These data are expected to help considerably the development of high-affinity ligands for TSPO-based therapies or diagnostics.
Collapse
Affiliation(s)
- Jean-Jacques Lacapere
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules (LBM), 4 Place Jussieu, F-75005 Paris, France.
| | - Luminita Duma
- CNRS Enzyme and Cell Engineering Laboratory, Sorbonne Université, Université de Technologie de Compiègne, 60203 Compiègne Cedex, France
| | - Stephanie Finet
- IMPMC, UMR 7590 CNRS Sorbonne Université, 4 Place Jussieu, F-75005 Paris, France
| | - Michael Kassiou
- School of Chemistry, Faculty of Science, The University of Sydney, F11, Eastern Ave, Sydney, NSW 2006, Australia
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
12
|
Fan J, Campioli E, Sottas C, Zirkin B, Papadopoulos V. Amhr2-Cre-Mediated Global Tspo Knockout. J Endocr Soc 2020; 4:bvaa001. [PMID: 32099945 PMCID: PMC7031085 DOI: 10.1210/jendso/bvaa001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 01/09/2020] [Indexed: 12/27/2022] Open
Abstract
Although the role of translocator protein (TSPO) in cholesterol transport in steroid-synthesizing cells has been studied extensively, recent studies of TSPO genetic depletion have questioned its role. Amhr2-Cre mice have been used to generate Leydig cell-specific Tspo conditional knockout (cKO) mice. Using the same Cre line, we were unable to generate Tspo cKO mice possibly because of genetic linkage between Tspo and Amhr2 and coexpression of Amhr2-Cre and Tspo in early embryonic development. We found that Amhr2-Cre is expressed during preimplantation stages, resulting in global heterozygous mice (gHE; Amhr2-Cre+/-,Tspo -/+). Two gHE mice were crossed, generating Amhr2-Cre-mediated Tspo global knockout (gKO; Tspo -/-) mice. We found that 33.3% of blastocysts at E3.5 to E4.5 showed normal morphology, whereas 66.7% showed delayed development, which correlates with the expected Mendelian proportions of Tspo +/+ (25%), Tspo -/- (25%), and Tspo +/- (50%) genotypes from crossing 2 Tspo -/+ mice. Adult Tspo gKO mice exhibited disturbances in neutral lipid homeostasis and reduced intratesticular and circulating testosterone levels, but no change in circulating basal corticosterone levels. RNA-sequencing data from mouse adrenal glands and lungs revealed transcriptome changes in response to the loss of TSPO, including changes in several cholesterol-binding and transfer proteins. This study demonstrates that Amhr2-Cre can be used to produce Tspo gKO mice instead of cKO, and can serve as a new global "Cre deleter." Moreover, our results show that Tspo deletion causes delayed preimplantation embryonic development, alters neutral lipid storage and steroidogenesis, and leads to transcriptome changes that may reflect compensatory mechanisms in response to the loss of function of TSPO.
Collapse
Affiliation(s)
- Jinjiang Fan
- The Research Institute of the McGill University Health Centre
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Enrico Campioli
- The Research Institute of the McGill University Health Centre
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Chantal Sottas
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California, US
| | - Barry Zirkin
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, US
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Centre
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California, US
| |
Collapse
|
13
|
Tiepolt S, Patt M, Aghakhanyan G, Meyer PM, Hesse S, Barthel H, Sabri O. Current radiotracers to image neurodegenerative diseases. EJNMMI Radiopharm Chem 2019; 4:17. [PMID: 31659510 PMCID: PMC6660543 DOI: 10.1186/s41181-019-0070-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/16/2019] [Indexed: 12/11/2022] Open
Abstract
The term of neurodegenerative diseases covers a heterogeneous group of disorders that are distinguished by progressive degeneration of the structure and function of the nervous system such as dementias, movement disorders, motor neuron disorders, as well as some prion disorders. In recent years, a paradigm shift started for the diagnosis of neurodegenerative diseases, for which successively clinical testing is supplemented by biomarker information. In research scenarios, it was even proposed recently to substitute the current syndromic by a biological definition of Alzheimer's diseases. PET examinations with various radiotracers play an important role in providing non-invasive biomarkers and co-morbidity information in neurodegeneration. Information on co-morbidity, e.g. Aβ plaques and Lewy-bodies or Aβ plaques in patients with aphasia or the absence of Aβ plaques in clinical AD patients are of interest to expand our knowledge about the pathogenesis of different phenotypically defined neurodegenerative diseases. Moreover, this information is also important in therapeutic trials targeting histopathological abnormalities.The aim of this review is to present an overview of the currently available radiotracers for imaging neurodegenerative diseases in research and in routine clinical settings. In this context, we also provide a short summary of the most frequent neurodegenerative diseases from a nuclear medicine point of view, their clinical and pathophysiological as well as nuclear imaging characteristics, and the resulting need for new radiotracers.
Collapse
Affiliation(s)
- Solveig Tiepolt
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Marianne Patt
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Gayane Aghakhanyan
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Philipp M. Meyer
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Swen Hesse
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| |
Collapse
|
14
|
VanElzakker MB, Brumfield SA, Lara Mejia PS. Neuroinflammation and Cytokines in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): A Critical Review of Research Methods. Front Neurol 2019; 9:1033. [PMID: 30687207 PMCID: PMC6335565 DOI: 10.3389/fneur.2018.01033] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 11/16/2018] [Indexed: 01/18/2023] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is the label given to a syndrome that can include long-term flu-like symptoms, profound fatigue, trouble concentrating, and autonomic problems, all of which worsen after exertion. It is unclear how many individuals with this diagnosis are suffering from the same condition or have the same underlying pathophysiology, and the discovery of biomarkers would be clarifying. The name "myalgic encephalomyelitis" essentially means "muscle pain related to central nervous system inflammation" and many efforts to find diagnostic biomarkers have focused on one or more aspects of neuroinflammation, from periphery to brain. As the field uncovers the relationship between the symptoms of this condition and neuroinflammation, attention must be paid to the biological mechanisms of neuroinflammation and issues with its potential measurement. The current review focuses on three methods used to study putative neuroinflammation in ME/CFS: (1) positron emission tomography (PET) neuroimaging using translocator protein (TSPO) binding radioligand (2) magnetic resonance spectroscopy (MRS) neuroimaging and (3) assays of cytokines circulating in blood and cerebrospinal fluid. PET scanning using TSPO-binding radioligand is a promising option for studies of neuroinflammation. However, methodological difficulties that exist both in this particular technique and across the ME/CFS neuroimaging literature must be addressed for any results to be interpretable. We argue that the vast majority of ME/CFS neuroimaging has failed to use optimal techniques for studying brainstem, despite its probable centrality to any neuroinflammatory causes or autonomic effects. MRS is discussed as a less informative but more widely available, less invasive, and less expensive option for imaging neuroinflammation, and existing studies using MRS neuroimaging are reviewed. Studies seeking to find a peripheral circulating cytokine "profile" for ME/CFS are reviewed, with attention paid to the biological and methodological reasons for lack of replication among these studies. We argue that both the biological mechanisms of cytokines and the innumerable sources of potential variance in their measurement make it unlikely that a consistent and replicable diagnostic cytokine profile will ever be discovered.
Collapse
Affiliation(s)
- Michael B. VanElzakker
- Division of Neurotherapeutics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | | | | |
Collapse
|
15
|
Rajkovic O, Potjewyd G, Pinteaux E. Regenerative Medicine Therapies for Targeting Neuroinflammation After Stroke. Front Neurol 2018; 9:734. [PMID: 30233484 PMCID: PMC6129611 DOI: 10.3389/fneur.2018.00734] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/13/2018] [Indexed: 12/15/2022] Open
Abstract
Inflammation is a major pathological event following ischemic stroke that contributes to secondary brain tissue damage leading to poor functional recovery. Following the initial ischemic insult, post-stroke inflammatory damage is driven by initiation of a central and peripheral innate immune response and disruption of the blood-brain barrier (BBB), both of which are triggered by the release of pro-inflammatory cytokines and infiltration of circulating immune cells. Stroke therapies are limited to early cerebral blood flow reperfusion, and whilst current strategies aim at targeting neurodegeneration and/or neuroinflammation, innovative research in the field of regenerative medicine aims at developing effective treatments that target both the acute and chronic phase of inflammation. Anti-inflammatory regenerative strategies include the use of nanoparticles and hydrogels, proposed as therapeutic agents and as a delivery vehicle for encapsulated therapeutic biological factors, anti-inflammatory drugs, stem cells, and gene therapies. Biomaterial strategies-through nanoparticles and hydrogels-enable the administration of treatments that can more effectively cross the BBB when injected systemically, can be injected directly into the brain, and can be 3D-bioprinted to create bespoke implants within the site of ischemic injury. In this review, these emerging regenerative and anti-inflammatory approaches will be discussed in relation to ischemic stroke, with a perspective on the future of stroke therapies.
Collapse
Affiliation(s)
- Olivera Rajkovic
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Geoffrey Potjewyd
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
16
|
Schain M, Zanderigo F, Ogden RT, Kreisl WC. Non-invasive estimation of [11C]PBR28 binding potential. Neuroimage 2018; 169:278-285. [DOI: 10.1016/j.neuroimage.2017.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 12/01/2017] [Indexed: 01/14/2023] Open
|
17
|
Papadopoulos V, Fan J, Zirkin B. Translocator protein (18 kDa): an update on its function in steroidogenesis. J Neuroendocrinol 2018; 30:10.1111/jne.12500. [PMID: 28667781 PMCID: PMC5748373 DOI: 10.1111/jne.12500] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 06/26/2017] [Accepted: 06/27/2017] [Indexed: 12/15/2022]
Abstract
Translocator protein (18 kDa) (TSPO) is a ubiquitous mitochondrial protein. Studies of its responses to drug and endogenous ligands have shown TSPO to be involved either directly or indirectly in numerous biological functions, including mitochondrial cholesterol transport and steroid hormone biosynthesis, porphyrin transport and heme synthesis, apoptosis, cell proliferation, and anion transport. Localised to the outer mitochondrial membrane of steroidogenic cells, TSPO has been shown to associate with cytosolic and mitochondrial proteins as part of a large multiprotein complex involved in mitochondrial cholesterol transport, the rate-limiting step in steroidogenesis. There is general agreement as to the structure and pharmacology of TSPO. Stimulation of TSPO has been shown to have therapeutic use as anxiolytics by inducing allopregnanolone production in the brain, and also potentially for re-establishing androgen levels in hypogonadal ageing animals. Until recently, there has been general agreement regarding the role of TSPO in steroidogenesis. However, recent studies involving genetic depletion of TSPO in mice have created controversy about the role of this protein in steroid and heme synthesis. We review the data on the structure and function of TSPO, as well as the recent results obtained using various genetic animal models. Taken together, these studies suggest that TSPO is a unique mitochondrial pharmacological target for diseases that involve increased mitochondrial activity, including steroidogenesis. Although there is no known mammalian species that lacks TSPO, it is likely that, because of the importance of this ancient protein in evolution and mitochondrial function, redundant mechanisms may exist to replace it under circumstances when it is removed.
Collapse
Affiliation(s)
- Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089
- Research Institute of the McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, H4A 3J1, Canada
| | - Jinjiang Fan
- Research Institute of the McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, H4A 3J1, Canada
| | - Barry Zirkin
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| |
Collapse
|
18
|
Zanotti-Fregonara P, Pascual B, Rizzo G, Yu M, Pal N, Beers D, Carter R, Appel SH, Atassi N, Masdeu JC. Head-to-Head Comparison of 11C-PBR28 and 18F-GE180 for Quantification of the Translocator Protein in the Human Brain. J Nucl Med 2018; 59:1260-1266. [DOI: 10.2967/jnumed.117.203109] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/06/2017] [Indexed: 01/29/2023] Open
|
19
|
Uptake of 18F-FET and 18F-FCH in Human Glioblastoma T98G Cell Line after Irradiation with Photons or Carbon Ions. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:6491674. [PMID: 29097931 PMCID: PMC5612615 DOI: 10.1155/2017/6491674] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/10/2016] [Accepted: 12/26/2016] [Indexed: 12/02/2022]
Abstract
The differential diagnosis between recurrence of gliomas or brain metastases and this phenomenon is important in order to choose the best therapy and predict the prognosis but is still a big problem for physicians. The new emerging MRI, CT, and PET diagnostic modalities still lack sufficient accuracy. Radiolabeled choline and amino acids have been reported to show great tumor specificity. We studied the uptake kinetics of [18F]fluoromethyl-choline (FCH) and O-(2-[18F]fluoroethyl)-L-tyrosine (FET) by the T98G human glioblastoma cells from 20 to 120 min after irradiation either with photons at 2-10-20 Gy or with carbon ions at 2 Gy (at the National Centre for Oncological Hadrontherapy (CNAO), Pavia, Italy). We also evaluated the cell death and morphology changes induced by radiation treatment. Both FET and FCH are able to trace tumor behavior in terms of higher uptake for increased doses of radiation treatment, due to the upregulation of cells attempts to repair nonlethal damage. Our data suggest that both FCH and FET could be useful to analyze the metabolic pathways of glioblastoma cells before and after radiotherapy. Physicians will have to consider the different kinetics pathways of uptake concerning the two radiopharmaceuticals.
Collapse
|
20
|
Neuroimaging Techniques to Assess Inflammation in Multiple Sclerosis. Neuroscience 2017; 403:4-16. [PMID: 28764938 DOI: 10.1016/j.neuroscience.2017.07.055] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/21/2017] [Accepted: 07/21/2017] [Indexed: 01/07/2023]
Abstract
Multiple Sclerosis (MS) is a chronic neurological disease that represents a leading cause of disability in young adults and is characterized by inflammation and degeneration of both white matter (WM) and gray matter (GM). Defining the presence or absence of inflammation on individual basis is a key point in choosing the therapy and monitoring the treatment response. Magnetic resonance imaging (MRI) represents the most sensitive non-invasive tool to monitor inflammation in the clinical practice. Indeed, in the early phase of inflammation MRI detects new lesions as extrusion of gadolinium contrast agents across the altered blood-brain-barrier (BBB). The occurrence of MRI lesions is used to confirm diagnosis and has been validated as surrogate marker of relapse to monitor response to treatments. However, focal gadolinium-enhancing lesions represent only an aspect of neuroinflammation. Recent studies have suggested the presence of a widespread inflammation of the central nervous system (CNS), which is mainly related to microglial cells activation occurring both at the edge of chronic focal lesions and throughout the normal-appearing brain tissue. New imaging techniques have been developed to study diffuse inflammation taking place outside the focal plaques. The scope of this review is to examine the various neuroimaging techniques and those biophysical quantities that can be non-invasively detected to enlighten the different aspects of neuroinflammation. Some techniques are commonly used in the clinical practice, while others are used in the research field to better understand the pathophysiological mechanisms of the disease and the role of inflammation.
Collapse
|
21
|
Schain M, Kreisl WC. Neuroinflammation in Neurodegenerative Disorders—a Review. Curr Neurol Neurosci Rep 2017; 17:25. [DOI: 10.1007/s11910-017-0733-2] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
22
|
Albrecht DS, Granziera C, Hooker JM, Loggia ML. In Vivo Imaging of Human Neuroinflammation. ACS Chem Neurosci 2016; 7:470-83. [PMID: 26985861 DOI: 10.1021/acschemneuro.6b00056] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation is implicated in the pathophysiology of a growing number of human disorders, including multiple sclerosis, chronic pain, traumatic brain injury, and amyotrophic lateral sclerosis. As a result, interest in the development of novel methods to investigate neuroinflammatory processes, for the purpose of diagnosis, development of new therapies, and treatment monitoring, has surged over the past 15 years. Neuroimaging offers a wide array of non- or minimally invasive techniques to characterize neuroinflammatory processes. The intent of this Review is to provide brief descriptions of currently available neuroimaging methods to image neuroinflammation in the human central nervous system (CNS) in vivo. Specifically, because of the relatively widespread accessibility of equipment for nuclear imaging (positron emission tomography [PET]; single photon emission computed tomography [SPECT]) and magnetic resonance imaging (MRI), we will focus on strategies utilizing these technologies. We first provide a working definition of "neuroinflammation" and then discuss available neuroimaging methods to study human neuroinflammatory processes. Specifically, we will focus on neuroimaging methods that target (1) the activation of CNS immunocompetent cells (e.g. imaging of glial activation with TSPO tracer [(11)C]PBR28), (2) compromised BBB (e.g. identification of MS lesions with gadolinium-enhanced MRI), (3) CNS-infiltration of circulating immune cells (e.g. tracking monocyte infiltration into brain parenchyma with iron oxide nanoparticles and MRI), and (4) pathological consequences of neuroinflammation (e.g. imaging apoptosis with [(99m)Tc]Annexin V or iron accumulation with T2* relaxometry). This Review provides an overview of state-of-the-art techniques for imaging human neuroinflammation which have potential to impact patient care in the foreseeable future.
Collapse
Affiliation(s)
| | - Cristina Granziera
- Neuro-Immunology,
Neurology Division, Department of Clinical Neurosciences, Centre Hospitalier
Universitaire Vaudois and University of Lausanne, CH-1011 Lausanne, Switzerland
- LTS5, Ecole
Polytechnique
Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | | | | |
Collapse
|