1
|
Phillips RD. Neural and immune interactions linking early life stress and anhedonia. Brain Behav Immun Health 2024; 42:100881. [PMID: 39415844 PMCID: PMC11480252 DOI: 10.1016/j.bbih.2024.100881] [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: 02/16/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024] Open
Abstract
Early experiences of stress and adversity are associated with blunted reward sensitivity and altered reward learning. Meanwhile, anhedonia is characterized by impairments in reward processing, including motivation, effort, and pleasure. Early life stress (ELS) and anhedonia share psychological, behavioral, and neurobiological correlates, and the system-level interactions that give rise to anhedonia have yet to be fully appreciated. The proposed framework uses a multilevel, multisystem approach to aid in understanding neural-immune interactions that link ELS and anhedonia. The interactions linking anhedonia and ELS presented here include reduced reward sensitivity, alterations in hypothalamic-pituitary-adrenal (HPA) axis response, elevated inflammatory cytokines or physiological markers of stress, and blunted reward circuitry functioning along the mesocorticolimbic pathway. The clinical implications and areas for future research are also discussed. Ultimately, this research may inform the development of more specific and individualized treatments for anhedonia.
Collapse
Affiliation(s)
- Rachel Deanna Phillips
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, USA
| |
Collapse
|
2
|
Herzog S, Bartlett EA, Zanderigo F, Galfalvy HC, Burke A, Mintz A, Schmidt M, Hauser E, Huang YY, Melhem N, Sublette ME, Miller JM, Mann JJ. Neuroinflammation, Stress-Related Suicidal Ideation, and Negative Mood in Depression. JAMA Psychiatry 2024:2825422. [PMID: 39504032 PMCID: PMC11541744 DOI: 10.1001/jamapsychiatry.2024.3543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/17/2024] [Indexed: 11/09/2024]
Abstract
Importance Brain translocator protein 18k Da (TSPO) binding, a putative marker of neuroinflammatory processes (eg, gliosis), is associated with stress and elevated in depressed and suicidal populations. However, it is unclear whether neuroinflammation moderates the impact of daily life stress on suicidal ideation and negative affect, thereby increasing risk for suicidal behavior. Objective To examine the association of TSPO binding in participants with depression with real-world daily experiences of acute stress-related suicidal ideation and negative affect, as well as history of suicidal behavior and clinician-rated suicidal ideation. Design, Setting, and Participants Data for this cross-sectional study were collected from June 2019 through July 2023. Procedures were conducted at a hospital-based research center in New York, New York. Participants were recruited via clinical referrals, the Columbia University research subject web portal, and from responses to internet advertisements. Of 148 participants who signed informed consent for study protocols, 53 adults aged 18 to 60 years who met DSM-5 diagnostic criteria for current major depressive disorder completed procedures with approved data and were enrolled. Participants were free of schizophrenia spectrum disorders, active physical illness, cognitive impairment, and substance intoxication or withdrawal at the time of scan. Exposures All participants underwent positron emission tomography imaging of TSPO binding with 11C-ER176 and concurrent arterial blood sampling. Main Outcome and Measures A weighted average of 11C-ER176 total distribution volume (VT) was computed across 11 a priori brain regions and made up the primary outcome measure. Clinician-rated suicidal ideation was measured via the Beck Scale for Suicidal Ideation (BSS). A subset of participants (n = 21) completed 7 days of ecological momentary assessment (EMA), reporting daily on suicidal ideation, negative affect, and stressors. Results In the overall sample of 53 participants (mean [SD] age, 29.5 [9.8] years; 37 [69.8%] female and 16 [30.2%] male), 11C-ER176 VT was associated at trend levels with clinician-rated suicidal ideation severity (β, 0.19; 95% CI, -0.03 to 0.39; P = .09) and did not differ by suicide attempt history (n = 15; β, 0.18; 95% CI, -0.04 to 0.37; P = .11). Exploratory analyses indicated that presence of suicidal ideation (on BSS or EMA) was associated with higher 11C-ER176 VT (β, 0.21; 95% CI, 0.01 to 0.98; P = .045). In 21 participants who completed EMA, 11C-ER176 VT was associated with greater suicidal ideation and negative affect during EMA periods with stressors compared with nonstress periods (β, 0.12; SE, 0.06; 95% CI, 0.01 to 0.23; P = .03 and β, 0.19; SE, 0.06; 95% CI, 0.08 to 0.30; P < .001, respectively). Conclusion and Relevance TSPO binding in individuals with depression may be a marker of vulnerability to acute stress-related increases in suicidal ideation and negative affect. Continued study is needed to determine the causal direction of TSPO binding and stress-related suicidal ideation or negative affect and whether targeting neuroinflammation may improve resilience to life stress in patients with depression.
Collapse
Affiliation(s)
- Sarah Herzog
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Elizabeth A. Bartlett
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Francesca Zanderigo
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Hanga C. Galfalvy
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
- Department of Biostatistics, Columbia University, New York, New York
| | - Ainsley Burke
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Akiva Mintz
- Department of Radiology, Columbia University Irving Medical Center, New York, New York
| | - Mike Schmidt
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Eric Hauser
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Yung-yu Huang
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Nadine Melhem
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - M. Elizabeth Sublette
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Jeffrey M. Miller
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - J. John Mann
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
- Department of Radiology, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
3
|
Hattori M, Kikutani K, Hosokawa K, Kyo M, Nishikimi M, Ota K, Ohshimo S, Aizawa H, Shime N. Diagnostic utility of plasma translocator protein 18 kDa (TSPO) in sepsis: A case-control study. Medicine (Baltimore) 2024; 103:e40396. [PMID: 39495982 PMCID: PMC11537663 DOI: 10.1097/md.0000000000040396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 10/16/2024] [Indexed: 11/06/2024] Open
Abstract
Translocator protein 18 kDa (TSPO) is a mitochondrial membrane protein that is involved in inflammation, oxidative stress, and steroidogenesis. TSPO may be a marker of inflammatory responses in the brain and other organs, but there have been few studies of the potential clinical significance of measuring the circulating TSPO concentration, especially in patients with sepsis. In this study, we compared the circulating TSPO concentrations of patients with sepsis and healthy controls to investigate the utility of plasma TSPO for the diagnosis of sepsis. Patients with sepsis admitted to the intensive care unit of Hiroshima University Hospital between January 2020 and April 2024 were enrolled. Plasma samples were collected from patients within 24 hours of admission and also from healthy volunteers, and their plasma TSPO concentrations were compared. Receiver operating characteristic analysis was used to evaluate the usefulness of plasma TSPO concentration for the diagnosis of sepsis. We also investigated the relationships of TSPO concentration with the severity of sepsis, complications, and prognosis of the patients. Eighty subjects (52 patients and 28 controls) were included in this study. The plasma TSPO concentrations of the patients with sepsis were significantly lower than those of the healthy controls (0.094 vs 0.25 ng/mL, P < .001), and receiver operating characteristic analysis generated an area under the curve of 0.81 (95% confidence interval: 0.72-0.91). In patients with sepsis, the TSPO concentration was not associated with the severity of sepsis, complications, or prognosis. Plasma TSPO may be a useful biomarker for the diagnosis of sepsis.
Collapse
Affiliation(s)
- Miyuki Hattori
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Neurobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuya Kikutani
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Neurobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Koji Hosokawa
- Department of Anesthesiology and Reanimatology, Faculty of Medicine Sciences, University of Fukui, Fukui, Japan
| | - Michihito Kyo
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Radiation Disaster Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Mitsuaki Nishikimi
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kohei Ota
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shinichiro Ohshimo
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hidenori Aizawa
- Department of Neurobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobuaki Shime
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
4
|
Treaba CA, Herranz E, Barletta VT, Mehndiratta A, Sloane JA, Granberg T, Miscioscia A, Bomprezzi R, Loggia ML, Mainero C. Phenotyping in vivo chronic inflammation in multiple sclerosis by combined 11C-PBR28 MR-PET and 7T susceptibility-weighted imaging. Mult Scler 2024:13524585241284157. [PMID: 39436837 DOI: 10.1177/13524585241284157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
BACKGROUND 11C-PBR28 positron emission tomography (PET), targeting the translocator protein, and paramagnetic rim lesions (PRL) have emerged as promising imaging markers of MS chronic inflammation. No consensus on which is the optimal marker exists. OBJECTIVES To investigate the ability of 11C-PBR28 PET and PRL assessment to identify chronic inflammation in white matter (WM) MS lesions and their relation to neurological impairment. METHODS Based on 11C-PBR28 uptake, brain WM lesions from 30 MS patients were classified as PET active or inactive. The PRL presence was assessed on 7T phase reconstructions, T1/T2 ratio was calculated to measure WM microstructural integrity. RESULTS Less than half (44%) of non-PRL WM lesions were active on 11C-PBR28 imaging either throughout the lesion (whole active) or at its periphery. PET peripherally active lesions and PRL did not differ in T1/T2 ratio and 11C-PBR28 uptake. A positive correlation was observed between PRL and active PET lesion count. Whole active PET lesion volume was the strongest predictor (β = 0.97, p < 0.001) of increased Expanded Disability Status Scale scores. CONCLUSION 11C-PBR28 imaging reveals more active WM lesions than 7T PRL assessment. Although PRL and PET active lesion counts are related, neurological disability is better explained by PET whole active lesion volume.
Collapse
Affiliation(s)
- Constantina A Treaba
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Elena Herranz
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Valeria T Barletta
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ambica Mehndiratta
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Jacob A Sloane
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Tobias Granberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Alessandro Miscioscia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Neuroscience, University of Padua, Padua, Italy
| | - Roberto Bomprezzi
- Department of Neurology, UMass Chan Medical School, Worcester, MA, USA
| | - Marco L Loggia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care & Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Caterina Mainero
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Bartos LM, Quach S, Zenatti V, Kirchleitner SV, Blobner J, Wind-Mark K, Kolabas ZI, Ulukaya S, Holzgreve A, Ruf VC, Kunze LH, Kunte ST, Hoermann L, Härtel M, Park HE, Groß M, Franzmeier N, Zatcepin A, Zounek A, Kaiser L, Riemenschneider MJ, Perneczky R, Rauchmann BS, Stöcklein S, Ziegler S, Herms J, Ertürk A, Tonn JC, Thon N, von Baumgarten L, Prestel M, Tahirovic S, Albert NL, Brendel M. Remote Neuroinflammation in Newly Diagnosed Glioblastoma Correlates with Unfavorable Clinical Outcome. Clin Cancer Res 2024; 30:4618-4634. [PMID: 39150564 PMCID: PMC11474166 DOI: 10.1158/1078-0432.ccr-24-1563] [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: 05/22/2024] [Revised: 07/15/2024] [Accepted: 08/14/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE Current therapy strategies still provide only limited success in the treatment of glioblastoma, the most frequent primary brain tumor in adults. In addition to the characterization of the tumor microenvironment, global changes in the brain of patients with glioblastoma have been described. However, the impact and molecular signature of neuroinflammation distant of the primary tumor site have not yet been thoroughly elucidated. EXPERIMENTAL DESIGN We performed translocator protein (TSPO)-PET in patients with newly diagnosed glioblastoma (n = 41), astrocytoma WHO grade 2 (n = 7), and healthy controls (n = 20) and compared TSPO-PET signals of the non-lesion (i.e., contralateral) hemisphere. Back-translation into syngeneic SB28 glioblastoma mice was used to characterize Pet alterations on a cellular level. Ultimately, multiplex gene expression analyses served to profile immune cells in remote brain. RESULTS Our study revealed elevated TSPO-PET signals in contralateral hemispheres of patients with newly diagnosed glioblastoma compared to healthy controls. Contralateral TSPO was associated with persisting epileptic seizures and shorter overall survival independent of the tumor phenotype. Back-translation into syngeneic glioblastoma mice pinpointed myeloid cells as the predominant source of contralateral TSPO-PET signal increases and identified a complex immune signature characterized by myeloid cell activation and immunosuppression in distant brain regions. CONCLUSIONS Neuroinflammation within the contralateral hemisphere can be detected with TSPO-PET imaging and associates with poor outcome in patients with newly diagnosed glioblastoma. The molecular signature of remote neuroinflammation promotes the evaluation of immunomodulatory strategies in patients with detrimental whole brain inflammation as reflected by high TSPO expression.
Collapse
Affiliation(s)
- Laura M. Bartos
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Stefanie Quach
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
| | - Valerio Zenatti
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
| | | | - Jens Blobner
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
| | - Karin Wind-Mark
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Zeynep Ilgin Kolabas
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Munich, Germany.
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany.
| | - Selin Ulukaya
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Munich, Germany.
- Faculty of Biology, Master of Science Program in Molecular and Cellular Biology, Ludwig-Maximilians-Universität München, Planegg, Germany.
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Viktoria C. Ruf
- Center for Neuropathology and Prion Research, University Hospital, LMU Munich, Munich, Germany.
| | - Lea H. Kunze
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Sebastian T. Kunte
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Leonie Hoermann
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Marlies Härtel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Ha Eun Park
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Mattes Groß
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Artem Zatcepin
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
| | - Adrian Zounek
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Lena Kaiser
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | | | - Robert Perneczky
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), University of Munich, Munich, Germany.
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom.
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, United Kingdom.
| | | | - Sophia Stöcklein
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany.
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Jochen Herms
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
- Center for Neuropathology and Prion Research, University Hospital, LMU Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), University of Munich, Munich, Germany.
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Munich, Germany.
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), University of Munich, Munich, Germany.
| | - Joerg C. Tonn
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Niklas Thon
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Louisa von Baumgarten
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Matthias Prestel
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
| | - Sabina Tahirovic
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
| | - Nathalie L. Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Munich Cluster for Systems Neurology (SyNergy), University of Munich, Munich, Germany.
| |
Collapse
|
6
|
Shraim MA, Massé-Alarie H, Farrell MJ, Cavaleri R, Loggia ML, Hodges PW. Neuroinflammatory activation in sensory and motor regions of the cortex is related to sensorimotor function in individuals with low back pain maintained by nociplastic mechanisms: A preliminary proof-of-concept study. Eur J Pain 2024; 28:1607-1626. [PMID: 39007713 DOI: 10.1002/ejp.2313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 06/26/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Chronic pain involves communication between neural and immune systems. Recent data suggest localization of glial (brain immune cells) activation to the sensorimotor regions of the brain cortex (S1/M1) in chronic low back pain (LBP). As glia perform diverse functions that impact neural function, activation might contribute to sensorimotor changes, particularly in LBP maintained by increased nervous system sensitivity (i.e., nociplastic pain). This preliminary proof-of-concept study aimed to: (i) compare evidence of neuroinflammatory activation in S1/M1 between individuals with and without LBP (and between nociceptive and nociplastic LBP phenotypes), and (ii) evaluate relationships between neuroinflammatory activation and sensorimotor function. METHODS Simultaneous PET-fMRI measured neuroinflammatory activation in functionally defined S1/M1 in pain-free individuals (n = 8) and individuals with chronic LBP (n = 9; nociceptive: n = 4, nociplastic: n = 5). Regions of S1/M1 related to the back were identified using fMRI during motor tasks and thermal stimuli. Sensorimotor measures included single and paired-pulse transcranial magnetic stimulation (TMS) and quantitative sensory testing (QST). Sleep, depression, disability and pain questionnaires were administered. RESULTS Neuroinflammatory activation was greater in the lower back cortical representation of S1/M1 of the nociplastic LBP group than both nociceptive LBP and pain-free groups. Neuroinflammatory activation in S1/M1 was positively correlated with sensitivity to hot (r = 0.52) and cold (r = 0.55) pain stimuli, poor sleep, depression, disability and BMI, and negatively correlated with intracortical facilitation (r = -0.41). CONCLUSION This preliminary proof-of-concept study suggests that neuroinflammation in back regions of S1/M1 in individuals with nociplastic LBP could plausibly explain some characteristic features of this LBP phenotype. SIGNIFICANCE STATEMENT Neuroinflammatory activation localized to sensorimotor areas of the brain in individuals with nociplastic pain might contribute to changes in sensory and motor function and aspects of central sensitization. If cause-effect relationships are established in longitudinal studies, this may direct development of therapies that target neuroinflammatory activation.
Collapse
Affiliation(s)
- Muath A Shraim
- The University of Queensland, School of Health & Rehabilitation Sciences, St Lucia, Queensland, Australia
| | - Hugo Massé-Alarie
- The University of Queensland, School of Health & Rehabilitation Sciences, St Lucia, Queensland, Australia
- Centre Interdisciplinaire de Recherche en réadaptation et Integration Sociale (CIRRIS), Université Laval, Québec City, Québec, Canada
| | - Michael J Farrell
- Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| | - Rocco Cavaleri
- Brain Stimulation and Rehabilitation Lab, Western Sydney University, School of Health Sciences, Sydney, New South Wales, Australia
| | - Marco L Loggia
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Paul W Hodges
- The University of Queensland, School of Health & Rehabilitation Sciences, St Lucia, Queensland, Australia
| |
Collapse
|
7
|
Kara F, Kantarci K. Understanding Proton Magnetic Resonance Spectroscopy Neurochemical Changes Using Alzheimer's Disease Biofluid, PET, Postmortem Pathology Biomarkers, and APOE Genotype. Int J Mol Sci 2024; 25:10064. [PMID: 39337551 PMCID: PMC11432594 DOI: 10.3390/ijms251810064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/15/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
In vivo proton (1H) magnetic resonance spectroscopy (MRS) is a powerful non-invasive method that can measure Alzheimer's disease (AD)-related neuropathological alterations at the molecular level. AD biomarkers include amyloid-beta (Aβ) plaques and hyperphosphorylated tau neurofibrillary tangles. These biomarkers can be detected via postmortem analysis but also in living individuals through positron emission tomography (PET) or biofluid biomarkers of Aβ and tau. This review offers an overview of biochemical abnormalities detected by 1H MRS within the biologically defined AD spectrum. It includes a summary of earlier studies that explored the association of 1H MRS metabolites with biofluid, PET, and postmortem AD biomarkers and examined how apolipoprotein e4 allele carrier status influences brain biochemistry. Studying these associations is crucial for understanding how AD pathology affects brain homeostasis throughout the AD continuum and may eventually facilitate the development of potential novel therapeutic approaches.
Collapse
Affiliation(s)
- Firat Kara
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
8
|
Meng H, He L, Chunyu H, Zhou Q, Wang J, Qu Q, Hai W, Zhang Y, Li B, Zhang M, Chen S. 18F-DPA714 PET/MRI as a potential imaging tool for detecting possible antibody-negative autoimmune encephalitis: a prospective study. J Neurol 2024:10.1007/s00415-024-12690-w. [PMID: 39294471 DOI: 10.1007/s00415-024-12690-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/06/2024] [Accepted: 09/07/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND AND OBJECTIVES Conventional magnetic resonance imaging (MRI) used for detecting possible antibody-negative autoimmune encephalitis (AIE) often fails to meet the diagnostic requirements of this disease. Positron emission tomography (PET) with a translocator protein radioligand can help visualize microglia distribution density in inflammation-related diseases, thereby offering potentially incremental value to conventional MRI for the in vivo assessment of possible antibody-negative AIE. METHODS In this prospective study, 15 participants diagnosed with possible antibody-negative AIE and 10 healthy controls were enrolled (ClinicalTrials.gov: NCT05293405, dated March 15, 2022). All participants underwent hybrid 18F-DPA714 PET/MRI and evaluation for modified Rankin scale (mRS) score, clinical assessment scale for AIE (CASE), and appropriate antibodies. A positive finding was defined as the intensity of 18F-DPA714 uptake that was above a threshold of mean standardized uptake value ratio (SUVR) + two standard deviations of SUVR within the corresponding brain regions of healthy controls. RESULTS The positive detection rate of 18F-DPA714 PET for possible antibody-negative AIE was significantly higher than that of brain MRI (10/15 [67%] vs. 3/15 [20%]; P = 0.039). In addition, both the intensity and extent of 18F-DPA714 uptake were significantly associated with the CASE score (P = 0.002 and 0.001). Meanwhile, SUVR levels in the cerebellar region were significantly higher in patients with ataxia than in those without ataxia (P = 0.006). Furthermore, 18F-DPA714 uptake decreased in 5/10 [50%] patients who underwent follow-up PET/MRI, which mirrored their symptom relief. CONCLUSION 18F-DPA714 PET demonstrated its potentially incremental value to conventional MRI for detecting possible antibody-negative AIE.
Collapse
Affiliation(s)
- Huanyu Meng
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Lu He
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Hangxing Chunyu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qinming Zhou
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jin Wang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qian Qu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wangxi Hai
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanxi Medical University-Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi, China
| | - Min Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanxi Medical University-Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi, China.
| | - Sheng Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.
- Department of Neurology, Xinrui Hospital, Wuxi, China.
| |
Collapse
|
9
|
Fairley LH, Lai KO, Grimm A, Eckert A, Barron AM. The mitochondrial translocator protein (TSPO) in Alzheimer's disease: Therapeutic and immunomodulatory functions. Biochimie 2024; 224:120-131. [PMID: 38971458 DOI: 10.1016/j.biochi.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
The translocator protein (TSPO) has been widely investigated as a PET-imaging biomarker of neuroinflammation and, more recently, as a therapeutic target for the treatment of neurodegenerative disease. TSPO ligands have been shown to exert neuroprotective effects in vivo and in vitro models of Alzheimer's disease (AD), by reducing toxic beta amyloid peptides, and attenuating brain atrophy. Recent transcriptomic and proteomic analyses, and the generation of TSPO-KO mice, have enabled new insights into the mechanistic function of TSPO in AD. Using a multi-omics approach in both TSPO-KO- and TSPO ligand-treated mice, we have demonstrated a key role for TSPO in microglial respiratory metabolism and phagocytosis in AD. In this review, we discuss emerging evidence for therapeutic and immunomodulatory functions of TSPO in AD, and new tools for studying TSPO in the brain.
Collapse
Affiliation(s)
- Lauren H Fairley
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 308232, Singapore
| | - Kei Onn Lai
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 308232, Singapore
| | - Amandine Grimm
- Transfaculty Research Platform, Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland; Psychiatric University Clinics, Basel, Switzerland
| | - Anne Eckert
- Transfaculty Research Platform, Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland; Psychiatric University Clinics, Basel, Switzerland
| | - Anna M Barron
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 308232, Singapore.
| |
Collapse
|
10
|
Bonomi R, Hillmer AT, Woodcock E, Bhatt S, Rusowicz A, Angarita GA, Carson RE, Davis MT, Esterlis I, Nabulsi N, Huang Y, Krystal JH, Pietrzak RH, Cosgrove KP. Microglia-mediated neuroimmune suppression in PTSD is associated with anhedonia. Proc Natl Acad Sci U S A 2024; 121:e2406005121. [PMID: 39172786 PMCID: PMC11363315 DOI: 10.1073/pnas.2406005121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/16/2024] [Indexed: 08/24/2024] Open
Abstract
Dynamic brain immune function in individuals with posttraumatic stress disorder is rarely studied, despite evidence of peripheral immune dysfunction. Positron emission tomography brain imaging using the radiotracer [11C]PBR28 was used to measure the 18-kDa translocator protein (TSPO), a microglial marker, at baseline and 3 h after administration of lipopolysaccharide (LPS), a potent immune activator. Data were acquired in 15 individuals with PTSD and 15 age-matched controls. The PTSD group exhibited a significantly lower magnitude LPS-induced increase in TSPO availability in an a priori prefrontal-limbic circuit compared to controls. Greater anhedonic symptoms in the PTSD group were associated with a more suppressed neuroimmune response. In addition, while a reduced granulocyte-macrophage colony-stimulating factor response to LPS was observed in the PTSD group, other measured cytokine responses and self-reported sickness symptoms did not differ between groups; these findings highlight group differences in central-peripheral immune system relationships. The results of this study provide evidence of a suppressed microglia-mediated neuroimmune response to a direct immune system insult in individuals with PTSD that is associated with the severity of symptoms. They also provide further support to an emerging literature challenging traditional concepts of microglial and immune function in psychiatric disease.
Collapse
Affiliation(s)
- Robin Bonomi
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
| | - Ansel T. Hillmer
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT06520
- Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT06519
| | - Eric Woodcock
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
| | - Shivani Bhatt
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
| | | | | | - Richard E. Carson
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT06520
- Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT06519
| | - Margaret T. Davis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, Veterans Affairs Connecticut Healthcare System, West Haven, CT06516
| | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT06520
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, Veterans Affairs Connecticut Healthcare System, West Haven, CT06516
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT06520
- Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT06519
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT06520
- Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT06519
| | - John H. Krystal
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, Veterans Affairs Connecticut Healthcare System, West Haven, CT06516
| | - Robert H. Pietrzak
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, Veterans Affairs Connecticut Healthcare System, West Haven, CT06516
| | - Kelly P. Cosgrove
- Department of Psychiatry, Yale School of Medicine, New Haven, CT06511
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT06520
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, Veterans Affairs Connecticut Healthcare System, West Haven, CT06516
| |
Collapse
|
11
|
Rousseau C, Metz R, Kerdraon O, Ouldamer L, Boiffard F, Renaudeau K, Ferrer L, Vercouillie J, Doutriaux-Dumoulin I, Mouton A, Le Thiec M, Morel A, Rusu D, Santiago-Ribeiro MJ, Campion L, Arlicot N, Kraeber-Bodéré F. Pilot Feasibility Study: 18 F-DPA-714 PET/CT Macrophage Imaging in Triple-Negative Breast Cancers (EITHICS). Clin Nucl Med 2024; 49:701-708. [PMID: 38913962 DOI: 10.1097/rlu.0000000000005338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
ABSTRACT Tumor-associated macrophages are targets of interest in triple-negative breast cancer (TNBC). The translocator protein 18 kDa (TSPO) is a sensitive marker for macrophages and holds potential relevance in TNBC stratification. This pilot prospective study (EITHICS, NCT04320030) aimed to assess the potential of TSPO PET/CT imaging using 18 F-DPA-714 in primary TNBC, compared with immunohistochemistry, autoradiography, and TSPO polymorphism. PATIENTS AND METHODS Thirteen TNBC patients were included. They underwent TSPO genotyping (HAB, MAB, LAB), 18 F-FDG PET/CT, and breast MRI. Semiquantitative PET parameters were computed. VOIs were defined on the tumor lesion, healthy breast tissue, and pectoral muscle to obtain SUV, tumor-to-background ratio (TBR), and time-activity curves (TACs). Additionally, immunohistochemistry, 3 H-DPA-714, and 3 H-PK-11195 autoradiography were conducted. RESULTS The majority of TNBC tumors (11/13, 84%) had a preponderance of M2-polarized macrophages with a median proportion of 82% (range, 44%-94%). 18 F-DPA-714 PET/CT clearly identified TNBC tumors with an excellent TBR. Three distinct patterns of 18 F-DPA-714 TACs were identified, categorized as "above muscular," "equal to muscular," and "below muscular" with reference to the muscular background. For the "above muscular" group (2 HAB and 2 MAB), "equal muscular" group (3 HAB, 3 MAB, and 1 LAB), and "below muscular" group (1 LAB and 1 MAB), tumor TACs showed a 18 F-DPA-714 accumulation slope of 1.35, 0.62, and 0.22, respectively, and a median SUV mean of 4.02 (2.09-5.31), 1.66 (0.93-3.07), and 0.61 (0.43-1.02). CONCLUSIONS This study successfully demonstrated TNBC tumor targeting by 18 F-DPA-714 with an excellent TBR, allowing to stratify 3 patterns of uptake potentially influenced by the TSPO polymorphism status. Further studies in larger populations should be performed to evaluate the prognostic value of this new biomarker.
Collapse
Affiliation(s)
| | - Raphaël Metz
- From the ICO René Gauducheau, F-44800, Saint-Herblain, France
| | | | | | | | | | | | | | | | - Alexis Mouton
- From the ICO René Gauducheau, F-44800, Saint-Herblain, France
| | - Maelle Le Thiec
- From the ICO René Gauducheau, F-44800, Saint-Herblain, France
| | - Agnès Morel
- From the ICO René Gauducheau, F-44800, Saint-Herblain, France
| | - Daniela Rusu
- From the ICO René Gauducheau, F-44800, Saint-Herblain, France
| | | | | | | | | |
Collapse
|
12
|
Hemond CC, Gaitán MI, Absinta M, Reich DS. New Imaging Markers in Multiple Sclerosis and Related Disorders: Smoldering Inflammation and the Central Vein Sign. Neuroimaging Clin N Am 2024; 34:359-373. [PMID: 38942521 PMCID: PMC11213979 DOI: 10.1016/j.nic.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Concepts of multiple sclerosis (MS) biology continue to evolve, with observations such as "progression independent of disease activity" challenging traditional phenotypic categorization. Iron-sensitive, susceptibility-based imaging techniques are emerging as highly translatable MR imaging sequences that allow for visualization of at least 2 clinically useful biomarkers: the central vein sign and the paramagnetic rim lesion (PRL). Both biomarkers demonstrate high specificity in the discrimination of MS from other mimics and can be seen at 1.5 T and 3 T field strengths. Additionally, PRLs represent a subset of chronic active lesions engaged in "smoldering" compartmentalized inflammation behind an intact blood-brain barrier.
Collapse
Affiliation(s)
- Christopher C Hemond
- Department of Neurology, University of Massachusetts Memorial Medical Center and University of Massachusetts Chan Medical School, Worcester, MA, USA; National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - María I Gaitán
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Martina Absinta
- Translational Neuropathology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
13
|
Maccioni L, Michelle CM, Brusaferri L, Silvestri E, Bertoldo A, Schubert JJ, Nettis MA, Mondelli V, Howes O, Turkheimer FE, Bottlaender M, Bodini B, Stankoff B, Loggia ML, Veronese M. A blood-free modeling approach for the quantification of the blood-to-brain tracer exchange in TSPO PET imaging. Front Neurosci 2024; 18:1395769. [PMID: 39104610 PMCID: PMC11299498 DOI: 10.3389/fnins.2024.1395769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Introduction Recent evidence suggests the blood-to-brain influx rate (K1 ) in TSPO PET imaging as a promising biomarker of blood-brain barrier (BBB) permeability alterations commonly associated with peripheral inflammation and heightened immune activity in the brain. However, standard compartmental modeling quantification is limited by the requirement of invasive and laborious procedures for extracting an arterial blood input function. In this study, we validate a simplified blood-free methodologic framework for K1 estimation by fitting the early phase tracer dynamics using a single irreversible compartment model and an image-derived input function (1T1K-IDIF). Methods The method is tested on a multi-site dataset containing 177 PET studies from two TSPO tracers ([11C]PBR28 and [18F]DPA714). Firstly, 1T1K-IDIF K1 estimates were compared in terms of both bias and correlation with standard kinetic methodology. Then, the method was tested on an independent sample of [11C]PBR28 scans before and after inflammatory interferon-α challenge, and on test-retest dataset of [18F]DPA714 scans. Results Comparison with standard kinetic methodology showed good-to-excellent intra-subject correlation for regional 1T1K-IDIF-K1 (ρintra = 0.93 ± 0.08), although the bias was variable depending on IDIF ability to approximate blood input functions (0.03-0.39 mL/cm3/min). 1T1K-IDIF-K1 unveiled a significant reduction of BBB permeability after inflammatory interferon-α challenge, replicating results from standard quantification. High intra-subject correlation (ρ = 0.97 ± 0.01) was reported between K1 estimates of test and retest scans. Discussion This evidence supports 1T1K-IDIF as blood-free alternative to assess TSPO tracers' unidirectional blood brain clearance. K1 investigation could complement more traditional measures in TSPO studies, and even allow further mechanistic insight in the interpretation of TSPO signal.
Collapse
Affiliation(s)
- Lucia Maccioni
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Carranza Mellana Michelle
- Department of Information Engineering, University of Padova, Padova, Italy
- Paris Brain Institute, ICM, CNRS, Inserm, Sorbonne Université, Paris, France
| | - Ludovica Brusaferri
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
- Computer Science and Informatics, School of Engineering, London South Bank University, London, United Kingdom
| | - Erica Silvestri
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Alessandra Bertoldo
- Department of Information Engineering, University of Padova, Padova, Italy
- Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Julia J. Schubert
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Maria A. Nettis
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Valeria Mondelli
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Oliver Howes
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Federico E. Turkheimer
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Michel Bottlaender
- BioMaps, Service Hospitalier Frédéric Joliot CEA, CNRS Inserm, Université Paris-Saclay, Orsay, France
| | - Benedetta Bodini
- Paris Brain Institute, ICM, CNRS, Inserm, Sorbonne Université, Paris, France
| | - Bruno Stankoff
- Paris Brain Institute, ICM, CNRS, Inserm, Sorbonne Université, Paris, France
| | - Marco L. Loggia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Mattia Veronese
- Department of Information Engineering, University of Padova, Padova, Italy
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| |
Collapse
|
14
|
Zarkali A, Thomas GEC, Zetterberg H, Weil RS. Neuroimaging and fluid biomarkers in Parkinson's disease in an era of targeted interventions. Nat Commun 2024; 15:5661. [PMID: 38969680 PMCID: PMC11226684 DOI: 10.1038/s41467-024-49949-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 06/19/2024] [Indexed: 07/07/2024] Open
Abstract
A major challenge in Parkinson's disease is the variability in symptoms and rates of progression, underpinned by heterogeneity of pathological processes. Biomarkers are urgently needed for accurate diagnosis, patient stratification, monitoring disease progression and precise treatment. These were previously lacking, but recently, novel imaging and fluid biomarkers have been developed. Here, we consider new imaging approaches showing sensitivity to brain tissue composition, and examine novel fluid biomarkers showing specificity for pathological processes, including seed amplification assays and extracellular vesicles. We reflect on these biomarkers in the context of new biological staging systems, and on emerging techniques currently in development.
Collapse
Affiliation(s)
- Angeliki Zarkali
- Dementia Research Centre, Institute of Neurology, UCL, London, UK.
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Rimona S Weil
- Dementia Research Centre, Institute of Neurology, UCL, London, UK
- Department of Advanced Neuroimaging, UCL, London, UK
- Movement Disorders Centre, UCL, London, UK
| |
Collapse
|
15
|
Jolly AA, Brown RB, Tozer DJ, Hong YT, Fryer TD, Aigbirhio FI, O’Brien JT, Markus HS. Are central and systemic inflammation associated with fatigue in cerebral small vessel disease? Int J Stroke 2024; 19:705-713. [PMID: 38533609 PMCID: PMC11292988 DOI: 10.1177/17474930241245613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Fatigue is a common symptom in cerebral small vessel disease (SVD), but its pathogenesis is poorly understood. It has been suggested that inflammation may play a role. We determined whether central (neuro) inflammation and peripheral inflammation were associated with fatigue in SVD. METHODS Notably, 36 patients with moderate-to-severe SVD underwent neuropsychometric testing, combined positron emission tomography and magnetic resonance imaging (PET-MRI) scan, and blood draw for the analysis of inflammatory blood biomarkers. Microglial signal was taken as a proxy for neuroinflammation, assessed with radioligand 11C-PK11195. Of these, 30 subjects had full PET datasets for analysis. We assessed global 11C-PK11195 binding and hotspots of 11C-PK11195 binding in the normal-appearing white matter, lesioned tissue, and combined total white matter. Peripheral inflammation was assessed with serum C-reactive protein (CRP) and using the Olink cardiovascular III proteomic panel comprising 92 biomarkers of cardiovascular inflammation and endothelial activation. Fatigue was assessed using the fatigue severity scale (FSS), the visual analog fatigue scale, and a subscale of the Geriatric Depression Scale. RESULTS Mean (SD) age was 68.7 (11.2) years, and 63.9% were male. Of these, 55.6% showed fatigue on the FSS. Fatigued participants had higher disability scores (p = 0.02), higher total GDS scores (p = 0.02), and more commonly reported a history of depression (p = 0.04). 11C-PK11195 ligand binding in the white matter was not associated with any measure of fatigue. Serum CRP was significantly associated with average fatigue score on FSS (ρ = 0.48, p = 0.004); this association persisted when controlling for age, sex, disability score, and depression (β = 0.49, 95% CI (0.17, 2.26), p = 0.03). Blood biomarkers from the Olink panel showed no association with fatigue. CONCLUSION In symptomatic SVD patients, neuroinflammation, assessed with microglial marker 11C-PK11195, was not associated with fatigue. We found some evidence for a role of systematic inflammation, evidenced by an association between fatigue severity and raised CRP, but further studies are required to understand this relationship and inform whether it could be therapeutically modified to reduce fatigue severity. DATA ACCESS STATEMENT Data for this study are available from the corresponding author upon reasonable request.
Collapse
Affiliation(s)
- Amy A Jolly
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Robin B Brown
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Daniel J Tozer
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Young T Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Franklin I Aigbirhio
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - John T O’Brien
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
16
|
VanElzakker MB, Bues HF, Brusaferri L, Kim M, Saadi D, Ratai EM, Dougherty DD, Loggia ML. Neuroinflammation in post-acute sequelae of COVID-19 (PASC) as assessed by [ 11C]PBR28 PET correlates with vascular disease measures. Brain Behav Immun 2024; 119:713-723. [PMID: 38642615 PMCID: PMC11225883 DOI: 10.1016/j.bbi.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/28/2024] [Accepted: 04/16/2024] [Indexed: 04/22/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has triggered a consequential public health crisis of post-acute sequelae of COVID-19 (PASC), sometimes referred to as long COVID. The mechanisms of the heterogeneous persistent symptoms and signs that comprise PASC are under investigation, and several studies have pointed to the central nervous and vascular systems as being potential sites of dysfunction. In the current study, we recruited individuals with PASC with diverse symptoms, and examined the relationship between neuroinflammation and circulating markers of vascular dysfunction. We used [11C]PBR28 PET neuroimaging, a marker of neuroinflammation, to compare 12 PASC individuals versus 43 normative healthy controls. We found significantly increased neuroinflammation in PASC versus controls across a wide swath of brain regions including midcingulate and anterior cingulate cortex, corpus callosum, thalamus, basal ganglia, and at the boundaries of ventricles. We also collected and analyzed peripheral blood plasma from the PASC individuals and found significant positive correlations between neuroinflammation and several circulating analytes related to vascular dysfunction. These results suggest that an interaction between neuroinflammation and vascular health may contribute to common symptoms of PASC.
Collapse
Affiliation(s)
- Michael B VanElzakker
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; PolyBio Research Foundation, Medford, MA, USA.
| | - Hannah F Bues
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ludovica Brusaferri
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Computer Science And Informatics, School of Engineering, London South Bank University, London, UK
| | - Minhae Kim
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deena Saadi
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eva-Maria Ratai
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Darin D Dougherty
- Division of Neurotherapeutics, Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco L Loggia
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Barateau L, Krache A, Da Costa A, Lecendreux M, Chenini S, Arlicot N, Vourc'h P, Alonso M, Salabert AS, Beziat S, Jaussent I, Mariano-Goulart D, Payoux P, Dauvilliers Y. PET Study of Microglial Activation in Kleine-Levin Syndrome. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200263. [PMID: 38885456 PMCID: PMC11186701 DOI: 10.1212/nxi.0000000000200263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/10/2024] [Indexed: 06/20/2024]
Abstract
OBJECTIVES Kleine-Levin syndrome (KLS) is a rare recurrent hypersomnolence disorder associated with cognitive and behavioral disturbances, of unknown origin, but inflammatory mechanisms could be involved. We aimed to explore in vivo microglia activation using [18F]DPA-714 PET imaging in patients with KLS compared with controls, and during symptomatic vs asymptomatic periods. METHODS Patients with KLS and controls underwent a standardized clinical evaluation and PET imaging, using a radiolabeled ligand specific to the 18 kDa translocator protein. Images were processed on the PMOD (peripheral module) interface using a standard uptake value (SUV). Five regions of interest (ROIs) were analyzed: hypothalamus, thalamus, frontal area, cerebellum, and whole brain. SUV ratios (SUVr) were calculated by normalizing SUV with cerebellum uptake. RESULTS Images of 17 consecutive patients with KLS (7 during episodes, 10 out of episodes) and 14 controls were analyzed. We found no SUV/SUVr difference between KLS and controls, between patients in and out episodes in all ROIs, and no correlation between SUVr and episode duration at the time of PET scan. No association was found between SUVr and sex, disease duration, or orexin levels. DISCUSSION Our findings do not support the presence of neuroinflammation in KLS. Further research is needed to identify relevant biomarkers in KLS.
Collapse
Affiliation(s)
- Lucie Barateau
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| | - Anis Krache
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| | - Alexandre Da Costa
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| | - Michel Lecendreux
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| | - Sofiene Chenini
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| | - Nicolas Arlicot
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| | - Patrick Vourc'h
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| | - Mathieu Alonso
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| | - Anne-Sophie Salabert
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| | - Séverine Beziat
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| | - Isabelle Jaussent
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| | - Denis Mariano-Goulart
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| | - Pierre Payoux
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| | - Yves Dauvilliers
- From the Sleep-Wake Disorders Unit (L.B., S.C., Y.D.), Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (L.B., Y.D.), Montpellier; Institute of Neurosciences of Montpellier (L.B., S.B., I.J., Y.D.), University of Montpellier, INSERM; ToNIC (A.K., A.D.C., A.-S.S., P.P.), Toulouse NeuroImaging Center, UMR 1214, INSERM, Université Paul-Sabatier; Pediatric Sleep Centre (M.L.), Hospital Robert-Debré; National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome (M.L.), Paris; CHRU de Tours - UMR 1253 iBraiN (N.A., P.V.), Université de Tours, Inserm, Inserm CIC 1415; Radiopharmacy Department (M.A., A.-S.S.), CHU Toulouse; Department of Nuclear Medicine (D.M.-G.), CHU Montpellier; PhyMedExp (D.M.-G.), University of Montpellier, INSERM, CNRS; and Nuclear Medicine Department (P.P.), CHU Toulouse, France
| |
Collapse
|
18
|
Ottoy J, Kang MS, Tan JXM, Boone L, Vos de Wael R, Park BY, Bezgin G, Lussier FZ, Pascoal TA, Rahmouni N, Stevenson J, Fernandez Arias J, Therriault J, Hong SJ, Stefanovic B, McLaurin J, Soucy JP, Gauthier S, Bernhardt BC, Black SE, Rosa-Neto P, Goubran M. Tau follows principal axes of functional and structural brain organization in Alzheimer's disease. Nat Commun 2024; 15:5031. [PMID: 38866759 PMCID: PMC11169286 DOI: 10.1038/s41467-024-49300-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 05/24/2024] [Indexed: 06/14/2024] Open
Abstract
Alzheimer's disease (AD) is a brain network disorder where pathological proteins accumulate through networks and drive cognitive decline. Yet, the role of network connectivity in facilitating this accumulation remains unclear. Using in-vivo multimodal imaging, we show that the distribution of tau and reactive microglia in humans follows spatial patterns of connectivity variation, the so-called gradients of brain organization. Notably, less distinct connectivity patterns ("gradient contraction") are associated with cognitive decline in regions with greater tau, suggesting an interaction between reduced network differentiation and tau on cognition. Furthermore, by modeling tau in subject-specific gradient space, we demonstrate that tau accumulation in the frontoparietal and temporo-occipital cortices is associated with greater baseline tau within their functionally and structurally connected hubs, respectively. Our work unveils a role for both functional and structural brain organization in pathology accumulation in AD, and supports subject-specific gradient space as a promising tool to map disease progression.
Collapse
Affiliation(s)
- Julie Ottoy
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Min Su Kang
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | | | - Lyndon Boone
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Reinder Vos de Wael
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Bo-Yong Park
- Department of Data Science, Inha University, Incheon, Republic of Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea
| | - Gleb Bezgin
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
- Neuroinformatics for Personalized Medicine lab, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nesrine Rahmouni
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Jenna Stevenson
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Jaime Fernandez Arias
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Joseph Therriault
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Seok-Jun Hong
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Bojana Stefanovic
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - JoAnne McLaurin
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Jean-Paul Soucy
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Serge Gauthier
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Boris C Bernhardt
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Sandra E Black
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
- Department of Medicine (Division of Neurology), University of Toronto, Toronto, ON, Canada
| | - Pedro Rosa-Neto
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
- Translational Neuroimaging laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Maged Goubran
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Physical Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
19
|
Tseng CEJ, Canales C, Marcus RE, Parmar AJ, Hightower BG, Mullett JE, Makary MM, Tassone AU, Saro HK, Townsend PH, Birtwell K, Nowinski L, Thom RP, Palumbo ML, Keary C, Catana C, McDougle CJ, Hooker JM, Zürcher NR. In vivo translocator protein in females with autism spectrum disorder: a pilot study. Neuropsychopharmacology 2024; 49:1193-1201. [PMID: 38615126 PMCID: PMC11109261 DOI: 10.1038/s41386-024-01859-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/21/2024] [Accepted: 04/02/2024] [Indexed: 04/15/2024]
Abstract
Sex-based differences in the prevalence of autism spectrum disorder (ASD) are well-documented, with a male-to-female ratio of approximately 4:1. The clinical presentation of the core symptoms of ASD can also vary between sexes. Previously, positron emission tomography (PET) studies have identified alterations in the in vivo levels of translocator protein (TSPO)-a mitochondrial protein-in primarily or only male adults with ASD, with our group reporting lower TSPO relative to whole brain mean in males with ASD. However, whether in vivo TSPO levels are altered in females with ASD, specifically, is unknown. This is the first pilot study to measure in vivo TSPO in the brain in adult females with ASD using [11C]PBR28 PET-magnetic resonance imaging (MRI). Twelve adult females with ASD and 10 age- and TSPO genotype-matched controls (CON) completed one or two [11C]PBR28 PET-MRI scans. Females with ASD exhibited elevated [11C]PBR28 standardized uptake value ratio (SUVR) in the midcingulate cortex and splenium of the corpus callosum compared to CON. No brain area showed lower [11C]PBR28 SUVR in females with ASD compared to CON. Test-retest over several months showed stable [11C]PBR28 SUVR across time in both groups. Elevated regional [11C]PBR28 SUVR in females with ASD stand in stark contrast to our previous findings of lower regional [11C]PBR28 SUVR in males with ASD. Preliminary evidence of regionally elevated mitochondrial protein TSPO relative to whole brain mean in ASD females may reflect neuroimmuno-metabolic alterations specific to females with ASD.
Collapse
Affiliation(s)
- Chieh-En Jane Tseng
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Camila Canales
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Rachel E Marcus
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Anjali J Parmar
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Baileigh G Hightower
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Jennifer E Mullett
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
- Department of Pediatrics, Indiana University, Indianapolis, IN, USA
| | - Meena M Makary
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
- Systems and Biomedical Engineering Department, Faculty of Engineering, Cairo University, Cairo, Egypt
| | - Alison U Tassone
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Hannah K Saro
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Paige Hickey Townsend
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Kirstin Birtwell
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Lisa Nowinski
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Robyn P Thom
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Michelle L Palumbo
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Christopher Keary
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Ciprian Catana
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Christopher J McDougle
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Nicole R Zürcher
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA.
| |
Collapse
|
20
|
Brown RB, Tozer DJ, Loubière L, Harshfield EL, Hong YT, Fryer TD, Williams GB, Graves MJ, Aigbirhio FI, O'Brien JT, Markus HS. MINocyclinE to Reduce inflammation and blood-brain barrier leakage in small Vessel diseAse (MINERVA): A phase II, randomized, double-blind, placebo-controlled experimental medicine trial. Alzheimers Dement 2024; 20:3852-3863. [PMID: 38629936 PMCID: PMC11180856 DOI: 10.1002/alz.13830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/13/2024] [Accepted: 03/16/2024] [Indexed: 06/18/2024]
Abstract
INTRODUCTION Cerebral small vessel disease (SVD) is a common cause of stroke/vascular dementia with few effective treatments. Neuroinflammation and increased blood-brain barrier (BBB) permeability may influence pathogenesis. In rodent models, minocycline reduced inflammation/BBB permeability. We determined whether minocycline had a similar effect in patients with SVD. METHODS MINERVA was a single-center, phase II, randomized, double-blind, placebo-controlled trial. Forty-four participants with moderate-to-severe SVD took minocycline or placebo for 3 months. Co-primary outcomes were microglial signal (determined using 11C-PK11195 positron emission tomography) and BBB permeability (using dynamic contrast-enhanced MRI). RESULTS Forty-four participants were recruited between September 2019 and June 2022. Minocycline had no effect on 11C-PK11195 binding (relative risk [RR] 1.01, 95% confidence interval [CI] 0.98-1.04), or BBB permeability (RR 0.97, 95% CI 0.91-1.03). Serum inflammatory markers were not affected. DISCUSSION 11C-PK11195 binding and increased BBB permeability are present in SVD; minocycline did not reduce either process. Whether these pathophysiological mechanisms are disease-causing remains unclear. INTERNATIONAL CLINICAL TRIALS REGISTRY PORTAL IDENTIFIER ISRCTN15483452 HIGHLIGHTS: We found focal areas of increased microglial signal and increased blood-brain barrier permeability in patients with small vessel disease. Minocycline treatment was not associated with a change in these processes measured using advanced neuroimaging. Blood-brain barrier permeability was dynamic but MRI-derived measurements correlated well with CSF/serum albumin ratio. Advanced neuroimaging is a feasible outcome measure for mechanistic clinical trials.
Collapse
Affiliation(s)
- Robin B. Brown
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Daniel J. Tozer
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Laurence Loubière
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | | | - Young T. Hong
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
| | - Tim D. Fryer
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
| | - Guy B. Williams
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
| | - Martin J. Graves
- Department of RadiologyUniversity of CambridgeCambridgeCambridgeUK
| | - Franklin I. Aigbirhio
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
| | | | - Hugh S. Markus
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| |
Collapse
|
21
|
Qiu L, Jiang H, Cho K, Yu Y, Jones LA, Huang T, Perlmutter JS, Gropler RJ, Brier MR, Patti GJ, Benzinger TLS, Tu Z. Metabolite Study and Structural Authentication for the First-in-Human Use Sphingosine-1-phosphate Receptor 1 Radiotracer. ACS Chem Neurosci 2024; 15:1882-1892. [PMID: 38634759 PMCID: PMC11103254 DOI: 10.1021/acschemneuro.4c00077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024] Open
Abstract
The sphingosine-1-phosphate receptor 1 (S1PR1) radiotracer [11C]CS1P1 has shown promise in proof-of-concept PET imaging of neuroinflammation in multiple sclerosis (MS). Our HPLC radiometabolite analysis of human plasma samples collected during PET scans with [11C]CS1P1 detected a radiometabolite peak that is more lipophilic than [11C]CS1P1. Radiolabeled metabolites that cross the blood-brain barrier complicate quantitative modeling of neuroimaging tracers; thus, characterizing such radiometabolites is important. Here, we report our detailed investigation of the metabolite profile of [11C]CS1P1 in rats, nonhuman primates, and humans. CS1P1 is a fluorine-containing ligand that we labeled with C-11 or F-18 for preclinical studies; the brain uptake was similar for both radiotracers. The same lipophilic radiometabolite found in human studies also was observed in plasma samples of rats and NHPs for CS1P1 labeled with either C-11 or F-18. We characterized the metabolite in detail using rats after injection of the nonradioactive CS1P1. To authenticate the molecular structure of this radiometabolite, we injected rats with 8 mg/kg of CS1P1 to collect plasma for solvent extraction and HPLC injection, followed by LC/MS analysis of the same metabolite. The LC/MS data indicated in vivo mono-oxidation of CS1P1 produces the metabolite. Subsequently, we synthesized three different mono-oxidized derivatives of CS1P1 for further investigation. Comparing the retention times of the mono-oxidized derivatives with the metabolite observed in rats injected with CS1P1 identified the metabolite as N-oxide 1, also named TZ82121. The MS fragmentation pattern of N-oxide 1 also matched that of the major metabolite in rat plasma. To confirm that metabolite TZ82121 does not enter the brain, we radiosynthesized [18F]TZ82121 by the oxidation of [18F]FS1P1. Radio-HPLC analysis confirmed that [18F]TZ82121 matched the radiometabolite observed in rat plasma post injection of [18F]FS1P1. Furthermore, the acute biodistribution study in SD rats and PET brain imaging in a nonhuman primate showed that [18F]TZ82121 does not enter the rat or nonhuman primate brain. Consequently, we concluded that the major lipophilic radiometabolite N-oxide [11C]TZ82121, detected in human plasma post injection of [11C]CS1P1, does not enter the brain to confound quantitative PET data analysis. [11C]CS1P1 is a promising S1PR1 radiotracer for detecting S1PR1 expression in the CNS.
Collapse
Affiliation(s)
- Lin Qiu
- Department of Radiology, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
| | - Hao Jiang
- Department of Radiology, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
| | - Kevin Cho
- Center for Mass Spectrometry and Metabolic Tracing, Department of Chemistry, Department of Medicine, Washington University, Saint Louis, Missouri 63130, United States
| | - Yanbo Yu
- Department of Radiology, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
| | - Lynne A Jones
- Department of Radiology, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
| | - Tianyu Huang
- Department of Radiology, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
- Departments of Neurology, Neuroscience, Physical Therapy and Occupational Therapy, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
| | - Robert J Gropler
- Department of Radiology, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
| | - Matthew R Brier
- Departments of Neurology, Neuroscience, Physical Therapy and Occupational Therapy, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
| | - Gary J Patti
- Center for Mass Spectrometry and Metabolic Tracing, Department of Chemistry, Department of Medicine, Washington University, Saint Louis, Missouri 63130, United States
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
| |
Collapse
|
22
|
Shippy DC, Oliai SF, Ulland TK. Zinc utilization by microglia in Alzheimer's disease. J Biol Chem 2024; 300:107306. [PMID: 38648940 PMCID: PMC11103939 DOI: 10.1016/j.jbc.2024.107306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia defined by two key pathological characteristics in the brain, amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau. Microglia, the primary innate immune cells of the central nervous system (CNS), provide neuroprotection through Aβ and tau clearance but may also be neurotoxic by promoting neuroinflammation to exacerbate Aβ and tau pathogenesis in AD. Recent studies have demonstrated the importance of microglial utilization of nutrients and trace metals in controlling their activation and effector functions. Trace metals, such as zinc, have essential roles in brain health and immunity, and zinc dyshomeostasis has been implicated in AD pathogenesis. As a result of these advances, the mechanisms by which zinc homeostasis influences microglial-mediated neuroinflammation in AD is a topic of continuing interest since new strategies to treat AD are needed. Here, we review the roles of zinc in AD, including zinc activation of microglia, the associated neuroinflammatory response, and the application of these findings in new therapeutic strategies.
Collapse
Affiliation(s)
- Daniel C Shippy
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Sophia F Oliai
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Tyler K Ulland
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA; Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA.
| |
Collapse
|
23
|
Tenovuo O, Loane DJ. Neuroinflammation is a player in coma, but in which role? Brain 2024; 147:1121-1123. [PMID: 38574286 DOI: 10.1093/brain/awae082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 04/06/2024] Open
Abstract
This scientific commentary refers to ‘Neuroimmune activation is associated with neurological outcome in anoxic and traumatic coma’ by Sarton et al. (https://doi.org/10.1093/brain/awae045).
Collapse
Affiliation(s)
- Olli Tenovuo
- Department of Clinical Neurosciences, University of Turku, 20520 Turku, Finland
| | - David J Loane
- School of Biochemistry and Immunology, Trinity College Dublin, D02R590 Dublin, Ireland
| |
Collapse
|
24
|
Sarton B, Tauber C, Fridman E, Péran P, Riu B, Vinour H, David A, Geeraerts T, Bounes F, Minville V, Delmas C, Salabert AS, Albucher JF, Bataille B, Olivot JM, Cariou A, Naccache L, Payoux P, Schiff N, Silva S. Neuroimmune activation is associated with neurological outcome in anoxic and traumatic coma. Brain 2024; 147:1321-1330. [PMID: 38412555 PMCID: PMC10994537 DOI: 10.1093/brain/awae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 12/22/2023] [Accepted: 01/09/2024] [Indexed: 02/29/2024] Open
Abstract
The pathophysiological underpinnings of critically disrupted brain connectomes resulting in coma are poorly understood. Inflammation is potentially an important but still undervalued factor. Here, we present a first-in-human prospective study using the 18-kDa translocator protein (TSPO) radioligand 18F-DPA714 for PET imaging to allow in vivo neuroimmune activation quantification in patients with coma (n = 17) following either anoxia or traumatic brain injuries in comparison with age- and sex-matched controls. Our findings yielded novel evidence of an early inflammatory component predominantly located within key cortical and subcortical brain structures that are putatively implicated in consciousness emergence and maintenance after severe brain injury (i.e. mesocircuit and frontoparietal networks). We observed that traumatic and anoxic patients with coma have distinct neuroimmune activation profiles, both in terms of intensity and spatial distribution. Finally, we demonstrated that both the total amount and specific distribution of PET-measurable neuroinflammation within the brain mesocircuit were associated with the patient's recovery potential. We suggest that our results can be developed for use both as a new neuroprognostication tool and as a promising biometric to guide future clinical trials targeting glial activity very early after severe brain injury.
Collapse
Affiliation(s)
- Benjamine Sarton
- Critical Care Unit, University Teaching Hospital of Purpan, F-31059 Toulouse Cedex 9, France
- Toulouse NeuroImaging Center, Toulouse University, Inserm 1214, UPS, F-31300 Toulouse, France
| | - Clovis Tauber
- Imaging and Brain laboratory, UMRS Inserm U930, Université de Tours, F-37000 Tours, France
| | - Estéban Fridman
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA
| | - Patrice Péran
- Toulouse NeuroImaging Center, Toulouse University, Inserm 1214, UPS, F-31300 Toulouse, France
| | - Beatrice Riu
- Critical Care Unit, University Teaching Hospital of Purpan, F-31059 Toulouse Cedex 9, France
| | - Hélène Vinour
- Critical Care Unit, University Teaching Hospital of Purpan, F-31059 Toulouse Cedex 9, France
| | - Adrian David
- Critical Care Unit, University Teaching Hospital of Purpan, F-31059 Toulouse Cedex 9, France
| | - Thomas Geeraerts
- Neurocritical Care Unit, University Teaching Hospital of Purpan, F-31059 Toulouse Cedex 9, France
| | - Fanny Bounes
- Critical Care Unit, University Teaching Hospital of Rangueil, F-31400 Toulouse Cedex 9, France
| | - Vincent Minville
- Critical Care Unit, University Teaching Hospital of Rangueil, F-31400 Toulouse Cedex 9, France
| | - Clément Delmas
- Cardiology Department, University Teaching Hospital of Purpan, F-31059 Toulouse Cedex 9, France
| | - Anne-Sophie Salabert
- Toulouse NeuroImaging Center, Toulouse University, Inserm 1214, UPS, F-31300 Toulouse, France
| | - Jean François Albucher
- Neurology Department, University Teaching Hospital of Purpan, F-31059 Toulouse Cedex 9, France
| | - Benoit Bataille
- Critical Care Unit, Hôtel Dieu Hospital, F-11100 Narbonne, France
| | - Jean Marc Olivot
- Neurology Department, University Teaching Hospital of Purpan, F-31059 Toulouse Cedex 9, France
| | - Alain Cariou
- Critical Care Unit, APHP, Cochin Hospital, F-75014 Paris, France
| | - Lionel Naccache
- Institut du Cerveau et de la Moelle épinière, ICM, PICNIC Lab, F-75013 Paris, France
| | - Pierre Payoux
- Toulouse NeuroImaging Center, Toulouse University, Inserm 1214, UPS, F-31300 Toulouse, France
| | - Nicholas Schiff
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA
| | - Stein Silva
- Critical Care Unit, University Teaching Hospital of Purpan, F-31059 Toulouse Cedex 9, France
- Toulouse NeuroImaging Center, Toulouse University, Inserm 1214, UPS, F-31300 Toulouse, France
| |
Collapse
|
25
|
Jaisa-Aad M, Muñoz-Castro C, Healey MA, Hyman BT, Serrano-Pozo A. Characterization of monoamine oxidase-B (MAO-B) as a biomarker of reactive astrogliosis in Alzheimer's disease and related dementias. Acta Neuropathol 2024; 147:66. [PMID: 38568475 PMCID: PMC10991006 DOI: 10.1007/s00401-024-02712-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 02/03/2024] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
Reactive astrogliosis accompanies the two neuropathological hallmarks of Alzheimer's disease (AD)-Aβ plaques and neurofibrillary tangles-and parallels neurodegeneration in AD and AD-related dementias (ADRD). Thus, there is growing interest in developing imaging and fluid biomarkers of reactive astrogliosis for AD/ADRD diagnosis and prognostication. Monoamine oxidase-B (MAO-B) is emerging as a target for PET imaging radiotracers of reactive astrogliosis. However, a thorough characterization of MAO-B expression in postmortem control and AD/ADRD brains is lacking. We sought to: (1) identify the primary cell type(s) expressing MAO-B in control and AD brains; (2) quantify MAO-B immunoreactivity in multiple brain regions of control and AD donors as a proxy for PET radiotracer uptake; (3) correlate MAO-B level with local AD neuropathological changes, reactive glia, and cortical atrophy; (4) determine whether the MAOB rs1799836 SNP genotype impacts MAO-B expression level; (5) compare MAO-B immunoreactivity across AD/ADRD, including Lewy body diseases (LBD) and frontotemporal lobar degenerations with tau (FTLD-Tau) and TDP-43 (FTLD-TDP). We found that MAO-B is mainly expressed by subpial and perivascular cortical astrocytes as well as by fibrous white matter astrocytes in control brains, whereas in AD brains, MAO-B is significantly upregulated by both cortical reactive astrocytes and white matter astrocytes across temporal, frontal, and occipital lobes. By contrast, MAO-B expression level was unchanged and lowest in cerebellum. Cortical MAO-B expression was independently associated with cortical atrophy and local measures of reactive astrocytes and microglia, and significantly increased in reactive astrocytes surrounding Thioflavin-S+ dense-core Aβ plaques. MAO-B expression was not affected by the MAOB rs1799836 SNP genotype. MAO-B expression was also significantly increased in the frontal cortex and white matter of donors with corticobasal degeneration, Pick's disease, and FTLD-TDP, but not in LBD or progressive supranuclear palsy. These findings support ongoing efforts to develop MAO-B-based PET radiotracers to image reactive astrogliosis in AD/ADRD.
Collapse
Affiliation(s)
- Methasit Jaisa-Aad
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Clara Muñoz-Castro
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Molly A Healey
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
26
|
Ibrahim W, An J, Yang Y, Cosgrove KP, Matuskey D. Does seasonal variation affect the neuroimmune system? A retrospective [ 11C]PBR28 PET study in healthy individuals. Neurosci Lett 2024; 828:137766. [PMID: 38583505 PMCID: PMC11073647 DOI: 10.1016/j.neulet.2024.137766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/31/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
INTRODUCTION The neuroimmune system performs a wide range of functions in the brain and the central nervous system. The microglial translocator protein (TSPO) has an established role as a cell marker in identification of the neuroimmune system. Previously, human studies have shown TSPO differences in neuropsychiatric disorders. Seasonal variability has also been demonstrated in multiple systems of healthy individuals. Therefore, in this study, we attempt to understand whether seasonal changes affect brain TSPO levels using [11C]PBR28 positron emission tomography (PET) imaging. METHODS 46 healthy subjects (mean age ± SD = 32.5 ± 10); sex (M/F) = 32/14)) underwent PET imaging with [11C]PBR28 in a retrospectively conducted analysis. All PET scans were performed on the HRRT scanner. Volume of distribution (VT) values were generated for cortical and subcortical regions and the cerebellum. Spring/summer months were defined as March to August while fall/winter months were defined as September to February and were compared through 2-tailed t-tests (SciPy library v.1.10.1 and Pinguoin library on Python v.3.8.8). Average daylight hours and temperature in New Haven, CT were obtained online (www.wunderground.com) and compared to VT with Spearman's correlations. RESULTS There were no significant differences observed between the TSPO levels of spring/summer and fall/winter months in the brain (t = 0.52, p = 0.61). Additional analysis on all individual brain regions also indicated non-significance. Likewise, no significant correlations were found between TSPO levels in the whole brain and brain regions against daylight hours (ρ= 0.05, p = 0.74), temperature (ρ = 0.04, p = 0.81), or month (ρ = 0.08, p = 0.60). Controlling TSPO gene polymorphisms and other variables had no significant effect on the outcome. CONCLUSION To the best of our knowledge, this is the first human study to investigate seasonal changes in TSPO expression. Our results can be interpreted as the lack of seasonal variability in the neuroimmune system, but important limitations include high interindividual variability, test-retest variability, specificity of the tracer, and a limited sample size. Limitations notwithstanding, our results conclude that TSPO levels in the brain are not impacted by light and temperature changes in different seasons.
Collapse
Affiliation(s)
- Waleed Ibrahim
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Jeonghyun An
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Yanghong Yang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Kelly P. Cosgrove
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - David Matuskey
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
27
|
Rossano SM, Johnson AS, Smith A, Ziaggi G, Roetman A, Guzman D, Okafor A, Klein J, Tomljanovic Z, Stern Y, Brickman AM, Lee S, Kreisl WC, Lao P. Microglia measured by TSPO PET are associated with Alzheimer's disease pathology and mediate key steps in a disease progression model. Alzheimers Dement 2024; 20:2397-2407. [PMID: 38298155 PMCID: PMC11032543 DOI: 10.1002/alz.13699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/30/2023] [Accepted: 12/18/2023] [Indexed: 02/02/2024]
Abstract
INTRODUCTION Evidence suggests microglial activation precedes regional tau and neurodegeneration in Alzheimer's disease (AD). We characterized microglia with translocator protein (TSPO) positron emission tomography (PET) within an AD progression model where global amyloid beta (Aβ) precedes local tau and neurodegeneration, resulting in cognitive impairment. METHODS Florbetaben, PBR28, and MK-6240 PET, T1 magnetic resonance imaging, and cognitive measures were performed in 19 cognitively unimpaired older adults and 22 patients with mild cognitive impairment or mild AD to examine associations among microglia activation, Aβ, tau, and cognition, adjusting for neurodegeneration. Mediation analyses evaluated the possible role of microglial activation along the AD progression model. RESULTS Higher PBR28 uptake was associated with higher Aβ, higher tau, and lower MMSE score, independent of neurodegeneration. PBR28 mediated associations between tau in early and middle Braak stages, between tau and neurodegeneration, and between neurodegeneration and cognition. DISCUSSION Microglia are associated with AD pathology and cognition and may mediate relationships between subsequent steps in AD progression.
Collapse
Affiliation(s)
- Samantha M. Rossano
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Aubrey S. Johnson
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Anna Smith
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Galen Ziaggi
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Andrew Roetman
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Diana Guzman
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Amarachukwu Okafor
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Julia Klein
- Department of Anesthesiology and Perioperative MedicineUniversity of California Los Angeles HealthLos AngelesCaliforniaUSA
| | - Zeljko Tomljanovic
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Yaakov Stern
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Adam M. Brickman
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Seonjoo Lee
- Department of Psychiatry and BiostatisticsColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - William C. Kreisl
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Patrick Lao
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
28
|
Alzghool OM, Aarnio R, Helin JS, Wahlroos S, Keller T, Matilainen M, Solis J, Danon JJ, Kassiou M, Snellman A, Solin O, Rinne JO, Haaparanta-Solin M. Glial reactivity in a mouse model of beta-amyloid deposition assessed by PET imaging of P2X7 receptor and TSPO using [ 11C]SMW139 and [ 18F]F-DPA. EJNMMI Res 2024; 14:25. [PMID: 38446249 PMCID: PMC10917722 DOI: 10.1186/s13550-024-01085-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/22/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND P2X7 receptor has emerged as a potentially superior PET imaging marker to TSPO, the gold standard for imaging glial reactivity. [11C]SMW139 is the most recently developed radiotracer to image P2X7 receptor. The aim of this study was to image reactive glia in the APP/PS1-21 transgenic (TG) mouse model of Aβ deposition longitudinally using [11C]SMW139 targeting P2X7 receptor and to compare tracer uptake to that of [18F]F-DPA targeting TSPO at the final imaging time point. TG and wild type (WT) mice underwent longitudinal in vivo PET imaging using [11C]SMW139 at 5, 8, 11, and 14 months, followed by [18F]F-DPA PET scan only at 14 months. In vivo imaging results were verified by ex vivo brain autoradiography, immunohistochemical staining, and analysis of [11C]SMW139 unmetabolized fraction in TG and WT mice. RESULTS Longitudinal change in [11C]SMW139 standardized uptake values (SUVs) showed no statistically significant increase in the neocortex and hippocampus of TG or WT mice, which was consistent with findings from ex vivo brain autoradiography. Significantly higher [18F]F-DPA SUVs were observed in brain regions of TG compared to WT mice. Quantified P2X7-positive staining in the cortex and thalamus of TG mice showed a minor increase in receptor expression with ageing, while TSPO-positive staining in the same regions showed a more robust increase in expression in TG mice as they aged. [11C]SMW139 was rapidly metabolized in mice, with 33% of unmetabolized fraction in plasma and 29% in brain homogenates 30 min after injection. CONCLUSIONS [11C]SMW139, which has a lower affinity for the rodent P2X7 receptor than the human version of the receptor, was unable to image the low expression of P2X7 receptor in the APP/PS1-21 mouse model. Additionally, the rapid metabolism of [11C]SMW139 in mice and the presence of several brain-penetrating radiometabolites significantly impacted the analysis of in vivo PET signal of the tracer. Finally, [18F]F-DPA targeting TSPO was more suitable for imaging reactive glia and neuroinflammatory processes in the APP/PS1-21 mouse model, based on the findings presented in this study and previous studies with this mouse model.
Collapse
Affiliation(s)
- Obada M Alzghool
- PET Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland.
- Medicity Research Laboratory, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland.
- Drug Research Doctoral Programme, University of Turku, Turku, Finland.
- Turku University Hospital, Turku PET Centre, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
| | - Richard Aarnio
- PET Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland
- Medicity Research Laboratory, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland
- Drug Research Doctoral Programme, University of Turku, Turku, Finland
| | - Jatta S Helin
- PET Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland
- Medicity Research Laboratory, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland
| | - Saara Wahlroos
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Thomas Keller
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Markus Matilainen
- Turku University Hospital, Turku PET Centre, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Junel Solis
- Turku BioImaging, Åbo Akademi University and University of Turku, Turku, Finland
| | - Jonathan J Danon
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Anniina Snellman
- PET Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland
| | - Olof Solin
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland
- Department of Chemistry, University of Turku, Henrikinkatu 2, 20500, Turku, Finland
- Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Juha O Rinne
- Turku University Hospital, Turku PET Centre, Kiinamyllynkatu 4-8, 20520, Turku, Finland
- Department of Neurology, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Merja Haaparanta-Solin
- PET Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland
- Medicity Research Laboratory, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland
| |
Collapse
|
29
|
Biechele G, Rauchmann BS, Janowitz D, Buerger K, Franzmeier N, Weidinger E, Guersel S, Schuster S, Finze A, Harris S, Lindner S, Albert NL, Wetzel C, Rupprecht R, Rominger A, Palleis C, Katzdobler S, Burow L, Kurz C, Zaganjori M, Trappmann LK, Goldhardt O, Grimmer T, Haeckert J, Keeser D, Stoecklein S, Morenas-Rodriguez E, Bartenstein P, Levin J, Höglinger GU, Simons M, Perneczky R, Brendel M. Associations between sex, body mass index and the individual microglial response in Alzheimer's disease. J Neuroinflammation 2024; 21:30. [PMID: 38263017 PMCID: PMC10804830 DOI: 10.1186/s12974-024-03020-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/11/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVES 18-kDa translocator protein position-emission-tomography (TSPO-PET) imaging emerged for in vivo assessment of neuroinflammation in Alzheimer's disease (AD) research. Sex and obesity effects on TSPO-PET binding have been reported for cognitively normal humans (CN), but such effects have not yet been systematically evaluated in patients with AD. Thus, we aimed to investigate the impact of sex and obesity on the relationship between β-amyloid-accumulation and microglial activation in AD. METHODS 49 patients with AD (29 females, all Aβ-positive) and 15 Aβ-negative CN (8 female) underwent TSPO-PET ([18F]GE-180) and β-amyloid-PET ([18F]flutemetamol) imaging. In 24 patients with AD (14 females), tau-PET ([18F]PI-2620) was additionally available. The brain was parcellated into 218 cortical regions and standardized-uptake-value-ratios (SUVr, cerebellar reference) were calculated. Per region and tracer, the regional increase of PET SUVr (z-score) was calculated for AD against CN. The regression derived linear effect of regional Aβ-PET on TSPO-PET was used to determine the Aβ-plaque-dependent microglial response (slope) and the Aβ-plaque-independent microglial response (intercept) at the individual patient level. All read-outs were compared between sexes and tested for a moderation effect of sex on associations with body mass index (BMI). RESULTS In AD, females showed higher mean cortical TSPO-PET z-scores (0.91 ± 0.49; males 0.30 ± 0.75; p = 0.002), while Aβ-PET z-scores were similar. The Aβ-plaque-independent microglial response was stronger in females with AD (+ 0.37 ± 0.38; males with AD - 0.33 ± 0.87; p = 0.006), pronounced at the prodromal stage. On the contrary, the Aβ-plaque-dependent microglial response was not different between sexes. The Aβ-plaque-independent microglial response was significantly associated with tau-PET in females (Braak-II regions: r = 0.757, p = 0.003), but not in males. BMI and the Aβ-plaque-independent microglial response were significantly associated in females (r = 0.44, p = 0.018) but not in males (BMI*sex interaction: F(3,52) = 3.077, p = 0.005). CONCLUSION While microglia response to fibrillar Aβ is similar between sexes, women with AD show a stronger Aβ-plaque-independent microglia response. This sex difference in Aβ-independent microglial activation may be associated with tau accumulation. BMI is positively associated with the Aβ-plaque-independent microglia response in females with AD but not in males, indicating that sex and obesity need to be considered when studying neuroinflammation in AD.
Collapse
Affiliation(s)
- Gloria Biechele
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
- Institute of Neuroradiology, LMU University Hospital, LMU Munich, Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Gothenburg, Sweden
| | - Endy Weidinger
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Selim Guersel
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Schuster
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Anika Finze
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Stefanie Harris
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Christian Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- Department of Nuclear Medicine, University of Bern, Inselspital, Bern, Switzerland
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sabrina Katzdobler
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lena Burow
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Carolin Kurz
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Mirlind Zaganjori
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lena-Katharina Trappmann
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Oliver Goldhardt
- Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Jan Haeckert
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Daniel Keeser
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sophia Stoecklein
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Neuronal Cell Biology, TU Munich, Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
30
|
Malpetti M, Franzmeier N, Brendel M. PET Imaging to Measure Neuroinflammation In Vivo. Methods Mol Biol 2024; 2785:177-193. [PMID: 38427195 DOI: 10.1007/978-1-0716-3774-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
This paper provides an overview of the role of neuroinflammation in Alzheimer's disease and other neurodegenerative diseases, highlighting the potential of anti-inflammatory treatments to slow or prevent decline. This research focuses on the use of positron emission tomography (PET) imaging to visualize and quantify molecular brain changes in patients, specifically microglial activation and reactive astrogliosis. We discuss the development and application of several PET radioligands, including first-generation ligands like PK11195 and Ro5-4864, as well as second- and third-generation ligands such as [11C]PBR28, [18F]DPA-714, [18F]GE-180, and [11C]ER176. These ligands target the 18-kDa translocator protein (TSPO), which is overexpressed in activated microglia and upregulated in astrocytes. We also address the limitations of these ligands, such as low brain uptake, poor penetration of the blood-brain barrier, short half-life, and variable kinetic behavior. Furthermore, we demonstrate the impact of genetic polymorphisms on ligand binding.
Collapse
Affiliation(s)
- Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal and Gothenburg, Sweden
| | - Matthias Brendel
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| |
Collapse
|
31
|
Ottoy J, De Picker L, Kang MS. Microglial Positron Emission Tomography Imaging In Vivo : Positron Emission Tomography Radioligands: Utility in Research and Clinical Practice. ADVANCES IN NEUROBIOLOGY 2024; 37:579-589. [PMID: 39207714 DOI: 10.1007/978-3-031-55529-9_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia, the resident immune cells of the central nervous system (CNS) play a key role in regulating and maintaining homeostasis in the brain. However, the CNS is also vulnerable to infections and inflammatory processes. In response to CNS perturbations, microglia become reactive, notably with expression of the translocator protein (TSPO), primarily on their outer mitochondrial membrane. Despite TSPO being commonly used as a marker for microglia, it is also present in other cell types such as astrocytes. Positron emission tomography (PET) ligands that target the TSPO enable the noninvasive detection and quantification of glial reactivity. While some limitations were raised, TSPO PET remains an attractive biomarker of CNS infection and inflammation. This book chapter delves into the development and application of microglial PET imaging with a focus on the TSPO PET. First, we provide an overview of the evolution of TSPO PET radioligands from first-generation to second-generation ligands and their applications in studying neuroinflammation (or CNS inflammation). Subsequently, we discuss the limitations and challenges associated with TSPO PET. Then we go on to explore non-TSPO targets for microglial PET imaging. Finally, we conclude with future directions for research and clinical practice in this field.
Collapse
Affiliation(s)
- Julie Ottoy
- Dr. Sandra E. Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Livia De Picker
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium
- University Psychiatric Hospital Campus Duffel, Duffel, Belgium
| | - Min Su Kang
- Dr. Sandra E. Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
Kipp M. Astrocytes: Lessons Learned from the Cuprizone Model. Int J Mol Sci 2023; 24:16420. [PMID: 38003609 PMCID: PMC10671869 DOI: 10.3390/ijms242216420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
A diverse array of neurological and psychiatric disorders, including multiple sclerosis, Alzheimer's disease, and schizophrenia, exhibit distinct myelin abnormalities at both the molecular and histological levels. These aberrations are closely linked to dysfunction of oligodendrocytes and alterations in myelin structure, which may be pivotal factors contributing to the disconnection of brain regions and the resulting characteristic clinical impairments observed in these conditions. Astrocytes, which significantly outnumber neurons in the central nervous system by a five-to-one ratio, play indispensable roles in the development, maintenance, and overall well-being of neurons and oligodendrocytes. Consequently, they emerge as potential key players in the onset and progression of a myriad of neurological and psychiatric disorders. Furthermore, targeting astrocytes represents a promising avenue for therapeutic intervention in such disorders. To gain deeper insights into the functions of astrocytes in the context of myelin-related disorders, it is imperative to employ appropriate in vivo models that faithfully recapitulate specific aspects of complex human diseases in a reliable and reproducible manner. One such model is the cuprizone model, wherein metabolic dysfunction in oligodendrocytes initiates an early response involving microglia and astrocyte activation, culminating in multifocal demyelination. Remarkably, following the cessation of cuprizone intoxication, a spontaneous process of endogenous remyelination occurs. In this review article, we provide a historical overview of studies investigating the responses and putative functions of astrocytes in the cuprizone model. Following that, we list previously published works that illuminate various aspects of the biology and function of astrocytes in this multiple sclerosis model. Some of the studies are discussed in more detail in the context of astrocyte biology and pathology. Our objective is twofold: to provide an invaluable overview of this burgeoning field, and, more importantly, to inspire fellow researchers to embark on experimental investigations to elucidate the multifaceted functions of this pivotal glial cell subpopulation.
Collapse
Affiliation(s)
- Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
33
|
Pradhan AK, Neumüller T, Klug C, Fuchs S, Schlegel M, Ballmann M, Tartler KJ, Pianos A, Garcia MS, Liere P, Schumacher M, Kreuzer M, Rupprecht R, Rammes G. Chronic administration of XBD173 ameliorates cognitive deficits and neuropathology via 18 kDa translocator protein (TSPO) in a mouse model of Alzheimer's disease. Transl Psychiatry 2023; 13:332. [PMID: 37891168 PMCID: PMC10611770 DOI: 10.1038/s41398-023-02630-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of β-amyloid peptide (Aβ). It affects cognition and leads to memory impairment. The mitochondrial translocator protein (TSPO) plays an essential role in maintaining mitochondrial homeostasis and has been implicated in several neuronal disorders or neuronal injuries. Ligands targeting the mitochondrial translocator protein (18 kDa), promote neurosteroidogenesis and may be neuroprotective. To study whether the TSPO ligand XBD173 may exert early neuroprotective effects in AD pathology we investigated the impact of XBD173 on amyloid toxicity and neuroplasticity in mouse models of AD. We show that XBD173 (emapunil), via neurosteroid-mediated signaling and delta subunit-containing GABAA receptors, prevents the neurotoxic effect of Aβ on long-term potentiation (CA1-LTP) in the hippocampus and prevents the loss of spines. Chronic but not acute administration of XBD173 ameliorates spatial learning deficits in transgenic AD mice with arctic mutation (ArcAβ). The heterozygous TSPO-knockout crossed with the transgenic arctic mutation model of AD mice (het TSPOKO X ArcAβ) treated with XBD173 does not show this improvement in spatial learning suggesting TSPO is needed for procognitive effects of XBD173. The neuroprotective profile of XBD173 in AD pathology is further supported by a reduction in plaques and soluble Aβ levels in the cortex, increased synthesis of neurosteroids, rescued spine density, reduction of complement protein C1q deposits, and reduced astrocytic phagocytosis of functional synapses both in the hippocampus and cortex. Our findings suggest that XBD173 may exert therapeutic effects via TSPO in a mouse model of AD.
Collapse
Affiliation(s)
- Arpit Kumar Pradhan
- Klinik für Anaesthesiologie und Intensivmedizin der Technischen Universität München, Klinikum rechts der Isar, Munich, Germany.
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität München, Martinsried, Germany.
| | - Tatjana Neumüller
- Klinik für Anaesthesiologie und Intensivmedizin der Technischen Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Claudia Klug
- Klinik für Anaesthesiologie und Intensivmedizin der Technischen Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Severin Fuchs
- Klinik für Anaesthesiologie und Intensivmedizin der Technischen Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Martin Schlegel
- Klinik für Anaesthesiologie und Intensivmedizin der Technischen Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Markus Ballmann
- Klinik für Anaesthesiologie und Intensivmedizin der Technischen Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Katharina Johanna Tartler
- Klinik für Anaesthesiologie und Intensivmedizin der Technischen Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Antoine Pianos
- U1195 Inserm and University Paris-Saclay, 80 rue du Général Leclerc, Le Kremlin-Bicêtre, 94276, France
| | - Maria Sanchez Garcia
- U1195 Inserm and University Paris-Saclay, 80 rue du Général Leclerc, Le Kremlin-Bicêtre, 94276, France
| | - Philippe Liere
- U1195 Inserm and University Paris-Saclay, 80 rue du Général Leclerc, Le Kremlin-Bicêtre, 94276, France
| | - Michael Schumacher
- U1195 Inserm and University Paris-Saclay, 80 rue du Général Leclerc, Le Kremlin-Bicêtre, 94276, France
| | - Matthias Kreuzer
- Klinik für Anaesthesiologie und Intensivmedizin der Technischen Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University Regensburg, Regensburg, Germany
| | - Gerhard Rammes
- Klinik für Anaesthesiologie und Intensivmedizin der Technischen Universität München, Klinikum rechts der Isar, Munich, Germany
| |
Collapse
|
34
|
Orbe EB, Benros ME. Immunological Biomarkers as Predictors of Treatment Response in Psychotic Disorders. J Pers Med 2023; 13:1382. [PMID: 37763150 PMCID: PMC10532612 DOI: 10.3390/jpm13091382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/05/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
Psychotic disorders, notably schizophrenia, impose a detrimental burden on both an individual and a societal level. The mechanisms leading to psychotic disorders are multifaceted, with genetics and environmental factors playing major roles. Increasing evidence additionally implicates neuro-inflammatory processes within at least a subgroup of patients with psychosis. While numerous studies have investigated anti-inflammatory add-on treatments to current antipsychotics, the exploration of immunological biomarkers as a predictor of treatment response remains limited. This review outlines the current evidence from trials exploring the potential of baseline inflammatory biomarkers as predictors of the treatment effect of anti-inflammatory drugs as add-ons to antipsychotics and of antipsychotics alone. Several of the studies have found correlations between baseline immunological biomarkers and treatment response; however, only a few studies incorporated baseline biomarkers as a primary endpoint, and the findings thus need to be interpreted with caution. Our review emphasizes the need for additional research on the potential of repurposing anti-inflammatory drugs while utilizing baseline inflammatory biomarkers as a predictor of treatment response and to identify subgroups of individuals with psychotic disorders where add-on treatment with immunomodulating agents would be warranted. Future studies investigating the correlation between baseline inflammatory markers and treatment responses can pave the way for personalized medicine approaches in psychiatry centred around biomarkers such as specific baseline inflammatory biomarkers in psychotic disorders.
Collapse
Affiliation(s)
- Elif Bayram Orbe
- Copenhagen Research Centre for Biological and Precision Psychiatry, Mental Health Centre Copenhagen, Copenhagen University Hospital, 2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Michael Eriksen Benros
- Copenhagen Research Centre for Biological and Precision Psychiatry, Mental Health Centre Copenhagen, Copenhagen University Hospital, 2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| |
Collapse
|
35
|
Nutma E, Fancy N, Weinert M, Tsartsalis S, Marzin MC, Muirhead RCJ, Falk I, Breur M, de Bruin J, Hollaus D, Pieterman R, Anink J, Story D, Chandran S, Tang J, Trolese MC, Saito T, Saido TC, Wiltshire KH, Beltran-Lobo P, Phillips A, Antel J, Healy L, Dorion MF, Galloway DA, Benoit RY, Amossé Q, Ceyzériat K, Badina AM, Kövari E, Bendotti C, Aronica E, Radulescu CI, Wong JH, Barron AM, Smith AM, Barnes SJ, Hampton DW, van der Valk P, Jacobson S, Howell OW, Baker D, Kipp M, Kaddatz H, Tournier BB, Millet P, Matthews PM, Moore CS, Amor S, Owen DR. Translocator protein is a marker of activated microglia in rodent models but not human neurodegenerative diseases. Nat Commun 2023; 14:5247. [PMID: 37640701 PMCID: PMC10462763 DOI: 10.1038/s41467-023-40937-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023] Open
Abstract
Microglial activation plays central roles in neuroinflammatory and neurodegenerative diseases. Positron emission tomography (PET) targeting 18 kDa Translocator Protein (TSPO) is widely used for localising inflammation in vivo, but its quantitative interpretation remains uncertain. We show that TSPO expression increases in activated microglia in mouse brain disease models but does not change in a non-human primate disease model or in common neurodegenerative and neuroinflammatory human diseases. We describe genetic divergence in the TSPO gene promoter, consistent with the hypothesis that the increase in TSPO expression in activated myeloid cells depends on the transcription factor AP1 and is unique to a subset of rodent species within the Muroidea superfamily. Finally, we identify LCP2 and TFEC as potential markers of microglial activation in humans. These data emphasise that TSPO expression in human myeloid cells is related to different phenomena than in mice, and that TSPO-PET signals in humans reflect the density of inflammatory cells rather than activation state.
Collapse
Affiliation(s)
- Erik Nutma
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
- Department of Neurobiology and Aging, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Nurun Fancy
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Maria Weinert
- Department of Brain Sciences, Imperial College London, London, UK
| | - Stergios Tsartsalis
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Manuel C Marzin
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - Robert C J Muirhead
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Irene Falk
- Viral Immunology Section, NIH, Bethesda, MD, USA
- Flow and Imaging Cytometry Core Facility, NIH, Bethesda, MD, USA
| | - Marjolein Breur
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - Joy de Bruin
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - Robin Pieterman
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - Jasper Anink
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - David Story
- UK Dementia Research Institute at Edinburgh, Edinburgh, UK
| | | | - Jiabin Tang
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Maria C Trolese
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako-shi, Saitama, Japan
| | - Takaomi C Saido
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University, Nagoya, Japan
| | | | - Paula Beltran-Lobo
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alexandra Phillips
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Jack Antel
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Luke Healy
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Marie-France Dorion
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, Canada
| | - Dylan A Galloway
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, Canada
| | - Rochelle Y Benoit
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, Canada
| | - Quentin Amossé
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Kelly Ceyzériat
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | | | - Enikö Kövari
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Caterina Bendotti
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Carola I Radulescu
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Jia Hui Wong
- Neurobiology of Aging and Disease Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Anna M Barron
- Neurobiology of Aging and Disease Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Amy M Smith
- UK Dementia Research Institute at Imperial College London, London, UK
- Centre for Brain Research and Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Samuel J Barnes
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | | | - Paul van der Valk
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | | | - Owain W Howell
- Institute of Life Science (ILS), Swansea University Medical School, Swansea, UK
| | - David Baker
- Department of Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, UK
| | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, 18057, Rostock, Germany
| | - Hannes Kaddatz
- Institute of Anatomy, Rostock University Medical Center, 18057, Rostock, Germany
| | | | - Philippe Millet
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
- Division of Adult Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Craig S Moore
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, Canada
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands.
- Department of Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, UK.
- Institute of Anatomy, Rostock University Medical Center, 18057, Rostock, Germany.
| | - David R Owen
- Department of Brain Sciences, Imperial College London, London, UK.
- UK Dementia Research Institute at Imperial College London, London, UK.
| |
Collapse
|
36
|
Mrdjen D, Amouzgar M, Cannon B, Liu C, Spence A, McCaffrey E, Bharadwaj A, Tebaykin D, Bukhari S, Hartmann FJ, Kagel A, Vijayaragavan K, Oliveria JP, Yakabi K, Serrano GE, Corrada MM, Kawas CH, Camacho C, Bosse M, Tibshirani R, Beach TG, Angelo M, Montine T, Bendall SC. Spatial proteomics reveals human microglial states shaped by anatomy and neuropathology. RESEARCH SQUARE 2023:rs.3.rs-2987263. [PMID: 37398389 PMCID: PMC10312937 DOI: 10.21203/rs.3.rs-2987263/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Microglia are implicated in aging, neurodegeneration, and Alzheimer's disease (AD). Traditional, low-plex, imaging methods fall short of capturing in situ cellular states and interactions in the human brain. We utilized Multiplexed Ion Beam Imaging (MIBI) and data-driven analysis to spatially map proteomic cellular states and niches in healthy human brain, identifying a spectrum of microglial profiles, called the microglial state continuum (MSC). The MSC ranged from senescent-like to active proteomic states that were skewed across large brain regions and compartmentalized locally according to their immediate microenvironment. While more active microglial states were proximal to amyloid plaques, globally, microglia significantly shifted towards a, presumably, dysfunctional low MSC in the AD hippocampus, as confirmed in an independent cohort (n=26). This provides an in situ single cell framework for mapping human microglial states along a continuous, shifting existence that is differentially enriched between healthy brain regions and disease, reinforcing differential microglial functions overall.
Collapse
Affiliation(s)
- Dunja Mrdjen
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Meelad Amouzgar
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Bryan Cannon
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Candace Liu
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Angie Spence
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Erin McCaffrey
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Anusha Bharadwaj
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Dmitry Tebaykin
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Syed Bukhari
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Felix J. Hartmann
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
- Systems Immunology and Single-Cell Biology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Adam Kagel
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Kausalia Vijayaragavan
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - John Paul Oliveria
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Koya Yakabi
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | | | - Maria M. Corrada
- Department of Neurology, University of California, Irvine, 9269, CA, USA
| | - Claudia H. Kawas
- Department of Neurology, University of California, Irvine, 9269, CA, USA
| | - Christine Camacho
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Marc Bosse
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Robert Tibshirani
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Thomas G. Beach
- Banner Sun Health Research Institute, Sun City, 85351, AZ, USA
| | - Michael Angelo
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Thomas Montine
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| | - Sean C. Bendall
- Department of Pathology, Stanford University, School of Medicine, Palo Alto 94304, CA, USA
| |
Collapse
|