1
|
Liss A, Siddiqi MT, Marsland P, Varodayan FP. Neuroimmune regulation of the prefrontal cortex tetrapartite synapse. Neuropharmacology 2025; 269:110335. [PMID: 39904409 DOI: 10.1016/j.neuropharm.2025.110335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
The prefrontal cortex (PFC) is an essential driver of cognitive, affective, and motivational behavior. There is clear evidence that the neuroimmune system directly influences PFC synapses, in addition to its role as the first line of defense against toxins and pathogens. In this review, we first describe the core structures that form the tetrapartite PFC synapse, focusing on the signaling microdomain created by astrocytic cradling of the synapse as well as the emerging role of the extracellular matrix in synaptic organization and plasticity. Neuroimmune signals (e.g. pro-inflammatory interleukin 1β) can impact the function of each core structure within the tetrapartite synapse, as well as promote intra-synaptic crosstalk, and we will provide an overview of recent advances in this field. Finally, evidence from post mortem human brain tissue and preclinical studies indicate that inflammation may be a key contributor to PFC dysfunction. Therefore, we conclude with a mechanistic discussion of neuroimmune-mediated maladaptive plasticity in neuropsychiatric disorders, with a focus on alcohol use disorder (AUD). Growing recognition of the neuroimmune system's role as a critical regulator of the PFC tetrapartite synapse provides strong support for targeting the neuroimmune system to develop new pharmacotherapeutics.
Collapse
Affiliation(s)
- Andrea Liss
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Mahum T Siddiqi
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Paige Marsland
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Florence P Varodayan
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA.
| |
Collapse
|
2
|
Diaz MR, Barney TM, Marsland P, Deak T. Age- and cytokine-dependent modulation of GABAergic transmission within the basolateral amygdala of male Sprague Dawley rats. Neuropharmacology 2025; 267:110304. [PMID: 39827996 DOI: 10.1016/j.neuropharm.2025.110304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/02/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Alcohol binge drinking has a multitude of effects on CNS function, including changes in inflammatory cytokines such as IL-6 and IL-1β that may contribute to mood fluctuations associated with the intoxication-withdrawal cycle. Widely throughout the brain, including the amygdala, IL-6 mRNA is enhanced during intoxication, whereas IL-1β is initially suppressed during alcohol intoxication, with increased expression seen shortly after ethanol clearance, during acute hangover. Furthermore, induction of neuroimmune genes appears to be muted during adolescence in the amygdala, suggesting a broader functional immaturity of the adolescent neuroimmune system in structures involved in negative affect associated with ethanol exposure. However, neither the effect of IL-6 or IL-1β on synaptic function within the amygdala nor the impact of acute intoxication and withdrawal on these cytokines' function are known. To test this, we used whole-cell patch-clamp electrophysiology to assess the effects of IL-6 and IL-1β on GABA-mediated spontaneous inhibitory postsynaptic currents (sIPSCs) in BLA pyramidal neurons from male rats in early adolescence (P28-40) or adulthood (P70+). These experiments were done in naïve, intoxicated (3-4 h following an intraperitoneal injection of 3.5 g/kg ethanol), and during acute hangover (11-18 h post ethanol injection). In naïve males, we found that IL-6 (10 ng/ml) significantly enhanced sIPSC amplitude only in adults, with no apparent effect in adolescents; this effect of IL-6 in adults was not different during intoxication. Conversely, IL-1β (10 ng/ml) did not alter sIPSC frequency in any group (naïve or hangover adolescents or adults). Unlike our previous work in adult rats, here we found that contextual fear conditioning was not altered in adolescents when conditioned during acute hangover. Together, these observations suggest that IL-6, but not IL-1β, regulation of BLA GABA transmission emerges as a function of age, but is not affected by acute ethanol exposure or hangover for adolescents or adults. Importantly, these findings provide additional evidence to support functional immaturity of the neuroimmune system in adolescence.
Collapse
Affiliation(s)
- Marvin R Diaz
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton, NY, 13902-6000, USA
| | - Thaddeus M Barney
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton, NY, 13902-6000, USA
| | - Paige Marsland
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton, NY, 13902-6000, USA
| | - Terrence Deak
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton, NY, 13902-6000, USA.
| |
Collapse
|
3
|
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
|
4
|
Benarroch E. What Is the Role of Cytokines in Synaptic Transmission? Neurology 2024; 103:e209928. [PMID: 39303183 DOI: 10.1212/wnl.0000000000209928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
|
5
|
Liss A, Siddiqi M, Podder D, Scroger M, Vessey G, Martin K, Paperny N, Vo K, Astefanous A, Belachew N, Idahor E, Varodayan F. Ethanol drinking sex-dependently alters cortical IL-1β synaptic signaling and cognitive behavior in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617276. [PMID: 39416094 PMCID: PMC11483015 DOI: 10.1101/2024.10.08.617276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Individuals with alcohol use disorder (AUD) struggle with inhibitory control, decision making, and emotional processing. These cognitive symptoms reduce treatment adherence, worsen clinical outcomes, and promote relapse. Neuroimmune activation is a key factor in the pathophysiology of AUD, and targeting this modulatory system is less likely to produce unwanted side effects compared to directly targeting neurotransmitter dysfunction. Notably, the cytokine interleukin-1β (IL-1β) has been broadly associated with the cognitive symptoms of AUD, though the underlying mechanisms are not well understood. Here we investigated how chronic intermittent 24-hour access two bottle choice ethanol drinking affects medial prefrontal cortex (mPFC)-related cognitive function and IL-1 synaptic signaling in male and female C57BL/6J mice. In both sexes, ethanol drinking decreased reference memory and increased mPFC IL-1 receptor 1 (IL-1R1) mRNA levels. In neurons, IL-1β can activate either pro-inflammatory or neuroprotective intracellular pathways depending on the isoform of the accessory protein (IL-1RAcP) recruited to the IL-1R1 complex. Moreover, ethanol drinking sex-dependently shifted mPFC IL-1RAcP isoform gene expression and IL-1β regulation of mPFC GABA synapses, both of which may contribute to female mPFC resiliency and male mPFC susceptibility. This type of signaling bias has become a recent focus of rational drug development. Therefore, in addition to increasing our understanding of how IL-1β sex-dependently contributes to mPFC dysfunction in AUD, our current findings also support the development of a new class of pharmacotherapeutics based on biased IL-1 signaling.
Collapse
|
6
|
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
|
7
|
Borgonetti V, Roberts AJ, Bajo M, Galeotti N, Roberto M. Chronic alcohol induced mechanical allodynia by promoting neuroinflammation: A mouse model of alcohol-evoked neuropathic pain. Br J Pharmacol 2023; 180:2377-2392. [PMID: 37050867 PMCID: PMC10898491 DOI: 10.1111/bph.16091] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Chronic pain is considered a key factor contributing to alcohol use disorder (AUD). The mechanisms responsible for chronic pain associated with chronic alcohol consumption are unknown. We evaluated the development of chronic pain in a mouse model of alcohol dependence and investigate the role of neuroinflammation. EXPERIMENTAL APPROACH The chronic-intermittent ethanol two-bottle choice CIE-2BC paradigm generates three groups: alcohol-dependent with escalating alcohol intake, nondependent (moderate drinking) and alcohol-naïve control male and female mice. We measured mechanical allodynia during withdrawal and after the last voluntary drinking. Immunoblotting was used to evaluate the protein levels of IBA-1, CSFR, IL-6, p38 and ERK2/1 in spinal cord tissue of dependent and non-dependent animals. KEY RESULTS We found significant escalation of drinking in the dependent group in male and female compared with the non-dependent group. The dependent group developed mechanical allodynia during 72 h of withdrawal, which was completely reversed after voluntary drinking. We observed an increased pain hypersensitivity compared with the naïve in 50% of non-dependent group. Increased IBA-1 and CSFR expression was observed in spinal cord tissue of both hypersensitivity-abstinence related and neuropathy-alcohol mice, and increased IL-6 expression and ERK1/2 activation in mice with hypersensitivity-related to abstinence, but not in mice with alcohol-evoked neuropathic pain. CONCLUSIONS AND IMPLICATIONS The CIE-2BC model induces two distinct pain conditions specific to the type of ethanol exposure: abstinence-related hypersensitivity in dependent mice and alcohol-evoked neuropathic pain in about a half of the non-dependent mice.
Collapse
Affiliation(s)
- Vittoria Borgonetti
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, Florence, 50139, Italy
- Department of Molecular Medicine and Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Amanda J. Roberts
- Animal Models Core, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Michal Bajo
- Department of Molecular Medicine and Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, Florence, 50139, Italy
| | - Marisa Roberto
- Department of Molecular Medicine and Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| |
Collapse
|
8
|
Mondello JE, Gano A, Vore AS, Deak T. Cues associated with repeated ethanol exposure facilitate the corticosterone response to ethanol and immunological challenges in adult male Sprague Dawley rats: implications for neuroimmune regulation. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2023; 49:359-369. [PMID: 36862971 PMCID: PMC10474242 DOI: 10.1080/00952990.2023.2169831] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 01/06/2023] [Accepted: 01/14/2023] [Indexed: 03/04/2023]
Abstract
Background: We previously found a conditioned increase in central neuroinflammatory markers (Interleukin 6; IL-6) following exposure to alcohol-associated cues. Recent studies suggest (unconditioned) induction of IL-6 is entirely dependent on ethanol-induced corticosterone.Objectives: The goals of these present studies were to test whether alcohol-paired cues facilitated the hypothalamic-pituitary-adrenal (HPA) axis response to either a subthreshold priming alcohol dose or an immune or psychological stress challengeMethods: In Experiment 1 (N = 64), adult male Sprague Dawley rats were trained (paired or unpaired, four pairings total) with either vehicle or 2 g/kg alcohol [intragastric (i.g.) or intraperitoneal (i.p.)] injections. In Experiments 2 (N = 28) and 3 (N = 30), male rats were similarly trained but with 4 g/kg alcohol i.g. intubations. On test day, all rats were either administered a 0.5 g/kg alcohol dose (i.p. or i.g. Experiment 1), a 100 µg/kg i.p. lipopolysaccharide (LPS) challenge (Experiment 2), or a restraint challenge (Experiment 3), and exposed to alcohol-associated cues. Blood plasma was collected for analysis.Results: Alcohol-associated cues facilitated the plasma corticosterone response to a subthreshold dose of alcohol (F1,28 = 4.85, p < .05) and an immune challenge (F8,80 = 6.23, p < .001), but not a restraint challenge (F2,27 = 0.18, p > .05).Conclusion: These findings reveal that the impact of the cues associated with alcohol intoxication on the HPA axis may be context-specific. This work illustrates how HPA axis learning processes form in the early stages of alcohol use and has important implications for how the HPA and neuroimmune conditioning may develop in alcohol use disorder in humans and facilitate the response to a later immune challenge.
Collapse
Affiliation(s)
- Jamie E. Mondello
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000, USA
| | - Anny Gano
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000, USA
| | - Andrew S. Vore
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000, USA
| | - Terrence Deak
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000, USA
| |
Collapse
|
9
|
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
|
10
|
Gano A, Lebonville CL, Becker HC. TLR3 activation with poly I:C exacerbates escalated alcohol consumption in dependent male C57BL/6J mice. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2022:1-12. [PMID: 36095319 DOI: 10.1080/00952990.2022.2092492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 06/15/2023]
Abstract
Background: Activation of TLR3 receptors, which are sensitive to viral infection, has emerged as a possible mechanism that increases alcohol intake in rodents.Objectives: These studies examined whether a history of ethanol dependence exacerbated the increase in drinking driven by the TLR3 agonist poly I:C.Methods: Male C57BL/6J mice (>10 per group) were given access to ethanol (20% v/v) 2 hours a day following a history of home cage drinking or after having been rendered ethanol-dependent using a chronic intermittent ethanol (CIE) vapor model. After testing multiple doses, a 5 mg/kg repeated poly I:C challenge was used to probe the effects of repeated immune challenge, alone or in conjunction with repeated cycles of CIE, on voluntary drinking. An ethanol (12% v/v) operant self-administration model was used to test the effects of poly I:C on stress-induced reinstatement of ethanol seeking and consumption.Results: Poly I:C in naive animals resulted in transient, modest increases in ethanol intake in the home cage and in self-administration (p < 0.05). However, poly I:C challenge resulted in sensitized stress-induced ethanol consumption and evoked a strong and persistent escalation of drinking in mice with a history of dependence (p < 0.05 for both).Conclusion: Activation of viral immune defense may affect ethanol consumption in dependence and sensitivity to future stressors. As patients who suffer from alcohol use disorder are at a heightened risk for viral infection, this interaction could generate risk factors for exacerbating behaviors associated with Alcohol Use Disorders via an immune mechanism.
Collapse
Affiliation(s)
- Anny Gano
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Christina L Lebonville
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- RHJ Department of Veterans Affairs Medical Center, Charleston, SC, USA
| |
Collapse
|
11
|
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: 3] [Impact Index Per Article: 1.0] [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
|
12
|
Nwachukwu KN, King DM, Healey KL, Swartzwelder HS, Marshall SA. Sex-specific effects of adolescent intermittent ethanol exposure-induced dysregulation of hippocampal glial cells in adulthood. Alcohol 2022; 100:31-39. [PMID: 35182671 PMCID: PMC8983575 DOI: 10.1016/j.alcohol.2022.02.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 12/11/2022]
Abstract
Adolescent alcohol abuse is a significant public health concern, with approximately 4.3 million U.S. adolescents reporting monthly binge drinking. Excessive ethanol consumption during adolescence has been linked to dysregulation of the neuroimmune system, particularly in the hippocampus. Because there are sex differences in both neuroimmune responses and ethanol's pharmacologic actions, this study tested whether there were disparate effects based on sex in glial cells and neurodegeneration in adulthood after the adolescent intermittent ethanol (AIE) model. Male and female adolescent Sprague-Dawley rats underwent AIE. In adulthood, immunohistochemical techniques were utilized to determine the effects of AIE on astrocytes and microglia, and Fluoro-Jade C (FJC) was used to assess neurodegeneration in the hippocampus. AIE exposure significantly increased astrocyte activation in the cornu ammonis 1 (CA1), CA2/3, and dentate gyrus (DG) in both male and female rats with no discernible sex differences in immunoreactivity. Likewise, the number of GFAP + cells was significantly increased by AIE across the hippocampus. In our microglial assessment, AIE only led to increased Iba1 immunoreactivity in the CA1 but not CA2/3 or DG regions. However, the number of Iba1+ cells was increased by AIE in both the CA1 and DG subregions. In the DG, the ethanol effect was observed in both sexes, but in the CA1, AIE-induced increased Iba1 cells were only observed in females. In regard to neurodegeneration, there were no persisting AIE effects on FJC + cells. These findings indicate that AIE alters hippocampal glial cells in adulthood, in the absence of active neurodegeneration. However, while AIE induced long-term elevation of astroglial measures in both males and females, persisting AIE-induced microglial activation was more sparse and sex-dependent. While the majority of these findings suggest that AIE has similar effects on glial morphology and number between males and females, additional work should determine whether there are molecular differences as well as innate sex differences in glial interaction with AIE's influence on glial functions in behavior.
Collapse
Affiliation(s)
- Kala N Nwachukwu
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, 27707, United States
| | - Dantae M King
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, 27707, United States
| | - Kati L Healey
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, 27708, United States
| | - H Scott Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, 27708, United States
| | - S Alex Marshall
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, 27707, United States.
| |
Collapse
|
13
|
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
|
14
|
Phillips TJ. Genetic and Brain Mechanisms of Addictive Behavior and Neuroadaptation. Brain Sci 2021; 12:51. [PMID: 35053795 PMCID: PMC8773497 DOI: 10.3390/brainsci12010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 11/23/2022] Open
Abstract
Genetic differences play a role in the susceptibility to addictive drug use, the probability that the use of these drugs will escalate and result in a drug use disorder, and whether relapse to use will occur during or after treatment [...].
Collapse
Affiliation(s)
- Tamara J. Phillips
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA;
- Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, USA
- Veterans Affairs Portland Health Care System, Portland, OR 97239, USA
| |
Collapse
|
15
|
Tang Y, Tang Z, Yang J, Liu T, Tang Y. MicroRNA-7-5p Inhibits Migration, Invasion and Metastasis of Intrahepatic Cholangiocarcinoma by Inhibiting MyD88. J Clin Transl Hepatol 2021; 9:809-817. [PMID: 34966644 PMCID: PMC8666375 DOI: 10.14218/jcth.2021.00021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/29/2021] [Accepted: 04/08/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS Intrahepatic cholangiocarcinoma (ICC) is a malignant tumor derived from intrahepatic bile duct epithelial cells. Accumulating studies report that microRNAs are widely involved in tumor migration and metastasis by regulation of target genes. miR-7-5p has been confirmed to inhibit tumor metastasis and to be related to prognosis for several malignant tumors. Our study investigated the underlying functions of miR-7-5p in ICC. METHODS The expression of miR-7-5p in ICC tissues but also in ICC cell lines was analyzed by real-time PCR. By analyzing the relationship between the clinicopathological parameters of 60 ICC patients and the expression level of miR-7-5p, the effect of miR-7-5p on the prognosis was clarified. After transfected with miR-7-5p mimics or miR-7-5p inhibitor, cell counting kit-8 assay was applied to evaluate the cells proliferation, flow cytometry was applied to analyze the cells apoptosis, wound healing assay and transwell chamber assay were applied to analyze the cell invasion and migration. A luciferase reporter assay was identified the relationship of miR-7-5p and myeloid differentiation factor 88 (MyD88). Western blotting was used to analyze the proteins expression. And immunochemistry was performed to determine the expression of MYD88 in ICC tissues. RESULTS Our data showed the expression of miR-7-5p was down-regulated not only in ICC tissues but also in ICC cell lines compared with normal controls. Low expression of miR-7-5p was notably associated with poor prognosis in ICC patients. miR-7-5p negatively regulated cell proliferation, migration, invasion and apoptosis in ICC cells. We further verified that MyD88 was a novel target of miR-7-5p and was significantly overexpressed in ICC tissues. Overexpression of MyD88 counteracted the effects of miR-7-5p in ICC cells. CONCLUSIONS The present findings suggest that miR-7-5p plays a pivotal role in ICC invasion by regulating MyD88. Ampliative insight into the key factors of ICC invasion may result in the development of new treatment options for ICC.
Collapse
Affiliation(s)
| | | | | | | | - Yuntian Tang
- Correspondence to: Yuntian Tang, Department of Hepatobiliary Surgery, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China. ORCID: https://orcid.org/0000-0001-7658-870X. Tel/Fax: +86-771-218-6308, E-mail:
| |
Collapse
|
16
|
Abstract
Interleukin-1 (IL-1) is an inflammatory cytokine that has been shown to modulate neuronal signaling in homeostasis and diseases. In homeostasis, IL-1 regulates sleep and memory formation, whereas in diseases, IL-1 impairs memory and alters affect. Interestingly, IL-1 can cause long-lasting changes in behavior, suggesting IL-1 can alter neuroplasticity. The neuroplastic effects of IL-1 are mediated via its cognate receptor, Interleukin-1 Type 1 Receptor (IL-1R1), and are dependent on the distribution and cell type(s) of IL-1R1 expression. Recent reports found that IL-1R1 expression is restricted to discrete subpopulations of neurons, astrocytes, and endothelial cells and suggest IL-1 can influence neural circuits directly through neuronal IL-1R1 or indirectly via non-neuronal IL-1R1. In this review, we analyzed multiple mechanisms by which IL-1/IL-1R1 signaling might impact neuroplasticity based upon the most up-to-date literature and provided potential explanations to clarify discrepant and confusing findings reported in the past.
Collapse
Affiliation(s)
- Daniel P. Nemeth
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| |
Collapse
|
17
|
Yang J, Jia Z, Xiao Z, Zhao J, Lu Y, Chu L, Shao H, Pei L, Zhang S, Chen Y. Baicalin Rescues Cognitive Dysfunction, Mitigates Neurodegeneration, and Exerts Anti-Epileptic Effects Through Activating TLR4/MYD88/Caspase-3 Pathway in Rats. Drug Des Devel Ther 2021; 15:3163-3180. [PMID: 34321866 PMCID: PMC8312624 DOI: 10.2147/dddt.s314076] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose This study aims to evaluate the beneficial effects of anti-epileptic mechanisms of baicalin (BA) on cognitive dysfunction and neurodegeneration in pentylenetetrazol (PTZ)-induced epileptic rats. Methods First, PTZ-induced epileptic rats were administered intraperitoneally a sub-convulsive dose of PTZ (40 mg/kg) daily, and the seizure susceptibility (the degree of seizures and latency) was evaluated using Racine’s criterion. Then, classical behavioral experiments were performed to test whether BA ameliorated cognitive dysfunction. Neurodegeneration was assessed using Fluoro Jade-B (FJB), and NeuN staining was used to determine whether BA offered a neuroprotective role. After BA had been proven to possess anti-epileptic effects, its possible mechanisms were analyzed through network pharmacology. Finally, the key targets for predictive mechanisms were experimentally verified. Results The epileptic model was successfully established, and BA had anti-epileptic effects. Epileptic rats displayed significant cognitive dysfunction, and BA markedly ameliorated cognitive dysfunction. Further, we also discovered that BA treatment mitigated neurodegeneration of the hippocampus CA3 regions, thereby ameliorated cognitive dysfunction of epileptic rats. Subsequent network pharmacology analysis was implemented to reveal a possible mechanism of BA in the anti-epileptic process and the TLR4/MYD88/Caspase-3 pathway was predicted. Finally, experimental studies showed that BA exerted an anti-epileptic effect by activating the TLR4/MYD88/Caspase-3 pathway in PTZ-induced epileptic rats. Conclusion In conclusion, BA had a protective effect against PTZ-induced seizures. BA improved cognitive dysfunction and exerted a neuroprotective action. The anti-epileptic effects of BA may be potentially through activation of the TLR4/MYD88/Caspase-3 pathway.
Collapse
Affiliation(s)
- Jiali Yang
- School of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People's Republic of China
| | - Zhixia Jia
- School of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People's Republic of China
| | - Zhigang Xiao
- School of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People's Republic of China
| | - Jing Zhao
- Hebei Key Laboratory of Turbidity, Hebei Academy of Chinese Medicine Sciences, Shijiazhuang, Hebei, 050011, People's Republic of China
| | - Ye Lu
- Hebei Key Laboratory of Turbidity, Hebei Academy of Chinese Medicine Sciences, Shijiazhuang, Hebei, 050011, People's Republic of China
| | - Li Chu
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People's Republic of China
| | - Hui Shao
- School of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People's Republic of China.,Hebei Key Laboratory of Turbidity, Hebei Academy of Chinese Medicine Sciences, Shijiazhuang, Hebei, 050011, People's Republic of China
| | - Lin Pei
- School of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, People's Republic of China.,Hebei Key Laboratory of Turbidity, Hebei Academy of Chinese Medicine Sciences, Shijiazhuang, Hebei, 050011, People's Republic of China
| | - Shaodan Zhang
- Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Yuan Chen
- Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
| |
Collapse
|
18
|
IL-10 normalizes aberrant amygdala GABA transmission and reverses anxiety-like behavior and dependence-induced escalation of alcohol intake. Prog Neurobiol 2020; 199:101952. [PMID: 33197496 DOI: 10.1016/j.pneurobio.2020.101952] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/13/2020] [Accepted: 11/06/2020] [Indexed: 12/27/2022]
Abstract
Alcohol elicits a neuroimmune response in the brain contributing to the development and maintenance of alcohol use disorder (AUD). While pro-inflammatory mediators initiate and drive the neuroimmune response, anti-inflammatory mediators provide an important homeostatic mechanism to limit inflammation and prevent pathological damage. However, our understanding of the role of anti-inflammatory signaling on neuronal physiology in critical addiction-related brain regions and pathological alcohol-dependence induced behaviors is limited, precluding our ability to identify promising therapeutic targets. Here, we hypothesized that chronic alcohol exposure compromises anti-inflammatory signaling in the central amygdala, a brain region implicated in anxiety and addiction, consequently perpetuating a pro-inflammatory state driving aberrant neuronal activity underlying pathological behaviors. We found that alcohol dependence alters the global brain immune landscape increasing IL-10 producing microglia and T-regulatory cells but decreasing local amygdala IL-10 levels. Amygdala IL-10 overexpression decreases anxiety-like behaviors, suggesting its local role in regulating amygdala-mediated behaviors. Mechanistically, amygdala IL-10 signaling through PI3K and p38 MAPK modulates GABA transmission directly at presynaptic terminals and indirectly through alterations in spontaneous firing. Alcohol dependence-induces neuroadaptations in IL-10 signaling leading to an overall IL-10-induced decrease in GABA transmission, which normalizes dependence-induced elevated amygdala GABA transmission. Notably, amygdala IL-10 overexpression abolishes escalation of alcohol intake, a diagnostic criterion of AUD, in dependent mice. This highlights the importance of amygdala IL-10 signaling in modulating neuronal activity and underlying anxiety-like behavior and aberrant alcohol intake, providing a new framework for therapeutic intervention.
Collapse
|
19
|
Chen J, Xia D, Xu M, Su R, Lin W, Guo D, Chen G, Liu S. Expression and Significance of MyD88 in Patients With Gastric Cardia Cancer in a High-Incidence Area of China. Front Oncol 2020; 10:559. [PMID: 32477927 PMCID: PMC7239990 DOI: 10.3389/fonc.2020.00559] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 03/27/2020] [Indexed: 02/05/2023] Open
Abstract
Background: Gastric cardia cancer (GCC) arises in the area of the stomach adjoining the esophageal-gastric junction and has unique risk factors. It was suggested that the involvement of Helicobacter pylori is associated with GCC from high-risk population. Myeloid differentiation factor 88 (MyD88) is a crucial adaptor molecule in Toll-like signaling pathway recognizing H. pylori. Its role in GCC has not been elucidated yet. In this study, our purpose is to investigate the expression and significance of MyD88 in GCC tissue. Methods: Expression of MyD88 and nuclear factor κB (NF-κB) p105/p50 and infection of H. pylori were detected by immunohistochemistry in gastric cardia tissue. The correlation of MyD88 expression to NF-κB p105/p50 expression, H. pylori infection, and clinicopathologic characteristics in gastric cardia tissue was analyzed. The involvement of MyD88 in patient prognosis was also analyzed. Results: Our data showed that the expression of MyD88 elevated from normal mucosa to inflammation (p = 0.071). The expression of MyD88 was enhanced in GCC tissues by contrast to non-malignant cardia mucosa (p = 0.025). What's more, overexpression of MyD88 was detected in intestinal-type adenocarcinoma with inflammation. Patients with high MyD88 staining revealed a better differentiation (p = 0.02). MyD88 also positively correlated with NF-κB p105/p50 expression (p = 0.012) in cancer tissue. Expression of MyD88 was increased but not significantly in biopsies with H. pylori infection compared with non-infected biopsies. Multivariate analyses revealed lymph node metastasis but not MyD88 expression was an independent predictor for patient survival. Conclusion: These findings provide pathological evidence that upregulating MyD88 and inducing inflammation might be involved in gastric cardia carcinogenesis in high-risk population. MyD88 plays a role in gastric cardia carcinogenesis with NF-κB pathway activation. Higher MyD88 expression is not a major prognostic determinant in GCC, but it may relate to the tumor cell differentiation.
Collapse
Affiliation(s)
- Jingyao Chen
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Di Xia
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Muming Xu
- Department of Abdominal Surgery, The Tumor Hospital of Shantou University Medical College, Shantou, China
| | - Ruibing Su
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Wenting Lin
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Dan Guo
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Guangcan Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Guangcan Chen
| | - Shuhui Liu
- Department of Pathology, Shantou University Medical College, Shantou, China
- Shuhui Liu
| |
Collapse
|