1
|
James BC, Cox AJ, Lewohl JM. Current trends in the role of neuroinflammation & α-synuclein in alcohol use disorder: A systematic quantitative literature review. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1209-1220. [PMID: 38724887 DOI: 10.1111/acer.15340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 07/11/2024]
Abstract
The neurodegenerative effects alcohol use disorder (AUD) have been well characterized and are likely due to the long-term effects of alcohol on the brain. The molecular events that underlie regional neuronal loss are a focus of current research. Chronic inflammation in the central nervous system, termed neuroinflammation, contributes to the progressive loss of neurons in the brain. Using data from genome-wide association studies and genetic and gene expression data, α-synuclein was identified as a gene of interest for AUD almost 10 years ago. Despite this and the well-recognized role of α-synuclein in mediating neuroinflammation in other neurodegenerative diseases, its role in alcohol-induced brain damage and AUD is yet to be elucidated. This systematic literature review quantifies and analyzes relationships between AUD, α-synuclein, and neuroinflammation. The review identified fewer studies focused on the role in AUD of α-synuclein (30) than on neuroinflammation (177), with published studies heavily centered on the myeloid differentiation primary response 88 (MyD88)-dependent toll-like receptor 4 (TLR4) pathway. The systematic review revealed that no original literature investigates the roles of α-synuclein and neuroinflammation in AUD and that there are significantly fewer published articles on the role of α-synuclein in AUD than in other neuroinflammatory conditions. Studies of the role of neuroinflammation in AUD are largely centered on the TLR4 signaling cascade, followed by TLR2 and TLR3, and soluble cytokines such as IL-10, IL-1β, and TNF-α. Key research themes identified in other neurodegenerative disorders provide new insights for further investigation in AUD.
Collapse
Affiliation(s)
- Brandon C James
- School of Pharmacy and Medical Sciences, Griffith University, Brisbane, Queensland, Australia
| | - Amanda J Cox
- School of Pharmacy and Medical Sciences, Griffith University, Brisbane, Queensland, Australia
| | - Joanne M Lewohl
- School of Pharmacy and Medical Sciences, Griffith University, Brisbane, Queensland, Australia
| |
Collapse
|
2
|
Li X, Ramos-Rolón AP, Kass G, Pereira-Rufino LS, Shifman N, Shi Z, Volkow ND, Wiers CE. Imaging neuroinflammation in individuals with substance use disorders. J Clin Invest 2024; 134:e172884. [PMID: 38828729 PMCID: PMC11142750 DOI: 10.1172/jci172884] [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] [Indexed: 06/05/2024] Open
Abstract
Increasing evidence suggests a role of neuroinflammation in substance use disorders (SUDs). This Review presents findings from neuroimaging studies assessing brain markers of inflammation in vivo in individuals with SUDs. Most studies investigated the translocator protein 18 kDa (TSPO) using PET; neuroimmune markers myo-inositol, choline-containing compounds, and N-acetyl aspartate using magnetic resonance spectroscopy; and fractional anisotropy using MRI. Study findings have contributed to a greater understanding of neuroimmune function in the pathophysiology of SUDs, including its temporal dynamics (i.e., acute versus chronic substance use) and new targets for SUD treatment.
Collapse
Affiliation(s)
- Xinyi Li
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, Department of Psychiatry, Philadelphia, Pennsylvania, USA
| | - Astrid P. Ramos-Rolón
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, Department of Psychiatry, Philadelphia, Pennsylvania, USA
| | - Gabriel Kass
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, Department of Psychiatry, Philadelphia, Pennsylvania, USA
| | - Lais S. Pereira-Rufino
- Departamento de Morfologia e Genética, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Naomi Shifman
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, Department of Psychiatry, Philadelphia, Pennsylvania, USA
| | - Zhenhao Shi
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, Department of Psychiatry, Philadelphia, Pennsylvania, USA
| | - Nora D. Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland, USA
| | - Corinde E. Wiers
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, Department of Psychiatry, Philadelphia, Pennsylvania, USA
| |
Collapse
|
3
|
Berríos-Cárcamo P, Núñez S, Castañeda J, Gallardo J, Bono MR, Ezquer F. Two-Month Voluntary Ethanol Consumption Promotes Mild Neuroinflammation in the Cerebellum but Not in the Prefrontal Cortex, Hippocampus, or Striatum of Mice. Int J Mol Sci 2024; 25:4173. [PMID: 38673763 PMCID: PMC11050159 DOI: 10.3390/ijms25084173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/29/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Chronic ethanol exposure often triggers neuroinflammation in the brain's reward system, potentially promoting the drive for ethanol consumption. A main marker of neuroinflammation is the microglia-derived monocyte chemoattractant protein 1 (MCP1) in animal models of alcohol use disorder in which ethanol is forcefully given. However, there are conflicting findings on whether MCP1 is elevated when ethanol is taken voluntarily, which challenges its key role in promoting motivation for ethanol consumption. Here, we studied MCP1 mRNA levels in areas implicated in consumption motivation-specifically, the prefrontal cortex, hippocampus, and striatum-as well as in the cerebellum, a brain area highly sensitive to ethanol, of C57BL/6 mice subjected to intermittent and voluntary ethanol consumption for two months. We found a significant increase in MCP1 mRNA levels in the cerebellum of mice that consumed ethanol compared to controls, whereas no significant changes were observed in the prefrontal cortex, hippocampus, or striatum or in microglia isolated from the hippocampus and striatum. To further characterize cerebellar neuroinflammation, we measured the expression changes in other proinflammatory markers and chemokines, revealing a significant increase in the proinflammatory microRNA miR-155. Notably, other classical proinflammatory markers, such as TNFα, IL6, and IL-1β, remained unaltered, suggesting mild neuroinflammation. These results suggest that the onset of neuroinflammation in motivation-related areas is not required for high voluntary consumption in C57BL/6 mice. In addition, cerebellar susceptibility to neuroinflammation may be a trigger to the cerebellar degeneration that occurs after chronic ethanol consumption in humans.
Collapse
Affiliation(s)
- Pablo Berríos-Cárcamo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago 7610615, Chile; (J.G.); (F.E.)
| | - Sarah Núñez
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Los Leones 7510602, Chile;
- Centro Ciencia & Vida, Santiago 8580702, Chile
| | - Justine Castañeda
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile; (J.C.); (M.R.B.)
| | - Javiera Gallardo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago 7610615, Chile; (J.G.); (F.E.)
| | - María Rosa Bono
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile; (J.C.); (M.R.B.)
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago 7610615, Chile; (J.G.); (F.E.)
- Research Center for the Development of Novel Therapeutics Alternatives for Alcohol Use Disorders, Santiago 7610658, Chile
| |
Collapse
|
4
|
Deng S, Guo A, Huang Z, Guan K, Zhu Y, Chan C, Gui J, Song C, Li X. The exploration of neuroinflammatory mechanism by which CRHR2 deficiency induced anxiety disorder. Prog Neuropsychopharmacol Biol Psychiatry 2024; 128:110844. [PMID: 37640149 DOI: 10.1016/j.pnpbp.2023.110844] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/07/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023]
Abstract
Inflammation stimulates the hypothalamic-pituitary adrenal (HPA) axis and triggers glial neuroinflammatory phenotypes, which reduces monoamine neurotransmitters by activating indoleamine 2,3-dioxygenase enzyme. These changes can induce psychiatric diseases, including anxiety. Corticotropin releasing hormone receptor 2 (CRHR2) in the HPA axis is involved in the etiology of anxiety. Omega(n)-3 polyunsaturated fatty acids (PUFAs) can attenuate anxiety through anti-inflammation and HPA axis modulation. However, the underlying molecular mechanism by CRHR2 modulates anxiety and its correlation with neuroinflammation remain unclear. Here, we first constructed a crhr2 zebrafish mutant line, and evaluated anxiety-like behaviors, gene expression associated with the HPA axis, neuroinflammatory response, neurotransmitters, and PUFAs profile in crhr2+/+ and crhr2-/- zebrafish. The crhr2 deficiency decreased cortisol levels and up-regulated crhr1 and down-regulated crhb, crhbp, ucn3l and proopiomelanocortin a (pomc a) in zebrafish. Interestingly, a significant increase in the neuroinflammatory markers, translocator protein (TSPO) and the activation of microglia M1 phenotype (CD11b) were found in crhr2-/- zebrafish. As a consequence, the expression of granulocyte-macrophage colony-stimulating factor, pro-inflammatory cytokines vascular endothelial growth factor, and astrocyte A1 phenotype c3 were up-regulated. While microglia anti-inflammatory phenotype (CD206), central anti-inflammatory cytokine interleukin-4, arginase-1, and transforming growth factor-β were downregulated. In parallel, crhr2-deficient zebrafish showed an upregulation of vdac1 protein, a TSPO ligand, and its downstream caspase-3. Furthermore, 5-HT/5-HIAA ratio was decreased and n-3 PUFAs deficiency was identified in crhr2-/- zebrafish. In conclusion, anxiety-like behavior displayed by crhr2-deficient zebrafish may be caused by the HPA axis dysfunction and enhanced neuroinflammation, which resulted in n-3 PUFAs and monoamine neurotransmitter reductions.
Collapse
Affiliation(s)
- Shuyi Deng
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Anqi Guo
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou, Zhejiang 325000, China
| | - Zhengwei Huang
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou, Zhejiang 325000, China
| | - Kaiyu Guan
- Wenzhou Seventh People's Hospital, Wenzhou, Zhejiang 325000, China
| | - Ya Zhu
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou, Zhejiang 325000, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, University of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Cheekai Chan
- College of Science and Technology, Wenzhou-Kean University, Zhejiang 325000, China
| | - Jianfang Gui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, University of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Cai Song
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China.
| | - Xi Li
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
5
|
Cao H, Shinn AK, Guo W. Editorial: Cerebellar structure and function in psychotic disorders: from mechanisms to clinics. Front Psychiatry 2023; 14:1344882. [PMID: 38146281 PMCID: PMC10749355 DOI: 10.3389/fpsyt.2023.1344882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/27/2023] Open
Affiliation(s)
- Hengyi Cao
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, NY, United States
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Ann K. Shinn
- Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Wenbin Guo
- Department of Psychiatry, National Clinical Research Center for Mental Disorders and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
6
|
De Picker LJ, Morrens M, Branchi I, Haarman BCM, Terada T, Kang MS, Boche D, Tremblay ME, Leroy C, Bottlaender M, Ottoy J. TSPO PET brain inflammation imaging: A transdiagnostic systematic review and meta-analysis of 156 case-control studies. Brain Behav Immun 2023; 113:415-431. [PMID: 37543251 DOI: 10.1016/j.bbi.2023.07.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 06/26/2023] [Accepted: 07/30/2023] [Indexed: 08/07/2023] Open
Abstract
INTRODUCTION The 18-kDa translocator protein (TSPO) is increasingly recognized as a molecular target for PET imaging of inflammatory responses in various central nervous system (CNS) disorders. However, the reported sensitivity and specificity of TSPO PET to identify brain inflammatory processes appears to vary greatly across disorders, disease stages, and applied quantification methods. To advance TSPO PET as a potential biomarker to evaluate brain inflammation and anti-inflammatory therapies, a better understanding of its applicability across disorders is needed. We conducted a transdiagnostic systematic review and meta-analysis of all in vivo human TSPO PET imaging case-control studies in the CNS. Specifically, we investigated the direction, strength, and heterogeneity associated with the TSPO PET signal across disorders in pre-specified brain regions, and explored the demographic and methodological sources of heterogeneity. METHODS We searched for English peer-reviewed articles that reported in vivo human case-control TSPO PET differences. We extracted the demographic details, TSPO PET outcomes, and technical variables of the PET procedure. A random-effects meta-analysis was applied to estimate case-control standardized mean differences (SMD) of the TSPO PET signal in the lobar/whole-brain cortical grey matter (cGM), thalamus, and cortico-limbic circuitry between different illness categories. Heterogeneity was evaluated with the I2 statistic and explored using subgroup and meta-regression analyses for radioligand generation, PET quantification method, age, sex, and publication year. Significance was set at the False Discovery Rate (FDR)-corrected P < 0.05. RESULTS 156 individual case-control studies were included in the systematic review, incorporating data for 2381 healthy controls and 2626 patients. 139 studies documented meta-analysable data and were grouped into 11 illness categories. Across all the illness categories, we observed a significantly higher TSPO PET signal in cases compared to controls for the cGM (n = 121 studies, SMD = 0.358, PFDR < 0.001, I2 = 68%), with a significant difference between the illness categories (P = 0.004). cGM increases were only significant for Alzheimer's disease (SMD = 0.693, PFDR < 0.001, I2 = 64%) and other neurodegenerative disorders (SMD = 0.929, PFDR < 0.001, I2 = 73%). Cortico-limbic increases (n = 97 studies, SMD = 0.541, P < 0.001, I2 = 67%) were most prominent for Alzheimer's disease, mild cognitive impairment, other neurodegenerative disorders, mood disorders and multiple sclerosis. Thalamic involvement (n = 79 studies, SMD = 0.393, P < 0.001, I2 = 71%) was observed for Alzheimer's disease, other neurodegenerative disorders, multiple sclerosis, and chronic pain and functional disorders (all PFDR < 0.05). Main outcomes for systemic immunological disorders, viral infections, substance use disorders, schizophrenia and traumatic brain injury were not significant. We identified multiple sources of between-study variance to the TSPO PET signal including a strong transdiagnostic effect of the quantification method (explaining 25% of between-study variance; VT-based SMD = 0.000 versus reference tissue-based studies SMD = 0.630; F = 20.49, df = 1;103, P < 0.001), patient age (9% of variance), and radioligand generation (5% of variance). CONCLUSION This study is the first overarching transdiagnostic meta-analysis of case-control TSPO PET findings in humans across several brain regions. We observed robust increases in the TSPO signal for specific types of disorders, which were widespread or focal depending on illness category. We also found a large and transdiagnostic horizontal (positive) shift of the effect estimates of reference tissue-based compared to VT-based studies. Our results can support future studies to optimize experimental design and power calculations, by taking into account the type of disorder, brain region-of-interest, radioligand, and quantification method.
Collapse
Affiliation(s)
- Livia J De Picker
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Campus Duffel, Duffel, Belgium.
| | - Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Campus Duffel, Duffel, Belgium
| | - Igor Branchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Roma, Italy
| | - Bartholomeus C M Haarman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tatsuhiro Terada
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Min Su Kang
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences School, Faculty of Medicine, University of Southampton, UK
| | - Marie-Eve Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, BC, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Claire Leroy
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay (BioMaps), Orsay, France
| | - Michel Bottlaender
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay (BioMaps), Orsay, France; Université Paris-Saclay, UNIACT, Neurospin, CEA, Gif-sur-Yvette, France
| | - Julie Ottoy
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
7
|
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: 0] [Impact Index Per Article: 0] [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
|
8
|
Zhao Y, Hong Z, Lin Y, Shen W, Yang Y, Zuo Z, Hu X. Exercise pretreatment alleviates neuroinflammation and oxidative stress by TFEB-mediated autophagic flux in mice with ischemic stroke. Exp Neurol 2023; 364:114380. [PMID: 36914085 DOI: 10.1016/j.expneurol.2023.114380] [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: 01/10/2023] [Revised: 02/20/2023] [Accepted: 03/08/2023] [Indexed: 03/13/2023]
Abstract
BACKGROUND Neuroinflammation and oxidative stress are important pathological mechanisms underlying cerebral ischemic stroke. Increasing evidence suggests that regulation autophagy in ischemic stroke may improve neurological functions. In this study, we aimed to explore whether exercise pretreatment attenuates neuroinflammation and oxidative stress in ischemic stroke by improving autophagic flux. METHODS 2,3,5-Triphenyltetrazolium chloride staining was used to determine the infarction volume, and modified Neurological Severity Scores and rotarod test were used to evaluate neurological functions after ischemic stroke. The levels of oxidative stress, neuroinflammation, neuronal apoptosis and degradation, autophagic flux, and signaling pathway proteins were determined using immunofluorescence, dihydroethidium, TUNEL, and Fluoro-Jade B staining, western blotting, and co-immunoprecipitation. RESULTS Our results showed that, in middle cerebral artery occlusion (MCAO) mice, exercise pretreatment improved neurological functions and defective autophagy, and reduced neuroinflammation and oxidative stress. Mechanistically, after using chloroquine, impaired autophagy abolished the neuroprotection of exercise pretreatment. And transcription factor EB (TFEB) activation mediated by exercise pretreatment contributes to improving autophagic flux after MCAO. Furthermore, we showed that TFEB activation mediated by exercise pretreatment in MCAO was regulated by the AMPK-mTOR and AMPK-FOXO3a-SKP2-CARM1 signaling pathways. CONCLUSIONS Exercise pretreatment has the potential to improve the prognosis of ischemic stroke patients, and it can exert neuroprotective effects in ischemic stroke by inhibiting neuroinflammation and oxidative stress, which might be due to the TFEB-mediated autophagic flux. And targeting autophagic flux may be promising strategies for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yun Zhao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou 510630, Guangdong, China
| | - Zhongqiu Hong
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou 510630, Guangdong, China
| | - Yao Lin
- Department of Pediatrics, Taizhou First People's Hospital, 218 Hengjie Road, Taizhou 318020, Zhejiang, China
| | - Weimin Shen
- Department of Respiratory Care, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Qingchun East Road No. 3, Hangzhou 310016, Zhejiang, China
| | - Yuhan Yang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou 510630, Guangdong, China
| | - Zejie Zuo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou 510630, Guangdong, China.
| | - Xiquan Hu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou 510630, Guangdong, China.
| |
Collapse
|
9
|
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
|
10
|
Carlson ER, Guerin SP, Nixon K, Fonken LK. The neuroimmune system - Where aging and excess alcohol intersect. Alcohol 2023; 107:153-167. [PMID: 36150610 PMCID: PMC10023388 DOI: 10.1016/j.alcohol.2022.08.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/21/2022] [Accepted: 08/31/2022] [Indexed: 01/05/2023]
Abstract
As the percentage of the global population over age 65 grows, and with it a subpopulation of individuals with alcohol use disorder (AUD), understanding the effect of alcohol on the aged brain is of utmost importance. Neuroinflammation is implicated in both natural aging as well as alcohol use, and its role in alterations to brain morphology and function may be exacerbated in aging individuals who drink alcohol to excess. The neuroimmune response to alcohol in aging is complex. The few studies investigating this issue have reported heightened basal activity and either hypo- or hyper-reactivity to an alcohol challenge. This review of preclinical research will first introduce key players of the immune system, then explore changes in neuroimmune function with aging or alcohol alone, with discussion of vulnerable brain regions, changes in cytokines, and varied reactions of microglia and astrocytes. We will then consider different levels of alcohol exposure, relevant animal models of AUD, and neuroimmune activation by alcohol across the lifespan. By identifying key findings, challenges, and targets for future research, we hope to bring more attention and resources to this underexplored area of inquiry.
Collapse
Affiliation(s)
- Erika R Carlson
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, United States
| | - Steven P Guerin
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, United States
| | - Kimberly Nixon
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, United States
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
11
|
Adams C, Perry N, Conigrave J, Hurzeler T, Stevens J, Yacou Dunbar KP, Sweeney A, Lee K, Sutherland G, Haber P, Morley KC. Central markers of neuroinflammation in alcohol use disorder: A meta-analysis of neuroimaging, cerebral spinal fluid, and postmortem studies. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:197-208. [PMID: 36852781 DOI: 10.1111/acer.14992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 03/01/2023]
Abstract
INTRODUCTION AND AIMS There is emerging evidence that heavy long-term alcohol consumption may alter the neuroimmune profile. We conducted a meta-analysis of the association between alcohol use disorder (AUD) and the extent of neuroinflammation using cerebrospinal (CSF), PET (Positron Emission Tomography), and postmortem studies. DESIGN AND METHODS A comprehensive search of electronic databases was conducted using the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols (PRISMA-P) for AUD-related terms in combination with neuroinflammatory markers and cytokine- and chemokine-related terms for CSF, PET, and postmortem studies. Participants had to meet established criteria for AUD and/or heavy alcohol consumption with dependence features and be compared with healthy controls. Papers retrieved were assessed for inclusion criteria and a critical appraisal was completed using the Newcastle-Ottawa Scale. A meta-analysis was conducted on postmortem and PET studies. RESULTS Eleven papers met the inclusion criteria with CSF, PET, and postmortem studies included in the final analysis. Postmortem studies demonstrate significant heterogeneity (𝑄 (14) = 62.02, 𝑝 < 0.001), with the alcohol group showing higher levels of neuroimmune markers than controls (𝑑 = 1.50 [95% CI 0.56, 2.45]). PET studies demonstrated a lower [11 C] PBR28 total volume of distribution (V T ) for translocator protein in the hippocampus (g = -1.95 [95% CI -2.72, -1.18], p < 0.001) of the alcohol group compared to controls. CONCLUSION There is emerging evidence across multiple diagnostic modalities that alcohol impacts neuroimmune signaling in the human brain.
Collapse
Affiliation(s)
- Claire Adams
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Drug Health Services, Sydney Local Health District, Sydney, New South Wales, Australia
| | - Nina Perry
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - James Conigrave
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Drug Health Services, Sydney Local Health District, Sydney, New South Wales, Australia
| | - Tristan Hurzeler
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Drug Health Services, Sydney Local Health District, Sydney, New South Wales, Australia
| | - Julia Stevens
- NSW Brain Tissue Resource Centre and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Kristiane P Yacou Dunbar
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Alicia Sweeney
- NSW Brain Tissue Resource Centre and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Kylie Lee
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Drug Health Services, Sydney Local Health District, Sydney, New South Wales, Australia
| | - Greg Sutherland
- NSW Brain Tissue Resource Centre and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul Haber
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Drug Health Services, Sydney Local Health District, Sydney, New South Wales, Australia
| | - Kirsten C Morley
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Drug Health Services, Sydney Local Health District, Sydney, New South Wales, Australia
| |
Collapse
|
12
|
Bach P, de Timary P, Gründer G, Cumming P. Molecular Imaging Studies of Alcohol Use Disorder. Curr Top Behav Neurosci 2023. [PMID: 36639552 DOI: 10.1007/7854_2022_414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Alcohol use disorder (AUD) is a serious public health problem in many countries, bringing a gamut of health risks and impairments to individuals and a great burden to society. Despite the prevalence of a disease model of AUD, the current pharmacopeia does not present reliable treatments for AUD; approved treatments are confined to a narrow spectrum of medications engaging inhibitory γ-aminobutyric acid (GABA) neurotransmission and possibly excitatory N-methyl-D-aspartate (NMDA) receptors, and opioid receptor antagonists. Molecular imaging with positron emission tomography (PET) and single-photon emission computed tomography (SPECT) can open a window into the living brain and has provided diverse insights into the pathology of AUD. In this narrative review, we summarize the state of molecular imaging findings on the pharmacological action of ethanol and the neuropathological changes associated with AUD. Laboratory and preclinical imaging results highlight the interactions between ethanol and GABA A-type receptors (GABAAR), but the interpretation of such results is complicated by subtype specificity. An abundance of studies with the glucose metabolism tracer fluorodeoxyglucose (FDG) concur in showing cerebral hypometabolism after ethanol challenge, but there is relatively little data on long-term changes in AUD. Alcohol toxicity evokes neuroinflammation, which can be tracked using PET with ligands for the microglial marker translocator protein (TSPO). Several PET studies show reversible increases in TSPO binding in AUD individuals, and preclinical results suggest that opioid-antagonists can rescue from these inflammatory responses. There are numerous PET/SPECT studies showing changes in dopaminergic markers, generally consistent with an impairment in dopamine synthesis and release among AUD patients, as seen in a number of other addictions; this may reflect the composite of an underlying deficiency in reward mechanisms that predisposes to AUD, in conjunction with acquired alterations in dopamine signaling. There is little evidence for altered serotonin markers in AUD, but studies with opioid receptor ligands suggest a specific up-regulation of the μ-opioid receptor subtype. Considerable heterogeneity in drinking patterns, gender differences, and the variable contributions of genetics and pre-existing vulnerability traits present great challenges for charting the landscape of molecular imaging in AUD.
Collapse
Affiliation(s)
- Patrick Bach
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.
| | - Philippe de Timary
- Department of Adult Psychiatry, Cliniques Universitaires Saint-Luc and Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Gerhard Gründer
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, Australia
- International Centre for Education and Research in Neuropsychiatry (ICERN), Samara State Medical University, Samara, Russia
| |
Collapse
|
13
|
Mineur YS, Garcia-Rivas V, Thomas MA, Soares AR, McKee SA, Picciotto MR. Sex differences in stress-induced alcohol intake: a review of preclinical studies focused on amygdala and inflammatory pathways. Psychopharmacology (Berl) 2022; 239:2041-2061. [PMID: 35359158 PMCID: PMC9704113 DOI: 10.1007/s00213-022-06120-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/14/2022] [Indexed: 02/06/2023]
Abstract
Clinical studies suggest that women are more likely than men to relapse to alcohol drinking in response to stress; however, the mechanisms underlying this sex difference are not well understood. A number of preclinical behavioral models have been used to study stress-induced alcohol intake. Here, we review paradigms used to study effects of stress on alcohol intake in rodents, focusing on findings relevant to sex differences. To date, studies of sex differences in stress-induced alcohol drinking have been somewhat limited; however, there is evidence that amygdala-centered circuits contribute to effects of stress on alcohol seeking. In addition, we present an overview of inflammatory pathways leading to microglial activation that may contribute to alcohol-dependent behaviors. We propose that sex differences in neuronal function and inflammatory signaling in circuits centered on the amygdala are involved in sex-dependent effects on stress-induced alcohol seeking and suggest that this is an important area for future studies.
Collapse
Affiliation(s)
- Yann S Mineur
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Vernon Garcia-Rivas
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Merrilee A Thomas
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Alexa R Soares
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
- Yale Interdepartmental Neuroscience Program, New Haven, CT, USA
| | - Sherry A McKee
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA.
- Yale Interdepartmental Neuroscience Program, New Haven, CT, USA.
| |
Collapse
|
14
|
Fang YHD, McConathy JE, Yacoubian TA, Zhang Y, Kennedy RE, Standaert DG. Image Quantification for TSPO PET with a Novel Image-Derived Input Function Method. Diagnostics (Basel) 2022; 12:1161. [PMID: 35626315 PMCID: PMC9140104 DOI: 10.3390/diagnostics12051161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 01/27/2023] Open
Abstract
There is a growing interest in using 18F-DPA-714 PET to study neuroinflammation and microglial activation through imaging the 18-kDa translocator protein (TSPO). Although quantification of 18F-DPA-714 binding can be achieved through kinetic modeling analysis with an arterial input function (AIF) measured with blood sampling procedures, the invasiveness of such procedures has been an obstacle for wide application. To address these challenges, we developed an image-derived input function (IDIF) that noninvasively estimates the arterial input function from the images acquired for 18F-DPA-714 quantification. Methods: The method entails three fully automatic steps to extract the IDIF, including a segmentation of voxels with highest likelihood of being the arterial blood over the carotid artery, a model-based matrix factorization to extract the arterial blood signal, and a scaling optimization procedure to scale the extracted arterial blood signal into the activity concentration unit. Two cohorts of human subjects were used to evaluate the extracted IDIF. In the first cohort of five subjects, arterial blood sampling was performed, and the calculated IDIF was validated against the measured AIF through the comparison of distribution volumes from AIF (VT,AIF) and IDIF (VT,IDIF). In the second cohort, PET studies from twenty-eight healthy controls without arterial blood sampling were used to compare VT,IDIF with VT,REF measured using a reference region-based analysis to evaluate whether it can distinguish high-affinity (HAB) and mixed-affinity (MAB) binders. Results: In the arterial blood-sampling cohort, VT derived from IDIF was found to be an accurate surrogate of the VT from AIF. The bias of VT, IDIF was −5.8 ± 7.8% when compared to VT,AIF, and the linear mixed effect model showed a high correlation between VT,AIF and VT, IDIF (p < 0.001). In the nonblood-sampling cohort, VT, IDIF showed a significance difference between the HAB and MAB healthy controls. VT, IDIF and standard uptake values (SUV) showed superior results in distinguishing HAB from MAB subjects than VT,REF. Conclusions: A novel IDIF method for 18F-DPA-714 PET quantification was developed and evaluated in this study. This IDIF provides a noninvasive alternative measurement of VT to quantify the TSPO binding of 18F-DPA-714 in the human brain through dynamic PET scans.
Collapse
Affiliation(s)
- Yu-Hua Dean Fang
- Department of Radiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.A.Y.); (D.G.S.)
| | - Jonathan E. McConathy
- Department of Radiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Talene A. Yacoubian
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.A.Y.); (D.G.S.)
| | - Yue Zhang
- Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Y.Z.); (R.E.K.)
| | - Richard E. Kennedy
- Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (Y.Z.); (R.E.K.)
| | - David G. Standaert
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (T.A.Y.); (D.G.S.)
| |
Collapse
|
15
|
Rupprecht R, Rupprecht C, Di Benedetto B, Rammes G. Neuroinflammation and psychiatric disorders: Relevance of C1q, translocator protein (18 kDa) (TSPO), and neurosteroids. World J Biol Psychiatry 2022; 23:257-263. [PMID: 34320915 DOI: 10.1080/15622975.2021.1961503] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
There is increasing evidence that neuroinflammatory processes may play a role in the pathophysiology of psychiatric disorders. Recently, the complement protein C1q and the translocator protein (18 kDa) (TSPO) have attracted considerable interest in this context. C1q is a small molecule which is involved into synaptic pruning mechanisms, increases during ageing and may contribute to neurodegenerative disorders. TSPO is a transmembrane channel protein and mediates numerous biological functions such as bioenergetics and steroid synthesis. Meanwhile, there is evidence that both C1q and TSPO may be elevated in psychiatric disorders, e.g. major depression. Moreover, preclinical and first clinical studies suggest that TSPO ligands may exert antidepressant and anxiolytic properties by promoting endogenous neurosteroid synthesis. In addition, certain neurosteroids, e.g. allopregnanolone, are potent positive allosteric modulators of GABAA receptors and their composition is altered in depression and anxiety disorders. Recently, neurosteroid compounds such as brexanolone or zuranolone have been reported to reduce depressive and anxiety symptoms in postpartum depression and major depressive disorder. In conclusion, compounds enhancing GABAergic neurotransmission such as neurosteroids and TSPO ligands, which also may exert anti-inflammatory properties in concert with immunomodulators such as C1q may open new avenues for the treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University Regensburg, Regensburg, Germany
| | - Christian Rupprecht
- Experimental Neuropharmacology, Department of Anesthesiology, Technische Universität München, Munich, Germany
| | - Barbara Di Benedetto
- Department of Psychiatry and Psychotherapy, University Regensburg, Regensburg, Germany
| | - Gerhard Rammes
- Experimental Neuropharmacology, Department of Anesthesiology, Technische Universität München, Munich, Germany
| |
Collapse
|
16
|
Agarwal K, Manza P, Chapman M, Nawal N, Biesecker E, McPherson K, Dennis E, Johnson A, Volkow ND, Joseph PV. Inflammatory Markers in Substance Use and Mood Disorders: A Neuroimaging Perspective. Front Psychiatry 2022; 13:863734. [PMID: 35558424 PMCID: PMC9086785 DOI: 10.3389/fpsyt.2022.863734] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/17/2022] [Indexed: 12/21/2022] Open
Abstract
Chronic exposure to addictive drugs in substance use disorders and stressors in mood disorders render the brain more vulnerable to inflammation. Inflammation in the brain, or neuroinflammation, is characterized by gliosis, microglial activation, and sustained release of cytokines, chemokines, and pro-inflammatory factors compromising the permeability of the blood-brain barrier. There is increased curiosity in understanding how substance misuse and/or repeated stress exposure affect inflammation and contribute to abnormal neuronal activity, altered neuroplasticity, and impaired cognitive control, which eventually promote compulsive drug-use behaviors and worsen mood disorders. This review will emphasize human imaging studies to explore the link between brain function and peripheral markers of inflammation in substance use disorders and mood disorders.
Collapse
Affiliation(s)
- Khushbu Agarwal
- Section of Sensory Science and Metabolism Unit, Division of Intramural Research, Department of Health and Human Services, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States.,Section of Sensory Science and Metabolism, Division of Intramural Research, U.S. Department of Health and Human Services, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Peter Manza
- Laboratory of Neuroimaging, Department of Health and Human Services, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Marquis Chapman
- Section of Sensory Science and Metabolism Unit, Division of Intramural Research, Department of Health and Human Services, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Nafisa Nawal
- Section of Sensory Science and Metabolism Unit, Division of Intramural Research, Department of Health and Human Services, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Erin Biesecker
- Section of Sensory Science and Metabolism Unit, Division of Intramural Research, Department of Health and Human Services, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Katherine McPherson
- Section of Sensory Science and Metabolism Unit, Division of Intramural Research, Department of Health and Human Services, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Evan Dennis
- Section of Sensory Science and Metabolism Unit, Division of Intramural Research, Department of Health and Human Services, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Allison Johnson
- Section of Sensory Science and Metabolism Unit, Division of Intramural Research, Department of Health and Human Services, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Nora D Volkow
- Laboratory of Neuroimaging, Department of Health and Human Services, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Paule V Joseph
- Section of Sensory Science and Metabolism Unit, Division of Intramural Research, Department of Health and Human Services, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States.,Section of Sensory Science and Metabolism, Division of Intramural Research, U.S. Department of Health and Human Services, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
17
|
Morcuende A, Navarrete F, Nieto E, Manzanares J, Femenía T. Inflammatory Biomarkers in Addictive Disorders. Biomolecules 2021; 11:biom11121824. [PMID: 34944470 PMCID: PMC8699452 DOI: 10.3390/biom11121824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022] Open
Abstract
Substance use disorders are a group of diseases that are associated with social, professional, and family impairment and that represent a high socio-economic impact on the health systems of countries around the world. These disorders present a very complex diagnosis and treatment regimen due to the lack of suitable biomarkers supporting the correct diagnosis and classification and the difficulty of selecting effective therapies. Over the last few years, several studies have pointed out that these addictive disorders are associated with systemic and central nervous system inflammation, which could play a relevant role in the onset and progression of these diseases. Therefore, identifying different immune system components as biomarkers of such addictive disorders could be a crucial step to promote appropriate diagnosis and treatment. Thus, this work aims to provide an overview of the immune system alterations that may be biomarkers of various addictive disorders.
Collapse
Affiliation(s)
- Alvaro Morcuende
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| | - Elena Nieto
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| | - Teresa Femenía
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (A.M.); (F.N.); (E.N.); (J.M.)
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-965-919-553
| |
Collapse
|
18
|
Leroy C, Saba W. Contribution of TSPO imaging in the understanding of the state of gliosis in substance use disorders. Eur J Nucl Med Mol Imaging 2021; 49:186-200. [PMID: 34041563 DOI: 10.1007/s00259-021-05408-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/10/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE Recent research in last years in substance use disorders (SUD) synthesized a proinflammatory hypothesis of SUD based on reported pieces of evidence of non-neuronal central immune signalling pathways modulated by drug of abuse and that contribute to their pharmacodynamic actions. Positron emission tomography has been shown to be a precious imaging technique to study in vivo neurochemical processes involved in SUD and to highlight the central immune signalling actions of drugs of abuse. METHODS In this review, we investigate the contribution of the central immune system, with a particular focus on translocator protein 18 kDa (TSPO) imaging, associated with a series of drugs involved in substance use disorders (SUD) specifically alcohol, opioids, tobacco, methamphetamine, cocaine, and cannabis. RESULTS The large majority of preclinical and clinical studies presented in this review converges towards SUD modulation of the neuroimmune responses and TSPO expression and speculated a pivotal positioning in the pathogenesis of SUD. However, some contradictions concerning the same drug or between preclinical and clinical studies make it difficult to draw a clear picture about the significance of glial state in SUD. DISCUSSION Significant disparities in clinical and biological characteristics are present between investigated populations among studies. Heterogeneity in genetic factors and other clinical co-morbidities, difficult to be reproduced in animal models, may affect findings. On the other hand, technical aspects including study designs, radioligand limitations, or PET imaging quantification methods could impact the study results and should be considered to explain discrepancies in outcomes. CONCLUSION The supposed neuroimmune component of SUD provides new therapeutic approaches in the prediction and treatment of SUD pointing to the central immune signalling.
Collapse
Affiliation(s)
- Claire Leroy
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4 place du général Leclerc, 91401, Orsay, France
| | - Wadad Saba
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4 place du général Leclerc, 91401, Orsay, France.
| |
Collapse
|
19
|
NRM 2021 Abstract Booklet. J Cereb Blood Flow Metab 2021; 41:11-309. [PMID: 34905986 PMCID: PMC8851538 DOI: 10.1177/0271678x211061050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
20
|
Chauveau F, Becker G, Boutin H. Have (R)-[ 11C]PK11195 challengers fulfilled the promise? A scoping review of clinical TSPO PET studies. Eur J Nucl Med Mol Imaging 2021; 49:201-220. [PMID: 34387719 PMCID: PMC8712292 DOI: 10.1007/s00259-021-05425-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE The prototypical TSPO radiotracer (R)-[11C]PK11195 has been used in humans for more than thirty years to visualize neuroinflammation in several pathologies. Alternative radiotracers have been developed to improve signal-to-noise ratio and started to be tested clinically in 2008. Here we examined the scientific value of these "(R)-[11C]PK11195 challengers" in clinical research to determine if they could supersede (R)-[11C]PK11195. METHODS A systematic MEDLINE (PubMed) search was performed (up to end of year 2020) to extract publications reporting TSPO PET in patients with identified pathologies, excluding studies in healthy subjects and methodological studies. RESULTS Of the 288 publications selected, 152 used 13 challengers, and 142 used (R)-[11C]PK11195. Over the last 20 years, the number of (R)-[11C]PK11195 studies remained stable (6 ± 3 per year), but was surpassed by the total number of challenger studies for the last 6 years. In total, 3914 patients underwent a TSPO PET scan, and 47% (1851 patients) received (R)-[11C]PK11195. The 2 main challengers were [11C]PBR28 (24%-938 patients) and [18F]FEPPA (11%-429 patients). Only one-in-ten patients (11%-447) underwent 2 TSPO scans, among whom 40 (1%) were scanned with 2 different TSPO radiotracers. CONCLUSIONS Generally, challengers confirmed disease-specific initial (R)-[11C]PK11195 findings. However, while their better signal-to-noise ratio seems particularly useful in diseases with moderate and widespread neuroinflammation, most challengers present an allelic-dependent (Ala147Thr polymorphism) TSPO binding and genetic stratification is hindering their clinical implementation. As new challengers, insensitive to TSPO human polymorphism, are about to enter clinical evaluation, we propose this systematic review to be regularly updated (living review).
Collapse
Affiliation(s)
- Fabien Chauveau
- University of Lyon, Lyon Neuroscience Research Center (CRNL), CNRS UMR5292, INSERM U1028, University Lyon 1, Lyon, France.
| | - Guillaume Becker
- GIGA - CRC In Vivo Imaging, University Liege, Liege, Belgium
- University of Lyon, CarMeN Laboratory, INSERM U1060, University Lyon 1, Hospices Civils Lyon, Lyon, France
| | - Hervé Boutin
- Faculty of Biology Medicine and Health, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
| |
Collapse
|
21
|
Wimberley C, Lavisse S, Hillmer A, Hinz R, Turkheimer F, Zanotti-Fregonara P. Kinetic modeling and parameter estimation of TSPO PET imaging in the human brain. Eur J Nucl Med Mol Imaging 2021; 49:246-256. [PMID: 33693967 PMCID: PMC8712306 DOI: 10.1007/s00259-021-05248-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/07/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE Translocator protein 18-kDa (TSPO) imaging with positron emission tomography (PET) is widely used in research studies of brain diseases that have a neuro-immune component. Quantification of TSPO PET images, however, is associated with several challenges, such as the lack of a reference region, a genetic polymorphism affecting the affinity of the ligand for TSPO, and a strong TSPO signal in the endothelium of the brain vessels. These challenges have created an ongoing debate in the field about which type of quantification is most useful and whether there is an appropriate simplified model. METHODS This review focuses on the quantification of TSPO radioligands in the human brain. The various methods of quantification are summarized, including the gold standard of compartmental modeling with metabolite-corrected input function as well as various alternative models and non-invasive approaches. Their advantages and drawbacks are critically assessed. RESULTS AND CONCLUSIONS Researchers employing quantification methods for TSPO should understand the advantages and limitations associated with each method. Suggestions are given to help researchers choose between these viable alternative methods.
Collapse
Affiliation(s)
| | - Sonia Lavisse
- CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Université Paris-Saclay, 92265, Fontenay-aux-Roses, France
| | - Ansel Hillmer
- Departments of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Departments of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, USA
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, M20 3LJ, UK
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Centre for Neuroimaging Sciences, King's College London, De Crespigny Park, London, SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE1 1UL, UK
| | - Paolo Zanotti-Fregonara
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
22
|
Nutt D, Hayes A, Fonville L, Zafar R, Palmer EO, Paterson L, Lingford-Hughes A. Alcohol and the Brain. Nutrients 2021; 13:3938. [PMID: 34836193 PMCID: PMC8625009 DOI: 10.3390/nu13113938] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/20/2022] Open
Abstract
Alcohol works on the brain to produce its desired effects, e.g., sociability and intoxication, and hence the brain is an important organ for exploring subsequent harms. These come in many different forms such as the consequences of damage during intoxication, e.g., from falls and fights, damage from withdrawal, damage from the toxicity of alcohol and its metabolites and altered brain structure and function with implications for behavioral processes such as craving and addiction. On top of that are peripheral factors that compound brain damage such as poor diet, vitamin deficiencies leading to Wernicke-Korsakoff syndrome. Prenatal alcohol exposure can also have a profound impact on brain development and lead to irremediable changes of fetal alcohol syndrome. This chapter briefly reviews aspects of these with a particular focus on recent brain imaging results. Cardiovascular effects of alcohol that lead to brain pathology are not covered as they are dealt with elsewhere in the volume.
Collapse
Affiliation(s)
- David Nutt
- Neuropsychopharmacology Unit, Division of Psychiatry, Department of Brain Sciences, Hammersmith Hospital, Imperial College London, London W12 ONN, UK; (A.H.); (L.F.); (R.Z.); (E.O.C.P.); (L.P.); (A.L.-H.)
| | | | | | | | | | | | | |
Collapse
|
23
|
Liu Y, Li J, Bu H, Wang H, Zhang Y, Shen Q, Li M, Lu Z, Rong X, Zheng D, Peng Y. Circular RNA expression alteration identifies a novel circulating biomarker in serum exosomal for detection of alcohol dependence. Addict Biol 2021; 26:e13031. [PMID: 33821559 DOI: 10.1111/adb.13031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/22/2020] [Accepted: 03/05/2021] [Indexed: 12/15/2022]
Abstract
Alcohol dependence (AD) is one of the most common and detrimental neuropsychological disorders. Recently, more and more studies have focused on circular RNA as markers for central nervous system (CNS) diseases. The present study was conducted to evaluate the circular RNA expression alteration in serum exosomal and to identify a novel circulating biomarker for the detection of AD. We first isolated exosomes from serum and then investigated the circRNA expression alterations by high throughput whole transcriptome sequencing. The data were then analyzed using bioinformatics methods. Moreover, we verified the circRNA-seq by qRT-PCR. Furthermore, we analyzed the correlations between the levels of hsa_circ_0004771 and both Severity of Alcohol Dependence Questionnaire (SADQ) and Alcohol Dependence Scale (ADS). The diagnostic value of hsa_circ_0004771 in AD patients was evaluated by receiver operating characteristic (ROC). In this study, 254 differentially expressed circRNAs were identified, with 149 upregulated and 105 downregulated. GO analysis showed that these differentially expressed circRNAs from exosomes might be associated with the regulation of neuron projection and axon regeneration. KEGG analysis revealed that T cell receptor signaling and antigen processing and presentation pathway had a regulating effect on upstream levels. We found that hsa_circ_0004771 was related to the severity of AD. The AUC for the diagnostic value of hsa_circ_0004771 in AD patients was 0.874. These findings indicated that circRNA in serum exosomes provide novel targets for further research on molecular mechanisms of AD. Among these, hsa_circ_0004771 may be a sensitive biomarker that was related to the severity of AD.
Collapse
Affiliation(s)
- Yunyun Liu
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Jiande Li
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Huanhuan Bu
- Department of Neurology The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
| | - Hongxuan Wang
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Yuanpei Zhang
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Qingyu Shen
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Mei Li
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Zijing Lu
- Department of Plastic and Cosmetic Surgery Nanfang Hospital, Southern Medical University Guangzhou China
| | - Xiaoming Rong
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Dong Zheng
- Department of Neurology The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
| | - Ying Peng
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| |
Collapse
|
24
|
Sander CY, Bovo S, Torrado-Carvajal A, Albrecht D, Deng H, Napadow V, Price JC, Hooker JM, Loggia ML. [ 11C]PBR28 radiotracer kinetics are not driven by alterations in cerebral blood flow. J Cereb Blood Flow Metab 2021; 41:3069-3084. [PMID: 34159823 PMCID: PMC8756484 DOI: 10.1177/0271678x211023387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The positron emission tomography (PET) radiotracer [11C]PBR28 has been increasingly used to image the translocator protein (TSPO) as a marker of neuroinflammation in a variety of brain disorders. Interrelatedly, similar clinical populations can also exhibit altered brain perfusion, as has been shown using arterial spin labelling in magnetic resonance imaging (MRI) studies. Hence, an unsolved debate has revolved around whether changes in perfusion could alter delivery, uptake, or washout of the radiotracer [11C]PBR28, and thereby influence outcome measures that affect interpretation of TSPO upregulation. In this simultaneous PET/MRI study, we demonstrate that [11C]PBR28 signal elevations in chronic low back pain patients are not accompanied, in the same regions, by increases in cerebral blood flow (CBF) compared to healthy controls, and that areas of marginal hypoperfusion are not accompanied by decreases in [11C]PBR28 signal. In non-human primates, we show that hypercapnia-induced increases in CBF during radiotracer delivery or washout do not alter [11C]PBR28 outcome measures. The combined results from two methodologically distinct experiments provide support from human data and direct experimental evidence from non-human primates that changes in CBF do not influence outcome measures reported by [11C]PBR28 PET imaging studies and corresponding interpretations of the biological meaning of TSPO upregulation.
Collapse
Affiliation(s)
- Christin Y Sander
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Stefano Bovo
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Information Engineering, University of Padova, Padova, Italy
| | - Angel Torrado-Carvajal
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Medical Image Analysis and Biometry Laboratory, Universidad Rey Juan Carlos, Madrid, Spain
| | - Daniel Albrecht
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA
| | - Hongping Deng
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA
| | - Vitaly Napadow
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Julie C Price
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Jacob M Hooker
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Marco L Loggia
- Department of Radiology, Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Deri Y, Clouston SAP, DeLorenzo C, Gardus JD, Bartlett EA, Santiago-Michels S, Bangiyev L, Kreisl WC, Kotov R, Huang C, Slifstein M, Parsey RV, Luft BJ. Neuroinflammation in World Trade Center responders at midlife: A pilot study using [ 18F]-FEPPA PET imaging. Brain Behav Immun Health 2021; 16:100287. [PMID: 34589784 PMCID: PMC8474562 DOI: 10.1016/j.bbih.2021.100287] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 06/20/2021] [Indexed: 02/08/2023] Open
Abstract
Background Neuroinflammation has long been theorized to arise from exposures to fine particulate matter and to be modulated when individuals experience chronic stress, both of which are also though to cause cognitive decline in part as a result of neuroinflammation. Objectives Hypothesizing that neuroinflammation might be linked to experiences at the World Trade Center (WTC) events, this study explored associations between glial activation and neuropsychological measures including post-traumatic stress disorder (PTSD) symptom severity and WTC exposure duration. Methods Translocator protein 18-kDa (TSPO) is overexpressed by activated glial cells, predominantly microglia and astrocytes, making TSPO distribution a putative biomarker for neuroinflammation. Twenty WTC responders completed neuropsychological assessments and in vivo PET brain scan with [18F]-FEPPA. Generalized linear modeling was used to test associations between PTSD, and WTC exposure duratiioni as the predictor and both global and regional [18F]-FEPPA total distribution volumes as the outcomes. Result Responders were 56.0 ± 4.7 years-old, and 75% were police officers on 9/11/2001, and all had at least a high school education. Higher PTSD symptom severity was associated with global and regional elevations in [18F]-FEPPA binding predominantly in the hippocampus (d = 0.72, P = 0.001) and frontal cortex (d = 0.64, P = 0.004). Longer exposure duration to WTC sites was associated with higher [18F]-FEPPA binding in the parietal cortex. Conclusion Findings from this study of WTC responders at midlife suggest that glial activation is associated with PTSD symptoms, and WTC exposure duration. Future investigation is needed to understand the important role of neuroinflammation in highly exposed WTC responders. We examined the theory that glial activation is associated with 9/11 exposures. TSPO-Vt was examined using PET in 20 responders adjusting for TSPO genotype. Responders with PTSD had increased TSPO distribution volume in the hippocampus. Heavily exposed responders had increased TSPO distribution in the parietal cortex.
Collapse
Affiliation(s)
- Yael Deri
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Sean A P Clouston
- Program in Public Health and Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Christine DeLorenzo
- Department of Psychiatry, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.,Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - John D Gardus
- Department of Psychiatry, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Elizabeth A Bartlett
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY, USA.,Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Stephanie Santiago-Michels
- Stony Brook World Trade Center Wellness Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Lev Bangiyev
- Department of Radiology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - William C Kreisl
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Roman Kotov
- Department of Psychiatry, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Chuan Huang
- Department of Psychiatry, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.,Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.,Department of Radiology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Mark Slifstein
- Department of Psychiatry, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Ramin V Parsey
- Department of Psychiatry, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.,Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Benjamin J Luft
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.,Stony Brook World Trade Center Wellness Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
26
|
Rahimian R, Wakid M, O'Leary LA, Mechawar N. The emerging tale of microglia in psychiatric disorders. Neurosci Biobehav Rev 2021; 131:1-29. [PMID: 34536460 DOI: 10.1016/j.neubiorev.2021.09.023] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/18/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022]
Abstract
As the professional phagocytes of the brain, microglia orchestrate the immunological response and play an increasingly important role in maintaining homeostatic brain functions. Microglia are activated by pathological events or slight alterations in brain homeostasis. This activation is dependent on the context and type of stressor or pathology. Through secretion of cytokines, chemokines and growth factors, microglia can strongly influence the response to a stressor and can, therefore, determine the pathological outcome. Psychopathologies have repeatedly been associated with long-lasting priming and sensitization of cerebral microglia. This review focuses on the diversity of microglial phenotype and function in health and psychiatric disease. We first discuss the diverse homeostatic functions performed by microglia and then elaborate on context-specific spatial and temporal microglial heterogeneity. Subsequently, we summarize microglia involvement in psychopathologies, namely major depressive disorder, schizophrenia and bipolar disorder, with a particular focus on post-mortem studies. Finally, we postulate microglia as a promising novel therapeutic target in psychiatry through antidepressant and antipsychotic treatment.
Collapse
Affiliation(s)
- Reza Rahimian
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada
| | - Marina Wakid
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Liam Anuj O'Leary
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
27
|
Yokokura M, Takebasashi K, Takao A, Nakaizumi K, Yoshikawa E, Futatsubashi M, Suzuki K, Nakamura K, Yamasue H, Ouchi Y. In vivo imaging of dopamine D1 receptor and activated microglia in attention-deficit/hyperactivity disorder: a positron emission tomography study. Mol Psychiatry 2021; 26:4958-4967. [PMID: 32439845 DOI: 10.1038/s41380-020-0784-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/05/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Alterations in the cortical dopamine system and microglial activation have been implicated in the pathophysiology of attention-deficit/hyperactivity disorder (ADHD), one of neurodevelopmental disorders that can be conventionally treated with a dopamine enhancer (methylphenidate) albeit unsatisfactorily. Here, we investigated the contributions of the dopamine D1 receptor (D1R) and activated microglia and their interactions to the clinical severities in ADHD individuals using positron emission tomography (PET). Twenty-four psychotropic-naïve ADHD individuals and 24 age- and sex-matched typically developing (TD) subjects underwent PET measurements with [11C]SCH23390 for the D1R and [11C](R)PK11195 for activated microglia as well as assessments of clinical symptoms and cognitive functions. The ADHD individuals showed decreased D1R in the anterior cingulate cortex (ACC) and increased activated microglia in the dorsolateral prefrontal cortex (DLPFC) and orbitofrontal cortex (OFC) compared with the TD subjects. The decreased D1R in the ACC was associated with severe hyperactivity in the participants with ADHD. Microglial activation in the DLPFC were associated with deficits in processing speed and attentional ability, and that in the OFC was correlated with lower processing speed in the ADHD individuals. Furthermore, positive correlations between the D1R and activated microglia in both the DLPFC and the OFC were found to be significantly specific to the ADHD group and not to the TD group. The current findings suggest that microglial activation and the D1R reduction as well as their aberrant interactions underpin the neurophysiological mechanism of ADHD and indicate these biomolecular changes as a novel therapeutic target.
Collapse
Affiliation(s)
- Masamichi Yokokura
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kiyokazu Takebasashi
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | - Kyoko Nakaizumi
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Etsuji Yoshikawa
- Central Research Laboratory, Hamamatsu Photonics K.K, Hamamatsu, Japan
| | - Masami Futatsubashi
- Global Strategic Challenge Center, Hamamatsu Photonics K.K, Hamamatsu, Japan.,Hamamatsu PET Imaging Center, Hamamatsu Medical Photonics Foundation, Hamamatsu, Japan
| | - Katsuaki Suzuki
- Department of Biofunctional Imaging, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | - Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yasuomi Ouchi
- Department of Biofunctional Imaging, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| |
Collapse
|
28
|
Salavrakos M, Leclercq S, De Timary P, Dom G. Microbiome and substances of abuse. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110113. [PMID: 32971216 DOI: 10.1016/j.pnpbp.2020.110113] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022]
Abstract
There is a growing amount of evidence showing a reciprocal relation between the gut microbiota and the brain. Substance use disorders (SUD), which are a major cause of preventable morbidity and mortality worldwide, have an influence on the gut microbiota and on the gut-brain axis. The communication between the microbiota and the brain exists through different pathways: (1) the immune response elicited by bacterial products, coupled with alterations of the intestinal barrier allowing these products to enter the bloodstream, (2) the direct and indirect effects of bacterial metabolites such as short chain fatty acids (SCFAs) or tryptophan on the brain, (3) and the hypothalamic-pituitary-adrenal (HPA) axis, whose peripheral afferents can be influenced by the microbiota, and can in turn activate microglia. Among substances of abuse, alcohol has been the subject of the greatest number of studies in this field. In some but not all patients suffering from alcohol-use-disorder (AUD), alcohol alters the composition of the gut microbiota and the permeability of the intestinal barrier, directly and through dysbiosis. It has also been well demonstrated that alcohol induces a peripheral inflammation; it is still unclear whether it induces a central inflammation, as there are contradictory results in human studies. In animal studies, it has been shown that neuroinflammation increases during alcohol withdrawal. Literature on opioids and stimulants is less numerous. Chronic morphine intake induces dysbiosis, increased intestinal permeability and a probable neuroinflammation, which could explain symptoms such as tolerance, hyperalgesia and deficit in reward behavior. Cocaine induces a dysbiosis and conversely the microbiome can modulate the behavioral response to stimulant drugs. Tobacco cessation is associated with an increase in microbiota diversity. Taken together, the findings of our narrative literature review suggest a bidirectional influence in the pathogenesis of substance use disorders.
Collapse
Affiliation(s)
- M Salavrakos
- Target Journal Progress in Neuropsychopharmacology and Biological Psychiatry, Belgium
| | - S Leclercq
- Target Journal Progress in Neuropsychopharmacology and Biological Psychiatry, Belgium
| | - P De Timary
- Target Journal Progress in Neuropsychopharmacology and Biological Psychiatry, Belgium
| | - G Dom
- Target Journal Progress in Neuropsychopharmacology and Biological Psychiatry, Belgium.
| |
Collapse
|
29
|
De Carvalho LM, Wiers CE, Sun H, Wang G, Volkow ND. Increased transcription of TSPO, HDAC2, and HDAC6 in the amygdala of males with alcohol use disorder. Brain Behav 2021; 11:e01961. [PMID: 33216461 PMCID: PMC7882159 DOI: 10.1002/brb3.1961] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Repeated exposure to high doses of alcohol triggers neuroinflammatory processes that contribute to craving and mood dysfunction in alcohol use disorder (AUD). The upregulation of the translocator protein (TSPO) is considered a biomarker of neuroinflammation, and TSPO ligands have been used as neuroimaging biomarkers of neuroinflammation. Epigenetic mechanisms are also implicated in neuroinflammatory responses to alcohol, and elevated expression of HDAC2 and HDAC6 has been reported in the brain of animals exposed to chronic alcohol. METHODS The present study examined the transcriptional regulation of TSPO, HDAC2, and HDAC6 in human postmortem brain tissue from males previously diagnosed with AUD (n = 11) compared to age-matched nondependent males (n = 13) in four brain regions relevant to AUD: prefrontal cortex (PFC), nucleus accumbens (NAc), hippocampus (HPP), and amygdala (AMY). RESULTS Translocator protein mRNA levels in AMY and PFC and HDAC2 and HDAC6 mRNA levels in AMY were upregulated in AUD compared to controls. In AMY, TSPO mRNA levels were positively associated with HDAC2 and HDAC6 mRNA levels, suggesting a possible regulation of TSPO by HDAC2 and HDAC6 in this brain region. In contrast, there were no group differences for TSPO, HDAC2, and HDAC6 in NAc and HPP. CONCLUSION Our study is the first to find upregulated TSPO mRNA levels in AMY and PFC in postmortem brains from AUD consistent with neuroinflammation, and in the amygdala, they implicate epigenetic regulation of TSPO by HDAC2 and HDAC6.
Collapse
Affiliation(s)
- Luana Martins De Carvalho
- National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMDUSA
- Center for Alcohol Research in Epigenetics, Department of PsychiatryUniversity of Illinois at ChicagoChicagoILUSA
| | - Corinde E. Wiers
- National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMDUSA
- Department of PsychiatryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Hui Sun
- National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMDUSA
| | - Gene‐Jack Wang
- National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMDUSA
| | - Nora D. Volkow
- National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMDUSA
- National Institute on Drug AbuseNational Institutes of HealthRockvilleMDUSA
| |
Collapse
|
30
|
Alvarez Cooper I, Beecher K, Chehrehasa F, Belmer A, Bartlett SE. Tumour Necrosis Factor in Neuroplasticity, Neurogenesis and Alcohol Use Disorder. Brain Plast 2020; 6:47-66. [PMID: 33680846 PMCID: PMC7903009 DOI: 10.3233/bpl-190095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Alcohol use disorder is a pervasive and detrimental condition that involves changes in neuroplasticity and neurogenesis. Alcohol activates the neuroimmune system and alters the inflammatory status of the brain. Tumour necrosis factor (TNF) is a well characterised neuroimmune signal but its involvement in alcohol use disorder is unknown. In this review, we discuss the variable findings of TNF's effect on neuroplasticity and neurogenesis. Acute ethanol exposure reduces TNF release while chronic alcohol intake generally increases TNF levels. Evidence suggests TNF potentiates excitatory transmission, promotes anxiety during alcohol withdrawal and is involved in drug use in rodents. An association between craving for alcohol and TNF is apparent during withdrawal in humans. While anti-inflammatory therapies show efficacy in reversing neurogenic deficit after alcohol exposure, there is no evidence for TNF's essential involvement in alcohol's effect on neurogenesis. Overall, defining TNF's role in alcohol use disorder is complicated by poor understanding of its variable effects on synaptic transmission and neurogenesis. While TNF may be of relevance during withdrawal, the neuroimmune system likely acts through a larger group of inflammatory cytokines to alter neuroplasticity and neurogenesis. Understanding the individual relevance of TNF in alcohol use disorder awaits a more comprehensive understanding of TNF's effects within the brain.
Collapse
Affiliation(s)
- Ignatius Alvarez Cooper
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
| | - Kate Beecher
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
- School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Fatemeh Chehrehasa
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
| | - Arnauld Belmer
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
- School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Selena E. Bartlett
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
- School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
31
|
Meyer JH, Cervenka S, Kim MJ, Kreisl WC, Henter ID, Innis RB. Neuroinflammation in psychiatric disorders: PET imaging and promising new targets. Lancet Psychiatry 2020; 7:1064-1074. [PMID: 33098761 PMCID: PMC7893630 DOI: 10.1016/s2215-0366(20)30255-8] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 01/14/2023]
Abstract
Neuroinflammation is a multifaceted physiological and pathophysiological response of the brain to injury and disease. Given imaging findings of 18 kDa translocator protein (TSPO) and the development of radioligands for other inflammatory targets, PET imaging of neuroinflammation is at a particularly promising stage. This Review critically evaluates PET imaging results of inflammation in psychiatric disorders, including major depressive disorder, schizophrenia and psychosis disorders, substance use, and obsessive-compulsive disorder. We also consider promising new targets that can be measured in the brain, such as monoamine oxidase B, cyclooxygenase-1 and cyclooxygenase-2, colony stimulating factor 1 receptor, and the purinergic P2X7 receptor. Thus far, the most compelling TSPO imaging results have arguably been found in major depressive disorder, for which consistent increases have been observed, and in schizophrenia and psychosis, for which patients show reduced TSPO levels. This pattern highlights the importance of validating brain biomarkers of neuroinflammation for each condition separately before moving on to patient stratification and treatment monitoring trials.
Collapse
Affiliation(s)
- Jeffrey H Meyer
- Campbell Family Mental Health Research Institute, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Simon Cervenka
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Stockholm, Sweden
| | - Min-Jeong Kim
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - William C Kreisl
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Ioline D Henter
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA.
| |
Collapse
|
32
|
García-Marchena N, Maza-Quiroga R, Serrano A, Barrios V, Requena-Ocaña N, Suárez J, Chowen JA, Argente J, Rubio G, Torrens M, López-Gallardo M, Marco EM, Castilla-Ortega E, Santín LJ, Rodríguez de Fonseca F, Pavón FJ, Araos P. Abstinent patients with alcohol use disorders show an altered plasma cytokine profile: Identification of both interleukin 6 and interleukin 17A as potential biomarkers of consumption and comorbid liver and pancreatic diseases. J Psychopharmacol 2020; 34:1250-1260. [PMID: 32536325 DOI: 10.1177/0269881120928176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Recent studies have demonstrated that alcohol consumption can modulate the immune system by directly activating natural immunity and triggering inflammatory processes in the central nervous system and in peripheral organs, such as the liver and pancreas. Patients with alcohol use disorders have an elevated frequency of comorbid mental disorders and gut diseases (i.e. fatty liver and pancreatitis) that complicate diagnosis, treatment and prognosis. AIMS The present study aims to explore possible associations in circulating plasma cytokine concentrations in abstinent patients diagnosed with alcohol use disorders. METHODS To this end, 85 abstinent subjects with alcohol use disorders from an outpatient setting and 55 healthy subjects were evaluated for both substance and mental disorders. The plasma levels of cytokines interleukin 1 beta, interleukin 4, interleukin 6, interleukin 17A, interferon gamma and tumour necrosis alpha were determined and their association with (a) history of alcohol consumption, (b) psychiatric comorbidity and (c) liver/pancreas comorbidities was explored. RESULTS We found that plasma concentrations of interleukin 1 beta, interleukin 6 and tumour necrosis alpha were increased, whereas plasma concentrations of interleukin 4, interleukin 17A and interferon gamma were decreased in abstinent alcohol use disorder patients as compared with control subjects. Moreover, we found that changes in interleukin 6 and interleukin 17A plasma concentrations in alcohol use disorder patients were associated with the presence of liver and pancreatic diseases. CONCLUSION The present results suggest alcohol use disorder is associated with alterations of plasma cytokines, being interleukin 6 and interleukin 17A potential biomarkers of the presence of comorbidities of digestive organs. The clinical relevance of these findings is discussed in the context of alcohol-induced inflammatory processes.
Collapse
Affiliation(s)
- Nuria García-Marchena
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Málaga, Spain.,Unidad de Adicciones, Servicio de Medicina Interna, Institut Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Rosa Maza-Quiroga
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Antonia Serrano
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Vicente Barrios
- Departments of Pediatrics and Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain.,IMDEA Food Institute, Madrid, Spain
| | - Nerea Requena-Ocaña
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Juan Suárez
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Julie Ann Chowen
- Departments of Pediatrics and Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain.,IMDEA Food Institute, Madrid, Spain
| | - Jesús Argente
- Departments of Pediatrics and Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain.,Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain.,IMDEA Food Institute, Madrid, Spain
| | - Gabriel Rubio
- Servicio de Psiquiatría, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Marta Torrens
- Institut de Neuropsiquiatria i Addiccions del Parc de Salut Mar, Barcelona, Spain
| | - Meritxell López-Gallardo
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Eva María Marco
- Departamento de Fisiología II, Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - Estela Castilla-Ortega
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, Málaga, Spain
| | - Luis Javier Santín
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Francisco Javier Pavón
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Málaga, Spain.,Unidad de Gestión Clínica del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Pedro Araos
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Málaga, Spain.,Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, Málaga, Spain
| |
Collapse
|
33
|
Chang L, Liang H, Kandel SR, He JJ. Independent and Combined Effects of Nicotine or Chronic Tobacco Smoking and HIV on the Brain: A Review of Preclinical and Clinical Studies. J Neuroimmune Pharmacol 2020; 15:658-693. [PMID: 33108618 DOI: 10.1007/s11481-020-09963-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023]
Abstract
Tobacco smoking is highly prevalent among HIV-infected individuals. Chronic smokers with HIV showed greater cognitive deficits and impulsivity, and had more psychopathological symptoms and greater neuroinflammation than HIV non-smokers or smokers without HIV infection. However, preclinical studies that evaluated the combined effects of HIV-infection and tobacco smoking are scare. The preclinical models typically used cell cultures or animal models that involved specific HIV viral proteins or the administration of nicotine to rodents. These preclinical models consistently demonstrated that nicotine had neuroprotective and anti-inflammatory effects, leading to cognitive enhancement. Although the major addictive ingredient in tobacco smoking is nicotine, chronic smoking does not lead to improved cognitive function in humans. Therefore, preclinical studies designed to unravel the interactive effects of chronic tobacco smoking and HIV infection are needed. In this review, we summarized the preclinical studies that demonstrated the neuroprotective effects of nicotine, the neurotoxic effects of the HIV viral proteins, and the scant literature on nicotine or tobacco smoke in HIV transgenic rat models. We also reviewed the clinical studies that evaluated the neurotoxic effects of tobacco smoking, HIV infection and their combined effects on the brain, including studies that evaluated the cognitive and behavioral assessments, as well as neuroimaging measures. Lastly, we compared the different approaches between preclinical and clinical studies, identified some gaps and proposed some future directions. Graphical abstract Independent and combined effects of HIV and tobacco/nicotine. Left top and bottom panels: Both clinical studies of HIV infected persons and preclinical studies using viral proteins in vitro or in vivo in animal models showed that HIV infection could lead to neurotoxicity and neuroinflammation. Right top and bottom panels: While clinical studies of tobacco smoking consistently showed deleterious effects of smoking, clinical and preclinical studies that used nicotine show mild cognitive enhancement, neuroprotective and possibly anti-inflammatory effects. In the developing brain, however, nicotine is neurotoxic. Middle overlapping panels: Clinical studies of persons with HIV who were smokers typically showed additive deleterious effects of HIV and tobacco smoking. However, in the preclinical studies, when nicotine was administered to the HIV-1 Tg rats, the neurotoxic effects of HIV were attenuated, but tobacco smoke worsened the inflammatory cascade.
Collapse
Affiliation(s)
- Linda Chang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 670 W. Baltimore Street, HSF III, Baltimore, MD, 21201, USA.
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Medicine, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA.
| | - Huajun Liang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 670 W. Baltimore Street, HSF III, Baltimore, MD, 21201, USA
| | - Suresh R Kandel
- Department of Microbiology and Immunology, Chicago Medical School, Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, 3333 Green Bay Road, Basic Science Building 2.300, North Chicago, IL, 60064, USA
| | - Johnny J He
- Department of Microbiology and Immunology, Chicago Medical School, Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, 3333 Green Bay Road, Basic Science Building 2.300, North Chicago, IL, 60064, USA.
| |
Collapse
|
34
|
Lowe PP, Morel C, Ambade A, Iracheta-Vellve A, Kwiatkowski E, Satishchandran A, Furi I, Cho Y, Gyongyosi B, Catalano D, Lefebvre E, Fischer L, Seyedkazemi S, Schafer DP, Szabo G. Chronic alcohol-induced neuroinflammation involves CCR2/5-dependent peripheral macrophage infiltration and microglia alterations. J Neuroinflammation 2020; 17:296. [PMID: 33036616 PMCID: PMC7547498 DOI: 10.1186/s12974-020-01972-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/30/2020] [Indexed: 02/11/2023] Open
Abstract
Background Chronic alcohol consumption is associated with neuroinflammation, neuronal damage, and behavioral alterations including addiction. Alcohol-induced neuroinflammation is characterized by increased expression of proinflammatory cytokines (including TNFα, IL-1β, and CCL2) and microglial activation. We hypothesized chronic alcohol consumption results in peripheral immune cell infiltration to the CNS. Since chemotaxis through the CCL2-CCR2 signaling axis is critical for macrophage recruitment peripherally and centrally, we further hypothesized that blockade of CCL2 signaling using the dual CCR2/5 inhibitor cenicriviroc (CVC) would prevent alcohol-induced CNS infiltration of peripheral macrophages and alter the neuroinflammatory state in the brain after chronic alcohol consumption. Methods C57BL/6J female mice were fed an isocaloric or 5% (v/v) ethanol Lieber DeCarli diet for 6 weeks. Some mice received daily injections of CVC. Microglia and infiltrating macrophages were characterized and quantified by flow cytometry and visualized using CX3CR1eGFP/+ CCR2RFP/+ reporter mice. The effect of ethanol and CVC treatment on the expression of inflammatory genes was evaluated in various regions of the brain, using a Nanostring nCounter inflammation panel. Microglia activation was analyzed by immunofluorescence. CVC-treated and untreated mice were presented with the two-bottle choice test. Results Chronic alcohol consumption induced microglia activation and peripheral macrophage infiltration in the CNS, particularly in the hippocampus. Treatment with CVC abrogated ethanol-induced recruitment of peripheral macrophages and partially reversed microglia activation. Furthermore, the expression of proinflammatory markers was upregulated by chronic alcohol consumption in various regions of the brain, including the cortex, hippocampus, and cerebellum. Inhibition of CCR2/5 decreased alcohol-mediated expression of inflammatory markers. Finally, microglia function was impaired by chronic alcohol consumption and restored by CVC treatment. CVC treatment did not change the ethanol consumption or preference of mice in the two-bottle choice test. Conclusions Together, our data establish that chronic alcohol consumption promotes the recruitment of peripheral macrophages into the CNS and microglia alterations through the CCR2/5 axis. Therefore, further exploration of the CCR2/5 axis as a modulator of neuroinflammation may offer a potential therapeutic approach for the treatment of alcohol-associated neuroinflammation.
Collapse
Affiliation(s)
- Patrick P Lowe
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Caroline Morel
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, ST-214B, Boston, MA, 02215, USA
| | - Aditya Ambade
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Arvin Iracheta-Vellve
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Erica Kwiatkowski
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Istvan Furi
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Yeonhee Cho
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.,Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, ST-214B, Boston, MA, 02215, USA
| | - Benedek Gyongyosi
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Donna Catalano
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, ST-214B, Boston, MA, 02215, USA
| | | | | | | | - Dorothy P Schafer
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Gyongyi Szabo
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA. .,Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, ST-214B, Boston, MA, 02215, USA.
| |
Collapse
|
35
|
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
|
36
|
|
37
|
Woodcock EA, Schain M, Cosgrove KP, Hillmer AT. Quantification of [ 11C]PBR28 data after systemic lipopolysaccharide challenge. EJNMMI Res 2020; 10:19. [PMID: 32166497 PMCID: PMC7067964 DOI: 10.1186/s13550-020-0605-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 01/31/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Lipopolysaccharide (LPS) is a classic immune stimulus. LPS combined with positron emission tomography (PET) 18 kDa translocator protein (TSPO) brain imaging provides a robust human laboratory model to study neuroimmune signaling. To evaluate optimal analysis of these data, this work compared the sensitivity of six quantification approaches. METHODS [11C]PBR28 data from healthy volunteers (N = 8) were collected before and 3 h after LPS challenge (1.0 ng/kg IV). Quantification approaches included total volume of distribution estimated with two tissue, and two tissue plus irreversible uptake in whole blood, compartment models (2TCM and 2TCM-1k, respectively) and multilinear analysis-1 (MA-1); binding potential estimated with simultaneous estimation (SIME); standardized uptake values (SUV); and SUV ratio (SUVR). RESULTS The 2TCM, 2TCM-1k, MA-1, and SIME approaches each yielded substantive effect sizes for LPS effects (partial η2 = 0.56-0.89, ps <. 05), whereas SUV and SUVR did not. CONCLUSION These findings highlight the importance of incorporating AIF measurements to quantify LPS-TSPO studies.
Collapse
Affiliation(s)
- Eric A Woodcock
- Department of Pscyhiatry, Yale School of Medicine, 300 George St., New Haven, CT, USA
| | - Martin Schain
- Neurobiology Research Unit, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kelly P Cosgrove
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, 330 Cedar St., New Haven, CT, USA
| | - Ansel T Hillmer
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, 330 Cedar St., New Haven, CT, USA.
| |
Collapse
|
38
|
Hillmer AT, Matuskey D, Huang Y, Nabulsi N, Ropchan J, Carson RE, O'Malley SS, Cosgrove KP. Tobacco Smoking in People Is Not Associated with Altered 18-kDa Translocator Protein Levels: A PET Study. J Nucl Med 2020; 61:1200-1204. [PMID: 32005773 DOI: 10.2967/jnumed.119.237735] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/19/2019] [Indexed: 11/16/2022] Open
Abstract
The effects of tobacco smoking on the immune system of the brain are not well elucidated. Although nicotine is immunosuppressive, other constituents in tobacco smoke have inflammatory effects. PET imaging of the 18-kDa translocator protein (TSPO) provides a biomarker for microglia, the primary immunocompetent cells of the brain. This work compared brain TSPO levels in 20 tobacco smokers (abstinent for at least 2 h) and 20 nonsmokers using a fully quantitative modeling approach for the first time, to our knowledge. Methods: 11C-PBR28 (N-((2-(methoxy-11C)-phenyl)methyl)-N-(6-phenoxy-3-pyridinyl)acetamide) PET scans were acquired with arterial blood sampling to estimate the metabolite-corrected input function. 11C-PBR28 volumes of distribution were estimated throughout the brain with multilinear analysis. Results: Statistical analyses revealed no evidence of significant differences in regional 11C-PBR28 volumes of distribution between smokers and nonsmokers (whole-brain Cohen d = 0.09) despite adequate power to detect medium effect sizes. Conclusion: These findings inform previous PET studies reporting lower TSPO radiotracer concentrations in the brain (measured as SUV) for tobacco smokers than for nonsmokers by demonstrating the importance of accounting for radiotracer concentrations in plasma. These findings suggest that nonsmokers and smokers have comparable TSPO levels in the brain. Additional work with other biomarkers is needed to fully characterize the effects of tobacco smoking on the brain immune system.
Collapse
Affiliation(s)
- Ansel T Hillmer
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut .,Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,Yale University PET Center, Yale University School of Medicine, New Haven, Connecticut.,Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut
| | - David Matuskey
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut.,Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,Yale University PET Center, Yale University School of Medicine, New Haven, Connecticut.,Department of Neurology, Yale University School of Medicine, New Haven, Connecticut; and
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut.,Yale University PET Center, Yale University School of Medicine, New Haven, Connecticut
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut.,Yale University PET Center, Yale University School of Medicine, New Haven, Connecticut
| | - Jim Ropchan
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut.,Yale University PET Center, Yale University School of Medicine, New Haven, Connecticut
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut.,Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,Yale University PET Center, Yale University School of Medicine, New Haven, Connecticut.,Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut
| | - Stephanie S O'Malley
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Kelly P Cosgrove
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut.,Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,Yale University PET Center, Yale University School of Medicine, New Haven, Connecticut.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
39
|
De Picker L, Morrens M. Perspective: Solving the Heterogeneity Conundrum of TSPO PET Imaging in Psychosis. Front Psychiatry 2020; 11:362. [PMID: 32425835 PMCID: PMC7206714 DOI: 10.3389/fpsyt.2020.00362] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 04/09/2020] [Indexed: 12/11/2022] Open
Abstract
Positron emission tomography using ligands targeting translocator protein 18 kDa (TSPO PET) is an innovative method to visualize and quantify glial inflammatory responses in the central nervous system in vivo. Compared to some other neuropsychiatric disorders, findings of TSPO PET in schizophrenia and related psychotic disorders have been considerably more heterogeneous. Two conflicting meta-analyses have been published on the topic within the last year: one asserting evidence for decreased TSPO uptake, while the other observed increased TSPO uptake in a selection of studies. In this paper, we review and discuss five hypotheses which may explain the observed variability of TSPO PET findings in psychotic illness, namely that (1) an inflammatory phenotype is only present in a subgroup of psychosis patients; (2) heterogeneity is caused by interference of antipsychotic medication; (3) interference of other clinical confounders in the study populations (such as age, sex, BMI, smoking, and substance use); or (4) methodological variability between studies (such as choice of tracer and kinetic model, genotyping, study power, and diurnal effects); and (5) the glial responses underlying changes in TSPO expression are themselves heterogeneous and dynamic. Finally, we propose four key recommendations for future research proposals to mitigate these different causes of heterogeneity.
Collapse
Affiliation(s)
- Livia De Picker
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium.,SINAPS, University Psychiatric Hospital Campus Duffel, Duffel, Belgium
| | - Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium.,SINAPS, University Psychiatric Hospital Campus Duffel, Duffel, Belgium
| |
Collapse
|
40
|
Liu Y, Zhang Y, Peng J, Wang H, Li X, Li X, Rong X, Pan J, Peng Y. Autophagy alleviates ethanol-induced memory impairment in association with anti-apoptotic and anti-inflammatory pathways. Brain Behav Immun 2019; 82:63-75. [PMID: 31376498 DOI: 10.1016/j.bbi.2019.07.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 07/28/2019] [Accepted: 07/30/2019] [Indexed: 12/15/2022] Open
Abstract
Chronic excessive drinking leads to a wide spectrum of neurological disorders, including cognitive deficits, such as learning and memory impairment. However, the neurobiological mechanisms underlying these deleterious changes are still poorly understood. We conducted a comprehensive study to investigate the role and mechanism of autophagy in alcohol-induced memory impairment. To establish an ethanol-induced memory impairment mouse model, we allowed C57BL/6J mice intermittent access to 20% ethanol (four-bottle choice) to escalate ethanol drinking levels. Memory impairment was confirmed by a Morris water maze test. We found that mice exposed to EtOH (ethanol) and EtOH combined with the autophagy inhibitor 3-methyladenine (3-MA) showed high alcohol intake and blood alcohol concentration. We confirmed that the EtOH group exhibited notable memory impairment. Inhibition of autophagy by 3-MA worsened ethanol-induced memory impairment. Ethanol induced autophagy in the hippocampus of mice as indicated by western blotting, electron microscopy, RT-qPCR, and fluorescence confocal microscopy. We determined that the mTOR/BECN1 (S14) pathway is involved in ethanol-induced autophagy in vivo. Further, ethanol-induced autophagy suppressed the NLRP3 inflammatory and apoptosis pathways in the hippocampus in mice and in vitro. These findings suggest that autophagy activation in hippocampal cells alleviates ethanol-induced memory impairment in association with anti-apoptotic and anti-inflammatory pathways.
Collapse
Affiliation(s)
- Yunyun Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yuanpei Zhang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jialing Peng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Hongxuan Wang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiangpen Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaoyu Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaoming Rong
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jingrui Pan
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ying Peng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
41
|
Targeting neuroinflammation with minocycline in heavy drinkers. Psychopharmacology (Berl) 2019; 236:3013-3021. [PMID: 30919006 PMCID: PMC6764907 DOI: 10.1007/s00213-019-05205-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/18/2019] [Indexed: 12/27/2022]
Abstract
RATIONALE Alcohol has both acute and chronic effects on neuroimmune signaling, including triggering pro-inflammatory cytokine release by microglia. Minocycline, a second-generation tetracycline antibiotic, inhibits microglial activation and reduces neuroinflammation in preclinical studies. In mice, minocycline also reduces ethanol intake, attenuates ethanol-induced conditioned place preference, and inhibits ethanol-induced microglial activation and pro-inflammatory cytokine release. OBJECTIVE Here, for the first time, we tested the effects of minocycline on subjective response to ethanol and acute ethanol-induced inflammation in humans. METHODS Forty-eight heavy drinkers participated in a double-blind, placebo-controlled trial in which they were randomized to receive placebo, 100 mg, or 200 mg of minocycline for 10 days. Each subject then underwent two experimental sessions in which they were given a fixed dose of intravenous ethanol using a "clamp" procedure (100 mg%) or placebo (normal saline) on days 8 and 10 of treatment. RESULTS Minocycline was well tolerated, but there was no effect of either dose of minocycline on subjective response to ethanol or ethanol-induced craving; minocycline effects on cognitive function seem to interact with age. Minocycline treatment did not alter serum cytokine levels at baseline or during ethanol-exposure, although certain baseline cytokine levels predict sedative response to ethanol. CONCLUSION These findings indicate that a short-term treatment with minocycline may not alter ethanol-related inflammation or subjective response to ethanol in humans. Further research is needed to identify pharmacological agents with robust effects on ethanol-induced inflammation to determine whether neuroimmune modulation represents a viable treatment strategy for alcohol use disorder.
Collapse
|
42
|
A potential role for microglia in stress- and drug-induced plasticity in the nucleus accumbens: A mechanism for stress-induced vulnerability to substance use disorder. Neurosci Biobehav Rev 2019; 107:360-369. [PMID: 31550452 DOI: 10.1016/j.neubiorev.2019.09.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/16/2019] [Accepted: 09/05/2019] [Indexed: 12/16/2022]
Abstract
Stress is an important risk factor for the development of substance use disorder (SUD). Exposure to both stress and drugs abuse lead to changes in synaptic plasticity and stress-induced alterations in synaptic plasticity may contribute to later vulnerability to SUD. Recent developmental neuroscience studies have identified microglia as regulators of synaptic plasticity. As both stress and drugs of abuse lead to microglial activation, we propose this as a potential mechanism underlying their ability to change synaptic plasticity. This review focuses on three components of synaptic plasticity: spine density, brain-derived neurotrophic factor (BDNF) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor expression. Their roles in addiction, stress, and development will be reviewed, as well as possible mechanisms by which microglia could regulate their function. Potential links between stress, vulnerability to addiction, and microglial activity will be explored.
Collapse
|
43
|
Ezquer F, Quintanilla ME, Morales P, Santapau D, Ezquer M, Kogan MJ, Salas-Huenuleo E, Herrera-Marschitz M, Israel Y. Intranasal delivery of mesenchymal stem cell-derived exosomes reduces oxidative stress and markedly inhibits ethanol consumption and post-deprivation relapse drinking. Addict Biol 2019; 24:994-1007. [PMID: 30239077 DOI: 10.1111/adb.12675] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/19/2018] [Accepted: 07/30/2018] [Indexed: 01/01/2023]
Abstract
Chronic ethanol consumption leads to brain oxidative stress and neuroinflammation, conditions known to potentiate and perpetuate each other. Several studies have shown that neuroinflammation results in increases in chronic ethanol consumption. Recent reports showed that the intra-cerebroventricular administration of mesenchymal stem cells to rats consuming alcohol chronically markedly inhibited oxidative-stress, abolished neuroinflammation and greatly reduced chronic alcohol intake and post deprivation relapse-like alcohol intake. However, the intra-cerebroventricular administration of living cells is not suitable as a treatment of a chronic condition. The present study aimed at inhibiting ethanol intake by the non-invasive intranasal administration of human mesenchymal stem cell products: exosomes, microvesicles (40 to 150 nm) with marked antioxidant activity extruded from mesenchymal stem cells. The exosome membrane can fuse with the plasma membrane of cells in different tissues, thus delivering their content intracellularly. The study showed that the weekly intranasal administration of mesenchymal stem cell-derived exosomes to rats consuming alcohol chronically (1) inhibited their ethanol intake by 84 percent and blunted the relapse-like 'binge' drinking that follows an alcohol deprivation period and ethanol re-access. (2) Intranasally administered exosomes were found in the brain within 24 hours; (3) fully reversed both alcohol-induced hippocampal oxidative-stress, evidenced by a lower ratio of oxidized to reduced glutathione, and neuroinflammation, shown by a reduced astrocyte activation and microglial density; and (4) increased glutamate transporter GLT1 expression in nucleus accumbens, counteracting the inhibition of glutamate transporter activity, reportedly depressed under oxidative-stress conditions. Possible translational implications are envisaged.
Collapse
Affiliation(s)
- Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo; Chile
| | - María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences; Faculty of Medicine, University of Chile; Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences; Faculty of Medicine, University of Chile; Chile
- Department of Neuroscience; University of Chile; Chile
| | - Daniela Santapau
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo; Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo; Chile
| | - Marcelo J. Kogan
- Faculty of Medicine, Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile; Chile
- Advanced Center for Chronic Diseases (ACCDis); Chile
| | - Edison Salas-Huenuleo
- Faculty of Medicine, Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile; Chile
- Advanced Center for Chronic Diseases (ACCDis); Chile
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences; Faculty of Medicine, University of Chile; Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences; Faculty of Medicine, University of Chile; Chile
| |
Collapse
|
44
|
Melbourne JK, Thompson KR, Peng H, Nixon K. Its complicated: The relationship between alcohol and microglia in the search for novel pharmacotherapeutic targets for alcohol use disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 167:179-221. [PMID: 31601404 DOI: 10.1016/bs.pmbts.2019.06.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alcohol use disorder (AUD) is a chronic relapsing disorder with wide-ranging health consequences. Alcohol targets the central nervous system producing neurodegeneration and subsequent cognitive and behavioral deficits, but the mechanisms behind these effects remain unclear. Recently, evidence has been mounting for the role of neuroimmune activation in the pathogenesis of AUDs, but our nascent state of knowledge about the interaction of alcohol with the neuroimmune system supports that the relationship is complicated. As the resident macrophage of the central nervous system, microglia are a central focus. Human and animal research on the interplay between microglia and alcohol in AUDs has proven to be complex, and though early research focused on a pro-inflammatory phenotype of microglia, the anti-inflammatory and homeostatic roles of microglia must be considered. How these new roles for microglia should be incorporated into our thinking about the neuroimmune system in AUDs is discussed in the context of developing novel pharmacotherapies for AUDs.
Collapse
Affiliation(s)
- Jennifer K Melbourne
- The University of Texas at Austin, College of Pharmacy, Division of Pharmacology & Toxicology, Austin, TX, United States
| | - K Ryan Thompson
- The University of Texas at Austin, College of Pharmacy, Division of Pharmacology & Toxicology, Austin, TX, United States
| | - Hui Peng
- University of Kentucky, College of Pharmacy, Department of Pharmaceutical Sciences, Lexington, KY, United States
| | - Kimberly Nixon
- The University of Texas at Austin, College of Pharmacy, Division of Pharmacology & Toxicology, Austin, TX, United States.
| |
Collapse
|
45
|
Tyler RE, Kim SW, Guo M, Jang YJ, Damadzic R, Stodden T, Vendruscolo LF, Koob GF, Wang GJ, Wiers CE, Volkow ND. Detecting neuroinflammation in the brain following chronic alcohol exposure in rats: A comparison between in vivo and in vitro TSPO radioligand binding. Eur J Neurosci 2019; 50:1831-1842. [PMID: 30803059 PMCID: PMC10714130 DOI: 10.1111/ejn.14392] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/17/2019] [Accepted: 02/08/2019] [Indexed: 12/18/2022]
Abstract
Excessive alcohol consumption is associated with neuroinflammation, which likely contributes to alcohol-related pathology. However, positron emission tomography (PET) studies using radioligands for the 18-kDa translocator protein (TSPO), which is considered a biomarker of neuroinflammation, reported decreased binding in alcohol use disorder (AUD) participants compared to controls. In contrast, autoradiographic findings in alcohol exposed rats reported increases in TSPO radioligand binding. To assess if these discrepancies reflected differences between in vitro and in vivo methodologies, we compared in vitro autoradiography (using [3 H]PBR28 and [3 H]PK11195) with in vivo PET (using [11 C]PBR28) in male, Wistar rats exposed to chronic alcohol-vapor (dependent n = 10) and in rats exposed to air-vapor (nondependent n = 10). PET scans were obtained with [11 C]PBR28, after which rats were euthanized and the brains were harvested for autoradiography with [3 H]PBR28 and [3 H]PK11195 (n = 7 dependent and n = 7 nondependent), and binding quantified in hippocampus, thalamus, and parietal cortex. Autoradiography revealed significantly higher binding in alcohol-dependent rats for both radioligands in thalamus and hippocampus (trend level for [3 H]PBR28) compared to nondependent rats, and these group differences were stronger for [3 H]PK11195 than [3 H]PBR28. In contrast, PET measures obtained in the same rats showed no group difference in [11 C]PBR28 binding. Our in vitro data are consistent with neuroinflammation associated with chronic alcohol exposure. Failure to observe similar increases in [11 C]PBR28 binding in vivo suggests the possibility that a mechanism mediated by chronic alcohol exposure interferes with [11 C]PBR28 binding to TSPO in vivo. These data question the sensitivity of PBR28 PET as a methodology to assess neuroinflammation in AUD.
Collapse
Affiliation(s)
- Ryan E. Tyler
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Sung Won Kim
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Min Guo
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Yeon Joo Jang
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Ruslan Damadzic
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Tyler Stodden
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Leandro F. Vendruscolo
- National Institute on Drug Abuse, National Institutes of Health, NIH, Baltimore, Maryland
| | - George F. Koob
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
- National Institute on Drug Abuse, National Institutes of Health, NIH, Baltimore, Maryland
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Corinde E. Wiers
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Nora D. Volkow
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
- National Institute on Drug Abuse, National Institutes of Health, NIH, Baltimore, Maryland
| |
Collapse
|
46
|
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
|
47
|
Catale C, Bussone S, Lo Iacono L, Carola V. Microglial alterations induced by psychoactive drugs: A possible mechanism in substance use disorder? Semin Cell Dev Biol 2019; 94:164-175. [PMID: 31004753 DOI: 10.1016/j.semcdb.2019.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/19/2019] [Accepted: 03/29/2019] [Indexed: 12/11/2022]
Abstract
Recently, the xenobiotic hypothesis has implicated the immune system in targeting substances of abuse as foreign molecules and stimulating inflammatory responses. Microglial cells are the resident immune cells of the central nervous system and function in homeostatic surveillance. Microglial changes that are induced by exposure to substances of abuse appear to mediate in part the establishment of addiction and the persistence of drug-mediated biological and behavioral changes. In this context, interest in the study of drug-microglia interactions has increased recently. This review summarizes the most recent preclinical rodent and clinical studies on the interaction between microglia and various classes of drugs of abuse, such as ethanol, psychostimulants, and opioids. The principal biological mechanisms of the communication between substances of abuse and microglia will be described to consider putative mechanisms of the establishment of drug addiction and future potential targets for treating substance use disorder.
Collapse
Affiliation(s)
- Clarissa Catale
- Department of Psychology, University of Rome "La Sapienza", Via dei Marsi, 78, 00185 Rome, Italy
| | - Silvia Bussone
- Department of Dynamic and Clinical Psychology, University of Rome "La Sapienza", Via degli Apuli 1, 00185 Rome, Italy
| | - Luisa Lo Iacono
- Department of Psychology, University of Rome "La Sapienza", Via dei Marsi, 78, 00185 Rome, Italy; IRCCS Santa Lucia Foundation, Via Fosso di Fiorano 64, 00143 Rome, Italy
| | - Valeria Carola
- Department of Dynamic and Clinical Psychology, University of Rome "La Sapienza", Via degli Apuli 1, 00185 Rome, Italy; IRCCS Santa Lucia Foundation, Via Fosso di Fiorano 64, 00143 Rome, Italy.
| |
Collapse
|
48
|
Da Silva T, Hafizi S, Rusjan PM, Houle S, Wilson AA, Prce I, Sailasuta N, Mizrahi R. GABA levels and TSPO expression in people at clinical high risk for psychosis and healthy volunteers: a PET-MRS study. J Psychiatry Neurosci 2019; 44:111-119. [PMID: 30255837 PMCID: PMC6397035 DOI: 10.1503/jpn.170201] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND γ-Aminobutyric acidergic (GABAergic) dysfunction and immune activation have been implicated in the pathophysiology of schizophrenia. Preclinical evidence suggests that inflammation-related abnormalities may contribute to GABAergic alterations in the brain, but this has never been investigated in vivo in humans. In this multimodal imaging study, we quantified cerebral GABA plus macromolecule (GABA+) levels in antipsychotic-naive people at clinical high risk for psychosis and in healthy volunteers. We investigated for the first time the association between GABA+ levels and expression of translocator protein 18 kDa (TSPO; a marker of microglial activation) using positron emission tomography (PET). METHODS Thirty-five people at clinical high risk for psychosis and 18 healthy volunteers underwent 3 T proton magnetic resonance spectroscopy to obtain GABA+ levels in the medial prefrontal cortex (mPFC). A subset (29 people at clinical high risk for psychosis and 15 healthy volunteers) also underwent a high-resolution [18F]FEPPA PET scan to quantify TSPO expression. Each participant was genotyped for the TSPO rs6971 polymorphism. RESULTS We found that GABA+ levels were significantly associated with TSPO expression in the mPFC (F1,40 = 10.45, p = 0.002). We found no significant differences in GABA+ levels in the mPFC (F1,51 = 0.00, p > 0.99) between people at clinical high risk for psychosis and healthy volunteers. We found no significant correlations between GABA+ levels or residuals of the association with TSPO expression and the severity of prodromal symptoms or cognition. LIMITATIONS Given the cross-sectional nature of this study, we could determine no cause-and-effect relationships for GABA alterations and TSPO expression. CONCLUSION Our findings suggest that TSPO expression is negatively associated with GABA+ levels in the prefrontal cortex, independent of disease status.
Collapse
Affiliation(s)
- Tania Da Silva
- From the Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Silva, Hafizi, Rusjan, Houle, Wilson, Prce, Sailasuta, Mizrahi); the Institute of Medical Science, University of Toronto, Toronto, Ont., Canada (Silva, Rusjan, Mizrahi); the Department of Psychiatry, University of Toronto, Toronto, Ont., Canada (Rusjan, Houle, Wilson, Mizrahi); and the Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Rusjan, Houle, Sailasuta, Mizrahi)
| | - Sina Hafizi
- From the Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Silva, Hafizi, Rusjan, Houle, Wilson, Prce, Sailasuta, Mizrahi); the Institute of Medical Science, University of Toronto, Toronto, Ont., Canada (Silva, Rusjan, Mizrahi); the Department of Psychiatry, University of Toronto, Toronto, Ont., Canada (Rusjan, Houle, Wilson, Mizrahi); and the Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Rusjan, Houle, Sailasuta, Mizrahi)
| | - Pablo M Rusjan
- From the Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Silva, Hafizi, Rusjan, Houle, Wilson, Prce, Sailasuta, Mizrahi); the Institute of Medical Science, University of Toronto, Toronto, Ont., Canada (Silva, Rusjan, Mizrahi); the Department of Psychiatry, University of Toronto, Toronto, Ont., Canada (Rusjan, Houle, Wilson, Mizrahi); and the Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Rusjan, Houle, Sailasuta, Mizrahi)
| | - Sylvain Houle
- From the Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Silva, Hafizi, Rusjan, Houle, Wilson, Prce, Sailasuta, Mizrahi); the Institute of Medical Science, University of Toronto, Toronto, Ont., Canada (Silva, Rusjan, Mizrahi); the Department of Psychiatry, University of Toronto, Toronto, Ont., Canada (Rusjan, Houle, Wilson, Mizrahi); and the Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Rusjan, Houle, Sailasuta, Mizrahi)
| | - Alan A Wilson
- From the Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Silva, Hafizi, Rusjan, Houle, Wilson, Prce, Sailasuta, Mizrahi); the Institute of Medical Science, University of Toronto, Toronto, Ont., Canada (Silva, Rusjan, Mizrahi); the Department of Psychiatry, University of Toronto, Toronto, Ont., Canada (Rusjan, Houle, Wilson, Mizrahi); and the Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Rusjan, Houle, Sailasuta, Mizrahi)
| | - Ivana Prce
- From the Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Silva, Hafizi, Rusjan, Houle, Wilson, Prce, Sailasuta, Mizrahi); the Institute of Medical Science, University of Toronto, Toronto, Ont., Canada (Silva, Rusjan, Mizrahi); the Department of Psychiatry, University of Toronto, Toronto, Ont., Canada (Rusjan, Houle, Wilson, Mizrahi); and the Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Rusjan, Houle, Sailasuta, Mizrahi)
| | - Napapon Sailasuta
- From the Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Silva, Hafizi, Rusjan, Houle, Wilson, Prce, Sailasuta, Mizrahi); the Institute of Medical Science, University of Toronto, Toronto, Ont., Canada (Silva, Rusjan, Mizrahi); the Department of Psychiatry, University of Toronto, Toronto, Ont., Canada (Rusjan, Houle, Wilson, Mizrahi); and the Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Rusjan, Houle, Sailasuta, Mizrahi)
| | - Romina Mizrahi
- From the Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Silva, Hafizi, Rusjan, Houle, Wilson, Prce, Sailasuta, Mizrahi); the Institute of Medical Science, University of Toronto, Toronto, Ont., Canada (Silva, Rusjan, Mizrahi); the Department of Psychiatry, University of Toronto, Toronto, Ont., Canada (Rusjan, Houle, Wilson, Mizrahi); and the Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ont., Canada (Rusjan, Houle, Sailasuta, Mizrahi)
| |
Collapse
|
49
|
Peltier MR, Verplaetse TL, Mineur YS, Petrakis IL, Cosgrove KP, Picciotto MR, McKee SA. Sex differences in stress-related alcohol use. Neurobiol Stress 2019; 10:100149. [PMID: 30949562 PMCID: PMC6430711 DOI: 10.1016/j.ynstr.2019.100149] [Citation(s) in RCA: 255] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/30/2019] [Accepted: 01/30/2019] [Indexed: 01/12/2023] Open
Abstract
Rates of alcohol use disorder (AUD) have increased in women by 84% over the past ten years relative to a 35% increase in men. This substantive increase in female drinking is alarming given that women experience greater alcohol-related health consequences compared to men. Stress is strongly associated with all phases of alcohol addiction, including drinking initiation, maintenance, and relapse for both women and men, but plays an especially critical role for women. The purpose of the present narrative review is to highlight what is known about sex differences in the relationship between stress and drinking. The critical role stress reactivity and negative affect play in initiating and maintaining alcohol use in women is addressed, and the available evidence for sex differences in drinking for negative reinforcement as it relates to brain stress systems is presented. This review discusses the critical structures and neurotransmitters that may underlie sex differences in stress-related alcohol use (e.g., prefrontal cortex, amygdala, norepinephrine, corticotropin releasing factor, and dynorphin), the involvement of sex and stress in alcohol-induced neurodegeneration, and the role of ovarian hormones in stress-related drinking. Finally, the potential avenues for the development of sex-appropriate pharmacological and behavioral treatments for AUD are identified. Overall, women are generally more likely to drink to regulate negative affect and stress reactivity. Sex differences in the onset and maintenance of alcohol use begin to develop during adolescence, coinciding with exposure to early life stress. These factors continue to affect alcohol use into adulthood, when reduced responsivity to stress, increased affect-related psychiatric comorbidities and alcohol-induced neurodegeneration contribute to chronic and problematic alcohol use, particularly for women. However, current research is limited regarding the examination of sex in the initiation and maintenance of alcohol use. Probing brain stress systems and associated brain regions is an important future direction for developing sex-appropriate treatments to address the role of stress in AUD.
Collapse
Affiliation(s)
| | | | - Yann S. Mineur
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Ismene L. Petrakis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06519, USA
- VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Kelly P. Cosgrove
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06519, USA
- Department of Diagnostic Radiology, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Marina R. Picciotto
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Sherry A. McKee
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06519, USA
| |
Collapse
|
50
|
Kohno M, Link J, Dennis LE, McCready H, Huckans M, Hoffman WF, Loftis JM. Neuroinflammation in addiction: A review of neuroimaging studies and potential immunotherapies. Pharmacol Biochem Behav 2019; 179:34-42. [PMID: 30695700 DOI: 10.1016/j.pbb.2019.01.007] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 01/22/2019] [Accepted: 01/25/2019] [Indexed: 12/29/2022]
Abstract
Addiction is a worldwide public health problem and this article reviews scientific advances in identifying the role of neuroinflammation in the genesis, maintenance, and treatment of substance use disorders. With an emphasis on neuroimaging techniques, this review examines human studies of addiction using positron emission tomography to identify binding of translocator protein (TSPO), which is upregulated in reactive glial cells and activated microglia during pathological states. High TSPO levels have been shown in methamphetamine use but exhibits variable patterns in cocaine use. Alcohol and nicotine use, however, are associated with lower TSPO levels. We discuss how mechanistic differences at the neurotransmitter and circuit level in the neural effects of these agents and subsequent immune response may explain these observations. Finally, we review the potential of anti-inflammatory drugs, including ibudilast, minocycline, and pioglitazone, to ameliorate the behavioral and cognitive consequences of addiction.
Collapse
Affiliation(s)
- Milky Kohno
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Jeanne Link
- Center for Radiochemistry Research, Knight Cardiovascular Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA
| | - Laura E Dennis
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Holly McCready
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Marilyn Huckans
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Mental Health and Clinical Neurosciences Division, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - William F Hoffman
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Mental Health and Clinical Neurosciences Division, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Jennifer M Loftis
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA.
| |
Collapse
|