1
|
Jobling AI, Greferath U, Dixon MA, Quiriconi P, Eyar B, van Koeverden AK, Mills SA, Vessey KA, Bui BV, Fletcher EL. Microglial regulation of the retinal vasculature in health and during the pathology associated with diabetes. Prog Retin Eye Res 2025; 106:101349. [PMID: 40020909 DOI: 10.1016/j.preteyeres.2025.101349] [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: 11/20/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The high metabolic demand of retinal neurons requires a precisely regulated vascular system that can deliver rapid changes in blood flow in response to neural need. In the retina, this is achieved via the action of a coordinated group of cells that form the neurovascular unit. While cells such as pericytes, Müller cells, and astrocytes have long been linked to neurovascular coupling, more recently the resident microglial population have also been implicated. In the healthy retina, microglia make extensive contact with blood vessels, as well as neuronal synapses, and are important in vascular patterning during development. Work in the brain and retina has recently indicated that microglia can directly regulate the local vasculature. In the retina, the fractalkine-Cx3cr1 signalling axis has been shown to induce local capillary constriction within the superficial vascular plexus via a mechanism involving components of the renin-angiotensin system. Furthermore, aberrant microglial induced vasoconstriction may be at the centre of early vascular reactivity changes observed in those with diabetes. This review summarizes the recent emerging evidence that microglia play multiple roles in retinal homeostasis especially in regulating the vasculature. We highlight what is known about the role of microglia under normal circumstances, and then build on this to discuss how microglia contribute to early vascular compromise during diabetes. Further understanding of the mechanisms of microglial-vascular regulation may allow alternate treatment strategies to be devised to reduce vascular pathology in diseases such as diabetic retinopathy.
Collapse
Affiliation(s)
- Andrew I Jobling
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Ursula Greferath
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Michael A Dixon
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Pialuisa Quiriconi
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Belinda Eyar
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Anna K van Koeverden
- Department of Optometry and Vision Sciences, The University of Melbourne, Victoria, Australia
| | - Samuel A Mills
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Kirstan A Vessey
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Victoria, Australia
| | - Erica L Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia.
| |
Collapse
|
2
|
Perez-Castro L, Nawas AF, Kilgore JA, Garcia R, Lafita-Navarro M, Acosta PH, Nogueira PAS, Williams NS, Conacci-Sorrell M. Tryptophan metabolite atlas uncovers organ, age, and sex-specific variations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.23.630041. [PMID: 39763948 PMCID: PMC11703250 DOI: 10.1101/2024.12.23.630041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Although tryptophan (Trp) is the largest and most structurally complex amino acid, it is the least abundant in the proteome. Its distinct indole ring and high carbon content enable it to generate various biologically active metabolites such as serotonin, kynurenine (Kyn), and indole-3-pyruvate (I3P). Dysregulation of Trp metabolism has been implicated in diseases ranging from depression to cancer. Investigating Trp and its metabolites in healthy tissues offers pathways to target disease-associated disruptions selectively, while preserving essential functions. In this study, we comprehensively mapped Trp metabolites across the Kyn, serotonin, and I3P pathways, as well as the microbiome-derived metabolite tryptamine, in C57BL/6 mice. Our comprehensive analysis covered 12 peripheral organs, the central nervous system, and serum in both male and female mice at three life stages: young (3 weeks), adult (54 weeks), and aged (74 weeks). We found significant tissue-, sex-, and age-specific variations in Trp metabolism, with notably higher levels of the oncometabolites I3P and Kyn in aging males. These findings emphasize the value of organ-specific analysis of Trp metabolism for understanding its role in disease progression and identifying targeted therapeutic opportunities.
Collapse
Affiliation(s)
- Lizbeth Perez-Castro
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Afshan F. Nawas
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Jessica A. Kilgore
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Roy Garcia
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - M.Carmen Lafita-Navarro
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Paul H. Acosta
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Pedro A. S. Nogueira
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Noelle S. Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Maralice Conacci-Sorrell
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
3
|
Daugherty-Lopès A, Pérez-Guijarro E, Gopalan V, Rappaport J, Chen Q, Huang A, Lam KC, Chin S, Ebersole J, Wu E, Needle GA, Church I, Kyriakopoulos G, Xie S, Zhao Y, Gruen C, Sassano A, Araya RE, Thorkelsson A, Smith C, Lee MP, Hannenhalli S, Day CP, Merlino G, Goldszmid RS. IMMUNE AND MOLECULAR CORRELATES OF RESPONSE TO IMMUNOTHERAPY REVEALED BY BRAIN-METASTATIC MELANOMA MODELS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609785. [PMID: 39372744 PMCID: PMC11451731 DOI: 10.1101/2024.08.26.609785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Despite the promising results of immune checkpoint blockade (ICB) therapy, outcomes for patients with brain metastasis (BrM) remain poor. Identifying resistance mechanisms has been hindered by limited access to patient samples and relevant preclinical models. Here, we developed two mouse melanoma BrM models that recapitulate the disparate responses to ICB seen in patients. We demonstrate that these models capture the cellular and molecular complexity of human disease and reveal key factors shaping the tumor microenvironment and influencing ICB response. BR1-responsive tumor cells express inflammatory programs that polarize microglia into reactive states, eliciting robust T cell recruitment. In contrast, BR3-resistant melanoma cells are enriched in neurological programs and exploit tolerance mechanisms to maintain microglia homeostasis and limit T cell infiltration. In humans, BR1 and BR3 expression signatures correlate positively or negatively with T cell infiltration and BrM patient outcomes, respectively. Our study provides clinically relevant models and uncovers mechanistic insights into BrM ICB responses, offering potential biomarkers and therapeutic targets to improve therapy efficacy.
Collapse
Affiliation(s)
- Amélie Daugherty-Lopès
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Eva Pérez-Guijarro
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Vishaka Gopalan
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jessica Rappaport
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Quanyi Chen
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Kelly Government Solutions, Bethesda, MD, USA
| | - April Huang
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Kelly Government Solutions, Bethesda, MD, USA
| | - Khiem C. Lam
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Sung Chin
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21701, USA
| | - Jessica Ebersole
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21701, USA
| | - Emily Wu
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Gabriel A. Needle
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Isabella Church
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - George Kyriakopoulos
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Shaojun Xie
- CCR-SF Bioinformatics Team, Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Yongmei Zhao
- CCR-SF Bioinformatics Team, Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Charli Gruen
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Antonella Sassano
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Romina E. Araya
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Andres Thorkelsson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Cari Smith
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21701, USA
| | - Maxwell P. Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Romina S. Goldszmid
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
4
|
Li X, He E, Chen G, Cao X, Zhao L, Xu X, Fu Z, Qiu H. Intergenerational neurotoxicity of polystyrene nanoplastics in offspring mice is mediated by dysfunctional microbe-gut-brain axis. ENVIRONMENT INTERNATIONAL 2024; 192:109026. [PMID: 39321539 DOI: 10.1016/j.envint.2024.109026] [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: 05/21/2024] [Revised: 08/28/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Nanoplastics (NPs) are ubiquitous in daily life, posing potential risks to the environment and human. While their negative effects on parental organisms have been extensively studied, intergenerational effects are still in the early stages of investigation. Here, we aimed to investigate the impact of maternal exposure to an environmentally relevant level of polystyrene NPs (PSNPs, 100 nm) during gestation and lactation (∼32 days, 50 μg/mouse/day) on neurotoxicity mediated by the microbe-gut-brain axis in offspring mice. Maternal PSNPs exposure significantly increased brain TNF-α level and microglia by 1.43 and 1.48 folds respectively, compared to control, accompanied by nuclear pyknosis and cell vacuolization in cortex and hippocampus. Targeted neurotransmitter metabolomics analysis revealed dysregulation in dopamine and serotonin metabolism. Specifically, dopamine levels increased significantly from 0.007 ng/L to 0.015 ng/L, while N-acetylseroton and 3,4-dihydroxyphenylacetic acid decreased significantly from 0.002 and 0.929 ng/L to 0.001 and 0.680 ng/L, respectively. Through a combination of 16S rRNA sequencing and biochemical analysis, we discovered that maternal PSNPs exposure led to a depletion of anti-inflammatory bacteria and an enrichment of pro-inflammatory bacteria resulting in intestinal barrier damage, elevated levels of lipopolysaccharide in blood, and subsequent activation of neuroinflammation. Meanwhile, gut bacteria dysbiosis interfered with communication between gut and brain by dysregulating neurotransmitter synthesis, as evidenced by significant associations between neurotransmitter-related bacteria (Akkermansia, Family_XIII_AD3011_group, Lachnoclostridium) and dopamine/serotonin related metabolites. Furthermore, transcriptional alterations in dopamine and serotonin related pathways were observed in the enteric nervous system, suggesting abnormal signal transduction from gut to brain contributes to neurotoxicity. This study provides new insights into NPs-induced neurotoxicity within the context of microbe-gut-brain axis and highlights the risk of cerebral dysfunction in offspring with maternal NPs exposure.
Collapse
Affiliation(s)
- Xing Li
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Erkai He
- School of Geographic Sciences, East China Normal University, Shanghai 200241, China
| | - Guangquan Chen
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Xinde Cao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ling Zhao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyun Xu
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhuozhong Fu
- School of Geographic Sciences, East China Normal University, Shanghai 200241, China
| | - Hao Qiu
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
5
|
Zhao Q, Chen J, Wu M, Yin X, Jiang Q, Gao H, Zheng H. Microbiota from healthy mice alleviates cognitive decline via reshaping the gut-brain metabolic axis in diabetic mice. Chem Biol Interact 2023; 382:110638. [PMID: 37473910 DOI: 10.1016/j.cbi.2023.110638] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Diabetic cognitive decline has been associated with the gut microbial disorders, but its potential gut-brain axis mechanisms remain unclear. Herein we transplanted the gut microbiota from healthy mice into type 1 diabetic (T1D) mice and then investigated the effect of fecal microbiota transplantation (FMT) on cognitive function and the gut-brain metabolic axis. The results demonstrate that FMT from healthy mice effectively improved the learning and memory abilities in T1D mice, and significantly reduced neuroinflammation and neuron injury in the cortex and hippocampus. Moreover, FMT partly reversed the gut microbiota and gut-brain metabolic disorders, particularly glutamate metabolism. In vitro study, we found that glutamate notably decreased microglia activation and the expression levels of proinflammatory factor. Hence, our study suggests that glutamate serves as a key signal metabolite connecting the gut to brain and affects cognitive functions.
Collapse
Affiliation(s)
- Qihui Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Junli Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Mengjun Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiaoli Yin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qiaoying Jiang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hongchang Gao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Hong Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
6
|
Matrisciano F. Epigenetic regulation of metabotropic glutamate 2/3 receptors: Potential role for ultra-resistant schizophrenia? Pharmacol Biochem Behav 2023:173589. [PMID: 37348609 DOI: 10.1016/j.pbb.2023.173589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023]
Abstract
Schizophrenia is a severe and debilitating psychiatric disorder characterized by early cognitive deficits, emotional and behavioral abnormalities resulted by a dysfunctional gene x environment interaction. Genetic and epigenetic abnormalities in cortical parvalbumin-positive GABAergic interneurons lead to alterations in glutamate-mediated excitatory neurotransmission, synaptic plasticity, and neuronal development. Epigenetic alterations during pregnancy or early phases of postnatal life are associated with schizophrenia vulnerability as well as inflammatory processes which are at the basis of brain pathology. An epigenetic animal model of schizophrenia showed specific changes in promoter DNA methylation activity of genes related to schizophrenia such as reelin, BDNF and GAD67, and altered expression and function of mGlu2/3 receptors in the frontal cortex. Although antipsychotic medications represent the main treatment for schizophrenia and generally show an optimal efficacy profile for positive symptoms and relatively poor efficacy for negative or cognitive symptoms, a considerable percentage of individuals show poor response, do not achieve a complete remission, and approximately 30 % of patients show treatment-resistance. Here, we explore the potential role of epigenetic abnormalities linked to metabotropic glutamate 2/3 receptors changes in expression and function as key molecular factors underlying the difference in response to antipsychotics.
Collapse
Affiliation(s)
- Francesco Matrisciano
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago (UIC), Chicago, IL, USA.
| |
Collapse
|
7
|
Balbi M, Bonanno G, Bonifacino T, Milanese M. The Physio-Pathological Role of Group I Metabotropic Glutamate Receptors Expressed by Microglia in Health and Disease with a Focus on Amyotrophic Lateral Sclerosis. Int J Mol Sci 2023; 24:5240. [PMID: 36982315 PMCID: PMC10048889 DOI: 10.3390/ijms24065240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
Microglia cells are the resident immune cells of the central nervous system. They act as the first-line immune guardians of nervous tissue and central drivers of neuroinflammation. Any homeostatic alteration that can compromise neuron and tissue integrity could activate microglia. Once activated, microglia exhibit highly diverse phenotypes and functions related to either beneficial or harmful consequences. Microglia activation is associated with the release of protective or deleterious cytokines, chemokines, and growth factors that can in turn determine defensive or pathological outcomes. This scenario is complicated by the pathology-related specific phenotypes that microglia can assume, thus leading to the so-called disease-associated microglia phenotypes. Microglia express several receptors that regulate the balance between pro- and anti-inflammatory features, sometimes exerting opposite actions on microglial functions according to specific conditions. In this context, group I metabotropic glutamate receptors (mGluRs) are molecular structures that may contribute to the modulation of the reactive phenotype of microglia cells, and this is worthy of exploration. Here, we summarize the role of group I mGluRs in shaping microglia cells' phenotype in specific physio-pathological conditions, including some neurodegenerative disorders. A significant section of the review is specifically focused on amyotrophic lateral sclerosis (ALS) since it represents an entirely unexplored topic of research in the field.
Collapse
Affiliation(s)
- Matilde Balbi
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy (M.M.)
| | - Giambattista Bonanno
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy (M.M.)
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy (M.M.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Marco Milanese
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy (M.M.)
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| |
Collapse
|
8
|
Holton K. The potential role of dietary intervention for the treatment of neuroinflammation. TRANSLATIONAL NEUROIMMUNOLOGY, VOLUME 7 2023:239-266. [DOI: 10.1016/b978-0-323-85841-0.00022-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
9
|
Liu C, She Y, Huang J, Liu Y, Li W, Zhang C, Zhang T, Yu L. HMGB1-NLRP3-P2X7R pathway participates in PM 2.5-induced hippocampal neuron impairment by regulating microglia activation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 239:113664. [PMID: 35605331 DOI: 10.1016/j.ecoenv.2022.113664] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 06/15/2023]
Abstract
Neuroinflammation is a key mechanism underlying the cognitive impairment induced by PM2.5, and activated microglia plays an important role in this process. However, the mechanisms by which activated microglia induced by PM2.5 impair hippocampal neurons have not been fully elucidated. In this study, we focused on the role of HMGB1-NLRP3-P2X7R pathway which mediated the microglia activation in hippocampal neurons impairment induced by PM2.5 using a co-culture model of microglia and hippocampal neurons. We found that PM2.5 resulted in activated microglia and HMGB1-NLRP3 inflammatory pathway, and elevated proinflammatory cytokines of IL-18 and IL-1β in a dose-dependent manner. Notably, we next utilized previously reported pharmacological inhibitors or siRNA for HMGB1 and found that they significantly inhibited the activation of downstream NLRP3 and MAPK pathways derived from PM2.5 exposure, and down-regulated IL-18 and IL-1β in microglia. Furthermore, we employed co-cultured hippocampal neurons and microglia and found that reducing HMGB1 significantly decreased neuron impairment, apoptosis related protein of cl-caspase3, synaptic damage, and neurotransmitter receptor of 5-HT2A, along with notably elevated presynaptic and postsynaptic proteins of SYP and PSD-95, as well as learning and memory related proteins of p-CREB and BDNF. The neuronal impairment induced by PM2.5 could not be prevented in the case of simultaneous employment of HMGB1 siRNA and NLRP3 agonist. After silencing NLRP3 alone in microglia, hippocampal neurons demonstrated decreased excessive autophagy and up-regulated synaptic protein of GAP43 as well as learning and memory related protein of NCAM1. Therefore, we further studied how hippocampal neurons affected microglia under PM2.5 exposure, Further investigation indicated that silencing HMGB1 could affect the activation of P2X7R and reduce the release of ATP from hippocampal neurons, thus protecting the interaction between microglia and hippocampal neurons. The present work suggests that regulation of HMGB1-NLRP3-P2X7R pathway can inhibit the microglia activation induced by PM2.5 to alleviate hippocampal neuron impairment and stabilize the microenvironment between microglia and neurons. This contributes to maintaining the normal function of hippocampal neurons and alleviating the cognitive impairment derived from PM2.5 exposure.
Collapse
Affiliation(s)
- Chong Liu
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China
| | - Yingjie She
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China
| | - Jia Huang
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China
| | - Yongping Liu
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China
| | - Wanwei Li
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Tianliang Zhang
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China.
| | - Li Yu
- School of Basic Medical Sciences, Experimental Center for Medical Research, Neurologic Disorders and Regeneration Repair Lab of Shandong Higher Education, Weifang Medical University, Weifang, China.
| |
Collapse
|
10
|
Biological Potential, Gastrointestinal Digestion, Absorption, and Bioavailability of Algae-Derived Compounds with Neuroprotective Activity: A Comprehensive Review. Mar Drugs 2022; 20:md20060362. [PMID: 35736165 PMCID: PMC9227170 DOI: 10.3390/md20060362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
Currently, there is no known cure for neurodegenerative disease. However, the available therapies aim to manage some of the symptoms of the disease. Human neurodegenerative diseases are a heterogeneous group of illnesses characterized by progressive loss of neuronal cells and nervous system dysfunction related to several mechanisms such as protein aggregation, neuroinflammation, oxidative stress, and neurotransmission dysfunction. Neuroprotective compounds are essential in the prevention and management of neurodegenerative diseases. This review will focus on the neurodegeneration mechanisms and the compounds (proteins, polyunsaturated fatty acids (PUFAs), polysaccharides, carotenoids, phycobiliproteins, phenolic compounds, among others) present in seaweeds that have shown in vivo and in vitro neuroprotective activity. Additionally, it will cover the recent findings on the neuroprotective effects of bioactive compounds from macroalgae, with a focus on their biological potential and possible mechanism of action, including microbiota modulation. Furthermore, gastrointestinal digestion, absorption, and bioavailability will be discussed. Moreover, the clinical trials using seaweed-based drugs or extracts to treat neurodegenerative disorders will be presented, showing the real potential and limitations that a specific metabolite or extract may have as a new therapeutic agent considering the recent approval of a seaweed-based drug to treat Alzheimer’s disease.
Collapse
|
11
|
Zhang Y, Chu JMT, Wong GTC. Cerebral Glutamate Regulation and Receptor Changes in Perioperative Neuroinflammation and Cognitive Dysfunction. Biomolecules 2022; 12:biom12040597. [PMID: 35454185 PMCID: PMC9029551 DOI: 10.3390/biom12040597] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 12/23/2022] Open
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system and is intricately linked to learning and memory. Its activity depends on the expression of AMPA and NMDA receptors and excitatory amino transporters on neurons and glial cells. Glutamate transporters prevent the excess accumulation of glutamate in synapses, which can lead to aberrant synaptic signaling, excitotoxicity, or cell death. Neuroinflammation can occur acutely after surgical trauma and contributes to the development of perioperative neurocognitive disorders, which are characterized by impairment in multiple cognitive domains. In this review, we aim to examine how glutamate handling and glutamatergic function are affected by neuroinflammation and their contribution to cognitive impairment. We will first summarize the current data regarding glutamate in neurotransmission, its receptors, and their regulation and trafficking. We will then examine the impact of inflammation on glutamate handling and neurotransmission, focusing on changes in glial cells and the effect of cytokines. Finally, we will discuss these changes in the context of perioperative neuroinflammation and the implications they have for perioperative neurocognitive disorders.
Collapse
|
12
|
Gómez-Piñeiro RJ, Dali M, Mansuy D, Boucher JL. Unstability of cinnabarinic acid, an endogenous metabolite of tryptophan, under situations mimicking physiological conditions. Biochimie 2022; 199:150-157. [DOI: 10.1016/j.biochi.2022.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 12/25/2022]
|
13
|
Dyomina AV, Kovalenko AA, Zakharova MV, Postnikova TY, Griflyuk AV, Smolensky IV, Antonova IV, Zaitsev AV. MTEP, a Selective mGluR5 Antagonist, Had a Neuroprotective Effect but Did Not Prevent the Development of Spontaneous Recurrent Seizures and Behavioral Comorbidities in the Rat Lithium-Pilocarpine Model of Epilepsy. Int J Mol Sci 2022; 23:ijms23010497. [PMID: 35008924 PMCID: PMC8745728 DOI: 10.3390/ijms23010497] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 02/08/2023] Open
Abstract
Metabotropic glutamate receptors (mGluRs) are expressed predominantly on neurons and glial cells and are involved in the modulation of a wide range of signal transduction cascades. Therefore, different subtypes of mGluRs are considered a promising target for the treatment of various brain diseases. Previous studies have demonstrated the seizure-induced upregulation of mGluR5; however, its functional significance is still unclear. In the present study, we aimed to clarify the effect of treatment with the selective mGluR5 antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]-pyridine (MTEP) on epileptogenesis and behavioral impairments in rats using the lithium–pilocarpine model. We found that the administration of MTEP during the latent phase of the model did not improve survival, prevent the development of epilepsy, or attenuate its manifestations in rats. However, MTEP treatment completely prevented neuronal loss and partially attenuated astrogliosis in the hippocampus. An increase in excitatory amino acid transporter 2 expression, which has been detected in treated rats, may prevent excitotoxicity and be a potential mechanism of neuroprotection. We also found that MTEP administration did not prevent the behavioral comorbidities such as depressive-like behavior, motor hyperactivity, reduction of exploratory behavior, and cognitive impairments typical in the lithium–pilocarpine model. Thus, despite the distinct neuroprotective effect, the MTEP treatment was ineffective in preventing epilepsy.
Collapse
|
14
|
The Glutamate Receptor Plays a Role in Defense against Botrytis cinerea through Electrical Signaling in Tomato. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app112311217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Plant glutamate-like receptor genes (GLRs) are homologous to mammalian ionotropic glutamate receptors genes (iGluRs). Although GLRs have been implicated in plant defenses to biotic stress, the relationship between GLR-mediated plant immunity against fungal pathogens and electrical signals remains poorly understood. Here, we found that pretreatment with a GLR inhibitor, 6,7-dinitriquinoxaline-2,3-dione (DNQX), increased the susceptibility of tomato plants to the necrotrophic fungal pathogen Botrytis cinerea. Assessment of the glr3.3, glr3.5 and glr3.3/glr3.5 double-mutants upon B. cinerea infection showed that tomato GLR3.3 and GLR3.5 are essential for plant immunity against B. cinerea, wherein GLR3.3 plays the main role. Analysis of the membrane potential changes induced by glutamate (Glu) or glycine (Gly) revealed that amplitude was significantly reduced by knocking out GLR3.3 in tomato. While treatment with Glu or Gly significantly increased immunity against B. cinerea in wild-type plants, this effect was significantly attenuated in glr3.3 mutants. Thus, our data demonstrate that GLR3.3- and GLR3.5-mediated plant immunity against B. cinerea is associated with electrical signals in tomato plants.
Collapse
|
15
|
Glutamate triggers the expression of functional ionotropic and metabotropic glutamate receptors in mast cells. Cell Mol Immunol 2021; 18:2383-2392. [PMID: 32313211 PMCID: PMC8484602 DOI: 10.1038/s41423-020-0421-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 03/16/2020] [Indexed: 02/07/2023] Open
Abstract
Mast cells are emerging as players in the communication between peripheral nerve endings and cells of the immune system. However, it is not clear the mechanism by which mast cells communicate with peripheral nerves. We previously found that mast cells located within healing tendons can express glutamate receptors, raising the possibility that mast cells may be sensitive to glutamate signaling. To evaluate this hypothesis, we stimulated primary mast cells with glutamate and showed that glutamate induced the profound upregulation of a panel of glutamate receptors of both the ionotropic type (NMDAR1, NMDAR2A, and NMDAR2B) and the metabotropic type (mGluR2 and mGluR7) at both the mRNA and protein levels. The binding of glutamate to glutamate receptors on the mast cell surface was confirmed. Further, glutamate had extensive effects on gene expression in the mast cells, including the upregulation of pro-inflammatory components such as IL-6 and CCL2. Glutamate also induced the upregulation of transcription factors, including Egr2, Egr3 and, in particular, FosB. The extensive induction of FosB was confirmed by immunofluorescence assessment. Glutamate receptor antagonists abrogated the responses of the mast cells to glutamate, supporting the supposition of a functional glutamate-glutamate receptor axis in mast cells. Finally, we provide in vivo evidence supporting a functional glutamate-glutamate receptor axis in the mast cells of injured tendons. Together, these findings establish glutamate as an effector of mast cell function, thereby introducing a novel principle for how cells in the immune system can communicate with nerve cells.
Collapse
|
16
|
Qiu W, Go KA, Lamers Y, Galea LAM. Postpartum corticosterone and fluoxetine shift the tryptophan-kynurenine pathway in dams. Psychoneuroendocrinology 2021; 130:105273. [PMID: 34051656 DOI: 10.1016/j.psyneuen.2021.105273] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/29/2021] [Accepted: 05/13/2021] [Indexed: 02/05/2023]
Abstract
Perinatal depression (PND) affects 15% of mothers. Selective serotonin reuptake inhibitors (SSRIs) are currently the first-line of treatment for PND but are not always efficacious. Previously, we found significant reductions in plasma tryptophan concentrations and higher hippocampal proinflammatory cytokine, IL-1β levels, due to maternal SSRI treatment. Both inflammation and tryptophan-kynurenine metabolic pathway (TKP) are associated with SSRI efficacy in individuals with major depressive disorder (MDD). TKP is divided into neuroprotective and neurotoxic pathways. Higher metabolite concentrations of the neurotoxic pathway are associated with depression onset and implicated in SSRI efficacy. Metabolites in TKP were investigated in a rodent model of de novo postpartum depression (PPD) given treatment with the SSRI, fluoxetine (FLX). Dams were administered corticosterone (CORT) (40 mg/kg, s.c.), and treated with the SSRI, fluoxetine (FLX) (10 mg/kg, s.c.), during the postpartum for 22 days after parturition. Plasma TKP metabolite concentrations were quantified on the last day of treatment. Maternal postpartum CORT increased neurotoxic metabolites and co-enzyme/cofactors in dams (3-hydroxykynurenine, 3-hydroxyanthranilic acid, vitamin B2, flavin adenine dinucleotide). The combination of both CORT and FLX shifted the neuroprotective-to-neurotoxic ratio towards neurotoxicity. Postpartum FLX decreased plasma xanthurenic acid concentrations. Together, our data indicate higher neurotoxic TKP expression due to maternal postpartum CORT treatment, similar to clinical presentation of MDD. Moreover, maternal FLX treatment showed limited efficacy to influence TKP metabolites, which may correspond to its limited efficacy to treat depressive-like endophenotypes in the postpartum. Overall suggesting changes in TKP may be used as a biomarker of de novo PPD and antidepressant efficacy and targeting this pathway may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Canada
| | - Kimberly A Go
- Department of Psychology, University of British Columbia, Canada
| | - Yvonne Lamers
- Food Nutrition and Health Program, Faculty of Land and Food Systems, University of British Columbia, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Canada; Department of Psychology, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada.
| |
Collapse
|
17
|
Castillo CA, Ballesteros-Yáñez I, León-Navarro DA, Albasanz JL, Martín M. Early Effects of the Soluble Amyloid β 25-35 Peptide in Rat Cortical Neurons: Modulation of Signal Transduction Mediated by Adenosine and Group I Metabotropic Glutamate Receptors. Int J Mol Sci 2021; 22:ijms22126577. [PMID: 34205261 PMCID: PMC8234864 DOI: 10.3390/ijms22126577] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/04/2021] [Accepted: 06/17/2021] [Indexed: 12/20/2022] Open
Abstract
The amyloid β peptide (Aβ) is a central player in the neuropathology of Alzheimer’s disease (AD). The alteration of Aβ homeostasis may impact the fine-tuning of cell signaling from the very beginning of the disease, when amyloid plaque is not deposited yet. For this reason, primary culture of rat cortical neurons was exposed to Aβ25-35, a non-oligomerizable form of Aβ. Cell viability, metabotropic glutamate receptors (mGluR) and adenosine receptors (AR) expression and signalling were assessed. Aβ25-35 increased mGluR density and affinity, mainly due to a higher gene expression and protein presence of Group I mGluR (mGluR1 and mGluR5) in the membrane of cortical neurons. Intriguingly, the main effector of group I mGluR, the phospholipase C β1 isoform, was less responsive. Also, the inhibitory action of group II and group III mGluR on adenylate cyclase (AC) activity was unaltered or increased, respectively. Interestingly, pre-treatment of cortical neurons with an antagonist of group I mGluR reduced the Aβ25-35-induced cell death. Besides, Aβ25-35 increased the density of A1R and A2AR, along with an increase in their gene expression. However, while A1R-mediated AC inhibition was increased, the A2AR-mediated stimulation of AC remained unchanged. Therefore, one of the early events that takes place after Aβ25-35 exposure is the up-regulation of adenosine A1R, A2AR, and group I mGluR, and the different impacts on their corresponding signaling pathways. These results emphasize the importance of deciphering the early events and the possible involvement of metabotropic glutamate and adenosine receptors in AD physiopathology.
Collapse
Affiliation(s)
- Carlos Alberto Castillo
- Department of Nursing, Physiotherapy and Occupational Therapy, School of Physiotherapy and Nursing, University of Castilla-La Mancha, 45071 Toledo, Spain;
- Regional Center for Biomedical Research (CRIB), University of Castilla-La Mancha, 02071 Albacete, Spain; (I.B.-Y.); (D.A.L.-N.); (M.M.)
| | - Inmaculada Ballesteros-Yáñez
- Regional Center for Biomedical Research (CRIB), University of Castilla-La Mancha, 02071 Albacete, Spain; (I.B.-Y.); (D.A.L.-N.); (M.M.)
- Department of Inorganic, School of Medicine of Ciudad Real, Organic and Biochemistry, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - David Agustín León-Navarro
- Regional Center for Biomedical Research (CRIB), University of Castilla-La Mancha, 02071 Albacete, Spain; (I.B.-Y.); (D.A.L.-N.); (M.M.)
- Department of Inorganic, Faculty of Chemical and Technological Sciences, Organic and Biochemistry, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - José Luis Albasanz
- Regional Center for Biomedical Research (CRIB), University of Castilla-La Mancha, 02071 Albacete, Spain; (I.B.-Y.); (D.A.L.-N.); (M.M.)
- Department of Inorganic, School of Medicine of Ciudad Real, Organic and Biochemistry, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
- Correspondence:
| | - Mairena Martín
- Regional Center for Biomedical Research (CRIB), University of Castilla-La Mancha, 02071 Albacete, Spain; (I.B.-Y.); (D.A.L.-N.); (M.M.)
- Department of Inorganic, Faculty of Chemical and Technological Sciences, Organic and Biochemistry, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| |
Collapse
|
18
|
Targeting the dysfunction of glutamate receptors for the development of novel antidepressants. Pharmacol Ther 2021; 226:107875. [PMID: 33901503 DOI: 10.1016/j.pharmthera.2021.107875] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2021] [Indexed: 12/19/2022]
Abstract
Increasing evidence indicates that dysfunction of glutamate receptors is involved in the pathophysiology of major depressive disorder (MDD). Although accumulating efforts have been made to elucidate the applications and mechanisms underlying antidepressant-like effects of ketamine, a non-selective antagonist of N-methyl-d-aspartate receptor (NMDAR), the role of specific glutamate receptor subunit in regulating depression is not completely clear. The current review aims to discuss the relationships between glutamate receptor subunits and depressive-like behaviors. Research literatures were searched from inception to July 2020. We summarized the alterations of glutamate receptor subunits in patients with MDD and animal models of depression. Animal behaviors in response to dysfunction of glutamate receptor subunits were also surveyed. To fully understand mechanisms underlying antidepressant-like effects of modulators targeting glutamate receptors, we discussed effects of each glutamate receptor subunit on serotonin system, synaptic plasticity, neurogenesis and neuroinflammation. Finally, we collected most recent clinical applications of glutamate receptor modulators and pointed out the limitations of these candidates in the treatment of MDD.
Collapse
|
19
|
Namba MD, Leyrer-Jackson JM, Nagy EK, Olive MF, Neisewander JL. Neuroimmune Mechanisms as Novel Treatment Targets for Substance Use Disorders and Associated Comorbidities. Front Neurosci 2021; 15:650785. [PMID: 33935636 PMCID: PMC8082184 DOI: 10.3389/fnins.2021.650785] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Recent studies examining the neurobiology of substance abuse have revealed a significant role of neuroimmune signaling as a mechanism through which drugs of abuse induce aberrant changes in synaptic plasticity and contribute to substance abuse-related behaviors. Immune signaling within the brain and the periphery critically regulates homeostasis of the nervous system. Perturbations in immune signaling can induce neuroinflammation or immunosuppression, which dysregulate nervous system function including neural processes associated with substance use disorders (SUDs). In this review, we discuss the literature that demonstrates a role of neuroimmune signaling in regulating learning, memory, and synaptic plasticity, emphasizing specific cytokine signaling within the central nervous system. We then highlight recent preclinical studies, within the last 5 years when possible, that have identified immune mechanisms within the brain and the periphery associated with addiction-related behaviors. Findings thus far underscore the need for future investigations into the clinical potential of immunopharmacology as a novel approach toward treating SUDs. Considering the high prevalence rate of comorbidities among those with SUDs, we also discuss neuroimmune mechanisms of common comorbidities associated with SUDs and highlight potentially novel treatment targets for these comorbid conditions. We argue that immunopharmacology represents a novel frontier in the development of new pharmacotherapies that promote long-term abstinence from drug use and minimize the detrimental impact of SUD comorbidities on patient health and treatment outcomes.
Collapse
Affiliation(s)
- Mark D. Namba
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | | | - Erin K. Nagy
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - M. Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | | |
Collapse
|
20
|
Gipson CD, Rawls S, Scofield MD, Siemsen BM, Bondy EO, Maher EE. Interactions of neuroimmune signaling and glutamate plasticity in addiction. J Neuroinflammation 2021; 18:56. [PMID: 33612110 PMCID: PMC7897396 DOI: 10.1186/s12974-021-02072-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/05/2021] [Indexed: 02/28/2023] Open
Abstract
Chronic use of drugs of abuse affects neuroimmune signaling; however, there are still many open questions regarding the interactions between neuroimmune mechanisms and substance use disorders (SUDs). Further, chronic use of drugs of abuse can induce glutamatergic changes in the brain, but the relationship between the glutamate system and neuroimmune signaling in addiction is not well understood. Therefore, the purpose of this review is to bring into focus the role of neuroimmune signaling and its interactions with the glutamate system following chronic drug use, and how this may guide pharmacotherapeutic treatment strategies for SUDs. In this review, we first describe neuroimmune mechanisms that may be linked to aberrant glutamate signaling in addiction. We focus specifically on the nuclear factor-kappa B (NF-κB) pathway, a potentially important neuroimmune mechanism that may be a key player in driving drug-seeking behavior. We highlight the importance of astroglial-microglial crosstalk, and how this interacts with known glutamatergic dysregulations in addiction. Then, we describe the importance of studying non-neuronal cells with unprecedented precision because understanding structure-function relationships in these cells is critical in understanding their role in addiction neurobiology. Here we propose a working model of neuroimmune-glutamate interactions that underlie drug use motivation, which we argue may aid strategies for small molecule drug development to treat substance use disorders. Together, the synthesis of this review shows that interactions between glutamate and neuroimmune signaling may play an important and understudied role in addiction processes and may be critical in developing more efficacious pharmacotherapies to treat SUDs.
Collapse
Affiliation(s)
- Cassandra D Gipson
- Department of Family and Community Medicine, University of Kentucky, 741 S. Limestone, BBSRB, Room 363, Lexington, KY, 40536-0509, USA.
| | - Scott Rawls
- Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| | - Michael D Scofield
- Department of Anesthesiology, Medical University of South Carolina, Charleston, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, USA
| | - Benjamin M Siemsen
- Department of Anesthesiology, Medical University of South Carolina, Charleston, USA
| | - Emma O Bondy
- Department of Family and Community Medicine, University of Kentucky, 741 S. Limestone, BBSRB, Room 363, Lexington, KY, 40536-0509, USA
| | - Erin E Maher
- Department of Family and Community Medicine, University of Kentucky, 741 S. Limestone, BBSRB, Room 363, Lexington, KY, 40536-0509, USA
| |
Collapse
|
21
|
Yu J, Zhu H, Taheri S, Mondy W, Perry S, Kindy MS. Plant-Based Nutritional Supplementation Attenuates LPS-Induced Low-Grade Systemic Activation. Int J Mol Sci 2021; 22:ijms22020573. [PMID: 33430045 PMCID: PMC7826722 DOI: 10.3390/ijms22020573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/09/2020] [Accepted: 01/04/2021] [Indexed: 11/21/2022] Open
Abstract
Plant-based nutritional supplementation has been shown to attenuate and reduce mortality in the processes of both acute and chronic disorders, including diabetes, obesity, cardiovascular disease, cancer, inflammatory diseases, and neurological and neurodegenerative disorders. Low-level systemic inflammation is an important contributor to these afflictions and diets enriched in phytochemicals can slow the progression. The goal of this study was to determine the impact of lipopolysaccharide (LPS)-induced inflammation on changes in glucose and insulin tolerance, performance enhancement, levels of urinary neopterin and concentrations of neurotransmitters in the striatum in mouse models. Both acute and chronic injections of LPS (2 mg/kg or 0.33 mg/kg/day, respectively) reduced glucose and insulin tolerance and elevated neopterin levels, which are indicative of systemic inflammatory responses. In addition, there were significant decreases in striatal neurotransmitter levels (dopamine and DOPAC), while serotonin (5-HT) levels were essentially unchanged. LPS resulted in impaired execution in the incremental loading test, which was reversed in mice on a supplemental plant-based diet, improving their immune function and maintaining skeletal muscle mitochondrial activity. In conclusion, plant-based nutritional supplementation attenuated the metabolic changes elicited by LPS injections, causing systemic inflammatory activity that contributed to both systemic and neurological alterations.
Collapse
Affiliation(s)
- Jin Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (J.Y.); (H.Z.); (S.T.); (W.M.)
| | - Hong Zhu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (J.Y.); (H.Z.); (S.T.); (W.M.)
| | - Saeid Taheri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (J.Y.); (H.Z.); (S.T.); (W.M.)
| | - William Mondy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (J.Y.); (H.Z.); (S.T.); (W.M.)
| | | | - Mark S. Kindy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; (J.Y.); (H.Z.); (S.T.); (W.M.)
- Department of Neurology, College of Medicine, University of South Florida, Tampa, FL 33620, USA
- James A. Haley VA Medical Center, Tampa, FL 33612, USA
- Shriners Hospital for Children, Tampa, FL 33612, USA
- Correspondence:
| |
Collapse
|
22
|
Purinergic signaling orchestrating neuron-glia communication. Pharmacol Res 2020; 162:105253. [PMID: 33080321 DOI: 10.1016/j.phrs.2020.105253] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/29/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022]
Abstract
This review discusses the evidence supporting a role for ATP signaling (operated by P2X and P2Y receptors) and adenosine signaling (mainly operated by A1 and A2A receptors) in the crosstalk between neurons, astrocytes, microglia and oligodendrocytes. An initial emphasis will be given to the cooperation between adenosine receptors to sharpen information salience encoding across synapses. The interplay between ATP and adenosine signaling in the communication between astrocytes and neurons will then be presented in context of the integrative properties of the astrocytic syncytium, allowing to implement heterosynaptic depression processes in neuronal networks. The process of microglia 'activation' and its control by astrocytes and neurons will then be analyzed under the perspective of an interplay between different P2 receptors and adenosine A2A receptors. In spite of these indications of a prominent role of purinergic signaling in the bidirectional communication between neurons and glia, its therapeutical exploitation still awaits obtaining an integrated view of the spatio-temporal action of ATP signaling and adenosine signaling, clearly distinguishing the involvement of both purinergic signaling systems in the regulation of physiological processes and in the control of pathogenic-like responses upon brain dysfunction or damage.
Collapse
|
23
|
Ulivieri M, Wierońska JM, Lionetto L, Martinello K, Cieslik P, Chocyk A, Curto M, Di Menna L, Iacovelli L, Traficante A, Liberatore F, Mascio G, Antenucci N, Giannino G, Vergassola M, Pittaluga A, Bruno V, Battaglia G, Fucile S, Simmaco M, Nicoletti F, Pilc A, Fazio F. The Trace Kynurenine, Cinnabarinic Acid, Displays Potent Antipsychotic-Like Activity in Mice and Its Levels Are Reduced in the Prefrontal Cortex of Individuals Affected by Schizophrenia. Schizophr Bull 2020; 46:1471-1481. [PMID: 32506121 PMCID: PMC7846105 DOI: 10.1093/schbul/sbaa074] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cinnabarinic acid (CA) is a kynurenine metabolite that activates mGlu4 metabotropic glutamate receptors. Using a highly sensitive ultra-performance liquid chromatography/tandem mass spectrometry (UPLC/MS-MS) method, we found that CA is present in trace amounts in human brain tissue. CA levels were largely reduced in the prefrontal cortex (PFC) of individuals affected by schizophrenia. This reduction did not correlate with age, sex, duration of the disease, and duration and type of antipsychotic medication and might, therefore, represent a trait of schizophrenia. Interestingly, systemic treatment with low doses of CA (<1 mg/kg, i.p.) showed robust efficacy in several behavioral tests useful to study antipsychotic-like activity in mice and rats and attenuated MK-801-evoked glutamate release. CA failed to display antipsychotic-like activity and inhibit excitatory synaptic transmission in mice lacking mGlu4 receptors. These findings suggest that CA is a potent endogenous antipsychotic-like molecule and reduced CA levels in the PFC might contribute to the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
| | | | - Luana Lionetto
- Department of Medical-Surgical Sciences and Translational Medicine, DiMA (Advanced Molecular Diagnosis), Sant’Andrea Hospital—Sapienza University, Rome, Italy
| | | | - Paulina Cieslik
- Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Agnieszka Chocyk
- Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Martina Curto
- Department of Neurology and Psychiatry, Sapienza University, Rome, Italy,Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy,Bipolar & Psychotic Disorders Program, McLean Hospital, Belmont, MA
| | | | - Luisa Iacovelli
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | | | | | | | - Nico Antenucci
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Giuseppe Giannino
- School of Medicine and Psychology NESMOS Department, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | | | - Anna Pittaluga
- Department of Pharmacy, DiFAR, University of Genoa, Genoa, Italy,I.R.C.C.S. San Martino Hospital, Genoa, Italy
| | - Valeria Bruno
- I.R.C.C.S. Neuromed, Pozzilli, Italy,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Giuseppe Battaglia
- I.R.C.C.S. Neuromed, Pozzilli, Italy,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Sergio Fucile
- I.R.C.C.S. Neuromed, Pozzilli, Italy,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Maurizio Simmaco
- Department of Medical-Surgical Sciences and Translational Medicine, DiMA (Advanced Molecular Diagnosis), Sant’Andrea Hospital—Sapienza University, Rome, Italy
| | - Ferdinando Nicoletti
- I.R.C.C.S. Neuromed, Pozzilli, Italy,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Andrzej Pilc
- Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Francesco Fazio
- I.R.C.C.S. Neuromed, Pozzilli, Italy,To whom correspondence should be addressed; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Rose F. Kennedy Center, 1410 Pelham Parkway South, room 610, New York City, NY, USA; tel: +1-718-430-2160, fax: +1-718-430-8932, e-mail:
| |
Collapse
|
24
|
Alim MA, Peterson M, Pejler G. Do Mast Cells Have a Role in Tendon Healing and Inflammation? Cells 2020; 9:cells9051134. [PMID: 32375419 PMCID: PMC7290807 DOI: 10.3390/cells9051134] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/15/2022] Open
Abstract
Understanding the links between the tendon healing process, inflammatory mechanisms, and tendon homeostasis/pain after tissue damage is crucial in developing novel therapeutics for human tendon disorders. The inflammatory mechanisms that are operative in response to tendon injury are not fully understood, but it has been suggested that inflammation occurring in response to nerve signaling, i.e., neurogenic inflammation, has a pathogenic role. The mechanisms driving such neurogenic inflammation are presently not clear. However, it has recently been demonstrated that mast cells present within the injured tendon can express glutamate receptors, raising the possibility that mast cells may be sensitive to glutamate signaling and thereby modulate neurogenic inflammation following tissue injury. In this review, we discuss the role of mast cells in the communication with peripheral nerves, and their emerging role in tendon healing and inflammation after injury.
Collapse
Affiliation(s)
- Md Abdul Alim
- Department of Public Health and Caring Sciences, General Medicine, Uppsala University, 751 22 Uppsala, Sweden;
- Department of Medical Biochemistry and Microbiology, Uppsala University, 75123 Uppsala, Sweden
- Correspondence: (M.A.A.); (G.P.)
| | - Magnus Peterson
- Department of Public Health and Caring Sciences, General Medicine, Uppsala University, 751 22 Uppsala, Sweden;
- Academic Primary Health Care, Region Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, 75123 Uppsala, Sweden
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, 756 51 Uppsala, Sweden
- Correspondence: (M.A.A.); (G.P.)
| |
Collapse
|
25
|
Regulation of Microglial Functions by Purinergic Mechanisms in the Healthy and Diseased CNS. Cells 2020; 9:cells9051108. [PMID: 32365642 PMCID: PMC7290360 DOI: 10.3390/cells9051108] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Microglial cells, the resident macrophages of the central nervous system (CNS), exist in a process-bearing, ramified/surveying phenotype under resting conditions. Upon activation by cell-damaging factors, they get transformed into an amoeboid phenotype releasing various cell products including pro-inflammatory cytokines, chemokines, proteases, reactive oxygen/nitrogen species, and the excytotoxic ATP and glutamate. In addition, they engulf pathogenic bacteria or cell debris and phagocytose them. However, already resting/surveying microglia have a number of important physiological functions in the CNS; for example, they shield small disruptions of the blood–brain barrier by their processes, dynamically interact with synaptic structures, and clear surplus synapses during development. In neurodegenerative illnesses, they aggravate the original disease by a microglia-based compulsory neuroinflammatory reaction. Therefore, the blockade of this reaction improves the outcome of Alzheimer’s Disease, Parkinson’s Disease, multiple sclerosis, amyotrophic lateral sclerosis, etc. The function of microglia is regulated by a whole array of purinergic receptors classified as P2Y12, P2Y6, P2Y4, P2X4, P2X7, A2A, and A3, as targets of endogenous ATP, ADP, or adenosine. ATP is sequentially degraded by the ecto-nucleotidases and 5′-nucleotidase enzymes to the almost inactive inosine as an end product. The appropriate selective agonists/antagonists for purinergic receptors as well as the respective enzyme inhibitors may profoundly interfere with microglial functions and reconstitute the homeostasis of the CNS disturbed by neuroinflammation.
Collapse
|
26
|
Zhang X, Wang D, Zhang B, Zhu J, Zhou Z, Cui L. Regulation of microglia by glutamate and its signal pathway in neurodegenerative diseases. Drug Discov Today 2020; 25:1074-1085. [PMID: 32320851 DOI: 10.1016/j.drudis.2020.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 03/10/2020] [Accepted: 04/03/2020] [Indexed: 02/06/2023]
Abstract
Microglia are an essential component of the central nervous system (CNS) and are involved in the primary response to microorganisms, neuroinflammation, homeostasis, and tissue regeneration, as well as contributing to the pathogenesis of neurodegenerative diseases. Research has shown that microglial diversity, multifunctionality, and their relationship with glutamate are crucial to determining their roles in these diseases. In this review, we focus on recent progress in determining microglial characteristics and the role of glutamate and its receptors in microglia regulation, which could be a novel therapeutic strategy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Xinyue Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China; Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrics, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden.
| | - Dan Wang
- Department of Ophthalmology, the First Hospital of Jilin University, Changchun, China.
| | - Bo Zhang
- Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrics, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden; Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China.
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China; Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrics, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden.
| | - Zhulin Zhou
- Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrics, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden.
| | - Li Cui
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
27
|
Behavioral responses of mGluR3-KO mice to the lipopolysaccharide-induced innate inflammatory reaction. Pharmacol Biochem Behav 2020; 190:172852. [DOI: 10.1016/j.pbb.2020.172852] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/09/2020] [Accepted: 01/12/2020] [Indexed: 12/13/2022]
|
28
|
Serum xanthurenic acid levels: Reduced in subjects at ultra high risk for psychosis. Schizophr Res 2019; 208:465-466. [PMID: 30837204 DOI: 10.1016/j.schres.2019.02.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 11/22/2022]
|
29
|
Pereira V, Goudet C. Emerging Trends in Pain Modulation by Metabotropic Glutamate Receptors. Front Mol Neurosci 2019; 11:464. [PMID: 30662395 PMCID: PMC6328474 DOI: 10.3389/fnmol.2018.00464] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022] Open
Abstract
Pain is an essential protective mechanism meant to prevent tissue damages in organisms. On the other hand, chronic or persistent pain caused, for example, by inflammation or nerve injury is long lasting and responsible for long-term disability in patients. Therefore, chronic pain and its management represents a major public health problem. Hence, it is critical to better understand chronic pain molecular mechanisms to develop innovative and efficient drugs. Over the past decades, accumulating evidence has demonstrated a pivotal role of glutamate in pain sensation and transmission, supporting glutamate receptors as promising potential targets for pain relieving drug development. Glutamate is the most abundant excitatory neurotransmitter in the brain. Once released into the synapse, glutamate acts through ionotropic glutamate receptors (iGluRs), which are ligand-gated ion channels triggering fast excitatory neurotransmission, and metabotropic glutamate receptors (mGluRs), which are G protein-coupled receptors modulating synaptic transmission. Eight mGluRs subtypes have been identified and are divided into three classes based on their sequence similarities and their pharmacological and biochemical properties. Of note, all mGluR subtypes (except mGlu6 receptor) are expressed within the nociceptive pathways where they modulate pain transmission. This review will address the role of mGluRs in acute and persistent pain processing and emerging pharmacotherapies for pain management.
Collapse
Affiliation(s)
- Vanessa Pereira
- IGF, CNRS, INSERM, Univ. de Montpellier, Montpellier, France
| | - Cyril Goudet
- IGF, CNRS, INSERM, Univ. de Montpellier, Montpellier, France
| |
Collapse
|
30
|
Nicoletti F, Orlando R, Di Menna L, Cannella M, Notartomaso S, Mascio G, Iacovelli L, Matrisciano F, Fazio F, Caraci F, Copani A, Battaglia G, Bruno V. Targeting mGlu Receptors for Optimization of Antipsychotic Activity and Disease-Modifying Effect in Schizophrenia. Front Psychiatry 2019; 10:49. [PMID: 30890967 PMCID: PMC6413697 DOI: 10.3389/fpsyt.2019.00049] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/23/2019] [Indexed: 01/03/2023] Open
Abstract
Metabotropic glutamate (mGlu) receptors are considered as candidate drug targets for the treatment of schizophrenia. These receptors form a family of eight subtypes (mGlu1 to -8), of which mGlu1 and -5 are coupled to Gq/11, and all other subtypes are coupled to Gi/o. Here, we discuss the possibility that selective ligands of individual mGlu receptor subtypes may be effective in controlling the core symptoms of schizophrenia, and, in some cases, may impact mechanisms underlying the progression of the disorder. Recent evidence indicates that activation of mGlu1 receptors inhibits dopamine release in the meso-striatal system. Hence, selective positive allosteric modulators (PAMs) of mGlu1 receptors hold promise for the treatment of positive symptoms of schizophrenia. mGlu5 receptors are widely expressed in the CNS and regulate the activity of cells that are involved in the pathophysiology of schizophrenia, such as cortical GABAergic interneurons and microglial cells. mGlu5 receptor PAMs are under development for the treatment of schizophrenia and cater the potential to act as disease modifiers by restraining neuroinflammation. mGlu2 receptors have attracted considerable interest because they negatively modulate 5-HT2A serotonin receptor signaling in the cerebral cortex. Both mGlu2 receptor PAMs and orthosteric mGlu2/3 receptor agonists display antipsychotic-like activity in animal models, and the latter drugs are inactive in mice lacking mGlu2 receptors. So far, mGlu3 receptors have been left apart as drug targets for schizophrenia. However, activation of mGlu3 receptors boosts mGlu5 receptor signaling, supports neuronal survival, and drives microglial cells toward an antiinflammatory phenotype. This strongly encourages research of mGlu3 receptors in schizophrenia. Finally, preclical studies suggest that mGlu4 receptors might be targeted by novel antipsychotic drugs, whereas studies of mGlu7 and mGlu8 receptors in animal models of psychosis are still at their infancy.
Collapse
Affiliation(s)
- Ferdinando Nicoletti
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Rosamaria Orlando
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | - Luisa Iacovelli
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Francesco Matrisciano
- Department of Psychiatry, The Psychiatric Institute, College of Medicine, University of Illinois, Chicago, IL, United States
| | | | - Filippo Caraci
- Department of Drug Sciences, University of Catania, Catania, Italy.,Oasi Research Institute (IRCCS), Troina, Italy
| | - Agata Copani
- Department of Drug Sciences, University of Catania, Catania, Italy.,Institute of Biostructure and Bioimaging, National Research Council, Catania, Italy
| | | | - Valeria Bruno
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
31
|
Presta I, Vismara M, Novellino F, Donato A, Zaffino P, Scali E, Pirrone KC, Spadea MF, Malara N, Donato G. Innate Immunity Cells and the Neurovascular Unit. Int J Mol Sci 2018; 19:E3856. [PMID: 30513991 PMCID: PMC6321635 DOI: 10.3390/ijms19123856] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/26/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022] Open
Abstract
Recent studies have clarified many still unknown aspects related to innate immunity and the blood-brain barrier relationship. They have also confirmed the close links between effector immune system cells, such as granulocytes, macrophages, microglia, natural killer cells and mast cells, and barrier functionality. The latter, in turn, is able to influence not only the entry of the cells of the immune system into the nervous tissue, but also their own activation. Interestingly, these two components and their interactions play a role of great importance not only in infectious diseases, but in almost all the pathologies of the central nervous system. In this paper, we review the main aspects in the field of vascular diseases (cerebral ischemia), of primitive and secondary neoplasms of Central Nervous System CNS, of CNS infectious diseases, of most common neurodegenerative diseases, in epilepsy and in demyelinating diseases (multiple sclerosis). Neuroinflammation phenomena are constantly present in all diseases; in every different pathological state, a variety of innate immunity cells responds to specific stimuli, differentiating their action, which can influence the blood-brain barrier permeability. This, in turn, undergoes anatomical and functional modifications, allowing the stabilization or the progression of the pathological processes.
Collapse
Affiliation(s)
- Ivan Presta
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Marco Vismara
- Department of Cell Biotechnologies and Hematology, University "La Sapienza" of Rome, 00185 Rome, Italy.
| | - Fabiana Novellino
- Institute of Molecular Bioimaging and Physiology, National Research Council, 88100 Catanzaro, Italy.
| | - Annalidia Donato
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Paolo Zaffino
- Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Elisabetta Scali
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Krizia Caterina Pirrone
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Maria Francesca Spadea
- Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Natalia Malara
- Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Giuseppe Donato
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| |
Collapse
|
32
|
Spampinato SF, Copani A, Nicoletti F, Sortino MA, Caraci F. Metabotropic Glutamate Receptors in Glial Cells: A New Potential Target for Neuroprotection? Front Mol Neurosci 2018; 11:414. [PMID: 30483053 PMCID: PMC6243036 DOI: 10.3389/fnmol.2018.00414] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 10/25/2018] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative disorders are characterized by excitotoxicity and neuroinflammation that finally lead to slow neuronal degeneration and death. Although neurons are the principal target, glial cells are important players as they contribute by either exacerbating or dampening the events that lead to neuroinflammation and neuronal damage. A dysfunction of the glutamatergic system is a common event in the pathophysiology of these diseases. Metabotropic glutamate (mGlu) receptors belong to a large family of G protein-coupled receptors largely expressed in neurons as well as in glial cells. They often appear overexpressed in areas involved in neurodegeneration, where they can modulate glutamatergic transmission. Of note, mGlu receptor upregulation may involve microglia or, even more frequently, astrocytes, where their activation causes release of factors potentially able to influence neuronal death. The expression of mGlu receptors has been also reported on oligodendrocytes, a glial cell type specifically involved in the development of multiple sclerosis. Here we will provide a general overview on the possible involvement of mGlu receptors expressed on glial cells in the pathogenesis of different neurodegenerative disorders and the potential use of subtype-selective mGlu receptor ligands as candidate drugs for the treatment of neurodegenerative disorders. Negative allosteric modulators (NAM) of mGlu5 receptors might represent a relevant pharmacological tool to develop new neuroprotective strategies in these diseases. Recent evidence suggests that targeting astrocytes and microglia with positive allosteric modulators (PAM) of mGlu3 receptor or oligodendrocytes with mGlu4 PAMS might represent novel pharmacological approaches for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Agata Copani
- Department of Drug Sciences, University of Catania, Catania, Italy.,Institute of Biostructure and Bioimaging, National Research Council, Catania, Italy
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neuromed, Istituto di Ricovero e Cura a Carattere Scientifico, Pozzilli, Italy
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Filippo Caraci
- Department of Drug Sciences, University of Catania, Catania, Italy.,Oasi Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Troina, Italy
| |
Collapse
|
33
|
Szepesi Z, Manouchehrian O, Bachiller S, Deierborg T. Bidirectional Microglia-Neuron Communication in Health and Disease. Front Cell Neurosci 2018; 12:323. [PMID: 30319362 PMCID: PMC6170615 DOI: 10.3389/fncel.2018.00323] [Citation(s) in RCA: 307] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022] Open
Abstract
Microglia are ramified cells that exhibit highly motile processes, which continuously survey the brain parenchyma and react to any insult to the CNS homeostasis. Although microglia have long been recognized as a crucial player in generating and maintaining inflammatory responses in the CNS, now it has become clear, that their function are much more diverse, particularly in the healthy brain. The innate immune response and phagocytosis represent only a little segment of microglia functional repertoire that also includes maintenance of biochemical homeostasis, neuronal circuit maturation during development and experience-dependent remodeling of neuronal circuits in the adult brain. Being equipped by numerous receptors and cell surface molecules microglia can perform bidirectional interactions with other cell types in the CNS. There is accumulating evidence showing that neurons inform microglia about their status and thus are capable of controlling microglial activation and motility while microglia also modulate neuronal activities. This review addresses the topic: how microglia communicate with other cell types in the brain, including fractalkine signaling, secreted soluble factors and extracellular vesicles. We summarize the current state of knowledge of physiological role and function of microglia during brain development and in the mature brain and further highlight microglial contribution to brain pathologies such as Alzheimer’s and Parkinson’s disease, brain ischemia, traumatic brain injury, brain tumor as well as neuropsychiatric diseases (depression, bipolar disorder, and schizophrenia).
Collapse
Affiliation(s)
- Zsuzsanna Szepesi
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Oscar Manouchehrian
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Sara Bachiller
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
34
|
Joffe ME, Centanni SW, Jaramillo AA, Winder DG, Conn PJ. Metabotropic Glutamate Receptors in Alcohol Use Disorder: Physiology, Plasticity, and Promising Pharmacotherapies. ACS Chem Neurosci 2018; 9:2188-2204. [PMID: 29792024 PMCID: PMC6192262 DOI: 10.1021/acschemneuro.8b00200] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Developing efficacious treatments for alcohol use disorder (AUD) has proven difficult. The insidious nature of the disease necessitates a deep understanding of its underlying biology as well as innovative approaches to ameliorate ethanol-related pathophysiology. Excessive ethanol seeking and relapse are generated by long-term changes to membrane properties, synaptic physiology, and plasticity throughout the limbic system and associated brain structures. Each of these factors can be modulated by metabotropic glutamate (mGlu) receptors, a diverse set of G protein-coupled receptors highly expressed throughout the central nervous system. Here, we discuss how different components of the mGlu receptor family modulate neurotransmission in the limbic system and other brain regions involved in AUD etiology. We then describe how these processes are dysregulated following ethanol exposure and speculate about how mGlu receptor modulation might restore such pathophysiological changes. To that end, we detail the current understanding of the behavioral pharmacology of mGlu receptor-directed drug-like molecules in animal models of AUD. Together, this review highlights the prominent position of the mGlu receptor system in the pathophysiology of AUD and provides encouragement that several classes of mGlu receptor modulators may be translated as viable treatment options.
Collapse
Affiliation(s)
- Max E. Joffe
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
| | - Samuel W. Centanni
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Anel A. Jaramillo
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Danny G. Winder
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee 37232-0697, United States
| |
Collapse
|