1
|
Zeiss CJ, Huttner A, Nairn AC, Arnsten A, Datta D, Strittmatter SM, Wyk BV, Duque A. The neuropathologic basis for translational biomarker development in the macaque model of late-onset Alzheimer's disease. J Alzheimers Dis 2025:13872877251323787. [PMID: 40095666 DOI: 10.1177/13872877251323787] [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] [Indexed: 03/19/2025]
Abstract
BackgroundAccurate placement of the macaque within the Alzheimer's disease (AD) research framework is essential to discover early-stage predictive biomarkers.ObjectiveTo assess utility of the aging macaque in advancing translational biomarker development for preclinical AD, we evaluated relative signal strength of comparable neuropathologic phenomena in macaques and patients.MethodsWe compared pathology in patient and macaque formalin-fixed paraffin embedded (FFPE) tissues using identical criteria. We quantified expression of amyloid-β (Aβ), pTau, and inflammatory and senescence markers across species. Distribution of AD-relevant markers were compared in FFPE and perfused frozen macaque brain to assess expression of labile proteins that could inform in-life fluid biomarkers.ResultsAβ pathology in macaques closely approximated patient pathology. Complex plaque composition in macaques implied significant disruption of synaptic connectivity. In FFPE tissue, pretangle pTau immunoreactivity placed the macaque in Braak Stage 1b. In perfused frozen tissue, soluble pTau distribution approximated Braak Stage III-IV. In macaque, Aβ, pTau, and acetylcholinesterase labeling co-localized to AD-vulnerable circuits. Significant association of glial fibrillary acidic protein with Aβ occurred in humans only. The senescence marker p16 correlated positively with pTau expression and negatively with Aβ in patients only. Macaques lacked neuropathologic co-morbidities.ConclusionsAD-relevant neuropathologic signals in the macaque support biomarker discovery in the areas of Aβ plaque evolution and associated synaptic disruption as well as early-stage tau phosphorylation. Relative protection from accumulation of senescence markers, fibrillar tau and neuropathologic co-morbidities in macaque implicate species difference in rates of biological brain aging. We provide over 4000 digital slides for further study.
Collapse
Affiliation(s)
- Caroline J Zeiss
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Anita Huttner
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Amy Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Dibyadeep Datta
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Stephen M Strittmatter
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Brent Vander Wyk
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Alvaro Duque
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
2
|
Gu J, Zheng MQ, Holden D, Fowles K, Qiu L, Felchner Z, Zhang L, Ropchan J, Gropler RJ, Carson RE, Tu Z, Huang Y, Hillmer AT. PET Imaging of Sphingosine-1-Phosphate Receptor 1 with [ 18F]TZ4877 in Nonhuman Primates. Mol Imaging Biol 2025; 27:54-63. [PMID: 39779653 DOI: 10.1007/s11307-024-01979-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/08/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE The sphingosine-1-phosphate receptor-1 (S1PR1) is involved in regulating responses to neuroimmune stimuli. There is a need for S1PR1-specific radioligands with clinically suitable brain pharmcokinetic properties to complement existing radiotracers. This work evaluated a promising S1PR1 radiotracer, [18F]TZ4877, in nonhuman primates. PROCEDURES [18F]TZ4877 was produced via nucleophilic substitution of tosylate precursor with K[18F]/F- followed by deprotection. Brain PET imaging data were acquired with a Focus220 scanner in two Macaca mulatta (6, 13 years old) for 120-180 min following bolus injection of 118-163 MBq [18F]TZ4877, with arterial blood sampling and metabolite analysis to measure the parent input function and plasma free fraction (fP). Each animal was scanned at baseline, 15-18 min after 0.047-0.063 mg/kg of the S1PR1 inhibitor ponesimod, 33 min after 0.4-0.8 mg/kg of the S1PR1-specific compound TZ82112, and 167-195 min after 1 ng/kg of the immune stimulus endotoxin. Kinetic analysis with metabolite-corrected input function was performed to estimate the free fraction corrected total distribution volume (VT/fP). Whole-body dosimetry scans were acquired in 2 animals (1M, 1F) with a Biograph Vision PET/CT System, and absorbed radiation dose estimates were calculated with OLINDA. RESULTS [18F]TZ4877 exhibited fast kinetics that were described by the reversible 2-tissue compartment model. Baseline [18F]TZ4877 fP was low (<1%), and [18F]TZ4877 VT/fP values were 233-866 mL/cm3. TZ82112 dose-dependently reduced [18F]TZ4877 VT/fP, while ponesimod and endotoxin exhibited negligible effects on VT/fP, possibly due to scan timing relative to dosing. Dosimetry studies identified the critical organs of gallbladder (0.42 (M) and 0.31 (F) mSv/MBq) for anesthetized nonhuman primate. CONCLUSIONS [18F]TZ4877 exhibits reversible kinetic properties, but the low fP value limits reproducible quantification with this radiotracer. S1PR1 is a compelling PET imaging target, and these data support pursuing alternative F-18 labeled radiotracers for potential future human studies.
Collapse
Affiliation(s)
- Jiwei Gu
- Yale PET Center, Yale School of Medicine, New Haven, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Ming-Qiang Zheng
- Yale PET Center, Yale School of Medicine, New Haven, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Daniel Holden
- Yale PET Center, Yale School of Medicine, New Haven, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Krista Fowles
- Yale PET Center, Yale School of Medicine, New Haven, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Lin Qiu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, USA
| | - Zachary Felchner
- Yale PET Center, Yale School of Medicine, New Haven, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Li Zhang
- Yale PET Center, Yale School of Medicine, New Haven, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Jim Ropchan
- Yale PET Center, Yale School of Medicine, New Haven, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Robert J Gropler
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, USA
| | - Richard E Carson
- Yale PET Center, Yale School of Medicine, New Haven, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Zhude Tu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, USA
| | - Yiyun Huang
- Yale PET Center, Yale School of Medicine, New Haven, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Ansel T Hillmer
- Yale PET Center, Yale School of Medicine, New Haven, USA.
- Department of Psychiatry, Yale School of Medicine, New Haven, USA.
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA.
| |
Collapse
|
3
|
Renteln M. Toward Systemic Lipofuscin Removal. Rejuvenation Res 2024; 27:171-179. [PMID: 39041624 DOI: 10.1089/rej.2024.0034] [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: 07/24/2024] Open
Abstract
Lipofuscin is indigestible garbage that accumulates in the autophagic vesicles and cytosol of postmitotic cells with age. Drs. Brunk and Terman postulated that lipofuscin accumulation is the main or at least a major driving factor in aging. They even posited that the evolution of memory is the reason why we get lipofuscin at all, as stable synaptic connections must be maintained over time, meaning that the somas of neurons must also remain in the same locale. In other words, they cannot dilute out their garbage over time through cell division. Mechanistically, their position certainly makes sense given that rendering a large percentage of a postmitotic cell's lysosomes useless must almost certainly negatively affect that cell and the surrounding microenvironment. It may be the case that lipofuscin accumulation is the main issue with regard to current age-related disease. Degradation in situ may be an insurmountable task currently. However, a method of systemic lipofuscin removal is discussed herein.
Collapse
Affiliation(s)
- Michael Renteln
- Molecular Genetics and Biochemistry from USC, Los Angeles, California, USA
| |
Collapse
|
4
|
Greenberg EF, Voorbach MJ, Smith A, Reuter DR, Zhuang Y, Wang JQ, Wooten DW, Asque E, Hu M, Hoft C, Duggan R, Townsend M, Orsi K, Dalecki K, Amberg W, Duggan L, Knight H, Spina JS, He Y, Marsh K, Zhao V, Ybarra S, Mollon J, Fang Y, Vasanthakumar A, Westmoreland S, Droescher M, Finnema SJ, Florian H. Navitoclax safety, tolerability, and effect on biomarkers of senescence and neurodegeneration in aged nonhuman primates. Heliyon 2024; 10:e36483. [PMID: 39253182 PMCID: PMC11382177 DOI: 10.1016/j.heliyon.2024.e36483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024] Open
Abstract
Alzheimer's disease (AD) is the most common global dementia and is universally fatal. Most late-stage AD disease-modifying therapies are intravenous and target amyloid beta (Aβ), with only modest effects on disease progression: there remains a high unmet need for convenient, safe, and effective therapeutics. Senescent cells (SC) and the senescence-associated secretory phenotype (SASP) drive AD pathology and increase with AD severity. Preclinical senolytic studies have shown improvements in neuroinflammation, tau, Aβ, and CNS damage; most were conducted in transgenic rodent models with uncertain human translational relevance. In this study, aged cynomolgus monkeys had significant elevation of biomarkers of senescence, SASP, and neurological damage. Intermittent treatment with the senolytic navitoclax induced modest reversible thrombocytopenia; no serious drug-related toxicity was noted. Navitoclax reduced several senescence and SASP biomarkers, with CSF concentrations sufficient for senolysis. Finally, navitoclax reduced TSPO-PET frontal cortex binding and showed trends of improvement in CSF biomarkers of neuroinflammation, neuronal damage, and synaptic dysfunction. Overall, navitoclax administration was safe and well tolerated in aged monkeys, inducing trends of biomarker changes relevant to human neurodegenerative disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Min Hu
- AbbVie Inc., North Chicago, IL, United States
| | - Carolin Hoft
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Ryan Duggan
- AbbVie Inc., North Chicago, IL, United States
| | - Matthew Townsend
- AbbVie, Cambridge Research Center, 200 Sidney Street, Cambridge, MA, 02139, United States
| | - Karin Orsi
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | | | - Willi Amberg
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Lori Duggan
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Heather Knight
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Joseph S Spina
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Yupeng He
- AbbVie Inc., North Chicago, IL, United States
| | | | - Vivian Zhao
- AbbVie Bay Area, 1000 Gateway Boulevard, South San Francisco, CA, 94080, United States
| | - Suzanne Ybarra
- AbbVie Bay Area, 1000 Gateway Boulevard, South San Francisco, CA, 94080, United States
| | - Jennifer Mollon
- AbbVie Deutschland GmbH & Co. KG, Statistical Sciences and Analytics, Knollstrasse, 67061, Ludwigshafen, Germany
| | - Yuni Fang
- AbbVie Bay Area, 1000 Gateway Boulevard, South San Francisco, CA, 94080, United States
| | | | - Susan Westmoreland
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, United States
| | - Mathias Droescher
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061, Ludwigshafen, Germany
| | | | | |
Collapse
|
5
|
Gu J, Zheng MQ, Holden D, Fowles K, Qiu L, Felchner Z, Zhang L, Ropchan J, Gropler RJ, Carson RE, Tu Z, Huang Y, Hillmer AT. PET Imaging of Sphingosine-1-Phosphate Receptor 1 with [18F]TZ4877 in Nonhuman Primates. RESEARCH SQUARE 2024:rs.3.rs-4350862. [PMID: 38854065 PMCID: PMC11160920 DOI: 10.21203/rs.3.rs-4350862/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Purpose The sphingosine-1-phosphate receptor-1 (S1PR1) is involved in regulating responses to neuroimmune stimuli. There is a need for S1PR1-specific radioligands with clinically suitable brain pharmcokinetic properties to complement existing radiotracers. This work evaluated a promising S1PR1 radiotracer, [18F]TZ4877, in nonhuman primates. Procedures [18F]TZ4877 was produced via nucleophilic substitution of tosylate precursor with K[18F]/F- followed by deprotection. Brain PET imaging data were acquired with a Focus220 scanner in two Macaca mulatta (6, 13 years old) for 120-180 min following bolus injection of 118-163 MBq [18F]TZ4877, with arterial blood sampling and metabolite analysis to measure the parent input function and plasma free fraction (f P). Each animal was scanned at baseline, 15-18 min after 0.047-0.063 mg/kg of the S1PR1 inhibitor ponesimod, 33 min after 0.4-0.8 mg/kg of the S1PR1-specific compound TZ82112, and 167-195 min after 1 ng/kg of the immune stimulus endotoxin. Kinetic analysis with metabolite-corrected input function was performed to estimate the free fraction corrected total distribution volume (V T/f P). Whole-body dosimetry scans were acquired in 2 animals (1M, 1F) with a Biograph Vision PET/CT System, and absorbed radiation dose estimates were calculated with OLINDA. Results [18F]TZ4877 exhibited fast kinetics that were described by the reversible 2-tissue compartment model. Baseline [18F]TZ4877 f P was low (< 1%), and [18F]TZ4877 V T/f P values were 233-866 mL/cm3. TZ82112 dose-dependently reduced [18F]TZ4877 V T/f P, while ponesimod and endotoxin exhibited negligible effects on V T/f P, possibly due to scan timing relative to dosing. Dosimetry studies identified the critical organs of gallbladder (0.42 (M) and 0.31 (F) mSv/MBq) for anesthetized nonhuman primate. Conclusions [18F]TZ4877 exhibits reversible kinetic properties, but the low f P value limits quantification with this radiotracer. S1PR1 is a compelling PET imaging target, and these data support pursuing alternative F-18 labeled radiotracers for potential future human studies.
Collapse
Affiliation(s)
| | | | | | | | - Lin Qiu
- Washington University School of Medicine in Saint Louis: Washington University in St Louis School of Medicine
| | | | | | | | - Robert J Gropler
- Washington University School of Medicine in Saint Louis: Washington University in St Louis School of Medicine
| | | | - Zhude Tu
- Washington University School of Medicine in Saint Louis: Washington University in St Louis School of Medicine
| | | | | |
Collapse
|
6
|
Aceves-Serrano L, Neva JL, Munro J, Vavasour IM, Parent M, Boyd LA, Doudet DJ. Evaluation of microglia activation related markers following a clinical course of TBS: A non-human primate study. PLoS One 2024; 19:e0301118. [PMID: 38753646 PMCID: PMC11098425 DOI: 10.1371/journal.pone.0301118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 03/11/2024] [Indexed: 05/18/2024] Open
Abstract
While the applicability and popularity of theta burst stimulation (TBS) paradigms remain, current knowledge of their neurobiological effects is still limited, especially with respect to their impact on glial cells and neuroinflammatory processes. We used a multimodal imaging approach to assess the effects of a clinical course of TBS on markers for microglia activation and tissue injury as an indirect assessment of neuroinflammatory processes. Healthy non-human primates received continuous TBS (cTBS), intermittent TBS (iTBS), or sham stimulation over the motor cortex at 90% of resting motor threshold. Stimulation was delivered to the awake subjects 5 times a week for 3-4 weeks. Translocator protein (TSPO) expression was evaluated using Positron Emission Tomography and [11C]PBR28, and myo-inositol (mI) and N-acetyl-aspartate (NAA) concentrations were assessed with Magnetic Resonance Spectroscopy. Animals were then euthanized, and immunofluorescence staining was performed using antibodies against TSPO. Paired t-tests showed no significant changes in [11C]PBR28 measurements after stimulation. Similarly, no significant changes in mI and NAA concentrations were found. Post-mortem TSPO evaluation showed comparable mean immunofluorescence intensity after active TBS and sham delivery. The current study suggests that in healthy brains a clinical course of TBS, as evaluated with in-vivo imaging techniques (PET and MRS), did not measurably modulate the expression of glia related markers and metabolite associated with neural viability.
Collapse
Affiliation(s)
- Lucero Aceves-Serrano
- Department of Medicine, Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jason L. Neva
- Faculté de Médecine, École de Kinésiologie et des Sciences de l’activité Physique, Université de Montréal, Montreal, Quebec, Canada
- Centre de Recherche de l’institut Universitaire de Gériatrie de Montréal, Montreal, QC, Canada
| | - Jonathan Munro
- CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Irene M. Vavasour
- Faculty of Medicine, UBC MRI Research Center, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin Parent
- CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Lara A. Boyd
- Faculty of Medicine, Department of Physical Therapy, University of British Columbia, Vancouver, British Columbia, Canada
- Faculty of Medicine, Graduate Program of Rehabilitation Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Doris J. Doudet
- Department of Medicine, Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
7
|
Soares AR, Picciotto MR. Nicotinic regulation of microglia: potential contributions to addiction. J Neural Transm (Vienna) 2024; 131:425-435. [PMID: 37778006 PMCID: PMC11189589 DOI: 10.1007/s00702-023-02703-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/22/2023] [Indexed: 10/03/2023]
Abstract
Clinical and preclinical studies have identified immunosuppressive effects of nicotine, with potential implications for treating nicotine addiction. Here we review how nicotine can regulate microglia, the resident macrophages in the brain, and corresponding effects of nicotine on neuroimmune signaling. There is significant evidence that activation of α7 nicotinic acetylcholine receptors (nAChRs) on microglia can trigger an anti-inflammatory cascade that alters microglial polarization and activity, cytokine release, and intracellular calcium concentrations, leading to neuroprotection. These anti-inflammatory effects of nicotine-dependent α7 nAChR signaling are lost during withdrawal, suggesting that neuroimmune signaling is potentiated during abstinence, and thus, heightened microglial activity may drive circuit disruption that contributes to withdrawal symptoms and hyperkatifeia. In sum, the clinical literature has highlighted immunomodulatory effects of nicotine and the potential for anti-inflammatory compounds to treat addiction. The preclinical literature investigating the underlying mechanisms points to a role of microglial engagement in the circuit dysregulation and behavioral changes that occur during nicotine addiction and withdrawal, driven, at least in part, by activation of α7 nAChRs on microglia. Specifically targeting microglial signaling may help alleviate withdrawal symptoms in people with nicotine dependence and help to promote abstinence.
Collapse
Affiliation(s)
- Alexa R Soares
- Department of Psychiatry, Yale University, 34 Park Street-3rd floor Research, New Haven, CT, 06508, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, 06508, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University, 34 Park Street-3rd floor Research, New Haven, CT, 06508, USA.
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, 06508, USA.
| |
Collapse
|
8
|
Weyer MP, Strehle J, Schäfer MKE, Tegeder I. Repurposing of pexidartinib for microglia depletion and renewal. Pharmacol Ther 2024; 253:108565. [PMID: 38052308 DOI: 10.1016/j.pharmthera.2023.108565] [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: 09/28/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023]
Abstract
Pexidartinib (PLX3397) is a small molecule receptor tyrosine kinase inhibitor of colony stimulating factor 1 receptor (CSF1R) with moderate selectivity over other members of the platelet derived growth factor receptor family. It is approved for treatment of tenosynovial giant cell tumors (TGCT). CSF1R is highly expressed by microglia, which are macrophages of the central nervous system (CNS) that defend the CNS against injury and pathogens and contribute to synapse development and plasticity. Challenged by pathogens, apoptotic cells, debris, or inflammatory molecules they adopt a responsive state to propagate the inflammation and eventually return to a homeostatic state. The phenotypic switch may fail, and disease-associated microglia contribute to the pathophysiology in neurodegenerative or neuropsychiatric diseases or long-lasting detrimental brain inflammation after brain, spinal cord or nerve injury or ischemia/hemorrhage. Microglia also contribute to the growth permissive tumor microenvironment of glioblastoma (GBM). In rodents, continuous treatment for 1-2 weeks via pexidartinib food pellets leads to a depletion of microglia and subsequent repopulation from the remaining fraction, which is aided by peripheral monocytes that search empty niches for engraftment. The putative therapeutic benefit of such microglia depletion or forced renewal has been assessed in almost any rodent model of CNS disease or injury or GBM with heterogeneous outcomes, but a tendency of partial beneficial effects. So far, microglia monitoring e.g. via positron emission imaging is not standard of care for patients receiving Pexidartinib (e.g. for TGCT), so that the depletion and repopulation efficiency in humans is still largely unknown. Considering the virtuous functions of microglia, continuous depletion is likely no therapeutic option but short-lasting transient partial depletion to stimulate microglia renewal or replace microglia in genetic disease in combination with e.g. stem cell transplantation or as part of a multimodal concept in treatment of glioblastoma appears feasible. The present review provides an overview of the preclinical evidence pro and contra microglia depletion as a therapeutic approach.
Collapse
Affiliation(s)
- Marc-Philipp Weyer
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Faculty of Medicine, Frankfurt, Germany
| | - Jenny Strehle
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Faculty of Medicine, Frankfurt, Germany.
| |
Collapse
|
9
|
Wang W, Li Y, Ma F, Sheng X, Chen K, Zhuo R, Wang C, Zheng H, Zhang YW, Bu G, Chen XF, Zhong L. Microglial repopulation reverses cognitive and synaptic deficits in an Alzheimer's disease model by restoring BDNF signaling. Brain Behav Immun 2023; 113:275-288. [PMID: 37482204 DOI: 10.1016/j.bbi.2023.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/10/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023] Open
Abstract
Over the past decade, compelling genetic evidence has highlighted the crucial role of microglial dysregulation in the development of Alzheimer's disease (AD). As resident immune cells in the brain, microglia undergo dystrophy and senescence during the chronic progression of AD. To explore the potential therapeutic benefits of replenishing the brain with new microglia in AD, we utilized the CSF1R inhibitor PLX3397 to deplete existing microglia and induce repopulation after inhibitor withdrawal in 5xFAD transgenic mice. Our findings revealed the remarkable benefits of microglial repopulation in ameliorating AD-associated cognitive deficits, accompanied by a notable elevation in synaptic proteins and an enhancement of hippocampal long-term potentiation (LTP). Additionally, we observed the profound restoration of microglial morphology and synaptic engulfment following their self-renewal. The impact of microglial repopulation on amyloid pathology is dependent on the duration of repopulation. Transcriptome analysis revealed a high resemblance between the gene expression profiles of repopulated microglia from 5xFAD mice and those of microglia from WT mice. Importantly, the dysregulated neurotrophic signaling pathway and hippocampal neurogenesis in the AD brain are restored following microglial replenishment. Lastly, we demonstrated that the repopulation restores the expression of brain-derived neurotrophic factor (BDNF) in microglia, thereby contributing to synaptic plasticity. In conclusion, our findings provide compelling evidence to support the notion that microglial self-renewal confers substantial benefits to the AD brain by restoring the BDNF neurotrophic signaling pathway. Thus, targeted microglial repopulation emerges as a highly promising and novel therapeutic strategy for alleviating cognitive impairment in AD.
Collapse
Affiliation(s)
- Wanbing Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yanzhong Li
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Fangling Ma
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Xuan Sheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Kai Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Rengong Zhuo
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Chen Wang
- Department of Neurology, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Honghua Zheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Xiao-Fen Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China; Shenzhen Research Institute of Xiamen University, Shenzhen 518063, China.
| | - Li Zhong
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
10
|
Raval NR, Angarita G, Matuskey D, Miller R, Drake LR, Kapinos M, Nabulsi N, Huang Y, Carson RE, O'Malley SS, Cosgrove KP, Hillmer AT. Imaging the brain's immune response to alcohol with [ 11C]PBR28 TSPO Positron Emission Tomography. Mol Psychiatry 2023; 28:3384-3390. [PMID: 37532797 PMCID: PMC10743097 DOI: 10.1038/s41380-023-02198-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 08/04/2023]
Abstract
In humans, the negative effects of alcohol are linked to immune dysfunction in both the periphery and the brain. Yet acute effects of alcohol on the neuroimmune system and its relationships with peripheral immune function are not fully understood. To address this gap, immune response to an alcohol challenge was measured with positron emission tomography (PET) using the radiotracer [11C]PBR28, which targets the 18-kDa translocator protein, a marker sensitive to immune challenges. Participants (n = 12; 5 F; 25-45 years) who reported consuming binge levels of alcohol (>3 drinks for females; >4 drinks for males) 1-3 months before scan day were enrolled. Imaging featured a baseline [11C]PBR28 scan followed by an oral laboratory alcohol challenge over 90 min. An hour later, a second [11C]PBR28 scan was acquired. Dynamic PET data were acquired for at least 90 min with arterial blood sampling to measure the metabolite-corrected input function. [11C]PBR28 volume of distributions (VT) was estimated in the brain using multilinear analysis 1. Subjective effects, blood alcohol levels (BAL), and plasma cytokines were measured during the paradigm. Full completion of the alcohol challenge and data acquisition occurred for n = 8 (2 F) participants. Mean peak BAL was 101 ± 15 mg/dL. Alcohol significantly increased brain [11C]PBR28 VT (n = 8; F(1,49) = 34.72, p > 0.0001; Cohen's d'=0.8-1.7) throughout brain by 9-16%. Alcohol significantly altered plasma cytokines TNF-α (F(2,22) = 17.49, p < 0.0001), IL-6 (F(2,22) = 18.00, p > 0.0001), and MCP-1 (F(2,22) = 7.02, p = 0.004). Exploratory analyses identified a negative association between the subjective degree of alcohol intoxication and changes in [11C]PBR28 VT. These findings provide, to our knowledge, the first in vivo human evidence for an acute brain immune response to alcohol.
Collapse
Affiliation(s)
- Nakul R Raval
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Yale PET Center, Yale University, New Haven, CT, USA
| | - Gustavo Angarita
- Yale PET Center, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - David Matuskey
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Yale PET Center, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
- Department of Neurology, Yale University New Haven, New Haven, CT, USA
| | - Rachel Miller
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Lindsey R Drake
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Yale PET Center, Yale University, New Haven, CT, USA
| | - Michael Kapinos
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Yale PET Center, Yale University, New Haven, CT, USA
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Yale PET Center, Yale University, New Haven, CT, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Yale PET Center, Yale University, New Haven, CT, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Yale PET Center, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, CT, USA
| | | | - Kelly P Cosgrove
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Yale PET Center, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Ansel T Hillmer
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA.
- Yale PET Center, Yale University, New Haven, CT, USA.
- Department of Psychiatry, Yale University, New Haven, CT, USA.
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, CT, USA.
| |
Collapse
|
11
|
Temple S. Advancing cell therapy for neurodegenerative diseases. Cell Stem Cell 2023; 30:512-529. [PMID: 37084729 PMCID: PMC10201979 DOI: 10.1016/j.stem.2023.03.017] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/23/2023]
Abstract
Cell-based therapies are being developed for various neurodegenerative diseases that affect the central nervous system (CNS). Concomitantly, the roles of individual cell types in neurodegenerative pathology are being uncovered by genetic and single-cell studies. With a greater understanding of cellular contributions to health and disease and with the arrival of promising approaches to modulate them, effective therapeutic cell products are now emerging. This review examines how the ability to generate diverse CNS cell types from stem cells, along with a deeper understanding of cell-type-specific functions and pathology, is advancing preclinical development of cell products for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA.
| |
Collapse
|
12
|
Raval NR, Wetherill RR, Wiers CE, Dubroff JG, Hillmer AT. Positron Emission Tomography of Neuroimmune Responses in Humans: Insights and Intricacies. Semin Nucl Med 2023; 53:213-229. [PMID: 36270830 PMCID: PMC11261531 DOI: 10.1053/j.semnuclmed.2022.08.008] [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: 08/15/2022] [Accepted: 08/30/2022] [Indexed: 11/06/2022]
Abstract
The brain's immune system plays a critical role in responding to immune challenges and maintaining homeostasis. However, dysregulated neuroimmune function contributes to neurodegenerative disease and neuropsychiatric conditions. In vivo positron emission tomography (PET) imaging of the neuroimmune system has facilitated a greater understanding of its physiology and the pathology of some neuropsychiatric conditions. This review presents an in-depth look at PET findings from human neuroimmune function studies, highlighting their importance in current neuropsychiatric research. Although the majority of human PET studies feature radiotracers targeting the translocator protein 18 kDa (TSPO), this review also considers studies with other neuroimmune targets, including monoamine oxidase B, cyclooxygenase-1 and cyclooxygenase-2, nitric oxide synthase, and the purinergic P2X7 receptor. Promising new targets, such as colony-stimulating factor 1, Sphingosine-1-phosphate receptor 1, and the purinergic P2Y12 receptor, are also discussed. The significance of validating neuroimmune targets and understanding their function and expression is emphasized in this review to better identify and interpret PET results.
Collapse
Affiliation(s)
- Nakul R Raval
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT; Yale PET Center, Yale University, New Haven, CT
| | - Reagan R Wetherill
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Corinde E Wiers
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jacob G Dubroff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ansel T Hillmer
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT; Yale PET Center, Yale University, New Haven, CT; Department of Psychiatry, Yale University, New Haven, CT.
| |
Collapse
|
13
|
CSF1R inhibitors induce a sex-specific resilient microglial phenotype and functional rescue in a tauopathy mouse model. Nat Commun 2023; 14:118. [PMID: 36624100 PMCID: PMC9829908 DOI: 10.1038/s41467-022-35753-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 12/23/2022] [Indexed: 01/10/2023] Open
Abstract
Microglia are central to pathogenesis in many neurological conditions. Drugs targeting colony-stimulating factor-1 receptor (CSF1R) to block microglial proliferation in preclinical disease models have shown mixed outcomes, thus the therapeutic potential of this approach remains unclear. Here, we show that CSF1R inhibitors given by multiple dosing paradigms in the Tg2541 tauopathy mouse model cause a sex-independent reduction in pathogenic tau and reversion of non-microglial gene expression patterns toward a normal wild type signature. Despite greater drug exposure in male mice, only female mice have functional rescue and extended survival. A dose-dependent upregulation of immediate early genes and neurotransmitter dysregulation are observed in the brains of male mice only, indicating that excitotoxicity may preclude functional benefits. Drug-resilient microglia in male mice exhibit morphological and gene expression patterns consistent with increased neuroinflammatory signaling, suggesting a mechanistic basis for sex-specific excitotoxicity. Complete microglial ablation is neither required nor desirable for neuroprotection and therapeutics targeting microglia must consider sex-dependent effects.
Collapse
|
14
|
Li H, Zhou B, Liao P, Liao D, Yang L, Wang J, Liu J, Jiang R, Chen L. Prolonged exposure of neonatal mice to sevoflurane leads to hyper-ramification in microglia, reduced contacts between microglia and synapses, and defects in adult behavior. Front Neurol 2023; 14:1142739. [PMID: 37025197 PMCID: PMC10072331 DOI: 10.3389/fneur.2023.1142739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/24/2023] [Indexed: 04/08/2023] Open
Abstract
Background Prolonged exposure to general anesthetics during development is known to cause neurobehavioral abnormalities, but the cellular and molecular mechanisms involved are unclear. Microglia are the resident immune cells in the central nervous system and play essential roles in normal brain development. Materials and methods In the study, postnatal day 7 (P7) C57BL/6 mice were randomly assigned to two groups. In the sevoflurane (SEVO), mice were exposed to 2.5% sevoflurane for 4 h. In the control group, mice were exposed to carrier gas (30% O2/70% N2) for 4 h. Fixed brain slices from P14 to P21 mice were immunolabeled for ionized calcium-binding adapter molecule 1 (IBA-1) to visualize microglia. The morphological analysis of microglia in the somatosensory cortex was performed using ImageJ and Imaris software. Serial block face scanning electron microscopy (SBF-SEM) was performed to assess the ultrastructure of the microglia and the contacts between microglia and synapse in P14 and P21 mice. The confocal imaging of brain slices was performed to assess microglia surveillance in resting and activated states in P14 and P21 mice. Behavioral tests were used to assess the effect of microglia depletion and repopulation on neurobehavioral abnormalities caused by sevoflurane exposure. Results The prolonged exposure of neonatal mice to sevoflurane induced microglia hyper-ramification with an increase in total branch length, arborization area, and branch complexity 14 days after exposure. Prolonged neonatal sevoflurane exposure reduced contacts between microglia and synapses, without affecting the surveillance of microglia in the resting state or responding to laser-induced focal brain injury. These neonatal changes in microglia were associated with anxiety-like behaviors in adult mice. Furthermore, microglial depletion before sevoflurane exposure and subsequent repopulation in the neonatal brain mitigated anxiety-like behaviors caused by sevoflurane exposure. Conclusion Our experiments indicate that general anesthetics may harm the developing brain, and microglia may be an essential target of general anesthetic-related developmental neurotoxicity.
Collapse
Affiliation(s)
- Hong Li
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Zhou
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Ping Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Daqing Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Linghui Yang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Wang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Ruotian Jiang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Ruotian Jiang,
| | - Lingmin Chen
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Lingmin Chen,
| |
Collapse
|
15
|
Tollefson S, Himes ML, Kozinski KM, Lopresti BJ, Mason NS, Hibbeln J, Muldoon MF, Narendran R. Imaging the Influence of Red Blood Cell Docosahexaenoic Acid Status on the Expression of the 18 kDa Translocator Protein in the Brain: A [ 11C]PBR28 Positron Emission Tomography Study in Young Healthy Men. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:998-1006. [PMID: 34607054 DOI: 10.1016/j.bpsc.2021.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/09/2021] [Accepted: 09/18/2021] [Indexed: 01/11/2023]
Abstract
BACKGROUND Docosahexaenoic acid (DHA) shows anti-inflammatory/proresolution effects in the brain. Higher red blood cell (RBC) DHA in humans is associated with improved cognitive performance and a lower risk for suicide. Here, we hypothesized that binding to the 18 kDa translocator protein (TSPO), a proxy for microglia levels, will be higher in individuals with low DHA relative to high DHA levels. We also postulated that higher TSPO would predict poor cognitive performance and impaired stress resilience. METHODS RBC DHA screening was performed in 320 healthy males. [11C]PBR28 positron emission tomography was used to measure binding to TSPO in 38 and 32 males in the lowest and highest RBC DHA quartiles. Volumes of distribution expressed relative to total plasma ligand concentration (VT) was derived using an arterial input function-based kinetic analysis in 14 brain regions. RESULTS [11C]PBR28 VT was significantly lower (by 12% and 20% in C/T and C/C rs6971 genotypes) in males with low RBC DHA than in males with high RBC DHA. Regional VT was correlated positively and negatively with RBC DHA and serum triglycerides, respectively. No relationships between VT and cognitive performance or stress resilience measures were present. CONCLUSIONS Contrary to our hypothesis, we found lower TSPO binding in low-DHA than in high-DHA subjects. It is unclear as to whether low TSPO binding reflects differences in microglia levels and/or triglyceride metabolism in this study. Future studies with specific targets are necessary to confirm the effect of DHA on microglia. These results underscore the need to consider lipid parameters as a factor when interpreting TSPO positron emission tomography clinical findings.
Collapse
Affiliation(s)
- Savannah Tollefson
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Michael L Himes
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Katelyn M Kozinski
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - N Scott Mason
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Joseph Hibbeln
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Matthew F Muldoon
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
16
|
Toloff K, Woodcock EA. Is the Neuroimmune System a Therapeutic Target for Opioid Use Disorder? A Systematic Review. MEDICAL RESEARCH ARCHIVES 2022; 10:2955. [PMID: 37744743 PMCID: PMC10516332 DOI: 10.18103/mra.v10i8.2955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Opioid use disorder (OUD) is an epidemic in the United States. In the past 12 months alone, there have been 75,000+ deaths attributed to opioid overdose: more than any other year in American history. Current pharmacotherapies for the treatment of OUD effectively suppress opioid withdrawal symptoms, but long-term relapse rates remain unacceptably high. Novel treatments for OUD are desperately needed to curb this epidemic. One target that has received considerable recent interest is the neuroimmune system. The neuroimmune system is anchored by glial cells, i.e., microglia and astrocytes, but neuroimmune signaling is known to influence neurons, including altering neurotransmission, synapse formation, and ultimately, brain function. Preclinical studies have shown that experimental attenuation of pro-inflammatory neuroimmune signaling modulates opioid addiction processes, including opioid reward, tolerance, and withdrawal symptoms, which suggests potential therapeutic benefit in patients. Whereas the peripheral immune system in OUD patients has been studied for decades and is well-understood, little is known about the neuroimmune system in OUD patients or its viability as a treatment target. Herein, we review the literature describing relationships between opioid administration and the neuroimmune system, the influence of neuroimmune signaling on opioid addiction processes, and the therapeutic potential for targeting the neuroimmune system in OUD subjects using glial modulator medications.
Collapse
Affiliation(s)
- Katelyn Toloff
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eric A. Woodcock
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
17
|
Nicotinic Acetylcholine Receptors and Microglia as Therapeutic and Imaging Targets in Alzheimer's Disease. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092780. [PMID: 35566132 PMCID: PMC9102429 DOI: 10.3390/molecules27092780] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022]
Abstract
Amyloid-β (Aβ) accumulation and tauopathy are considered the pathological hallmarks of Alzheimer’s disease (AD), but attenuation in choline signaling, including decreased nicotinic acetylcholine receptors (nAChRs), is evident in the early phase of AD. Currently, there are no drugs that can suppress the progression of AD due to a limited understanding of AD pathophysiology. For this, diagnostic methods that can assess disease progression non-invasively before the onset of AD symptoms are essential, and it would be valuable to incorporate the concept of neurotheranostics, which simultaneously enables diagnosis and treatment. The neuroprotective pathways activated by nAChRs are attractive targets as these receptors may regulate microglial-mediated neuroinflammation. Microglia exhibit both pro- and anti-inflammatory functions that could be modulated to mitigate AD pathogenesis. Currently, single-cell analysis is identifying microglial subpopulations that may have specific functions in different stages of AD pathologies. Thus, the ability to image nAChRs and microglia in AD according to the stage of the disease in the living brain may lead to the development of new diagnostic and therapeutic methods. In this review, we summarize and discuss the recent findings on the nAChRs and microglia, as well as their methods for live imaging in the context of diagnosis, prophylaxis, and therapy for AD.
Collapse
|
18
|
Han J, Chitu V, Stanley ER, Wszolek ZK, Karrenbauer VD, Harris RA. Inhibition of colony stimulating factor-1 receptor (CSF-1R) as a potential therapeutic strategy for neurodegenerative diseases: opportunities and challenges. Cell Mol Life Sci 2022; 79:219. [PMID: 35366105 PMCID: PMC8976111 DOI: 10.1007/s00018-022-04225-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/06/2022] [Accepted: 02/26/2022] [Indexed: 12/12/2022]
Abstract
Microglia are specialized dynamic immune cells in the central nervous system (CNS) that plays a crucial role in brain homeostasis and in disease states. Persistent neuroinflammation is considered a hallmark of many neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson's disease (PD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS) and primary progressive multiple sclerosis (MS). Colony stimulating factor 1-receptor (CSF-1R) is predominantly expressed on microglia and its expression is significantly increased in neurodegenerative diseases. Cumulative findings have indicated that CSF-1R inhibitors can have beneficial effects in preclinical neurodegenerative disease models. Research using CSF-1R inhibitors has now been extended into non-human primates and humans. This review article summarizes the most recent advances using CSF-1R inhibitors in different neurodegenerative conditions including AD, PD, HD, ALS and MS. Potential challenges for translating these findings into clinical practice are presented.
Collapse
Affiliation(s)
- Jinming Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - E. Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | | | - Virginija Danylaité Karrenbauer
- Department of Clinical Neuroscience, Center for Molecular Medicine L8:04, Karolinska Institutet, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Robert A. Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
19
|
Barca C, Foray C, Hermann S, Herrlinger U, Remory I, Laoui D, Schäfers M, Grauer OM, Zinnhardt B, Jacobs AH. The Colony Stimulating Factor-1 Receptor (CSF-1R)-Mediated Regulation of Microglia/Macrophages as a Target for Neurological Disorders (Glioma, Stroke). Front Immunol 2021; 12:787307. [PMID: 34950148 PMCID: PMC8688767 DOI: 10.3389/fimmu.2021.787307] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/17/2021] [Indexed: 12/11/2022] Open
Abstract
Immunomodulatory therapies have fueled interest in targeting microglial cells as part of the innate immune response after infection or injury. In this context, the colony-stimulating factor 1 (CSF-1) and its receptor (CSF-1R) have gained attention in various neurological conditions to deplete and reprogram the microglia/macrophages compartment. Published data in physiological conditions support the use of small-molecule inhibitors to study microglia/macrophages dynamics under inflammatory conditions and as a therapeutic strategy in pathologies where those cells support disease progression. However, preclinical and clinical data highlighted that the complexity of the spatiotemporal inflammatory response could limit their efficiency due to compensatory mechanisms, ultimately leading to therapy resistance. We review the current state-of-art in the field of CSF-1R inhibition in glioma and stroke and provide an overview of the fundamentals, ongoing research, potential developments of this promising therapeutic strategy and further application toward molecular imaging.
Collapse
Affiliation(s)
- Cristina Barca
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany
| | - Claudia Foray
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany
| | - Sven Hermann
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany
| | - Ulrich Herrlinger
- Division of Clinical Neuro-Oncology, Department of Neurology, University Hospital Bonn, Bonn, Germany.,Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | - Isabel Remory
- In vivo Cellular and Molecular Imaging laboratory (ICMI), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Damya Laoui
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium.,Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Michael Schäfers
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany.,Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Oliver M Grauer
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Bastian Zinnhardt
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany.,Biomarkers & Translational Technologies (BTT), Pharma Research & Early Development (pRED), F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany.,Centre of Integrated Oncology, University Hospital Bonn, Bonn, Germany.,Department of Geriatrics and Neurology, Johanniter Hospital, Bonn, Germany
| |
Collapse
|
20
|
Van Camp N, Lavisse S, Roost P, Gubinelli F, Hillmer A, Boutin H. TSPO imaging in animal models of brain diseases. Eur J Nucl Med Mol Imaging 2021; 49:77-109. [PMID: 34245328 PMCID: PMC8712305 DOI: 10.1007/s00259-021-05379-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/25/2021] [Indexed: 12/19/2022]
Abstract
Over the last 30 years, the 18-kDa TSPO protein has been considered as the PET imaging biomarker of reference to measure increased neuroinflammation. Generally assumed to image activated microglia, TSPO has also been detected in endothelial cells and activated astrocytes. Here, we provide an exhaustive overview of the recent literature on the TSPO-PET imaging (i) in the search and development of new TSPO tracers and (ii) in the understanding of acute and chronic neuroinflammation in animal models of neurological disorders. Generally, studies testing new TSPO radiotracers against the prototypic [11C]-R-PK11195 or more recent competitors use models of acute focal neuroinflammation (e.g. stroke or lipopolysaccharide injection). These studies have led to the development of over 60 new tracers during the last 15 years. These studies highlighted that interpretation of TSPO-PET is easier in acute models of focal lesions, whereas in chronic models with lower or diffuse microglial activation, such as models of Alzheimer's disease or Parkinson's disease, TSPO quantification for detection of neuroinflammation is more challenging, mirroring what is observed in clinic. Moreover, technical limitations of preclinical scanners provide a drawback when studying modest neuroinflammation in small brains (e.g. in mice). Overall, this review underlines the value of TSPO imaging to study the time course or response to treatment of neuroinflammation in acute or chronic models of diseases. As such, TSPO remains the gold standard biomarker reference for neuroinflammation, waiting for new radioligands for other, more specific targets for neuroinflammatory processes and/or immune cells to emerge.
Collapse
Affiliation(s)
- Nadja Van Camp
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Sonia Lavisse
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Pauline Roost
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Francesco Gubinelli
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Ansel Hillmer
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, CT, USA
| | - Hervé Boutin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Brain and Mental Health, University of Manchester, M13 9PL, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, 27 Palatine Road, M20 3LJ, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
| |
Collapse
|
21
|
Aceves-Serrano L, Sossi V, Doudet DJ. Comparison of Invasive and Non-invasive Estimation of [ 11C]PBR28 Binding in Non-human Primates. Mol Imaging Biol 2021; 24:404-415. [PMID: 34622422 DOI: 10.1007/s11307-021-01661-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE To identify a reliable alternative to the full blood [11C]PBR28 quantification method that would be easily replicated in multiple research and clinical settings. PROCEDURES Ten [11C]PBR28 scans were acquired from 7 healthy non-human primates (NHP). Arterial input functions (AIFs) were averaged to create a population template input function (TIF). Population-based input functions were created by scaling the TIF with injected activity per body weight (PBIF) or unmetabolized tracer activity in blood at 15-,30-, and 60-min post-injection (PBIF15, PBIF30, and PBIF60). Two additional input functions were used: the native unmetabolized total plasma activity (Totals) and the Totals curve metabolite corrected by a scaled template parent fraction from a 30-min sample (TPF30-IF). Total distribution volumes (VTs) were calculated using PBIF, PBIF30, PBIF15, PBIF60, Totals, TPF30-IF, and the individual AIF (VTAIF). Distribution volume ratios (DVR) were computed using the cerebellum and the centrum semiovale (CSO), as pseudo-reference regions (DVRCereb, DVRCSO). Results obtained with each method were compared to VTAIF. Applicability of these alternative methods was tested on an independent pharmacological challenge dataset of microglial activation and depletion. Evaluation was carried at baseline, immediately after intervention (acute), and weeks post-intervention (post-recovery). RESULTS VTs computed using PBIF15 and PBIF30 showed the best correlation to VTAIF (r > 0.90), while VT derived from the blood-free-scaled PBIF showed poor correlation (r = 0.46) and DVRCSO correlated the least (r = 0.26). In the pharmacological challenge study, most population-derived VT values were comparable to VTAIF at baseline and showed varied sensitivity to challenges at acute and post-recovery evaluation. DVR values did not detect relevant changes. CONCLUSIONS Population-based input functions scaled with a single blood sample might be a useful alternative to using AIF to compute [11C]PBR28 binding in healthy NHPs or animals with comparable metabolism and overall perform better than pseudo-reference regions approaches.
Collapse
Affiliation(s)
- Lucero Aceves-Serrano
- Department of Medicine, Division of Neurology, University of British Columbia, Rm M36 Purdy Pavilion, 2221 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada.
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| | - Doris J Doudet
- Department of Medicine, Division of Neurology, University of British Columbia, Rm M36 Purdy Pavilion, 2221 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
| |
Collapse
|
22
|
Oh SJ, Ahn H, Jung KH, Han SJ, Nam KR, Kang KJ, Park JA, Lee KC, Lee YJ, Choi JY. Evaluation of the Neuroprotective Effect of Microglial Depletion by CSF-1R Inhibition in a Parkinson's Animal Model. Mol Imaging Biol 2021; 22:1031-1042. [PMID: 32086763 DOI: 10.1007/s11307-020-01485-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Neuroinflammation in Parkinson's disease (PD) is known to play a pivotal role in progression to neuronal degeneration. It has been reported that colony-stimulation factor 1 receptor (CSF-1R) inhibition can effectively deplete microglia. However, its therapeutic efficacy in PD is unclear still now. PROCEDURES To elucidate this issue, we examined the contribution of microglial depletion to PD by behavioral testing, positron emission tomography (PET) imaging, and immunoassays in sham, PD, and microglial depletion PD model (PLX3397 was administered to PD groups, with n = 6 in each group). RESULTS The microglial depletion in PD model showed improved sensory motor function and depressive-like behavior. NeuroPET revealed that PLX3397 treatment resulted in partial recovery of striatal neuro-inflammatory functions (binding values of [18F]DPA-174 for PD, 1.47 ± 0.12, p < 0.01 vs. for PLX3397 in PD: 1.33 ± 0.26) and the dopaminergic (binding values of 18F-FP-CIT for PD, 1.32 ± 0.07 vs. for PLX3397 in PD: 1.54 ± 0.10, p < 0.01) and glutamatergic systems (binding values of [18F]FPEB for PD: 9.22 ± 0.54 vs. for PLX3397 Tx in PD: 9.83 ± 0.96, p > 0.05). Western blotting for microglia showed similar changes. CONCLUSION Microglial depletion has inflammation-related therapeutic effects, which have beneficial effects on motor and nonmotor symptoms of PD.
Collapse
Affiliation(s)
- Se Jong Oh
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, South Korea
| | - Heesu Ahn
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, South Korea.,Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon, South Korea
| | - Ki-Hye Jung
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, South Korea.,Medical Device-Bio Research Institute, Korea Testing and Research Institute, Gwacheon, Gyeonggi-do, South Korea
| | - Sang Jin Han
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, South Korea
| | - Kyung Rok Nam
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, South Korea
| | - Kyung Jun Kang
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, South Korea
| | - Ji-Ae Park
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, South Korea
| | - Kyo Chul Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, South Korea
| | - Yong Jin Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, South Korea
| | - Jae Yong Choi
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, South Korea.
| |
Collapse
|
23
|
Scott MC, Bedi SS, Olson SD, Sears CM, Cox CS. Microglia as therapeutic targets after neurological injury: strategy for cell therapy. Expert Opin Ther Targets 2021; 25:365-380. [PMID: 34029505 DOI: 10.1080/14728222.2021.1934447] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Microglia is the resident tissue macrophages of the central nervous system. Prolonged microglial activation often occurs after traumatic brain injury and is associated with deteriorating neurocognitive outcomes. Resolution of microglial activation is associated with limited tissue loss and improved neurocognitive outcomes. Limiting the prolonged pro-inflammatory response and the associated secondary tissue injury provides the rationale and scientific premise for considering microglia as a therapeutic target. AREAS COVERED In this review, we discuss markers of microglial activation, such as immunophenotype and microglial response to injury, including cytokine/chemokine release, free radical formation, morphology, phagocytosis, and metabolic shifts. We compare the origin and role in neuroinflammation of microglia and monocytes/macrophages. We review potential therapeutic targets to shift microglial polarization. Finally, we review the effect of cell therapy on microglia. EXPERT OPINION Dysregulated microglial activation after neurologic injury, such as traumatic brain injury, can worsen tissue damage and functional outcomes. There are potential targets in microglia to attenuate this activation, such as proteins and molecules that regulate microglia polarization. Cellular therapeutics that limit, but do not eliminate, the inflammatory response have improved outcomes in animal models by reducing pro-inflammatory microglial activation via secondary signaling. These findings have been replicated in early phase clinical trials.
Collapse
Affiliation(s)
- M Collins Scott
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston (Uthealth), USA
| | - Supinder S Bedi
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Scott D Olson
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Candice M Sears
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Charles S Cox
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| |
Collapse
|
24
|
Abstract
Positron emission tomography (PET) is a non-invasive imaging technology employed to describe metabolic, physiological, and biochemical processes in vivo. These include receptor availability, metabolic changes, neurotransmitter release, and alterations of gene expression in the brain. Since the introduction of dedicated small-animal PET systems along with the development of many novel PET imaging probes, the number of PET studies using rats and mice in basic biomedical research tremendously increased over the last decade. This article reviews challenges and advances of quantitative rodent brain imaging to make the readers aware of its physical limitations, as well as to inspire them for its potential applications in preclinical research. In the first section, we briefly discuss the limitations of small-animal PET systems in terms of spatial resolution and sensitivity and point to possible improvements in detector development. In addition, different acquisition and post-processing methods used in rodent PET studies are summarized. We further discuss factors influencing the test-retest variability in small-animal PET studies, e.g., different receptor quantification methodologies which have been mainly translated from human to rodent receptor studies to determine the binding potential and changes of receptor availability and radioligand affinity. We further review different kinetic modeling approaches to obtain quantitative binding data in rodents and PET studies focusing on the quantification of endogenous neurotransmitter release using pharmacological interventions. While several studies have focused on the dopamine system due to the availability of several PET tracers which are sensitive to dopamine release, other neurotransmitter systems have become more and more into focus and are described in this review, as well. We further provide an overview of latest genome engineering technologies, including the CRISPR/Cas9 and DREADD systems that may advance our understanding of brain disorders and function and how imaging has been successfully applied to animal models of human brain disorders. Finally, we review the strengths and opportunities of simultaneous PET/magnetic resonance imaging systems to study drug-receptor interactions and challenges for the translation of PET results from bench to bedside.
Collapse
|
25
|
Plavén-Sigray P, Matheson GJ, Coughlin JM, Hafizi S, Laurikainen H, Ottoy J, De Picker L, Rusjan P, Hietala J, Howes OD, Mizrahi R, Morrens M, Pomper MG, Cervenka S. Meta-analysis of the Glial Marker TSPO in Psychosis Revisited: Reconciling Inconclusive Findings of Patient-Control Differences. Biol Psychiatry 2021; 89:e5-e8. [PMID: 32682565 PMCID: PMC7899168 DOI: 10.1016/j.biopsych.2020.05.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/12/2020] [Accepted: 05/17/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Pontus Plavén-Sigray
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden,Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Granville J. Matheson
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Jennifer M. Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Sina Hafizi
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Heikki Laurikainen
- Department of Psychiatry, University of Turku and Neuropsychiatric Imaging Group, Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Julie Ottoy
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Livia De Picker
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | - Pablo Rusjan
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Jarmo Hietala
- Department of Psychiatry, University of Turku and Neuropsychiatric Imaging Group, Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Oliver D. Howes
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London,MRC London Institute of Medical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom,Hammersmith Hospital; and Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Romina Mizrahi
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | - Martin G. Pomper
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Simon Cervenka
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden.
| |
Collapse
|
26
|
Graykowski D, Cudaback E. Don't know what you got till it's gone: microglial depletion and neurodegeneration. Neural Regen Res 2021; 16:1921-1927. [PMID: 33642360 PMCID: PMC8343303 DOI: 10.4103/1673-5374.308078] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In the central nervous system, immunologic surveillance and response are carried out, in large part, by microglia. These resident macrophages derive from myeloid precursors in the embryonic yolk sac, migrating to the brain and eventually populating local tissue prior to blood-brain barrier formation. Preserved for the duration of lifespan, microglia serve the host as more than just a central arm of innate immunity, also contributing significantly to the development and maintenance of neurons and neural networks, as well as neuroregeneration. The critical nature of these varied functions makes the characterization of key roles played by microglia in neurodegenerative disorders, especially Alzheimer's disease, of paramount importance. While genetic models and rudimentary pharmacologic approaches for microglial manipulation have greatly improved our understanding of central nervous system health and disease, significant advances in the selective and near complete in vitro and in vivo depletion of microglia for neuroscience application continue to push the boundaries of research. Here we discuss the research efficacy and utility of various microglial depletion strategies, including the highly effective CSF1R inhibitor models, noteworthy insights into the relationship between microglia and neurodegeneration, and the potential for therapeutic repurposing of microglial depletion and repopulation.
Collapse
Affiliation(s)
- David Graykowski
- Department of Health Sciences, DePaul University, Chicago, IL, USA
| | - Eiron Cudaback
- Department of Health Sciences, DePaul University, Chicago, IL, USA
| |
Collapse
|
27
|
Woodcock EA, Hillmer AT, Sandiego CM, Maruff P, Carson RE, Cosgrove KP, Pietrzak RH. Acute neuroimmune stimulation impairs verbal memory in adults: A PET brain imaging study. Brain Behav Immun 2021; 91:784-787. [PMID: 33002632 PMCID: PMC7749814 DOI: 10.1016/j.bbi.2020.09.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/01/2020] [Accepted: 09/23/2020] [Indexed: 01/14/2023] Open
Abstract
Psychiatric and neurologic disorders are often characterized by both neuroinflammation and cognitive dysfunction. To date, however, the relationship between neuroinflammation and cognitive dysfunction remains understudied in humans. Preclinical research indicates that experimental induction of neuroinflammation reliably impairs memory processes. In this paradigm development study, we translated those robust preclinical findings to humans using positron emission tomography (PET) imaging with [11C]PBR28, a marker of microglia, and lipopolysaccharide (LPS), a potent neuroimmune stimulus. In a sample of 18 healthy adults, we extended our previous findings that LPS administration increased whole-brain [11C]PBR28 availability by 31-50%, demonstrating a robust neuroimmune response (Cohen's ds > 1.6). We now show that LPS specifically impaired verbal learning and recall, hippocampal memory processes, by 11% and 22%, respectively (Cohen's ds > 0.9), but did not alter attention, motor, or executive processes. The LPS-induced increase in [11C]PBR28 binding was correlated with significantly greater decrements in verbal learning performance in the hippocampus (r = -0.52, p = .028), putamen (r = -0.50, p = .04), and thalamus (r = -0.55, p = .02). This experimental paradigm may be useful in investigating mechanistic relationships between neuroinflammatory signaling and cognitive dysfunction in psychiatric and neurologic disorders. It may also provide a direct approach to evaluate medications designed to rescue cognitive deficits associated with neuroinflammatory dysfunction.
Collapse
Affiliation(s)
- Eric A. Woodcock
- Department of Psychiatry, Yale School of Medicine, New Haven, CT
| | - Ansel T. Hillmer
- Department of Psychiatry, Yale School of Medicine, New Haven, CT,Yale PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT
| | | | - Paul Maruff
- Cogstate Ltd., and The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia
| | - Richard E. Carson
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT
| | - Kelly P. Cosgrove
- Department of Psychiatry, Yale School of Medicine, New Haven, CT,Yale PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT,Correspondence to: Kelly P. Cosgrove, PhD, 2 Church Street S., Suite 511, New Haven, CT, 06519, Phone: 203.737.6969,
| | - Robert H. Pietrzak
- Department of Psychiatry, Yale School of Medicine, New Haven, CT,U.S. Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT
| |
Collapse
|
28
|
Enomoto S, Kato TA. Involvement of microglia in disturbed fear memory regulation: Possible microglial contribution to the pathophysiology of posttraumatic stress disorder. Neurochem Int 2020; 142:104921. [PMID: 33232758 DOI: 10.1016/j.neuint.2020.104921] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
Microglia, immune cells in the brain, play a crucial role in brain inflammation and synaptic plasticity by releasing inflammatory mediators and neurotrophic factors as well as, phagocytosing synaptic elements. Recent studies have shown peripheral inflammation, immune alteration in the brain are associated with post-traumatic stress disorder (PTSD) in humans. Several preclinical studies using Pavlovian fear conditioning have suggested that microglia are involved in fear memory dysregulation and altered fear neuronal networks. Microglial priming resulting from previous stressful experiences may also have an effect. This review will introduce the current knowledge of microglial contribution to disturbed fear memory regulation, a fundamental feature of PTSD.
Collapse
Affiliation(s)
- Shingo Enomoto
- Self Defense Force, Fukuoka Hospital, 1-61 Kokura Higashi, Kasuga-Si, Fukuoka, 816-0826, Japan; Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Takahiro A Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-Ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
29
|
Green KN, Crapser JD, Hohsfield LA. To Kill a Microglia: A Case for CSF1R Inhibitors. Trends Immunol 2020; 41:771-784. [PMID: 32792173 PMCID: PMC7484341 DOI: 10.1016/j.it.2020.07.001] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
Microglia, the brain's immune sentinels, have garnered much attention in recent years. Researchers have begun to identify the manifold roles that these cells play in the central nervous system (CNS), and this work has been greatly facilitated by microglial depletion paradigms. The varying degrees of spatiotemporal manipulation afforded by such techniques allow microglial ablation before, during, and/or following insult, injury, or disease. We review the major methods of microglial depletion, including toxin-based, genetic, and pharmacological approaches, which differ in key factors including depletion onset, duration, and off-target effects. We conclude that pharmacological CSF1R inhibitors afford the most extensive versatility in manipulating microglia, making them ideal candidates for future studies investigating microglial function in health and disease.
Collapse
Affiliation(s)
- Kim N Green
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA.
| | - Joshua D Crapser
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Lindsay A Hohsfield
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| |
Collapse
|
30
|
Laurell GL, Plavén-Sigray P, Jucaite A, Varrone A, Cosgrove KP, Svarer C, Knudsen GM, Ogden RT, Zanderigo F, Cervenka S, Hillmer AT, Schain M. Nondisplaceable Binding Is a Potential Confounding Factor in 11C-PBR28 Translocator Protein PET Studies. J Nucl Med 2020; 62:412-417. [PMID: 32680926 DOI: 10.2967/jnumed.120.243717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/23/2020] [Indexed: 01/08/2023] Open
Abstract
The PET ligand 11C-PBR28 (N-((2-(methoxy-11C)-phenyl)methyl)-N-(6-phenoxy-3-pyridinyl)acetamide) binds to the 18-kDa translocator protein (TSPO), a biomarker of glia. In clinical studies of TSPO, the ligand total distribution volume, VT, is frequently the reported outcome measure. Since VT is the sum of the ligand-specific distribution volume (VS) and the nondisplaceable-binding distribution volume (VND), differences in VND across subjects and groups will have an impact on VT Methods: Here, we used a recently developed method for simultaneous estimation of VND (SIME) to disentangle contributions from VND and VS Data from 4 previously published 11C-PBR28 PET studies were included: before and after a lipopolysaccharide challenge (8 subjects), in alcohol use disorder (14 patients, 15 controls), in first-episode psychosis (16 patients, 16 controls), and in Parkinson disease (16 patients, 16 controls). In each dataset, regional VT estimates were obtained with a standard 2-tissue-compartment model, and brain-wide VND was estimated with SIME. VS was then calculated as VT - VND VND and VS were then compared across groups, within each dataset. Results: A lower VND was found for individuals with alcohol-use disorder (34%, P = 0.00084) and Parkinson disease (34%, P = 0.0032) than in their corresponding controls. We found no difference in VND between first-episode psychosis patients and their controls, and the administration of lipopolysaccharide did not change VND Conclusion: Our findings suggest that in TSPO PET studies, nondisplaceable binding can differ between patient groups and conditions and should therefore be considered.
Collapse
Affiliation(s)
- Gjertrud L Laurell
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Pontus Plavén-Sigray
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Aurelija Jucaite
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden.,PET Science Centre, Precision Medicine and Genomics, R&D, AstraZeneca, Stockholm, Sweden
| | - Andrea Varrone
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Kelly P Cosgrove
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut.,Department of Psychiatry, Yale University, New Haven, Connecticut
| | - Claus Svarer
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Gitte M Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - R Todd Ogden
- Department of Biostatistics, Columbia University, New York, New York.,Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York
| | - Francesca Zanderigo
- Department of Biostatistics, Columbia University, New York, New York.,Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, New York; and
| | - Simon Cervenka
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Ansel T Hillmer
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut.,Department of Psychiatry, Yale University, New Haven, Connecticut.,Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Martin Schain
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
31
|
Bhatt S, Hillmer AT, Girgenti MJ, Rusowicz A, Kapinos M, Nabulsi N, Huang Y, Matuskey D, Angarita GA, Esterlis I, Davis MT, Southwick SM, Friedman MJ, Duman RS, Carson RE, Krystal JH, Pietrzak RH, Cosgrove KP. PTSD is associated with neuroimmune suppression: evidence from PET imaging and postmortem transcriptomic studies. Nat Commun 2020; 11:2360. [PMID: 32398677 PMCID: PMC7217830 DOI: 10.1038/s41467-020-15930-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 03/31/2020] [Indexed: 12/22/2022] Open
Abstract
Despite well-known peripheral immune activation in posttraumatic stress disorder (PTSD), there are no studies of brain immunologic regulation in individuals with PTSD. [11C]PBR28 Positron Emission Tomography brain imaging of the 18-kDa translocator protein (TSPO), a microglial biomarker, was conducted in 23 individuals with PTSD and 26 healthy individuals-with or without trauma exposure. Prefrontal-limbic TSPO availability in the PTSD group was negatively associated with PTSD symptom severity and was significantly lower than in controls. Higher C-reactive protein levels were also associated with lower prefrontal-limbic TSPO availability and PTSD severity. An independent postmortem study found no differential gene expression in 22 PTSD vs. 22 controls, but showed lower relative expression of TSPO and microglia-associated genes TNFRSF14 and TSPOAP1 in a female PTSD subgroup. These findings suggest that peripheral immune activation in PTSD is associated with deficient brain microglial activation, challenging prevailing hypotheses positing neuroimmune activation as central to stress-related pathophysiology.
Collapse
Affiliation(s)
- Shivani Bhatt
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, 06510, USA
| | - Ansel T Hillmer
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Matthew J Girgenti
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Aleksandra Rusowicz
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Michael Kapinos
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - David Matuskey
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Gustavo A Angarita
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Irina Esterlis
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, 06510, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Margaret T Davis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Steven M Southwick
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | | | - Ronald S Duman
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - John H Krystal
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Robert H Pietrzak
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Kelly P Cosgrove
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, 06510, USA.
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA.
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA.
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA.
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, 06516, USA.
| |
Collapse
|
32
|
Elevated serum chemokine CCL22 levels in first-episode psychosis: associations with symptoms, peripheral immune state and in vivo brain glial cell function. Transl Psychiatry 2020; 10:94. [PMID: 32179746 PMCID: PMC7075957 DOI: 10.1038/s41398-020-0776-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/18/2020] [Accepted: 03/03/2020] [Indexed: 02/08/2023] Open
Abstract
Several lines of research support immune system dysregulation in psychotic disorders. However, it remains unclear whether the immunological marker alterations are stable and how they associate with brain glial cell function. This longitudinal study aimed at investigating whether peripheral immune functions are altered in the early phases of psychotic disorders, whether the changes are associated with core symptoms, remission, brain glial cell function, and whether they persist in a one-year follow-up. Two independent cohorts comprising in total of 129 first-episode psychosis (FEP) patients and 130 controls were assessed at baseline and at the one-year follow-up. Serum cyto-/chemokines were measured using a 38-plex Luminex assay. The FEP patients showed a marked increase in chemokine CCL22 levels both at baseline (p < 0.0001; Cohen's d = 0.70) and at the 12-month follow-up (p = 0.0007) compared to controls. The group difference remained significant (p = 0.0019) after accounting for relevant covariates including BMI, smoking, and antipsychotic medication. Elevated serum CCL22 levels were significantly associated with hallucinations (ρ = 0.20) and disorganization (ρ = 0.23), and with worse verbal performance (ρ = -0.23). Brain glial cell activity was indexed with positron emission tomography and the translocator protein radiotracer [11C]PBR28 in subgroups of 15 healthy controls and 14 FEP patients with serum CCL22/CCL17 measurements. The distribution volume (VT) of [11C]PBR28 was lower in patients compared to controls (p = 0.026; Cohen's d = 0.94) without regionally specific effects, and was inversely associated with serum CCL22 and CCL17 levels (p = 0.036). Our results do not support the over-active microglia hypothesis of psychosis, but indicate altered CCR4 immune signaling in early psychosis with behavioral correlates possibly mediated through cross-talk between chemokine networks and dysfunctional or a decreased number of glial cells.
Collapse
|
33
|
Nettis MA, Veronese M, Nikkheslat N, Mariani N, Lombardo G, Sforzini L, Enache D, Harrison NA, Turkheimer FE, Mondelli V, Pariante CM. PET imaging shows no changes in TSPO brain density after IFN-α immune challenge in healthy human volunteers. Transl Psychiatry 2020; 10:89. [PMID: 32152285 PMCID: PMC7063038 DOI: 10.1038/s41398-020-0768-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/18/2020] [Accepted: 02/24/2020] [Indexed: 12/22/2022] Open
Abstract
Depression is associated with peripheral inflammation, but its link with brain microglial activity remains unclear. In seven healthy males, we used repeated translocator protein-Positron Emission Tomography (TSPO-PET) dynamic scans with [11C]PBR28 to image brain microglial activation before and 24 h after the immune challenge interferon (IFN)-α. We also investigated the association between changes in peripheral inflammation, changes in microglial activity, and changes in mood. IFN-α administration decreased [11C]PBR28 PET tissue volume of distribution (Vt) across the brain (-20 ± 4%; t6 = 4.1, p = 0.01), but after correction for radioligand free-plasma fraction there were no longer any changes (+23 ± 31%; t = 0.1, p = 0.91). IFN-α increased serum IL-6 (1826 ± 513%, t6 = -7.5, p < 0.001), IL-7 (39 ± 12%, t6 = -3.6, p = 0.01), IL-10 (328 ± 48%, t6 = -12.8, p < 0.001), and IFN-γ (272 ± 64%, t6 = -7.0, p < 0.001) at 4-6 h, and increased serum TNF-α (49 ± 7.6%, t6 = -7.5, p < 0.001), IL-8 (39 ± 12%, t6 = -3.5, p = 0.013), and C-reactive protein (1320 ± 459%, t6 = -7.2, p < 0.001) at 24 h. IFN-α induced temporary mood changes and sickness symptoms after 4-6 h, measured as an increase in POMS-2 total mood score, confusion and fatigue, and a decrease in vigor and friendliness (all p ≤ 0.04). No association was found between changes in peripheral inflammation and changes in PET or mood measures. Our work suggests that brain TSPO-PET signal is highly dependent of inflammation-induced changes in ligand binding to plasma proteins. This limits its usefulness as a sensitive marker of neuroinflammation and consequently, data interpretation. Thus, our results can be interpreted as showing either that [11C]PBR28 is not sensitive enough under these conditions, or that there is simply no microglial activation in this model.
Collapse
Affiliation(s)
- M A Nettis
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK.
- National Institute for Health and Research Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK.
| | - M Veronese
- National Institute for Health and Research Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
- Institute of Psychiatry, Psychology and Neuroscience, King's College London Department of Neuroimaging, London, UK
| | - N Nikkheslat
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK
| | - N Mariani
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK
| | - G Lombardo
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK
| | - L Sforzini
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK
- Universita' degli Studi di Milano, Psychiatry Unit, Department of Biomedical and Clinical Sciences, Luigi Sacco Hospital, Milan, Italy
| | - D Enache
- Karolinska Institute, Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Stockholm, Sweden
| | - N A Harrison
- Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff University, Cardiff, UK
| | - F E Turkheimer
- National Institute for Health and Research Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
- Institute of Psychiatry, Psychology and Neuroscience, King's College London Department of Neuroimaging, London, UK
| | - V Mondelli
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK
- National Institute for Health and Research Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - C M Pariante
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK
- National Institute for Health and Research Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
34
|
Abstract
Microglia are resident macrophages of the CNS that are involved in its development, homeostasis and response to infection and damage. Microglial activation is a common feature of neurological disorders, and although in some instances this activation can be damaging, protective and regenerative functions of microglia have been revealed. The most prominent example of the regenerative functions is a role for microglia in supporting regeneration of myelin after injury, a process that is critical for axonal health and relevant to numerous disorders in which loss of myelin integrity is a prevalent feature, such as multiple sclerosis, Alzheimer disease and motor neuron disease. Although drugs that are intended to promote remyelination are entering clinical trials, the mechanisms by which remyelination is controlled and how microglia are involved are not completely understood. In this Review, we discuss work that has identified novel regulators of microglial activation - including molecular drivers, population heterogeneity and turnover - that might influence their pro-remyelination capacity. We also discuss therapeutic targeting of microglia as a potential approach to promoting remyelination.
Collapse
|
35
|
Nettis MA, Pariante CM. Is there neuroinflammation in depression? Understanding the link between the brain and the peripheral immune system in depression. STRESS AND BRAIN HEALTH: IN CLINICAL CONDITIONS 2020; 152:23-40. [DOI: 10.1016/bs.irn.2019.12.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
36
|
Substrate Reduction Therapy for Sandhoff Disease through Inhibition of Glucosylceramide Synthase Activity. Mol Ther 2019; 27:1495-1506. [PMID: 31208914 DOI: 10.1016/j.ymthe.2019.05.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/10/2019] [Accepted: 05/19/2019] [Indexed: 12/12/2022] Open
Abstract
Neuronopathic glycosphingolipidoses are a sub-group of lysosomal storage disorders for which there are presently no effective therapies. Here, we evaluated the potential of substrate reduction therapy (SRT) using an inhibitor of glucosylceramide synthase (GCS) to decrease the synthesis of glucosylceramide (GL1) and related glycosphingolipids. The substrates that accumulate in Sandhoff disease (e.g., ganglioside GM2 and its nonacylated derivative, lyso-GM2) are distal to the drug target, GCS. Treatment of Sandhoff mice with a GCS inhibitor that has demonstrated CNS access (Genz-682452) reduced the accumulation of GL1 and GM2, as well as a variety of disease-associated substrates in the liver and brain. Concomitant with these effects was a significant decrease in the expression of CD68 and glycoprotein non-metastatic melanoma B protein (Gpnmb) in the brain, indicating a reduction in microgliosis in the treated mice. Moreover, using in vivo imaging, we showed that the monocytic biomarker translocator protein (TSPO), which was elevated in Sandhoff mice, was normalized following Genz-682452 treatment. These positive effects translated in turn into a delay (∼28 days) in loss of motor function and coordination, as measured by rotarod latency, and a significant increase in longevity (∼17.5%). Together, these results support the development of SRT for the treatment of gangliosidoses, particularly in patients with residual enzyme activity.
Collapse
|
37
|
Woodcock EA, Hillmer AT, Mason GF, Cosgrove KP. Imaging Biomarkers of the Neuroimmune System among Substance Use Disorders: A Systematic Review. MOLECULAR NEUROPSYCHIATRY 2019; 5:125-146. [PMID: 31312635 PMCID: PMC6597912 DOI: 10.1159/000499621] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/13/2019] [Indexed: 12/14/2022]
Abstract
There is tremendous interest in the role of the neuroimmune system and inflammatory processes in substance use disorders (SUDs). Imaging biomarkers of the neuroimmune system in vivo provide a vital translational bridge between preclinical and clinical research. Herein, we examine two imaging techniques that measure putative indices of the neuroimmune system and review their application among SUDs. Positron emission tomography (PET) imaging of 18 kDa translocator protein availability is a marker associated with microglia. Proton magnetic resonance spectroscopy quantification of myo-inositol levels is a putative glial marker found in astrocytes. Neuroinflammatory responses are initiated and maintained by microglia and astrocytes, and thus represent important imaging markers. The goal of this review is to summarize neuroimaging findings from the substance use literature that report data using these markers and discuss possible mechanisms of action. The extant literature indicates abused substances exert diverse and complex neuroimmune effects. Moreover, drug effects may change across addiction stages, i.e. the neuroimmune effects of acute drug administration may differ from chronic use. This burgeoning field has considerable potential to improve our understanding and treatment of SUDs. Future research is needed to determine how targeting the neuroimmune system may improve treatment outcomes.
Collapse
Affiliation(s)
| | | | | | - Kelly P. Cosgrove
- Departments of Psychiatry, and of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
38
|
Elmore MRP, Hohsfield LA, Kramár EA, Soreq L, Lee RJ, Pham ST, Najafi AR, Spangenberg EE, Wood MA, West BL, Green KN. Replacement of microglia in the aged brain reverses cognitive, synaptic, and neuronal deficits in mice. Aging Cell 2018; 17:e12832. [PMID: 30276955 PMCID: PMC6260908 DOI: 10.1111/acel.12832] [Citation(s) in RCA: 231] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 07/02/2018] [Accepted: 07/21/2018] [Indexed: 12/11/2022] Open
Abstract
Microglia, the resident immune cell of the brain, can be eliminated via pharmacological inhibition of the colony‐stimulating factor 1 receptor (CSF1R). Withdrawal of CSF1R inhibition then stimulates microglial repopulation, effectively replacing the microglial compartment. In the aged brain, microglia take on a “primed” phenotype and studies indicate that this coincides with age‐related cognitive decline. Here, we investigated the effects of replacing the aged microglial compartment with new microglia using CSF1R inhibitor‐induced microglial repopulation. With 28 days of repopulation, replacement of resident microglia in aged mice (24 months) improved spatial memory and restored physical microglial tissue characteristics (cell densities and morphologies) to those found in young adult animals (4 months). However, inflammation‐related gene expression was not broadly altered with repopulation nor the response to immune challenges. Instead, microglial repopulation resulted in a reversal of age‐related changes in neuronal gene expression, including expression of genes associated with actin cytoskeleton remodeling and synaptogenesis. Age‐related changes in hippocampal neuronal complexity were reversed with both microglial elimination and repopulation, while microglial elimination increased both neurogenesis and dendritic spine densities. These changes were accompanied by a full rescue of age‐induced deficits in long‐term potentiation with microglial repopulation. Thus, several key aspects of the aged brain can be reversed by acute noninvasive replacement of microglia.
Collapse
Affiliation(s)
- Monica R. P. Elmore
- Department of Neurobiology and Behavior; University of California; Irvine California
- Institute for Memory Impairments and Neurological Disorders (UCI MIND); Irvine California
| | - Lindsay A. Hohsfield
- Department of Neurobiology and Behavior; University of California; Irvine California
- Institute for Memory Impairments and Neurological Disorders (UCI MIND); Irvine California
| | - Enikö A. Kramár
- Department of Neurobiology and Behavior; University of California; Irvine California
| | - Lilach Soreq
- University College London; London UK
- The Francis Crick Institute; London UK
| | - Rafael J. Lee
- Department of Neurobiology and Behavior; University of California; Irvine California
- Institute for Memory Impairments and Neurological Disorders (UCI MIND); Irvine California
| | - Stephanie T. Pham
- Department of Neurobiology and Behavior; University of California; Irvine California
- Institute for Memory Impairments and Neurological Disorders (UCI MIND); Irvine California
| | - Allison R. Najafi
- Department of Neurobiology and Behavior; University of California; Irvine California
- Institute for Memory Impairments and Neurological Disorders (UCI MIND); Irvine California
| | - Elizabeth E. Spangenberg
- Department of Neurobiology and Behavior; University of California; Irvine California
- Institute for Memory Impairments and Neurological Disorders (UCI MIND); Irvine California
| | - Marcelo A. Wood
- Department of Neurobiology and Behavior; University of California; Irvine California
| | | | - Kim N. Green
- Department of Neurobiology and Behavior; University of California; Irvine California
- Institute for Memory Impairments and Neurological Disorders (UCI MIND); Irvine California
| |
Collapse
|
39
|
Abstract
Viral infection in the brain can be acute or chronic, with the responses often producing foci of increasingly cytotoxic inflammation. This can lead to effects beyond the central nervous system (CNS). To stimulate discussion, this commentary addresses four questions: What drives the development of human immunodeficiency virus (HIV)-associated neurocognitive disorders, does the phenotype of macrophages in the CNS spur development of HIV encephalitis (HIVE), does continual activation of astrocytes drive the development of HIV-associated neurocognitive disorders/subclinical disease, and neuroinflammation: friend or foe? A unifying theory that connects each question is the issue of continued activation of glial cells, even in the apparent absence of simian immunodeficiency virus/HIV in the CNS. As the CNS innate immune system is distinct from the rest of the body, it is likely there could be a number of activation profiles not observed elsewhere.
Collapse
Affiliation(s)
- Elizabeth C. Delery
- Tulane National Primate Research Center, Covington, Louisiana
- Tulane Program in Biomedical Sciences, Tulane Medical School, New Orleans, Louisiana
- Department of Microbiology and Immunology, Tulane Medical School, New Orleans, Louisiana
| | - Andrew G. MacLean
- Tulane National Primate Research Center, Covington, Louisiana
- Tulane Program in Biomedical Sciences, Tulane Medical School, New Orleans, Louisiana
- Department of Microbiology and Immunology, Tulane Medical School, New Orleans, Louisiana
- Tulane Brain Institute, Tulane University, New Orleans, Louisiana
- Center for Aging, School of Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
40
|
TSPO in diverse CNS pathologies and psychiatric disease: A critical review and a way forward. Pharmacol Ther 2018; 194:44-58. [PMID: 30189290 DOI: 10.1016/j.pharmthera.2018.09.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The use of Translocator Protein 18 kDa (TSPO) as a clinical neuroimaging biomarker of brain injury and neuroinflammation has increased exponentially in the last decade. There has been a furious pace in the development of new radiotracers for TSPO positron emission tomography (PET) imaging and its use has now been extensively described in many neurological and mental disorders. This fast pace of research and the ever-increasing number of new laboratories entering the field often times lack an appreciation of the historical perspective of the field and introduce dogmatic, but unproven facts, related to the underlying neurobiology of the TSPO response to brain injury and neuroinflammation. Paradoxically, while in neurodegenerative disorders and in all types of CNS pathologies brain TSPO levels increase, a new observation in psychiatric disorders such as schizophrenia is decreased brain levels of TSPO measured by PET. The neurobiological bases for this new finding is currently not known, but rigorous experimental design using multiple experimental approaches and careful interpretation of results is critically important to provide the methodological and/or biological underpinnings to this new observation. This review provides a perspective of the early history of validating TSPO as a biomarker of brain injury and neuroinflammation and a critical analysis of controversial topics in the literature related to the cellular sources of the TSPO response. The latter is important in order to provide the correct interpretation of PET studies in neurodegenerative and psychiatric disorders. Furthermore, this review proposes some yet to be explored explanations to new findings in psychiatric disorders and new approaches to quantitatively assess the glial sources of the TSPO response in order to move the field forward.
Collapse
|
41
|
Schrepf A, Kaplan CM, Ichesco E, Larkin T, Harte SE, Harris RE, Murray AD, Waiter GD, Clauw DJ, Basu N. A multi-modal MRI study of the central response to inflammation in rheumatoid arthritis. Nat Commun 2018; 9:2243. [PMID: 29884867 PMCID: PMC5993749 DOI: 10.1038/s41467-018-04648-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 05/14/2018] [Indexed: 12/17/2022] Open
Abstract
It is unknown how chronic inflammation impacts the brain. Here, we examined whether higher levels of peripheral inflammation were associated with brain connectivity and structure in 54 rheumatoid arthritis patients using functional and structural MRI. We show that higher levels of inflammation are associated with more positive connections between the inferior parietal lobule (IPL), medial prefrontal cortex, and multiple brain networks, as well as reduced IPL grey matter, and that these patterns of connectivity predicted fatigue, pain and cognitive dysfunction. At a second scan 6 months later, some of the same patterns of connectivity were again associated with higher peripheral inflammation. A graph theoretical analysis of whole-brain functional connectivity revealed a pattern of connections spanning 49 regions, including the IPL and medial frontal cortex, that are associated with peripheral inflammation. These regions may play a critical role in transducing peripheral inflammatory signals to the central changes seen in rheumatoid arthritis. Many diseases, such as rheumatoid arthritis, are characterized by a chronic inflammatory state, but it is not clear whether or how this affects the brain. Here, the authors show that the severity of on-going inflammation predicts altered functional brain connectivity in people with rheumatoid arthritis.
Collapse
Affiliation(s)
- Andrew Schrepf
- Chronic Pain and Fatigue Research Center, Department of Anesthesiology, University of Michigan Health System, Domino's Farms, Lobby M, PO Box 385, 24 Frank Lloyd Wright Drive, Ann Arbor, MI, 48106, USA.
| | - Chelsea M Kaplan
- Chronic Pain and Fatigue Research Center, Department of Anesthesiology, University of Michigan Health System, Domino's Farms, Lobby M, PO Box 385, 24 Frank Lloyd Wright Drive, Ann Arbor, MI, 48106, USA
| | - Eric Ichesco
- Chronic Pain and Fatigue Research Center, Department of Anesthesiology, University of Michigan Health System, Domino's Farms, Lobby M, PO Box 385, 24 Frank Lloyd Wright Drive, Ann Arbor, MI, 48106, USA
| | - Tony Larkin
- Chronic Pain and Fatigue Research Center, Department of Anesthesiology, University of Michigan Health System, Domino's Farms, Lobby M, PO Box 385, 24 Frank Lloyd Wright Drive, Ann Arbor, MI, 48106, USA
| | - Steven E Harte
- Chronic Pain and Fatigue Research Center, Department of Anesthesiology, University of Michigan Health System, Domino's Farms, Lobby M, PO Box 385, 24 Frank Lloyd Wright Drive, Ann Arbor, MI, 48106, USA
| | - Richard E Harris
- Chronic Pain and Fatigue Research Center, Department of Anesthesiology, University of Michigan Health System, Domino's Farms, Lobby M, PO Box 385, 24 Frank Lloyd Wright Drive, Ann Arbor, MI, 48106, USA
| | - Alison D Murray
- School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Gordon D Waiter
- School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Daniel J Clauw
- Chronic Pain and Fatigue Research Center, Department of Anesthesiology, University of Michigan Health System, Domino's Farms, Lobby M, PO Box 385, 24 Frank Lloyd Wright Drive, Ann Arbor, MI, 48106, USA
| | - Neil Basu
- School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, UK.,Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, G12 8QQ, UK
| |
Collapse
|
42
|
Tay TL, Béchade C, D'Andrea I, St-Pierre MK, Henry MS, Roumier A, Tremblay ME. Microglia Gone Rogue: Impacts on Psychiatric Disorders across the Lifespan. Front Mol Neurosci 2018; 10:421. [PMID: 29354029 PMCID: PMC5758507 DOI: 10.3389/fnmol.2017.00421] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022] Open
Abstract
Microglia are the predominant immune response cells and professional phagocytes of the central nervous system (CNS) that have been shown to be important for brain development and homeostasis. These cells present a broad spectrum of phenotypes across stages of the lifespan and especially in CNS diseases. Their prevalence in all neurological pathologies makes it pertinent to reexamine their distinct roles during steady-state and disease conditions. A major question in the field is determining whether the clustering and phenotypical transformation of microglial cells are leading causes of pathogenesis, or potentially neuroprotective responses to the onset of disease. The recent explosive growth in our understanding of the origin and homeostasis of microglia, uncovering their roles in shaping of the neural circuitry and synaptic plasticity, allows us to discuss their emerging functions in the contexts of cognitive control and psychiatric disorders. The distinct mesodermal origin and genetic signature of microglia in contrast to other neuroglial cells also make them an interesting target for the development of therapeutics. Here, we review the physiological roles of microglia, their contribution to the effects of environmental risk factors (e.g., maternal infection, early-life stress, dietary imbalance), and their impact on psychiatric disorders initiated during development (e.g., Nasu-Hakola disease (NHD), hereditary diffuse leukoencephaly with spheroids, Rett syndrome, autism spectrum disorders (ASDs), and obsessive-compulsive disorder (OCD)) or adulthood (e.g., alcohol and drug abuse, major depressive disorder (MDD), bipolar disorder (BD), schizophrenia, eating disorders and sleep disorders). Furthermore, we discuss the changes in microglial functions in the context of cognitive aging, and review their implication in neurodegenerative diseases of the aged adult (e.g., Alzheimer’s and Parkinson’s). Taking into account the recent identification of microglia-specific markers, and the availability of compounds that target these cells selectively in vivo, we consider the prospect of disease intervention via the microglial route.
Collapse
Affiliation(s)
- Tuan Leng Tay
- Institute of Neuropathology, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Catherine Béchade
- INSERM UMR-S 839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Ivana D'Andrea
- INSERM UMR-S 839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris, France.,Institut du Fer à Moulin, Paris, France
| | | | - Mathilde S Henry
- Axe Neurosciences, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - Anne Roumier
- INSERM UMR-S 839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Marie-Eve Tremblay
- Axe Neurosciences, CRCHU de Québec-Université Laval, Québec, QC, Canada.,Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
| |
Collapse
|
43
|
Radioligand binding analysis of α 2 adrenoceptors with [ 11C]yohimbine in brain in vivo: Extended Inhibition Plot correction for plasma protein binding. Sci Rep 2017; 7:15979. [PMID: 29167492 PMCID: PMC5700124 DOI: 10.1038/s41598-017-16020-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 10/23/2017] [Indexed: 11/23/2022] Open
Abstract
We describe a novel method of kinetic analysis of radioligand binding to neuroreceptors in brain in vivo, here applied to noradrenaline receptors in rat brain. The method uses positron emission tomography (PET) of [11C]yohimbine binding in brain to quantify the density and affinity of α2 adrenoceptors under condition of changing radioligand binding to plasma proteins. We obtained dynamic PET recordings from brain of Spraque Dawley rats at baseline, followed by pharmacological challenge with unlabeled yohimbine (0.3 mg/kg). The challenge with unlabeled ligand failed to diminish radioligand accumulation in brain tissue, due to the blocking of radioligand binding to plasma proteins that elevated the free fractions of the radioligand in plasma. We devised a method that graphically resolved the masking of unlabeled ligand binding by the increase of radioligand free fractions in plasma. The Extended Inhibition Plot introduced here yielded an estimate of the volume of distribution of non-displaceable ligand in brain tissue that increased with the increase of the free fraction of the radioligand in plasma. The resulting binding potentials of the radioligand declined by 50–60% in the presence of unlabeled ligand. The kinetic unmasking of inhibited binding reflected in the increase of the reference volume of distribution yielded estimates of receptor saturation consistent with the binding of unlabeled ligand.
Collapse
|