1
|
Nemeth DP, Liu X, Monet MC, Niu H, Maxey G, Schrier MS, Smirnova MI, McGovern SJ, Herd A, DiSabato DJ, Floyd T, Atluri RR, Nusstein AC, Oliver B, Witcher KG, Juste Ellis JS, Yip J, Crider AD, McKim DB, Gajewski-Kurdziel PA, Godbout JP, Zhang Q, Blakely RD, Sheridan JF, Quan N. Localization of brain neuronal IL-1R1 reveals specific neural circuitries responsive to immune signaling. J Neuroinflammation 2024; 21:303. [PMID: 39563437 PMCID: PMC11575132 DOI: 10.1186/s12974-024-03287-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
Interleukin-1 (IL-1) is a pro-inflammatory cytokine that exerts a wide range of neurological and immunological effects throughout the central nervous system (CNS) and is associated with the etiology of affective and cognitive disorders. The cognate receptor for IL-1, Interleukin-1 Receptor Type 1 (IL-1R1), is primarily expressed on non-neuronal cells (e.g., endothelial cells, choroidal cells, ventricular ependymal cells, astrocytes, etc.) throughout the brain. However, the presence and distribution of neuronal IL-1R1 (nIL-1R1) has been controversial. Here, for the first time, a novel genetic mouse line that allows for the visualization of IL-1R1 mRNA and protein expression (Il1r1GR/GR) was used to map all brain nuclei and determine the neurotransmitter systems which express nIL-1R1 in adult male mice. The direct responsiveness of nIL-1R1-expressing neurons to both inflammatory and physiological levels of IL-1β in vivo was tested. Neuronal IL-1R1 expression across the brain was found in discrete glutamatergic and serotonergic neuronal populations in the somatosensory cortex, piriform cortex, dentate gyrus, and dorsal raphe nucleus. Glutamatergic nIL-1R1 comprises most of the nIL-1R1 expression and, using Vglut2-Cre-Il1r1r/r mice, which restrict IL-1R1 expression to only glutamatergic neurons, an atlas of glutamatergic nIL-1R1 expression across the brain was generated. Analysis of functional outputs of these nIL-1R1-expressing nuclei, in both Il1r1GR/GR and Vglut2-Cre-Il1r1r/r mice, reveals IL-1R1+ nuclei primarily relate to sensory detection, processing, and relay pathways, mood regulation, and spatial/cognitive processing centers. Intracerebroventricular (i.c.v.) injections of IL-1 (20 ng) induces NFκB signaling in IL-1R1+ non-neuronal cells but not in IL-1R1+ neurons, and in Vglut2-Cre-Il1r1r/r mice IL-1 did not change gene expression in the dentate gyrus of the hippocampus (DG). GO pathway analysis of spatial RNA sequencing 1mo following restoration of nIL-1R1 in the DG neurons reveals IL-1R1 expression downregulates genes related to both synaptic function and mRNA binding while increasing select complement markers (C1ra, C1qb). Further, DG neurons exclusively express an alternatively spliced IL-1R Accessory protein isoform (IL-1RAcPb), a known synaptic adhesion molecule. Altogether, this study reveals a unique network of neurons that can respond directly to IL-1 via nIL-1R1 through non-autonomous transcriptional pathways; earmarking these circuits as potential neural substrates for immune signaling-triggered sensory, affective, and cognitive disorders.
Collapse
Affiliation(s)
- Daniel P Nemeth
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA.
| | - Xiaoyu Liu
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Marianne C Monet
- The International Max Planck Research School (IMPRS) for Synapses and Circuits, Max Planck Florida Institute for Neuroscience Jupiter, Jupiter, FL, 33458, USA
- Department of Biological Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Haichen Niu
- Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Gabriella Maxey
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
- Wilkes Honors College, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Matt S Schrier
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Maria I Smirnova
- The International Max Planck Research School (IMPRS) for Synapses and Circuits, Max Planck Florida Institute for Neuroscience Jupiter, Jupiter, FL, 33458, USA
- Department of Biological Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL, 33458, USA
| | | | - Anu Herd
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Damon J DiSabato
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
| | - Trey Floyd
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Rohit R Atluri
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH, 43614, USA
| | - Alex C Nusstein
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Braedan Oliver
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Kristina G Witcher
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
| | - Joshua St Juste Ellis
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Jasmine Yip
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Andrew D Crider
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Daniel B McKim
- Department of Animal Science, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | | | - Jonathan P Godbout
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
| | - Qi Zhang
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
- Department of Biological Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL, 33458, USA
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - John F Sheridan
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, 43210, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA.
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA.
| |
Collapse
|
2
|
Zhang Y, Tan X, Tang C. Estrogen-immuno-neuromodulation disorders in menopausal depression. J Neuroinflammation 2024; 21:159. [PMID: 38898454 PMCID: PMC11188190 DOI: 10.1186/s12974-024-03152-1] [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: 04/20/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024] Open
Abstract
A significant decrease in estrogen levels puts menopausal women at high risk for major depression, which remains difficult to cure despite its relatively clear etiology. With the discovery of abnormally elevated inflammation in menopausal depressed women, immune imbalance has become a novel focus in the study of menopausal depression. In this paper, we examined the characteristics and possible mechanisms of immune imbalance caused by decreased estrogen levels during menopause and found that estrogen deficiency disrupted immune homeostasis, especially the levels of inflammatory cytokines through the ERα/ERβ/GPER-associated NLRP3/NF-κB signaling pathways. We also analyzed the destruction of the blood-brain barrier, dysfunction of neurotransmitters, blockade of BDNF synthesis, and attenuation of neuroplasticity caused by inflammatory cytokine activity, and investigated estrogen-immuno-neuromodulation disorders in menopausal depression. Current research suggests that drugs targeting inflammatory cytokines and NLRP3/NF-κB signaling molecules are promising for restoring homeostasis of the estrogen-immuno-neuromodulation system and may play a positive role in the intervention and treatment of menopausal depression.
Collapse
Affiliation(s)
- Yuling Zhang
- College of Life Science, Henan Normal University, Xinxiang, 453007, Henan, China
| | - Xiying Tan
- Department of Neurology, Xinxiang City First People's Hospital, Xinxiang, 453000, Henan, China
| | - Chaozhi Tang
- College of Life Science, Henan Normal University, Xinxiang, 453007, Henan, China.
| |
Collapse
|
3
|
Sowa JE, Tokarski K, Hess G. Activation of the CXCR4 Receptor by Chemokine CXCL12 Increases the Excitability of Neurons in the Rat Central Amygdala. J Neuroimmune Pharmacol 2024; 19:9. [PMID: 38430337 DOI: 10.1007/s11481-024-10112-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 02/23/2024] [Indexed: 03/03/2024]
Abstract
Primarily regarded as immune proteins, chemokines are emerging as a family of molecules serving neuromodulatory functions in the developing and adult brain. Among them, CXCL12 is constitutively and widely expressed in the CNS, where it was shown to act on cellular, synaptic, network, and behavioral levels. Its receptor, CXCR4, is abundant in the amygdala, a brain structure involved in pathophysiology of anxiety disorders. Dysregulation of CXCL12/CXCR4 signaling has been implicated in anxiety-related behaviors. Here we demonstrate that exogenous CXCL12 at 2 nM but not at 5 nM increased neuronal excitability in the lateral division of the rat central amygdala (CeL) which was evident in the Late-Firing but not Regular-Spiking neurons. These effects were blocked by AMD3100, a CXCR4 antagonist. Moreover, CXCL12 increased the excitability of the neurons of the basolateral amygdala (BLA) that is known to project to the CeL. However, CXCL12 increased neither the spontaneous excitatory nor spontaneous inhibitory synaptic transmission in the CeL. In summary, the data reveal specific activation of Late-Firing CeL cells along with BLA neurons by CXCL12 and suggest that this chemokine may alter information processing by the amygdala that likely contributes to anxiety and fear conditioning.
Collapse
Affiliation(s)
- Joanna Ewa Sowa
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, Krakow, 31-343, Poland.
| | - Krzysztof Tokarski
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, Krakow, 31-343, Poland
| | - Grzegorz Hess
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, Krakow, 31-343, Poland
| |
Collapse
|
4
|
Munshi S, Albrechet-Souza L, Dos-Santos RC, Stelly CE, Secci ME, Gilpin NW, Tasker JG. Acute Ethanol Modulates Synaptic Inhibition in the Basolateral Amygdala via Rapid NLRP3 Inflammasome Activation and Regulates Anxiety-Like Behavior in Rats. J Neurosci 2023; 43:7902-7912. [PMID: 37739795 PMCID: PMC10669756 DOI: 10.1523/jneurosci.1744-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 08/22/2023] [Accepted: 09/14/2023] [Indexed: 09/24/2023] Open
Abstract
Chronic alcohol exposure leads to a neuroinflammatory response involving activation of the nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome and proinflammatory cytokine production. Acute ethanol (EtOH) exposure activates GABAergic synapses in the central and basolateral amygdala (BLA) ex vivo, but whether this rapid modulation of synaptic inhibition is because of an acute inflammatory response and alters anxiety-like behavior in male and female animals is not known. Here, we tested the hypotheses that acute EtOH facilitates inhibitory synaptic transmission in the BLA by activating the NLRP3 inflammasome-dependent acute inflammatory response, that the alcohol-induced increase in inhibition is cell type and sex dependent, and that acute EtOH in the BLA reduces anxiety-like behavior. Acute EtOH application at a binge-like concentration (22-44 mm) stimulated synaptic GABA release from putative parvalbumin (PV) interneurons onto BLA principal neurons in ex vivo brain slices from male, but not female, rats. The EtOH facilitation of synaptic inhibition was blocked by antagonists of the Toll-like receptor 4 (TLR4), the NLRP3 inflammasome, and interleukin-1 receptors, suggesting it was mediated by a rapid local neuroinflammatory response in the BLA. In vivo, bilateral injection of EtOH directly into the BLA produced an acute concentration-dependent reduction in anxiety-like behavior in male but not female rats. These findings demonstrate that acute EtOH in the BLA regulates anxiety-like behavior in a sex-dependent manner and suggest that this effect is associated with presynaptic facilitation of parvalbumin-expressing interneuron inputs to BLA principal neurons via a local NLRP3 inflammasome-dependent neuroimmune response.SIGNIFICANCE STATEMENT Chronic alcohol exposure produces a neuroinflammatory response, which contributes to alcohol-associated pathologies. Acute alcohol administration increases inhibitory synaptic signaling in the brain, but the mechanism for the rapid alcohol facilitation of inhibitory circuits is unknown. We found that acute ethanol at binge-like concentrations in the basolateral amygdala (BLA) facilitates GABA release from parvalbumin-expressing (PV) interneuron synapses onto principal neurons in ex vivo brain slices from male rats and that intra-BLA ethanol reduces anxiety-like behavior in vivo in male rats, but not female rats. The ethanol (EtOH) facilitation of inhibition in the BLA is mediated by Toll-like receptor 4 (TLR4) and nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome activation and proinflammatory IL-1β signaling, which suggests a rapid NLRP3 inflammasome-dependent neuroimmune cascade that plays a critical role in acute alcohol intoxication.
Collapse
Affiliation(s)
- Soumyabrata Munshi
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Lucas Albrechet-Souza
- Department of Cell Biology and Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Alcohol and Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | | | - Claire E Stelly
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
| | - Maria E Secci
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Alcohol and Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Southeast Louisiana Veterans Affairs Healthcare System (SLVHCS), New Orleans, Louisiana 70119
| | - Nicholas W Gilpin
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Alcohol and Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
- Southeast Louisiana Veterans Affairs Healthcare System (SLVHCS), New Orleans, Louisiana 70119
| | - Jeffrey G Tasker
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana 70118
- Tulane Brain Institute, Tulane University, New Orleans, Louisiana 70118
- Southeast Louisiana Veterans Affairs Healthcare System (SLVHCS), New Orleans, Louisiana 70119
| |
Collapse
|
5
|
Kokhan VS, Anokhin PK, Proskuryakova TV, Shokhonova VA, Ageldinov RA, Shamakina IY. Interleukin-1β and TNF-α are elevated in the amygdala of adult rats prenatally exposed to ethanol. BIOMEDITSINSKAIA KHIMIIA 2023; 69:300-306. [PMID: 37937432 DOI: 10.18097/pbmc20236905300] [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: 11/09/2023]
Abstract
Affective disorders, including anxiety and depression, developed in adult offspring of the mothers who consumed alcohol during pregnancy could be associated with an imbalance in neuroimmune factors in the amygdala (corpus amygdaloideum) resulted in impaired emotional stimulus processing. The aim of this study was to compare the content of cytokines TNF-α, IL-1α, IL-1β, IL-10, and IL-17 in the amygdala of adult female rats exposed to alcohol in utero and control rats. Cytokine levels were evaluated using a multiplex immunoassay system; mRNA expression was investigated using a real-time reverse transcription-polymerase chain reaction (RT-qPCR) assay. Prenatal alcohol exposure led to the increase in the content of TNF-α and IL-1β without significant changes in the mRNA expression level. Our data suggest that ethanol exposure to the fetus during pregnancy can result in long-term alterations in the content of the key neuroinflammatory factors in the amygdala, which in turn can be a risk factor for affective disorders in the adulthood.
Collapse
Affiliation(s)
- V S Kokhan
- National Scientific Center for Narcology - Branch of the V.P. Serbsky National Medical Research Centre for Psychiatry and Narcology, Moscow, Russia
| | - P K Anokhin
- National Scientific Center for Narcology - Branch of the V.P. Serbsky National Medical Research Centre for Psychiatry and Narcology, Moscow, Russia
| | - T V Proskuryakova
- National Scientific Center for Narcology - Branch of the V.P. Serbsky National Medical Research Centre for Psychiatry and Narcology, Moscow, Russia
| | - V A Shokhonova
- National Scientific Center for Narcology - Branch of the V.P. Serbsky National Medical Research Centre for Psychiatry and Narcology, Moscow, Russia
| | - R A Ageldinov
- Scientific Center of Biomedical Technologies of the Federal Medical and Biological Agency of Russia, Svetlye gory, Moscow Region, Russia
| | - I Yu Shamakina
- National Scientific Center for Narcology - Branch of the V.P. Serbsky National Medical Research Centre for Psychiatry and Narcology, Moscow, Russia
| |
Collapse
|
6
|
Cruz B, Borgonetti V, Bajo M, Roberto M. Sex-dependent factors of alcohol and neuroimmune mechanisms. Neurobiol Stress 2023; 26:100562. [PMID: 37601537 PMCID: PMC10432974 DOI: 10.1016/j.ynstr.2023.100562] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/25/2023] [Accepted: 08/02/2023] [Indexed: 08/22/2023] Open
Abstract
Excessive alcohol use disrupts neuroimmune signaling across various cell types, including neurons, microglia, and astrocytes. The present review focuses on recent, albeit limited, evidence of sex differences in biological factors that mediate neuroimmune responses to alcohol and underlying neuroimmune systems that may influence alcohol drinking behaviors. Females are more vulnerable than males to the neurotoxic and negative consequences of chronic alcohol drinking, reflected by elevations of pro-inflammatory cytokines and inflammatory mediators. Differences in cytokine, microglial, astrocytic, genomic, and transcriptomic evidence suggest females are more reactive than males to neuroinflammatory changes after chronic alcohol exposure. The growing body of evidence supports that innate immune factors modulate synaptic transmission, providing a mechanistic framework to examine sex differences across neurocircuitry. Targeting neuroimmune signaling may be a viable strategy for treating AUD, but more research is needed to understand sex-specific differences in alcohol drinking and neuroimmune mechanisms.
Collapse
Affiliation(s)
- Bryan Cruz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| | - Vittoria Borgonetti
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| | - Michal Bajo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA, 92073
| |
Collapse
|
7
|
Li M, Sun X, Wang Z, Li Y. Caspase-1 affects chronic restraint stress-induced depression-like behaviors by modifying GABAergic dysfunction in the hippocampus. Transl Psychiatry 2023; 13:229. [PMID: 37369673 DOI: 10.1038/s41398-023-02527-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Major depression disorder (MDD) is one of the most common psychiatric disorders and one of the leading causes of disability in worldwide. Both inflammation and GABAergic dysfunction have been implicated in the pathophysiology of MDD. Caspase-1, a classic inflammatory caspase, regulates AMPARs-mediated glutamatergic neurotransmission. However, the role of caspase-1 in chronic stress-induced GABAergic dysfunction remains largely unknown. In this study, we found that serum and hippocampal caspase-1-IL-1β levels increased significantly in chronic restraint stress (CRS) mice, and a significant negative correlation occurred between levels of caspase-1 and depression-like behaviors. Furthermore, CRS significantly decreased GAD67 mRNA levels and GABAergic neurotransmission accompanied by the reduction of GABA concentration, reduced the amplitude and frequency of mIPSCs inhibitory postsynaptic currents (mIPSCs) and the decreased surface expression of GABAARs γ2 subunit in the hippocampus. Genetic deficiency of caspase-1 not only blocked CRS-induced depression-like behaviors, but also alleviated CRS-induced impairments in GABAergic neurotransmission. Finally, reexpression of caspase-1 in the hippocampus of Caspase-1-/- mice increased susceptibility to stress-induced anxiety- and depression-like behaviors through inhibiting GAD67 expression and GABAARs-mediated synaptic transmission. Our study suggests that CRS dysregulates GABAergic neurotransmission via increasing the levels of caspase-1-mediated neuroinflammation in the hippocampus, ultimately leading to depression-like behaviors. This work illustrates that targeting caspase-1 may provide potential therapeutic benefits to stress-related GABAergic dysfunction in the pathogenesis of MDD.
Collapse
Affiliation(s)
- Mingxing Li
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430012, China.
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, 430012, China.
| | - Xuejiao Sun
- Department of Rehabilitation Medicine, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Zongqin Wang
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430012, China
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, 430012, China
| | - Yi Li
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430012, China.
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, 430012, China.
| |
Collapse
|
8
|
Varodayan FP, Pahng AR, Davis TD, Gandhi P, Bajo M, Steinman MQ, Kiosses WB, Blednov YA, Burkart MD, Edwards S, Roberts AJ, Roberto M. Chronic ethanol induces a pro-inflammatory switch in interleukin-1β regulation of GABAergic signaling in the medial prefrontal cortex of male mice. Brain Behav Immun 2023; 110:125-139. [PMID: 36863493 PMCID: PMC10106421 DOI: 10.1016/j.bbi.2023.02.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/20/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023] Open
Abstract
Neuroimmune pathways regulate brain function to influence complex behavior and play a role in several neuropsychiatric diseases, including alcohol use disorder (AUD). In particular, the interleukin-1 (IL-1) system has emerged as a key regulator of the brain's response to ethanol (alcohol). Here we investigated the mechanisms underlying ethanol-induced neuroadaptation of IL-1β signaling at GABAergic synapses in the prelimbic region of the medial prefrontal cortex (mPFC), an area responsible for integrating contextual information to mediate conflicting motivational drives. We exposed C57BL/6J male mice to the chronic intermittent ethanol vapor-2 bottle choice paradigm (CIE-2BC) to induce ethanol dependence, and conducted ex vivo electrophysiology and molecular analyses. We found that the IL-1 system regulates basal mPFC function through its actions at inhibitory synapses on prelimbic layer 2/3 pyramidal neurons. IL-1β can selectively recruit either neuroprotective (PI3K/Akt) or pro-inflammatory (MyD88/p38 MAPK) mechanisms to produce opposing synaptic effects. In ethanol naïve conditions, there was a strong PI3K/Akt bias leading to a disinhibition of pyramidal neurons. Ethanol dependence produced opposite IL-1 effects - enhanced local inhibition via a switch in IL-1β signaling to the canonical pro-inflammatory MyD88 pathway. Ethanol dependence also increased cellular IL-1β in the mPFC, while decreasing expression of downstream effectors (Akt, p38 MAPK). Thus, IL-1β may represent a key neural substrate in ethanol-induced cortical dysfunction. As the IL-1 receptor antagonist (kineret) is already FDA-approved for other diseases, this work underscores the high therapeutic potential of IL-1 signaling/neuroimmune-based treatments for AUD.
Collapse
Affiliation(s)
- F P Varodayan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA; Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - A R Pahng
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, USA; Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| | - T D Davis
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University-SUNY, Binghamton, NY, USA
| | - P Gandhi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - M Bajo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - M Q Steinman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - W B Kiosses
- Microscopy Core Imaging Facility, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Y A Blednov
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - M D Burkart
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA, USA
| | - S Edwards
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - A J Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, USA
| | - M Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
9
|
Khan KM, Bierlein-De La Rosa G, Biggerstaff N, Pushpavathi Selvakumar G, Wang R, Mason S, Dailey ME, Marcinkiewcz CA. Adolescent ethanol drinking promotes hyperalgesia, neuroinflammation and serotonergic deficits in mice that persist into adulthood. Brain Behav Immun 2023; 107:419-431. [PMID: 35907582 PMCID: PMC10289137 DOI: 10.1016/j.bbi.2022.07.160] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 02/09/2023] Open
Abstract
Adolescent alcohol use can permanently alter brain function and lead to poor health outcomes in adulthood. Emerging evidence suggests that alcohol use can predispose individuals to pain disorders or exacerbate existing pain conditions, but the underlying neural mechanisms are currently unknown. Here we report that mice exposed to adolescent intermittent access to ethanol (AIE) exhibit increased pain sensitivity and depressive-like behaviors that persist for several weeks after alcohol cessation and are accompanied by elevated CD68 expression in microglia and reduced numbers of serotonin (5-HT)-expressing neurons in the dorsal raphe nucleus (DRN). 5-HT expression was also reduced in the thalamus, anterior cingulate cortex (ACC) and amygdala as well as the lumbar dorsal horn of the spinal cord. We further demonstrate that chronic minocycline administration after AIE alleviated hyperalgesia and social deficits, while chemogenetic activation of microglia in the DRN of ethanol-naïve mice reproduced the effects of AIE on pain and social behavior. Chemogenetic activation of microglia also reduced tryptophan hydroxylase 2 (Tph2) expression and was negatively correlated with the number of 5-HT-immunoreactive cells in the DRN. Taken together, these results indicate that microglial activation in the DRN may be a primary driver of pain, negative affect, and 5-HT depletion after AIE.
Collapse
Affiliation(s)
- Kanza M Khan
- Department of Neuroscience and Pharmacology, University of Iowa, United States
| | | | - Natalie Biggerstaff
- Department of Neuroscience and Pharmacology, University of Iowa, United States
| | | | - Ruixiang Wang
- Department of Neuroscience and Pharmacology, University of Iowa, United States
| | - Suzanne Mason
- Department of Neuroscience and Pharmacology, University of Iowa, United States
| | - Michael E Dailey
- Iowa Neuroscience Institute, University of Iowa, United States; Department of Biology, University of Iowa, United States
| | - Catherine A Marcinkiewcz
- Department of Neuroscience and Pharmacology, University of Iowa, United States; Iowa Neuroscience Institute, University of Iowa, United States.
| |
Collapse
|
10
|
Melkumyan M, Silberman Y. Subregional Differences in Alcohol Modulation of Central Amygdala Neurocircuitry. Front Mol Neurosci 2022; 15:888345. [PMID: 35866156 PMCID: PMC9294740 DOI: 10.3389/fnmol.2022.888345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Alcohol use disorder is a highly significant medical condition characterized by an impaired ability to stop or control alcohol use, compulsive alcohol seeking behavior, and withdrawal symptoms in the absence of alcohol. Understanding how alcohol modulates neurocircuitry critical for long term and binge-like alcohol use, such as the central amygdala (CeA), may lead to the development of novel therapeutic strategies to treat alcohol use disorder. In clinical studies, reduction in the volume of the amygdala has been linked with susceptibility to relapse to alcohol use. Preclinical studies have shown the involvement of the CeA in the effects of alcohol use, with lesions of the amygdala showing a reduction in alcohol drinking, and manipulations of cells in the CeA altering alcohol drinking. A great deal of work has shown that acute alcohol, as well as chronic alcohol exposure via intake or dependence models, alters glutamatergic and GABAergic transmission in the CeA. The CeA, however, contains heterogeneous cell populations and distinct subregional differences in neurocircuit architecture which may influence the mechanism by which alcohol modulates CeA function overall. The current review aimed to parse out the differences in alcohol effects on the medial and lateral subregions of the CeA, and what role neuroinflammatory cells and markers, the endocannabinoid system, and the most commonly studied neuropeptide systems play in mediating these effects. A better understanding of alcohol effects on CeA subregional cell type and neurocircuit function may lead to development of more selective pharmacological interventions for alcohol use disorder.
Collapse
Affiliation(s)
- Mariam Melkumyan
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, United States
| | - Yuval Silberman
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
11
|
Borrego MB, Chan AE, Ozburn AR. Regulation of alcohol drinking by ventral striatum and extended amygdala circuitry. Neuropharmacology 2022; 212:109074. [PMID: 35487273 PMCID: PMC9677601 DOI: 10.1016/j.neuropharm.2022.109074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/24/2022] [Accepted: 04/20/2022] [Indexed: 02/07/2023]
Abstract
Alcohol use disorder is a complex psychiatric disorder that can be modeled in rodents using a number of drinking paradigms. Drinking-in-the-dark (DID) is widely used to model the binge/intoxication stage of addiction, and chronic intermittent ethanol vapor procedures (CIE) are used to induce dependence and model withdrawal/negative affect induced escalation of drinking. We discuss experiments showing the ventral striatum (vStr) and extended amygdala (EA) are engaged in response to ethanol in rodents through c-Fos/Fos immunoreactivity studies. We also discuss experiments in rodents that span a wide variety of techniques where the function of vStr and EA structures are changed following DID or CIE, and the role of neurotransmitter and neuropeptide systems studies in these ethanol-related outcomes. We note where signaling systems converge across regions and paradigms and where there are still gaps in the literature. Dynorphin/κ-opioid receptor (KOR) signaling, as well as corticotropin releasing factor (CRF)/CRF receptor signaling were found to be important regulators of drinking behaviors across brain regions and drinking paradigms. Future research will require that females and a variety of rodent strains are used in preclinical experiments in order to strengthen the generalizability of findings and improve the likelihood of success for testing potential therapeutics in human laboratory studies.
Collapse
Affiliation(s)
- Marissa B Borrego
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Amy E Chan
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA
| | - Angela R Ozburn
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR, 97239, USA.
| |
Collapse
|
12
|
Synaptic effects of IL-1β and CRF in the central amygdala after protracted alcohol abstinence in male rhesus macaques. Neuropsychopharmacology 2022; 47:847-856. [PMID: 34837077 PMCID: PMC8882167 DOI: 10.1038/s41386-021-01231-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/14/2021] [Accepted: 11/02/2021] [Indexed: 12/21/2022]
Abstract
A major barrier to remission from an alcohol use disorder (AUD) is the continued risk of relapse during abstinence. Assessing the neuroadaptations after chronic alcohol and repeated abstinence is important to identify mechanisms that may contribute to relapse. In this study, we used a rhesus macaque model of long-term alcohol use and repeated abstinence, providing a platform to extend mechanistic findings from rodents to primates. The central amygdala (CeA) displays elevated GABA release following chronic alcohol in rodents and in abstinent male macaques, highlighting this neuroadaptation as a conserved mechanism that may underlie excessive alcohol consumption. Here, we determined circulating interleukin-1β (IL-1β) levels, CeA transcriptomic changes, and the effects of IL-1β and corticotropin releasing factor (CRF) signaling on CeA GABA transmission in male controls and abstinent drinkers. While no significant differences in peripheral IL-1β or the CeA transcriptome were observed, pathway analysis identified several canonical immune-related pathways. We addressed this potential dysregulation of CeA immune signaling in abstient drinkers with an electrophysiological approach. We found that IL-1β decreased CeA GABA release in controls while abstinent drinkers were less sensitive to IL-1β's effects, suggesting adaptations in the neuromodulatory role of IL-1β. In contrast, CRF enhanced CeA GABA release similarly in controls and abstinent drinkers, consistent with rodent studies. Notably, CeA CRF expression was inversely correlated with intoxication, suggesting that CRF levels during abstinence may predict future intoxication. Together, our findings highlight conserved and divergent actions of chronic alcohol on neuroimmune and stress signaling on CeA GABA transmission across rodents and macaques.
Collapse
|
13
|
Astrocytes play a critical role in mediating the effect of acute ethanol on central amygdala glutamatergic transmission. Neuropharmacology 2022; 205:108918. [PMID: 34896402 PMCID: PMC8792276 DOI: 10.1016/j.neuropharm.2021.108918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 11/21/2022]
Abstract
The Central Amygdala (CeA) has been heavily implicated in many aspects of alcohol use disorder. Ethanol (EtOH) has been shown to modulate glutamatergic transmission in the lateral subdivision of the CeA, however, the exact mechanism of this modulation is still unclear. EtOH exposure is associated with increased pro-inflammatory cytokines in the CeA, and inhibition of neuroimmune cells (microglia and astrocytes) has previously been shown to reduce EtOH drinking in animal models. Since neuroimmune activation seems to be involved in many of the effects of EtOH, we hypothesized that acute EtOH exposure will increase excitatory glutamatergic transmission in the CeA via modulation of neuroimmune cells. Using ex vivo brain slice whole-cell patch clamp electrophysiology, it was found that a physiologically relevant concentration of EtOH (20 mM) significantly increased presynaptic glutamatergic transmission in the CeA. Pharmacologic and chemogenetic inhibition of astrocyte function significantly reduced the ability of EtOH to modulate CeA glutamatergic transmission with minimal impact of microglia inhibition. This finding prompted additional studies examining whether direct neuroimmune activation through lipopolysaccharide (LPS) might lead to an increase in the glutamatergic transmission in the CeA. It was found that LPS modulation of glutamatergic transmission was limited by microglia activation and required astrocyte signaling. Taken together these results support the hypothesis that acute EtOH enhances lateral CeA glutamatergic transmission through an astrocyte mediated mechanism.
Collapse
|
14
|
Wang Z, Zhu X, Ni X, Wen Y, Shang D. Knowledge atlas of the involvement of glutamate and GABA in alcohol use disorder: A bibliometric and scientometric analysis. Front Psychiatry 2022; 13:965142. [PMID: 36032235 PMCID: PMC9411946 DOI: 10.3389/fpsyt.2022.965142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Abnormal neurotransmission of glutamate and γ-aminobutyric acid (GABA) is a key characteristic of alcohol-related disorders. To track research output, we conducted a bibliometric analysis to explore the current status and trends in this field over the past decades. METHODS Studies related to neurotransmitters and alcohol use disorder published in English from 2005-2021 were retrieved from the Web of Science Core Collection and Scopus databases. The R-bibliometrix package was used for a descriptive analysis of the publications. Citespace, WOSviewer, and R-bibliometrix were used to construct networks of countries/institutions/authors based on co-authorship, co-citation analysis of cited references and co-occurrence as well as burst detection of keywords. RESULTS A total of 4,250 unique articles and reviews were included in the final analysis. The annual growth rate of publications was 5.4%. The USA was the most productive country in this field, contributing nearly half of the total documents. The top ten most productive institutions were all located in the USA. The most frequent worldwide collaboration was between the USA and Italy. The most productive and influential institution was the University of California. The author contributing the most productions to this field was Marisa Roberto from the Scripps Research Institute. The top co-cited reference was a review titled "Neurocircuitry of addiction." The top journal in terms of the number of records and citations was Alcoholism: Clinical and Experimental Research. Comprehensive analyses have been conducted over past decades based on co-cited reference analysis, including modulators, transporters, receptor subtypes, and animal models. In recent years, the research frontiers have been shifting to the identification of risk factors/biomarkers, drug development for alcohol use disorder, and mechanisms related to alcoholic and non-alcoholic fatty liver. CONCLUSION Our bibliometric analysis shows that glutamate and GABA continue to be of interest in alcohol use disorder. The focus has evolved from mechanisms and medications related to glutamate and GABA in alcohol use disorder, to novel drug development, risk factor/biomarker identification targeting neurotransmitters, and the mechanisms of related diseases.
Collapse
Affiliation(s)
- Zhanzhang Wang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiuqing Zhu
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaojia Ni
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuguan Wen
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dewei Shang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
15
|
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
|
16
|
Nwachukwu KN, Evans WA, Sides TR, Trevisani CP, Davis A, Marshall SA. Chemogenetic manipulation of astrocytic signaling in the basolateral amygdala reduces binge-like alcohol consumption in male mice. J Neurosci Res 2021; 99:1957-1972. [PMID: 33844860 DOI: 10.1002/jnr.24841] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 03/21/2021] [Indexed: 12/18/2022]
Abstract
Binge drinking is a common occurrence in the United States, but a high concentration of alcohol in the blood has been shown to have reinforcing and reciprocal effects on the neuroimmune system in both dependent and non-dependent scenarios. The first part of this study examined alcohol's effects on the astrocytic response in the central amygdala and basolateral amygdala (BLA) in a non-dependent model. C57BL/6J mice were given access to either ethanol, water, or sucrose during a "drinking in the dark" paradigm, and astrocyte number and astrogliosis were measured using immunohistochemistry. Results indicate that non-dependent consumption increased glial fibrillary acidic protein (GFAP) density but not the number of GFAP+ cells, suggesting that non-dependent ethanol is sufficient to elicit astrocyte activation. The second part of this study examined how astrocytes impacted behaviors and the neurochemistry related to alcohol using the chemogenetic tool, DREADDs (designer receptors exclusively activated by designer drugs). Transgenic GFAP-hM3Dq mice were administered clozapine N-oxide both peripherally, affecting the entire central nervous system (CNS), or directly into the BLA. In both instances, GFAP-Gq-signaling activation significantly reduced ethanol consumption and correlating blood ethanol concentrations. However, GFAP-Gq-DREADD activation throughout the CNS had more broad effects resulting in decreased locomotor activity and sucrose consumption. More targeted GFAP-Gq-signaling activation in the BLA only impacted ethanol consumption. Finally, a glutamate assay revealed that after GFAP-Gq-signaling activation glutamate concentrations in the amygdala were partially normalized to control levels. Altogether, these studies support the theory that astrocytes represent a viable target for alcohol use disorder therapies.
Collapse
Affiliation(s)
- Kala N Nwachukwu
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, USA
| | - William A Evans
- Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, NC, USA
| | - Tori R Sides
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, USA
| | - Christopher P Trevisani
- Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, NC, USA
| | - Ambryia Davis
- Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, NC, USA
| | - S Alex Marshall
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, USA.,Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, NC, USA.,Department of Psychology & Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
17
|
Warden AS, Wolfe SA, Khom S, Varodayan FP, Patel RR, Steinman MQ, Bajo M, Montgomery SE, Vlkolinsky R, Nadav T, Polis I, Roberts AJ, Mayfield RD, Harris RA, Roberto M. Microglia Control Escalation of Drinking in Alcohol-Dependent Mice: Genomic and Synaptic Drivers. Biol Psychiatry 2020; 88:910-921. [PMID: 32680583 PMCID: PMC7674270 DOI: 10.1016/j.biopsych.2020.05.011] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 04/10/2020] [Accepted: 05/06/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Microglia, the primary immune cells of the brain, are implicated in alcohol use disorder. However, it is not known if microglial activation contributes to the transition from alcohol use to alcohol use disorder or is a consequence of alcohol intake. METHODS We investigated the role of microglia in a mouse model of alcohol dependence using a colony stimulating factor 1 receptor inhibitor (PLX5622) to deplete microglia and a chronic intermittent ethanol vapor two-bottle choice drinking procedure. Additionally, we examined anxiety-like behavior during withdrawal. We then analyzed synaptic neuroadaptations in the central nucleus of the amygdala (CeA) and gene expression changes in the medial prefrontal cortex and CeA from the same animals used for behavioral studies. RESULTS PLX5622 prevented escalations in voluntary alcohol intake and decreased anxiety-like behavior associated with alcohol dependence. PLX5622 also reversed expression changes in inflammatory-related genes and glutamatergic and GABAergic (gamma-aminobutyric acidergic) genes in the medial prefrontal cortex and CeA. At the cellular level in these animals, microglia depletion reduced inhibitory GABAA and excitatory glutamate receptor-mediated synaptic transmission in the CeA, supporting the hypothesis that microglia regulate dependence-induced changes in neuronal function. CONCLUSIONS Our multifaceted approach is the first to link microglia to the molecular, cellular, and behavioral changes associated with the development of alcohol dependence, suggesting that microglia may also be critical for the development and progression of alcohol use disorder.
Collapse
Affiliation(s)
- Anna S Warden
- Waggoner Center for Alcoholism and Addiction Research, University of Texas at Austin, Austin, Texas; Institute for Neuroscience, University of Texas at Austin, Austin, Texas
| | - Sarah A Wolfe
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Sophia Khom
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Florence P Varodayan
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Reesha R Patel
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Michael Q Steinman
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Michal Bajo
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Sarah E Montgomery
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Roman Vlkolinsky
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Tali Nadav
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Ilham Polis
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Amanda J Roberts
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California
| | - R Dayne Mayfield
- Waggoner Center for Alcoholism and Addiction Research, University of Texas at Austin, Austin, Texas; Institute for Neuroscience, University of Texas at Austin, Austin, Texas
| | - R Adron Harris
- Waggoner Center for Alcoholism and Addiction Research, University of Texas at Austin, Austin, Texas; Institute for Neuroscience, University of Texas at Austin, Austin, Texas
| | - Marisa Roberto
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California.
| |
Collapse
|
18
|
Smith LC, Kimbrough A. Leveraging Neural Networks in Preclinical Alcohol Research. Brain Sci 2020; 10:E578. [PMID: 32825739 PMCID: PMC7565429 DOI: 10.3390/brainsci10090578] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/25/2022] Open
Abstract
Alcohol use disorder is a pervasive healthcare issue with significant socioeconomic consequences. There is a plethora of neural imaging techniques available at the clinical and preclinical level, including magnetic resonance imaging and three-dimensional (3D) tissue imaging techniques. Network-based approaches can be applied to imaging data to create neural networks that model the functional and structural connectivity of the brain. These networks can be used to changes to brain-wide neural signaling caused by brain states associated with alcohol use. Neural networks can be further used to identify key brain regions or neural "hubs" involved in alcohol drinking. Here, we briefly review the current imaging and neurocircuit manipulation methods. Then, we discuss clinical and preclinical studies using network-based approaches related to substance use disorders and alcohol drinking. Finally, we discuss how preclinical 3D imaging in combination with network approaches can be applied alone and in combination with other approaches to better understand alcohol drinking.
Collapse
Affiliation(s)
- Lauren C. Smith
- Department of Psychiatry, School of Medicine, University of California San Diego, MC 0667, La Jolla, CA 92093, USA;
| | - Adam Kimbrough
- Department of Psychiatry, School of Medicine, University of California San Diego, MC 0667, La Jolla, CA 92093, USA;
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN 47907, USA
| |
Collapse
|
19
|
Abstract
The pervasive and devastating nature of substance use disorders underlies the need for the continued development of novel pharmacotherapies. We now know that glia play a much greater role in neuronal processes than once believed. The various types of glial cells (e.g., astrocytes, microglial, oligodendrocytes) participate in numerous functions that are crucial to healthy central nervous system function. Drugs of abuse have been shown to interact with glia in ways that directly contribute to the pharmacodynamic effects responsible for their abuse potential. Through their effect upon glia, drugs of abuse also alter brain function resulting in behavioral changes associated with substance use disorders. Therefore, drug-induced changes in glia and inflammation within the central nervous system (neuroinflammation) have been investigated to treat various aspects of drug abuse and dependence. This article presents a brief overview of the effects of each of the major classes of addictive drugs on glia. Next, the paper reviews the pre-clinical and clinical studies assessing the effects that glial modulators have on abuse-related behavioral effects, such as pleasure, withdrawal, and motivation. There is a strong body of pre-clinical literature demonstrating the general effectiveness of several glia-modulating drugs in models of reward and relapse. Clinical studies have also yielded promising results, though not as robust. There is still much to disentangle regarding the integration between addictive drugs and glial cells. Improved understanding of the relationship between glia and the pathophysiology of drug abuse should allow for more precise exploration in the development and testing of glial-directed treatments for substance use disorders.
Collapse
Affiliation(s)
- Jermaine D. Jones
- Division on Substance Use Disorders, New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|
20
|
Zhang Y, Han Y, Chen Z, Zhao D, Xia Q. Inhibition of EZH2 attenuates inhibitory synaptic transmission via the pro-inflammatory pathway in rats. Neuropharmacology 2020; 171:108101. [PMID: 32298702 DOI: 10.1016/j.neuropharm.2020.108101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 04/01/2020] [Accepted: 04/05/2020] [Indexed: 11/24/2022]
Abstract
Enhancer of zeste homolog 2 (EZH2), a subunit of the polycomb repressive complex 2 (PRC2), is associated with seizure development and epileptogenesis, however, the underlying mechanism of the process remains to be elucidated. This study focused on exploring whether EZH2 regulated gamma-aminobutyric acid (GABA)-mediated neurotransmission during seizure generation. Hyperthermia-induced seizures were generated in Sprague-Dawley (SD) rats using a hot (43.5 °C) bath method, and seizure severity was evaluated according to the Racine scale. The effect of treatment with the EZH2 pharmacological inhibitor GSK 126 on the GABA and pro-inflammatory cytokine levels was tested using enzyme-linked immunosorbent assay (ELISA). Miniature inhibitory postsynaptic currents (mIPSCs) were recorded using whole-cell patch clamp. In this study, our results showed that intracerebroventricular (i.c.v) injection of the EZH2 pharmacological inhibitor GSK 126 (10 nM) increased seizure severity and shortened seizure latency in a rat model of FS, and these effects were accompanied by reduced GABA content. Furthermore, GSK 126 (1 μM) treatment decreased the mean amplitude and frequency of the mIPSCs in cultured hippocampal neurons subjected to hyperthermia. Importantly, the same results were also obtained in cultured neurons infected with lentivirus carrying EZH2 shRNA. In addition, a significant increase in the pro-inflammatory cytokine (IL-1β and TNF-α) levels was observed in rats after GSK 126 treatment, and IL-1β administration increased seizure severity, suggesting that the inflammatory response was involved in the regulation of seizure development by EZH2. This study helps clarify the role of EZH2 in FS and supports EZH2 administration as an effective target for the management of seizure generation.
Collapse
Affiliation(s)
- Yusong Zhang
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Provincial Cancer Hospital, Zhengzhou, 450008, China.
| | - Yadi Han
- Department of Clinical Laboratory, Affiliated Cancer Hospital of Zhengzhou University, Henan Provincial Cancer Hospital, Zhengzhou, 450008, China
| | - Zhiguo Chen
- Xinxiang Medical University, Xinxiang, 453003, China
| | - Dongmei Zhao
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Provincial Cancer Hospital, Zhengzhou, 450008, China
| | - Qingxin Xia
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Provincial Cancer Hospital, Zhengzhou, 450008, China
| |
Collapse
|
21
|
Munshi S, Loh MK, Ferrara N, DeJoseph MR, Ritger A, Padival M, Record MJ, Urban JH, Rosenkranz JA. Repeated stress induces a pro-inflammatory state, increases amygdala neuronal and microglial activation, and causes anxiety in adult male rats. Brain Behav Immun 2020; 84:180-199. [PMID: 31785394 PMCID: PMC7010555 DOI: 10.1016/j.bbi.2019.11.023] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/25/2022] Open
Abstract
A link exists between immune function and psychiatric conditions, particularly depressive and anxiety disorders. Psychological stress is a powerful trigger for these disorders and stress influences immune state. However, the nature of peripheral immune changes after stress conflicts across studies, perhaps due to the focus on few measures of pro-inflammatory or anti-inflammatory processes. The basolateral amygdala (BLA) is critical for emotion, and plays an important role in the effects of stress on anxiety. As such, it may be a primary central nervous system (CNS) mediator for the effects of peripheral immune changes on anxiety after stress. Therefore, this study aimed to delineate the influence of stress on peripheral pro-inflammatory and anti-inflammatory aspects, BLA immune activation, and its impact on BLA neuronal activity. To produce a more encompassing view of peripheral immune changes, this study used a less restrictive approach to categorize and group peripheral immune changes. We found that repeated social defeat stress in adult male Sprague-Dawley rats increased the frequencies of mature T-cells positive for intracellular type 2-like cytokine and serum pro-inflammatory cytokines. Principal component analysis and hierarchical clustering was used to guide grouping of T-cells and cytokines, producing unique profiles. Stress shifted the balance towards a specific set that included mostly type 2-like T-cells and pro-inflammatory cytokines. Within the CNS component, repeated stress caused an increase of activated microglia in the BLA, increased anxiety-like behaviors across several assays, and increased BLA neuronal firing in vivo that was prevented by blockade of microglia activation. Because repeated stress can trigger anxiety states by actions in the BLA, and altered immune function can trigger anxiety, these results suggest that repeated stress may trigger anxiety-like behaviors by inducing a pro-inflammatory state in the periphery and the BLA. These results begin to uncover how stress may recruit the immune system to alter the function of brain regions critical to emotion.
Collapse
Affiliation(s)
- Soumyabrata Munshi
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Department of Foundational Sciences and Humanities, Neuroscience, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Maxine K. Loh
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Nicole Ferrara
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - M. Regina DeJoseph
- Department of Foundational Sciences and Humanities, Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Alexandra Ritger
- Department of Foundational Sciences and Humanities, Neuroscience, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Mallika Padival
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Matthew J. Record
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Janice H. Urban
- Department of Foundational Sciences and Humanities, Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - J. Amiel Rosenkranz
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Corresponding Author: J. Amiel Rosenkranz, Ph.D., Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA., Telephone: 847-578-8680; Fax: 847-578-3268,
| |
Collapse
|
22
|
Bajo M, Patel RR, Hedges DM, Varodayan FP, Vlkolinsky R, Davis TD, Burkart MD, Blednov YA, Roberto M. Role of MyD88 in IL-1β and Ethanol Modulation of GABAergic Transmission in the Central Amygdala. Brain Sci 2019; 9:brainsci9120361. [PMID: 31817854 PMCID: PMC6956324 DOI: 10.3390/brainsci9120361] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 12/15/2022] Open
Abstract
Myeloid differentiation primary response protein (MyD88) is a critical neuroimmune adaptor protein in TLR (Toll-like receptor) and IL-1R (Interleukin-1 receptor) signaling complexes. These two pro-inflammatory families play an important role in the neurobiology of alcohol use disorder, specifically MyD88 regulates ethanol drinking, ethanol-induced sedation, and ethanol-induced deficits in motor coordination. In this study, we examined the role of MyD88 in mediating the effects of IL-1β and ethanol on GABAergic transmission in the central amygdala (CeA) of male mice using whole-cell patch-clamp recordings in combination with pharmacological (AS-1, a mimetic that prevents MyD88 recruitment by IL-1R) and genetic (Myd88 knockout mice) approaches. We demonstrate through both approaches that IL-1β and ethanol’s modulatory effects at CeA GABA synapses are not dependent on MyD88. Myd88 knockout potentiated IL-1β’s actions in reducing postsynaptic GABAA receptor function. Pharmacological inhibition of MyD88 modulates IL-1β’s action at CeA GABA synapses similar to Myd88 knockout mice. Additionally, ethanol-induced CeA GABA release was greater in Myd88 knockout mice compared to wildtype controls. Thus, MyD88 is not essential to IL-1β or ethanol regulation of CeA GABA synapses but plays a role in modulating the magnitude of their effects, which may be a potential mechanism by which it regulates ethanol-related behaviors.
Collapse
Affiliation(s)
- Michal Bajo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA (D.M.H.); (F.P.V.); (R.V.); (M.R.)
- Correspondence: ; Tel.: +1-858-784-7259
| | - Reesha R. Patel
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA (D.M.H.); (F.P.V.); (R.V.); (M.R.)
| | - David M. Hedges
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA (D.M.H.); (F.P.V.); (R.V.); (M.R.)
| | - Florence P. Varodayan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA (D.M.H.); (F.P.V.); (R.V.); (M.R.)
| | - Roman Vlkolinsky
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA (D.M.H.); (F.P.V.); (R.V.); (M.R.)
| | - Tony D. Davis
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA; (T.D.D.); (M.D.B.)
| | - Michael D. Burkart
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA; (T.D.D.); (M.D.B.)
| | - Yuri A. Blednov
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA (D.M.H.); (F.P.V.); (R.V.); (M.R.)
| |
Collapse
|
23
|
Roberts AJ, Khom S, Bajo M, Vlkolinsky R, Polis I, Cates-Gatto C, Roberto M, Gruol DL. Increased IL-6 expression in astrocytes is associated with emotionality, alterations in central amygdala GABAergic transmission, and excitability during alcohol withdrawal. Brain Behav Immun 2019; 82:188-202. [PMID: 31437534 PMCID: PMC6800653 DOI: 10.1016/j.bbi.2019.08.185] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 08/16/2019] [Accepted: 08/17/2019] [Indexed: 01/14/2023] Open
Abstract
Accumulating evidence from preclinical and clinical studies has implicated a role for the cytokine IL-6 in a variety of CNS diseases including anxiety-like and depressive-like behaviors, as well as alcohol use disorder. Here we use homozygous and heterozygous transgenic mice expressing elevated levels of IL-6 in the CNS due to increased astrocyte expression and non-transgenic littermates to examine a role for astrocyte-produced IL-6 in emotionality (response to novelty, anxiety-like, and depressive-like behaviors). Our results from homozygous IL-6 mice in a variety of behavioral tests (light/dark transfer, open field, digging, tail suspension, and forced swim tests) support a role for IL-6 in stress-coping behaviors. Ex vivo electrophysiological studies of neuronal excitability and inhibitory GABAergic synaptic transmission in the central nucleus of the amygdala (CeA) of the homozygous transgenic mice revealed increased inhibitory GABAergic signaling and increased excitability of CeA neurons, suggesting a role for astrocyte produced IL-6 in the amygdala in exploratory drive and depressive-like behavior. Furthermore, studies in the hippocampus of activation/expression of proteins associated with IL-6 signal transduction and inhibitory GABAergic mechanisms support a role for astrocyte produced IL-6 in depressive-like behaviors. Our studies indicate a complex and dose-dependent relationship between IL-6 and behavior and implicate IL-6 induced neuroadaptive changes in neuronal excitability and the inhibitory GABAergic system as important contributors to altered behavior associated with IL-6 expression in the CNS.
Collapse
Affiliation(s)
- Amanda J. Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA 92037 U.S.A
| | - Sophia Khom
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037 U.S.A
| | - Michal Bajo
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037 U.S.A
| | - Roman Vlkolinsky
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037 U.S.A
| | - Ilham Polis
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA 92037 U.S.A
| | - Chelsea Cates-Gatto
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA 92037 U.S.A
| | - Marisa Roberto
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037 U.S.A
| | - Donna L. Gruol
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA 92037 U.S.A,Corresponding Author: Dr. Donna L. Gruol, Neuroscience Department, SP30-1522, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, Phone: (858) 784-7060, Fax: (858) 784-7393,
| |
Collapse
|
24
|
Centanni SW, Bedse G, Patel S, Winder DG. Driving the Downward Spiral: Alcohol-Induced Dysregulation of Extended Amygdala Circuits and Negative Affect. Alcohol Clin Exp Res 2019; 43:2000-2013. [PMID: 31403699 PMCID: PMC6779502 DOI: 10.1111/acer.14178] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/07/2019] [Indexed: 12/17/2022]
Abstract
Alcohol use disorder (AUD) afflicts a large number of individuals, families, and communities globally. Affective disturbances, including stress, depression, and anxiety, are highly comorbid with AUD, contributing in some cases to initial alcohol use and continued use. Negative affect has a particularly strong influence on the withdrawal/abstinence stage of addiction as individuals with AUD frequently report stressful events, depression, and anxiety as key factors for relapse. Treatment options for negative affect associated with AUD are limited and often ineffective, highlighting the pressing need for preclinical studies examining the underlying neural circuitry driving AUD-associated negative affect. The extended amygdala (EA) is a set of brain areas collectively involved in generating and regulating affect, and extensive research has defined a critical role for the EA in all facets of substance use disorder. Here, we review the expansive historical literature examining the effects of ethanol exposure on the EA, with an emphasis on the complex EA neural circuitry driving negative affect in all phases of the alcohol addiction cycle. Specifically, this review focuses on the effects of alcohol exposure on the neural circuitry in 2 key components of the EA, the central nucleus of the amygdala and the bed nucleus of the stria terminalis. Additionally, future directions are proposed to advance our understanding of the relationship between AUD-associated negative affect and neural circuitry in the EA, with the long-term goal of developing better diagnostic tools and new pharmacological targets aimed at treating negative affect in AUD. The concepts detailed here will serve as the foundation for a companion review focusing on the potential for the endogenous cannabinoid system in the EA as a novel target for treating the stress, anxiety, and negative emotional state driving AUD.
Collapse
Affiliation(s)
- Samuel W. Centanni
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
| | - Gaurav Bedse
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Sachin Patel
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Danny G. Winder
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| |
Collapse
|
25
|
Abstract
Innate immune signaling is an important feature in the pathology of alcohol use disorders. Alcohol abuse causes persistent innate immune activation in the brain. This is seen in postmortem human alcoholic brain specimens, as well as in primate and rodent models of alcohol consumption. Further, in vitro models of alcohol exposure in neurons and glia also demonstrate innate immune activation. The activation of the innate immune system seems to be important in the development of alcohol use pathology, as anti-immune therapies reduce pathology and ethanol self-administration in rodent models. Further, innate immune activation has been identified in each of the stages of addiction: binge/intoxication, withdrawal/negative affect, and preoccupation/craving. This suggests that innate immune activation may play a role both in the development and maintenance of alcoholic pathology. In this chapter, we discuss the known contributions of innate immune signaling in the pathology of alcohol use disorders, and present potential therapeutic interventions that may be beneficial for alcohol use disorders.
Collapse
Affiliation(s)
- Leon G Coleman
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Fulton T Crews
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
26
|
Abstract
The innate immune system plays a critical role in the ethanol-induced neuroimmune response in the brain. Ethanol initiates the innate immune response via activation of the innate immune receptors Toll-like receptors (TLRs, e.g., TLR4, TLR3, TLR7) and NOD-like receptors (inflammasome NLRs) leading to a release of a plethora of chemokines and cytokines and development of the innate immune response. Cytokines and chemokines can have pro- or anti-inflammatory properties through which they regulate the immune response. In this chapter, we will focus on key cytokines (e.g., IL-1, IL-6, TNF-α) and chemokines (e.g., MCP-1/CCL2) that mediate the ethanol-induced neuroimmune responses. In this regard, we will use IL-1β, as an example cytokine, to discuss the neuromodulatory properties of cytokines on cellular properties and synaptic transmission. We will discuss their involvement through a set of evidence: (1) changes in gene and protein expression following ethanol exposure, (2) association of gene polymorphisms (humans) and alterations in gene expression (animal models) with increased alcohol intake, and (3) modulation of alcohol-related behaviors by transgenic or pharmacological manipulations of chemokine and cytokine systems. Over the last years, our understanding of the molecular mechanisms mediating cytokine- and chemokine-dependent regulation of immune responses has advanced tremendously, and we review evidence pointing to cytokines and chemokines serving as neuromodulators and regulators of neurotransmission.
Collapse
Affiliation(s)
- Marisa Roberto
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA.
| | - Reesha R Patel
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Michal Bajo
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
27
|
Grifasi IR, McIntosh SE, Thomas RD, Lysle DT, Thiele TE, Marshall SA. Characterization of the Hippocampal Neuroimmune Response to Binge-Like Ethanol Consumption in the Drinking in the Dark Model. Neuroimmunomodulation 2019; 26:19-32. [PMID: 30625475 PMCID: PMC6389401 DOI: 10.1159/000495210] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 11/08/2018] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVES Alcohol dependence leads to dysregulation of the neuroimmune system, but the effects of excessive alcohol consumption on key players of the neuroimmune response after episodic binge drinking in nondependence has not been readily assessed. These studies seek to determine how the neuroimmune system within the hippocampus responds to binge-like consumption prior to dependence or evidence of brain damage. METHODS C57BL/6J mice underwent the drinking in the dark (DID) paradigm to recapitulate binge consumption. Immunohistochemical techniques were employed to determine the effects of ethanol on cytokine and astrocyte responses within the hippocampus. Astrocyte activation was also assessed using qRT-PCR. RESULTS Our results indicated that binge-like ethanol consumption resulted in a 3.6-fold increase in the proinflammatory cytokine interleukin (IL)-1β immunoreactivity in various regions of the hippocampus. The opposite effect was seen in the anti-inflammatory cytokine IL-10. Binge-like consumption resulted in a 67% decrease in IL-10 immunoreactivity but had no effect on IL-4 or IL-6 compared with the water-drinking control group. Moreover, astrocyte activation occurred following ethanol exposure as GFAP immunoreactivity was increased over 120% in mice that experienced 3 cycles of ethanol binges. PCR analyses indicated that the mRNA increased by almost 4-fold after one cycle of DID, but this effect did not persist in abstinence. CONCLUSIONS Altogether, these findings suggest that binge-like ethanol drinking prior to dependence causes dysregulation to the neuroimmune system. This altered neuroimmune state may have an impact on behavior but could also result in a heightened neuroimmune response that is exacerbated from further ethanol exposure or other immune-modulating events.
Collapse
Affiliation(s)
- Isabella R Grifasi
- Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, North Carolina, USA
| | - Scot E McIntosh
- Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, North Carolina, USA
| | - Rhiannon D Thomas
- Department of Psychology and Neuroscience, The University of North Carolina, Chapel Hill, North Carolina, USA
| | - Donald T Lysle
- Department of Psychology and Neuroscience, The University of North Carolina, Chapel Hill, North Carolina, USA
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, North Carolina, USA
| | - Todd E Thiele
- Department of Psychology and Neuroscience, The University of North Carolina, Chapel Hill, North Carolina, USA
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, North Carolina, USA
| | - S Alex Marshall
- Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, North Carolina, USA,
- Department of Psychology and Neuroscience, The University of North Carolina, Chapel Hill, North Carolina, USA,
| |
Collapse
|
28
|
A comparison of hippocampal microglial responses in aged and young rodents following dependent and non-dependent binge drinking. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 148:305-343. [PMID: 31733666 PMCID: PMC9875180 DOI: 10.1016/bs.irn.2019.10.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Alcoholism is a highly visible and prevalent issue in the United States. Although binge-drinking is assumed to be a college-age problem, older adults (ages 65+) consume binge amounts of alcohol and have alcohol use disorders (AUDs). Moreover, individuals with alcohol dependence in their youth often continue to drink as they age. As such, this study tested the hypothesis that the effects of alcohol on hippocampal microglia are exacerbated in aged versus younger rodents in two AUD models. Briefly, adult (2-3 months) and aged (15+ months) Sprague-Dawley rats were administered alcohol or control diet using the Majchrowicz model to study alcohol-induced neurodegeneration. To study the effects of non-dependent binge consumption on microglia, adolescent (6-8 weeks) and aged (18+ months) C57/BL6N were subjected to the Drinking in the Dark paradigm. Microglia number and densitometry were assessed using immunohistochemistry. Hippocampal subregional and model/species-specific effects of alcohol were observed, but overall, aging did not appear to increase the alcohol-induced microglia reactivity as measured by Iba-1 densitometry. However, analysis of microglial counts revealed a significant decrease in the number microglia cells in both the alcohol-induced neurodegeneration and DID model across age groups. In the dentate gyrus, the loss of microglia was exacerbated by aging, particularly in mice after DID, non-dependent model. Using qRT-PCR, the persistence of alcohol and aging effects was assessed following the DID model. Allograft Inflammatory Factor 1 mRNA was increased in both young and aged mice by alcohol exposure; however, only in the aged mice did the alcohol effect persist. Overall, these data imply that the microglial response to alcohol is complex with evidence of depressed numbers of microglia but also increased reactivity with advanced age.
Collapse
|
29
|
Patel RR, Khom S, Steinman MQ, Varodayan FP, Kiosses WB, Hedges DM, Vlkolinsky R, Nadav T, Polis I, Bajo M, Roberts AJ, Roberto M. IL-1β expression is increased and regulates GABA transmission following chronic ethanol in mouse central amygdala. Brain Behav Immun 2019; 75:208-219. [PMID: 30791967 PMCID: PMC6383367 DOI: 10.1016/j.bbi.2018.10.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/09/2018] [Accepted: 10/25/2018] [Indexed: 12/20/2022] Open
Abstract
The interleukin-1 system (IL-1) is a prominent pro-inflammatory pathway responsible for the initiation and regulation of immune responses. Human genetic and preclinical studies suggest a critical role for IL-1β signaling in ethanol drinking and dependence, but little is known about the effects of chronic ethanol on the IL-1 system in addiction-related brain regions such as the central amygdala (CeA). In this study, we generated naïve, non-dependent (Non-Dep) and dependent (Dep) male mice using a paradigm of chronic-intermittent ethanol vapor exposure interspersed with two-bottle choice to examine 1) the expression of IL-1β, 2) the role of the IL-1 system on GABAergic transmission, and 3) the potential interaction with the acute effects of ethanol in the CeA. Immunohistochemistry with confocal microscopy was used to assess expression of IL-1β in microglia and neurons in the CeA, and whole-cell patch clamp recordings were obtained from CeA neurons to measure the effects of IL-1β (50 ng/ml) or the endogenous IL-1 receptor antagonist (IL-1ra; 100 ng/ml) on action potential-dependent spontaneous inhibitory postsynaptic currents (sIPSCs). Overall, we found that IL-1β expression is significantly increased in microglia and neurons of Dep compared to Non-Dep and naïve mice, IL-1β and IL-1ra bi-directionally modulate GABA transmission through both pre- and postsynaptic mechanisms in all three groups, and IL-1β and IL-1ra do not alter the facilitation of GABA release induced by acute ethanol. These data suggest that while ethanol dependence induces a neuroimmune response in the CeA, as indicated by increased IL-1β expression, this does not significantly alter the neuromodulatory role of IL-1β on synaptic transmission.
Collapse
Affiliation(s)
- Reesha R Patel
- The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Sophia Khom
- The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Michael Q Steinman
- The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Florence P Varodayan
- The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - William B Kiosses
- The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - David M Hedges
- The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Roman Vlkolinsky
- The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Tali Nadav
- The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Ilham Polis
- The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Michal Bajo
- The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Amanda J Roberts
- The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Marisa Roberto
- The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA.
| |
Collapse
|
30
|
Erickson EK, Grantham EK, Warden AS, Harris RA. Neuroimmune signaling in alcohol use disorder. Pharmacol Biochem Behav 2018; 177:34-60. [PMID: 30590091 DOI: 10.1016/j.pbb.2018.12.007] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/25/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023]
Abstract
Alcohol use disorder (AUD) is a widespread disease with limited treatment options. Targeting the neuroimmune system is a new avenue for developing or repurposing effective pharmacotherapies. Alcohol modulates innate immune signaling in different cell types in the brain by altering gene expression and the molecular pathways that regulate neuroinflammation. Chronic alcohol abuse may cause an imbalance in neuroimmune function, resulting in prolonged perturbations in brain function. Likewise, manipulating the neuroimmune system may change alcohol-related behaviors. Psychiatric disorders that are comorbid with AUD, such as post-traumatic stress disorder, major depressive disorder, and other substance use disorders, may also have underlying neuroimmune mechanisms; current evidence suggests that convergent immune pathways may be involved in AUD and in these comorbid disorders. In this review, we provide an overview of major neuroimmune cell-types and pathways involved in mediating alcohol behaviors, discuss potential mechanisms of alcohol-induced neuroimmune activation, and present recent clinical evidence for candidate immune-related drugs to treat AUD.
Collapse
Affiliation(s)
- Emma K Erickson
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA.
| | - Emily K Grantham
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA
| | - Anna S Warden
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA
| | - R A Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA
| |
Collapse
|
31
|
Vonder Haar C, Ferland JMN, Kaur S, Riparip LK, Rosi S, Winstanley CA. Cocaine self-administration is increased after frontal traumatic brain injury and associated with neuroinflammation. Eur J Neurosci 2018; 50:2134-2145. [PMID: 30118561 DOI: 10.1111/ejn.14123] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 07/30/2018] [Accepted: 08/07/2018] [Indexed: 12/18/2022]
Abstract
Traumatic brain injury (TBI) has been linked to the development of numerous psychiatric diseases, including substance use disorder. However, it can be difficult to ascertain from clinical data whether the TBI is cause or consequence of increased addiction vulnerability. Surprisingly few studies have taken advantage of animal models to investigate the causal nature of this relationship. In terms of a plausible neurobiological mechanism through which TBI could magnify the risk of substance dependence, numerous studies indicate that TBI can cause widespread disruption to monoaminergic signaling in striatal regions, and also increases neuroinflammation. In the current study, male Long-Evans rats received either a mild or severe TBI centered over the frontal cortex via controlled cortical impact, and were subsequently trained to self-administer cocaine over 10 6-hour sessions. At the end of the study, markers of striatal dopaminergic function, and levels of inflammatory cytokine levels in the frontal lobes, were assessed via western blot and multiplex ELISA, respectively. There was significantly higher cocaine intake in a subset of animals with either mild or severe TBI. However, many animals within both TBI groups failed to acquire self-administration. Principal components analysis suggested that both dopaminergic and neuroinflammatory proteins were associated with overall cocaine intake, yet only an inflammatory component was associated with acquisition of self-administration, suggesting neuroinflammation may make a more substantial contribution to the likelihood of drug-taking. Should neuroinflammation play a causal role in mediating TBI-induced addiction risk, anti-inflammatory therapy may reduce the likelihood of substance abuse in TBI populations.
Collapse
Affiliation(s)
- Cole Vonder Haar
- Injury and Recovery Laboratory, Department of Psychology, West Virginia University, PO Box 6040, 53 Campus Drive, Morgantown, WV, 26505, USA.,Laboratory of Molecular and Behavioural Neuroscience, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Jacqueline-Marie N Ferland
- Laboratory of Molecular and Behavioural Neuroscience, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Sukhbir Kaur
- Laboratory of Molecular and Behavioural Neuroscience, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Lara-Kirstie Riparip
- Brain and Spinal Injury Center, Departments of Physical Therapy Rehabilitation Science and Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Susanna Rosi
- Brain and Spinal Injury Center, Departments of Physical Therapy Rehabilitation Science and Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Catharine A Winstanley
- Laboratory of Molecular and Behavioural Neuroscience, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| |
Collapse
|
32
|
Seifi M, Rodaway S, Rudolph U, Swinny JD. GABA A Receptor Subtypes Regulate Stress-Induced Colon Inflammation in Mice. Gastroenterology 2018; 155:852-864.e3. [PMID: 29802853 DOI: 10.1053/j.gastro.2018.05.033] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 04/30/2018] [Accepted: 05/18/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Psychological stress, in early life or adulthood, is a significant risk factor for inflammatory disorders, including inflammatory bowel diseases. However, little is known about the mechanisms by which emotional factors affect the immune system. γ-Aminobutyric acid type A receptors (GABAARs) regulate stress and inflammation, but it is not clear whether specific subtypes of GABAARs mediate stress-induced gastrointestinal inflammation. We investigated the roles of different GABAAR subtypes in mouse colon inflammation induced by 2 different forms of psychological stress. METHODS C57BL/6J mice were exposed to early-life stress, and adult mice were exposed to acute-restraint stress; control mice were not exposed to either form of stress. We collected colon tissues and measured contractility using isometric tension recordings; colon inflammation, based on levels of cluster of differentiation 163 and tumor necrosis factor messenger RNA (mRNA) and protein and myeloperoxidase activity; and permeability, based on levels of tight junction protein 1 and occludin mRNA and protein. Mice were given fluorescently labeled dextran orally and systemic absorption was measured. We also performed studies of mice with disruption of the GABAAR subunit α3 gene (Gabra3-/- mice). RESULTS Mice exposed to early-life stress had significantly altered GABAAR-mediated colonic contractility and impaired barrier function, and their colon tissue had increased levels of Gabra3 mRNA compared with control mice. Restraint stress led to colon inflammation in C57/BL6J mice but not Gabra3-/- mice. Colonic inflammation was induced in vitro by an α3-GABAAR agonist, showing a proinflammatory role for this receptor subtype. In contrast, α1/4/5-GABAAR ligands decreased the expression of colonic inflammatory markers. CONCLUSIONS We found stress to increase expression of Gabra3 and induce inflammation in mouse colon, together with impaired barrier function. The in vitro pharmacologic activation of α3-GABAARs recapitulated colonic inflammation, whereas α1/4/5-GABAAR ligands were anti-inflammatory. These proteins might serve as therapeutic targets for treatment of colon inflammation or inflammatory bowel diseases.
Collapse
Affiliation(s)
- Mohsen Seifi
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Scott Rodaway
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Uwe Rudolph
- Laboratory of Genetic Neuropharmacology, McLean Hospital, Belmont, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Jerome D Swinny
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK.
| |
Collapse
|
33
|
Munshi S, Rosenkranz JA. Effects of Peripheral Immune Challenge on In Vivo Firing of Basolateral Amygdala Neurons in Adult Male Rats. Neuroscience 2018; 390:174-186. [PMID: 30170159 DOI: 10.1016/j.neuroscience.2018.08.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 08/07/2018] [Accepted: 08/20/2018] [Indexed: 12/15/2022]
Abstract
Peripheral inflammation often causes changes in mood and emergence of depressive behavior, and is characterized by a group of physical manifestations including lethargy, malaise, listlessness, decreased appetite, anhedonia, and fever. These behavioral changes are induced at the molecular level by pro-inflammatory cytokines like interleukin (IL)-1β, IL-6 and TNF-α. The basolateral amygdala (BLA) is a key brain region involved in mood and may mediate some of the behavioral effects of inflammation. However, it is unknown whether peripheral inflammatory state affects the activity of BLA neurons. To test this, adult male Sprague-Dawley rats were treated with IL-1β (1 μg, intraperitoneal (i.p.)), and behavioral and electrophysiological measures were obtained. IL-1β reduced locomotion in the open-field test and also reduced home-cage mobility, consistent with features of sickness-like behavior. Using in vivo single-unit extracellular electrophysiological recordings from anesthetized rats, we found that spontaneous BLA neuronal firing was acutely (<30 min) increased after IL-1β, followed by a return to baseline level, particularly in the basal nucleus of the BLA complex. To verify and expand on effects of peripheral inflammation, we tested whether another, long-lasting inflammagen also changes BLA neuronal firing. Lipopolysaccharide (250 μg/kg, i.p.) increased BLA firing rate acutely (<30 min) and persistently. The findings demonstrate a rapid effect of peripheral inflammation on BLA activity and suggest a link between BLA neuronal firing and triggering of behavioral consequences of peripheral inflammation. These findings are a first step toward understanding the neuronal basis of depressive behavior caused by acute peripheral inflammation.
Collapse
Affiliation(s)
- Soumyabrata Munshi
- Department of Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA; Department of Neuroscience, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - J Amiel Rosenkranz
- Department of Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA; Center for Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.
| |
Collapse
|
34
|
McCarthy GM, Warden AS, Bridges CR, Blednov YA, Harris RA. Chronic ethanol consumption: role of TLR3/TRIF-dependent signaling. Addict Biol 2018; 23:889-903. [PMID: 28840972 PMCID: PMC5828779 DOI: 10.1111/adb.12539] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 12/27/2022]
Abstract
Chronic ethanol consumption stimulates neuroimmune signaling in the brain, and Toll-like receptor (TLR) activation plays a key role in ethanol-induced inflammation. However, it is unknown which of the TLR signaling pathways, the myeloid differentiation primary response gene 88 (MyD88) dependent or the TIR-domain-containing adapter-inducing interferon-β (TRIF) dependent, is activated in response to chronic ethanol. We used voluntary (every-other-day) chronic ethanol consumption in adult C57BL/6J mice and measured expression of TLRs and their signaling molecules immediately following consumption and 24 hours after removing alcohol. We focused on the prefrontal cortex where neuroimmune changes are the most robust and also investigated the nucleus accumbens and amygdala. Tlr mRNA and components of the TRIF-dependent pathway (mRNA and protein) were increased in the prefrontal cortex 24 hours after ethanol and Cxcl10 expression increased 0 hour after ethanol. Expression of Tlr3 and TRIF-related components increased in the nucleus accumbens, but slightly decreased in the amygdala. In addition, we demonstrate that the IKKε/TBK1 inhibitor Amlexanox decreases immune activation of TRIF-dependent pathway in the brain and reduces ethanol consumption, suggesting the TRIF-dependent pathway regulates drinking. Our results support the importance of TLR3 and the TRIF-dependent pathway in ethanol-induced neuroimmune signaling and suggest that this pathway could be a target in the treatment of alcohol use disorders.
Collapse
Affiliation(s)
- Gizelle M. McCarthy
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX USA
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX USA
| | - Anna S. Warden
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX USA
- Insitute for Neuroscience, University of Texas at Austin, Austin, TX USA
| | - Courtney R. Bridges
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX USA
| | - Yuri A. Blednov
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX USA
| | - R. Adron Harris
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX USA
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX USA
- Insitute for Neuroscience, University of Texas at Austin, Austin, TX USA
| |
Collapse
|
35
|
Varodayan FP, Sidhu H, Kreifeldt M, Roberto M, Contet C. Morphological and functional evidence of increased excitatory signaling in the prelimbic cortex during ethanol withdrawal. Neuropharmacology 2018; 133:470-480. [PMID: 29471053 PMCID: PMC5865397 DOI: 10.1016/j.neuropharm.2018.02.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/26/2018] [Accepted: 02/16/2018] [Indexed: 02/06/2023]
Abstract
Excessive alcohol consumption in humans induces deficits in decision making and emotional processing, which indicates a dysfunction of the prefrontal cortex (PFC). The present study aimed to determine the impact of chronic intermittent ethanol (CIE) inhalation on mouse medial PFC pyramidal neurons. Data were collected 6-8 days into withdrawal from 7 weeks of CIE exposure, a time point when mice exhibit behavioral symptoms of withdrawal. We found that spine maturity in prelimbic (PL) layer 2/3 neurons was increased, while dendritic spines in PL layer 5 neurons or infralimbic (IL) neurons were not affected. Corroborating these morphological observations, CIE enhanced glutamatergic transmission in PL layer 2/3 pyramidal neurons, but not IL layer 2/3 neurons. Contrary to our predictions, these cellular alterations were associated with improved, rather than impaired, performance in reversal learning and strategy switching tasks in the Barnes maze at an earlier stage of chronic ethanol exposure (5-7 days withdrawal from 3 to 4 weeks of CIE), which could result from the anxiety-like behavior associated with ethanol withdrawal. Altogether, this study adds to a growing body of literature indicating that glutamatergic activity in the PFC is upregulated following chronic ethanol exposure, and identifies PL layer 2/3 pyramidal neurons as a sensitive target of synaptic remodeling. It also indicates that the Barnes maze is not suitable to detect deficits in cognitive flexibility in CIE-withdrawn mice.
Collapse
Affiliation(s)
| | - Harpreet Sidhu
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA
| | - Max Kreifeldt
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA
| | - Marisa Roberto
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA
| | - Candice Contet
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA.
| |
Collapse
|
36
|
Zahr NM. Peripheral TNFα elevations in abstinent alcoholics are associated with hepatitis C infection. PLoS One 2018; 13:e0191586. [PMID: 29408932 PMCID: PMC5800541 DOI: 10.1371/journal.pone.0191586] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 01/08/2018] [Indexed: 12/31/2022] Open
Abstract
Substantial evidence supports the view that inflammatory processes contribute to brain alterations in HIV infection. Mechanisms recently proposed to underlie neuropathology in Alcohol Use Disorder (AUD) include elevations in peripheral cytokines that sensitize the brain to the damaging effects of alcohol. This study included 4 groups: healthy controls, individuals with AUD (abstinent from alcohol at examination), those infected with HIV, and those comorbid for HIV and AUD. The aim was to determine whether inflammatory cytokines are elevated in AUD as they are in HIV infection. Cytokines showing group differences included interferon gamma-induced protein 10 (IP-10) and tumor necrosis factor α (TNFα). Follow-up t-tests revealed that TNFα and IP-10 were higher in AUD than controls but only in AUD patients who were seropositive for Hepatitis C virus (HCV). Specificity of TNFα and IP-10 elevations to HCV infection status was provided by correlations between cytokine levels and HCV viral load and indices of liver integrity including albumin/globulin ratio, fibrosis scores, and AST/platelet count ratio. Because TNFα levels were mediated by HCV infection, this study provides no evidence for elevations in peripheral cytokines in "uncomplicated", abstinent alcoholics, independent of liver disease or HCV infection. Nonetheless, these results corroborate evidence for elevations in IP-10 and TNFα in HIV and for IP-10 levels in HIV+HCV co-infection.
Collapse
Affiliation(s)
- Natalie M. Zahr
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States of America
- Neuroscience Department, SRI International, Menlo Park, CA, United States of America
- * E-mail:
| |
Collapse
|
37
|
Microglia and alcohol meet at the crossroads: Microglia as critical modulators of alcohol neurotoxicity. Toxicol Lett 2018; 283:21-31. [DOI: 10.1016/j.toxlet.2017.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 11/01/2017] [Accepted: 11/05/2017] [Indexed: 12/17/2022]
|
38
|
Brown KT, Levis SC, O’Neill CE, Northcutt AL, Fabisiak TJ, Watkins LR, Bachtell RK. Innate immune signaling in the ventral tegmental area contributes to drug-primed reinstatement of cocaine seeking. Brain Behav Immun 2018; 67:130-138. [PMID: 28813640 PMCID: PMC6252252 DOI: 10.1016/j.bbi.2017.08.012] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/20/2017] [Accepted: 08/11/2017] [Indexed: 02/05/2023] Open
Abstract
Cocaine addiction is a chronic relapsing disorder characterized by persistent perturbations to an organism's homeostatic processes that result in maladaptive drug seeking. Although considerable attention has been directed at the consequences of neuronal changes following chronic cocaine taking, few studies have examined the role of microglia, the brain's resident immune cells, following chronic cocaine administration. Toll-Like Receptor 4 (TLR4) is a molecular pattern receptor that recognizes pathogens, danger signals, and xenobiotics and induces proinflammatory signaling in the central nervous system. TLR4 is generally considered to be expressed primarily by microglia. Here, we used a rodent model of cocaine addiction to investigate the role of TLR4 in the ventral tegmental area (VTA) in cocaine seeking. Male Sprague-Dawley rats were trained to self-administer cocaine in daily 2-h sessions for 15days. Following self-administration, rats underwent extinction training and were tested in a drug-primed reinstatement paradigm. Pharmacological antagonism of TLR4 in the VTA using lipopolysaccharide from the bacterium Rhodobacter sphaeroides (LPS-RS) significantly reduced cocaine-primed reinstatement of drug seeking but had no effect on sucrose seeking. TLR4 activation within the VTA using the TLR4 activator, lipopolysaccharide, was sufficient to moderately reinstate cocaine seeking. We also assessed changes in proinflammatory cytokine expression in the VTA following cocaine self-administration. Cocaine self-administration increased the expression of mRNA for the proinflammatory cytokine interleukin-1ß, but not tumor necrosis factor alpha, in the VTA. Pharmacological antagonism of the interleukin-1 receptor in the VTA reduced cocaine-primed drug seeking. These results are consistent with the hypothesis that chronic cocaine produces inflammatory signaling that contributes to cocaine seeking.
Collapse
Affiliation(s)
- Kyle T. Brown
- Corresponding author at: Department of Psychology and Neuroscience, University of Colorado, Muenzinger Building, Boulder, CO 80309, United States., (K.T. Brown)
| | | | | | | | | | | | | |
Collapse
|
39
|
Marshall SA, McKnight KH, Blose AK, Lysle DT, Thiele TE. Modulation of Binge-like Ethanol Consumption by IL-10 Signaling in the Basolateral Amygdala. J Neuroimmune Pharmacol 2017; 12:249-259. [PMID: 27640210 PMCID: PMC5357210 DOI: 10.1007/s11481-016-9709-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/09/2016] [Indexed: 12/20/2022]
Abstract
Excessive ethanol consumption alters the neuroimmune system and particularly impacts the cytokine milieu of the CNS. Cytokine dysregulation has been shown to underlie addictive-like behaviors including alcohol abuse; however, many studies focus primarily on the proinflammatory cytokine profile during alcohol dependence. The current study furthers this research by determining the impact of excessive ethanol consumption on interleukin-10 (IL-10) and interleukin-4 (IL-4) activity in a model of non-dependent binge consumption called the "drinking in the dark" (DID) paradigm. Furthermore, the ability of IL-10 to modulate ethanol consumption was tested using site-directed pharmacology. Immunohistochemistry analyses determined that ethanol decreased IL-10 by 50 % in the basolateral amygdala (BLA) but had no effect on IL-4. Neither IL-10 nor IL-4, however, were altered in the central amygdala (CEA). Enzyme linked immunosorbent assays confirmed that IL-10 was decreased in the amygdala but not in the serum, suggesting changes of this cytokine with the DID paradigm are restricted to the central nervous system. Finally, bilateral infusions of IL-10 into the BLA, but not CeA, reduced binge-like drinking and corresponding blood ethanol concentrations without impacting either locomotor activity or anxiety-like behavioral correlates. Together, these data support the idea that alcohol abuse dysregulates specific anti-inflammatory cytokines; however, ameliorating alcohol-induced effects on cytokines, like IL-10, may prove to be an effective therapy in curbing excessive consumption.
Collapse
Affiliation(s)
- S Alex Marshall
- Department of Psychology & Neuroscience, The University of North Carolina, Davie Hall, CB# 3270, Chapel Hill, NC, 27599-3270, USA
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Kyle H McKnight
- Department of Psychology & Neuroscience, The University of North Carolina, Davie Hall, CB# 3270, Chapel Hill, NC, 27599-3270, USA
| | - Allyson K Blose
- Department of Psychology & Neuroscience, The University of North Carolina, Davie Hall, CB# 3270, Chapel Hill, NC, 27599-3270, USA
| | - Donald T Lysle
- Department of Psychology & Neuroscience, The University of North Carolina, Davie Hall, CB# 3270, Chapel Hill, NC, 27599-3270, USA
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Todd E Thiele
- Department of Psychology & Neuroscience, The University of North Carolina, Davie Hall, CB# 3270, Chapel Hill, NC, 27599-3270, USA.
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
40
|
Hutson LW, Lebonville CL, Jones ME, Fuchs RA, Lysle DT. Interleukin-1 signaling in the basolateral amygdala is necessary for heroin-conditioned immunosuppression. Brain Behav Immun 2017; 62:171-179. [PMID: 28131792 PMCID: PMC5828772 DOI: 10.1016/j.bbi.2017.01.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/14/2017] [Accepted: 01/23/2017] [Indexed: 12/22/2022] Open
Abstract
Heroin administration suppresses the production of inducible nitric oxide (NO), as indicated by changes in splenic inducible nitric oxide synthase (iNOS) and plasma nitrate/nitrite. Since NO is a measure of host defense against infection and disease, this provides evidence that heroin can increase susceptibility to pathogens by directly interacting with the immune system. Previous research in our laboratory has demonstrated that these immunosuppressive effects of heroin can also be conditioned to environmental stimuli by repeatedly pairing heroin administration with a unique environmental context. Re-exposure to a previously drug-paired context elicits immunosuppressive effects similar to heroin administration alone. In addition, our laboratory has reported that the basolateral amygdala (BLA) and medial nucleus accumbens shell (mNAcS) are critical neural substrates that mediate this conditioned effect. However, our understanding of the contributing mechanisms within these brain regions is limited. It is known that the cytokine interleukin-1 (IL-1) plays an important role in learning and memory. In fact, our laboratory has demonstrated that inhibition of IL-1β expression in the dorsal hippocampus (DH) prior to re-exposure to a heroin-paired context prevents the suppression of measures of NO production. Therefore, the present studies sought to further investigate the role of IL-1 in heroin-conditioned immunosuppression. Blockade of IL-1 signaling in the BLA, but not in the caudate putamen or mNAcS, using IL-1 receptor antagonist (IL-1Ra) attenuated heroin-conditioned immunosuppression of NO production as measured by plasma nitrate/nitrite and iNOS mRNA expression in spleen tissue. Taken together, these findings suggest that IL-1 signaling in the BLA is necessary for the expression of heroin-conditioned immunosuppression of NO production and may be a target for interventions that normalize immune function in heroin users and patient populations exposed to opiate regimens.
Collapse
Affiliation(s)
- Lee W Hutson
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, Chapel Hill, NC, USA
| | - Christina L Lebonville
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, Chapel Hill, NC, USA
| | - Meghan E Jones
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, Chapel Hill, NC, USA
| | - Rita A Fuchs
- Washington State University, College of Veterinary Medicine, Department of Integrative Physiology and Neuroscience, Pullman, WA, USA
| | - Donald T Lysle
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, Chapel Hill, NC, USA.
| |
Collapse
|
41
|
Blednov YA, Black M, Benavidez JM, Da Costa A, Mayfield J, Harris RA. Sedative and Motor Incoordination Effects of Ethanol in Mice Lacking CD14, TLR2, TLR4, or MyD88. Alcohol Clin Exp Res 2017; 41:531-540. [PMID: 28160299 PMCID: PMC5332292 DOI: 10.1111/acer.13314] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 12/14/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND In our companion article, we examined the role of MyD88-dependent signaling in ethanol (EtOH) consumption in mice lacking key components of this inflammatory pathway and observed differential effects on drinking. Here, we studied the role of these same signaling components in the acute sedative, intoxicating, and physiological effects of EtOH. Toll-like receptor 4 (TLR4) has been reported to strongly reduce the duration of EtOH-induced sedation, although most studies do not support its direct involvement in EtOH consumption. We examined TLR4 and other MyD88 pathway molecules to determine signaling specificity in acute EtOH-related behaviors. We also studied other GABAergic sedatives to gauge the EtOH specificity and potential role for GABA in EtOH's sedative and intoxicating effects in the mutant mice. METHODS Loss of righting reflex (LORR) and recovery from motor incoordination were studied following acute injection of EtOH or other sedative drugs in male and female control C57BL/6J mice versus mice lacking CD14, TLR2, TLR4 (C57BL/10ScN), or MyD88. We also examined EtOH-induced hypothermia and blood EtOH clearance in these mice. RESULTS Male and female mice lacking TLR4 or MyD88 showed reduced duration of EtOH-induced LORR and faster recovery from EtOH-induced motor incoordination in the rotarod test. MyD88 knockout mice had slightly faster recovery from EtOH-induced hypothermia compared to control mice. None of the mutants differed from control mice in the rate of blood EtOH clearance. All of the mutants showed similar decreases in the duration of gaboxadol-induced LORR, but only mice lacking TLR4 were less sensitive to the sedative effects of pentobarbital. Faster recovery from diazepam-induced motor impairment was observed in CD14, TLR4, and MyD88 null mice of both sexes. CONCLUSIONS TLR4 and MyD88 were key mediators of the sedative and intoxicating effects of EtOH and GABAergic sedatives, indicating a strong influence of TLR4-MyD88 signaling on GABAergic function. Despite the involvement of TLR4 in EtOH's acute behaviors, it did not regulate EtOH consumption in any drinking model as shown in our companion article. Collectively, our studies demonstrate differential effects of TLR-MyD88 components in the acute versus chronic actions of EtOH.
Collapse
Affiliation(s)
- Yuri A Blednov
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, Texas
| | - Mendy Black
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, Texas
| | - Jillian M Benavidez
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, Texas
| | - Adriana Da Costa
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, Texas
| | - Jody Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, Texas
| | - R Adron Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
42
|
The role of neuroimmune signaling in alcoholism. Neuropharmacology 2017; 122:56-73. [PMID: 28159648 DOI: 10.1016/j.neuropharm.2017.01.031] [Citation(s) in RCA: 216] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 01/24/2017] [Accepted: 01/29/2017] [Indexed: 02/07/2023]
Abstract
Alcohol consumption and stress increase brain levels of known innate immune signaling molecules. Microglia, the innate immune cells of the brain, and neurons respond to alcohol, signaling through Toll-like receptors (TLRs), high-mobility group box 1 (HMGB1), miRNAs, pro-inflammatory cytokines and their associated receptors involved in signaling between microglia, other glia and neurons. Repeated cycles of alcohol and stress cause a progressive, persistent induction of HMGB1, miRNA and TLR receptors in brain that appear to underlie the progressive and persistent loss of behavioral control, increased impulsivity and anxiety, as well as craving, coupled with increasing ventral striatal responses that promote reward seeking behavior and increase risk of developing alcohol use disorders. Studies employing anti-oxidant, anti-inflammatory, anti-depressant, and innate immune antagonists further link innate immune gene expression to addiction-like behaviors. Innate immune molecules are novel targets for addiction and affective disorders therapies. This article is part of the Special Issue entitled "Alcoholism".
Collapse
|
43
|
Suryanarayanan A, Carter JM, Landin JD, Morrow AL, Werner DF, Spigelman I. Role of interleukin-10 (IL-10) in regulation of GABAergic transmission and acute response to ethanol. Neuropharmacology 2016; 107:181-188. [PMID: 27016017 PMCID: PMC5076550 DOI: 10.1016/j.neuropharm.2016.03.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/08/2016] [Accepted: 03/21/2016] [Indexed: 12/27/2022]
Abstract
Mounting evidence indicates that ethanol (EtOH) exposure activates neuroimmune signaling. Alterations in pro-inflammatory cytokines after acute and chronic EtOH exposure have been heavily investigated. In contrast, little is known about the regulation of neurotransmission and/or modulation by anti-inflammatory cytokines in the brain after an acute EtOH exposure. Recent evidence suggests that interleukin-10 (IL-10), an anti-inflammatory cytokine, is upregulated during withdrawal from chronic EtOH exposure. In the present study, we show that IL-10 is increased early (1 h) after a single intoxicating dose of EtOH (5 g/kg, intragastric) in Sprague Dawley rats. We also show that IL-10 rapidly regulates GABAergic transmission in dentate gyrus neurons. In brain slice recordings, IL-10 application dose-dependently decreases miniature inhibitory postsynaptic current (mIPSC) area and frequency, and decreases the magnitude of the picrotoxin sensitive tonic current (Itonic), indicating both pre- and postsynaptic mechanisms. A PI3K inhibitor LY294002 (but not the negative control LY303511) ablated the inhibitory effects of IL-10 on mIPSC area and Itonic, but not on mIPSC frequency, indicating the involvement of PI3K in postsynaptic effects of IL-10 on GABAergic transmission. Lastly, we also identify a novel neurobehavioral regulation of EtOH sensitivity by IL-10, whereby IL-10 attenuates acute EtOH-induced hypnosis. These results suggest that EtOH causes an early release of IL-10 in the brain, which may contribute to neuronal hyperexcitability as well as disturbed sleep seen after binge exposure to EtOH. These results also identify IL-10 signaling as a potential therapeutic target in alcohol-use disorders and other CNS disorders where GABAergic transmission is altered.
Collapse
Affiliation(s)
- A Suryanarayanan
- Department of Pharmaceutical Sciences, University of the Sciences, Philadelphia College of Pharmacy, Philadelphia, PA 19104, USA.
| | - J M Carter
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY 13902, USA
| | - J D Landin
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY 13902, USA
| | - A L Morrow
- Departments of Psychiatry and Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - D F Werner
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY 13902, USA
| | - I Spigelman
- Division of Oral Biology & Medicine, School of Dentistry, 63-078 CHS, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
44
|
Breese GR, Knapp DJ. Persistent adaptation by chronic alcohol is facilitated by neuroimmune activation linked to stress and CRF. Alcohol 2016; 52:9-23. [PMID: 27139233 PMCID: PMC4855305 DOI: 10.1016/j.alcohol.2016.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 12/10/2015] [Accepted: 01/24/2016] [Indexed: 01/11/2023]
Abstract
This review updates the conceptual basis for the association of alcohol abuse with an insidious adaptation that facilitates negative affect during withdrawal from chronic intermittent alcohol (CIA) exposure - a change that later supports sensitization of stress-induced anxiety following alcohol abstinence. The finding that a CRF1-receptor antagonist (CRF1RA) minimized CIA withdrawal-induced negative affect supported an association of alcohol withdrawal with a stress mechanism. The finding that repeated stresses or multiple CRF injections into selected brain sites prior to a single 5-day chronic alcohol (CA) exposure induced anxiety during withdrawal provided critical support for a linkage of CIA withdrawal with stress. The determination that CRF1RA injection into positive CRF-sensitive brain sites prevented CIA withdrawal-induced anxiety provided support that neural path integration maintains the persistent CIA adaptation. Based upon reports that stress increases neuroimmune function, an effort was undertaken to test whether cytokines would support the adaptation induced by stress/CA exposure. Twenty-four hours after withdrawal from CIA, cytokine mRNAs were found to be increased in cortex as well as other sites in brain. Further, repeated cytokine injections into previously identified brain sites substituted for stress and CRF induction of anxiety during CA withdrawal. Discovery that a CRF1RA prevented the brain cytokine mRNA increase induced by CA withdrawal provided critical evidence for CRF involvement in this neuroimmune induction after CA withdrawal. However, the CRF1RA did not block the stress increase in cytokine mRNA increases in controls. The latter data supported the hypothesis that distinct mechanisms linked to stress and CA withdrawal can support common neuroimmune functions within a brain site. As evidence evolves concerning neural involvement in brain neuroimmune function, a better understanding of the progressive adaptation associated with CIA exposure will advance new knowledge that could possibly lead to strategies to combat alcohol abuse.
Collapse
Affiliation(s)
- George R Breese
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Curriculum in Neurobiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; The UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA.
| | - Darin J Knapp
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA
| |
Collapse
|
45
|
Marshall SA, Casachahua JD, Rinker JA, Blose AK, Lysle DT, Thiele TE. IL-1 receptor signaling in the basolateral amygdala modulates binge-like ethanol consumption in male C57BL/6J mice. Brain Behav Immun 2016; 51:258-267. [PMID: 26365025 PMCID: PMC4679505 DOI: 10.1016/j.bbi.2015.09.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/26/2015] [Accepted: 09/09/2015] [Indexed: 12/26/2022] Open
Abstract
Proinflammatory cytokines have been implicated in alcohol-induced neurodegeneration, but the role of the neuroimmune system in alcohol related behaviors has only recently come to the forefront. Herein, the effects of binge-like drinking on IL-1β mRNA and immunoreactivity within the amygdala were measured following the "drinking in the dark" (DID) paradigm, a model of binge-like ethanol drinking in C57BL/6J mice. Moreover, the role of IL-1 receptor signaling in the amygdala on ethanol consumption was assessed. Results indicated that a history of binge-like ethanol drinking promoted a significant increase of IL-1β mRNA expression within the amygdala, and immunohistochemistry analyses revealed that the basolateral amygdala (BLA), but not central amygdala (CeA), exhibited significantly increased IL-1β immunoreactivity. However, Fluoro-Jade® C labeling indicated that multiple cycles of the DID paradigm were not sufficient to elicit neuronal death. Bilateral infusions of IL-1 receptor antagonist (IL-1Ra) reduced ethanol consumption when infused into the BLA but not the CeA. These observations were specific to ethanol drinking as the IL-1Ra did not alter either sucrose drinking or open-field locomotor activity. The current findings highlight a specific role for IL-1 receptor signaling in modulating binge-like ethanol consumption and indicate that proinflammatory cytokines can be induced prior to dependence or any evidence of neuronal cell death. These findings provide a framework in which to understand how neuroimmune adaptations may alter ethanol consumption and therein contribute to alcohol abuse.
Collapse
Affiliation(s)
- S Alex Marshall
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - John D Casachahua
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jennifer A Rinker
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Allyson K Blose
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Donald T Lysle
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Todd E Thiele
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
46
|
Olive MF, Gass JT. Editorial: structural plasticity induced by drugs of abuse. Front Pharmacol 2015; 6:88. [PMID: 26029104 PMCID: PMC4432576 DOI: 10.3389/fphar.2015.00088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 04/10/2015] [Indexed: 01/11/2023] Open
Affiliation(s)
- M Foster Olive
- Behavioral Neuroscience Area, Department of Psychology, Arizona State University Tempe, AZ, USA
| | - Justin T Gass
- Department of Neurosciences, Medical University of South Carolina Charleston, SC, USA
| |
Collapse
|