1
|
Liss A, Siddiqi MT, Marsland P, Varodayan FP. Neuroimmune regulation of the prefrontal cortex tetrapartite synapse. Neuropharmacology 2025; 269:110335. [PMID: 39904409 DOI: 10.1016/j.neuropharm.2025.110335] [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: 11/14/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
The prefrontal cortex (PFC) is an essential driver of cognitive, affective, and motivational behavior. There is clear evidence that the neuroimmune system directly influences PFC synapses, in addition to its role as the first line of defense against toxins and pathogens. In this review, we first describe the core structures that form the tetrapartite PFC synapse, focusing on the signaling microdomain created by astrocytic cradling of the synapse as well as the emerging role of the extracellular matrix in synaptic organization and plasticity. Neuroimmune signals (e.g. pro-inflammatory interleukin 1β) can impact the function of each core structure within the tetrapartite synapse, as well as promote intra-synaptic crosstalk, and we will provide an overview of recent advances in this field. Finally, evidence from post mortem human brain tissue and preclinical studies indicate that inflammation may be a key contributor to PFC dysfunction. Therefore, we conclude with a mechanistic discussion of neuroimmune-mediated maladaptive plasticity in neuropsychiatric disorders, with a focus on alcohol use disorder (AUD). Growing recognition of the neuroimmune system's role as a critical regulator of the PFC tetrapartite synapse provides strong support for targeting the neuroimmune system to develop new pharmacotherapeutics.
Collapse
Affiliation(s)
- Andrea Liss
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Mahum T Siddiqi
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Paige Marsland
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Florence P Varodayan
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA.
| |
Collapse
|
2
|
Kastner-Blasczyk AR, Hester SW, Reasons SE, Scofield MD, Woodward JJ. Effect of an astrocyte calcium exporter on orbitofrontal cortex neuron excitability, astrocyte-synaptic interaction, and alcohol consumption. Neuropharmacology 2025; 269:110365. [PMID: 39952350 PMCID: PMC11995387 DOI: 10.1016/j.neuropharm.2025.110365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/30/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Previous electrophysiology studies show that acute ethanol inhibits firing of orbitofrontal (OFC) cortex neurons while chronic intermittent ethanol (CIE) exposure increases firing accompanied by enhanced ethanol drinking. The acute ethanol inhibition of OFC neuronal firing is mediated by inhibitory glycine receptors and is reduced by expressing a plasma membrane calcium ATPase (PMCA) in OFC astrocytes. In this study, we tested the effects of astrocyte PMCA on CIE-induced increases in excitability and alcohol consumption and the physical interaction between OFC astrocytes and neurons. CIE increased neuronal firing in male mice as compared to Air controls while PMCA itself increased firing in Air control male mice. In contrast, PMCA reduced CIE-mediated hyperexcitability of firing in females. CIE did not affect OFC astrocyte size in control or PMCA male mice but increased astrocyte size in female mice. Similar to spiking, PMCA and CIE both increased the number of GluA1 containing synapses within the vicinity of virally labeled astrocytes in male mice but had differential effects in females. The astrocytic interaction with GluA1 labeled synapses was not affected by CIE treatment in male or female control mice, but there was a treatment-dependent effect of PMCA in male mice. CIE increased alcohol consumption in control but not PMCA male mice and had no effect on drinking in female mice. Lastly, OFC astrocyte PMCA expression had no effect on behavioral measures of locomotion, anxiety, spontaneous alternation, or spatial memory. These findings reveal important sex-dependent differences in the physiological, structural and behavioral actions of OFC astrocytes.
Collapse
Affiliation(s)
- A R Kastner-Blasczyk
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - S W Hester
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - S E Reasons
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - M D Scofield
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - J J Woodward
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
3
|
Peyton L, Haroon H, Umpierre A, Essa H, Bruce R, Wu LJ, Choi DS. In vivo calcium extrusion from accumbal astrocytes reduces anxiety-like behaviors but increases compulsive-like responses and compulsive ethanol drinking in mice. Neuropharmacology 2025; 268:110320. [PMID: 39842625 PMCID: PMC11830519 DOI: 10.1016/j.neuropharm.2025.110320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
The ventral striatum is crucially involved in reward processing. The present study investigates the behavioral effects of astrocyte-specific calcium extrusion virus "CalEx" on perseverative responses in the operant five-choice serial reaction time task and ethanol-conditioned place preference. Mice were injected with CalEx via the GfaABC1D promoter to extrude cytosolic calcium from astrocytes within the ventral striatum. We found that CalEx transfection in the ventral striatum reduced evoked response duration, the maximum amplitude, and the response frequency to 500 μM ATP as measured by ΔF/F fluorescence intensity of the genetically encoded calcium indicator targeting astrocytes GCaMP6f. During the five-choice serial reaction time task, CalEx mice persisted in perseverative responses compared to their counterparts. Additionally, during ethanol-conditioned place preference, CalEx mice showed increased place preference for a low ethanol concentration compared to control group. Furthermore, we found that accumbal astrocytic calcium extrusion increased quinine adulterated ethanol drinking. Our findings suggest that diminishing ventral striatum astrocyte calcium activity contributes to compulsive behaviors, ethanol drinking, and enhanced ethanol drug reward.
Collapse
Affiliation(s)
- Lee Peyton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Humza Haroon
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | | | - Hesham Essa
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Robert Bruce
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA; Neuroscience Program, Mayo Clinic College of Medicine and Science, MN, 55905, USA; Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA; Neuroscience Program, Mayo Clinic College of Medicine and Science, MN, 55905, USA; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, 59905, USA.
| |
Collapse
|
4
|
D'Sa K, Choi ML, Wagen AZ, Setó-Salvia N, Kopach O, Evans JR, Rodrigues M, Lopez-Garcia P, Lachica J, Clarke BE, Singh J, Ghareeb A, Bayne J, Grant-Peters M, Garcia-Ruiz S, Chen Z, Rodriques S, Athauda D, Gustavsson EK, Gagliano Taliun SA, Toomey C, Reynolds RH, Young G, Strohbuecker S, Warner T, Rusakov DA, Patani R, Bryant C, Klenerman DA, Gandhi S, Ryten M. Astrocytic RNA editing regulates the host immune response to alpha-synuclein. SCIENCE ADVANCES 2025; 11:eadp8504. [PMID: 40215316 PMCID: PMC11988446 DOI: 10.1126/sciadv.adp8504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 03/07/2025] [Indexed: 04/14/2025]
Abstract
RNA editing is a posttranscriptional mechanism that targets changes in RNA transcripts to modulate innate immune responses. We report the role of astrocyte-specific, ADAR1-mediated RNA editing in neuroinflammation in Parkinson's disease (PD). We generated human induced pluripotent stem cell-derived astrocytes, neurons and cocultures and exposed them to small soluble alpha-synuclein aggregates. Oligomeric alpha-synuclein triggered an inflammatory glial state associated with Toll-like receptor activation, viral responses, and cytokine secretion. This reactive state resulted in loss of neurosupportive functions and the induction of neuronal toxicity. Notably, interferon response pathways were activated leading to up-regulation and isoform switching of the RNA deaminase enzyme, ADAR1. ADAR1 mediates A-to-I RNA editing, and increases in RNA editing were observed in inflammatory pathways in cells, as well as in postmortem human PD brain. Aberrant, or dysregulated, ADAR1 responses and RNA editing may lead to sustained inflammatory reactive states in astrocytes triggered by alpha-synuclein aggregation, and this may drive the neuroinflammatory cascade in Parkinson's.
Collapse
Affiliation(s)
- Karishma D'Sa
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Minee L. Choi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Brain & Cognitive Sciences, KAIST, 921 Dehak-ro, Daejeon, Republic of Korea
| | - Aaron Z. Wagen
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Núria Setó-Salvia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Olga Kopach
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Neuroscience and Cell Biology Research Institute, City St George’s, University of London, Cranmer Terrace, London SW17 0RE, UK
| | - James R. Evans
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Margarida Rodrigues
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute at The University of Cambridge, Cambridge CB2 0AH, UK
| | - Patricia Lopez-Garcia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Joanne Lachica
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Benjamin E. Clarke
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Jaijeet Singh
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ali Ghareeb
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Applied Biotechnology Lab, The Francis Crick Institute, London NW1 1AT, UK
| | - James Bayne
- Applied Biotechnology Lab, The Francis Crick Institute, London NW1 1AT, UK
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Melissa Grant-Peters
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Sonia Garcia-Ruiz
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Zhongbo Chen
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Samuel Rodriques
- Applied Biotechnology Lab, The Francis Crick Institute, London NW1 1AT, UK
- FutureHouse, 1405 Minnesota Street, San Francisco, CA 94107, USA
| | - Dilan Athauda
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Emil K. Gustavsson
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Sarah A. Gagliano Taliun
- Montréal Heart Institute, Montréal, QC, Canada
- Department of Medicine and Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Christina Toomey
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Regina H. Reynolds
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - George Young
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- MRC Laboratory of Medical Sciences, London W12 0HS, UK
| | - Stephanie Strohbuecker
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Thomas Warner
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Dmitri A. Rusakov
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Rickie Patani
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Clare Bryant
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | - David A. Klenerman
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute at The University of Cambridge, Cambridge CB2 0AH, UK
| | - Sonia Gandhi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Mina Ryten
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UK Dementia Research Institute at The University of Cambridge, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| |
Collapse
|
5
|
Nentwig TB, Obray JD, Kruyer A, Wilkes ET, Vaughan DT, Scofield MD, Chandler LJ. Central amygdala astrocyte plasticity underlies GABAergic dysregulation in ethanol dependence. Transl Psychiatry 2025; 15:132. [PMID: 40199844 PMCID: PMC11978928 DOI: 10.1038/s41398-025-03337-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/21/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025] Open
Abstract
Dependence is a hallmark of alcohol use disorder characterized by excessive alcohol intake and withdrawal symptoms. The central nucleus of the amygdala (CeA) is a key brain structure underlying the synaptic and behavioral consequences of ethanol dependence. While accumulating evidence suggests that astrocytes regulate synaptic transmission and behavior, there is a limited understanding of the role astrocytes play in ethanol dependence. The present study used a combination of viral labeling, super resolution confocal microscopy, 3D image analysis, and slice electrophysiology to determine the effects of chronic intermittent ethanol (CIE) exposure on astrocyte plasticity in the CeA. During withdrawal from CIE exposure, we observed increased GABA transmission, an upregulation in astrocytic GAT3 levels, and an increased proximity of astrocyte processes near CeA synapses. Furthermore, GAT3 levels and synaptic proximity were positively associated with voluntary ethanol drinking in dependent rats. Slice electrophysiology confirmed that the upregulation in astrocytic GAT3 levels was functional, as CIE exposure unmasked a GAT3-sensitive tonic GABA current in the CeA. A causal role for astrocytic GAT3 in ethanol dependence was assessed using viral-mediated GAT3 overexpression and knockdown approaches. However, GAT3 knockdown or overexpression had no effect on somatic withdrawal symptoms, dependence-escalated ethanol intake, aversion-resistant drinking, or post-dependent ethanol drinking in male or female rats. Moreover, intra-CeA pharmacological inhibition of GAT3 did not alter dependent ethanol drinking. Together, these findings indicate that ethanol dependence induces GABAergic dysregulation and astrocyte plasticity in the CeA. However, these changes in astrocytic GAT3 do not appear to be necessary for the drinking related phenotypes associated with dependence.
Collapse
Affiliation(s)
- Todd B Nentwig
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - J Daniel Obray
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - Erik T Wilkes
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Dylan T Vaughan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - L Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
6
|
Díaz-Ubilla M, Figueroa-Valdés AI, Tobar HE, Quintanilla ME, Díaz E, Morales P, Berríos-Cárcamo P, Santapau D, Gallardo J, de Gregorio C, Ugalde J, Rojas C, Gonzalez-Madrid A, Ezquer M, Israel Y, Alcayaga-Miranda F, Ezquer F. Gut Microbiota-Derived Extracellular Vesicles Influence Alcohol Intake Preferences in Rats. J Extracell Vesicles 2025; 14:e70059. [PMID: 40098337 PMCID: PMC11913890 DOI: 10.1002/jev2.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 01/03/2025] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Growing preclinical and clinical evidence suggests a link between gut microbiota dysbiosis and problematic alcohol consumption. Extracellular vesicles (EVs) are key mediators involved in bacteria-to-host communication. However, their potential role in mediating addictive behaviour remains unexplored. This study investigates the role of gut microbiota-derived bacterial extracellular vesicles (bEVs) in driving high alcohol consumption. bEVs were isolated from the gut microbiota of a high alcohol-drinking rat strain (UChB rats), either ethanol-naïve or following chronic alcohol consumption and administered intraperitoneally or orally to alcohol-rejecting male and female Wistar rats. Both types of UChB-derived bEVs increased Wistar's voluntary alcohol consumption (three bottle choice test) up to 10-fold (p < 0.0001), indicating that bEVs are able and sufficient to transmit drinking behaviour across different rat strains. Molecular analysis revealed that bEVs administration did not induce systemic or brain inflammation in the recipient animals, suggesting that the increased alcohol intake triggered by UChB-derived bEVs operates through an inflammation-independent mechanism. Furthermore, we demonstrate that the vagus nerve mediates the bEV-induced increase in alcohol consumption, as bilateral vagotomy completely abolished the high drinking behaviour induced by both intraperitoneally injected and orally administered bEVs. Thus, this study identifies bEVs as a novel mechanism underlying gut microbiota-induced high alcohol intake in a vagus nerve-dependent manner.
Collapse
Affiliation(s)
- Macarena Díaz-Ubilla
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Aliosha I Figueroa-Valdés
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Santiago, Chile
| | - Hugo E Tobar
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Santiago, Chile
| | - María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Specialized Center for Prevention of Substance Use and Treatment of Addictions (CESA), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Eugenio Díaz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Specialized Center for Prevention of Substance Use and Treatment of Addictions (CESA), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Pablo Berríos-Cárcamo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Daniela Santapau
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Javiera Gallardo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Cristian de Gregorio
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Juan Ugalde
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Carolina Rojas
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- Faculty of Dentistry, Universidad de Los Andes, Santiago, Chile
| | - Antonia Gonzalez-Madrid
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Marcelo Ezquer
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Specialized Center for Prevention of Substance Use and Treatment of Addictions (CESA), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Francisca Alcayaga-Miranda
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Santiago, Chile
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago, Chile
| |
Collapse
|
7
|
Warden AS, Salem NA, Brenner E, Sutherland GT, Stevens J, Kapoor M, Goate AM, Mayfield RD. Integrative genomics approach identifies glial transcriptomic dysregulation and risk in the cortex of individuals with Alcohol Use Disorder. Biol Psychiatry 2025:S0006-3223(25)00994-1. [PMID: 40024496 DOI: 10.1016/j.biopsych.2025.02.895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/24/2025] [Accepted: 02/14/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Alcohol use disorder (AUD) is a prevalent neuropsychiatric disorder that is a major global health concern, affecting millions of people worldwide. Past studies of AUD used underpowered single cell analysis or bulk homogenates of postmortem brain tissue, which obscures gene expression changes in specific cell types. Therefore, we sought to conduct the largest-to-date single-nuclei RNAseq (snRNA-seq) postmortem brain study in AUD to elucidate transcriptomic pathology with cell type-specific resolution. METHODS Here we performed snRNA-seq and high dimensional network analysis of 73 post-mortem samples from individuals with AUD (N=36, Nnuclei= 248,873) and neurotypical controls (N=37, Nnuclei= 210,573) in the dorsolateral prefrontal cortex from both male and female donors. Additionally, we performed analysis for cell type-specific enrichment of aggregate genetic risk for AUD as well as integration of the AUD proteome for secondary validation. RESULTS We identified 32 distinct cell clusters and found widespread cell type-specific transcriptomic changes across the cortex in AUD, particularly affecting glial populations. We found the greatest dysregulation in novel microglial and astrocytic subtypes that accounted for the majority of differential gene expression and co-expression modules linked to AUD. Differential gene expression was secondarily validated by integration of a publicly available AUD proteome. Finally, analysis for aggregate genetic risk for AUD identified subtypes of glia as potential key players not only affected by but causally linked to the progression of AUD. CONCLUSIONS These results highlight the importance of cell type-specific molecular changes in AUD and offer opportunities to identify novel targets for treatment on the single-nuclei level.
Collapse
Affiliation(s)
- Anna S Warden
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Nihal A Salem
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA; Institute for Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Eric Brenner
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Greg T Sutherland
- School of Medical Sciences and Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Julia Stevens
- New South Wales Brain Tissue Resource Centre, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Manav Kapoor
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Alison M Goate
- Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, 1425 Madison Ave, New York, NY, 10029, USA; Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mt. Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA; Institute for Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
8
|
Singh S, Kannan M, Oladapo A, Deshetty UM, Ray S, Buch S, Periyasamy P. Ethanol modulates astrocyte activation and neuroinflammation via miR-339/NLRP6 inflammasome signaling. Free Radic Biol Med 2025; 226:1-12. [PMID: 39522566 DOI: 10.1016/j.freeradbiomed.2024.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/22/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Alcohol is the most abused substance among adolescents and has a profound impact on health, society, and the economy. Alcohol intoxication is linked to neuroinflammation and neuronal damage, which result in behavioral alterations such as motor dysfunction, neuronal injury, cognitive deficits, and inflammation. Alcohol-induced neuroinflammation is associated with the activation of central nervous system cells, including astrocytes, and the release of proinflammatory cytokines. In this study, we investigated the role of the NLRP6 inflammasome signaling pathway in inducing cellular activation and neuroinflammation in human primary astrocytes exposed to ethanol. Our results demonstrated that ethanol upregulates the expression of NLRP6 inflammasome signaling mediators, including NLRP6, caspase 1, and proinflammatory cytokines IL-1β and IL-18, in human primary astrocytes. Gene silencing studies using NLRP6 siRNA further validate ethanol-mediated activation of NLRP6, cleavage of caspase 1, IL-1β, and IL-18 in human primary astrocytes. miR array analysis of ethanol-exposed human primary astrocytes reveals decreased levels of miR-339, accompanied by an upregulation of NLRP6 inflammasome signaling and astrocyte activation. Through bioinformatics analyses, Argonaute immunoprecipitation assays, and miR-339 overexpression experiments, we identify NLRP6 as a novel 3'-UTR target of miR-339. Overall, our findings confirmed the involvement of miR-339 in NLRP6 inflammasome signaling and its association with cellular activation and neuroinflammation in human primary astrocytes exposed to ethanol and provide novel insights highlighting a previously unrecognized mechanism in alcohol-induced neuroinflammation.
Collapse
Affiliation(s)
- Seema Singh
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Muthukumar Kannan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Abiola Oladapo
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Uma Maheswari Deshetty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Sudipta Ray
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA.
| |
Collapse
|
9
|
Mignogna KM, Tatom Z, Macleod L, Sergi Z, Nguyen A, Michenkova M, Smith ML, Miles MF. Identification of novel genetic loci and candidate genes for progressive ethanol consumption in diversity outbred mice. Neuropsychopharmacology 2024; 49:1892-1904. [PMID: 38951586 PMCID: PMC11473901 DOI: 10.1038/s41386-024-01902-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/26/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024]
Abstract
Mouse behavioral genetic mapping studies can identify genomic intervals modulating complex traits under well-controlled environmental conditions and have been used to study ethanol behaviors to aid in understanding genetic risk and the neurobiology of alcohol use disorder (AUD). However, historically such studies have produced large confidence intervals, thus complicating identification of potential causal candidate genes. Diversity Outbred (DO) mice offer the ability to perform high-resolution quantitative trait loci (QTL) mapping on a very genetically diverse background, thus facilitating identification of candidate genes. Here, we studied a population of 636 male DO mice with four weeks of intermittent ethanol access via a three-bottle choice procedure, producing a progressive ethanol consumption phenotype. QTL analysis identified 3 significant (Chrs 3, 4, and 12) and 13 suggestive loci for ethanol-drinking behaviors with narrow confidence intervals (1-4 Mbp for significant QTLs). Results suggested that genetic influences on initial versus progressive ethanol consumption were localized to different genomic intervals. A defined set of positional candidate genes were prioritized using haplotype analysis, identified coding polymorphisms, prefrontal cortex transcriptomics data, human GWAS data and prior rodent gene set data for ethanol or other misused substances. These candidates included Car8, the lone gene with a significant cis-eQTL within a Chr 4 QTL for week four ethanol consumption. These results represent the highest-resolution genetic mapping of ethanol consumption behaviors in mice to date, providing identification of novel loci and candidate genes for study in relation to the neurobiology of AUD.
Collapse
Affiliation(s)
- Kristin M Mignogna
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Zachary Tatom
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Lorna Macleod
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Zachary Sergi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Angel Nguyen
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Marie Michenkova
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Maren L Smith
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael F Miles
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Neurology, Virginia Commonwealth University, Richmond, VA, USA.
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
10
|
Warden AS, Salem NA, Brenner E, Sutherland GT, Stevens J, Kapoor M, Goate AM, Dayne Mayfield R. Integrative genomics approach identifies glial transcriptomic dysregulation and risk in the cortex of individuals with Alcohol Use Disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.607185. [PMID: 39211266 PMCID: PMC11360965 DOI: 10.1101/2024.08.16.607185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Alcohol use disorder (AUD) is a prevalent neuropsychiatric disorder that is a major global health concern, affecting millions of people worldwide. Past molecular studies of AUD used underpowered single cell analysis or bulk homogenates of postmortem brain tissue, which obscures gene expression changes in specific cell types. Here we performed single nuclei RNA-sequencing analysis of 73 post-mortem samples from individuals with AUD (N=36, N nuclei = 248,873) and neurotypical controls (N=37, N nuclei = 210,573) in both sexes across two institutional sites. We identified 32 clusters and found widespread cell type-specific transcriptomic changes across the cortex in AUD, particularly affecting glia. We found the greatest dysregulation in novel microglial and astrocytic subtypes that accounted for the majority of differential gene expression and co-expression modules linked to AUD. Analysis for cell type-specific enrichment of aggregate genetic risk for AUD identified subtypes of microglia and astrocytes as potential key players not only affected by but causally linked to the progression of AUD. These results highlight the importance of cell-type specific molecular changes in AUD and offer opportunities to identify novel targets for treatment.
Collapse
|
11
|
Tavakoli NS, Malone SG, Anderson TL, Neeley RE, Asadipooya A, Bardo MT, Ortinski PI. Astrocyte Ca 2+ in the dorsal striatum suppresses neuronal activity to oppose cue-induced reinstatement of cocaine seeking. Front Cell Neurosci 2024; 18:1347491. [PMID: 39280793 PMCID: PMC11393831 DOI: 10.3389/fncel.2024.1347491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 08/12/2024] [Indexed: 09/18/2024] Open
Abstract
Recent literature supports a prominent role for astrocytes in regulation of drug-seeking behaviors. The dorsal striatum, specifically, is known to play a role in reward processing with neuronal activity that can be influenced by astrocyte Ca2+. However, the manner in which Ca2+ in dorsal striatum astrocytes impacts neuronal signaling after exposure to self-administered cocaine remains unclear. We addressed this question following over-expression of the Ca2+ extrusion pump, hPMCA2w/b, in dorsal striatum astrocytes and the Ca2+ indicator, GCaMP6f, in dorsal striatum neurons of rats that were trained to self-administer cocaine. Following extinction of cocaine-seeking behavior, the rats over-expressing hMPCA2w/b showed a significant increase in cue-induced reinstatement of cocaine seeking. Suppression of astrocyte Ca2+ increased the amplitude of neuronal Ca2+ transients in brain slices, but only after cocaine self-administration. This was accompanied by decreased duration of neuronal Ca2+ events in the cocaine group and no changes in Ca2+ event frequency. Acute administration of cocaine to brain slices decreased amplitude of neuronal Ca2+ in both the control and cocaine self-administration groups regardless of hPMCA2w/b expression. These results indicated that astrocyte Ca2+ control over neuronal Ca2+ transients was enhanced by cocaine self-administration experience, although sensitivity to acutely applied cocaine remained comparable across all groups. To explore this further, we found that neither the hMPCA2w/b expression nor the cocaine self-administration experience altered regulation of neuronal Ca2+ events by NPS-2143, a Ca2+ sensing receptor (CaSR) antagonist, suggesting that plasticity of neuronal signaling after hPMCA2w/b over-expression was unlikely to result from elevated extracellular Ca2+. We conclude that astrocyte Ca2+ in the dorsal striatum impacts neurons via cell-intrinsic mechanisms (e.g., gliotransmission, metabolic coupling, etc.) and impacts long-term neuronal plasticity after cocaine self-administration differently from neuronal response to acute cocaine. Overall, astrocyte Ca2+ influences neuronal output in the dorsal striatum to promote resistance to cue-induced reinstatement of cocaine seeking.
Collapse
Affiliation(s)
- Navid S Tavakoli
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Samantha G Malone
- Department of Psychology, University of Kentucky, Lexington, KY, United States
| | - Tanner L Anderson
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Ryson E Neeley
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Artin Asadipooya
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Michael T Bardo
- Department of Psychology, University of Kentucky, Lexington, KY, United States
| | - Pavel I Ortinski
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
12
|
Costa PA, Everett NA, Turner AJ, Umpierrez LS, Baracz SJ, Cornish JL. Adolescent alcohol binge drinking and withdrawal: behavioural, brain GFAP-positive astrocytes and acute methamphetamine effects in adult female rats. Psychopharmacology (Berl) 2024; 241:1539-1554. [PMID: 38705893 PMCID: PMC11269403 DOI: 10.1007/s00213-024-06580-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/25/2024] [Indexed: 05/07/2024]
Abstract
RATIONALE Alcopop beverages are generally the first alcoholic beverage that young females drink which contain high levels of sugar and alcohol. The over-consumption of these drinks may encourage alcohol co-administration with methamphetamine (METH) impacting on drinking behaviour and glial function. AIMS The aims of this study were to evaluate the effect of adolescent binge alcohol exposure on consumption level, anxiety-like behaviour, cross-sensitization with METH and on astrocyte expression in reward related brain regions. METHODS Adolescent female Sprague-Dawley rats had daily 1-hour oral alcohol consumption of alcopop (ALCP; with sucrose) or ethanol-only (ETOH; without sucrose), transitioned from 5 to 15% (v/v) ethanol content for 34 days. Water and sucrose groups act as controls. During alcohol withdrawal, rats were tested for anxiety on the elevated plus maze (EPM) and locomotor activity following saline or METH (1 mg/kg i.p) treatment. Brains were then collected to assess astrocyte immunofluorescence for glial fibrillary acidic protein (GFAP) in reward-related brain regions. RESULTS Rats pretreated with 5% ALCP consumed significantly more volume and ethanol intake when compared to 5% EtOH rats. Both ALCP and EtOH groups had a higher preference ratio for 5% than 15% alcohol solutions and ALCP rats had greater ethanol intake at 15% than EtOH rats. Alcohol withdrawal showed no significant differences between groups on anxiety, METH cross-sensitization effects or GFAP intensity in the regions studied. CONCLUSIONS Overall, the addition of sucrose to alcoholic solutions encouraged female rats to consume larger volumes and greater ethanol intake compared to ethanol-only solutions, yet did not have long lasting effects on behaviour and astrocytes.
Collapse
Affiliation(s)
- Priscila A Costa
- Behavioural Neuropharmacology Laboratory, School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia.
| | - Nicholas A Everett
- Behavioural Neuropharmacology Laboratory, School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Anita J Turner
- Behavioural Neuropharmacology Laboratory, School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Laísa S Umpierrez
- Behavioural Neuropharmacology Laboratory, School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Sarah J Baracz
- Behavioural Neuropharmacology Laboratory, School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia
| | - Jennifer L Cornish
- Behavioural Neuropharmacology Laboratory, School of Psychological Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, 2109, Australia.
| |
Collapse
|
13
|
Nentwig TB, Obray JD, Kruyer A, Wilkes ET, Vaughan DT, Scofield MD, Chandler LJ. Central Amygdala Astrocyte Plasticity Underlies GABAergic Dysregulation in Ethanol Dependence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598470. [PMID: 38915577 PMCID: PMC11195260 DOI: 10.1101/2024.06.11.598470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Dependence is a hallmark of alcohol use disorder characterized by excessive alcohol intake and withdrawal symptoms. The central nucleus of the amygdala (CeA) is a key brain structure underlying the synaptic and behavioral consequences of ethanol dependence. While accumulating evidence suggests that astrocytes regulate synaptic transmission and behavior, there is a limited understanding of the role astrocytes play in ethanol dependence. The present study used a combination of viral labeling, super resolution confocal microscopy, 3D image analysis, and slice electrophysiology to determine the effects of chronic intermittent ethanol (CIE) exposure on astrocyte plasticity in the CeA. During withdrawal from CIE exposure, we observed increased GABA transmission, an upregulation in astrocytic GAT3 levels, and an increased proximity of astrocyte processes near CeA synapses. Furthermore, GAT3 levels and synaptic proximity were positively associated with voluntary ethanol drinking in dependent rats. Slice electrophysiology confirmed that the upregulation in astrocytic GAT3 levels was functional, as CIE exposure unmasked a GAT3-sensitive tonic GABA current in the CeA. A causal role for astrocytic GAT3 in ethanol dependence was assessed using viral-mediated GAT3 overexpression and knockdown approaches. However, GAT3 knockdown or overexpression had no effect on somatic withdrawal symptoms, dependence-escalated ethanol intake, aversion-resistant drinking, or post-dependent ethanol drinking in male or female rats. Moreover, intra-CeA pharmacological inhibition of GAT3 also did not alter dependent ethanol drinking. Together, these findings indicate that ethanol dependence induces GABAergic dysregulation and astrocyte plasticity in the CeA. However, astrocytic GAT3 does not appear necessary for the drinking related phenotypes associated with dependence.
Collapse
Affiliation(s)
- Todd B. Nentwig
- Department of Neuroscience, Medical University of South Carolina, Charleston SC 29425, United States
| | - J. Daniel Obray
- Department of Neuroscience, Medical University of South Carolina, Charleston SC 29425, United States
| | - Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston SC 29425, United States
- Current affiliation: Department of Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - Erik T Wilkes
- Department of Neuroscience, Medical University of South Carolina, Charleston SC 29425, United States
| | - Dylan T. Vaughan
- Department of Neuroscience, Medical University of South Carolina, Charleston SC 29425, United States
- Current affiliation: Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael D. Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston SC 29425, United States
| | - L. Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, Charleston SC 29425, United States
| |
Collapse
|
14
|
Bansal Y, Codeluppi SA, Banasr M. Astroglial Dysfunctions in Mood Disorders and Rodent Stress Models: Consequences on Behavior and Potential as Treatment Target. Int J Mol Sci 2024; 25:6357. [PMID: 38928062 PMCID: PMC11204179 DOI: 10.3390/ijms25126357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
Astrocyte dysfunctions have been consistently observed in patients affected with depression and other psychiatric illnesses. Although over the years our understanding of these changes, their origin, and their consequences on behavior and neuronal function has deepened, many aspects of the role of astroglial dysfunction in major depressive disorder (MDD) and post-traumatic stress disorder (PTSD) remain unknown. In this review, we summarize the known astroglial dysfunctions associated with MDD and PTSD, highlight the impact of chronic stress on specific astroglial functions, and how astroglial dysfunctions are implicated in the expression of depressive- and anxiety-like behaviors, focusing on behavioral consequences of astroglial manipulation on emotion-related and fear-learning behaviors. We also offer a glance at potential astroglial functions that can be targeted for potential antidepressant treatment.
Collapse
Affiliation(s)
- Yashika Bansal
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada
| | - Sierra A. Codeluppi
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5G 2C8, Canada
| | - Mounira Banasr
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5G 2C8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M2J 4A6, Canada
| |
Collapse
|
15
|
Holt LM, Nestler EJ. Astrocytic transcriptional and epigenetic mechanisms of drug addiction. J Neural Transm (Vienna) 2024; 131:409-424. [PMID: 37940687 PMCID: PMC11066772 DOI: 10.1007/s00702-023-02716-4] [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: 08/21/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
Addiction is a leading cause of disease burden worldwide and remains a challenge in current neuroscience research. Drug-induced lasting changes in gene expression are mediated by transcriptional and epigenetic regulation in the brain and are thought to underlie behavioral adaptations. Emerging evidence implicates astrocytes in regulating drug-seeking behaviors and demonstrates robust transcriptional response to several substances of abuse. This review focuses on the astrocytic transcriptional and epigenetic mechanisms of drug action.
Collapse
Affiliation(s)
- Leanne M Holt
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
16
|
Wulaer B, Holtz MA, Nagai J. Homeostasis to Allostasis: Prefrontal Astrocyte Roles in Cognitive Flexibility and Stress Biology. ADVANCES IN NEUROBIOLOGY 2024; 39:137-163. [PMID: 39190074 DOI: 10.1007/978-3-031-64839-7_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
In the intricate landscape of neurophysiology, astrocytes have been traditionally cast as homeostatic cells; however, their mechanistic involvement in allostasis-particularly how they modulate the adaptive response to stress and its accumulative impact that disrupts cognitive functions and precipitates psychiatric disorders-is now starting to be unraveled. Here, we address the gap by positing astrocytes as crucial allostatic players whose molecular adaptations underlie cognitive flexibility in stress-related neuropsychiatric conditions. We review how astrocytes, responding to stress mediators such as glucocorticoid and epinephrine/norepinephrine, undergo morphological and functional transformations that parallel the maladaptive changes. Our synthesis of recent findings reveals that these glial changes, especially in the metabolically demanding prefrontal cortex, may underlie some of the neuropsychiatric mechanisms characterized by the disruption of energy metabolism and astrocytic networks, compromised glutamate clearance, and diminished synaptic support. We argue that astrocytes extend beyond their homeostatic role, actively participating in the brain's allostatic response, especially by modulating energy substrates critical for cognitive functions.
Collapse
Affiliation(s)
- Bolati Wulaer
- Laboratory for Glia-Neuron Circuit Dynamics, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Mika A Holtz
- Laboratory for Glia-Neuron Circuit Dynamics, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Jun Nagai
- Laboratory for Glia-Neuron Circuit Dynamics, RIKEN Center for Brain Science, Wako, Saitama, Japan.
| |
Collapse
|
17
|
Salem NA, Manzano L, Keist MW, Ponomareva O, Roberts AJ, Roberto M, Mayfield RD. Cell-type brain-region specific changes in prefrontal cortex of a mouse model of alcohol dependence. Neurobiol Dis 2024; 190:106361. [PMID: 37992784 PMCID: PMC10874299 DOI: 10.1016/j.nbd.2023.106361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/31/2023] [Accepted: 11/18/2023] [Indexed: 11/24/2023] Open
Abstract
The prefrontal cortex is a crucial regulator of alcohol drinking, and dependence, and other behavioral phenotypes associated with AUD. Comprehensive identification of cell-type specific transcriptomic changes in alcohol dependence will improve our understanding of mechanisms underlying the excessive alcohol use associated with alcohol dependence and will refine targets for therapeutic development. We performed single nucleus RNA sequencing (snRNA-seq) and Visium spatial gene expression profiling on the medial prefrontal cortex (mPFC) obtained from C57BL/6 J mice exposed to the two-bottle choice-chronic intermittent ethanol (CIE) vapor exposure (2BC-CIE, defined as dependent group) paradigm which models phenotypes of alcohol dependence including escalation of alcohol drinking. Gene co-expression network analysis and differential expression analysis identified highly dysregulated co-expression networks in multiple cell types. Dysregulated modules and their hub genes suggest novel understudied targets for studying molecular mechanisms contributing to the alcohol dependence state. A subtype of inhibitory neurons was the most alcohol-sensitive cell type and contained a downregulated gene co-expression module; the hub gene for this module is Cpa6, a gene previously identified by GWAS to be associated with excessive alcohol consumption. We identified an astrocytic Gpc5 module significantly upregulated in the alcohol-dependent group. To our knowledge, there are no studies linking Cpa6 and Gpc5 to the alcohol-dependent phenotype. We also identified neuroinflammation related gene expression changes in multiple cell types, specifically enriched in microglia, further implicating neuroinflammation in the escalation of alcohol drinking. Here, we present a comprehensive atlas of cell-type specific alcohol dependence mediated gene expression changes in the mPFC and identify novel cell type-specific targets implicated in alcohol dependence.
Collapse
Affiliation(s)
- Nihal A Salem
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Lawrence Manzano
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - Michael W Keist
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - Olga Ponomareva
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - Amanda J Roberts
- Animal Models Core Facility, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Marisa Roberto
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
18
|
Harder EV, Franklin JP, VanRyzin JW, Reissner KJ. Astrocyte-Neuron Interactions in Substance Use Disorders. ADVANCES IN NEUROBIOLOGY 2024; 39:165-191. [PMID: 39190075 DOI: 10.1007/978-3-031-64839-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Engagement of astrocytes within the brain's reward circuitry has been apparent for approximately 30 years, when noncontingent drug administration was observed to lead to cytological markers of reactive astrocytes. Since that time, advanced approaches in rodent behavior and astrocyte monitoring have revealed complex interactions between astrocytes with drug type, animal sex, brain region, and dose and duration of drug administration. A number of studies now collectively reveal that rodent drug self-administration followed by prolonged abstinence results in decreased features of structure and synaptic colocalization of astrocytes. In addition, stimulation of astrocytes in the nucleus accumbens with DREADD receptors or pharmacological compounds opposes drug-seeking behavior. These findings provide a clear path for ongoing investigation into astrocytes as mediators of drug action in the brain and underscore the potential therapeutic utility of astrocytes in the regulation of drug craving and relapse vulnerability.
Collapse
Affiliation(s)
- Eden V Harder
- Department of Psychology & Neuroscience, Neuroscience Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Janay P Franklin
- Department of Psychology & Neuroscience, Neuroscience Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Jonathan W VanRyzin
- Department of Psychology & Neuroscience, Neuroscience Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Kathryn J Reissner
- Department of Psychology & Neuroscience, Neuroscience Center, UNC Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
19
|
Padilla-Valdez MM, Díaz-Iñiguez MI, Ortuño-Sahagún D, Rojas-Mayorquín AE. Neuroinflammation in fetal alcohol spectrum disorders and related novel therapeutic approaches. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166854. [PMID: 37611676 DOI: 10.1016/j.bbadis.2023.166854] [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: 04/03/2023] [Revised: 07/21/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
Fetal alcohol spectrum disorders (FASD) is an umbrella term to describe the neurological effects of prenatal alcohol exposure (PAE). It has been extensively characterized that PAE causes cell proliferation disruption, heterotopias, and malformations in various brain regions and there is increasing evidence that neuroinflammation is responsible for some of these neurotoxic effects. Despite evidence of its importance, neuroinflammation is not usually considered at diagnosis or treatment for FASD. Here, we discuss the literature regarding anti- inflammatory drugs and nutraceuticals, which hold promise for future therapeutical interventions in these disorders.
Collapse
Affiliation(s)
- Mayra Madeleine Padilla-Valdez
- Departamento de Ciencias Ambientales, Universidad de Guadalajara, Centro Universitario de Ciencias Biológicas y Agropecuarias, Guadalajara 45200, Mexico; Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, C.P 44340 Guadalajara, JAL, Mexico
| | - María Isabel Díaz-Iñiguez
- Departamento de Ciencias Ambientales, Universidad de Guadalajara, Centro Universitario de Ciencias Biológicas y Agropecuarias, Guadalajara 45200, Mexico; Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, C.P 44340 Guadalajara, JAL, Mexico
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, C.P 44340 Guadalajara, JAL, Mexico.
| | - Argelia Esperanza Rojas-Mayorquín
- Departamento de Ciencias Ambientales, Universidad de Guadalajara, Centro Universitario de Ciencias Biológicas y Agropecuarias, Guadalajara 45200, Mexico.
| |
Collapse
|
20
|
Coulter OR, Walker CD, Risher ML. Astrocyte-specific Ca 2+ activity: Mechanisms of action, experimental tools, and roles in ethanol-induced dysfunction. Biochem Cell Biol 2023; 101:410-421. [PMID: 36989534 DOI: 10.1139/bcb-2023-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
Astrocytes are a subtype of non-neuronal glial cells that reside in the central nervous system. Astrocytes have extensive peripheral astrocytic processes that ensheathe synapses to form the tripartite synapse. Through a multitude of pathways, astrocytes can influence synaptic development and structural maturation, respond to neuronal signals, and modulate synaptic transmission. Over the last decade, strong evidence has emerged demonstrating that astrocytes can influence behavioral outcomes in various animal models of cognition. However, the full extent of how astrocytes influence brain function is still being revealed. Astrocyte calcium (Ca2+) signaling has emerged as an important driver of astrocyte-neuronal communication allowing intricate crosstalk through mechanisms that are still not fully understood. Here, we will review the field's current understanding of astrocyte Ca2+ signaling and discuss the sophisticated state-of-the-art tools and approaches used to continue unraveling astrocytes' interesting role in brain function. Using the field of pre-clinical ethanol (EtOH) studies in the context of alcohol use disorder, we focus on how these novel approaches have helped to reveal an important role for astrocyte Ca2+ function in regulating EtOH consumption and how astrocyte Ca2+ dysfunction contributes to the cognitive deficits that emerge after EtOH exposure in a rodent model.
Collapse
Affiliation(s)
- O R Coulter
- Department of Biomedical Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - C D Walker
- Department of Biomedical Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
- Neurobiology Research Laboratory, Hershel 'Woody' Williams Veterans Affairs Medical Center, Huntington, WV 25704, USA
| | - M-L Risher
- Department of Biomedical Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
- Neurobiology Research Laboratory, Hershel 'Woody' Williams Veterans Affairs Medical Center, Huntington, WV 25704, USA
| |
Collapse
|
21
|
Kang S, Hong SI, Kang S, Song M, Yang MA, Essa H, Baker M, Lee J, Bruce RA, Lee SW, Choi DS. Astrocyte activities in the external globus pallidus regulate action-selection strategies in reward-seeking behaviors. SCIENCE ADVANCES 2023; 9:eadh9239. [PMID: 37327345 PMCID: PMC10275597 DOI: 10.1126/sciadv.adh9239] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/12/2023] [Indexed: 06/18/2023]
Abstract
An imbalance in goal-directed and habitual behavioral control is a hallmark of decision-making-related disorders, including addiction. Although external globus pallidus (GPe) is critical for action selection, which harbors enriched astrocytes, the role of GPe astrocytes involved in action-selection strategies remained unknown. Using in vivo calcium signaling with fiber photometry, we found substantially attenuated GPe astrocytic activity during habitual learning compared to goal-directed learning. The support vector machine analysis predicted the behavioral outcomes. Chemogenetic activation of the astrocytes or inhibition of GPe pan-neuronal activities facilitates the transition from habit to goal-directed reward-seeking behavior. Next, we found increased astrocyte-specific GABA (γ-aminobutyric acid) transporter type 3 (GAT3) messenger RNA expression during habit learning. Notably, the pharmacological inhibition of GAT3 occluded astrocyte activation-induced transition from habitual to goal-directed behavior. On the other hand, attentional stimuli shifted the habit to goal-directed behaviors. Our findings suggest that the GPe astrocytes regulate the action selection strategy and behavioral flexibility.
Collapse
Affiliation(s)
- Shinwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Sa-Ik Hong
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Seungwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Minryung Song
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Minsu Abel Yang
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Program of Brain and Cognitive Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hesham Essa
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Matthew Baker
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Jeyeon Lee
- Department of Radiology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Robert A. Bruce
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Sang Wan Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Kim Jaechul Graduate School of AI, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| |
Collapse
|
22
|
Gruol DL, Calderon D, French K, Melkonian C, Huitron-Resendiz S, Cates-Gatto C, Roberts AJ. Neuroimmune interactions with binge alcohol drinking in the cerebellum of IL-6 transgenic mice. Neuropharmacology 2023; 228:109455. [PMID: 36775097 PMCID: PMC10029700 DOI: 10.1016/j.neuropharm.2023.109455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 01/20/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023]
Abstract
The neuroimmune system of the brain, which is comprised primarily of astrocytes and microglia, regulates a variety of homeostatic mechanisms that underlie normal brain function. Numerous conditions, including alcohol consumption, can disrupt this regulatory process by altering brain levels of neuroimmune factors. Alcohol and neuroimmune factors, such as proinflammatory cytokines IL-6 and TNF-alpha, act at similar targets in the brain, including excitatory and inhibitory synaptic transmission. Thus, alcohol-induced production of IL-6 and/or TNF-alpha could be important contributing factors to the effects of alcohol on the brain. Recent studies indicate that IL-6 plays a role in alcohol drinking and the effects of alcohol on the brain activity following the cessation of alcohol consumption (post-alcohol period), however information on these topics is limited. Here we used homozygous and heterozygous female and male transgenic mice with increased astrocyte expression of IL-6 to examined further the interactions between alcohol and IL-6 with respect to voluntary alcohol drinking, brain activity during the post-alcohol period, IL-6 signal transduction, and expression of synaptic proteins. Wildtype littermates (WT) served as controls. The transgenic mice model brain neuroimmune status with respect to IL-6 in subjects with a history of persistent alcohol use. Results showed a genotype dependent reduction in voluntary alcohol consumption in the Drinking in the Dark protocol and in frequency-dependent relationships between brain activity in EEG recordings during the post-alcohol period and alcohol consumption. IL-6, TNF-alpha, IL-6 signal transduction partners pSTAT3 and c/EBP beta, and synaptic proteins were shown to play a role in these genotypic effects.
Collapse
Affiliation(s)
- Donna L Gruol
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| | - Delilah Calderon
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Katharine French
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Claudia Melkonian
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | | | - Chelsea Cates-Gatto
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Amanda J Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
23
|
Munier J, Shen S, Rahal D, Hanna A, Marty V, O'Neill P, Fanselow M, Spigelman I. Chronic intermittent ethanol exposure disrupts stress-related tripartite communication to impact affect-related behavioral selection in male rats. Neurobiol Stress 2023; 24:100539. [PMID: 37131490 PMCID: PMC10149313 DOI: 10.1016/j.ynstr.2023.100539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/12/2023] [Accepted: 04/16/2023] [Indexed: 05/04/2023] Open
Abstract
Alcohol use disorder (AUD) is characterized by loss of intake control, increased anxiety, and susceptibility to relapse inducing stressors. Both astrocytes and neurons contribute to behavioral and hormonal consequences of chronic intermittent ethanol (CIE) exposure in animal models. Details on how CIE disrupts hypothalamic neuro-glial communication, which mediates stress responses are lacking. We conducted a behavioral battery (grooming, open field, reactivity to a single, uncued foot-shock, intermittent-access two-bottle choice ethanol drinking) followed by Ca2+ imaging in ex-vivo slices of paraventricular nucleus of the hypothalamus (PVN) from male rats exposed to CIE vapor or air-exposed controls. Ca2+ signals were evaluated in response to norepinephrine (NE) with or without selective α-adrenergic receptor (αAR) or GluN2B-containing N-methyl-D-aspartate receptor (NMDAR) antagonists, followed by dexamethasone (DEX) to mock a pharmacological stress response. Expectedly, CIE rats had altered anxiety-like, rearing, grooming, and drinking behaviors. Importantly, NE-mediated reductions in Ca2+ event frequency were blunted in both CIE neurons and astrocytes. Administration of the selective α1AR antagonist, prazosin, reversed this CIE-induced dysfunction in both cell types. Additionally, the pharmacological stress protocol reversed the altered basal Ca2+ signaling profile of CIE astrocytes. Signaling changes in astrocytes in response to NE were correlated with anxiety-like behaviors, such as the grooming:rearing ratio, suggesting tripartite synaptic function plays a role in switching between exploratory and stress-coping behavior. These data show how CIE exposure causes persistent changes to PVN neuro-glial function and provides the groundwork for how these physiological changes manifest in behavioral selection.
Collapse
Affiliation(s)
- J.J. Munier
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
- Corresponding author.
| | - S. Shen
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
| | - D. Rahal
- Edna Bennett Pierce Prevention Research Center, The Pennsylvania State University, United States
| | - A. Hanna
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
| | - V.N. Marty
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
| | - P.R. O'Neill
- Hatos Center for Neuropharmacology, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, UCLA, United States
| | - M.S. Fanselow
- Department of Psychology, College of Life Sciences, Department of Psychiatry & Biobehavioral Science, David Geffen School of Medicine, UCLA, United States
| | - I. Spigelman
- Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, United States
- Corresponding author. Laboratory of Neuropharmacology, Section of Biosystems & Function, School of Dentistry, UCLA, 10833 Le Conte Avenue, 63-078 CHS, Los Angeles, CA, 90095-1668, United States.
| |
Collapse
|
24
|
Wang F, Ruppell KT, Zhou S, Qu Y, Gong J, Shang Y, Wu J, Liu X, Diao W, Li Y, Xiang Y. Gliotransmission and adenosine signaling promote axon regeneration. Dev Cell 2023; 58:660-676.e7. [PMID: 37028426 PMCID: PMC10173126 DOI: 10.1016/j.devcel.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 11/18/2022] [Accepted: 03/08/2023] [Indexed: 04/08/2023]
Abstract
How glia control axon regeneration remains incompletely understood. Here, we investigate glial regulation of regenerative ability differences of closely related Drosophila larval sensory neuron subtypes. Axotomy elicits Ca2+ signals in ensheathing glia, which activates regenerative neurons through the gliotransmitter adenosine and mounts axon regenerative programs. However, non-regenerative neurons do not respond to glial stimulation or adenosine. Such neuronal subtype-specific responses result from specific expressions of adenosine receptors in regenerative neurons. Disrupting gliotransmission impedes axon regeneration of regenerative neurons, and ectopic adenosine receptor expression in non-regenerative neurons suffices to activate regenerative programs and induce axon regeneration. Furthermore, stimulating gliotransmission or activating the mammalian ortholog of Drosophila adenosine receptors in retinal ganglion cells (RGCs) promotes axon regrowth after optic nerve crush in adult mice. Altogether, our findings demonstrate that gliotransmission orchestrates neuronal subtype-specific axon regeneration in Drosophila and suggest that targeting gliotransmission or adenosine signaling is a strategy for mammalian central nervous system repair.
Collapse
Affiliation(s)
- Fei Wang
- Department of Neurobiology, Program of Neuroscience, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Kendra Takle Ruppell
- Department of Neurobiology, Program of Neuroscience, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Songlin Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Yun Qu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jiaxin Gong
- Department of Neurobiology, Program of Neuroscience, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Ye Shang
- Department of Neurobiology, Program of Neuroscience, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Jinglin Wu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xin Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Wenlin Diao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yi Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China; The National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China.
| | - Yang Xiang
- Department of Neurobiology, Program of Neuroscience, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA.
| |
Collapse
|
25
|
Shigetomi E, Koizumi S. The role of astrocytes in behaviors related to emotion and motivation. Neurosci Res 2023; 187:21-39. [PMID: 36181908 DOI: 10.1016/j.neures.2022.09.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 10/14/2022]
Abstract
Astrocytes are present throughout the brain and intimately interact with neurons and blood vessels. Three decades of research have shown that astrocytes reciprocally communicate with neurons and other non-neuronal cells in the brain and dynamically regulate cell function. Astrocytes express numerous receptors for neurotransmitters, neuromodulators, and cytokines and receive information from neurons, other astrocytes, and other non-neuronal cells. Among those receptors, the main focus has been G-protein coupled receptors. Activation of G-protein coupled receptors leads to dramatic changes in intracellular signaling (Ca2+ and cAMP), which is considered a form of astrocyte activity. Methodological improvements in measurement and manipulation of astrocytes have advanced our understanding of the role of astrocytes in circuits and have begun to reveal unexpected functions of astrocytes in behavior. Recent studies have suggested that astrocytic activity regulates behavior flexibility, such as coping strategies for stress exposure, and plays an important role in behaviors related to emotion and motivation. Preclinical evidence suggests that impairment of astrocytic function contributes to psychiatric diseases, especially major depression. Here, we review recent progress on the role of astrocytes in behaviors related to emotion and motivation.
Collapse
Affiliation(s)
- Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Japan; Yamanashi GLIA Center, Graduate School of Medical Science, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Japan.
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Japan; Yamanashi GLIA Center, Graduate School of Medical Science, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Japan.
| |
Collapse
|
26
|
Nuñez-delMoral A, Bianchi PC, Brocos-Mosquera I, Anesio A, Palombo P, Camarini R, Cruz FC, Callado LF, Vialou V, Erdozain AM. The Matricellular Protein Hevin Is Involved in Alcohol Use Disorder. Biomolecules 2023; 13:biom13020234. [PMID: 36830603 PMCID: PMC9953008 DOI: 10.3390/biom13020234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
Astrocytic-secreted matricellular proteins have been shown to influence various aspects of synaptic function. More recently, they have been found altered in animal models of psychiatric disorders such as drug addiction. Hevin (also known as Sparc-like 1) is a matricellular protein highly expressed in the adult brain that has been implicated in resilience to stress, suggesting a role in motivated behaviors. To address the possible role of hevin in drug addiction, we quantified its expression in human postmortem brains and in animal models of alcohol abuse. Hevin mRNA and protein expression were analyzed in the postmortem human brain of subjects with an antemortem diagnosis of alcohol use disorder (AUD, n = 25) and controls (n = 25). All the studied brain regions (prefrontal cortex, hippocampus, caudate nucleus and cerebellum) in AUD subjects showed an increase in hevin levels either at mRNA or/and protein levels. To test if this alteration was the result of alcohol exposure or indicative of a susceptibility factor to alcohol consumption, mice were exposed to different regimens of intraperitoneal alcohol administration. Hevin protein expression was increased in the nucleus accumbens after withdrawal followed by a ethanol challenge. The role of hevin in AUD was determined using an RNA interference strategy to downregulate hevin expression in nucleus accumbens astrocytes, which led to increased ethanol consumption. Additionally, ethanol challenge after withdrawal increased hevin levels in blood plasma. Altogether, these results support a novel role for hevin in the neurobiology of AUD.
Collapse
Affiliation(s)
- Amaia Nuñez-delMoral
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Paula C. Bianchi
- Department of Pharmacology, Universidade Federal de São Paulo-UNIFESP, São Paulo 04023-062, Brazil
| | - Iria Brocos-Mosquera
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain
| | - Augusto Anesio
- Department of Pharmacology, Universidade Federal de São Paulo-UNIFESP, São Paulo 04023-062, Brazil
| | - Paola Palombo
- Department of Pharmacology, Universidade Federal de São Paulo-UNIFESP, São Paulo 04023-062, Brazil
| | - Rosana Camarini
- Department of Pharmacology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | - Fabio C. Cruz
- Department of Pharmacology, Universidade Federal de São Paulo-UNIFESP, São Paulo 04023-062, Brazil
| | - Luis F. Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
- Biocruces-Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Vincent Vialou
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, Sorbonne Université, 75005 Paris, France
- Correspondence: (V.V.); (A.M.E.); Tel.: +33-1-44-27-60-98 (V.V.); +34-601-28-48 (A.M.E.)
| | - Amaia M. Erdozain
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
- Correspondence: (V.V.); (A.M.E.); Tel.: +33-1-44-27-60-98 (V.V.); +34-601-28-48 (A.M.E.)
| |
Collapse
|
27
|
Kruyer A, Kalivas PW, Scofield MD. Astrocyte regulation of synaptic signaling in psychiatric disorders. Neuropsychopharmacology 2023; 48:21-36. [PMID: 35577914 PMCID: PMC9700696 DOI: 10.1038/s41386-022-01338-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/27/2022] [Accepted: 05/01/2022] [Indexed: 02/07/2023]
Abstract
Over the last 15 years, the field of neuroscience has evolved toward recognizing the critical role of astroglia in shaping neuronal synaptic activity and along with the pre- and postsynapse is now considered an equal partner in tripartite synaptic transmission and plasticity. The relative youth of this recognition and a corresponding deficit in reagents and technologies for quantifying and manipulating astroglia relative to neurons continues to hamper advances in understanding tripartite synaptic physiology. Nonetheless, substantial advances have been made and are reviewed herein. We review the role of astroglia in synaptic function and regulation of behavior with an eye on how tripartite synapses figure into brain pathologies underlying behavioral impairments in psychiatric disorders, both from the perspective of measures in postmortem human brains and more subtle influences on tripartite synaptic regulation of behavior in animal models of psychiatric symptoms. Our goal is to provide the reader a well-referenced state-of-the-art understanding of current knowledge and predict what we may discover with deeper investigation of tripartite synapses using reagents and technologies not yet available.
Collapse
Affiliation(s)
- Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
- Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
28
|
Grantham EK, Barchiesi R, Salem NA, Mayfield RD. Neuroimmune pathways as targets to reduce alcohol consumption. Pharmacol Biochem Behav 2023; 222:173491. [PMID: 36400266 PMCID: PMC9906983 DOI: 10.1016/j.pbb.2022.173491] [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: 07/13/2022] [Revised: 10/04/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022]
Affiliation(s)
| | - Riccardo Barchiesi
- Waggoner Center for Alcohol and Addiction Research and the Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, United States.
| | | | | |
Collapse
|
29
|
Tremblay S, Zeng Y, Yue A, Chabot K, Mynahan A, Desrochers S, Bridges S, Ahmad ST. Caffeine Delays Ethanol-Induced Sedation in Drosophila. BIOLOGY 2022; 12:biology12010063. [PMID: 36671755 PMCID: PMC9855986 DOI: 10.3390/biology12010063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022]
Abstract
Caffeine and ethanol are among the most widely available and commonly consumed psychoactive substances. Both interact with adenosine receptor-mediated signaling which regulates numerous neurological processes including sleep and waking behaviors. In mammals, caffeine is an adenosine receptor antagonist and thus acts as a stimulant. Conversely, ethanol is a sedative because it promotes GABAergic neurotransmission, inhibits glutamatergic neurotransmission, and increases the amount of adenosine in the brain. Despite seemingly overlapping interactions, not much is known about the effect of caffeine on ethanol-induced sedation in Drosophila. In this study, using Drosophila melanogaster as a model, we show that caffeine supplementation in food delays the onset of ethanol-induced sedation in males and females of different strains. The resistance to sedation reverses upon caffeine withdrawal. Heterozygous adenosine receptor mutant flies are resistant to sedation. These findings suggest that caffeine and adenosine receptors modulate the sedative effects of ethanol in Drosophila.
Collapse
Affiliation(s)
- Sonia Tremblay
- Department of Biology, Colby College, Waterville, ME 04901, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80310, USA
| | - Yanqiqi Zeng
- Department of Biology, Colby College, Waterville, ME 04901, USA
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Aixin Yue
- Department of Biology, Colby College, Waterville, ME 04901, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Kiana Chabot
- Department of Biology, Colby College, Waterville, ME 04901, USA
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA
| | - Abigail Mynahan
- Department of Biology, Colby College, Waterville, ME 04901, USA
| | - Stephanie Desrochers
- Department of Biology, Colby College, Waterville, ME 04901, USA
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Sarra Bridges
- Department of Biology, Colby College, Waterville, ME 04901, USA
| | - S. Tariq Ahmad
- Department of Biology, Colby College, Waterville, ME 04901, USA
- Correspondence: ; Tel.: +1-207-859-5722
| |
Collapse
|
30
|
Kruyer A. Astrocyte Heterogeneity in Regulation of Synaptic Activity. Cells 2022; 11:cells11193135. [PMID: 36231097 PMCID: PMC9562199 DOI: 10.3390/cells11193135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/02/2022] [Accepted: 10/02/2022] [Indexed: 02/07/2023] Open
Abstract
Our awareness of the number of synapse regulatory functions performed by astroglia is rapidly expanding, raising interesting questions regarding astrocyte heterogeneity and specialization across brain regions. Whether all astrocytes are poised to signal in a multitude of ways, or are instead tuned to surrounding synapses and how astroglial signaling is altered in psychiatric and cognitive disorders are fundamental questions for the field. In recent years, molecular and morphological characterization of astroglial types has broadened our ability to design studies to better analyze and manipulate specific functions of astroglia. Recent data emerging from these studies will be discussed in depth in this review. I also highlight remaining questions emerging from new techniques recently applied toward understanding the roles of astrocytes in synapse regulation in the adult brain.
Collapse
Affiliation(s)
- Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
31
|
Walker CD, Sexton HG, Hyde J, Greene B, Risher ML. Diverging Effects of Adolescent Ethanol Exposure on Tripartite Synaptic Development across Prefrontal Cortex Subregions. Cells 2022; 11:3111. [PMID: 36231073 PMCID: PMC9561972 DOI: 10.3390/cells11193111] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 11/26/2022] Open
Abstract
Adolescence is a developmental period that encompasses, but is not limited to, puberty and continues into early adulthood. During this period, maturation and refinement are observed across brain regions such as the prefrontal cortex (PFC), which is critical for cognitive function. Adolescence is also a time when excessive alcohol consumption in the form of binge drinking peaks, increasing the risk of long-term cognitive deficits and the risk of developing an alcohol use disorder later in life. Animal models have revealed that adolescent ethanol (EtOH) exposure results in protracted disruption of neuronal function and performance on PFC-dependent tasks that require higher-order decision-making. However, the role of astrocytes in EtOH-induced disruption of prefrontal cortex-dependent function has yet to be elucidated. Astrocytes have complex morphologies with an extensive network of peripheral astrocyte processes (PAPs) that ensheathe pre- and postsynaptic terminals to form the 'tripartite synapse.' At the tripartite synapse, astrocytes play several critical roles, including synaptic maintenance, dendritic spine maturation, and neurotransmitter clearance through proximity-dependent interactions. Here, we investigate the effects of adolescent binge EtOH exposure on astrocyte morphology, PAP-synaptic proximity, synaptic stabilization proteins, and dendritic spine morphology in subregions of the PFC that are important in the emergence of higher cognitive function. We found that adolescent binge EtOH exposure resulted in subregion specific changes in astrocyte morphology and astrocyte-neuronal interactions. While this did not correspond to a loss of astrocytes, synapses, or dendritic spines, there was a corresponding region-specific and EtOH-dependent shift in dendritic spine phenotype. Lastly, we found that changes in astrocyte-neuronal interactions were not a consequence of changes in the expression of key synaptic structural proteins neurexin, neuroligin 1, or neuroligin 3. These data demonstrate that adolescent EtOH exposure results in enduring effects on neuron-glia interactions that persist into adulthood in a subregion-specific PFC manner, suggesting selective vulnerability. Further work is necessary to understand the functional and behavioral implications.
Collapse
Affiliation(s)
- Christopher Douglas Walker
- Department of Biomedical Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
- Neurobiology Research Laboratory, Hershel ‘Woody’ Williams Veterans Affairs Medical Center, Huntington, WV 25704, USA
| | - Hannah Gray Sexton
- Department of Biomedical Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
- Neurobiology Research Laboratory, Hershel ‘Woody’ Williams Veterans Affairs Medical Center, Huntington, WV 25704, USA
| | - Jentre Hyde
- Department of Biomedical Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Brittani Greene
- Department of Biomedical Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Mary-Louise Risher
- Department of Biomedical Research, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
- Neurobiology Research Laboratory, Hershel ‘Woody’ Williams Veterans Affairs Medical Center, Huntington, WV 25704, USA
| |
Collapse
|
32
|
Ortinski PI, Reissner KJ, Turner J, Anderson TA, Scimemi A. Control of complex behavior by astrocytes and microglia. Neurosci Biobehav Rev 2022; 137:104651. [PMID: 35367512 PMCID: PMC9119927 DOI: 10.1016/j.neubiorev.2022.104651] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/28/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023]
Abstract
Evidence that glial cells influence behavior has been gaining a steady foothold in scientific literature. Out of the five main subtypes of glial cells in the brain, astrocytes and microglia have received an outsized share of attention with regard to shaping a wide spectrum of behavioral phenomena and there is growing appreciation that the signals intrinsic to these cells as well as their interactions with surrounding neurons reflect behavioral history in a brain region-specific manner. Considerable regional diversity of glial cell phenotypes is beginning to be recognized and may contribute to behavioral outcomes arising from circuit-specific computations within and across discrete brain nuclei. Here, we summarize current knowledge on the impact of astrocyte and microglia activity on behavioral outcomes, with a specific focus on brain areas relevant to higher cognitive control, reward-seeking, and circadian regulation.
Collapse
Affiliation(s)
- P I Ortinski
- Department of Neuroscience, University of Kentucky, USA
| | - K J Reissner
- Department of Psychology and Neuroscience, University of North Carolina Chapel Hill, USA
| | - J Turner
- Department of Pharmaceutical Sciences, University of Kentucky, USA
| | - T A Anderson
- Department of Neuroscience, University of Kentucky, USA
| | - A Scimemi
- Department of Biology, State University of New York at Albany, USA
| |
Collapse
|
33
|
Villa-González M, Martín-López G, Pérez-Álvarez MJ. Dysregulation of mTOR Signaling after Brain Ischemia. Int J Mol Sci 2022; 23:ijms23052814. [PMID: 35269956 PMCID: PMC8911477 DOI: 10.3390/ijms23052814] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
In this review, we provide recent data on the role of mTOR kinase in the brain under physiological conditions and after damage, with a particular focus on cerebral ischemia. We cover the upstream and downstream pathways that regulate the activation state of mTOR complexes. Furthermore, we summarize recent advances in our understanding of mTORC1 and mTORC2 status in ischemia–hypoxia at tissue and cellular levels and analyze the existing evidence related to two types of neural cells, namely glia and neurons. Finally, we discuss the potential use of mTORC1 and mTORC2 as therapeutic targets after stroke.
Collapse
Affiliation(s)
- Mario Villa-González
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.V.-G.); (G.M.-L.)
- Centro de Biología Molecular “Severo Ochoa” (CBMSO), Universidad Autónoma de Madrid/CSIC, 28049 Madrid, Spain
| | - Gerardo Martín-López
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.V.-G.); (G.M.-L.)
| | - María José Pérez-Álvarez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.V.-G.); (G.M.-L.)
- Centro de Biología Molecular “Severo Ochoa” (CBMSO), Universidad Autónoma de Madrid/CSIC, 28049 Madrid, Spain
- Correspondence: ; Tel.: +34-91-497-2819
| |
Collapse
|
34
|
León BE, Kang S, Franca-Solomon G, Shang P, Choi DS. Alcohol-Induced Neuroinflammatory Response and Mitochondrial Dysfunction on Aging and Alzheimer's Disease. Front Behav Neurosci 2022; 15:778456. [PMID: 35221939 PMCID: PMC8866940 DOI: 10.3389/fnbeh.2021.778456] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/07/2021] [Indexed: 12/27/2022] Open
Abstract
Mitochondria are essential organelles central to various cellular functions such as energy production, metabolic pathways, signaling transduction, lipid biogenesis, and apoptosis. In the central nervous system, neurons depend on mitochondria for energy homeostasis to maintain optimal synaptic transmission and integrity. Deficiencies in mitochondrial function, including perturbations in energy homeostasis and mitochondrial dynamics, contribute to aging, and Alzheimer's disease. Chronic and heavy alcohol use is associated with accelerated brain aging, and increased risk for dementia, especially Alzheimer's disease. Furthermore, through neuroimmune responses, including pro-inflammatory cytokines, excessive alcohol use induces mitochondrial dysfunction. The direct and indirect alcohol-induced neuroimmune responses, including pro-inflammatory cytokines, are critical for the relationship between alcohol-induced mitochondrial dysfunction. In the brain, alcohol activates microglia and increases inflammatory mediators that can impair mitochondrial energy production, dynamics, and initiate cell death pathways. Also, alcohol-induced cytokines in the peripheral organs indirectly, but synergistically exacerbate alcohol's effects on brain function. This review will provide recent and advanced findings focusing on how alcohol alters the aging process and aggravates Alzheimer's disease with a focus on mitochondrial function. Finally, we will contextualize these findings to inform clinical and therapeutic approaches towards Alzheimer's disease.
Collapse
Affiliation(s)
- Brandon Emanuel León
- Regenerative Sciences Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Shinwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Gabriela Franca-Solomon
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Pei Shang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| |
Collapse
|
35
|
Lyon KA, Allen NJ. From Synapses to Circuits, Astrocytes Regulate Behavior. Front Neural Circuits 2022; 15:786293. [PMID: 35069124 PMCID: PMC8772456 DOI: 10.3389/fncir.2021.786293] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/05/2021] [Indexed: 12/21/2022] Open
Abstract
Astrocytes are non-neuronal cells that regulate synapses, neuronal circuits, and behavior. Astrocytes ensheath neuronal synapses to form the tripartite synapse where astrocytes influence synapse formation, function, and plasticity. Beyond the synapse, recent research has revealed that astrocyte influences on the nervous system extend to the modulation of neuronal circuitry and behavior. Here we review recent findings on the active role of astrocytes in behavioral modulation with a focus on in vivo studies, primarily in mice. Using tools to acutely manipulate astrocytes, such as optogenetics or chemogenetics, studies reviewed here have demonstrated a causal role for astrocytes in sleep, memory, sensorimotor behaviors, feeding, fear, anxiety, and cognitive processes like attention and behavioral flexibility. Current tools and future directions for astrocyte-specific manipulation, including methods for probing astrocyte heterogeneity, are discussed. Understanding the contribution of astrocytes to neuronal circuit activity and organismal behavior will be critical toward understanding how nervous system function gives rise to behavior.
Collapse
Affiliation(s)
- Krissy A Lyon
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Nicola J Allen
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
| |
Collapse
|
36
|
Miguel-Hidalgo JJ. Astroglia in the Vulnerability and Maintenance of Alcohol Use Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:255-279. [PMID: 34888838 DOI: 10.1007/978-3-030-77375-5_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Changes induced in the morphology and the multiplicity of functional roles played by astrocytes in brain regions critical to the establishment and maintenance of alcohol abuse suggest that they make an important contribution to the vulnerability to alcohol use disorders. The understanding of the relevant mechanisms accounting for that contribution is complicated by the fact that alcohol itself acts directly on astrocytes altering their metabolism, gene expression, and plasticity, so that the ultimate result is a complex interaction of various cellular pathways, including intracellular calcium regulation, neuroimmune responses, and regulation of neurotransmitter and gliotransmitter release and uptake. The recent years have seen a steady increase in the characterization of several of the relevant mechanisms, but much remains to be done for a full understanding of the astrocytes' contribution to the vulnerability to alcohol dependence and abuse and for using that knowledge in designing effective therapies for AUDs.
Collapse
Affiliation(s)
- José Javier Miguel-Hidalgo
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
37
|
O'Donovan B, Neugornet A, Neogi R, Xia M, Ortinski P. Cocaine experience induces functional adaptations in astrocytes: Implications for synaptic plasticity in the nucleus accumbens shell. Addict Biol 2021; 26:e13042. [PMID: 33864336 DOI: 10.1111/adb.13042] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 03/22/2021] [Accepted: 03/26/2021] [Indexed: 11/24/2022]
Abstract
Astrocytes have become established as an important regulator of neuronal activity in the brain. Accumulating literature demonstrates that cocaine self-administration in rodent models induces structural changes within astrocytes that may influence their interaction with the surrounding neurons. Here, we provide evidence that cocaine impacts astrocytes at the functional level and alters neuronal sensitivity to astrocyte-derived glutamate. We report that a 14-day period of short access to cocaine (2 h/day) decreases spontaneous astrocytic Ca2+ transients and precipitates changes in astrocyte network activity in the nucleus accumbens shell. This is accompanied by increased prevalence of slow inward currents, a physiological marker of neuronal activation by astrocytic glutamate, in a subset of medium spiny neurons. Within, but not outside, of this subset, we observe an increase in duration and frequency of N-methyl-D-aspartate (NMDA) receptor-mediated synaptic events. Additionally, we find that the link between synaptic NMDA receptor plasticity and neuron sensitivity to astrocytic glutamate is maintained independent of drug exposure and is observed in both cocaine and saline control animals. Imaging analyses of neuronal Ca2+ activity show no effect of cocaine self-administration on individual cells or on neuronal network activity in brain slices. Therefore, our data indicate that cocaine self-administration promotes astrocyte-specific functional changes that can be linked to increased glutamate-mediated coupling with principal neurons in the nucleus accumbens. Such coupling may be spatially restricted as it does not result in a broad impact on network structure of local neuronal circuits.
Collapse
Affiliation(s)
- Bernadette O'Donovan
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
| | - Austin Neugornet
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
| | - Richik Neogi
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
- Integrated Biomedical Sciences University of Kentucky Lexington Kentucky USA
| | - Mengfan Xia
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
| | - Pavel Ortinski
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
| |
Collapse
|
38
|
Hong SI, Kang S, Baker M, Choi DS. Astrocyte-neuron interaction in the dorsal striatum-pallidal circuits and alcohol-seeking behaviors. Neuropharmacology 2021; 198:108759. [PMID: 34433087 DOI: 10.1016/j.neuropharm.2021.108759] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/21/2021] [Accepted: 08/11/2021] [Indexed: 12/31/2022]
Abstract
In the striatum, two main types of GABAergic medium spiny neurons (MSNs), denoted striatonigral (or direct-pathway MSNs, dMSNs) and striatopallidal neurons (indirect-pathway MSNs, iMSNs), form circuits with distinct pallidal nuclei, which sends "GO" or "NO-GO" signals through the thalamus. These striatopallidal circuits evaluate and execute reward-seeking and taking behaviors. Especially, the dorsal striatum can be further divided into the dorsomedial striatum (DMS, equivalent to caudate in primates and humans) and dorsolateral striatum (DLS, equivalent to putamen), which orchestrates goal-directed and habitual reward-seeking and taking behaviors, respectively. Using optogenetics, chemogenetics and in vivo calcium imaging technologies combined with electrophysiology and digitalized behavior phenotyping, recent studies have revealed cell-, circuit- and context-specific functions of these microcircuits in addictive behaviors. Also, region-specific astrocytes regulate the homeostatic activities of the dMSNs and iMSNs as well as the downstream circuits, which determine the net balance of cortico-striato-pallidal activities to the thalamic neurons. This review will summarize the recent progress of striatopallidal circuits focusing on astrocyte-neuron interaction and, reward- and alcohol-seeking behaviors. Our review will also discuss the translational and clinical implications of these microcircuit studies. This article is part of the special Issue on "Neurocircuitry Modulating Drug and Alcohol Abuse".
Collapse
Affiliation(s)
- Sa-Ik Hong
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, 55905, USA
| | - Seungwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, 55905, USA
| | - Matthew Baker
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, 55905, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, 55905, USA; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA.
| |
Collapse
|
39
|
Meredith LR, Burnette EM, Grodin EN, Irwin MR, Ray LA. Immune treatments for alcohol use disorder: A translational framework. Brain Behav Immun 2021; 97:349-364. [PMID: 34343618 PMCID: PMC9044974 DOI: 10.1016/j.bbi.2021.07.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 07/10/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022] Open
Abstract
While the immune system is essential for survival, an excessive or prolonged inflammatory response, such as that resulting from sustained heavy alcohol use, can damage the host and contribute to psychiatric disorders. A growing body of literature indicates that the immune system plays a critical role in the development and maintenance of alcohol use disorder (AUD). As such, there is enthusiasm for treatments that can restore healthy levels of inflammation as a mechanism to reduce drinking and promote recovery. In this qualitative literature review, we provide a conceptual rationale for immune therapies and discuss progress in medications development for AUD focused on the immune system as a treatment target. This review is organized into sections based on primary signaling pathways targeted by the candidate therapies, namely: (a) toll-like receptors, (b) phosphodiesterase inhibitors, (c) peroxisome proliferator-activated receptors, (d) microglia and astrocytes, (e) other immune pharmacotherapies, and (f) behavioral therapies. As relevant within each section, we examine the basic biological mechanisms of each class of therapy and evaluate preclinical research testing the role of the therapy on mitigating alcohol-related behaviors in animal models. To the extent available, translational findings are reviewed with discussion of completed and ongoing randomized clinical trials and their findings to date. An applied and clinically focused approach is taken to identify the potential clinical applications of the various treatments reviewed. We conclude by delineating the most promising candidate treatments and discussing future directions by considering opportunities for immune treatment development and personalized medicine for AUD.
Collapse
Affiliation(s)
- Lindsay R Meredith
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Elizabeth M Burnette
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Erica N Grodin
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael R Irwin
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA; Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, CA, USA; Cousins Center for Psychoneuroimmunology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lara A Ray
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
40
|
Vetreno RP, Qin L, Coleman LG, Crews FT. Increased Toll-like Receptor-MyD88-NFκB-Proinflammatory neuroimmune signaling in the orbitofrontal cortex of humans with alcohol use disorder. Alcohol Clin Exp Res 2021; 45:1747-1761. [PMID: 34415075 PMCID: PMC8526379 DOI: 10.1111/acer.14669] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/02/2021] [Accepted: 07/07/2021] [Indexed: 01/27/2023]
Abstract
BACKGROUND Many brain disorders, including alcohol use disorder (AUD), are associated with induction of multiple proinflammatory genes. One aspect of proinflammatory signaling is progressive increases in expression across cells and induction of other innate immune genes. High-mobility group box 1 (HMGB1) heteromers contribute to amplification by potentiating multiple proinflammatory responses, including Toll-like receptors (TLRs). TLR signaling recruits coupling proteins linked to nuclear transcription factors that induce proinflammatory cytokines and chemokines and their respective receptors. We tested the hypothesis that AUD induction of TLR expression increases levels of proinflammatory genes and cellular signaling cascades in association with neurodegeneration in the orbitofrontal cortex (OFC). METHODS Postmortem human OFC tissue samples (n = 10) from males diagnosed with AUD were compared to age-matched moderate drinking controls (CON). Neuroimmune signaling molecules were assessed using immunohistochemistry for protein and reverse transcription polymerase chain reaction for messenger RNA (mRNA). RESULTS In the AUD OFC, we report induction of the endogenous TLR agonist HMGB1 as well as all TLRs assessed (i.e., TLR2-TLR9) except TLR1. This was accompanied by increased expression of the TLR adaptor protein myeloid differentiation primary response 88 (MyD88), activation of the proinflammatory nuclear transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB), and downstream induction of proinflammatory cytokines, chemokines, and their corresponding receptors. Several of these proinflammatory signaling markers are expressed in glia and neurons. The induction of HMGB1-TLR-MyD88-NFκB proinflammatory signaling pathways correlates with neurodegeneration (i.e., Fluoro-Jade B), lifetime alcohol consumption, and age of drinking onset. CONCLUSION These data implicate the induction of HMGB1-TLR-MyD88-NFκB cascades through coordinated glial and neuronal signaling as contributors to the neurodegeneration seen in the postmortem human OFC of individuals with AUD.
Collapse
Affiliation(s)
- Ryan P. Vetreno
- Bowles Center for Alcohol StudiesSchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of PsychiatrySchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Liya Qin
- Bowles Center for Alcohol StudiesSchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Leon G. Coleman
- Bowles Center for Alcohol StudiesSchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of PharmacologySchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Fulton T. Crews
- Bowles Center for Alcohol StudiesSchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of PsychiatrySchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of PharmacologySchool of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
41
|
Kisby BR, Farris SP, McManus MM, Varodayan FP, Roberto M, Harris RA, Ponomarev I. Alcohol Dependence in Rats Is Associated with Global Changes in Gene Expression in the Central Amygdala. Brain Sci 2021; 11:1149. [PMID: 34573170 PMCID: PMC8468792 DOI: 10.3390/brainsci11091149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 08/06/2021] [Accepted: 08/21/2021] [Indexed: 12/20/2022] Open
Abstract
Alcohol dependence is associated with adverse consequences of alcohol (ethanol) use and is evident in most severe cases of alcohol use disorder (AUD). The central nucleus of the amygdala (CeA) plays a critical role in the development of alcohol dependence and escalation of alcohol consumption in dependent subjects. Molecular mechanisms underlying the CeA-driven behavioral changes are not well understood. Here, we examined the effects of alcohol on global gene expression in the CeA using a chronic intermittent ethanol (CIE) vapor model in rats and RNA sequencing (RNA-Seq). The CIE procedure resulted in robust changes in CeA gene expression during intoxication, as the number of differentially expressed genes (DEGs) was significantly greater than those expected by chance. Over-representation analysis of cell types, functional groups and molecular pathways revealed biological categories potentially important for the development of alcohol dependence in our model. Genes specific for astrocytes, myelinating oligodendrocytes, and endothelial cells were over-represented in the DEG category, suggesting that these cell types were particularly affected by the CIE procedure. The majority of the over-represented functional groups and molecular pathways were directly related to the functions of glial and endothelial cells, including extracellular matrix (ECM) organization, myelination, and the regulation of innate immune response. A coordinated regulation of several ECM metalloproteinases (e.g., Mmp2; Mmp14), their substrates (e.g., multiple collagen genes and myelin basic protein; Mbp), and a metalloproteinase inhibitor, Reck, suggests a specific mechanism for ECM re-organization in response to chronic alcohol, which may modulate neuronal activity and result in behavioral changes, such as an escalation of alcohol drinking. Our results highlight the importance of glial and endothelial cells in the effects of chronic alcohol exposure on the CeA, and demonstrate further insight into the molecular mechanisms of alcohol dependence in rats. These molecular targets may be used in future studies to develop therapeutics to treat AUD.
Collapse
Affiliation(s)
- Brent R. Kisby
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (B.R.K.); (M.M.M.)
| | - Sean P. Farris
- Department of Neuroscience, University of Texas at Austin, Austin, TX 78715, USA; (S.P.F.); (R.A.H.)
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - Michelle M. McManus
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (B.R.K.); (M.M.M.)
| | - Florence P. Varodayan
- Department of Psychology, Binghamton University-SUNY, Binghamton, NY 13902, USA;
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA;
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA;
| | - R. Adron Harris
- Department of Neuroscience, University of Texas at Austin, Austin, TX 78715, USA; (S.P.F.); (R.A.H.)
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78741, USA
| | - Igor Ponomarev
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (B.R.K.); (M.M.M.)
| |
Collapse
|
42
|
Vanacker C, Defazio RA, Sykes CM, Moenter SM. A role for glial fibrillary acidic protein (GFAP)-expressing cells in the regulation of gonadotropin-releasing hormone (GnRH) but not arcuate kisspeptin neuron output in male mice. eLife 2021; 10:68205. [PMID: 34292152 PMCID: PMC8337074 DOI: 10.7554/elife.68205] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/21/2021] [Indexed: 01/05/2023] Open
Abstract
GnRH neurons are the final central neural output regulating fertility. Kisspeptin neurons in the hypothalamic arcuate nucleus (KNDy neurons) are considered the main regulator of GnRH output. GnRH and KNDy neurons are surrounded by astrocytes, which can modulate neuronal activity and communicate over distances. Prostaglandin E2 (PGE2), synthesized primarily by astrocytes, increases GnRH neuron activity and downstream pituitary release of luteinizing hormone (LH). We hypothesized that glial fibrillary acidic protein (GFAP)-expressing astrocytes play a role in regulating GnRH and/or KNDy neuron activity and LH release. We used adeno-associated viruses to target designer receptors exclusively activated by designer drugs (DREADDs) to GFAP-expressing cells to activate Gq- or Gi-mediated signaling. Activating Gq signaling in the preoptic area, near GnRH neurons, but not in the arcuate, increases LH release in vivo and GnRH firing in vitro via a mechanism in part dependent upon PGE2. These data suggest that astrocytes can activate GnRH/LH release in a manner independent of KNDy neurons.
Collapse
Affiliation(s)
- Charlotte Vanacker
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, United States
| | - R Anthony Defazio
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, United States
| | - Charlene M Sykes
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, United States
| | - Suzanne M Moenter
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, United States.,Internal Medicine, University of Michigan, Ann Arbor, United States.,Obstetrics & Gynecology, University of Michigan, Ann Arbor, United States.,Reproductive Sciences Program, University of Michigan, Ann Arbor, United States
| |
Collapse
|
43
|
Pickering CA, Mazarakis ND. Viral Vector Delivery of DREADDs for CNS Therapy. Curr Gene Ther 2021; 21:191-206. [PMID: 33573551 DOI: 10.2174/1566523221666210211102435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/15/2021] [Accepted: 01/25/2021] [Indexed: 11/22/2022]
Abstract
Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) are genetically modified G-protein-coupled receptors (GPCRs), that can be activated by a synthetic ligand which is otherwise inert at endogenous receptors. DREADDs can be expressed in cells in the central nervous system (CNS) and subsequently offer the opportunity for remote and reversible silencing or activation of the target cells when the synthetic ligand is systemically administered. In neuroscience, DREADDs have thus far shown to be useful tools for several areas of research and offer considerable potential for the development of gene therapy strategies for neurological disorders. However, in order to design a DREADD-based gene therapy, it is necessary to first evaluate the viral vector delivery methods utilised in the literature to deliver these chemogenetic tools. This review evaluates each of the prominent strategies currently utilised for DREADD delivery, discussing their respective advantages and limitations. We focus on adeno-associated virus (AAV)-based and lentivirus-based systems, and the manipulation of these through cell-type specific promoters and pseudotyping. Furthermore, we address how virally mediated DREADD delivery could be improved in order to make it a viable gene therapy strategy and thus expand its translational potential.
Collapse
Affiliation(s)
- Ceri A Pickering
- Division of Neuroscience, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Nicholas D Mazarakis
- Division of Neuroscience, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
44
|
Lim EY, Ye L, Paukert M. Potential and Realized Impact of Astroglia Ca 2 + Dynamics on Circuit Function and Behavior. Front Cell Neurosci 2021; 15:682888. [PMID: 34163330 PMCID: PMC8215280 DOI: 10.3389/fncel.2021.682888] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/03/2021] [Indexed: 11/13/2022] Open
Abstract
Astroglia display a wide range of spontaneous and behavioral state-dependent Ca2+ dynamics. During heightened vigilance, noradrenergic signaling leads to quasi-synchronous Ca2+ elevations encompassing soma and processes across the brain-wide astroglia network. Distinct from this vigilance-associated global Ca2+ rise are apparently spontaneous fluctuations within spatially restricted microdomains. Over the years, several strategies have been pursued to shed light on the physiological impact of these signals including deletion of endogenous ion channels or receptors and reduction of intracellular Ca2+ through buffering, extrusion or inhibition of release. Some experiments that revealed the most compelling behavioral alterations employed chemogenetic and optogenetic manipulations to modify astroglia Ca2+ signaling. However, there is considerable contrast between these findings and the comparatively modest effects of inhibiting endogenous sources of Ca2+. In this review, we describe the underlying mechanisms of various forms of astroglia Ca2+ signaling as well as the functional consequences of their inhibition. We then discuss how the effects of exogenous astroglia Ca2+ modification combined with our knowledge of physiological mechanisms of astroglia Ca2+ activation could guide further refinement of behavioral paradigms that will help elucidate the natural Ca2+-dependent function of astroglia.
Collapse
Affiliation(s)
- Eunice Y. Lim
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Liang Ye
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Martin Paukert
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States,*Correspondence: Martin Paukert,
| |
Collapse
|
45
|
Marshall SA. Glial cells as influencers and maladaptive consequences of alcohol use disorders. J Neurosci Res 2021; 99:1905-1907. [PMID: 34062005 DOI: 10.1002/jnr.24868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 11/06/2022]
Affiliation(s)
- S Alex Marshall
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, USA.,Department of Psychology & Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
46
|
Hamada K, Ferguson LB, Mayfield RD, Krishnan HR, Maienschein-Cline M, Lasek AW. Binge-like ethanol drinking activates anaplastic lymphoma kinase signaling and increases the expression of STAT3 target genes in the mouse hippocampus and prefrontal cortex. GENES, BRAIN, AND BEHAVIOR 2021; 20:e12729. [PMID: 33641239 PMCID: PMC8944393 DOI: 10.1111/gbb.12729] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/04/2021] [Accepted: 02/26/2021] [Indexed: 02/03/2023]
Abstract
Alcohol use disorder (AUD) has a complex pathogenesis, making it a difficult disorder to treat. Identifying relevant signaling pathways in the brain may be useful for finding new pharmacological targets to treat AUD. The receptor tyrosine kinase anaplastic lymphoma kinase (ALK) activates the transcription factor STAT3 in response to ethanol in cell lines. Here, we show ALK activation and upregulation of known STAT3 target genes (Socs3, Gfap and Tnfrsf1a) in the prefrontal cortex (PFC) and ventral hippocampus (HPC) of mice after 4 days of binge-like ethanol drinking. Mice treated with the STAT3 inhibitor stattic drank less ethanol than vehicle-treated mice, demonstrating the behavioral importance of STAT3. To identify novel ethanol-induced target genes downstream of the ALK and STAT3 pathway, we analyzed the NIH LINCS L1000 database for gene signature overlap between ALK inhibitor (alectinib and NVP-TAE684) and STAT3 inhibitor (niclosamide) treatments on cell lines. These genes were then compared with differentially expressed genes in the PFC of mice after binge-like drinking. We found 95 unique gene candidates, out of which 57 had STAT3 binding motifs in their promoters. We further showed by qPCR that expression of the putative STAT3 genes Nr1h2, Smarcc1, Smarca4 and Gpnmb were increased in either the PFC or HPC after binge-like drinking. Together, these results indicate activation of the ALK-STAT3 signaling pathway in the brain after binge-like ethanol consumption, identify putative novel ethanol-responsive STAT3 target genes, and suggest that STAT3 inhibition may be a potential method to reduce binge drinking in humans.
Collapse
Affiliation(s)
- Kana Hamada
- Graduate Program in Neuroscience, University of Illinois at Chicago, Chicago, IL 60612 USA
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Laura B. Ferguson
- Waggoner Center for Alcohol Addiction Research and Department of Neuroscience, University of Texas at Austin, Austin, TX 78712 USA
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - R. Dayne Mayfield
- Waggoner Center for Alcohol Addiction Research and Department of Neuroscience, University of Texas at Austin, Austin, TX 78712 USA
| | - Harish R. Krishnan
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612 USA
| | | | - Amy W. Lasek
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612 USA
| |
Collapse
|
47
|
Wahis J, Holt MG. Astrocytes, Noradrenaline, α1-Adrenoreceptors, and Neuromodulation: Evidence and Unanswered Questions. Front Cell Neurosci 2021; 15:645691. [PMID: 33716677 PMCID: PMC7947346 DOI: 10.3389/fncel.2021.645691] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/03/2021] [Indexed: 12/27/2022] Open
Abstract
Noradrenaline is a major neuromodulator in the central nervous system (CNS). It is released from varicosities on neuronal efferents, which originate principally from the main noradrenergic nuclei of the brain - the locus coeruleus - and spread throughout the parenchyma. Noradrenaline is released in response to various stimuli and has complex physiological effects, in large part due to the wide diversity of noradrenergic receptors expressed in the brain, which trigger diverse signaling pathways. In general, however, its main effect on CNS function appears to be to increase arousal state. Although the effects of noradrenaline have been researched extensively, the majority of studies have assumed that noradrenaline exerts its effects by acting directly on neurons. However, neurons are not the only cells in the CNS expressing noradrenaline receptors. Astrocytes are responsive to a range of neuromodulators - including noradrenaline. In fact, noradrenaline evokes robust calcium transients in astrocytes across brain regions, through activation of α1-adrenoreceptors. Crucially, astrocytes ensheath neurons at synapses and are known to modulate synaptic activity. Hence, astrocytes are in a key position to relay, or amplify, the effects of noradrenaline on neurons, most notably by modulating inhibitory transmission. Based on a critical appraisal of the current literature, we use this review to argue that a better understanding of astrocyte-mediated noradrenaline signaling is therefore essential, if we are ever to fully understand CNS function. We discuss the emerging concept of astrocyte heterogeneity and speculate on how this might impact the noradrenergic modulation of neuronal circuits. Finally, we outline possible experimental strategies to clearly delineate the role(s) of astrocytes in noradrenergic signaling, and neuromodulation in general, highlighting the urgent need for more specific and flexible experimental tools.
Collapse
Affiliation(s)
- Jérôme Wahis
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
| | - Matthew G. Holt
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
48
|
Hwang SN, Lee JS, Seo K, Lee H. Astrocytic Regulation of Neural Circuits Underlying Behaviors. Cells 2021; 10:cells10020296. [PMID: 33535587 PMCID: PMC7912785 DOI: 10.3390/cells10020296] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/23/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
Astrocytes, characterized by a satellite-like morphology, are the most abundant type of glia in the central nervous system. Their main functions have been thought to be limited to providing homeostatic support for neurons, but recent studies have revealed that astrocytes actually actively interact with local neural circuits and play a crucial role in information processing and generating physiological and behavioral responses. Here, we review the emerging roles of astrocytes in many brain regions, particularly by focusing on intracellular changes in astrocytes and their interactions with neurons at the molecular and neural circuit levels.
Collapse
Affiliation(s)
- Sun-Nyoung Hwang
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea;
| | - Jae Seung Lee
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, Korea; (J.S.L.); (K.S.)
| | - Kain Seo
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, Korea; (J.S.L.); (K.S.)
| | - Hyosang Lee
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea;
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, Korea; (J.S.L.); (K.S.)
- Korea Brain Research Institute (KBRI), Daegu 41062, Korea
- Correspondence: ; Tel.: +82-53-785-6147
| |
Collapse
|
49
|
Alcohol. Alcohol 2021. [DOI: 10.1016/b978-0-12-816793-9.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
50
|
Giacometti LL, Chandran K, Figueroa LA, Barker JM. Astrocyte modulation of extinction impairments in ethanol-dependent female mice. Neuropharmacology 2020; 179:108272. [PMID: 32801026 DOI: 10.1016/j.neuropharm.2020.108272] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/17/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022]
Abstract
Rates of alcohol use disorders are increasing in women, and there is growing evidence that both the cognitive and biological consequences of alcohol dependence are distinct in women as compared to men. Despite this, the neurobehavioral outcomes of chronic alcohol exposure are poorly characterized in women and female animals. In this study, we find that ethanol dependence impaired extinction of reward seeking in a food conditioned place preference task in female mice. At the same time point, ethanol-dependent females exhibited astrocytic dysregulation as indicated by a brain region-specific reduction in glial fibrillary acidic protein (GFAP) expression. Using a chemogenetic strategy, we demonstrate that modulating astrocyte function via chemogenetic activation of Gq-signaling in nucleus accumbens astrocytes transiently rescued extinction in ethanol-dependent females without impacting basal reward seeking. These findings identify astrocyte function as a potential target for the restoration of behavioral flexibility following chronic alcohol exposure in females.
Collapse
Affiliation(s)
- Laura L Giacometti
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Kelsey Chandran
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Laura A Figueroa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Jacqueline M Barker
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA.
| |
Collapse
|