1
|
Neumann KD, Broshek DK, Newman BT, Druzgal TJ, Kundu BK, Resch JE. Concussion: Beyond the Cascade. Cells 2023; 12:2128. [PMID: 37681861 PMCID: PMC10487087 DOI: 10.3390/cells12172128] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
Sport concussion affects millions of athletes each year at all levels of sport. Increasing evidence demonstrates clinical and physiological recovery are becoming more divergent definitions, as evidenced by several studies examining blood-based biomarkers of inflammation and imaging studies of the central nervous system (CNS). Recent studies have shown elevated microglial activation in the CNS in active and retired American football players, as well as in active collegiate athletes who were diagnosed with a concussion and returned to sport. These data are supportive of discordance in clinical symptomology and the inflammatory response in the CNS upon symptom resolution. In this review, we will summarize recent advances in the understanding of the inflammatory response associated with sport concussion and broader mild traumatic brain injury, as well as provide an outlook for important research questions to better align clinical and physiological recovery.
Collapse
Affiliation(s)
- Kiel D. Neumann
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Donna K. Broshek
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA 22903, USA;
| | - Benjamin T. Newman
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (B.T.N.); (T.J.D.); (B.K.K.)
| | - T. Jason Druzgal
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (B.T.N.); (T.J.D.); (B.K.K.)
| | - Bijoy K. Kundu
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (B.T.N.); (T.J.D.); (B.K.K.)
| | - Jacob E. Resch
- Department of Kinesiology, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
2
|
St-Pierre MK, VanderZwaag J, Loewen S, Tremblay MÈ. All roads lead to heterogeneity: The complex involvement of astrocytes and microglia in the pathogenesis of Alzheimer’s disease. Front Cell Neurosci 2022; 16:932572. [PMID: 36035256 PMCID: PMC9413962 DOI: 10.3389/fncel.2022.932572] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/11/2022] [Indexed: 01/04/2023] Open
Abstract
In recent years, glial cells have been acknowledged as key players in the pathogenesis of Alzheimer’s disease (AD), a neurodegenerative condition in which an accumulation of intracellular neurofibrillary tangles and extracellular fibrillar amyloid beta is notably observed in the central nervous system. Genome-wide association studies have shown, both in microglia and astrocytes, an increase in gene variants associated with a higher risk of developing late-onset AD. Microglia, the resident innate immune cells of the brain, and astrocytes, glial cells crucial for vascular integrity and neuronal support, both agglomerate near amyloid beta plaques and dystrophic neurites where they participate in the elimination of these harmful parenchymal elements. However, their role in AD pathogenesis has been challenging to resolve due to the highly heterogeneous nature of these cell populations, i.e., their molecular, morphological, and ultrastructural diversity, together with their ever-changing responsiveness and functions throughout the pathological course of AD. With the recent expansions in the field of glial heterogeneity through innovative advances in state-of-the-art microscopy and -omics techniques, novel concepts and questions arose, notably pertaining to how the diverse microglial and astrocytic states interact with each other and with the AD hallmarks, and how their concerted efforts/actions impact the progression of the disease. In this review, we discuss the recent advances and findings on the topic of glial heterogeneity, particularly focusing on the relationships of these cells with AD hallmarks (e.g., amyloid beta plaques, neurofibrillary tangles, synaptic loss, and dystrophic neurites) in murine models of AD pathology and post-mortem brain samples of patients with AD.
Collapse
Affiliation(s)
- Marie-Kim St-Pierre
- Département de Médecine Moléculaire, Université Laval, Quebec City, QC, Canada
- Axe Neurosciences, Center de Recherche du CHU de Québec, Université Laval, Quebec City, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Jared VanderZwaag
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
| | - Sophia Loewen
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | - Marie-Ève Tremblay
- Département de Médecine Moléculaire, Université Laval, Quebec City, QC, Canada
- Axe Neurosciences, Center de Recherche du CHU de Québec, Université Laval, Quebec City, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- *Correspondence: Marie-Ève Tremblay,
| |
Collapse
|
3
|
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
|
4
|
Chauveau F, Becker G, Boutin H. Have (R)-[ 11C]PK11195 challengers fulfilled the promise? A scoping review of clinical TSPO PET studies. Eur J Nucl Med Mol Imaging 2021; 49:201-220. [PMID: 34387719 PMCID: PMC8712292 DOI: 10.1007/s00259-021-05425-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE The prototypical TSPO radiotracer (R)-[11C]PK11195 has been used in humans for more than thirty years to visualize neuroinflammation in several pathologies. Alternative radiotracers have been developed to improve signal-to-noise ratio and started to be tested clinically in 2008. Here we examined the scientific value of these "(R)-[11C]PK11195 challengers" in clinical research to determine if they could supersede (R)-[11C]PK11195. METHODS A systematic MEDLINE (PubMed) search was performed (up to end of year 2020) to extract publications reporting TSPO PET in patients with identified pathologies, excluding studies in healthy subjects and methodological studies. RESULTS Of the 288 publications selected, 152 used 13 challengers, and 142 used (R)-[11C]PK11195. Over the last 20 years, the number of (R)-[11C]PK11195 studies remained stable (6 ± 3 per year), but was surpassed by the total number of challenger studies for the last 6 years. In total, 3914 patients underwent a TSPO PET scan, and 47% (1851 patients) received (R)-[11C]PK11195. The 2 main challengers were [11C]PBR28 (24%-938 patients) and [18F]FEPPA (11%-429 patients). Only one-in-ten patients (11%-447) underwent 2 TSPO scans, among whom 40 (1%) were scanned with 2 different TSPO radiotracers. CONCLUSIONS Generally, challengers confirmed disease-specific initial (R)-[11C]PK11195 findings. However, while their better signal-to-noise ratio seems particularly useful in diseases with moderate and widespread neuroinflammation, most challengers present an allelic-dependent (Ala147Thr polymorphism) TSPO binding and genetic stratification is hindering their clinical implementation. As new challengers, insensitive to TSPO human polymorphism, are about to enter clinical evaluation, we propose this systematic review to be regularly updated (living review).
Collapse
Affiliation(s)
- Fabien Chauveau
- University of Lyon, Lyon Neuroscience Research Center (CRNL), CNRS UMR5292, INSERM U1028, University Lyon 1, Lyon, France.
| | - Guillaume Becker
- GIGA - CRC In Vivo Imaging, University Liege, Liege, Belgium
- University of Lyon, CarMeN Laboratory, INSERM U1060, University Lyon 1, Hospices Civils Lyon, Lyon, France
| | - Hervé Boutin
- Faculty of Biology Medicine and Health, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
| |
Collapse
|
5
|
Positron emission tomography in multiple sclerosis - straight to the target. Nat Rev Neurol 2021; 17:663-675. [PMID: 34545219 DOI: 10.1038/s41582-021-00537-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 02/08/2023]
Abstract
Following the impressive progress in the treatment of relapsing-remitting multiple sclerosis (MS), the major challenge ahead is the development of treatments to prevent or delay the irreversible accumulation of clinical disability in progressive forms of the disease. The substrate of clinical progression is neuro-axonal degeneration, and a deep understanding of the mechanisms that underlie this process is a precondition for the development of therapies for progressive MS. PET imaging involves the use of radiolabelled compounds that bind to specific cellular and metabolic targets, thereby enabling direct in vivo measurement of several pathological processes. This approach can provide key insights into the clinical relevance of these processes and their chronological sequence during the disease course. In this Review, we focus on the contribution that PET is making to our understanding of extraneuronal and intraneuronal mechanisms that are involved in the pathogenesis of irreversible neuro-axonal damage in MS. We consider the major challenges with the use of PET in MS and the steps necessary to realize clinical benefits of the technique. In addition, we discuss the potential of emerging PET tracers and future applications of existing compounds to facilitate the identification of effective neuroprotective treatments for patients with MS.
Collapse
|
6
|
Correale J, Halfon MJ, Jack D, Rubstein A, Villa A. Acting centrally or peripherally: A renewed interest in the central nervous system penetration of disease-modifying drugs in multiple sclerosis. Mult Scler Relat Disord 2021; 56:103264. [PMID: 34547609 DOI: 10.1016/j.msard.2021.103264] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 09/03/2021] [Accepted: 09/12/2021] [Indexed: 10/20/2022]
Abstract
With the recent approval of cladribine tablets, siponimod and ozanimod, there has been a renewed interest into the extent to which these current generation disease-modifying therapies (DMTs) are able to cross into the central nervous system (CNS), and how this penetration of the blood-brain barrier (BBB) may influence their ability to treat multiple sclerosis (MS). The integrity of the CNS is maintained by the BBB, blood-cerebrospinal fluid barrier, and the arachnoid barrier, which all play an important role in preserving the immunological environment and homeostasis within the CNS. The integrity of the BBB decreases during the course of MS, with a putative temporal relationship to disease worsening. Furthermore, it is currently considered that progression of the disease is mediated mainly by resident cells of the CNS. The existing literature provides evidence to show that some of the current generation DMTs for MS are able to penetrate the CNS and potentially exert direct effects on CNS-resident cells, in particular the CNS-penetrating prodrugs cladribine and fingolimod, and other sphingosine-1 phosphate receptor modulators; siponimod and ozanimod. Other current generation DMTs appear to be restricted to the periphery due to their high molecular weight or physicochemical properties. As more effective brain penetrant therapies are developed for the treatment of MS, there is a need to understand whether the potential for direct effects within the CNS are of significance, and whether this brings additional benefits over and above treatment effects mediated in the periphery. In turn, this will require an improved understanding of the structure and function of the BBB, the role it plays in MS and subsequent treatments. This narrative review summarizes the data supporting the biological plausibility of a potential benefit from therapeutic molecules entering the CNS, and discusses the potential significance in the current and future treatment of MS.
Collapse
Affiliation(s)
- Jorge Correale
- Department of Neurology, Fleni, Buenos Aires, Argentina.
| | | | - Dominic Jack
- Merck Serono Ltd, Feltham, United Kingdom (an affiliate of Merck KGaA)
| | - Adrián Rubstein
- Merck S.A., Buenos Aires, Argentina (an affiliate of Merck KGaA)
| | - Andrés Villa
- Hospital Ramos Mejía, Universidad de Buenos Aires, Argentina
| |
Collapse
|
7
|
Amor S, Nutma E, Marzin M, Puentes F. Imaging immunological processes from blood to brain in amyotrophic lateral sclerosis. Clin Exp Immunol 2021; 206:301-313. [PMID: 34510431 PMCID: PMC8561688 DOI: 10.1111/cei.13660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/18/2021] [Accepted: 08/29/2021] [Indexed: 12/12/2022] Open
Abstract
Neuropathology studies of amyotrophic lateral sclerosis (ALS) and animal models of ALS reveal a strong association between aberrant protein accumulation and motor neurone damage, as well as activated microglia and astrocytes. While the role of neuroinflammation in the pathology of ALS is unclear, imaging studies of the central nervous system (CNS) support the idea that innate immune activation occurs early in disease in both humans and rodent models of ALS. In addition, emerging studies also reveal changes in monocytes, macrophages and lymphocytes in peripheral blood as well as at the neuromuscular junction. To more clearly understand the association of neuroinflammation (innate and adaptive) with disease progression, the use of biomarkers and imaging modalities allow monitoring of immune parameters in the disease process. Such approaches are important for patient stratification, selection and inclusion in clinical trials, as well as to provide readouts of response to therapy. Here, we discuss the different imaging modalities, e.g. magnetic resonance imaging, magnetic resonance spectroscopy and positron emission tomography as well as other approaches, including biomarkers of inflammation in ALS, that aid the understanding of the underlying immune mechanisms associated with motor neurone degeneration in ALS.
Collapse
Affiliation(s)
- Sandra Amor
- Department of Pathology, Amsterdam UMC Location VUmc, Amsterdam, the Netherlands.,Department of Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Erik Nutma
- Department of Pathology, Amsterdam UMC Location VUmc, Amsterdam, the Netherlands
| | - Manuel Marzin
- Department of Pathology, Amsterdam UMC Location VUmc, Amsterdam, the Netherlands
| | - Fabiola Puentes
- Department of Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
8
|
Knudsen TB, Fitzpatrick SC, De Abrew KN, Birnbaum LS, Chappelle A, Daston GP, Dolinoy DC, Elder A, Euling S, Faustman EM, Fedinick KP, Franzosa JA, Haggard DE, Haws L, Kleinstreuer NC, Buck Louis GM, Mendrick DL, Rudel R, Saili KS, Schug TT, Tanguay RL, Turley AE, Wetmore BA, White KW, Zurlinden TJ. FutureTox IV Workshop Summary: Predictive Toxicology for Healthy Children. Toxicol Sci 2021; 180:198-211. [PMID: 33555348 PMCID: PMC8041457 DOI: 10.1093/toxsci/kfab013] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
FutureTox IV, a Society of Toxicology Contemporary Concepts in Toxicology workshop, was held in November 2018. Building upon FutureTox I, II, and III, this conference focused on the latest science and technology for in vitro profiling and in silico modeling as it relates to predictive developmental and reproductive toxicity (DART). Publicly available high-throughput screening data sets are now available for broad in vitro profiling of bioactivities across large inventories of chemicals. Coupling this vast amount of mechanistic data with a deeper understanding of molecular embryology and post-natal development lays the groundwork for using new approach methodologies (NAMs) to evaluate chemical toxicity, drug efficacy, and safety assessment for embryo-fetal development. NAM is a term recently adopted in reference to any technology, methodology, approach, or combination thereof that can be used to provide information on chemical hazard and risk assessment to avoid the use of intact animals (U.S. Environmental Protection Agency [EPA], Strategic plan to promote the development and implementation of alternative test methods within the tsca program, 2018, https://www.epa.gov/sites/production/files/2018-06/documents/epa_alt_strat_plan_6-20-18_clean_final.pdf). There are challenges to implementing NAMs to evaluate chemicals for developmental toxicity compared with adult toxicity. This forum article reviews the 2018 workshop activities, highlighting challenges and opportunities for applying NAMs for adverse pregnancy outcomes (eg, preterm labor, malformations, low birth weight) as well as disorders manifesting postnatally (eg, neurodevelopmental impairment, breast cancer, cardiovascular disease, fertility). DART is an important concern for different regulatory statutes and test guidelines. Leveraging advancements in such approaches and the accompanying efficiencies to detecting potential hazards to human development are the unifying concepts toward implementing NAMs in DART testing. Although use of NAMs for higher level regulatory decision making is still on the horizon, the conference highlighted novel testing platforms and computational models that cover multiple levels of biological organization, with the unique temporal dynamics of embryonic development, and novel approaches for estimating toxicokinetic parameters essential in supporting in vitro to in vivo extrapolation.
Collapse
Affiliation(s)
- Thomas B Knudsen
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| | | | | | - Linda S Birnbaum
- National Institute of Environmental Health Science, NIH, Research Triangle Park, North Carolina, USA
| | - Anne Chappelle
- Chappelle Toxicology Consulting, LLC, Chadds Ford, Pennsylvania, USA
| | | | | | - Alison Elder
- University of Rochester, Rochester, New York, USA
| | - Susan Euling
- U.S. Environmental Protection Agency, Office of Children’s Health Protection, Washington, District of Columbia, USA
| | | | | | - Jill A Franzosa
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| | - Derik E Haggard
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
- Oak Ridge Institute for Science and Education (ORISE);, Texas, USA
| | | | | | | | - Donna L Mendrick
- U.S. Food and Drug Administration, NCTR, Silver Spring, Maryland, USA
| | | | - Katerine S Saili
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| | - Thaddeus T Schug
- National Institute of Environmental Health Science, NIH, Research Triangle Park, North Carolina, USA
| | | | | | - Barbara A Wetmore
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| | - Kimberly W White
- American Chemistry Council, Washington, District of Columbia, USA
| | - Todd J Zurlinden
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| |
Collapse
|
9
|
Modo M. 19F Magnetic Resonance Imaging and Spectroscopy in Neuroscience. Neuroscience 2021; 474:37-50. [PMID: 33766776 DOI: 10.1016/j.neuroscience.2021.03.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/23/2022]
Abstract
1H magnetic resonance imaging (MRI) has established itself as a key diagnostic technique, affording the visualization of brain anatomy, blood flow, activity and connectivity. The detection of other atoms (e.g. 19F, 23Na, 31P), so called hetero-nuclear MRI and spectroscopy (MRS), provides investigative avenues that complement and extend the richness of information that can be gained from 1H MRI. Especially 19F MRI is increasingly emerging as a multi-nuclear (1H/19F) technique that can be exploited to visualize cell migration and trafficking. The lack of a 19F background signal in the brain affords an unequivocal detection suitable for quantification. Fluorine-based contrast material can be engineered as nanoemulsions, nanocapsules, or nanoparticles to label cells in vitro or in vivo. Fluorinated blood substitutes, typically nanoemulsions, can also carry oxygen and serve as a theranostic in poorly perfused brain regions. Brain tissue concentrations of fluorinated pharmaceuticals, including inhalation anesthetics (e.g. isoflurane) and anti-depressants (e.g. fluoxetine), can also be measured using MRS. However, the low signal from these compounds provides a challenge for imaging. Further methodological advances that accelerate signal acquisition (e.g. compressed sensing, cryogenic coils) are required to expand the applications of 19F MR imaging to, for instance, determine the regional pharmacokinetics of novel fluorine-based drugs. Improvements in 19F signal detection and localization, combined with the development of novel sensitive probes, will increase the utility of these multi-nuclear studies. These advances will provide new insights into cellular and molecular processes involved in neurodegenerative disease, as well as the mode of action of pharmaceutical compounds.
Collapse
Affiliation(s)
- Michel Modo
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Rathitharan G, Truong J, Tong J, McCluskey T, Meyer JH, Mizrahi R, Warsh J, Rusjan P, Kennedy JL, Houle S, Kish SJ, Boileau I. Microglia imaging in methamphetamine use disorder: a positron emission tomography study with the 18 kDa translocator protein radioligand [F-18]FEPPA. Addict Biol 2021; 26:e12876. [PMID: 32017280 DOI: 10.1111/adb.12876] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/08/2020] [Accepted: 01/12/2020] [Indexed: 01/19/2023]
Abstract
Activation of brain microglial cells, microgliosis, has been linked to methamphetamine (MA)-seeking behavior, suggesting that microglia could be a new therapeutic target for MA use disorder. Animal data show marked brain microglial activation following acute high-dose MA, but microglial status in human MA users is uncertain, with one positron emission tomography (PET) investigation reporting massively and globally increased translocator protein 18 kDa (TSPO; [C-11](R)-PK11195) binding, a biomarker for microgliosis, in MA users. Our aim was to measure binding of a second-generation TSPO radioligand, [F-18]FEPPA, in brain of human chronic MA users. Regional total volume of distribution (VT ) of [F-18]FEPPA was estimated with a two-tissue compartment model with arterial plasma input function for 10 regions of interest in 11 actively using MA users and 26 controls. A RM-ANOVA corrected for TSPO rs6971 polymorphism was employed to test significance. There was no main effect of group on [F-18]FEPPA VT (P = .81). No significant correlations between [F-18]FEPPA VT and MA use duration, weekly dosage, blood MA concentrations, regional brain volumes, and self-reported craving were observed. Our preliminary findings, consistent with our earlier postmortem data, do not suggest substantial brain microgliosis in MA use disorder but do not rule out microglia as a therapeutic target in MA addiction. Absence of increased [F-18]FEPPA TSPO binding might be related to insufficient MA dose or blunting of microglial response following repeated MA exposure, as suggested by some animal data.
Collapse
Affiliation(s)
- Gausiha Rathitharan
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Jennifer Truong
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Junchao Tong
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
| | - Tina McCluskey
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
| | - Jeffrey H. Meyer
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Department of Pharmacology and Toxicology University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Romina Mizrahi
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Department of Pharmacology and Toxicology University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Jerry Warsh
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Department of Pharmacology and Toxicology University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Pablo Rusjan
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Pharmacology and Toxicology University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - James L. Kennedy
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Sylvain Houle
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
| | - Stephen J. Kish
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Department of Pharmacology and Toxicology University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Isabelle Boileau
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| |
Collapse
|
11
|
Sharma AA, Nenert R, Mueller C, Maudsley AA, Younger JW, Szaflarski JP. Repeatability and Reproducibility of in-vivo Brain Temperature Measurements. Front Hum Neurosci 2020; 14:598435. [PMID: 33424566 PMCID: PMC7785722 DOI: 10.3389/fnhum.2020.598435] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Magnetic resonance spectroscopic imaging (MRSI) is a neuroimaging technique that may be useful for non-invasive mapping of brain temperature (i.e., thermometry) over a large brain volume. To date, intra-subject reproducibility of MRSI-based brain temperature (MRSI-t) has not been investigated. The objective of this repeated measures MRSI-t study was to establish intra-subject reproducibility and repeatability of brain temperature, as well as typical brain temperature range. Methods: Healthy participants aged 23-46 years (N = 18; 7 females) were scanned at two time points ~12-weeks apart. Volumetric MRSI data were processed by reconstructing metabolite and water images using parametric spectral analysis. Brain temperature was derived using the frequency difference between water and creatine (TCRE) for 47 regions of interest (ROIs) delineated by the modified Automated Anatomical Labeling (AAL) atlas. Reproducibility was measured using the coefficient of variation for repeated measures (COVrep), and repeatability was determined using the standard error of measurement (SEM). For each region, the upper and lower bounds of Minimal Detectable Change (MDC) were established to characterize the typical range of TCRE values. Results: The mean global brain temperature over all subjects was 37.2°C with spatial variations across ROIs. There was a significant main effect for time [F (1, 1,591) = 37.0, p < 0.0001] and for brain region [F (46, 1,591) = 2.66, p < 0.0001]. The time*brain region interaction was not significant [F (46, 1,591) = 0.80, p = 0.83]. Participants' TCRE was stable for each ROI across both time points, with ROIs' COVrep ranging from 0.81 to 3.08% (mean COVrep = 1.92%); majority of ROIs had a COVrep <2.0%. Conclusions: Brain temperature measurements were highly consistent between both time points, indicating high reproducibility and repeatability of MRSI-t. MRSI-t may be a promising diagnostic, prognostic, and therapeutic tool for non-invasively monitoring brain temperature changes in health and disease. However, further studies of healthy participants with larger sample size(s) and numerous repeated acquisitions are imperative for establishing a reference range of typical brain TCRE, as well as the threshold above which TCRE is likely pathological.
Collapse
Affiliation(s)
- Ayushe A. Sharma
- Department of Psychology, University of Alabama at Birmingham (UAB), Birmingham, AL, United States
- Department of Neurobiology, University of Alabama at Birmingham (UAB), Birmingham, AL, United States
- University of Alabama at Birmingham Epilepsy Center (UABEC), Birmingham, AL, United States
| | - Rodolphe Nenert
- University of Alabama at Birmingham Epilepsy Center (UABEC), Birmingham, AL, United States
- Department of Neurology, University of Alabama at Birmingham (UAB), Birmingham, AL, United States
| | - Christina Mueller
- Department of Psychology, University of Alabama at Birmingham (UAB), Birmingham, AL, United States
| | - Andrew A. Maudsley
- Department of Radiology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Jarred W. Younger
- Department of Psychology, University of Alabama at Birmingham (UAB), Birmingham, AL, United States
| | - Jerzy P. Szaflarski
- Department of Neurobiology, University of Alabama at Birmingham (UAB), Birmingham, AL, United States
- University of Alabama at Birmingham Epilepsy Center (UABEC), Birmingham, AL, United States
- Department of Neurology, University of Alabama at Birmingham (UAB), Birmingham, AL, United States
- Department of Neurosurgery, University of Alabama at Birmingham (UAB), Birmingham, AL, United States
| |
Collapse
|
12
|
Sharma AA, Szaflarski JP. In Vivo Imaging of Neuroinflammatory Targets in Treatment-Resistant Epilepsy. Curr Neurol Neurosci Rep 2020; 20:5. [PMID: 32166626 DOI: 10.1007/s11910-020-1025-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Recent evidence indicates that chronic, low-level neuroinflammation underlies epileptogenesis. Targeted imaging of key neuroinflammatory cells, receptors, and tissues may enable localizing epileptogenic onset zone, especially in those patients who are treatment-resistant and considered MRI-negative. Finding a specific, sensitive neuroimaging-based biomarker could aid surgical planning and improve overall prognosis in eligible patients. This article reviews recent research on in vivo imaging of neuroinflammatory targets in patients with treatment-resistant, non-lesional epilepsy. RECENT FINDINGS A number of advanced approaches based on imaging neuroinflammation are being implemented in order to assist localization of epileptogenic onset zone. The most exciting tools are based on radioligand-based nuclear imaging or revisiting of existing technology in novel ways. The greatest limitations stem from gaps in knowledge about the exact function of neuroinflammatory targets (e.g., neurotoxic or neuroprotective). Further, lingering questions about each approach's specificity, reliability, and sensitivity must be addressed, and clinical utility must be validated before any novel method is incorporated into mainstream clinical practice. Current applications of imaging neuroinflammation in humans are limited and underutilized, but offer hope for finding sensitive and specific neuroimaging-based biomarker(s). Future work necessitates appreciation of investigations to date, significant findings, and neuroinflammatory targets worth exploring further.
Collapse
Affiliation(s)
- Ayushe A Sharma
- Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of Neurology, UAB Epilepsy Center, University of Alabama at Birmingham, 1719 6th Avenue South, CIRC 312, Birmingham, AL, 35249-0021, USA.
| | - Jerzy P Szaflarski
- Department of Neurology, UAB Epilepsy Center, University of Alabama at Birmingham, 1719 6th Avenue South, CIRC 312, Birmingham, AL, 35249-0021, USA.,University of Alabama at Birmingham Epilepsy Center, Birmingham, AL, USA
| |
Collapse
|
13
|
Betlazar C, Middleton RJ, Banati R, Liu GJ. The Translocator Protein (TSPO) in Mitochondrial Bioenergetics and Immune Processes. Cells 2020; 9:cells9020512. [PMID: 32102369 PMCID: PMC7072813 DOI: 10.3390/cells9020512] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022] Open
Abstract
The translocator protein (TSPO) is an outer mitochondrial membrane protein that is widely used as a biomarker of neuroinflammation, being markedly upregulated in activated microglia in a range of brain pathologies. Despite its extensive use as a target in molecular imaging studies, the exact cellular functions of this protein remain in question. The long-held view that TSPO plays a fundamental role in the translocation of cholesterol through the mitochondrial membranes, and thus, steroidogenesis, has been disputed by several groups with the advent of TSPO knockout mouse models. Instead, much evidence is emerging that TSPO plays a fundamental role in cellular bioenergetics and associated mitochondrial functions, also part of a greater role in the innate immune processes of microglia. In this review, we examine the more direct experimental literature surrounding the immunomodulatory effects of TSPO. We also review studies which highlight a more central role for TSPO in mitochondrial processes, from energy metabolism, to the propagation of inflammatory responses through reactive oxygen species (ROS) modulation. In this way, we highlight a paradigm shift in approaches to TSPO functioning.
Collapse
Affiliation(s)
- Calina Betlazar
- Human Health, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia; (R.J.M.); (R.B.)
- 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
- Correspondence: (C.B.); (G-J.L.)
| | - Ryan J. Middleton
- Human Health, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia; (R.J.M.); (R.B.)
| | - Richard Banati
- Human Health, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia; (R.J.M.); (R.B.)
- 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
- Human Health, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia; (R.J.M.); (R.B.)
- 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
- Correspondence: (C.B.); (G-J.L.)
| |
Collapse
|
14
|
Cai L, Kirchleitner SV, Zhao D, Li M, Tonn JC, Glass R, Kälin RE. Glioblastoma Exhibits Inter-Individual Heterogeneity of TSPO and LAT1 Expression in Neoplastic and Parenchymal Cells. Int J Mol Sci 2020; 21:ijms21020612. [PMID: 31963507 PMCID: PMC7013601 DOI: 10.3390/ijms21020612] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 02/06/2023] Open
Abstract
Molecular imaging is essential for diagnosis and treatment planning for glioblastoma patients. Positron emission tomography (PET) with tracers for the detection of the solute carrier family 7 member 5 (SLC7A5; also known as the amino acid transporter light chain L system, LAT1) and for the mitochondrial translocator protein (TSPO) is successfully used to provide additional information on tumor volume and prognosis. The current approaches for TSPO-PET and the visualization of tracer ([18F] Fluoroethyltyrosine, FET) uptake by LAT1 (FET-PET) do not yet exploit the full diagnostic potential of these molecular imaging techniques. Therefore, we investigated the expression of TSPO and LAT1 in patient glioblastoma (GBM) samples, as well as in various GBM mouse models representing patient GBMs of different genetic subtypes. By immunohistochemistry, we found that TSPO and LAT1 are upregulated in human GBM samples compared to normal brain tissue. Next, we orthotopically implanted patient-derived GBM cells, as well as genetically engineered murine GBM cells, representing different genetic subtypes of the disease. To determine TSPO and LAT1 expression, we performed immunofluorescence staining. We found that both TSPO and LAT1 expression was increased in tumor regions of the implanted human or murine GBM cells when compared to the neighboring mouse brain tissue. While LAT1 was largely restricted to tumor cells, we found that TSPO was also expressed by microglia, tumor-associated macrophages, endothelial cells, and pericytes. The Cancer Genome Atlas (TCGA)-data analysis corroborates the upregulation of TSPO in a bigger cohort of GBM patient samples compared to tumor-free brain tissue. In addition, AIF1 (the gene encoding for the myeloid cell marker Iba1) was also upregulated in GBM compared to the control. Interestingly, TSPO, as well as AIF1, showed significantly different expression levels depending on the GBM genetic subtype, with the highest expression being exhibited in the mesenchymal subtype. High TSPO and AIF1 expression also correlated with a significant decrease in patient survival compared to low expression. In line with this finding, the expression levels for TSPO and AIF1 were also significantly higher in (isocitrate-dehydrogenase wild-type) IDHWT compared to IDH mutant (IDHMUT) GBM. LAT1 expression, on the other hand, was not different among the individual GBM subtypes. Therefore, we could conclude that FET- and TSPO-PET confer different information on pathological features based on different genetic GBM subtypes and may thus help in planning individualized strategies for brain tumor therapy in the future. A combination of TSPO-PET and FET-PET could be a promising way to visualize tumor-associated myeloid cells and select patients for treatment strategies targeting the myeloid compartment.
Collapse
Affiliation(s)
- Linzhi Cai
- Neurosurgical Research, Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany (S.V.K.)
| | - Sabrina V. Kirchleitner
- Neurosurgical Research, Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany (S.V.K.)
- Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Dongxu Zhao
- Neurosurgical Research, Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany (S.V.K.)
| | - Min Li
- Neurosurgical Research, Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany (S.V.K.)
| | - Jörg-Christian Tonn
- Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Rainer Glass
- Neurosurgical Research, Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany (S.V.K.)
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence:
| | - Roland E. Kälin
- Neurosurgical Research, Department of Neurosurgery, University Hospital, LMU Munich, 81377 Munich, Germany (S.V.K.)
- Walter Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| |
Collapse
|
15
|
The Molecular Effects of Ionizing Radiations on Brain Cells: Radiation Necrosis vs. Tumor Recurrence. Diagnostics (Basel) 2019; 9:diagnostics9040127. [PMID: 31554255 PMCID: PMC6963489 DOI: 10.3390/diagnostics9040127] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/13/2019] [Accepted: 09/20/2019] [Indexed: 12/12/2022] Open
Abstract
The central nervous system (CNS) is generally resistant to the effects of radiation, but higher doses, such as those related to radiation therapy, can cause both acute and long-term brain damage. The most important results is a decline in cognitive function that follows, in most cases, cerebral radionecrosis. The essence of radio-induced brain damage is multifactorial, being linked to total administered dose, dose per fraction, tumor volume, duration of irradiation and dependent on complex interactions between multiple brain cell types. Cognitive impairment has been described following brain radiotherapy, but the mechanisms leading to this adverse event remain mostly unknown. In the event of a brain tumor, on follow-up radiological imaging often cannot clearly distinguish between recurrence and necrosis, while, especially in patients that underwent radiation therapy (RT) post-surgery, positron emission tomography (PET) functional imaging, is able to differentiate tumors from reactive phenomena. More recently, efforts have been done to combine both morphological and functional data in a single exam and acquisition thanks to the co-registration of PET/MRI. The future of PET imaging to differentiate between radionecrosis and tumor recurrence could be represented by a third-generation PET tracer already used to reveal the spatial extent of brain inflammation. The aim of the following review is to analyze the effect of ionizing radiations on CNS with specific regard to effect of radiotherapy, focusing the attention on the mechanism underling the radionecrosis and the brain damage, and show the role of nuclear medicine techniques to distinguish necrosis from recurrence and to early detect of cognitive decline after treatment.
Collapse
|
16
|
|